Next Article in Journal
Phytosynthesis and Characterization of Silver Nanoparticles from Antigonon leptopus: Assessment of Antibacterial and Cytotoxic Properties
Previous Article in Journal
Preparation and Characterization of Ternary Complexes to Improve the Solubility and Dissolution Performance of a Proteolysis-Targeting Chimera Drug
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Herbal Medicine: Enhancing the Anticancer Potential of Natural Products in Hepatocellular Carcinoma Therapy Through Advanced Drug Delivery Systems

by
Ghazala Muteeb
1,*,
Manar T. El-Morsy
2,
Mustafa Ali Abo-Taleb
3,
Salma K. Mohamed
4 and
Doaa S. R. Khafaga
5,*
1
Department of Nursing, College of Applied Medical Sciences, King Faisal University, Al-Ahsa 31982, Saudi Arabia
2
Bio-Nanotechnology Department, Faculty of Nanotechnology, Cairo University, Giza 12613, Egypt
3
Biotechnology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
4
Faculty of Medicine, Galala University, Suez 43511, Egypt
5
Health Sector, Faculty of Science, Galala University, Suez 43511, Egypt
*
Authors to whom correspondence should be addressed.
Pharmaceutics 2025, 17(5), 673; https://doi.org/10.3390/pharmaceutics17050673
Submission received: 20 March 2025 / Revised: 14 May 2025 / Accepted: 14 May 2025 / Published: 20 May 2025

Abstract

:
Hepatocellular carcinoma (HCC) is an aggressive and prevalent liver cancer with a poor prognosis. Nanotechnology combined with natural products has emerged as a promising strategy to enhance HCC treatment efficacy. This review assesses the current literature on the application of nanotechnology in delivering natural products for HCC therapy. A comprehensive search was conducted in PubMed, Science Direct, Web of Science, and Google Scholar to identify relevant studies published up to the present articles focusing on nanotechnology-based drug delivery systems using natural products for HCC therapy, including different nanoparticle (NP) formulations and therapeutic interventions, were included. Natural products with anticancer properties have been encapsulated using various nanocarriers such as liposomes, polymeric nanoparticles, and quantum dots, which have improved drug stability, prolonged circulation time, and enhanced targeted delivery to HCC cells. These advancements have led to increased therapeutic efficacy and reduced side effects. Additionally, combining multiple natural products or integrating them with conventional therapies via nanocarriers enables personalized treatment approaches based on patient characteristics and molecular profiles. The integration of nanotechnology with natural products shows great potential for improving HCC treatment outcomes, representing a significant advancement in precision medicine for liver cancer and paving the way for more effective and personalized therapeutic strategies.

1. Introduction

Global epidemiological trends in liver cancer incidence exhibit significant geographical variation [1]. Recent surveillance data indicate a notable increase in incidence rates in economically developed countries, especially in Western Europe, North America, and Oceania [1]. In contrast, various traditionally high-risk Asian countries have indicated decreasing rates, implying differing effects of preventive strategies, environmental factors, and healthcare interventions among regions [1]. The male-to-female rate ratios also vary across regions, with higher ratios observed in France and Egypt compared to sub-Saharan African and South American registries [2]. The global burden of liver cancer is anticipated to rise significantly, with a notable increase in new cases and deaths expected by 2040, especially in Asia [3]. As the third most common cause of cancer-related death worldwide, liver cancer ranks as the sixth most common type of cancer. In 2020, there were expected to be 830,180 fatalities and 905,677 new cases globally [4,5], and the rate of liver cancer escalated with advancing age [5]. It is estimated that HCC accounts for 80% of occurrences of liver cancer [4,5]. Most HCC cases have chronic liver disease, and most of these cases have cirrhosis as a contributing factor [6]. Alpha-fetoprotein (AFP) levels and ultrasonography are the primary methods used in HCC screening. Magnetic resonance imaging (MRI) or computed tomography (CT) is used to diagnose abnormalities. Nevertheless, pathological diagnosis is necessary in a small number of cases [7].
Furthermore, the most common cancer treatment approaches used worldwide for HCC therapy are traditional therapeutic techniques, which include radiation, chemotherapy, and surgical procedures [8,9,10]. These approaches have certain limitations, including non-specific biological distribution and non-targeted therapeutic delivery with an inadequate dosage of anticancer agents in the selected tumor microenvironment and some biologic imaging and diagnosis constraints [11]. Also, traditional chemotherapy and natural products have different therapeutic efficacy in cancer treatment due to differences in toxicity, resistance, and mechanisms of action [12,13]. Chemotherapy drugs such as cisplatin and doxorubicin are beneficial, but they frequently lead to drug resistance and severe damage in many organs [13,14]. Resistance to chemotherapy is a major concern, resulting in 80–90% of deaths caused by cancer [15]. Natural products can improve chemotherapy efficiency by sensitizing cancer cells and overcoming chemoresistance [16,17].
On the other hand, recent studies have reported a variety of different natural products for the possible treatment of HCC such as resveratrol, curcumin, and ginger extract, have been investigated as safe and effective anticancer agents with potentially fewer side effects [18,19,20,21]. Also, natural products have shown underlying molecular mechanisms involving apoptosis, anti-metastasis, and anti-angiogenesis [22,23,24]. Nanotechnology plays a significant role in the treatment of HCC. It offers targeted and precise therapy, overcoming the limitations of conventional cancer treatments such as chemotherapy, radiation, and surgery. Nanoparticles can be engineered to specifically target cancer cells, delivering drugs directly to them and minimizing damage to healthy cells [25,26,27]. Functionalized nanomaterials have been developed to embolize tumor vasculature, achieve high drug loading efficacy, enable tumor targeting, and provide controlled sustained release of drugs [28]. These nanoparticles can control drug release and enhance drug stability; furthermore, they improve drug accumulation in the target tumor because they can specifically enter neoplastic sites through passive and/or active pathways, therefore reducing drug toxicity [29,30]. Natural products with nanocarriers have shown promise in HCC therapy [31]. These nanocarriers can encapsulate bioactives from medicinal plants, allowing for targeted delivery to tumor areas with minimal damage to healthy cells [32]. Furthermore, using natural bioactive-loaded nanocarriers has shown promising therapeutic efficacy in HCC treatment, providing targeted delivery of natural active ingredients. Overall, natural products offer a potential alternative to traditional chemotherapy in the treatment of HCC, with the advantage of potentially lower toxicity and multi-level effects. This review aims to focus on the effective role of delivery systems to enhance the anticancer effect of natural products to treat HCC without side effects on normal cells.

2. Methodology

A comprehensive search was conducted in PubMed, Science Direct, Web of Science, and Google Scholar to identify relevant studies published on the role of nanotechnology-based drug delivery systems using natural products for HCC therapy. The search included studies published from 1976 to 2025 using the following keywords: ‘nanotechnology’, ‘drug delivery systems’, ‘natural products’, ‘hepatocellular carcinoma’, and ‘cancer therapy’. Inclusion criteria encompassed peer-reviewed articles and preclinical trials that investigated the therapeutic potential of natural product-based nanocarriers in HCC treatment. Studies were excluded if they lacked a focus on nanotechnology or did not provide sufficient data on the efficacy of natural products in HCC therapy.

3. Liver and Its Disorders

Necrosis of the liver cells and their incapacity to regenerate are signs of liver damage. This poses a serious risk to patients’ lives and health because, in extreme cases, it can cause bleeding and liver failure [33]. Additionally, astaxanthin has been demonstrated to lessen liver damage brought on by a number of environmental pollutants, such as acetaminophen, carbon tetrachloride, and lipopolysaccharide [34]. High-fat, restricted-protein diets have been associated with liver damage as well as notable alterations in the gut microbiota, which include a decrease in Lactobacillus abundance and an increase in the Firmicutes/Bacteroidetes ratio. A larger bile acid pool and compositional changes, particularly higher levels of CDCA and LCA species, were also linked to these dietary modifications. The liver’s adaptive regulation of bile acid synthesis and transport, as well as the FXR-SHP pathway, was necessary to preserve the homeostasis of bile acid and hepatic lipid metabolism. By activating the FXR pathway, CDCA was found to lessen endoplasmic reticulum stress brought on by palmitic acid in HepG2 cells [35]. Last but not least, it is crucial to remember that a number of established risk factors, such as age, cirrhosis, sex, and co-infection with HIV or other hepatitis viruses like hepatitis C and D, are linked to HCC in people with chronic hepatitis B [36].

4. Hepatoprotective Natural Products

Natural products obtained from medicinal plants, including phenolics, flavonoids, alkaloids, and polysaccharides, are crucial in the treatment of HCC in response to their various bioactive properties [37,38,39]. Hence, natural products exhibit significant therapeutic potential by targeting many molecular pathways associated with the development of cancer [38,40]. Phenolic and flavonoid compounds exhibit anticancer activity via regulating signaling pathways, including STAT3, NF-κB, and CXCR4, while also influencing the tumor environment to improve the immune system [41,42]. Alkaloid substances, derived from traditional Chinese medicine, demonstrate anti-hepatocarcinogenic properties by suppressing proliferation, metastasis, and angiogenesis while enhancing the autophagy and apoptosis processes [40]. Furthermore, natural polysaccharides indicate the ability to minimize the adverse effects of chemotherapeutic agents and possess anti-HCC properties through mechanisms such as apoptosis and immune system stimulation [43]. Thus, phenolics, flavonoids, alkaloids, and polysaccharides from medicinal plants modulate important signaling pathways and enhance the immune response in treating HCC, making them valuable in developing new HCC therapies. The following section describes different natural products that are utilized in treating HCC, which have some advantages and disadvantages, as shown in Table 1.

4.1. Shikonin

Shikonin is a natural product that possesses anticancer properties and has been investigated for its potential use in HCC treatment [44]. Recent studies have demonstrated that shikonin has been shown to control pyruvate kinase type M2 (PKM2), which in turn causes apoptosis and inhibits proliferation and glycolysis in HCC cells [44,45,46]. Shikonin exhibits a dose-dependent effect in enhancing cell apoptosis and autophagy by reducing the expression of pyrroline-5-carboxylate reductase 1 (PYCR1) and inhibiting the PI3K/AKT/mTOR signaling pathway [22,46]. Overall, shikonin has considerable anticancer effects in HCC via various mechanisms, including PKM2 suppression, ROS-mediated apoptosis induction, and regulation of essential signaling pathways [45,47]. Lin et al. demonstrated that shikonin, with an IC50 value of 15 μM, causes cytotoxicity in HER2-enriched cells by stopping the cell cycle in the sub-G1 phase and triggering apoptosis within a day of treatment. Shikonin controls cell proliferation and death by regulating 38 differentially expressed genes [48]. These findings emphasize shikonin’s potential as a significant enhancement of current HCC treatments.

4.2. Fucoidan

Fucoidan, a naturally sulfated polysaccharide found in brown seaweed, has been investigated for its potential role in the treatment of HCC [49]. Studies have demonstrated that fucoidan inhibits the progression of HCC by a variety of pathways, including up-regulation of LINC00261, suppression of invasion, induction of apoptosis, and enhancement of cancer immunity [50]. Also, fucoidan has been shown to exhibit anticancer properties in both in vivo and in vitro investigations, showing its potential as a therapeutic drug for HCC [51].

4.3. Nigella sativa

Nigella sativa, also known as black seed, has been investigated for its potential properties against HCC [52,53]. Recent studies showed that Nigella sativa seed oil inhibits cellular proliferation and induces apoptosis in HCC cells by various mechanisms, including the p53 pathway and reactive oxygen species (ROS)-dependent mitochondrial apoptosis. Nigella sativa is considered a chemopreventive therapy due to its antioxidant mechanism in HCC and inhibition of HCC signaling pathways [54]. Moreover, thymoquinone is a principal bioactive compound of Nigella sativa, which has demonstrated the ability to induce apoptosis in cancer cells via multiple pathways. Furthermore, thymoquinone (TQ) has been identified as a modulator of essential signaling pathways, including the AKT pathway, which plays a vital role in the regulation of cell survival and death [55,56]. Studies indicate that Nigella sativa may inhibit metastasis in multiple cancer models [55]. In vivo, research indicated that the use of Nigella sativa essential oil significantly lowered tumor volume and avoided the development of metastasis in mice models. This indicates a possible function in inhibiting the migration of HCC to other organs [55,57]. Although Nigella sativa is usually thought to be safe, it can produce gastrointestinal problems and allergic reactions in particular individuals. High doses may cause toxicity, especially if combined with different drugs [58,59]. Despite its possible advantages, the use of Nigella sativa in the treatment of HCC is limited due to a lack of clinical data, potential toxicity, complex mechanisms of action, and obstacles to regulation. Additional investigation is needed to address these limitations and develop clear guidelines for clinical use in HCC therapy.

4.4. Curcumin

Curcumin has demonstrated significant outcomes, such as preventing the growth of HCC by several mechanisms. Studies have shown that curcumin inhibits the expression of vascular endothelial growth factor (VEGF), the PI3K/AKT signaling pathway, and microRNAs such as miR-21, resulting in reduced cell proliferation and elevated cell death in both in vitro and in vivo HCC models [20]. It has been demonstrated that curcumin inhibits the growth of HCC by blocking the Wnt/β-Catenin signaling pathway, causing cell cycle arrest, resulting in death [60]. Furthermore, curcumin exhibits crucial properties due to its anticancer, anti-inflammatory, and antioxidant properties [61]. Despite the potential characteristics of curcumin in HCC therapy, there are challenges to applying curcumin in clinical application, such as instability, poor bioavailability, and hydrophobicity [62]. Before curcumin is used therapeutically, these drawbacks can be resolved by functionalizing it using nanoparticles and phospholipid complexes [20,62]. Addressing these difficulties through novel studies and research is critical for achieving the full therapeutic potential of curcumin in HCC therapy.

4.5. Green Tea Catechins

Green Tea Catechins (GTCs), specifically Epigallocatechin-3-gallate (EGCG), have shown promise in both the prevention and the therapy of HCC, which is considered an aggressive type of liver cancer [63]. EGCG has been shown to inhibit the growth and spread of HCC by inducing apoptosis, regulating autophagy, and functioning as an anti-angiogenic agent [63,64]. EGCG inhibits human HCC cell growth by inhibiting IGF-1R phosphorylation, triggering Caspase-9 and Caspase-3, suppressing Bcl-2 and COX-2, and regulating signaling processes like NF-κB and ERK1-2 [64]. The beneficial impacts of GTC intake have been investigated in a variety of animal models, with results supporting their potential to inhibit HCC development, proliferation, and apoptosis. GTCs may decrease hepatocyte stem cell development and affect epigenetic expression in the hepatocytes [65]. Overall, recent studies suggest that GTCs, particularly EGCG, have the potential to prevent and treat HCC by targeting several pathways associated with tumor development and progression.
Table 1. Advantages and disadvantages of some natural plants used for HCC treatment.
Table 1. Advantages and disadvantages of some natural plants used for HCC treatment.
Natural ProductAdvantagesDisadvantagesReference
Shikonin
  • Suppresses growth, invasion, migration, and metastasis.
  • Viability of tumor cells by interacting with important signaling pathways such as PI3K/AKT, MAPK, and PKM2.
  • Low aqueous solubility.
  • Hazardous (particularly when injected intravenously or intra-peritonially).
[47,66,67]
Fucoidan
  • It may trigger HCC cells to:
  • Induce apoptosis.
  • Arrest cell cycles.
  • Molecular structure variation.
  • Fluctuating composition of several fucoidan preparations.
[68,69]
Nigella sativa
  • Promotes HCC cell apoptosis.
  • Arresting the cell cycle.
  • Suppresses the following in a dose-dependent manner: invasion, motility, and proliferation in HCC cells.
  • Allergic reactions.
  • Gastrointestinal disturbances.
[70,71,72]
Curcumin
  • Blocking critical signaling pathways in HCC, such as STAT3/VEGF/HIF-1α.
  • Prevent tumor growth and angiogenesis.
  • Cell cycle arrest, especially in the S phase.
  • Low bioavailability.
[62,73]
Green Tea Catechins
  • Antioxidant properties.
  • Exhibit hepatoprotective characteristics.
  • Have low toxicity compared to other chemotherapies.
  • May cause adverse consequences.
  • Potential risk of liver damage.
[74]

5. Challenges of Using Nature Products as Therapeutic Agents for HCC

The toxic effects of plants used in HCC therapy are a concern. However, medicinal plants include bioactive compounds that can target neoplastic pathways, and nanoparticle-based drug delivery systems can improve selectivity while minimizing damage to healthy cells [75,76]. Many natural products have failed to advance to the clinical trial stage due to insufficient information available about their pharmacokinetics and toxicity towards normal cells [77]. Several natural products, such as curcumin and resveratrol, have poor solubility, fast metabolism, and instability under physiological conditions, thereby limiting their therapeutic effectiveness [75,78]. For instance, the limited bioavailability of curcumin constrains its therapeutic application, despite its promising preclinical efficacy against HCC [78,79]. Furthermore, various plant-derived biologically active substances have low aqueous solubility, cellular absorption and bioavailability, causing clinical translation challenges [80,81]. Despite these limitations, natural products have the potential to be used as an alternative or complementary therapy in conjunction with current HCC chemotherapies [22,76]. Additional study is required to address the limitations and assess the safety and efficacy of plant-based treatments for HCC therapy [22,82,83].

6. Role of Nanotechnology in HCC Treatment

Systemic medication, transplantation of the liver, transarterial chemoembolization, surgical procedures, and excision are among the available forms of treatment [84,85]. Since most patients with liver cirrhosis have infrequent screenings and HCC is frequently asymptomatic in its early stages, the majority of HCC patients are detected in the intermediate or advanced stages [84]. Due to variables including late detection, quick development, poor response to treatment at the advanced stage, and easy recurrence, the five-year survival rate for HCC is just 20% [86]. Therefore, the necessity for an enhanced or novel diagnostic and therapeutic approach is critical. Research in biological nanotechnology has steadily increased recently [87,88,89]. Its use in the diagnosis and treatment of HCC has also been taken into account [90,91,92]. The best treatment for liver cancer is surgical excision, although only 15% of patients respond to this approach [93,94]. Chemotherapy is yet another important therapeutic approach for HCC patients. Chemotherapy is a general treatment that can impact cancerous and normally functioning cells alike. The development of multidrug resistance (MDR) and the uneven biodistribution of chemotherapeutic drugs at the tumor site are two further drawbacks [95]. Existing therapeutic techniques for HCC are not desired, highlighting an urgent need for the development of innovative approaches such as therapeutic antibodies. This is because most HCC patients have been diagnosed in more advanced stages in which existing standard chemotherapies are insufficient [96]. Accordingly, targeted therapy has been proposed for highly expressed proteins in cancer cells that have low expression in normal liver tissue. By using nanostructures to achieve specific therapeutic activities, nanotechnologies have had a major impact on the medical industry in the past few years [87,88,97,98]. Nanoparticles’ high specific surface area gives them various benefits over other molecular conjugates in terms of focused diagnostics and therapies: (1) they can have several targeting ligands, which are capable of binding to receptors on the surface of cells more affinitive; (2) they are able to carry a lot of molecules with effects and guard them from degradation; for instance, lipiodol-bridged Indocyanine Green (ICG) nano emulsion is capable of protecting the fluorochrome ICG from deterioration for a few days via its shielding effect, while the fluorescence levels of free ICG solution quickly drops to zero under identical conditions [99]; (3) they are adequate to hold different kinds of effector molecules; as well as (4) effector molecules can be released from nanoparticles in a way that best suits their mode of action [99]. Because of these benefits, nanomaterials can minimize the negative effects of anticancer medications while simultaneously increasing the sensitivity of biomarker or image detection and facilitating targeted drug delivery [99,100]. Researchers working on nanomedicine are attempting to improve medical constraints by the application of nanotechnology. A therapeutic agent or screening factor and a system for delivering it comprising a targeting moiety, nanocarrier, and stimuli-responsive units are the two parts needed to build a novel therapeutic or image device [101,102,103,104]. The development of a cleverly targeted nanocarrier necessitates a thorough understanding of the physicochemical properties of nano-based materials, as well as the properties of the targeted tissue and how they behave biologically when entering the body. Taking all of these into account, a therapeutic instrument based on nanotechnology may be developed, allowing for the carefully regulated delivery of cargo to target cells in response to certain stimuli while having no negative effects on healthy cells [105,106,107]. Nanotechnology has created two main goals in the realms of diagnostics and therapy for overcoming these drawbacks: effective delivery to reach adequate levels in targeted tumor areas without negative effects or little damage to normal cells, and selective and specific targeting [55]. In order to avoid the liver’s fast evacuation and to precisely address the HCC microenvironment, greater emphasis needs to be paid to the architectonics of nanostructures for either active or passive transport to the liver [108]. When it comes to passive targeting, the liver acquires more NPs than other organs after intravenous administration. The modulation of blood circulation lifespan is influenced significantly by surface modification of nanocarriers, leading to alterations in their physical, chemical, and biological identities [108,109]. To ensure proper biological distribution and effective delivery, a targeting moiety such as immunotoxins, radioimmunotherapeutics, and biocompatible and biodegradable carriers is critically needed [55,109]. For instance, tyrosine kinase inhibitors (TKIs) used to treat advanced HCC, such as staurosporine and sorafenib (SFN), have side effects similar to those of other chemotherapy drugs that affect normal cells and increase resistance to TKIs [110,111]. Thus, in order to overcome cancer cells’ resistance to anticancer TKIs, focused novel TKIs need to be developed with the ability to potentially target and inhibit particular signaling pathways in cancer cells [111]. Since some TKIs are quite harmful when used alone, nanomedicine handles them to increase TKI levels in tumors in a targeted, safe manner [110,111]. The use of several nanomedicines for HCC treatment is discussed in this review. Next, the readers are introduced to the nanocarriers used in HCC diagnosis and treatment, such as lipid, polymeric, and inorganic nanostructures. Lastly, emphasis is placed on the clinical translation and biological safety of nanomedicines in the identification and management of HCC.

7. Green Synthesis of Nanoparticles

The top-down approach is used to transform bulk material into tiny, nanoscale particles. Top-down methods are easy to employ; however, they cannot be used to produce very tiny and regularly shaped particles [112]. The bottom-up approach has become known as the constructive technique. The top-down strategy is completely opposed to bottom-up methods. Atoms and molecules expand and self-assemble to form NPs with different shapes, sizes, and chemical compositions [113,114,115]. Chemical vapor deposition (CVD), biological synthesis, sol-gel, spinning, and pyrolysis are examples of bottom-up techniques [115]. The following three techniques can be utilized to produce NPs, including chemical, physical, and biological [116]. Because it is straightforward, safe, and economical, the biological approach is ideal for creating NPs [115,117], as shown in Table 2. The biological approach is the most efficient and least expensive way to synthesize nanoparticles since it is straightforward and non-toxic [115]. The biological method is straightforward, usually requiring only one step, and good for the environment. In this sense, microbes and various plant parts can be used to create nanomaterials, as illustrated in Figure 1 [118]. The end product is harmless and safe for the environment in nanoparticle form. Using the ideas of green chemistry in nanoscience, safer, more environmentally friendly nanomaterials may be created and processed more quickly. To create and enhance both large- and small-scale processes for creating nanomaterials as well as to use nanomaterials in a variety of industries, green nanotechnology uses green chemistry [119].
The creation of multifunctional nanomaterials that can be used in high-capacity products that may be dangerous to human health and the environment is another goal, as is educating people about the properties of nanoparticles with regard to toxicological concerns [119,120]. It specifically seeks to develop synthesis systems and procedures that can lessen the need for hazardous chemicals while enhancing the efficiency of these already-used synthesis methods. In order to guarantee that the nanoproducts while they are being synthesized are safer, it also provides guidelines for evaluating ecological risks and hazards in connection to design [121]. Numerous species, including fungi, bacteria, and algae, can be used to produce a wide range of nanomaterials from an aqueous solution of metal salts [116]. Living organisms will engage in biomineralization, which includes the production of nanoparticles, by utilizing a protein. For example, magnetotactic bacteria prepare the magnetic particles as a compass to the direction of their chosen habitat at the bottom of the sea in anaerobic conditions using magnetosomes, which are protein-coated for the synthesis of nanosized magnetic iron oxide crystals [122,123]. It has been demonstrated that in vitro synthesis of homogeneous particles with a core diameter of 20–45 nm occurs [124,125]. In spite of this, magnetosomes exhibit potent magnetic properties in therapeutic contexts such as hyperthermia [125,126]. He et al. synthesized 10–20 nm-sized gold nanoparticles extracellularly using microorganisms that produce photosynthesis, such as Rhodopseudomonas capsulata. Nicotinamide Adenine Dinucleotide hydride-dependent reductase is a bacterial enzyme that plays a major role in reducing gold ions to gold nanoparticles. They found that the morphology and form of nanoparticles are regulated by the pH of the growth media [127]. Extracellular palladium nanoparticle production from Pseudomonas cells from the alpine location was reported by Schlüter et al. [128]. The idea for extracellular Au NPs was proposed by Singaravelu et al. using Sargassum wightii algae. Incubation took only 12 h to reach 95% of the yield [129].
Table 2. Exploring the anti-HCC properties of nanoparticles derived from natural products.
Table 2. Exploring the anti-HCC properties of nanoparticles derived from natural products.
NanoparticleNatural ProductParticle Size (nm)ShapeIC50The Effect on HCCReference
AuNPDendrobium officinale extract30 nmSpherical 200 μg/mLAnticancer and immune regulation. [130]
AuNPC. militaris extract5–25 nm Face-center-cubic 10–12.5 µg/mLActivating the gene expression of Bax, Bid, Caspase-3 and Caspase-9. [131]
AgNPNigella sativa10–20 nmSpherical7.16 μg/mLGrowth inhibition without harming healthy cells.[132]
CuONPMomordica cochinchinensis (Lour.) extract40–80 nmCubic 75 μg/mLInduce apoptosis and ROS.[133]
AgNPPodophyllum hexandrum Royal leaf extract14 nmSpherical -Apoptotic effect.[134]
AgNPAmla extract188 nmSpherical and cubic -Apoptotic effect.[134]
AuNPZiziphus spina-christi leaves 31.26–58.06 nmSpherical-Induce apoptosis in cancer cells.[135]
AuNPCordia myxa L. leaves56.49–89.38 nmSpherical -Induce apoptosis in cancer cells.[135]
AgNPLeucus aspera40.67–58.17 nmSpherical 158 µg/mLInduce apoptosis in cancer cells.[136]

8. Nanoparticles Used for HCC

8.1. Quantum Dots NP

Quantum dots (QDs) are tiny semiconductor particles with small diameters that range from 2 to 10 nm [137]. Furthermore, the distinctive features of QDs are achieved through the precise control of their size, shape, and surface chemistry, which is achieved via the use of modern nanotechnology methods in their synthesis. On the other hand, conventional nanoparticles produced through either bottom-up or top-down approaches may not provide the same degree of control and precision [138,139]. Unique optical features, like size-tunable emission wavelengths and strong photostability, are produced by the quantum confinement phenomenon in QDs [138,139,140]. These properties are not usually seen in larger nanoparticles fabricated via traditional methods [138,140]. They also, display distinctive optical and electrical characteristics as a result of quantum confinement effects. These characteristics make them very beneficial across various fields, including biology, medicine, and quantum technology [137]. QDs have been explored for their potential properties in enhancing drug delivery to HCC [141]. In drug delivery applications, there are significant pharmacological properties improved via QDs; furthermore, their ability to enhance the uptake pathways of cancer drugs [141,142]. Furthermore, QDs can be conjugated to other elements, such as magnetic chitosan, for nano-drug delivery systems that target cancer cells. These QDs can target HCC cells and induce ROS formation in high quantities, as illustrated in Figure 2 [143]. Recent studies reported that carbon quantum dots (CQDs) have shown effective progress in drug delivery systems for various types of cancer therapy, including HCC [143,144]. Cadmium-selenium quantum dots (CdSe QDs) have demonstrated low levels of cytotoxicity, indicating their potential as a promising tool for cancer therapy [145]. Another type of QD is trichrome-tryptophan-sorbitol CQDs, derived from tryptophan [146]. These CQDs showed high green fluorescence in HCC cells, demonstrating their potential for cancer detection and therapy [146].
Although QDs show significant potential for improving HCC therapy via targeted drug delivery and imaging, their application is limited by toxicity concerns mainly associated with their material composition. Furthermore, QDs can produce ROS, which may be damaging to normal cells [147,148,149]. QDs are taken up by cells, leading to lysosomal destabilization and the initiation of oxidative stress via excessive ROS generation [5]. This cascade results in DNA damage, lipid peroxidation, protein oxidation, inflammation, and caspase-mediated apoptosis, which include Caspase-9 and Caspase-3, ultimately leading to cell death, as illustrated in Figure 3 [150,151]. This feature also has applications in therapies such as photodynamic therapy to specifically target cancer cells [147,148,149].
Overall, Progress in surface modification and biofunctionalization is essential for addressing these challenges and ensuring safe and successful therapeutic applications in HCC.

8.2. Liposome NP

Liposomes are sealed vesicles with a bilayer structure identical to biological membranes that are made of cholesterol and/or phospholipids as the primary membrane material [152], as illustrated in Figure 4. Because of their excellent biocompatibility, biodegradable non-toxic nature, and non-immunogenicity, liposomes have been used extensively in drug delivery systems [152,153]. Liposomes can simultaneously deliver many therapeutic drugs, including cisplatin and curcumin, to produce a synergistic impact against HCC [154]. This combination amplifies antitumor efficacy and minimizes adverse effects relative to monotherapy [154]. In addition, liposomes can deliver doxorubicin and miR-101 to inhibit tumor development [155]. Also, liposomes can be engineered for targeted delivery, using galactosylated chitosan to specifically target the asialoglycoprotein receptor on HCC, thereby improving the antitumor activity of natural compounds such as oleanolic acid [156]. Liposomes coated with photosensitizers or dyes can be involved in photodynamic and photothermal therapy, enabling a non-invasive treatment approach that successfully targets and ablates HCC cells [157,158]. Liposome nanoparticles present significant promise for HCC treatment due to their capacity to improve drug delivery and specifically target cancer cells. However, challenges such as immunogenicity take place, as liposomal nanoparticles can trigger immune responses and demonstrate off-target toxicity, especially when applied along with immune checkpoint blockade therapies [159]. Additionally, keeping liposomes stable and controlling how they release their encapsulated drug in the acidic environment of tumors presents important challenges that need to be addressed [157,160]. Therefore, liposome nanoparticles loaded with natural products present a promising approach for HCC treatment by improving the administration of drugs, more efficiently targeting tumor cells, and minimizing systemic toxicity. These developments in nanomedicine may result in safer and more efficient treatment alternatives for people with HCC.

8.3. Polymeric NP

Polymeric nanoparticles (PNPs) are solid, colloidal particles composed of many types of polymers, including biodegradable polymers such as chitosan, collagen, polylactic acid (PLA), and poly(lactic-co-glycolic acid) (PLGA) [161]. To increase the bioavailability of drugs that are not highly soluble, PNP can be used to administer the drug orally [162]. This increases the amount of drug at the site of action since the smaller particle size can penetrate capillaries and cells, as illustrated in Figure 5. Furthermore, the use of nanocarriers loaded with natural products has been highlighted as a promising strategy for the treatment of HCC, drawing attention to the fact that natural products exhibit stimulus-responsive drug release and multi-target effects [163]. The use of PNPs that contain natural compounds, such as curcumin, has demonstrated potential in improving the therapy of HCC [164]. By applying these nanoparticles, targeted drug delivery to hepatoma cells is possible due to their enhanced permeability and retention properties [164]. This efficiency results in reduced drug dosage, delayed drug release, and an improved therapeutic effect overall, all while avoiding any apparent adverse effects [21]. However, PNPs often face challenges due to the restricted loading capacity for hydrophobic drugs and inconsistent drug release rates, which may block their therapeutic efficiency [165]. Although PNPs can target-specific areas, they must overcome numerous biological barriers, including systemic, microenvironmental, and cellular barriers, which may differ among patient populations and diseases [166]. Overall, PNPs containing natural products like curcumin have demonstrated the potential to improve HCC therapy by strengthening the targeting, bioavailability, and stability of these drugs. As a starting point for investigating other possible chemotherapeutics, these nanoparticles can give an innovative approach to revolutionary HCC treatment.

8.4. Nanozyme

Natural enzymes are well known for their catalytic activity toward specific substrates; however, they require specific circumstances to be stable under harsh conditions like high temperatures [167]. To solve this problem, scientists have identified immobilization techniques using nano- and microscale materials [168]. The combination of these materials in healthcare equipment reflects the characteristics of major biological environments. Numerous materials can enhance these nanomaterials, including artificial polymers, carbon-based nanoparticles, metal-organic frameworks, nanosized protein-based nano-delivery systems, lipid-based nanomaterials, and polysaccharide-based nanoparticles [168]. Hence, nanozymes provide the benefits of natural enzymes in addition to improved stability, ease of storage, and convenient preparation [167]. The early identification of HCC biomarkers through fluorescence imaging poses significant challenges due to the complex physical environment. Human carboxylesterase (CE) is an important biomarker to enable accurate diagnosis of HCC [169]. Current probes for CE demonstrate a sluggish response rate and limited selectivity. The amide group is recognized as CE-responsive points through the application of “the substrate-hydrolysis enzymatic reaction” approach. A series of off–on probes with leaving groups in the amide unit was performed, leading to the identification of the JFast probe, which exhibits the optimum combination of rapid response time along with superior selectivity toward CE. Both in vitro and in vivo experiments demonstrated that the probe activated solely in hepatic cancer cells. This technique offers a precise method for diagnosing HCC [169]. Current investigations indicate that specific metabolic enzymes in cancerous cells assume inappropriate functions, directly affecting gene expression via mechanisms including protein-protein interactions, protein transformations, or protein-kinase behavior [170,171]. For instance, NAD(P)H quinone oxidoreductase-1 interacts with SREBP1, thereby promoting the development and malignancy of HCC, thus contributing to cancer progression [172]. Recognizing the abnormal functions of metabolic enzymes in tumors may provide new strategies for treating neoplasms [173]. One example is magneto-gold nanozymes (AuNC@Fe3O4) demonstrate photothermal effects and peroxidase-like activity, increasing ROS generation and triggering death in HCC cells via a synergistic mechanism combining photothermal and nano-catalytic therapy [174]. Furthermore, cobalt nanozymes within ferritin nanocages exhibit promise in prognostic diagnosis by differentiating HCC tissues from normal tissues with notable sensitivity and specificity [175]. Also, nanozymes facilitate the detection of HCC-associated microRNA, improving sensitivity by strand displacement amplification and CRISPR-Cas12a systems, enabling visual and colorimetric identification [176]. The staining intensity of these nanozymes is associated with tumor differentiation and patient prognosis. Overall, nanozymes provide various benefits in HCC treatment; challenges remain, such as maintaining biocompatibility and improving delivery systems for clinical use. Further research is required to completely understand their potential in cancer treatment.

9. Overcoming Drug Resistance in HCC Using Natural Product-Nanoparticle Combinations

The complicated process of MDR in HCC involves several mechanisms, including altered metabolism, decreased drug uptake, aberrant apoptosis, intracellular sequestration, and autophagic signaling, slight changes to the tumor microenvironment [177,178]. Despite remarkable advances in HCC treatment, MDR remains a key problem, limiting the efficacy of chemotherapy and targeted therapy [177,178]. Experimental techniques have been noticed to combat drug resistance, but converting them into favorable clinical results has been challenging [178]. Nanomedicine opens new avenues in reversing MDR [177,179]. Several characteristics make the application of nanomedicine appropriate and potentially effective in combating the resistant phenotype of tumor cells. These include the drug carrier’s small size, the ability to target cancer cells passively or actively, enhanced pharmacokinetics and biodistribution, decreased side effects from the formulation, intrinsic inhibitory activity towards MDR efflux pump proteins, and the feasibility of delivering multiple therapeutic agents in a single formulation [177,180,181]. Various nano-drug delivery strategies, such as dendrimers, liposomes, micelles, polymeric, solid lipid, mesoporous silica, metal nanoparticles, and nanostructured lipid carriers, are highlighted in the current review as ways to circumvent the MDR process [178]. NPs can inhibit key proteins involved in drug resistance, such as Bcl-2, AKT, and P-glycoprotein, thereby enhancing the effectiveness of chemotherapeutic agents [182]. Also, natural compounds have many benefits, including high efficacy, minimal toxicity, and the capacity to address several MDR mechanism routes [183,184]. For MDR reversal agents, natural products increase the tumor cells’ sensitivity to chemotherapeutics and work in concert with them. When natural products and anticancer drugs are delivered together via nanocarriers, the synergistic effects against MDR in tumor cells are maximized [183], as illustrated by Figure 6. Hence, the integration of natural products with nanoparticles presents a synergistic strategy to combat drug resistance [141,179]. Natural products such as celastrol, when formulated with doxorubicin in nanoparticle structure, may increase aqueous solubility, decrease drug dosage, and augment cellular drug accumulation, resulting in apoptosis and autophagy in drug-resistant cells [185]. Despite the significant potential of nanoparticle-based drugs and natural product combinations, challenges persist in optimizing these structures for clinical application.

10. Natural Product Delivery Systems for HCC

The utilization of natural products in cancer treatment has garnered significant attention in recent years, primarily due to persistent challenges related to drug resistance particularly in HCC, which is well-recognized for its resistance to conventional therapies [186]. Green drug delivery systems enhance the effectiveness of anticancer treatments by utilizing natural materials, such as plant extracts, marine animals, and microorganisms like fungi and bacteria [186,187]. These materials develop target-specific activity, decrease side effects, and increase in vivo stability and bioavailability [130,187]. Furthermore, natural substances have been used as herbal remedies throughout history for the treatment of HCC, like dietary curcumin and resveratrol loaded with DSPE-PEG2000-SP94 nanoparticles and ganoderic acid coated with nano-lipid [188]. In addition, a hepatic cancer cell line (Hep-G2) could be inhibited by spherical AgNPs that were green synthesized by using these plants: Citrulluss colocynthis, Erythrina indica, Panax ginseng, and Rubus glaucus Benth [189]. Although various delivery systems exhibit potential, as shown in Table 3, challenges continue to occur, including the need for consistent targeting and the reduction of adverse reactions. Subsequent research should focus on improving these systems for clinical use, maybe using multidisciplinary strategies to improve their effectiveness and safety in HCC treatment.

11. Nano-Enabled Personalized Medicine Approaches in HCC

The integration of nanoparticles with natural products presents promising opportunities for personalized medicine in the treatment of HCC [75,199,200]. Theragnostic nanoparticles are a new method that helps in both treatment and diagnosis, allowing for personalized treatment plans by tracking how well drugs are delivered and how tumors respond in real time [201,202]. Furthermore, adaptive therapies can be developed by combining nanoparticles with natural products; these strategies can then be tailored to the specific tumor characteristics and therapeutic response of each patient [203,204]. Nanoparticles can be functionalized with ligands (e.g., antibodies, peptides, aptamers) that specifically bind to HCC receptors such as glypican-3, asialoglycoprotein, or transferrin receptor [163]. For example, sorafenib-loaded nanoparticles that target glypican-3 increase drug accumulation in tumors while preserving normal tissue. An alternative approach is dual-ligand systems. Nanoparticles can be tailored to specific HCC characteristics, including receptor overexpression or mutations in the genome. For instance, dual-targeting nanoparticles are modified to target A54 receptors and nuclear localization signals [205]. Overall, the incorporation of nanoparticles and natural products in HCC treatment provides considerable potential for tailored medicine. This approach improves drug distribution, effectiveness, and safety, providing a customized therapy strategy that targets the complex nature of HCC.

12. Limitations in Herbal Medication Based on Nanoparticles

Natural product-based nanotechnology therapy for HCC is limited because of poor aqueous solubility, low cellular absorption, and low bioavailability [32]. Studying the pharmacokinetics of natural products is critical for maximizing therapeutic outcomes and dealing with concerns such as rapid elimination, toxic effects, and inflammation associated with their application [77]. There are major challenges with controlling release in nanoparticles, including non-tunable size, structural heterogeneity, and low release consistency [206]. NPs can trigger or alter the immune response, causing inflammation, immunosuppression, or autoimmune diseases [205,207]. Furthermore, the toxicity of nanoliposomes and QDs in treating HCC is a concern that must be addressed. Both NPs can have adverse effects, including toxicity in normal cells [208,209]. Additionally, nanocarriers can improve the selectivity and delivery of bioactive substances to tumor sites while limiting damage to normal cells. However, challenges related to nanocarrier design and the HCC microenvironment must be eliminated for the effective clinical translation of nanotechnology-based HCC treatment with natural products.

13. Conclusions

HCC remains to be considered a primary global health concern, as traditional therapies are frequently constrained by toxicity, drug resistance, and low bioavailability. Herbal medicine, containing various bioactive substances, presents a viable approach for HCC treatment by simultaneously targeting multiple tumor-promoting pathways while minimizing adverse effects. Nanotechnology approaches such as drug delivery systems (DDS) have emerged as promising approaches for the treatment of HCC. Natural products such as Shikonin, Fucoidan, Nigella sativa, etc., have shown promising anticancer properties, but their clinical application is often limited by low bioavailability, poor solubility, and potential adverse effects. Encapsulating or attaching natural products to nanoparticles can improve their pharmacokinetic profile, allowing for controlled release, targeted delivery, overcoming drug resistance, and enhanced therapeutic efficacy. The conjugation of nanoparticles, such as quantum dots and nanoliposomes, with natural products offers a potential strategy to improve effectiveness and reduce the adverse effects of these natural products. Despite these advances, problems such as scalability, long-term safety, and clinical translation remain. Future research should optimize DDS for herbal substances, perform thorough preclinical and clinical trials, and develop personalized medicine strategies.

Author Contributions

Conceptualization, D.S.R.K. and M.T.E.-M.; supervision, D.S.R.K. and G.M.; writing—original draft preparation, M.T.E.-M., M.A.A.-T., S.K.M. and D.S.R.K.; review and editing the original draft, D.S.R.K. and G.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Deanship of Scientific Research, Vice Presidency for Graduate Studies and Scientific Research, King Faisal University, Saudi Arabia [Grant No. KFU251896].

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

HCCHepatocellular carcinoma
PKM2Pyruvate kinase type M2
Hep-G2Hepatic cancer cell line
PYCR1Pyroline-5-carboxylate reductase 1
ROSReactive oxygen species
GTCsGreen Tea Catechins
EGCGEpigallocatechin-3-gallate
AFP Alpha-fetoprotien
MRIMagnetic resonance imaging
CTComputed tomography
MDRMultidrug resistance
ICGIndocyanine Green
NPsNanoparticles
TKIsTyrosine kinase inhibitors
SFNSorafenib
QDsQuantum dots
TQThymoquinone
VEGFVascular endothelial growth factor
CdSe QDsCadmium-selenium quantum dots
PNPsPolymeric nanoparticles
PLAPolylactic acid
CVDChemical vapor deposition
PVAPolyvinyl alcohol
NARNPsNaringenin nanoparticles
QCQuercetin
C-GPLGalactose-modified PEGylated liposomes
MLNPsMorus nigra L.-derived lipid nanoparticles
PLGAPoly(lactic-co-glycolic acid)

References

  1. Center, M.M.; Jemal, A. Data from International Trends in Liver Cancer Incidence Rates; American Association for Cancer Research: Philadelphia, PA, USA, 2023. [Google Scholar]
  2. Wang, R.H.; Hu, M.; Yang, Z.Y.; Niu, Z.Y.; Chen, H.S.; Zhou, X.; Cao, G.W. Global liver cancer incidence and mortality and future trends from 2000 to 2020: GLOBOCAN data analysis. Zhonghua Gan Zang Bing Za Zhi (Chin. J. Hepatol.) 2023, 31, 271–280. [Google Scholar] [CrossRef]
  3. Serraino, D.; Fratino, L.; Piselli, P. Epidemiological Aspects of Hepatocellular Carcinoma. In Hepatocellular Carcinoma; Ettorre, G.M., Ed.; Updates in Surgery; Springer International Publishing: Cham, Switzerland, 2023; pp. 3–9. ISBN 978-3-031-09370-8. [Google Scholar]
  4. Islami, F.; Ward, E.M.; Sung, H.; Cronin, K.A.; Tangka, F.K.L.; Sherman, R.L.; Zhao, J.; Anderson, R.N.; Henley, S.J.; Yabroff, K.R.; et al. Annual Report to the Nation on the Status of Cancer, Part 1: National Cancer Statistics. J. Natl. Cancer Inst. 2021, 113, 1648–1669. [Google Scholar] [CrossRef] [PubMed]
  5. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  6. Gong, H.; Ong, S.C.; Li, F.; Shen, Y.; Weng, Z.; Zhao, K.; Jiang, Z.; Wang, M. Cost-Effectiveness of Immune Checkpoint Inhibitors as a First-Line Therapy for Advanced Hepatocellular Carcinoma: A Systematic Review. Health Econ. Rev. 2024, 14, 48. [Google Scholar] [CrossRef]
  7. Wang, W.; Wei, C. Advances in the Early Diagnosis of Hepatocellular Carcinoma. Genes. Dis. 2020, 7, 308–319. [Google Scholar] [CrossRef]
  8. Lurje, I.; Czigany, Z.; Bednarsch, J.; Roderburg, C.; Isfort, P.; Neumann, U.P.; Lurje, G. Treatment Strategies for Hepatocellular Carcinoma—A Multidisciplinary Approach. Int. J. Mol. Sci. 2019, 20, 1465. [Google Scholar] [CrossRef]
  9. Petrowsky, H.; Fritsch, R.; Guckenberger, M.; De Oliveira, M.L.; Dutkowski, P.; Clavien, P.-A. Modern Therapeutic Approaches for the Treatment of Malignant Liver Tumours. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 755–772. [Google Scholar] [CrossRef] [PubMed]
  10. Man, S.; Luo, C.; Yan, M.; Zhao, G.; Ma, L.; Gao, W. Treatment for Liver Cancer: From Sorafenib to Natural Products. Eur. J. Med. Chem. 2021, 224, 113690. [Google Scholar] [CrossRef]
  11. Doustmihan, A.; Fathi, M.; Mazloomi, M.; Salemi, A.; Hamblin, M.R.; Jahanban-Esfahlan, R. Molecular Targets, Therapeutic Agents and Multitasking Nanoparticles to Deal with Cancer Stem Cells: A Narrative Review. J. Control. Release 2023, 363, 57–83. [Google Scholar] [CrossRef]
  12. Talib, W.H.; Alsayed, A.R.; Barakat, M.; Abu-Taha, M.I.; Mahmod, A.I. Targeting Drug Chemo-Resistance in Cancer Using Natural Products. Biomedicines 2021, 9, 1353. [Google Scholar] [CrossRef]
  13. Tan, B.L.; Norhaizan, M.E. Curcumin Combination Chemotherapy: The Implication and Efficacy in Cancer. Molecules 2019, 24, 2527. [Google Scholar] [CrossRef] [PubMed]
  14. Dasari, S.; Njiki, S.; Mbemi, A.; Yedjou, C.G.; Tchounwou, P.B. Pharmacological Effects of Cisplatin Combination with Natural Products in Cancer Chemotherapy. Int. J. Mol. Sci. 2022, 23, 1532. [Google Scholar] [CrossRef] [PubMed]
  15. Yuan, R.; Hou, Y.; Sun, W.; Yu, J.; Liu, X.; Niu, Y.; Lu, J.; Chen, X. Natural Products to Prevent Drug Resistance in Cancer Chemotherapy: A Review. Ann. N. Y. Acad. Sci. 2017, 1401, 19–27. [Google Scholar] [CrossRef]
  16. Roy, N.K.; Tewari, D.; Esposito, M.T. Editorial: Chemosensitizing Effect of Natural Products against Cancers: Applications in Enhancing Chemotherapy and Immunotherapy. Front. Pharmacol. 2022, 13, 988226. [Google Scholar] [CrossRef]
  17. Scaria, B.; Sood, S.; Raad, C.; Khanafer, J.; Jayachandiran, R.; Pupulin, A.; Grewal, S.; Okoko, M.; Arora, M.; Miles, L.; et al. Natural Health Products (NHP’s) and Natural Compounds as Therapeutic Agents for the Treatment of Cancer; Mechanisms of Anti-Cancer Activity of Natural Compounds and Overall Trends. Int. J. Mol. Sci. 2020, 21, 8480. [Google Scholar] [CrossRef] [PubMed]
  18. Das Gupta, V. Quantitative Determination of Piperazine Citrate in Piperazine Citrate Syrup USP. Am. J. Hosp. Pharm. 1976, 33, 283–284. [Google Scholar]
  19. Hamza, A.A.; Heeba, G.H.; Hamza, S.; Abdalla, A.; Amin, A. Standardized Extract of Ginger Ameliorates Liver Cancer by Reducing Proliferation and Inducing Apoptosis through Inhibition Oxidative Stress/Inflammation Pathway. Biomed. Pharmacother. 2021, 134, 111102. [Google Scholar] [CrossRef]
  20. Li, J.; Wei, H.; Liu, Y.; Li, Q.; Guo, H.; Guo, Y.; Chang, Z. Curcumin Inhibits Hepatocellular Carcinoma via Regulating miR-21/TIMP3 Axis. Evid.-Based Complement. Altern. Med. 2020, 2020, 2892917. [Google Scholar] [CrossRef]
  21. 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. [Google Scholar] [CrossRef]
  22. Huang, M.; Lu, J.-J.; Ding, J. Natural Products in Cancer Therapy: Past, Present and Future. Nat. Prod. Bioprospect. 2021, 11, 5–13. [Google Scholar] [CrossRef]
  23. Chen, L.; Wu, L.; Zhang, L.; Sun, B.; Wu, W.; Lei, Y.; Zhu, L.; Sun, T.; Liang, B.; Zhao, H.; et al. Effect of Metformin on Hepatocellular Carcinoma Patients with Type II Diabetes Receiving Transarterial Chemoembolization: A Multicenter Retrospective Cohort Study. Int. J. Surg. 2025, 111, 828–838. [Google Scholar] [CrossRef]
  24. Zheng, P.; Xu, D.; Cai, Y.; Zhu, L.; Xiao, Q.; Peng, W.; Chen, B. A Multi-Omic Analysis Reveals That Gamabufotalin Exerts Anti-Hepatocellular Carcinoma Effects by Regulating Amino Acid Metabolism through Targeting STAMBPL1. Phytomedicine 2024, 135, 156094. [Google Scholar] [CrossRef] [PubMed]
  25. Li, Y.; Zou, H.; Zheng, Z.; Liu, Z.; Hu, H.; Wu, W.; Wang, T. Advances in the Study of Bioactive Nanoparticles for the Treatment of HCC and Its Postoperative Residual Cancer. Int. J. Nanomed. 2023, 18, 2721–2735. [Google Scholar] [CrossRef] [PubMed]
  26. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering Precision Nanoparticles for Drug Delivery. Nat. Rev. Drug Discov. 2021, 20, 101–124. [Google Scholar] [CrossRef] [PubMed]
  27. Dong, Q.; Jiang, Z. Platinum–Iron Nanoparticles for Oxygen-Enhanced Sonodynamic Tumor Cell Suppression. Inorganics 2024, 12, 331. [Google Scholar] [CrossRef]
  28. Mansour, W.; El Fedawy, S.F.; Atta, S.A.; Zarie, R.M.; Fouad, N.T.A.; Maher, S.; Hussein, T.M.; Abdel Aziz, D.M.; Kamel, M. Targeted Therapy for HCC Using Dumbbell-like Nanoparticles Conjugated to Monoclonal Antibodies against VEGF and Cancer Stem Cell Receptors in Mice. Cancer Nanotechnol. 2023, 14, 14. [Google Scholar] [CrossRef]
  29. Clemons, T.D.; Singh, R.; Sorolla, A.; Chaudhari, N.; Hubbard, A.; Iyer, K.S. Distinction Between Active and Passive Targeting of Nanoparticles Dictate Their Overall Therapeutic Efficacy. Langmuir 2018, 34, 15343–15349. [Google Scholar] [CrossRef]
  30. Khafaga, D.S.R.; Eid, M.M.; Mohamed, M.H.; Abdelmaksoud, M.D.E.; Afify, M.; El-Khawaga, A.M.; Abdelhakim, H.K. Enhanced Anticancer Activity of Silver Doped Zinc Oxide Magnetic Nanocarrier Loaded with Sorafenib for Hepatocellular Carcinoma Treatment. Sci. Rep. 2024, 14, 15538. [Google Scholar] [CrossRef]
  31. Zeng, M.; Guo, D.; Fernández-Varo, G.; Zhang, X.; Fu, S.; Ju, S.; Yang, H.; Liu, X.; Wang, Y.-C.; Zeng, Y.; et al. The Integration of Nanomedicine with Traditional Chinese Medicine: Drug Delivery of Natural Products and Other Opportunities. Mol. Pharm. 2023, 20, 886–904. [Google Scholar] [CrossRef]
  32. Basu, A.; Namporn, T.; Ruenraroengsak, P. Critical Review in Designing Plant-Based Anticancer Nanoparticles against Hepatocellular Carcinoma. Pharmaceutics 2023, 15, 1611. [Google Scholar] [CrossRef]
  33. Li, J.; Chen, Y.; Zhang, S.; Zhao, Y.; Gao, D.; Xing, J.; Cao, Y.; Xu, G. Purslane (Portulaca oleracea L.) Polysaccharide Attenuates Carbon Tetrachloride-Induced Acute Liver Injury by Modulating the Gut Microbiota in Mice. Genomics 2025, 117, 110983. [Google Scholar] [CrossRef]
  34. Tan, Q.; Chu, H.; Wei, J.; Yan, S.; Sun, X.; Wang, J.; Zhu, L.; Yang, F. Astaxanthin Alleviates Hepatic Lipid Metabolic Dysregulation Induced by Microcystin-LR. Toxins 2024, 16, 401. [Google Scholar] [CrossRef]
  35. Huang, L.; Li, Y.; Tang, R.; Yang, P.; Zhuo, Y.; Jiang, X.; Che, L.; Lin, Y.; Xu, S.; Li, J.; et al. Bile Acids Metabolism in the Gut-Liver Axis Mediates Liver Injury during Lactation. Life Sci. 2024, 338, 122380. [Google Scholar] [CrossRef]
  36. Campbell, C.; Wang, T.; McNaughton, A.L.; Barnes, E.; Matthews, P.C. Risk Factors for the Development of Hepatocellular Carcinoma (HCC) in Chronic Hepatitis B Virus (HBV) Infection: A Systematic Review and Meta-Analysis. J. Viral Hepat. 2021, 28, 493–507. [Google Scholar] [CrossRef]
  37. Gao, S.; Jiang, X.; Wang, L.; Jiang, S.; Luo, H.; Chen, Y.; Peng, C. The Pathogenesis of Liver Cancer and the Therapeutic Potential of Bioactive Substances. Front. Pharmacol. 2022, 13, 1029601. [Google Scholar] [CrossRef] [PubMed]
  38. Guo, J.; Yan, W.; Duan, H.; Wang, D.; Zhou, Y.; Feng, D.; Zheng, Y.; Zhou, S.; Liu, G.; Qin, X. Therapeutic Effects of Natural Products on Liver Cancer and Their Potential Mechanisms. Nutrients 2024, 16, 1642. [Google Scholar] [CrossRef] [PubMed]
  39. Sun, D.; Li, X.; Nie, S.; Liu, J.; Wang, S. Disorders of Cancer Metabolism: The Therapeutic Potential of Cannabinoids. Biomed. Pharmacother. 2023, 157, 113993. [Google Scholar] [CrossRef] [PubMed]
  40. Liu, C.; Yang, S.; Wang, K.; Bao, X.; Liu, Y.; Zhou, S.; Liu, H.; Qiu, Y.; Wang, T.; Yu, H. Alkaloids from Traditional Chinese Medicine against Hepatocellular Carcinoma. Biomed. Pharmacother. 2019, 120, 109543. [Google Scholar] [CrossRef]
  41. Bailly, C. Molecular and Cellular Basis of the Anticancer Activity of the Prenylated Flavonoid Icaritin in Hepatocellular Carcinoma. Chem. Biol. Interact. 2020, 325, 109124. [Google Scholar] [CrossRef]
  42. Jiang, C.-H.; Sun, T.-L.; Xiang, D.-X.; Wei, S.-S.; Li, W.-Q. Anticancer Activity and Mechanism of Xanthohumol: A Prenylated Flavonoid From Hops (Humulus lupulus L.). Front. Pharmacol. 2018, 9, 530. [Google Scholar] [CrossRef]
  43. Wang, D.; Zhang, Z.; Zhao, L.; Yang, L.; Lou, C. Recent Advances in Natural Polysaccharides against Hepatocellular Carcinoma: A Review. Int. J. Biol. Macromol. 2023, 253, 126766. [Google Scholar] [CrossRef] [PubMed]
  44. Guo, C.; He, J.; Song, X.; Tan, L.; Wang, M.; Jiang, P.; Li, Y.; Cao, Z.; Peng, C. Pharmacological Properties and Derivatives of Shikonin—A Review in Recent Years. Pharmacol. Res. 2019, 149, 104463. [Google Scholar] [CrossRef] [PubMed]
  45. Zhao, X.; Zhu, Y.; Hu, J.; Jiang, L.; Li, L.; Jia, S.; Zen, K. Shikonin Inhibits Tumor Growth in Mice by Suppressing Pyruvate Kinase M2-Mediated Aerobic Glycolysis. Sci. Rep. 2018, 8, 14517. [Google Scholar] [CrossRef]
  46. Boulos, J.C.; Rahama, M.; Hegazy, M.-E.F.; Efferth, T. Shikonin Derivatives for Cancer Prevention and Therapy. Cancer Lett. 2019, 459, 248–267. [Google Scholar] [CrossRef] [PubMed]
  47. Zhang, J.; Shang, L.; Jiang, W.; Wu, W. Shikonin Induces Apoptosis and Autophagy via Downregulation of Pyrroline-5-Carboxylate Reductase1 in Hepatocellular Carcinoma Cells. Bioengineered 2022, 13, 7904–7918. [Google Scholar] [CrossRef]
  48. Lin, K.-H.; Huang, M.-Y.; Cheng, W.-C.; Wang, S.-C.; Fang, S.-H.; Tu, H.-P.; Su, C.-C.; Hung, Y.-L.; Liu, P.-L.; Chen, C.-S.; et al. RNA-Seq Transcriptome Analysis of Breast Cancer Cell Lines under Shikonin Treatment. Sci. Rep. 2018, 8, 2672. [Google Scholar] [CrossRef]
  49. Cho, Y.; Yoon, J.-H.; Yoo, J.; Lee, M.; Lee, D.H.; Cho, E.J.; Lee, J.-H.; Yu, S.J.; Kim, Y.J.; Kim, C.Y. Fucoidan Protects Hepatocytes from Apoptosis and Inhibits Invasion of Hepatocellular Carcinoma by Up-Regulating P42/44 MAPK-Dependent NDRG-1/CAP43. Acta Pharm. Sin. B 2015, 5, 544–553. [Google Scholar] [CrossRef]
  50. Ma, D.; Wei, J.; Chen, S.; Wang, H.; Ning, L.; Luo, S.-H.; Liu, C.-L.; Song, G.; Yao, Q. Fucoidan Inhibits the Progression of Hepatocellular Carcinoma via Causing lncRNA LINC00261 Overexpression. Front. Oncol. 2021, 11, 653902. [Google Scholar] [CrossRef]
  51. Lin, Y.; Qi, X.; Liu, H.; Xue, K.; Xu, S.; Tian, Z. The Anti-Cancer Effects of Fucoidan: A Review of Both In Vivo and In Vitro Investigations. Cancer Cell Int. 2020, 20, 154. [Google Scholar] [CrossRef]
  52. Rashwan, H.K.; Mahgoub, S.; Abuelezz, N.Z.; Amin, H.K. Black Cumin Seed (Nigella sativa) in Inflammatory Disorders: Therapeutic Potential and Promising Molecular Mechanisms. Drugs Drug Candidates 2023, 2, 516–537. [Google Scholar] [CrossRef]
  53. Abd-Rabou, A.A.; Edris, A.E. Cytotoxic, Apoptotic, and Genetic Evaluations of Nigella sativa Essential Oil Nanoemulsion against Human Hepatocellular Carcinoma Cell Lines. Cancer Nanotechnol. 2021, 12, 28. [Google Scholar] [CrossRef]
  54. Sadeghi, E.; Imenshahidi, M.; Hosseinzadeh, H. Molecular Mechanisms and Signaling Pathways of Black Cumin (Nigella sativa) and Its Active Constituent, Thymoquinone: A Review. Mol. Biol. Rep. 2023, 50, 5439–5454. [Google Scholar] [CrossRef]
  55. Wang, X.; Yang, L.; Chen, Z.; Shin, D.M. Application of Nanotechnology in Cancer Therapy and Imaging. CA Cancer J. Clin. 2008, 58, 97–110. [Google Scholar] [CrossRef]
  56. Jehan, S.; Zhong, C.; Li, G.; Zulqarnain Bakhtiar, S.; Li, D.; Sui, G. Thymoquinone Selectively Induces Hepatocellular Carcinoma Cell Apoptosis in Synergism with Clinical Therapeutics and Dependence of P53 Status. Front. Pharmacol. 2020, 11, 555283. [Google Scholar] [CrossRef]
  57. Liu, Y.; Wu, Y.; Li, Z.; Wan, D.; Pan, J. Targeted Drug Delivery Strategies for the Treatment of Hepatocellular Carcinoma. Molecules 2024, 29, 4405. [Google Scholar] [CrossRef]
  58. Alberts, A.; Moldoveanu, E.-T.; Niculescu, A.-G.; Grumezescu, A.M. Nigella sativa: A Comprehensive Review of Its Therapeutic Potential, Pharmacological Properties, and Clinical Applications. Int. J. Mol. Sci. 2024, 25, 13410. [Google Scholar] [CrossRef]
  59. Elshnoudy, I.A.; Elkhouly, A.M.; Masoud, M.; Rabea, H.A.; Mansour, F.R. Medicinal Plants Cultivated in Egypt with Anticancer Potential; a Systematic Review. Phytochem. Rev. 2024, 24, 527–583. [Google Scholar] [CrossRef]
  60. Shao, J.; Shi, C.-J.; Li, Y.; Zhang, F.; Pan, F.; Fu, W.; Zhang, J. LincROR Mediates the Suppressive Effects of Curcumin on Hepatocellular Carcinoma Through Inactivating Wnt/β-Catenin Signaling. Front. Pharmacol. 2020, 11, 847. [Google Scholar] [CrossRef]
  61. Zhang, J.; Liu, Y.; Wang, X.; Wang, Z.; Xing, E.; Li, J.; Wang, D. Curcumin Inhibits Proliferation of Hepatocellular Carcinoma Cells by Blocking PTPN1 and PTPN11 Expression. Oncol. Lett. 2023, 26, 307. [Google Scholar] [CrossRef]
  62. Gull, N.; Arshad, F.; Naikoo, G.A.; Hassan, I.U.; Pedram, M.Z.; Ahmad, A.; Aljabali, A.A.A.; Mishra, V.; Satija, S.; Charbe, N.; et al. Recent Advances in Anticancer Activity of Novel Plant Extracts and Compounds from Curcuma Longa in Hepatocellular Carcinoma. J. Gastrointest. Canc 2023, 54, 368–390. [Google Scholar] [CrossRef]
  63. Oh, J.-W.; Muthu, M.; Pushparaj, S.S.C.; Gopal, J. Anticancer Therapeutic Effects of Green Tea Catechins (GTCs) When Integrated with Antioxidant Natural Components. Molecules 2023, 28, 2151. [Google Scholar] [CrossRef] [PubMed]
  64. Almatroodi, S.A.; Almatroudi, A.; Khan, A.A.; Alhumaydhi, F.A.; Alsahli, M.A.; Rahmani, A.H. Potential Therapeutic Targets of Epigallocatechin Gallate (EGCG), the Most Abundant Catechin in Green Tea, and Its Role in the Therapy of Various Types of Cancer. Molecules 2020, 25, 3146. [Google Scholar] [CrossRef] [PubMed]
  65. Cheng, Z.; Zhang, Z.; Han, Y.; Wang, J.; Wang, Y.; Chen, X.; Shao, Y.; Cheng, Y.; Zhou, W.; Lu, X.; et al. A Review on Anti-Cancer Effect of Green Tea Catechins. J. Funct. Foods 2020, 74, 104172. [Google Scholar] [CrossRef]
  66. Du, D.; Liu, C.; Qin, M.; Zhang, X.; Xi, T.; Yuan, S.; Hao, H.; Xiong, J. Metabolic Dysregulation and Emerging Therapeutical Targets for Hepatocellular Carcinoma. Acta Pharm. Sin. B 2022, 12, 558–580. [Google Scholar] [CrossRef]
  67. Iranzadeh, S.; Dalil, D.; Kohansal, S.; Isakhani, M. Shikonin in Breast Cancer Treatment: A Comprehensive Review of Molecular Pathways and Innovative Strategies. J. Pharm. Pharmacol. 2024, 76, 967–982. [Google Scholar] [CrossRef]
  68. Kiselevskiy, M.V.; Anisimova, N.Y.; Ustyuzhanina, N.E.; Vinnitskiy, D.Z.; Tokatly, A.I.; Reshetnikova, V.V.; Chikileva, I.O.; Shubina, I.Z.H.; Kirgizov, K.I.; Nifantiev, N.E. Perspectives for the Use of Fucoidans in Clinical Oncology. Int. J. Mol. Sci. 2022, 23, 11821. [Google Scholar] [CrossRef]
  69. Duan, Y.; Li, J.; Jing, X.; Ding, X.; Yu, Y.; Zhao, Q. Fucoidan Induces Apoptosis and Inhibits Proliferation of Hepatocellular Carcinoma via the P38 MAPK/ERK and PI3K/Akt Signal Pathways. Cancer Manag. Res. 2020, 12, 1713–1723. [Google Scholar] [CrossRef]
  70. Adam, S.H.; Mohd Nasri, N.; Kashim, M.I.A.M.; Abd Latib, E.H.; Ahmad Juhari, M.A.A.; Mokhtar, M.H. Potential Health Benefits of Nigella sativa on Diabetes Mellitus and Its Complications: A Review from Laboratory Studies to Clinical Trials. Front. Nutr. 2022, 9, 1057825. [Google Scholar] [CrossRef]
  71. Yimer, E.M.; Tuem, K.B.; Karim, A.; Ur-Rehman, N.; Anwar, F. Nigella sativa L. (Black Cumin): A Promising Natural Remedy for Wide Range of Illnesses. Evid.-Based Complement. Altern. Med. 2019, 2019, 1528635. [Google Scholar] [CrossRef]
  72. Berköz, M. Nigella sativa L. In Novel Drug Targets with Traditional Herbal Medicines: Scientific and Clinical Evidence; Gürağaç Dereli, F.T., Ilhan, M., Belwal, T., Eds.; Springer International Publishing: Cham, Switzerland, 2022; pp. 445–461. ISBN 978-3-031-07753-1. [Google Scholar]
  73. Wang, X.; Tian, Y.; Lin, H.; Cao, X.; Zhang, Z. Curcumin Induces Apoptosis in Human Hepatocellular Carcinoma Cells by Decreasing the Expression of STAT3/VEGF/HIF-1α Signaling. Open Life Sci. 2023, 18, 20220618. [Google Scholar] [CrossRef]
  74. Farhan, M. Green Tea Catechins: Nature’s Way of Preventing and Treating Cancer. Int. J. Mol. Sci. 2022, 23, 10713. [Google Scholar] [CrossRef] [PubMed]
  75. Singh, A.K.; Singh, S.V.; Kumar, R.; Kumar, S.; Senapati, S.; Pandey, A.K. Current Therapeutic Modalities and Chemopreventive Role of Natural Products in Liver Cancer: Progress and Promise. World J. Hepatol. 2023, 15, 1–18. [Google Scholar] [CrossRef]
  76. Andreani, T.; Cheng, R.; Elbadri, K.; Ferro, C.; Menezes, T.; Dos Santos, M.R.; Pereira, C.M.; Santos, H.A. Natural Compounds-Based Nanomedicines for Cancer Treatment: Future Directions and Challenges. Drug Deliv. Transl. Res. 2024, 14, 2845–2916. [Google Scholar] [CrossRef]
  77. Gupta, M.; Sarwat, M. Protective Effects of Plant-Derived Natural Products against Hepatocellular Carcinoma. In Herbal Medicines; Elsevier: Amsterdam, The Netherlands, 2022; pp. 609–627. ISBN 978-0-323-90572-5. [Google Scholar]
  78. Alnajjar, A.; Elsiesy, H. Natural Products and Hepatocellular Carcinoma: A Review. Hepatoma Res. 2015, 1, 119. [Google Scholar] [CrossRef]
  79. Rodriguez, S.; Skeet, K.; Mehmetoglu-Gurbuz, T.; Goldfarb, M.; Karri, S.; Rocha, J.; Shahinian, M.; Yazadi, A.; Poudel, S.; Subramani, R. Phytochemicals as an Alternative or Integrative Option, in Conjunction with Conventional Treatments for Hepatocellular Carcinoma. Cancers 2021, 13, 5753. [Google Scholar] [CrossRef]
  80. Sayed, N.; Khurana, A.; Godugu, C. Pharmaceutical Perspective on the Translational Hurdles of Phytoconstituents and Strategies to Overcome. J. Drug Deliv. Sci. Technol. 2019, 53, 101201. [Google Scholar] [CrossRef]
  81. Yang, J.; Li, K.; He, D.; Gu, J.; Xu, J.; Xie, J.; Zhang, M.; Liu, Y.; Tan, Q.; Zhang, J. Toward a Better Understanding of Metabolic and Pharmacokinetic Characteristics of Low-Solubility, Low-Permeability Natural Medicines. Drug Metab. Rev. 2020, 52, 19–43. [Google Scholar] [CrossRef]
  82. Sun, L.; Zhou, W.; Zhang, H.; Guo, Q.; Yang, W.; Li, B.; Sun, Z.; Gao, S.; Cui, R. Modulation of Multiple Signaling Pathways of the Plant-Derived Natural Products in Cancer. Front. Oncol. 2019, 9, 1153. [Google Scholar] [CrossRef]
  83. SR Khafaga, D.; Ewies, E. Drug Delivery Systems Designed to Maximize the Therapeutic Efficacy of Herbal Medication: A Review Article. Egypt. J. Chem. 2023, 66, 477–485. [Google Scholar] [CrossRef]
  84. Galle, P.R.; Forner, A.; Llovet, J.M.; Mazzaferro, V.; Piscaglia, F.; Raoul, J.-L.; Schirmacher, P.; Vilgrain, V. EASL Clinical Practice Guidelines: Management of Hepatocellular Carcinoma. J. Hepatol. 2018, 69, 182–236. [Google Scholar] [CrossRef]
  85. Marrero, J.A.; Kulik, L.M.; Sirlin, C.B.; Zhu, A.X.; Finn, R.S.; Abecassis, M.M.; Roberts, L.R.; Heimbach, J.K. Diagnosis, Staging, and Management of Hepatocellular Carcinoma: 2018 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology 2018, 68, 723–750. [Google Scholar] [CrossRef]
  86. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  87. Yang, Y.; Yu, Y.; Chen, H.; Meng, X.; Ma, W.; Yu, M.; Li, Z.; Li, C.; Liu, H.; Zhang, X.; et al. Illuminating Platinum Transportation While Maximizing Therapeutic Efficacy by Gold Nanoclusters via Simultaneous Near-Infrared-I/II Imaging and Glutathione Scavenging. ACS Nano 2020, 14, 13536–13547. [Google Scholar] [CrossRef] [PubMed]
  88. Ma, W.; Chen, Q.; Xu, W.; Yu, M.; Yang, Y.; Zou, B.; Zhang, Y.S.; Ding, J.; Yu, Z. Self-Targeting Visualizable Hyaluronate Nanogel for Synchronized Intracellular Release of Doxorubicin and Cisplatin in Combating Multidrug-Resistant Breast Cancer. Nano Res. 2021, 14, 846–857. [Google Scholar] [CrossRef]
  89. Jin, D.; Zhu, Y.; Liu, M.; Yu, W.; Yu, J.; Zheng, X.; Wang, L.; Wu, Y.; Wei, K.; Cheng, J.; et al. A Leaking-Proof Theranostic Nanoplatform for Tumor-Targeted and Dual-Modality Imaging-Guided Photodynamic Therapy. BME Front. 2023, 4, 0015. [Google Scholar] [CrossRef]
  90. Yang, J.D.; Heimbach, J.K. New Advances in the Diagnosis and Management of Hepatocellular Carcinoma. BMJ 2020, 371, m3544. [Google Scholar] [CrossRef] [PubMed]
  91. Wu, H.; Wang, M.; Liang, L.; Xing, H.; Zhang, C.; Shen, F.; Huang, D.; Yang, T. Nanotechnology for Hepatocellular Carcinoma: From Surveillance, Diagnosis to Management. Small 2021, 17, 2005236. [Google Scholar] [CrossRef]
  92. Xu, M.; Yang, L.; Lin, Y.; Lu, Y.; Bi, X.; Jiang, T.; Deng, W.; Zhang, L.; Yi, W.; Xie, Y.; et al. Emerging Nanobiotechnology for Precise Theranostics of Hepatocellular Carcinoma. J. Nanobiotechnol. 2022, 20, 427. [Google Scholar] [CrossRef]
  93. Leung, T.W.T.; Yu, S.; Johnson, P.J.; Geschwind, J.; Vogl, T.J.; Engelmann, K.; Gores, G.J.; Giovannini, M.; O’Grady, J.; Heneghan, M.; et al. Phase II Study of the Efficacy and Safety of Cisplatin-Epinephrine Injectable Gel Administered to Patients With Unresectable Hepatocellular Carcinoma. J. Clin. Oncol. 2003, 21, 652–658. [Google Scholar] [CrossRef]
  94. Zhou, Q.; Sun, X.; Zeng, L.; Liu, J.; Zhang, Z. A Randomized Multicenter Phase II Clinical Trial of Mitoxantrone-Loaded Nanoparticles in the Treatment of 108 Patients with Unresected Hepatocellular Carcinoma. Nanomed. Nanotechnol. Biol. Med. 2009, 5, 419–423. [Google Scholar] [CrossRef]
  95. Chi, X.; Liu, K.; Luo, X.; Yin, Z.; Lin, H.; Gao, J. Recent Advances of Nanomedicines for Liver Cancer Therapy. J. Mater. Chem. B 2020, 8, 3747–3771. [Google Scholar] [CrossRef] [PubMed]
  96. Niu, C.; Wang, L.; Ye, W.; Guo, S.; Bao, X.; Wang, Y.; Xia, Z.; Chen, R.; Liu, C.; Lin, X.; et al. CCAT2 Contributes to Hepatocellular Carcinoma Progression via Inhibiting miR-145 Maturation to Induce MDM2 Expression. J. Cell. Physiol. 2020, 235, 6307–6320. [Google Scholar] [CrossRef] [PubMed]
  97. Sedighi, M.; Rahimi, F.; Rezayan, A.H.; Shahbazi, M.-A.; Witzigmann, D.; Huwyler, J. Combined Cerium Oxide Nanocapping and Layer-by-Layer Coating of Porous Silicon Containers for Controlled Drug Release. J. Mater. Sci. 2018, 53, 14975–14988. [Google Scholar] [CrossRef]
  98. He, M.; Yu, L.; Yang, Y.; Zou, B.; Ma, W.; Yu, M.; Lu, J.; Xiong, G.; Yu, Z.; Li, A. Delivery of Triptolide with Reduction-Sensitive Polymer Nanoparticles for Liver Cancer Therapy on Patient-Derived Xenografts Models. Chin. Chem. Lett. 2020, 31, 3178–3182. [Google Scholar] [CrossRef]
  99. Zhu, J.; Chu, C.; Li, D.; Zhang, Y.; Cheng, Y.; Lin, H.; Wang, X.; Liu, J.; Pang, X.; Cheng, J.; et al. Superior Fluorescent Nanoemulsion Illuminates Hepatocellular Carcinoma for Surgical Navigation. Front. Bioeng. Biotechnol. 2022, 10, 890668. [Google Scholar] [CrossRef]
  100. Lai, X.; Zhao, Y.; Shi, Z.; Xing, L.; Li, X.; Jia, L.; Lin, K. Plant-Derived Paclitaxel-Loaded Ultra-Small Fe3O4 Nanoparticles for MR Imaging-Mediated Antitumor Therapy. Ind. Crops Prod. 2025, 228, 120902. [Google Scholar] [CrossRef]
  101. Herranz-Blanco, B.; Shahbazi, M.; Correia, A.R.; Balasubramanian, V.; Kohout, T.; Hirvonen, J.; Santos, H.A. pH-Switch Nanoprecipitation of Polymeric Nanoparticles for Multimodal Cancer Targeting and Intracellular Triggered Delivery of Doxorubicin. Adv. Healthc. Mater. 2016, 5, 1904–1916. [Google Scholar] [CrossRef]
  102. Shahbazi, M.-A.; Almeida, P.V.; Correia, A.; Herranz-Blanco, B.; Shrestha, N.; Mäkilä, E.; Salonen, J.; Hirvonen, J.; Santos, H.A. Intracellular Responsive Dual Delivery by Endosomolytic Polyplexes Carrying DNA Anchored Porous Silicon Nanoparticles. J. Control. Release 2017, 249, 111–122. [Google Scholar] [CrossRef]
  103. Sedighi, M.; Sieber, S.; Rahimi, F.; Shahbazi, M.-A.; Rezayan, A.H.; Huwyler, J.; Witzigmann, D. Rapid Optimization of Liposome Characteristics Using a Combined Microfluidics and Design-of-Experiment Approach. Drug Deliv. Transl. Res. 2019, 9, 404–413. [Google Scholar] [CrossRef]
  104. Zhang, D.; Song, J.; Jing, Z.; Qin, H.; Wu, Y.; Zhou, J.; Zang, X. Stimulus Responsive Nanocarrier for Enhanced Antitumor Responses Against Hepatocellular Carcinoma. Int. J. Nanomed. 2024, 19, 13339–13355. [Google Scholar] [CrossRef]
  105. Shahbazi, M.-A.; Shrestha, N.; Mäkilä, E.; Araújo, F.; Correia, A.; Ramos, T.; Sarmento, B.; Salonen, J.; Hirvonen, J.; Santos, H.A. A Prospective Cancer Chemo-Immunotherapy Approach Mediated by Synergistic CD326 Targeted Porous Silicon Nanovectors. Nano Res. 2015, 8, 1505–1521. [Google Scholar] [CrossRef]
  106. Shahbazi, M.-A.; Sedighi, M.; Bauleth-Ramos, T.; Kant, K.; Correia, A.; Poursina, N.; Sarmento, B.; Hirvonen, J.; Santos, H.A. Targeted Reinforcement of Macrophage Reprogramming Toward M2 Polarization by IL-4-Loaded Hyaluronic Acid Particles. ACS Omega 2018, 3, 18444–18455. [Google Scholar] [CrossRef]
  107. Figueiredo, P.; Balasubramanian, V.; Shahbazi, M.-A.; Correia, A.; Wu, D.; Palivan, C.G.; Hirvonen, J.T.; Santos, H.A. Angiopep2-Functionalized Polymersomes for Targeted Doxorubicin Delivery to Glioblastoma Cells. Int. J. Pharm. 2016, 511, 794–803. [Google Scholar] [CrossRef] [PubMed]
  108. Baboci, L.; Capolla, S.; Di Cintio, F.; Colombo, F.; Mauro, P.; Dal Bo, M.; Argenziano, M.; Cavalli, R.; Toffoli, G.; Macor, P. The Dual Role of the Liver in Nanomedicine as an Actor in the Elimination of Nanostructures or a Therapeutic Target. J. Oncol. 2020, 2020, 4638192. [Google Scholar] [CrossRef] [PubMed]
  109. Ding, H.; Wang, Y.; Zhang, H. CCND1 Silencing Suppresses Liver Cancer Stem Cell Differentiation and Overcomes 5-Fluorouracil Resistance in Hepatocellular Carcinoma. J. Pharmacol. Sci. 2020, 143, 219–225. [Google Scholar] [CrossRef] [PubMed]
  110. Kinoh, H.; Shibasaki, H.; Liu, X.; Yamasoba, T.; Cabral, H.; Kataoka, K. Nanomedicines Blocking Adaptive Signals in Cancer Cells Overcome Tumor TKI Resistance. J. Control. Release 2020, 321, 132–144. [Google Scholar] [CrossRef]
  111. Sedighi, M.; Rahimi, F.; Shahbazi, M.-A.; Rezayan, A.H.; Kettiger, H.; Einfalt, T.; Huwyler, J.; Witzigmann, D. Controlled Tyrosine Kinase Inhibitor Delivery to Liver Cancer Cells by Gate-Capped Mesoporous Silica Nanoparticles. ACS Appl. Bio Mater. 2020, 3, 239–251. [Google Scholar] [CrossRef]
  112. Abid, N.; Khan, A.M.; Shujait, S.; Chaudhary, K.; Ikram, M.; Imran, M.; Haider, J.; Khan, M.; Khan, Q.; Maqbool, M. Synthesis of Nanomaterials Using Various Top-down and Bottom-Up Approaches, Influencing Factors, Advantages, and Disadvantages: A Review. Adv. Colloid. Interface Sci. 2022, 300, 102597. [Google Scholar] [CrossRef]
  113. Anu Mary Ealia, S.; Saravanakumar, M.P. A Review on the Classification, Characterisation, Synthesis of Nanoparticles and Their Application. IOP Conf. Ser. Mater. Sci. Eng. 2017, 263, 032019. [Google Scholar] [CrossRef]
  114. Kolahalam, L.A.; Kasi Viswanath, I.V.; Diwakar, B.S.; Govindh, B.; Reddy, V.; Murthy, Y.L.N. Review on Nanomaterials: Synthesis and Applications. Mater. Today Proc. 2019, 18, 2182–2190. [Google Scholar] [CrossRef]
  115. Ijaz, I.; Gilani, E.; Nazir, A.; Bukhari, A. Detail Review on Chemical, Physical and Green Synthesis, Classification, Characterizations and Applications of Nanoparticles. Green. Chem. Lett. Rev. 2020, 13, 223–245. [Google Scholar] [CrossRef]
  116. Alhamad, A.A.; Zeghoud, S.; Ben Amor, I.; Zaater, A.; Ben Amor, A.; Aouadif, A.; Hemmami, A.; Chile Nleonu, E. A Short Review of Nanomaterials: Synthesis Methods, Properties, and Applications. Alger. J. Chem. Eng. 2023, 1, 01–07. [Google Scholar] [CrossRef]
  117. Rashwan, D.; Nagy, R.; El-deen, M.A.; Elhakim, H.; Mohamed, M.; Afify, M.; Abd El Hamed, M.; Mohamed, A.E.R. Green Synthesis of Zinc Oxide Nanocomposite Using Fusarium oxysporum and Evaluation of the Anticancer Effect on Hepatocellular Carcinoma. Egypt. J. Chem. 2022, 65, 197–207. [Google Scholar] [CrossRef]
  118. Roy, S.; Ghosh, C.K.; Sarkar, C.K. (Eds.) Nanotechnology: Synthesis to Applications, 1st ed.; CRC Press: Boca Raton, FL, USA, 2017; ISBN 978-1-315-11673-0. [Google Scholar]
  119. Yashveer, S.; Singh, V.; Kaswan, V.; Kaushik, A.; Tokas, J. Green Biotechnology, Nanotechnology and Bio-Fortification: Perspectives on Novel Environment-Friendly Crop Improvement Strategies. Biotechnol. Genet. Eng. Rev. 2014, 30, 113–126. [Google Scholar] [CrossRef]
  120. Musa, I.O.; Isibor, P.O.; Samuel, J.O.; Mustapha, A.; Mustapha, A.; Akande, S.; Oyewole, O.A.; Kayode-Edwards, I.I.; Adeniji, H. Introduction to Nanotoxicology. In Environmental Nanotoxicology; Isibor, P.O., Devi, G., Enuneku, A.A., Eds.; Springer Nature Switzerland: Cham, Switzerland, 2024; pp. 1–22. ISBN 978-3-031-54153-7. [Google Scholar]
  121. Parthiban, E.; Manivannan, N.; Ramanibai, R.; Mathivanan, N. Green Synthesis of Silver-Nanoparticles from Annona Reticulata Leaves Aqueous Extract and Its Mosquito Larvicidal and Anti-Microbial Activity on Human Pathogens. Biotechnol. Rep. 2019, 21, e00297. [Google Scholar] [CrossRef]
  122. Yao, S.; Jin, B.; Liu, Z.; Shao, C.; Zhao, R.; Wang, X.; Tang, R. Biomineralization: From Material Tactics to Biological Strategy. Adv. Mater. 2017, 29, 1605903. [Google Scholar] [CrossRef] [PubMed]
  123. Chen, Y.; Feng, Y.; Deveaux, J.G.; Masoud, M.A.; Chandra, F.S.; Chen, H.; Zhang, D.; Feng, L. Biomineralization Forming Process and Bio-Inspired Nanomaterials for Biomedical Application: A Review. Minerals 2019, 9, 68. [Google Scholar] [CrossRef]
  124. Faivre, D.; Schüler, D. Magnetotactic Bacteria and Magnetosomes. Chem. Rev. 2008, 108, 4875–4898. [Google Scholar] [CrossRef]
  125. Kotakadi, S.M.; Borelli, D.P.R.; Nannepaga, J.S. Therapeutic Applications of Magnetotactic Bacteria and Magnetosomes: A Review Emphasizing on the Cancer Treatment. Front. Bioeng. Biotechnol. 2022, 10, 789016. [Google Scholar] [CrossRef]
  126. Pucci, C.; Degl’Innocenti, A.; Belenli Gümüş, M.; Ciofani, G. Superparamagnetic Iron Oxide Nanoparticles for Magnetic Hyperthermia: Recent Advancements, Molecular Effects, and Future Directions in the Omics Era. Biomater. Sci. 2022, 10, 2103–2121. [Google Scholar] [CrossRef]
  127. He, S.; Guo, Z.; Zhang, Y.; Zhang, S.; Wang, J.; Gu, N. Biosynthesis of Gold Nanoparticles Using the Bacteria Rhodopseudomonas capsulata. Mater. Lett. 2007, 61, 3984–3987. [Google Scholar] [CrossRef]
  128. Schlüter, M.; Hentzel, T.; Suarez, C.; Koch, M.; Lorenz, W.G.; Böhm, L.; Düring, R.-A.; Koinig, K.A.; Bunge, M. Synthesis of Novel Palladium(0) Nanocatalysts by Microorganisms from Heavy-Metal-Influenced High-Alpine Sites for Dehalogenation of Polychlorinated Dioxins. Chemosphere 2014, 117, 462–470. [Google Scholar] [CrossRef] [PubMed]
  129. Singaravelu, G.; Arockiamary, J.S.; Kumar, V.G.; Govindaraju, K. A Novel Extracellular Synthesis of Monodisperse Gold Nanoparticles Using Marine Alga, Sargassum wightii Greville. Colloids Surf. B Biointerfaces 2007, 57, 97–101. [Google Scholar] [CrossRef] [PubMed]
  130. Zhao, W.; Li, J.; Zhong, C.; Zhang, X.; Bao, Y. Green Synthesis of Gold Nanoparticles from Dendrobium officinale and Its Anticancer Effect on Liver Cancer. Drug Deliv. 2021, 28, 985–994. [Google Scholar] [CrossRef]
  131. Ji, Y.; Cao, Y.; Song, Y. Green Synthesis of Gold Nanoparticles Using a Cordyceps militaris Extract and Their Antiproliferative Effect in Liver Cancer Cells (HepG2). Artif. Cells Nanomed. Biotechnol. 2019, 47, 2737–2745. [Google Scholar] [CrossRef]
  132. Usmani, A.; Mishra, A.; Jafri, A.; Arshad, M.; Siddiqui, M.A. Green Synthesis of Silver Nanocomposites of Nigella sativa Seeds Extract for Hepatocellular Carcinoma. Curr. Nanomater. 2019, 4, 191–200. [Google Scholar] [CrossRef]
  133. Liu, Y.; Zeng, Z.; Jiang, O.; Li, Y.; Xu, Q.; Jiang, L.; Yu, J.; Xu, D. Green Synthesis of CuO NPs, Characterization and Their Toxicity Potential against HepG2 Cells. Mater. Res. Express 2021, 8, 015011. [Google Scholar] [CrossRef]
  134. Rawat, M. Green Synthesis of Silver Nanoparticles via Various Plant Extracts for Anti-Cancer Applications. Glob. J. Nanomed. 2017, 2, 555590. [Google Scholar] [CrossRef]
  135. Abed, A.S.; Khalaf, Y.H.; Mohammed, A.M. Green Synthesis of Gold Nanoparticles as an Effective Opportunity for Cancer Treatment. Results Chem. 2023, 5, 100848. [Google Scholar] [CrossRef]
  136. Zhang, H.; Li, T.; Luo, W.; Peng, G.X.; Xiong, J. Green Synthesis of Ag Nanoparticles from Leucus Aspera and Its Application in Anticancer Activity Against Alveolar Cancer. J. Exp. Nanosci. 2022, 17, 47–60. [Google Scholar] [CrossRef]
  137. Mansur, H.S. Quantum Dots and Nanocomposites. WIREs Nanomed. Nanobiotechnol 2010, 2, 113–129. [Google Scholar] [CrossRef]
  138. Rempel, A.A.; Ovchinnikov, O.V.; Weinstein, I.A.; Rempel, S.V.; Kuznetsova, Y.V.; Naumov, A.V.; Smirnov, M.S.; Eremchev, I.U.; Vokhmintsev, A.S.; Savchenko, S.S. Quantum Dots: Modern Methods of Synthesis and Optical Properties. Russ. Chem. Rev. 2024, 93, RCR5114. [Google Scholar] [CrossRef]
  139. Agarwal, K.; Rai, H.; Mondal, S. Quantum Dots: An Overview of Synthesis, Properties, and Applications. Mater. Res. Express 2023, 10, 062001. [Google Scholar] [CrossRef]
  140. Wang, Y.; Hu, A. Carbon Quantum Dots: Synthesis, Properties and Applications. J. Mater. Chem. C 2014, 2, 6921. [Google Scholar] [CrossRef]
  141. Abtahi, M.S.; Fotouhi, A.; Rezaei, N.; Akalin, H.; Ozkul, Y.; Hossein-Khannazer, N.; Vosough, M. Nano-Based Drug Delivery Systems in Hepatocellular Carcinoma. J. Drug Target. 2024, 32, 977–995. [Google Scholar] [CrossRef]
  142. Xing, L.; Chen, Y.; Zheng, T. Research Progress of Nanoparticles in Diagnosis and Treatment of Hepatocellular Carcinoma. Open Life Sci. 2024, 19, 20220932. [Google Scholar] [CrossRef]
  143. Li, K.; Xia, C.; Wang, B.; Chen, H.; Wang, T.; He, Q.; Cao, H.; Wang, Y. Effects of Quantum Dots on the ROS Amount of Liver Cancer Stem Cells. Colloids Surf. B Biointerfaces 2017, 155, 193–199. [Google Scholar] [CrossRef]
  144. Liu, Q.; Wu, D.; Ma, Y.; Cao, Y.; Pang, Y.; Tang, M.; Pu, Y.; Zhang, T. Intracellular Reactive Oxygen Species Trigger Mitochondrial Dysfunction and Apoptosis in Cadmium Telluride Quantum Dots-Induced Liver Damage. NanoImpact 2022, 25, 100392. [Google Scholar] [CrossRef]
  145. Rahman, M.M.; Opo, F.A.D.M.; Asiri, A.M. Cytotoxicity Study of Cadmium-Selenium Quantum Dots (Cdse QDs) for Destroying the Human HepG2 Liver Cancer Cell. J. Biomed. Nanotechnol. 2021, 17, 2153–2164. [Google Scholar] [CrossRef]
  146. Wang, Y.; Chen, J.; Tian, J.; Wang, G.; Luo, W.; Huang, Z.; Huang, Y.; Li, N.; Guo, M.; Fan, X. Tryptophan-Sorbitol Based Carbon Quantum Dots for Theranostics against Hepatocellular Carcinoma. J. Nanobiotechnol. 2022, 20, 78. [Google Scholar] [CrossRef]
  147. Luo, G.; Long, J.; Zhang, B.; Liu, C.; Ji, S.; Xu, J.; Yu, X.; Ni, Q. Quantum Dots in Cancer Therapy. Expert Opin. Drug Deliv. 2012, 9, 47–58. [Google Scholar] [CrossRef]
  148. Mazahir, F.; Sharma, R.; Yadav, A.K. Bioinspired Theranostic Quantum Dots: Paving the Road to a New Paradigm for Cancer Diagnosis and Therapeutics. Drug Discov. Today 2023, 28, 103822. [Google Scholar] [CrossRef]
  149. Jovanović, S.; Marković, Z.; Budimir, M.; Prekodravac, J.; Zmejkoski, D.; Kepić, D.; Bonasera, A.; Marković, B.T. Lights and Dots toward Therapy—Carbon-Based Quantum Dots as New Agents for Photodynamic Therapy. Pharmaceutics 2023, 15, 1170. [Google Scholar] [CrossRef]
  150. Reshma, V.G.; Mohanan, P.V. Assessment of Immunotoxicity and Oxidative Stress Induced by Zinc Selenium/Zinc Sulphide Quantum Dots. Front. Nanotechnol. 2021, 2, 597382. [Google Scholar] [CrossRef]
  151. Chan, W.-H.; Shiao, N.-H.; Lu, P.-Z. CdSe Quantum Dots Induce Apoptosis in Human Neuroblastoma Cells via Mitochondrial-Dependent Pathways and Inhibition of Survival Signals. Toxicol. Lett. 2006, 167, 191–200. [Google Scholar] [CrossRef]
  152. Large, D.E.; Abdelmessih, R.G.; Fink, E.A.; Auguste, D.T. Liposome Composition in Drug Delivery Design, Synthesis, Characterization, and Clinical Application. Adv. Drug Deliv. Rev. 2021, 176, 113851. [Google Scholar] [CrossRef]
  153. Liu, Y.; Castro Bravo, K.M.; Liu, J. Targeted Liposomal Drug Delivery: A Nanoscience and Biophysical Perspective. Nanoscale Horiz. 2021, 6, 78–94. [Google Scholar] [CrossRef]
  154. Cheng, Y.; Zhao, P.; Wu, S.; Yang, T.; Chen, Y.; Zhang, X.; He, C.; Zheng, C.; Li, K.; Ma, X.; et al. Cisplatin and Curcumin Co-Loaded Nano-Liposomes for the Treatment of Hepatocellular Carcinoma. Int. J. Pharm. 2018, 545, 261–273. [Google Scholar] [CrossRef]
  155. Xu, F.; Liao, J.; Xiang, G.; Zhao, P.; Ye, F.; Zhao, Q.; He, X. MiR-101 and Doxorubicin Codelivered by Liposomes Suppressing Malignant Properties of Hepatocellular Carcinoma. Cancer Med. 2017, 6, 651–661. [Google Scholar] [CrossRef]
  156. Wei, X.; Yang, D.; Xing, Z.; Cai, J.; Wang, L.; Zhao, C.; Wei, X.; Jiang, M.; Sun, H.; Zhou, L.; et al. Hepatocyte-Targeted Delivery Using Oleanolic Acid-Loaded Liposomes for Enhanced Hepatocellular Carcinoma Therapy. Biomater. Sci. 2023, 11, 3952–3964. [Google Scholar] [CrossRef]
  157. Samaei, S.S.; Daryab, M.; Gholami, S.; Rezaee, A.; Fatehi, N.; Roshannia, R.; Hashemi, S.; Javani, N.; Rahmanian, P.; Amani-Beni, R.; et al. Multifunctional and Stimuli-Responsive Liposomes in Hepatocellular Carcinoma Diagnosis and Therapy. Transl. Oncol. 2024, 45, 101975. [Google Scholar] [CrossRef]
  158. Li, Y.; Zhang, R.; Xu, Z.; Wang, Z. Advances in Nanoliposomes for the Diagnosis and Treatment of Liver Cancer. Int. J. Nanomed. 2022, 17, 909–925. [Google Scholar] [CrossRef] [PubMed]
  159. Li, J.; Wei, R.; Yao, W.; Pang, X.; Wang, N.; Lai, S.; Wei, X.; Yuan, Y.; Jiang, X.; Yang, R. iRGD-Mediated Liposomal Nanoplatforms for Improving Hepatocellular Carcinoma Targeted Combination Immunotherapy and Monitoring Tumor Response via IVIM-MRI. J. Mater. Chem. B 2024, 12, 9963–9978. [Google Scholar] [CrossRef]
  160. Ashrafizadeh, M.; Delfi, M.; Zarrabi, A.; Bigham, A.; Sharifi, E.; Rabiee, N.; Paiva-Santos, A.C.; Kumar, A.P.; Tan, S.C.; Hushmandi, K.; et al. Stimuli-Responsive Liposomal Nanoformulations in Cancer Therapy: Pre-Clinical & Clinical Approaches. J. Control. Release 2022, 351, 50–80. [Google Scholar] [CrossRef] [PubMed]
  161. Wang, J.; Hu, X.; Xiang, D. Nanoparticle Drug Delivery Systems: An Excellent Carrier for Tumor Peptide Vaccines. Drug Deliv. 2018, 25, 1319–1327. [Google Scholar] [CrossRef]
  162. Gigliobianco, M.R.; Casadidio, C.; Censi, R.; Di Martino, P. Nanocrystals of Poorly Soluble Drugs: Drug Bioavailability and Physicochemical Stability. Pharmaceutics 2018, 10, 134. [Google Scholar] [CrossRef]
  163. Rahman, M.; Almalki, W.H.; Alrobaian, M.; Iqbal, J.; Alghamdi, S.; Alharbi, K.S.; Alruwaili, N.K.; Hafeez, A.; Shaharyar, A.; Singh, T.; et al. Nanocarriers-Loaded with Natural Actives as Newer Therapeutic Interventions for Treatment of Hepatocellular Carcinoma. Expert Opin. Drug Deliv. 2021, 18, 489–513. [Google Scholar] [CrossRef] [PubMed]
  164. Kalyane, D.; Raval, N.; Maheshwari, R.; Tambe, V.; Kalia, K.; Tekade, R.K. Employment of Enhanced Permeability and Retention Effect (EPR): Nanoparticle-Based Precision Tools for Targeting of Therapeutic and Diagnostic Agent in Cancer. Mater. Sci. Eng. C 2019, 98, 1252–1276. [Google Scholar] [CrossRef]
  165. Chen, M.; Quan, G.; Sun, Y.; Yang, D.; Pan, X.; Wu, C. Nanoparticles-Encapsulated Polymeric Microneedles for Transdermal Drug Delivery. J. Control. Release 2020, 325, 163–175. [Google Scholar] [CrossRef]
  166. Beach, M.A.; Nayanathara, U.; Gao, Y.; Zhang, C.; Xiong, Y.; Wang, Y.; Such, G.K. Polymeric Nanoparticles for Drug Delivery. Chem. Rev. 2024, 124, 5505–5616. [Google Scholar] [CrossRef]
  167. Zhang, X.; Zou, L.; Liao, H.; Ren, H.; Niu, H.; Li, Z.; Zhang, X.; Huang, X.; Liu, Y.; Zhou, Z.; et al. Nanoenzyme-Based Sensors for the Detection of Anti-Tumor Drugs. Microchim. Acta 2025, 192, 103. [Google Scholar] [CrossRef]
  168. Holyavka, M.G.; Goncharova, S.S.; Redko, Y.A.; Lavlinskaya, M.S.; Sorokin, A.V.; Artyukhov, V.G. Novel Biocatalysts Based on Enzymes in Complexes with Nano- and Micromaterials. Biophys. Rev. 2023, 15, 1127–1158. [Google Scholar] [CrossRef]
  169. Wen, Y.; Jing, N.; Zhang, M.; Huo, F.; Li, Z.; Yin, C. A Space-Dependent ‘Enzyme-Substrate’ Type Probe Based on ‘Carboxylesterase-Amide Group’ for Ultrafast Fluorescent Imaging Orthotopic Hepatocellular Carcinoma. Adv. Sci. 2023, 10, 2206681. [Google Scholar] [CrossRef] [PubMed]
  170. Liu, G.; Wang, N.; Zhang, C.; Li, M.; He, X.; Yin, C.; Tu, Q.; Shen, X.; Zhang, L.; Lv, J.; et al. Fructose-1,6-Bisphosphate Aldolase B Depletion Promotes Hepatocellular Carcinogenesis Through Activating Insulin Receptor Signaling and Lipogenesis. Hepatology 2021, 74, 3037–3055. [Google Scholar] [CrossRef] [PubMed]
  171. Rossi, M.; Altea-Manzano, P.; Demicco, M.; Doglioni, G.; Bornes, L.; Fukano, M.; Vandekeere, A.; Cuadros, A.M.; Fernández-García, J.; Riera-Domingo, C.; et al. PHGDH Heterogeneity Potentiates Cancer Cell Dissemination and Metastasis. Nature 2022, 605, 747–753. [Google Scholar] [CrossRef]
  172. Wang, X.; Liu, Y.; Han, A.; Tang, C.; Xu, R.; Feng, L.; Yang, Y.; Chen, L.; Lin, Z. The NQO1/P53/SREBP1 Axis Promotes Hepatocellular Carcinoma Progression and Metastasis by Regulating Snail Stability. Oncogene 2022, 41, 5107–5120. [Google Scholar] [CrossRef] [PubMed]
  173. Nie, D.; Tang, X.; Deng, H.; Yang, X.; Tao, J.; Xu, F.; Liu, Y.; Wu, K.; Wang, K.; Mei, Z.; et al. Metabolic Enzyme SLC27A5 Regulates PIP4K2A pre-mRNA Splicing as a Noncanonical Mechanism to Suppress Hepatocellular Carcinoma Metastasis. Adv. Sci. 2024, 11, 2305374. [Google Scholar] [CrossRef]
  174. Shi, X.; Liu, J.; Wang, G. A Peroxidase-like Magneto-Gold Nanozyme AuNC@Fe3O4 with Photothermal Effect for Induced Cell Apoptosis of Hepatocellular Carcinoma Cells In Vitro. Front. Bioeng. Biotechnol. 2023, 11, 1168750. [Google Scholar] [CrossRef]
  175. Jiang, B.; Yan, L.; Zhang, J.; Zhou, M.; Shi, G.; Tian, X.; Fan, K.; Hao, C.; Yan, X. Biomineralization Synthesis of the Cobalt Nanozyme in SP94-Ferritin Nanocages for Prognostic Diagnosis of Hepatocellular Carcinoma. ACS Appl. Mater. Interfaces 2019, 11, 9747–9755. [Google Scholar] [CrossRef]
  176. Luo, B.; Zhou, J.; Zhan, X.; Ying, B.; Lan, F.; Wu, Y. Visual and Colorimetric Detection of microRNA in Clinical Samples Based on Strand Displacement Amplification and Nanozyme-Mediated CRISPR-Cas12a System. Talanta 2024, 277, 126310. [Google Scholar] [CrossRef]
  177. Wen, L.; Liang, C.; Chen, E.; Chen, W.; Liang, F.; Zhi, X.; Wei, T.; Xue, F.; Li, G.; Yang, Q.; et al. Regulation of Multi-Drug Resistance in Hepatocellular Carcinoma Cells Is TRPC6/Calcium Dependent. Sci. Rep. 2016, 6, 23269. [Google Scholar] [CrossRef] [PubMed]
  178. Duan, B.; Huang, C.; Bai, J.; Zhang, Y.L.; Wang, X.; Yang, J.; Li, J. Multidrug Resistance in Hepatocellular Carcinoma. In Hepatocellular Carcinoma; Tirnitz-Parker, J.E.E., Ed.; Codon Publications: Singapore, 2019; pp. 141–158. ISBN 978-0-9944381-8-8. [Google Scholar]
  179. Majidinia, M.; Mirza-Aghazadeh-Attari, M.; Rahimi, M.; Mihanfar, A.; Karimian, A.; Safa, A.; Yousefi, B. Overcoming Multidrug Resistance in Cancer: Recent Progress in Nanotechnology and New Horizons. IUBMB Life 2020, 72, 855–871. [Google Scholar] [CrossRef]
  180. Md, S.; Alhakamy, N.A.; Sharma, P.; Ansari, M.S.; Gorain, B. Nanocarrier-Based Co-Delivery Approaches of Chemotherapeutics with Natural P-Glycoprotein Inhibitors in the Improvement of Multidrug Resistance Cancer Therapy. J. Drug Target. 2022, 30, 801–818. [Google Scholar] [CrossRef] [PubMed]
  181. Duan, C.; Yu, M.; Xu, J.; Li, B.-Y.; Zhao, Y.; Kankala, R.K. Overcoming Cancer Multi-Drug Resistance (MDR): Reasons, Mechanisms, Nanotherapeutic Solutions, and Challenges. Biomed. Pharmacother. 2023, 162, 114643. [Google Scholar] [CrossRef]
  182. Yoo, H.; Kim, Y.; Kim, J.; Cho, H.; Kim, K. Overcoming Cancer Drug Resistance with Nanoparticle Strategies for Key Protein Inhibition. Molecules 2024, 29, 3994. [Google Scholar] [CrossRef] [PubMed]
  183. Lepeltier, E.; Rijo, P.; Rizzolio, F.; Popovtzer, R.; Petrikaite, V.; Assaraf, Y.G.; Passirani, C. Nanomedicine to Target Multidrug Resistant Tumors. Drug Resist. Updates 2020, 52, 100704. [Google Scholar] [CrossRef]
  184. Stan, D.; Enciu, A.-M.; Mateescu, A.L.; Ion, A.C.; Brezeanu, A.C.; Stan, D.; Tanase, C. Natural Compounds With Antimicrobial and Antiviral Effect and Nanocarriers Used for Their Transportation. Front. Pharmacol. 2021, 12, 723233. [Google Scholar] [CrossRef]
  185. Xiao, Y.; Liu, J.; Guo, M.; Zhou, H.; Jin, J.; Liu, J.; Liu, Y.; Zhang, Z.; Chen, C. Synergistic Combination Chemotherapy Using Carrier-Free Celastrol and Doxorubicin Nanocrystals for Overcoming Drug Resistance. Nanoscale 2018, 10, 12639–12649. [Google Scholar] [CrossRef]
  186. Colone, M.; Calcabrini, A.; Stringaro, A. Drug Delivery Systems of Natural Products in Oncology. Molecules 2020, 25, 4560. [Google Scholar] [CrossRef]
  187. Kong, F.-H.; Ye, Q.-F.; Miao, X.-Y.; Liu, X.; Huang, S.-Q.; Xiong, L.; Wen, Y.; Zhang, Z.-J. Current Status of Sorafenib Nanoparticle Delivery Systems in the Treatment of Hepatocellular Carcinoma. Theranostics 2021, 11, 5464–5490. [Google Scholar] [CrossRef]
  188. Khafaga, D.S.R.; El-Khawaga, A.M.; Elfattah Mohammed, R.A.; Abdelhakim, H.K. Green Synthesis of Nano-Based Drug Delivery Systems Developed for Hepatocellular Carcinoma Treatment: A Review. Mol. Biol. Rep. 2023, 50, 10351–10364. [Google Scholar] [CrossRef] [PubMed]
  189. Ratan, Z.A.; Haidere, M.F.; Nurunnabi, M.; Shahriar, S.M.; Ahammad, A.J.S.; Shim, Y.Y.; Reaney, M.J.T.; Cho, J.Y. Green Chemistry Synthesis of Silver Nanoparticles and Their Potential Anticancer Effects. Cancers 2020, 12, 855. [Google Scholar] [CrossRef]
  190. Gao, Q.; Chen, N.; Li, B.; Zu, M.; Ma, Y.; Xu, H.; Zhu, Z.; Reis, R.L.; Kundu, S.C.; Xiao, B. Natural Lipid Nanoparticles Extracted from Morus nigra L. Leaves for Targeted Treatment of Hepatocellular Carcinoma via the Oral Route. J. Nanobiotechnol. 2024, 22, 4. [Google Scholar] [CrossRef] [PubMed]
  191. Chen, X.; Hu, X.; Hu, J.; Qiu, Z.; Yuan, M.; Zheng, G. Celastrol-Loaded Galactosylated Liposomes Effectively Inhibit AKT/c-Met-Triggered Rapid Hepatocarcinogenesis in Mice. Mol. Pharm. 2020, 17, 738–747. [Google Scholar] [CrossRef] [PubMed]
  192. Elwan, A.G.; Mohamed, T.M.; Beltagy, D.M.; El Gamal, D.M. The Therapeutic Role of Naringenin Nanoparticles on Hepatocellular Carcinoma. BMC Pharmacol. Toxicol. 2025, 26, 3. [Google Scholar] [CrossRef]
  193. Khaled, A.M.; Othman, M.S.; Obeidat, S.T.; Aleid, G.M.; Aboelnaga, S.M.; Fehaid, A.; Hathout, H.M.R.; Bakkar, A.A.; Moneim, A.E.A.; El-Garawani, I.M.; et al. Green-Synthesized Silver and Selenium Nanoparticles Using Berberine: A Comparative Assessment of In Vitro Anticancer Potential on Human Hepatocellular Carcinoma Cell Line (HepG2). Cells 2024, 13, 287. [Google Scholar] [CrossRef]
  194. Roquito, T.; Colaço, M.; Costa, J.P.; Borges, O. Curcumin-Encapsulated Glucan Nanoparticles as an Oxidative Stress Modulator against Human Hepatic Cancer Cells. Colloids Surf. B Biointerfaces 2025, 245, 114326. [Google Scholar] [CrossRef]
  195. Asl, A.M.; Kalaee, M.; Abdouss, M.; Homami, S.S. Novel Targeted Delivery of Quercetin for Human Hepatocellular Carcinoma Using Starch/Polyvinyl Alcohol Nanocarriers Based Hydrogel Containing Fe2O3 Nanoparticles. Int. J. Biol. Macromol. 2024, 257, 128626. [Google Scholar] [CrossRef]
  196. Leng, M.; Jiang, H.; Zhang, S.; Bao, Y. Green Synthesis of Gold Nanoparticles from Polygahatous Polysaccharides and Their Anticancer Effect on Hepatic Carcinoma through Immunoregulation. ACS Omega 2024, 9, 21144–21151. [Google Scholar] [CrossRef]
  197. Chen, S.; Wang, Q. Green Synthesis of AuNPs and Their Effect on Cell Apoptosis of HepG2 Cells for Applications in Treatment of Hepatocellular Carcinoma in Nursing Care. Plasmonics 2024, 19, 1553–1563. [Google Scholar] [CrossRef]
  198. Fakhar-E-Alam, M.; Shafiq, Z.; Mahmood, A.; Atif, M.; Anwar, H.; Hanif, A.; Yaqub, N.; Farooq, W.A.; Fatehmulla, A.; Ahmad, S.; et al. Assessment of Green and Chemically Synthesized Copper Oxide Nanoparticles against Hepatocellular Carcinoma. J. King Saud. Univ. Sci. 2021, 33, 101669. [Google Scholar] [CrossRef]
  199. Hsu, C.-Y.; Mustafa, M.A.; Kumar, A.; Pramanik, A.; Sharma, R.; Mohammed, F.; Jawad, I.A.; Mohammed, I.J.; Alshahrani, M.Y.; Ali Khalil, N.A.M.; et al. Exploiting the Immune System in Hepatic Tumor Targeting: Unleashing the Potential of Drugs, Natural Products, and Nanoparticles. Pathol. Res. Pract. 2024, 256, 155266. [Google Scholar] [CrossRef] [PubMed]
  200. Mandlik, D.S.; Mandlik, S.K. Herbal and Natural Dietary Products: Upcoming Therapeutic Approach for Prevention and Treatment of Hepatocellular Carcinoma. Nutr. Cancer 2021, 73, 2130–2154. [Google Scholar] [CrossRef] [PubMed]
  201. Bonferoni, M.C.; Gavini, E.; Rassu, G.; Maestri, M.; Giunchedi, P. Chitosan Nanoparticles for Therapy and Theranostics of Hepatocellular Carcinoma (HCC) and Liver-Targeting. Nanomaterials 2020, 10, 870. [Google Scholar] [CrossRef]
  202. Karimi, K.; Mojtabavi, S.; Tehrany, P.M.; Nejad, M.M.; Rezaee, A.; Mohtashamian, S.; Hamedi, E.; Yousefi, F.; Salmani, F.; Zandieh, M.A.; et al. Chitosan-Based Nanoscale Delivery Systems in Hepatocellular Carcinoma: Versatile Bio-Platform with Theranostic Application. Int. J. Biol. Macromol. 2023, 242, 124935. [Google Scholar] [CrossRef]
  203. Lee, H.D.; Yuan, L.-Y. Nano-Revolution in Hepatocellular Carcinoma: A Multidisciplinary Odyssey—Are We There Yet? World J. Hepatol. 2024, 16, 684–687. [Google Scholar] [CrossRef]
  204. Sun, Y.; Ma, W.; Yang, Y.; He, M.; Li, A.; Bai, L.; Yu, B.; Yu, Z. Cancer Nanotechnology: Enhancing Tumor Cell Response to Chemotherapy for Hepatocellular Carcinoma Therapy. Asian J. Pharm. Sci. 2019, 14, 581–594. [Google Scholar] [CrossRef]
  205. Ren, X.; Su, D.; Shi, D.; Xiang, X. The Improving Strategies and Applications of Nanotechnology-Based Drugs in Hepatocellular Carcinoma Treatment. Front. Bioeng. Biotechnol. 2023, 11, 1272850. [Google Scholar] [CrossRef]
  206. Liu, M.; Yan, C.; Han, J.; Guo, Z.; Wu, Y.; Huang, J.; Xiao, Z.; Zhu, W.-H. Engineering Photo-Controllable Fragrance Release with Flash Nanoprecipitation. Green. Chem. Eng. 2021, 2, 301–308. [Google Scholar] [CrossRef]
  207. Chavda, V.P.; Balar, P.C.; Patel, S.B. Interventional Nanotheranostics in Hepatocellular Carcinoma. Nanotheranostics 2023, 7, 128–141. [Google Scholar] [CrossRef]
  208. Ray, P.; Haideri, N.; Haque, I.; Mohammed, O.; Chakraborty, S.; Banerjee, S.; Quadir, M.; Brinker, A.; Banerjee, S. The Impact of Nanoparticles on the Immune System: A Gray Zone of Nanomedicine. J. Immunol. Sci. 2021, 5, 19–33. [Google Scholar] [CrossRef]
  209. Liu, J.; Liu, Z.; Pang, Y.; Zhou, H. The Interaction between Nanoparticles and Immune System: Application in the Treatment of Inflammatory Diseases. J. Nanobiotechnol. 2022, 20, 127. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Scheme of green synthesis method for nanoparticles.
Figure 1. Scheme of green synthesis method for nanoparticles.
Pharmaceutics 17 00673 g001
Figure 2. QD induces ROS formation as a promising strategy for treating HCC via apoptosis.
Figure 2. QD induces ROS formation as a promising strategy for treating HCC via apoptosis.
Pharmaceutics 17 00673 g002
Figure 3. The toxicological pathway of QDs resulting in the formation of ROS leading to cell death.
Figure 3. The toxicological pathway of QDs resulting in the formation of ROS leading to cell death.
Pharmaceutics 17 00673 g003
Figure 4. Liposome-based delivery system in HCC.
Figure 4. Liposome-based delivery system in HCC.
Pharmaceutics 17 00673 g004
Figure 5. Polymeric delivery system exhibits cytotoxic effects on HCC, highlighting their potential as a targeted therapeutic strategy for liver cancer treatment.
Figure 5. Polymeric delivery system exhibits cytotoxic effects on HCC, highlighting their potential as a targeted therapeutic strategy for liver cancer treatment.
Pharmaceutics 17 00673 g005
Figure 6. The synergistic effects against MDR in liver cancer cells by up-regulating ROS, Cytochrome C, and pro-apoptotic markers.
Figure 6. The synergistic effects against MDR in liver cancer cells by up-regulating ROS, Cytochrome C, and pro-apoptotic markers.
Pharmaceutics 17 00673 g006
Table 3. Delivery systems of natural products for HCC treatment.
Table 3. Delivery systems of natural products for HCC treatment.
NanoparticleNatural ProductSize of NanocompositeCharacterization TechniquesModelAdvantagesReference
Liposome of biocompatible matrix DSPE-PEG2000 decorated with short peptide ligand SP94Curcumin and resveratrol<200 nmTEM, DLS, and UV–VIS In vivo and in vitro
-
Increasing tumor apoptosis by increasing ROS generation.
-
Targeting peptide SP94 improves drug accumulation in tumors via the EPR effect.
[21]
Morus nigra L.-derived lipid nanoparticles (MLNPs)Fresh leaves of Morus nigra L.Approximately 100 nm
Hydrodynamic size 162.1 nm
AFM, DLS, and TEMIn vivo and In vitro
-
MLNPs caused cell cycle arrest at the G0/G1 phase and induced apoptosis.
-
They also triggered a surge in intracellular ROS levels and significantly inhibited the proliferation and migration of hepatoma cells.
[190]
Galactose-modified PEGylated liposomes (C-GPL)Celastrol139.4 ± 2.7 nmTEM, and DLSIn vivo and In vitro
-
Enhancing its cellular uptake and reducing its side effects.
-
Effectively inhibited the development of HCC by suppressing AKT activation.
-
Inducing cell apoptosis and retarding cell proliferation
[191]
Naringenin nanoparticles (NARNPs)Naringenin54.96 ± 18.6 nm TEM
31.79 ± 6.8 nm SEM
SEM, TEM, FT-IR, and XRDIn vitro
-
NARNPs effectively inhibit cell proliferation and induce apoptosis.
-
No cytotoxic effects on normal cells.
[192]
SeNPs and AgNPsBerberine171.5 ± 4.2 nmDLSIn vitro
-
Inhibit the growth and migration of HepG2 by triggering oxidative stress and apoptotic cascades.
[193]
Glucan nanoparticles Curcumin111.0 ± 49.0 nmDLS, ELS, and Cryo-SEM In vitro
-
Induce ROS in HepG2 cells, impacting oxidative stress.
-
NPs modulate cancer therapy by affecting inflammatory chemokines like RANTES.
-
Encapsulation enhances curcumin’s bioactivity, affecting cellular responses.
[194]
Fe2O3/Starch/Polyvinyl alcohol nanocarrier (Fe2O3/S/PVA NC)Quercetin (QC)240–340 nmFTIR, XRD, FE-SEM, DLS, and VSM In vitro
-
Suppressing HepG2 cancer cells by targeted delivery of QC drug.
-
Fe2O3/S/PVA caused 6 % of HepG2 cells to early apoptotic death.
[195]
AuNPsPolystyrene polysaccharide extracted from Polygonatum91–459 nmUV–VIS, TEM, AFM, FTIR, DLS, and EDXIn vitro and In vivo
-
Enhancing immunoregulation.
-
Higher cancer suppression rates.
[196]
AuNPsPanicum maximum leaf extract45∼50 nmTEM, and UV–VisIn vitro
-
Induce cell apoptosis and ROS.
[197]
CuONPsNeem leaves extract
Azadirachta indica
15∼16 nmXRD, FTIR, and UV–VISIn vitro
-
Decline of cell viability caused by DNA destruction.
[198]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Muteeb, G.; El-Morsy, M.T.; Abo-Taleb, M.A.; Mohamed, S.K.; Khafaga, D.S.R. Herbal Medicine: Enhancing the Anticancer Potential of Natural Products in Hepatocellular Carcinoma Therapy Through Advanced Drug Delivery Systems. Pharmaceutics 2025, 17, 673. https://doi.org/10.3390/pharmaceutics17050673

AMA Style

Muteeb G, El-Morsy MT, Abo-Taleb MA, Mohamed SK, Khafaga DSR. Herbal Medicine: Enhancing the Anticancer Potential of Natural Products in Hepatocellular Carcinoma Therapy Through Advanced Drug Delivery Systems. Pharmaceutics. 2025; 17(5):673. https://doi.org/10.3390/pharmaceutics17050673

Chicago/Turabian Style

Muteeb, Ghazala, Manar T. El-Morsy, Mustafa Ali Abo-Taleb, Salma K. Mohamed, and Doaa S. R. Khafaga. 2025. "Herbal Medicine: Enhancing the Anticancer Potential of Natural Products in Hepatocellular Carcinoma Therapy Through Advanced Drug Delivery Systems" Pharmaceutics 17, no. 5: 673. https://doi.org/10.3390/pharmaceutics17050673

APA Style

Muteeb, G., El-Morsy, M. T., Abo-Taleb, M. A., Mohamed, S. K., & Khafaga, D. S. R. (2025). Herbal Medicine: Enhancing the Anticancer Potential of Natural Products in Hepatocellular Carcinoma Therapy Through Advanced Drug Delivery Systems. Pharmaceutics, 17(5), 673. https://doi.org/10.3390/pharmaceutics17050673

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop