The role of medicinal plants in the management of hepatocellular carcinoma and its metastasis
Sections
Open Access Review
The role of medicinal plants in the management of hepatocellular carcinoma and its metastasis

Affiliation:

Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry 605006, India

ORCID: https://orcid.org/0009-0008-6006-8257

Manoj Kumar Nagar

Affiliation:

Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry 605006, India

ORCID: https://orcid.org/0009-0008-9471-1458

Deepthi Sudha

Affiliation:

Liver Diseases Research Lab, Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry 605006, India

Email: balasubramaniyan.v@jipmer.edu.in

ORCID: https://orcid.org/0000-0003-1708-4864

Balasubramaniyan Vairappan
*

Explor Dig Dis. 2025;4:100588 DOI: https://doi.org/10.37349/edd.2025.100588

Received: April 09, 2025 Accepted: July 30, 2025 Published: August 24, 2025

Academic Editor: Jose Carlos Fernandez-Checa, Institute of Biomedical Research of Barcelona (IIBB-CSIC), Spain

Abstract

Hepatocellular carcinoma (HCC) ranks as the sixth most diagnosed cancer and the third most common cancer-related death globally. The underlying precise molecular mechanisms for its progression remain poorly understood. Interestingly, approximately 90% of HCC-related deaths are not due to the primary tumor itself but rather to its difficult-to-treat metastatic spread. Despite sorafenib being the first-line therapy for HCC, challenges such as drug resistance, frequent recurrence, and metastasis contribute to poor prognosis. In this context, alternative therapeutic strategies are urgently needed. A broad spectrum of phytochemicals, including polyphenolic derivatives, flavonoids, carotenoids, alkaloids, terpenes, lignans, and saponins, has shown considerable promise as potential anti-cancer agents, both in vitro and in vivo. These natural plant-derived compounds exhibit distinct and overlapping mechanisms of action, characterized by their antioxidant, anti-inflammatory, and anti-cancer properties, offering a novel approach to HCC treatment. An extensive literature search was conducted from 2010 to 2024 using reputable electronic databases such as MEDLINE, Embase, Google Scholar, Science Direct, and other reliable sources using different keywords, including HCC, medicinal plants in HCC, HCC metastasis, and mechanism of action of medicinal plants in HCC, among others. This comprehensive review aims to summarize the potential role of plant-based bioactive components in combating HCC through various cellular mechanisms, highlighting their therapeutic potential in the management of both primary and metastatic disease.

Keywords

Anti-cancer drugs, epithelial-mesenchymal transition (EMT), liver cancer, metastasis, phytochemicals

Introduction

Hepatocellular carcinoma (HCC) is the 3rd leading cause of cancer-related death and 6th most common type of cancer globally. According to the Global Cancer Observatory (GLOBOCON 2022, version 1.1), approximately 870,000 new cases of liver cancer were reported globally, with around 760,000 deaths attributed to the disease, reflecting the varying incidence rates worldwide [1]. Notably, the expected incidence of HCC is projected to increase by 55% from 2020 to 2040. The majority of HCC cases are concentrated in East and Southeast Asia, with China accounting for 62.4% of cases, followed by Japan (7.0%), India (5.3%), Thailand (4.2%), and Vietnam (4.0%) [2, 3]. Most HCC cases occur in individuals with cirrhosis, with the highest mortality rate observed in alcoholic liver disease (ALD), followed by MAFLD (metabolic dysfunction-associated fatty liver disease), and HBV (hepatitis B virus) or HCV-related cirrhosis [2, 3].

Moreover, Mongolia has the highest HCC incidence globally, while China reports the majority of cases in Asia, followed by Japan, India, Thailand, and Vietnam [4]. HCC incidents vary by country due to differences in screening practices, etiological factors, and treatment strategies. Hepatic viral diseases, particularly HBV and HCV, were historically the leading cause of HCC, but MAFLD now surpasses them in many countries, except in Africa, where HBV and aflatoxin exposure remain dominant risk factors. In regions like Northern Africa, Western Europe, and the USA, HCV remains a significant contributor to HCC [5]. Furthermore, metabolic disorders such as alpha 1-antitrypsin deficiency, Wilson’s disease, porphyria, and hereditary hemochromatosis are linked to the development of HCC, often in the context of cirrhosis [6]. Autoimmune conditions, including autoimmune hepatitis and primary sclerosing cholangitis, also play a role in HCC pathogenesis [7].

Pathophysiology of HCC

In HCC, invasion typically begins within the liver parenchyma, known as intrahepatic metastasis, where cancer cells circulate and infiltrate adjacent liver tissues. This process is facilitated by various molecular mechanisms, including alterations in cell-cell and cell-extracellular matrix (ECM) interactions, dysregulation of signaling pathways, and remodeling of the ECM. Invasion can be classified as either micro or macro. The majority of invasion occurs in the portal vein (approximately 60%), while only 20% occurs in the hepatic vein [8]. On the other hand, metastasis refers to the spread of cancer cells from the primary malignancy site to distant parts of the body. Radiological reports indicate that metastasis occurs through various routes, including the hematogenous route, lymphatic spread, bile duct invasion, and direct extrahepatic invasion. The hematogenous route is the most common pathway for HCC metastasis, primarily due to the invasion of intrahepatic arterioles and venules that drain into the pulmonary circulation. The lungs (the most common site), bones, and adrenal glands are frequently affected via the hematogenous route, while the diaphragm, abdominal wall, and peritoneum are commonly involved through direct extrahepatic invasion [9, 10].

A key molecular mechanism driving HCC invasion is the deregulation of epithelial-mesenchymal transition (EMT). EMT is a biological process in which epithelial cells lose their polarity and cell-cell adhesion, acquiring mesenchymal traits that enable them to migrate, invade surrounding tissues, and resist apoptosis. Previously, EMT was viewed as a simple binary transition between epithelial and mesenchymal states. However, recent studies reveal that EMT involves varying degrees of phenotypic changes, including partial EMT, intermediate EMT, extreme EMT, and ameboid EMT. In all phases, the ameboid stage shows a high potential for extravasation and metastasis due to squeezing mobility [11]. EMT changes are driven by factors such as disruption of cell-cell adhesion proteins, activation of transcription factors, alterations in cell surface proteins, increased production of degrading enzymes, and activation of key signaling pathways. Molecular markers like E-cadherin and cytokeratin are associated with epithelial cells, while N-cadherin and vimentin are linked to mesenchymal cells with migratory capabilities. Mechanisms altering these protein expressions include upregulation of retinoic acid receptor γ (RARγ), a nuclear receptor that promotes cell migration and invasion by downregulating E-cadherin, and homeodomain-interacting protein kinase 8 (HDGF8), a histone demethylase that also facilitates HCC cell migration [12, 13]. In addition, cytoskeletal remodeling, regulated by RhoGAP (an enzyme that hydrolyzes GTP to GDP), plays a crucial role in cell migration by modulating E-cadherin expression and actin polymerization [14].

Angiogenesis is a key process in HCC invasion and metastasis, involving the activation of endothelial cells to form new blood vessels, which provide nutrients to cancer cells. This process is regulated by various mechanisms; under hypoxic conditions, increased expression of HIF-1α (hypoxia inducible factor-1α) (a hypoxia-induced transcription factor) activates several angiogenic factors in HCC [15]. Pro-angiogenic factors include epidermal growth factor (EGF) [16], platelet-derived growth factor (PDGF) [17], fibroblast growth factor (FGF) [18], endoglin [19], and leptin [20], while anti-angiogenic factors like thrombospondin-1 [21] and endostatin [22] counteract this process. Invasion also involves complex interactions within the tumor microenvironment (TME), which comprises stromal cells such as hepatic stellate cells (HSCs), cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), and tumor-associated neutrophils (TANs) [23]. HSCs play a critical role in invasion and metastasis by secreting pro-angiogenic factors like vascular endothelial growth factor receptor (VEGFR) and PDGF, along with IL-8, which stimulates new blood vessel formation [24]. Additionally, HSCs modulate immune responses by promoting the transition of macrophages from the M1 to M2 phenotype via the CCL2/CCR2 pathway [25].

Current therapeutic landscape

The treatment of HCC faces significant challenges due to limited therapeutic options. Clinical management is complex and varies depending on tumor stage, liver function, and the patient’s overall health. The Barcelona Clinic Liver Cancer (BCLC) staging system and the Child-Pugh score are widely accepted tools for guiding therapeutic decisions [26, 27]. Patients in the early stages (BCLC 0/A) with preserved liver function and minimal tumor burden are typically eligible for potentially curative treatments, including hepatic resection, local ablation [such as radiofrequency ablation (RFA) or microwave ablation (MWA)], or liver transplantation (LT). The choice among these options depends on factors such as portal hypertension, bilirubin levels, and the presence of multiple nodules [28]. Hepatic resection is best suited for non-cirrhotic or cirrhotic patients with a single nodule (BCLC stage 0 or A), Child-Pugh A liver function, and no significant portal hypertension. Surgical resection offers a 5-year survival rate of ~ 70% though tumor recurrence is common, occurring in 75–80% of patients within 5 years. Recurrence is especially prevalent in cases related to viral hepatitis, particularly HCV [29, 30]. For patients who are not suitable for surgery, LT is a viable alternative. Eligibility typically follows the Milan criteria (a single lesion ≤ 5 cm or 2–3 nodules ≤ 3 cm without vascular invasion). LT provides favourable long-term outcomes, with low recurrence rates and a 10-year survival rate of around 70% [28]. In regions with limited deceased donor organs, living donor LT is practiced. Post-transplant immunosuppressants like tacrolimus are essential to prevent graft rejection, which is closely linked to recurrence risk. Sirolimus, however, has shown potential in reducing tumor progression [31, 32]. For patients ineligible for surgery or transplantation, non-surgical locoregional therapies such as RFA, MWA, percutaneous ethanol injection (PEI), and transarterial chemoembolization (TACE) are preferred. RFA is especially effective for nodules smaller than 2 cm and serves as an alternate to surgery for early-stage HCC, with a median overall survival of about 60 months and 5-year recurrence rates of 50–70% [33]. TACE is the first-line therapy for intermediate-stage HCC (BCLC-B), particularly in patients with preserved liver function (Child-Pugh A) and no vascular invasion or metastasis. More recently, selective internal radiation therapy (SIRT) with yttrium-90 microspheres has been used palliatively in BCLC-B patients; however, phase 3 trials have not demonstrated superior survival compared to sorafenib, alone or combined [34, 35].

Advanced HCC with BCLC-D patients are treated with first-line or second-line systemic therapies. Current systemic therapies include targeted therapy and immunotherapy. Sorafenib, an oral multi-kinase inhibitor, targets serine/threonine kinases RAF-1 and RAF, as well as tyrosine kinases such as VEGFR-1, -2, -3, and PDGFR-β, which are crucial in cancer development. Approved in 2007 as the first-line treatment for advanced HCC, sorafenib has been associated with adverse effects including hand-foot syndrome (7.0%), asthenia (7.4%), and diarrhea (13.1%), which can impact quality of life and lead to treatment discontinuation [36, 37]. Another option, regorafenib also a multi-kinase inhibitor targets VEGFRs 1–3, TIE2, RET, and RAF-1. It inhibits tumor cell proliferation, induces apoptosis, and exerts anti-angiogenic effects by blocking multiple pathways [38]. However, it is also linked to adverse reactions such as fatigue, hypertension, hand-foot skin reactions, and gastrointestinal symptoms, including diarrhea, nausea, and anorexia. Hepatotoxicity is a particular concern in HCC patients, who often have compromised liver function [39].

Another treatment option for HCC is the combination of atezolizumab and bevacizumab. Atezolizumab is a monoclonal antibody that inhibits PD-L1 (programmed death-ligand 1), while bevacizumab targets VEGF, a key mediator of angiogenesis [40]. In 2020, this combination was approved as a new standard treatment for patients with unresectable HCC. However, atezolizumab may cause immune-related adverse events such as hepatitis, pneumonitis, and colitis, while bevacizumab is associated with hypertension, bleeding, thromboembolism, and proteinuria. The combination therapy can increase the overall risk of these side effects [41]. A comparative overview of these therapies, including their mechanisms, clinical outcomes, and toxicity profiles, is presented in Table 1.

 Systemic treatments for advanced HCC: a comparative analysis of efficacy, survival outcomes, and safety profiles

DrugTargetBenefitsMedian OS and hazard ratioCommon adverse effectsReference
SorafenibTKI—VEGFR, PDGFR, RAFFirst-line for advanced HCC, delays HCC progression6.5 months (5.56–7.56) and 0.68HFSR, diarrhea, fatigue, hypertension[36]
DonafenibTKI—RAF/MEK/ERKModified structure of sorafenib12.1 months and 0.831HFSR, diarrhea, and elevated LFT[42]
LenvatinibTKI—VEGFR, FGFR, PDGFR, RET, KITNon-inferior to sorafenib, higher response rate (24%)13.6 months (range from 12.1–14.9) and 0.92Palmar-plantar erythrodysesthesia, hypertension, weight loss, proteinuria, diarrhea[43]
RegorafenibTKI—VEGFR, PDGFR, FGFRSecond-line after sorafenib, OS benefit (~ 3 months)10.6 months (range from 9.1–12.1) and 0.63Fatigue, diarrhea, hypertension, HFSR[38]
CabozantinibTKI—MET, VEGFR, AXLSecond/Third-line, OS and PFS benefit, activity in bone/lung metastases10.2 months and 0.76Diarrhea, HFSR, fatigue, increased LFTs[44]
RamucirumabAnti-VEGFR-2 monoclonal antibodySecond-line if AFP > 400 ng/mL, improves OS in AFP-high HCC8.5 months (range from 7.0–10.6) and 0.710Hypertension, proteinuria, bleeding, fatigue[45]
ApatinibTKI of VEGFR-2Second-line treatment after chemotherapy failure8.7 months (range from 7.5–9.8) and 0.785Hypertension, HFSR[46]
FOLFOX (5-FU + leucovorin + oxaliplatin)Cytotoxic chemotherapySome benefit in Asia, used in advanced/metastatic HCC5.9 months and 0.75Hematological symptoms, diarrhea, neuropathy[47]
Nivolumab + ipilimumabCheckpoint inhibitors (PD-1 + CTLA-4)Active in metastatic HCC23.7 months (range from 18.8–29.4) and 0.76Hepatitis-colitis, endocrinopathies, skin rash[48]
Atezolizumab + bevacizumabPD-L1 + VEGF inhibitionFirst-line standard in advanced HCC19.2 months (range from 17.0–23.7) and 0.66Hypertension, GI bleed, fatigue, immune hepatitis[40]

5-FU: 5-fluorouracil; AFP: alpha-fetoprotein; GI: gastrointestinal; HCC: hepatocellular carcinoma; HFSR: hand-foot skin reaction; LFT: liver function test; OS: overall survival; PD-1: programmed death-1; PD-L1: programmed death-ligand 1; PFS: progression-free survival; TKI: tyrosine kinase inhibitor; VEGF: vascular endothelial growth factor; VEGFR: VEGF receptor

Another immunotherapy target is CTLA-4, a CD28 homolog that inhibits T-cell activation by preventing CD28 from binding to CD80/CD86. Monoclonal antibodies against CTLA-4 reduce helper T-cell activity and enhance regulatory T-cell (Treg) function, thereby suppressing immune responses. A phase I/II study (NCT02519348) evaluated tremelimumab (anti-CTLA-4 antibody) combined with durvalumab (T300 + D) versus monotherapy with either agent. The combination showed a higher overall response but also a greater incidence of adverse events compared to targeted therapies [49].

Need for developing plant-based alternative therapies for HCC

HCC remains a major clinical challenge due to limited treatment options and significant side effects associated with current therapies. Plant-derived phytochemicals offer a promising alternative, as they can reduce cancer burden without causing severe toxicity to normal tissues.

These compounds act through various mechanisms, including antioxidant, anti-inflammatory, and pro-apoptotic pathways (Figure 1). Phytochemicals are classified into groups such as polyphenols, alkaloids, and flavonoids, all of which have demonstrated anticancer properties. Compared to conventional chemotherapy, phytochemicals generally exhibit better metabolism and biotransformation, making them both safer and potentially more effective. Plant-based therapies not only enhance treatment efficacy but also help mitigate treatment-related side effects.

Overview of the etiological factors contributing to HCC. The figure also highlights the mechanisms of medicinal plants in suppressing tumor growth and metastasis. EMT: epithelial-mesenchymal transition; FDL: fatty liver disease; HCC: hepatocellular carcinoma

Medicinal plants as anticancer agents

Numerous phytochemicals have significantly advanced cancer research and show promising therapeutic potential in HCC, primarily by modulating key molecular pathways involved in tumor growth, survival, and metastasis.

Alkaloids

Alkaloids are cyclic compounds containing one or more basic nitrogen atoms, making them unique and valuable in medicine. They are classified based on their chemical structure and the plant source from which they are derived. For instance, vinblastine, extracted from Vinca rosea (Apocynaceae family), is classified as an indole alkaloid. It, along with its analogue vincristine, exhibits anticancer activity by inhibiting microtubule polymerization [50]. Another class, proto-alkaloids, consists of plant metabolites derived from aromatic amino acids such as tryptophan, tyrosine, or phenylalanine. An example is colchicine, which is derived from Colchicum autumnale and inhibits cancer progression by disrupting the cytoskeleton of tumor cells, thereby reducing tumor burden in HCC [51]. Camptothecin, a pyrroloquinoline alkaloid extracted from the plant Camptotheca acuminata, exhibits antioxidant properties by decreasing the expression of the Nrf2 protein in an HCC mouse model [52]. Moreover, some alkaloids can modulate immune responses, enhancing the immune system’s ability to recognize and eliminate malignant cells.

Flavanoids

Flavonoids are secondary metabolites classified as polyphenolic compounds, abundantly present in fruits, vegetables, tea, and medicinal plants. They are characterized by aromatic rings with specific degrees of hydroxylation and various substituents that distinguish each flavonoid [53]. These compounds have attracted significant attention due to their ability to modulate signaling pathways involved in tumor development, progression, angiogenesis, and metastasis. Quercetin, found in high concentrations in various berries, onions, apples, and red wine, inhibits the proliferation of liver cancer cells by reducing inflammation and inducing cell cycle arrest [54]. Apigenin, extracted from Petroselinum crispum (parsley), demonstrates antimetastatic potential in an in vivo HCC model by altering miRNA expression and inducing apoptosis [55]. Luteolin, a flavonoid mainly found in broccoli, pepper, thyme, and celery, exhibits anticancer activity in vitro HCC models by modulating the expression of the p53 protein [56]. Genistein, derived from dyer’s broom (Genista tinctoria), reverses the EMT markers and inhibits cancer cells’ migration and invasion [57].

Terpenoids

Terpenoids are among the most diverse groups of phytochemicals, derived from terpenes composed of repeating isoprene units that form characteristic carbon skeletons. Based on the number of isoprene units, terpenes are classified into several groups, including monoterpenoids, sesquiterpenoids, diterpenoids, and triterpenoids [58]. Tanshinone I, a diterpenoid found in lavender, peppermint, cherries, celery seeds, and lemongrass, exhibits anticancer activity by inducing apoptosis and inhibiting p53-mediated autophagy in the HepG2 cell line model [59]. Parthenolide, a sesquiterpene lactone derived from Tanacetum parthenium (feverfew), exerts anticancer effects by modulating the immune system, reducing inflammation, and interfering with signaling pathways involved in the inflammatory response [60]. Ursolic acid, a triterpenoid found in apple peel, rosemary, thyme, oregano, and lavender, functions as an antioxidant and anti-inflammatory agent, and also reverses sorafenib resistance in HCC cells in an in vitro model [61]. Furthermore, saponins, a class of triterpenoids, exhibit anticancer properties by inducing apoptosis and blocking the β-catenin signaling pathway in the HCC mouse model [62]. Additionally, various terpenoids have been shown to inhibit angiogenesis, migration, and invasion of cancer cells, further highlighting their potential in cancer therapy.

Polyphenols

Polyphenols have shown strong potential to inhibit cancer progression in both in vitro and in vivo models. These compounds are classified based on their structure and function. For example, flavonoids, found in the root bark of plant species such as Ramulus mori (Moraceae family) and Sophora flavescens (Leguminosae family), act as antioxidants and anti-inflammatory agents, and inhibit the proliferation of HCC cells in vitro by modulating the ERK signaling pathway [63]. Stilbenes, another class of polyphenols found in red grapes and peanuts, exhibit anticancer and antimetastatic properties in vitro HCC models by inducing autophagy [64]. Phenolic acids, primarily present in fruits (especially berries), vegetables, cereals, legumes, and beverages such as coffee and wine, demonstrate anticancer activity by inducing apoptosis and modulating the TME in both in vivo and in vitro [65]. Lignans combat HCC through multiple mechanisms, including the induction of apoptosis and the inhibition of angiogenesis. Sesame and flax seeds are among the most concentrated dietary sources of lignans [66].

Key medicinal plants and phytochemicals with proven efficacy against HCC metastasis: mechanistic insights and preclinical evidence

Several medicinal plants have shown promising therapeutic potential against HCC through mechanisms including the induction of apoptosis, inhibition of metastasis, anti-inflammatory effects, and modulation of key oncogenic signaling pathways (Table 2). Below is an overview of key phytochemicals derived from selected medicinal plants that have shown efficacy in preclinical HCC metastasis models, highlighting their molecular targets and proposed modes of action.

 Key medicinal plants and phytochemicals with proven efficacy against HCC metastasis

S. No.Scientific nameCommon nameActive compoundMechanism of actionTherapeutic potentialReference
Apoptosis and cell cycle arrest
1Ferula assa-foetidaAsafoetida-devil’s dungFarnesiferol C↑ caspase activation results in increased apoptosisInduction of apoptosis[67]
2Petroselinum crispumParsleyApigeninInduction of cell cycle arrest and apoptosis, inhibiting the PI3K/Akt signaling pathways by overexpression of miR-199a/b-5pCell cycle arrest, apoptosis[55]
Activates anti-angiogenic factor
3Berberis vulgarisBarberryBerberineBlock the HIF-1α/VEGF axis, reduced expression of Id-1 by inactivation of p16INK4a/RB pathwayAnti-angiogenic[68]
4Solanum lycopersicumTomatoLycopene↓ expression of HIF-1α, VEGF, CD31, MMP-2, and MMP-9Suppression of neovascularization[69]
Metabolic reprogram
5Picrorhiza kurroaKutkiPicroside II↓ glycolytic enzyme expression activityMetabolic reprogramming[70]
6Silybum marianumMilk thistleSilybinActivating the AMPK-DR5 pathway inhibits intracellular ATP levels and glycolysis↓ ATP production and glycolysis[71]
Reduces tissue invasion
7Arctium lappaGreater burdockArctigeninWnt/β-catenin signaling, ↑ expression of E-cadherin, ↓ N-cadherin, and vimentin, prevent EMTEMT suppression[72]
8Camptotheca acuminataHappy treeCamptothecin↓ expression of Nrf2, ↑ expression of E-cadherin, and ↓ N-cadherinEMT inhibition[73]
9Curcuma longaTurmericCurcumin↓ expression of MMPs-2/9, VEGF,
inhibit the PI3K/Akt/mTOR/NF-κB signaling
EMT suppression, angiogenesis inhibition[74]
10Dioscorea zingiberensisZingiber yamDiosgeninInhibit platelet activation, inhibit P2Y2 receptor activity, and ↑ E-cadherin expressionEMT inhibition and reducing metastatic potential[75]
11Glycyrrhiza inflataChinese licoriceLicochalcone ADownregulation of the MKK4/JNK signaling pathway and NF-κB transcriptional activationEMT inhibition[76]
12Glycine maxSoybeanGenisteinInhibiting the EIF5A2/PI3K/Akt pathway, ↑ miR-1275, attenuate the EMT and stemnessEMT & stemness inhibition[77]
13Panax quinquefoliusGinsengGinsenosideInhibit the HIF-1α and NF-κB signaling pathway, preventing EMT by ↓ vimentin and ↑ E-cadherinEMT suppression[78]
14Plumbago zeylanicaChitrakPlumbagin↑ expression of E-cadherin and ↓ N-cadherin, vimentin, and snail, preventing EMTEMT suppression[79]
15Rosmarinus officinalisRosemaryRosmarinic acidInhibition of PI3K/Akt/mTOR signal pathwayAnti-invasion, anti-survival[80]
16Salvia miltiorrhizaDanshenTanshinone IIA↓ expression of Rho GTPases regulates the cytoskeleton remodelingEMT suppression[81]
17Scutellaria baicalensisChinese skullcapBaicalein↓ expression of MMP-2, MMP-9, ↑ expression of TIMP-1 and 2Anti-invasion[82]
18Thymus vulgarisThymeCarvacrolReduced the activity of argyrophilic nucleolar organizing regions, proliferating cell nuclear antigen, and MMPs-2/9Suppresses proliferation & metastasis[83]
19Withania somniferaAshwagandhaWithaferin A↓ expression of CD44, CD90, and EpCAM, inhibition of PI3K/Akt signaling pathway by ↑ miR-200cEMT suppression[84]
Induces autophagy
20Allium sativum L.GarlicAllicinInduced autophagyCell survival regulation[85]
21Rheum palmatumTurkish rhubarbEmodinInduced S and G2/M phase cell cycle arrest, induced autophagy, suppressed Wnt/β-catenin pathwaysInhibits proliferation and metastasis[86]
Activation of the immune system
22Andrographis paniculataKing of bittersAndrographolide↑ miR-22-3p expression level and ↓ HMGB1 and MMP-9 expression levelsImmunomodulation[87]
23Crocus sativusSaffron crocusCrocetin↑ expression of SHP-1, reduced activation STAT3, ↓ expression of MMP-9Inhibition of inflammation and invasion[88]
24Gentiana macrophyllaQinjiaoLuteoloside↓ ROS level, ↓ expression of NLRP3, ↓ secretion of IL-1βAnti-inflammatory response[89]
25Salvia miltiorrhizaDanshenCryptotanshinone↓ IDO1 enzyme activity modulates immunoregulationT cell modulation, immunoregulation[90]
26Terminalia belliricaBaheraTanninsModulating tumor immune microenvironment, restoration of CD8+ T cell infiltration and functionReversal of immune suppression in the tumor microenvironment[91]

EMT: epithelial-mesenchymal transition; HCC: hepatocellular carcinoma; HIF-1α: hypoxia inducible factor-1α; Id-1: inhibitor of differentiation-1; IDO1: indoleamine 2,3-dioxygenase 1; MMP-2: matrix metalloproteinase-2; RB: retinoblastoma; VEGF: vascular endothelial growth factor

Curcuma longa

Curcumin, a potent polyphenol extracted from Curcuma longa of the Zingiberaceae family, is widely found in the southeastern and southern regions of tropical Asia. It has been a key component of Ayurvedic and traditional Chinese medicine for centuries [92]. It has gained attention for its anti-proliferative, antimetastatic, and anti-inflammatory properties, as well as its ability to regulate multiple signaling pathways, making it a promising anticancer agent. A study reported that curcumin modulates the expression of cell adhesion markers—E-cadherin, N-cadherin, vimentin, and fibronectin, thereby regulating EMT through the TGF-β1 (transforming growth factor-β1) pathway and inhibiting HIF-1α in HepG2 cells, ultimately reducing tumor invasion and migration [93].

Curcumin acts on multiple molecular targets, including TGF-β, toll-like receptors (TLRs), and matrix metalloproteinases (MMPs), and also inhibits HCV replication, highlighting its antiviral potential [94, 95]. Curcumin inhibits cancer cell proliferation by mechanisms such as suppressing CDK2 activity in colon cancer [96]. In vitro studies have further demonstrated that curcumin induces cell cycle arrest at the S phase by downregulating cyclin A1 and promotes apoptosis by upregulating pro-apoptotic proteins, including Bax and caspase-3 [97]. It further suppresses cancer progression by blocking key signaling pathways such as NF-κB and Wnt, which are involved in cell proliferation and migration [98]. The combination of curcumin with metformin enhances therapeutic efficacy compared to curcumin alone by targeting the PI3K/Akt/mTOR/NF-κB and EGFR/STAT3 pathways, thereby suppressing angiogenesis and metastasis [74]. However, curcumin faces limitations such as poor absorption, rapid metabolism, and high clearance, with most of it excreted in unmetabolized sulfated or glucuronidated forms. A study using pH-sensitive nanoparticles co-loaded with doxorubicin and curcumin demonstrated improved drug delivery and enhanced inhibition of HCC proliferation [99].

Panax ginseng

The Araliaceae family, particularly the Panax genus, produces ginsenosides—also known as gintonin—a group of triterpenoid saponins derived from ginseng with notable anti-HCC properties [100]. Ginseng, primarily found in China and India, contains various ginsenoside subtypes, including ocotillol-type pseudo-ginsenosides, oleanane, protopanaxatriol (PPT), and protopanaxadiol (PPD). Among these, PPD significantly inhibits HCC cell migration and invasion. A recent study reported that PPD reduces STAT3 phosphorylation, thereby preventing its dimerization and nuclear translocation, leading to downregulation of TWIST1 expression. This effect reverses EMT in HCC by increasing E-cadherin levels and decreasing N-cadherin levels [101]. Another PPD derivative, ginsenoside Rh2, inhibits autophagy and reduces β-catenin levels in HCC cells in a dose-dependent manner [102]. A modified form, 20(S)-PPD, induces apoptosis by suppressing the PI3K/Akt signaling pathway, further inhibiting HCC proliferation [100, 103]. Additionally, ginsenoside Rg3 has been shown to reduce long non-coding RNA (lncRNA) expression and inhibit the PI3K/Akt pathway in vitro studies, resulting in reduced HCC cell migration [104].

Glycyrrhiza glabra

Glycyrrhiza glabra (liquorice), a medicinal plant from the Leguminosae family, is widely used in Ayurvedic medicine and primarily found in Central Asia and China [105]. Its root extract contains glycyrrhizin, a sweet-tasting tetracyclic triterpenoid saponin, commonly used as a flavouring agent in food and medicine. Liquorice is rich in bioactive compounds, including glycyrrhizin, glycyrrhetinic acid (GA), liquiritigenin, isoliquiritigenin, licochalcone A, licopyrano-coumarin, and glabrocoumarin. Among these, glycyrrhizin and GA have shown significant anticancer and antimetastatic potential. Glycyrrhizin is metabolized into GA by gut microbiota, which is responsible for its therapeutic effects [106]. GA (also known as enoxolone) exhibits anti-inflammatory, anticancer, and pro-apoptotic activities. It suppresses HCC proliferation by inhibiting the JNK1 pathway, which is associated with malignant transformation, and reduces the self-renewal capacity of HSCs [107]. In combination with sorafenib, GA enhances anticancer efficacy. It also inhibits cell migration and metastasis by targeting the EGFR, ERK, and Akt signaling pathways [108]. GA binds specifically to a receptor on the sinusoidal surface of hepatocytes (GA receptor), enabling its use in targeted drug delivery. For instance, GA has been conjugated with 5-fluorouracil (5-FU) via an alkyl side chain, resulting in increased ROS (reactive oxygen species) production and enhanced apoptosis [109]. Additionally, licochalcone A, a compound from Glycyrrhiza inflata, shows synergistic effects with sorafenib by blocking the MKK-4/JNK signaling pathway, thereby inhibiting HCC metastasis [76].

Picrorhiza kurroa

Picrorhiza kurroa (kutki) is a medicinal plant native to high-altitude regions (3,000–5,500 meters) of the Himalayas, India, Pakistan, Nepal, and Tibet. Belonging to the Plantaginaceae family, it includes two species: P. kurroa Royle ex Benth and P. scrophulariiflora Pennell [110, 111]. Valued in Ayurvedic medicine, the plant’s roots and rhizomes produce two primary crystalline compounds—picroline and kutkin—which contain various bioactive phytochemicals, including glycosides, iridoids, alkaloids, and terpenes. Among these, iridoids are the major class, featuring compounds such as picroside I–V, verminoside, catalpol, veronicoside, specioside, 6-feruloylcatalpol, pikuroside, and aucubin [112]. Picroside II has demonstrated antimetastatic and anti-angiogenic effects in both cellular and animal models [113]. It reduces inflammation by lowering TNF-α, IL-1β, and IL-6 levels and inhibiting NF-κB signaling in rat lung tissue [114]. In HCC models, picroside II targets glycosylphosphatidylinositol (GPI)-anchored signaling, suppressing cell proliferation and migration. However, further studies are needed to fully understand its role in inhibiting tumor cell migration [70].

Silybum marianum

Silymarin is a bioactive extract derived from Milk thistle (Silybum marianum), a medicinal plant known for its distinctive milky-white-veined leaves. It remains stable in acidic environments but degrades in alkaline conditions, making it effective in the acidic microenvironment of tumors. Silymarin comprises several active compounds, including silybin (or silibinin), isosilibinin, silydianin, silychristin, isosilychristin, and taxifolin. Among these, silybin is the most prominent for its anti-cancer properties, demonstrating antioxidant, anti-inflammatory, anti-proliferative, pro-apoptotic, antimetastatic, and anti-angiogenic effects [115]. Silybin shows enhanced therapeutic efficacy when combined with other agents. For example, in combination with doxorubicin, it induces cell cycle arrest at the G2-M phase by regulating the cdc25C-cyclin B1-cdc2 pathway and promotes apoptosis [116]. When paired with sorafenib, silibinin effectively suppresses the self-renewal capacity of cancer stem cells by downregulating stemness-related markers such as Nanog and Klf4 [117]. Additionally, silybin inhibits tumor progression in HCC by downregulating MMPs and modulating key signaling pathways, including the ERK1/2 cascade, Slit-2/Robo-1, and by suppressing PD-L1 expression, thereby enhancing T-cell activation [118, 119].

Allium sativum L.

Garlic (Allium sativum L.), a member of the Liliaceae family, is a herbaceous plant widely cultivated in India, Central Asia, and surrounding regions. Known for its pungent aroma and flavor, garlic is rich in sulfur-containing phytochemicals that contribute to its extensive nutritional, physiological, and medicinal benefits [120, 121]. Approximately 82% of garlic’s sulfur compounds include allicin, diallyl sulfide (DAS), diallyl trisulfide (DATS), E-/Z-ajoene, and S-allyl cysteine (SAC) sulfoxide. It also contains other organosulfur compounds such as SAC, N-acetylcysteine, and S-allyl mercapto cysteine (SAMC) [121, 122]. Among these, allicin (diallyl thiosulfinate) is the principal bioactive component with significant anticancer activity [123].

Although naturally derived from garlic, allicin is liposoluble and unstable post-synthesis, rapidly converting in vitro into secondary metabolites such as DADS (diallyl disulfide), DATS, SAC, SAMC, and γ-glutamyl-SAC (GSAC), which can also be synthesized artificially [122, 124]. Studies show that allicin induces apoptosis via the p38/MAPK signaling pathway in gastric carcinoma cells [125] and through p53-mediated autophagy in Hep3B cells [85]. Additionally, allicin suppresses telomerase activity, contributing to cancer cell apoptosis and senescence [126, 127]. It also inhibits the NF-κB pathway and modulates inflammatory cytokines by promoting pro-inflammatory signaling and inducing autophagy through TAMs in hepatoma cells [128130].

Allicin exhibits antimetastatic properties across various cancers. In gastric cancer, it inhibits cell migration by upregulating miR-383-5p and blocking ERBB4/PI3K/Akt signaling, while in cholangiocarcinoma, it suppresses MMP-2 and MMP-9 activity [131, 132]. In breast cancer, it enhances doxorubicin sensitivity by inhibiting the Nrf2/HO-1 pathway and promoting apoptosis [133, 134]. DATS, another allicin metabolite, also shows anticancer activity in HCC. In HepG2 cells, DATS induces apoptosis by activating the AMPK/SIRT1 pathway [135] and disrupts the ubiquitination of APPL1 via inhibition of TRAF6-mediated K63-linked polyubiquitination, leading to the inactivation of STAT3, Akt, and ERK1/2 pathways and suppression of tumor progression [136]. However, these findings are based on in vitro models, and further in vivo studies are necessary to validate DATS’s therapeutic potential in HCC.

Azadirachta indica

Azadirachta indica, commonly known as neem or “The Village Pharmacy”, belongs to the Meliaceae family. Valued in modern medicine as well as traditional systems such as Unani, Ayurveda, and Homeopathy, neem is recognized for its wide range of therapeutic properties. Its bioactive compounds fall into two major categories: isoprenoids and non-isoprenoids. Key constituents include azadirachtin, nimbolide, gedunin, nimbin, nimbidin, quercetin, and limonoids. These compounds contribute to disease prevention by reducing inflammation, inducing apoptosis, inhibiting angiogenesis, modulating immune responses, and providing antioxidant effects. Notably, neem exhibits significant anticancer activity by altering cancer cell behavior, making it a promising agent for cancer therapy [137].

Nimbolide, a potent limonoid derived from neem flowers and leaves, possesses diverse pharmacological properties including antimalarial, antibacterial, antiviral, antioxidant, anti-inflammatory, anti-invasive, neuroprotective, hepatoprotective, and pro-apoptotic effects. It enhances antioxidant defence by increasing the activity of enzymes such as glutathione (GSH) peroxidase, catalase, and superoxide dismutase (SOD) [138, 139]. Nimbolide also regulates lipid metabolism by modulating genes like liver X receptor-α (LXR-α), peroxisome proliferator-activated receptor-γ (PPARγ), and sterol regulatory element-binding protein-1c (SREBP1c) in hepatocytes [140]. In autoimmune hepatitis, nimbolide exerts anti-inflammatory effects by targeting histone deacetylase 3 (HDAC3), thereby reducing the expression of proinflammatory cytokines such as IL-1β, IL-6, and TNF-α [141]. In HCC, nimbolide enhances tight junction integrity by upregulating proteins such as ZO-1 and occludin, thereby preserving membrane polarity and reducing cell migration. Our previous study confirmed these effects, along with its anti-inflammatory properties [142]. However, further investigation is needed to fully elucidate nimbolide’s role in metastasis prevention in HCC.

Furthermore, in prostate cancer, nimbolide downregulates metastasis-associated genes like MMP-9 and ICAM1, reducing cell migration and invasion [143]. In oral cancer, it induces autophagy-dependent apoptosis by inhibiting the PI3K/Akt signaling pathway [144].

Nimbolide also reverses EMT markers in triple-negative breast cancer (TNBC) cells and inhibits the integrin-FAK (ITG-FAK) signaling pathway, affecting actin cytoskeleton remodeling and promoting apoptosis [145]. In pancreatic cancer, combining nimbolide with docetaxel (DTX) enhances DTX sensitivity, suppressing tumor growth and metastasis through inhibition of NF-κB signaling and promotion of apoptosis [146, 147].

Numerous medicinal plants exhibit promising antimetastatic effects in HCC through well-defined molecular mechanisms. Curcumin longa inhibits PI3K/Akt/mTOR/NF-κB pathway and downregulates MMPs-2/9 and VEGF, thereby impeding angiogenesis and invasion. Andrographis paniculata suppresses tumor progression by modulating HMGB1 and MMP-9 expression via upregulation of miR-22-3p. Camptotheca acuminata affects EMT markers by downregulating Nrf2 and N-cadherin while upregulating E-cadherin. Silybum marianum activates the AMPK-DR5 pathway, inhibiting glycolysis and inducing apoptosis. Collectively, these phytochemicals target critical oncogenic processes.

Challenges and future perspectives

Herbal medicine is one of the oldest forms of therapeutic intervention and has significantly influenced modern pharmaceutical development. Many medicinal plants, including those used in traditional Indian healthcare systems like Ayurveda, have been historically employed to treat various diseases. However, several challenges limit the effective use of herbal remedies in treating complex diseases such as cancer, including HCC.

A major limitation is the lack of sufficient clinical studies to validate the efficacy of plant-based anticancer agents, particularly in preventing cancer metastasis. Additionally, poor systemic absorption—such as low gastrointestinal uptake—significantly reduces the therapeutic potency of compounds like curcumin in clinical trials. Other contributing factors to low bioavailability include rapid metabolism and fast systemic elimination [99]. While herbal medicines generally have fewer side effects compared to synthetic drugs, their therapeutic application is hindered by the difficulty in identifying and standardizing active compounds due to the complex mixture of constituents in plant extracts. More research is necessary to elucidate the molecular mechanisms of these active ingredients and improve their pharmacokinetic properties [148].

Conclusions

This comprehensive review highlights the potential of plant-based bioactive compounds in targeting HCC through diverse cellular mechanisms. Natural agents such as nimbolide, curcumin, crocetin, allicin, picroside II, and ginsenosides have shown promising anti-cancer effects in both in vitro and in vivo studies. Notably, many of these compounds are commonly found in dietary sources, including spices and fruits. Preclinical and clinical findings suggest that these phytochemicals can enhance the efficacy of conventional anti-cancer therapies while reducing their side effects, offering promising alternative strategies for controlling HCC progression and metastasis. Further exploration of their molecular pathways may pave the way for the development of more effective and less toxic cancer treatments.

Abbreviations

BCLC: Barcelona Clinic Liver Cancer

DATS: diallyl trisulfide

DTX: docetaxel

ECM: extracellular matrix

EMT: epithelial-mesenchymal transition

GA: glycyrrhetinic acid

HBV: hepatitis B virus

HCC: hepatocellular carcinoma

HIF-1α: hypoxia inducible factor-1α

HSCs: hepatic stellate cells

LT: liver transplantation

MAFLD: metabolic dysfunction-associated fatty liver disease

MMPs: matrix metalloproteinases

MWA: microwave ablation

PD-L1: programmed death-ligand 1

PDGF: platelet-derived growth factor

PPD: protopanaxadiol

RFA: radiofrequency ablation

SAC: S-allyl cysteine

SAMC: S-allyl mercapto cysteine

SIRT: selective internal radiation therapy

TACE: transarterial chemoembolization

TAMs: tumor-associated macrophages

TGF-β1: transforming growth factor-β1

TME: tumor microenvironment

VEGFR: vascular endothelial growth factor receptor

Declarations

Acknowledgments

We also extend our utmost gratitude to Dr. Dinesh Kumar Meena, Research Scientist-II, St John’s Research Institute, Bangalore, for his invaluable insights and feedback on this manuscript.

Author contributions

MKN: Writing—original draft. DS and BV: Writing—review & editing.

Conflicts of interest

The authors declare that there are no conflicts of interest.

Ethical approval

This article is a review of previously published studies and does not involve human participants, animal subjects, or the collection of primary data; therefore, ethical approval was not required.

Consent to participate

Not applicable.

Consent to publication

Not applicable.

Availability of data and materials

This article is a review of previously published studies, and no new data were generated or analyzed in the course of this study. All data supporting the findings of this article are available in the cited references.

Funding

This work was supported by the JIPMER intramural research grant [JIP/Res/Intramural/Phase-4/2023-2024]. The funder had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Copyright

© The Author(s) 2025.

Publisher’s note

Open Exploration maintains a neutral stance on jurisdictional claims in published institutional affiliations and maps. All opinions expressed in this article are the personal views of the author(s) and do not represent the stance of the editorial team or the publisher.

References

Zhang C, Cheng Y, Zhang S, Fan J, Gao Q. Changing epidemiology of hepatocellular carcinoma in Asia. Liver Int. 2022;42:202941. [DOI] [PubMed]
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:20949. [DOI] [PubMed]
Rumgay H, Arnold M, Ferlay J, Lesi O, Cabasag CJ, Vignat J, et al. Global burden of primary liver cancer in 2020 and predictions to 2040. J Hepatol. 2022;77:1598606. [DOI] [PubMed] [PMC]
Acharya SK. Epidemiology of hepatocellular carcinoma in India. J Clin Exp Hepatol. 2014;4:S2733. [DOI] [PubMed] [PMC]
Baecker A, Liu X, La Vecchia C, Zhang ZF. Worldwide incidence of hepatocellular carcinoma cases attributable to major risk factors. Eur J Cancer Prev. 2018;27:20512. [DOI] [PubMed]
Guo A, Pomenti S, Wattacheril J. Health Disparities in Screening, Diagnosis, and Treatment of Hepatocellular Carcinoma. Clin Liver Dis (Hoboken). 2021;17:3538. [DOI] [PubMed] [PMC]
Xi D, Lin H, Shah AA. Overview of autoimmune liver disease: Prevalence, risk factors, and role of autoantibodies. Clin Liver Dis (Hoboken). 2022;20:1115. [DOI] [PubMed] [PMC]
Sneag DB, Krajewski K, Giardino A, O’Regan KN, Shinagare AB, Jagannathan JP, et al. Extrahepatic spread of hepatocellular carcinoma: spectrum of imaging findings. AJR Am J Roentgenol. 2011;197:W65864. [DOI] [PubMed]
Guglielmi A, Ruzzenente A, Conci S, Valdegamberi A, Vitali M, Bertuzzo F, et al. Hepatocellular carcinoma: surgical perspectives beyond the barcelona clinic liver cancer recommendations. World J Gastroenterol. 2014;20:752533. [DOI] [PubMed] [PMC]
Mähringer-Kunz A, Steinle V, Düber C, Weinmann A, Koch S, Schmidtmann I, et al. Extent of portal vein tumour thrombosis in patients with hepatocellular carcinoma: The more, the worse? Liver Int. 2019;39:32431. [DOI] [PubMed]
Celià-Terrassa T, Kang Y. How important is EMT for cancer metastasis? PLoS Biol. 2024;22:e3002487. [DOI] [PubMed] [PMC]
Gan W, Wang J, Zhu X, He X, Guo P, Zhang S, et al. RARγ-induced E-cadherin downregulation promotes hepatocellular carcinoma invasion and metastasis. J Exp Clin Cancer Res. 2016;35:164. [DOI] [PubMed] [PMC]
Zhou W, Gong L, Wu Q, Xing C, Wei B, Chen T, et al. PHF8 upregulation contributes to autophagic degradation of E-cadherin, epithelial-mesenchymal transition and metastasis in hepatocellular carcinoma. J Exp Clin Cancer Res. 2018;37:215. Erratum in: J Exp Clin Cancer Res. 2019;38:445. [DOI] [PubMed] [PMC]
Han C, He S, Wang R, Gao X, Wang H, Qiao J, et al. The role of ARHGAP9: clinical implication and potential function in acute myeloid leukemia. J Transl Med. 2021;19:65. [DOI] [PubMed] [PMC]
Cheng W, Cheng Z, Weng L, Xing D, Zhang M. Asparagus Polysaccharide inhibits the Hypoxia-induced migration, invasion and angiogenesis of Hepatocellular Carcinoma Cells partly through regulating HIF1α/VEGF expression via MAPK and PI3K signaling pathway. J Cancer. 2021;12:39209. [DOI] [PubMed] [PMC]
Lacin S, Yalcin S. The Prognostic Value of Circulating VEGF-A Level in Patients With Hepatocellular Cancer. Technol Cancer Res Treat. 2020;19:1533033820971677. [DOI] [PubMed] [PMC]
Chen C, Wu S, Lin Y, Chi H, Lin S, Yeh C, et al. Induction of nuclear protein-1 by thyroid hormone enhances platelet-derived growth factor A mediated angiogenesis in liver cancer. Theranostics. 2019;9:236179. [DOI] [PubMed] [PMC]
Wang Y, Liu D, Zhang T, Xia L. FGF/FGFR Signaling in Hepatocellular Carcinoma: From Carcinogenesis to Recent Therapeutic Intervention. Cancers (Basel). 2021;13:1360. [DOI] [PubMed] [PMC]
Jeng K, Sheen I, Lin S, Leu C, Chang C. The Role of Endoglin in Hepatocellular Carcinoma. Int J Mol Sci. 2021;22:3208. [DOI] [PubMed] [PMC]
Huang H, Zhang J, Ling F, Huang Y, Yang M, Zhang Y, et al. Leptin Receptor (LEPR) promotes proliferation, migration, and invasion and inhibits apoptosis in hepatocellular carcinoma by regulating ANXA7. Cancer Cell Int. 2021;21:4. [DOI] [PubMed] [PMC]
Li Y, Turpin CP, Wang S. Role of thrombospondin 1 in liver diseases. Hepatol Res. 2017;47:18693. [DOI] [PubMed] [PMC]
Ji Y, Fan H, Yang M, Bai C, Yang W, Wang Z. Synergistic Effect of Baculovirus-Mediated Endostatin and Angiostatin Combined with Gemcitabine in Hepatocellular Carcinoma. Biol Pharm Bull. 2022;45:30915. [DOI] [PubMed]
Yao C, Wu S, Kong J, Sun Y, Bai Y, Zhu R, et al. Angiogenesis in hepatocellular carcinoma: mechanisms and anti-angiogenic therapies. Cancer Biol Med. 2023;20:2543. [DOI] [PubMed] [PMC]
Zhu B, Lin N, Zhang M, Zhu Y, Cheng H, Chen S, et al. Activated hepatic stellate cells promote angiogenesis via interleukin-8 in hepatocellular carcinoma. J Transl Med. 2015;13:365. [DOI] [PubMed] [PMC]
Xi S, Zheng X, Li X, Jiang Y, Wu Y, Gong J, et al. Activated Hepatic Stellate Cells Induce Infiltration and Formation of CD163+ Macrophages via CCL2/CCR2 Pathway. Front Med (Lausanne). 2021;8:627927. [DOI] [PubMed] [PMC]
Omata M, Cheng A, Kokudo N, Kudo M, Lee JM, Jia J, et al. Asia-Pacific clinical practice guidelines on the management of hepatocellular carcinoma: a 2017 update. Hepatol Int. 2017;11:31770. [DOI] [PubMed] [PMC]
European Association for the Study of the Liver. EASL Clinical Practice Guidelines: Management of hepatocellular carcinoma. J Hepatol. 2018;69:182236. Erratum in: J Hepatol. 2019;70:817. [DOI] [PubMed]
Vairappan B, Wright G, Ravikumar TS. Incidence, diagnosis, and management of hepatocellular carcinoma: current perspectives and future direction. J Dig Dis Hepatol. 2023;8:188. [DOI]
Ishizawa T, Hasegawa K, Aoki T, Takahashi M, Inoue Y, Sano K, et al. Neither multiple tumors nor portal hypertension are surgical contraindications for hepatocellular carcinoma. Gastroenterology. 2008;134:190816. [DOI] [PubMed]
Bruix J, Takayama T, Mazzaferro V, Chau G, Yang J, Kudo M, et al. Adjuvant sorafenib for hepatocellular carcinoma after resection or ablation (STORM): a phase 3, randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2015;16:134454. [DOI] [PubMed]
Saliba F, Duvoux C, Dharancy S, Dumortier J, Calmus Y, Gugenheim J, et al. Five-year outcomes in liver transplant patients receiving everolimus with or without a calcineurin inhibitor: Results from the CERTITUDE study. Liver Int. 2022;42:251323. [DOI] [PubMed] [PMC]
Kneteman NM, Oberholzer J, Saghier MA, Meeberg GA, Blitz M, Ma MM, et al. Sirolimus-based immunosuppression for liver transplantation in the presence of extended criteria for hepatocellular carcinoma. Liver Transpl. 2004;10:130111. [DOI] [PubMed]
Burrel M, Llovet JM, Ayuso C, Iglesias C, Sala M, Miquel R, et al.; Barcelona Clínic Liver Cancer Group. MRI angiography is superior to helical CT for detection of HCC prior to liver transplantation: an explant correlation. Hepatology. 2003;38:103442. [DOI] [PubMed]
Lencioni R, de Baere T, Soulen MC, Rilling WS, Geschwind JH. Lipiodol transarterial chemoembolization for hepatocellular carcinoma: A systematic review of efficacy and safety data. Hepatology. 2016;64:10616. [DOI] [PubMed]
Salem R, Gordon AC, Mouli S, Hickey R, Kallini J, Gabr A, et al. Y90 Radioembolization Significantly Prolongs Time to Progression Compared With Chemoembolization in Patients With Hepatocellular Carcinoma. Gastroenterology. 2016;151:115563.e2. [DOI] [PubMed] [PMC]
Cheng A, Kang Y, Chen Z, Tsao C, Qin S, Kim JS, et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2009;10:2534. [DOI] [PubMed]
Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc J, et al.; SHARP Investigators Study Group. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:37890. [DOI] [PubMed]
Bruix J, Qin S, Merle P, Granito A, Huang Y, Bodoky G, et al.; RESORCE Investigators. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;389:5666. Erratum in: Lancet. 2017;389:36. [DOI] [PubMed]
Granito A, Forgione A, Marinelli S, Renzulli M, Ielasi L, Sansone V, et al. Experience with regorafenib in the treatment of hepatocellular carcinoma. Therap Adv Gastroenterol. 2021;14:17562848211016959. [DOI] [PubMed] [PMC]
Cheng A, Qin S, Ikeda M, Galle PR, Ducreux M, Kim T, et al. Updated efficacy and safety data from IMbrave150: Atezolizumab plus bevacizumab vs. sorafenib for unresectable hepatocellular carcinoma. J Hepatol. 2022;76:86273. [DOI] [PubMed]
Zanuso V, Pirozzi A, Balsano R, Pressiani T, Rimassa L. Safety and Efficacy of Atezolizumab and Bevacizumab Combination as a First Line Treatment of Advanced Hepatocellular Carcinoma. J Hepatocell Carcinoma. 2023;10:1689708. [DOI] [PubMed] [PMC]
Qin S, Bi F, Gu S, Bai Y, Chen Z, Wang Z, et al. Donafenib Versus Sorafenib in First-Line Treatment of Unresectable or Metastatic Hepatocellular Carcinoma: A Randomized, Open-Label, Parallel-Controlled Phase II-III Trial. J Clin Oncol. 2021;39:300211. [DOI] [PubMed] [PMC]
Kudo M, Finn RS, Qin S, Han K, Ikeda K, Piscaglia F, et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial. Lancet. 2018;391:116373. [DOI] [PubMed]
Abou-Alfa GK, Meyer T, Cheng A, El-Khoueiry AB, Rimassa L, Ryoo B, et al. Cabozantinib in Patients with Advanced and Progressing Hepatocellular Carcinoma. N Engl J Med. 2018;379:5463. [DOI] [PubMed] [PMC]
Zhu AX, Kang Y, Yen C, Finn RS, Galle PR, Llovet JM, et al. Ramucirumab after sorafenib in patients with advanced hepatocellular carcinoma and increased α-fetoprotein concentrations (REACH-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2019;20:28296. [DOI] [PubMed]
Qin S, Li Q, Gu S, Chen X, Lin L, Wang Z, et al. Apatinib as second-line or later therapy in patients with advanced hepatocellular carcinoma (AHELP): a multicentre, double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Gastroenterol Hepatol. 2021;6:55968. [DOI] [PubMed]
Qin S, Cheng Y, Liang J, Shen L, Bai Y, Li J, et al. Efficacy and safety of the FOLFOX4 regimen versus doxorubicin in Chinese patients with advanced hepatocellular carcinoma: a subgroup analysis of the EACH study. Oncologist. 2014;19:116978. [DOI] [PubMed] [PMC]
Yau T, Galle PR, Decaens T, Sangro B, Qin S, da Fonseca LG, et al. Nivolumab plus ipilimumab versus lenvatinib or sorafenib as first-line treatment for unresectable hepatocellular carcinoma (CheckMate 9DW): an open-label, randomised, phase 3 trial. Lancet. 2025;405:185164. [DOI] [PubMed]
Song X, Kelley RK, Khan AA, Standifer N, Zhou D, Lim K, et al. Exposure-Response Analyses of Tremelimumab Monotherapy or in Combination with Durvalumab in Patients with Unresectable Hepatocellular Carcinoma. Clin Cancer Res. 2023;29:75463. [DOI] [PubMed] [PMC]
Dhyani P, Quispe C, Sharma E, Bahukhandi A, Sati P, Attri DC, et al. Anticancer potential of alkaloids: a key emphasis to colchicine, vinblastine, vincristine, vindesine, vinorelbine and vincamine. Cancer Cell Int. 2022;22:206. [DOI] [PubMed] [PMC]
Zhang H, Su X, Gu L, Tan M, Liu Y, Xu K, et al. Colchicine-mediated selective autophagic degradation of HBV core proteins inhibits HBV replication and HBV-related hepatocellular carcinoma progression. Cell Death Discov. 2024;10:352. [DOI] [PubMed] [PMC]
Wang H, Gao C, Li X, Chen F, Li G. Author Correction: Camptothecin enhances the anti-tumor effect of low-dose apatinib combined with PD-1 inhibitor on hepatocellular carcinoma. Sci Rep. 2024;14:8787. [DOI] [PubMed] [PMC]
Mir SA, Dar A, Hamid L, Nisar N, Malik JA, Ali T, et al. Flavonoids as promising molecules in the cancer therapy: An insight. Curr Res Pharmacol Drug Discov. 2023;6:100167. [DOI] [PubMed] [PMC]
Ren K, Li Y, Wu G, Ren J, Lu H, Li Z, et al. Quercetin nanoparticles display antitumor activity via proliferation inhibition and apoptosis induction in liver cancer cells. Int J Oncol. 2017;50:1299311. [DOI] [PubMed]
Wang S, Yang P, Feng X, Zhu Y, Qiu F, Hu X, et al. Apigenin Inhibits the Growth of Hepatocellular Carcinoma Cells by Affecting the Expression of microRNA Transcriptome. Front Oncol. 2021;11:657665. [DOI] [PubMed] [PMC]
Lee Y, Kwon YH. Regulation of apoptosis and autophagy by luteolin in human hepatocellular cancer Hep3B cells. Biochem Biophys Res Commun. 2019;517:61722. [DOI] [PubMed]
Dai W, Wang F, He L, Lin C, Wu S, Chen P, et al. Genistein inhibits hepatocellular carcinoma cell migration by reversing the epithelial-mesenchymal transition: partial mediation by the transcription factor NFAT1. Mol Carcinog. 2015;54:30111. [DOI] [PubMed]
Tholl D. Terpene synthases and the regulation, diversity and biological roles of terpene metabolism. Curr Opin Plant Biol. 2006;9:297304. [DOI] [PubMed]
Liu X, Liu J. Tanshinone I induces cell apoptosis by reactive oxygen species-mediated endoplasmic reticulum stress and by suppressing p53/DRAM-mediated autophagy in human hepatocellular carcinoma. Artif Cells Nanomed Biotechnol. 2020;48:48897. [DOI] [PubMed]
Liu X, Gao Z, Wang X, Shen Y. Parthenolide targets NF-κB (P50) to inhibit HIF-1α-mediated metabolic reprogramming of HCC. Aging (Albany NY). 2022;14:834656. [DOI] [PubMed] [PMC]
Fan Y, Pan F, Cui Z, Zheng H. The Antitumor and Sorafenib-resistant Reversal Effects of Ursolic Acid on Hepatocellular Carcinoma via Targeting ING5. Int J Biol Sci. 2024;20:4190208. [DOI] [PubMed] [PMC]
Cui X, Jiang X, Wei C, Xing Y, Tong G. Astragaloside IV suppresses development of hepatocellular carcinoma by regulating miR-150-5p/β-catenin axis. Environ Toxicol Pharmacol. 2020;78:103397. [DOI] [PubMed]
Liang R, Zhang S, Qi J, Wang Z, Li J, Liu P, et al. Preferential inhibition of hepatocellular carcinoma by the flavonoid Baicalein through blocking MEK-ERK signaling. Int J Oncol. 2012;41:96978. [DOI] [PubMed]
Sun M, Bai J, Wang H, Zhou L, Li S. The 3,3'-dimethoxy-4,4'-dihydroxy-stilbene Triazole (STT) Inhibits Liver Cancer Cell Growth by Targeting Akt/mTOR Pathway. Dokl Biochem Biophys. 2024;517:27784. [DOI] [PubMed]
Shaban NZ, Hegazy WA, Abu-Serie MM, Talaat IM, Awad OM, Habashy NH. Seedless black Vitis vinifera polyphenols suppress hepatocellular carcinoma in vitro and in vivo by targeting apoptosis, cancer stem cells, and proliferation. Biomed Pharmacother. 2024;175:116638. [DOI] [PubMed]
Alshehri SA, Almarwani WA, Albalawi AZ, Al-Atwi SM, Aljohani KK, Alanazi AA, et al. Role of Arctiin in Fibrosis and Apoptosis in Experimentally Induced Hepatocellular Carcinoma in Rats. Cureus. 2024;16:e51997. [DOI] [PubMed] [PMC]
Alafnan A, Alamri A, Alanazi J, Hussain T. Farnesiferol C Exerts Antiproliferative Effects on Hepatocellular Carcinoma HepG2 Cells by Instigating ROS-Dependent Apoptotic Pathway. Pharmaceuticals (Basel). 2022;15:1070. [DOI] [PubMed] [PMC]
Tsang CM, Cheung KCP, Cheung YC, Man K, Lui VW, Tsao SW, et al. Berberine suppresses Id-1 expression and inhibits the growth and development of lung metastases in hepatocellular carcinoma. Biochim Biophys Acta. 2015;1852:54151. [DOI] [PubMed]
Bhatia N, Gupta P, Singh B, Koul A. Lycopene Enriched Tomato Extract Inhibits Hypoxia, Angiogenesis, and Metastatic Markers in early Stage N-Nitrosodiethylamine Induced Hepatocellular Carcinoma. Nutr Cancer. 2015;67:126875. [DOI] [PubMed]
Lei X, Yang S, Peng K, Wang M, Wu H, Yang Y. Picroside Ⅱ Inhibits Glycolysis and Migration in HepG2 Cells. Pharmacogn Mag. 2024;21:192206. [DOI]
Xiao B, Jiang Y, Yuan S, Cai L, Xu T, Jia L. Silibinin, a potential fasting mimetic, inhibits hepatocellular carcinoma by triggering extrinsic apoptosis. MedComm (2020). 2024;5:e457. [DOI] [PubMed] [PMC]
Lu Z, Chang L, Zhou H, Liu X, Li Y, Mi T, et al. Arctigenin Attenuates Tumor Metastasis Through Inhibiting Epithelial-Mesenchymal Transition in Hepatocellular Carcinoma via Suppressing GSK3β-Dependent Wnt/β-Catenin Signaling Pathway In Vivo and In Vitro. Front Pharmacol. 2019;10:937. [DOI] [PubMed] [PMC]
Liu Q, Zhao S, Meng F, Wang H, Sun L, Li G, et al. Nrf2 Down-Regulation by Camptothecin Favors Inhibiting Invasion, Metastasis and Angiogenesis in Hepatocellular Carcinoma. Front Oncol. 2021;11:661157. [DOI] [PubMed] [PMC]
Zhang H, Zhang Y, Cheng Y, Gong F, Cao Z, Yu L, et al. Metformin incombination with curcumin inhibits the growth, metastasis, and angiogenesis of hepatocellular carcinoma in vitro and in vivo. Mol Carcinog. 2018;57:4456. [DOI] [PubMed]
Zhuang M, Xin G, Wei Z, Li S, Xing Z, Ji C, et al. Dihydrodiosgenin inhibits endothelial cell-derived factor VIII and platelet-mediated hepatocellular carcinoma metastasis. Cancer Manag Res. 2019;11:487182. [DOI] [PubMed] [PMC]
Tsai J, Hsiao P, Yang S, Hsieh S, Bau D, Ling C, et al. Licochalcone A suppresses migration and invasion of human hepatocellular carcinoma cells through downregulation of MKK4/JNK via NF-κB mediated urokinase plasminogen activator expression. PLoS One. 2014;9:e86537. [DOI] [PubMed] [PMC]
Yang X, Jiang W, Kong X, Zhou X, Zhu D, Kong L. Genistein Restricts the Epithelial Mesenchymal Transformation (EMT) and Stemness of Hepatocellular Carcinoma via Upregulating miR-1275 to Inhibit the EIF5A2/PI3K/Akt Pathway. Biology (Basel). 2022;11:1383. [DOI] [PubMed] [PMC]
Zhang J, Ma X, Fan D. Ginsenoside CK Inhibits Hypoxia-Induced Epithelial-Mesenchymal Transformation through the HIF-1α/NF-κB Feedback Pathway in Hepatocellular Carcinoma. Foods. 2021;10:1195. [DOI] [PubMed] [PMC]
Du Y, Yuan B, Ye Y, Zhou F, Liu H, Huang J, et al. Plumbagin Regulates Snail to Inhibit Hepatocellular Carcinoma Epithelial-Mesenchymal Transition in vivo and in vitro. J Hepatocell Carcinoma. 2024;11:56580. [DOI] [PubMed] [PMC]
Wang L, Yang H, Wang C, Shi X, Li K. Rosmarinic acid inhibits proliferation and invasion of hepatocellular carcinoma cells SMMC 7721 via PI3K/AKT/mTOR signal pathway. Biomed Pharmacother. 2019;120:109443. [DOI] [PubMed]
Liang E, Huang M, Chen Y, Zhang P, Shen Y, Tu X, et al. Tanshinone IIA modulates cancer cell morphology and movement via Rho GTPases-mediated actin cytoskeleton remodeling. Toxicol Appl Pharmacol. 2024;483:116839. [DOI] [PubMed]
Chen K, Zhang S, Ji Y, Li J, An P, Ren H, et al. Baicalein inhibits the invasion and metastatic capabilities of hepatocellular carcinoma cells via down-regulation of the ERK pathway. PLoS One. 2013;8:e72927. [DOI] [PubMed] [PMC]
Subramaniyan J, Krishnan G, Balan R, Mgj D, Ramasamy E, Ramalingam S, et al. Carvacrol modulates instability of xenobiotic metabolizing enzymes and downregulates the expressions of PCNA, MMP-2, and MMP-9 during diethylnitrosamine-induced hepatocarcinogenesis in rats. Mol Cell Biochem. 2014;395:6576. [DOI] [PubMed]
Tian H. Withaferin a attenuates epithelial-mesenchymal transition and cancer stem cells properties in hepatocellular carcinoma cells by inhibiting the PI3K/AKT pathway through miR-200c. Pharmacogn Mag. 2022;18:11905. [DOI]
Chu Y, Ho C, Chung J, Raghu R, Lo Y, Sheen L. Allicin induces anti-human liver cancer cells through the p53 gene modulating apoptosis and autophagy. J Agric Food Chem. 2013;61:983948. [DOI] [PubMed]
Qin B, Zeng Z, Xu J, Shangwen J, Ye ZJ, Wang S, et al. Emodin inhibits invasion and migration of hepatocellular carcinoma cells via regulating autophagy-mediated degradation of snail and β-catenin. BMC Cancer. 2022;22:671. [DOI] [PubMed] [PMC]
Luo Y, Hu J, Jiao Y, Liu L, Miao D, Song Y, et al. Andrographolide anti-proliferation and metastasis of hepatocellular carcinoma through LncRNA MIR22HG regulation. J Nat Med. 2024;78:12345. [DOI] [PubMed]
Mohan CD, Kim C, Siveen KS, Manu KA, Rangappa S, Chinnathambi A, et al. Crocetin imparts antiproliferative activity via inhibiting STAT3 signaling in hepatocellular carcinoma. IUBMB Life. 2021;73:134862. [DOI] [PubMed]
Fan S, Wang Y, Lu J, Zheng Y, Wu D, Li M, et al. Luteoloside suppresses proliferation and metastasis of hepatocellular carcinoma cells by inhibition of NLRP3 inflammasome. PLoS One. 2014;9:e89961. [DOI] [PubMed] [PMC]
Yang X, She X, Zhao Z, Ren J, Wang P, Dong H, et al. In vitro and vivo anti-tumor activity and mechanisms of the new cryptotanshinone derivative 11 against hepatocellular carcinoma. Eur J Pharmacol. 2024;971:176522. [DOI] [PubMed]
Chang Z, Zhang Q, Hu Q, Liu Y, Zhang L, Liu R. Tannins in Terminalia bellirica inhibits hepatocellular carcinoma growth via re-educating tumor-associated macrophages and restoring CD8+T cell function. Biomed Pharmacother. 2022;154:113543. [DOI] [PubMed]
PubChem Compound Summary for CID 969516, Curcumin [Internet]. PubChem; [cited 2024 Nov 29]. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/Curcumin
Duan W, Chang Y, Li R, Xu Q, Lei J, Yin C, et al. Curcumin inhibits hypoxia inducible factor-1α-induced epithelial-mesenchymal transition in HepG2 hepatocellular carcinoma cells. Mol Med Rep. 2014;10:250510. [DOI] [PubMed]
Aggarwal BB, Sundaram C, Malani N, Ichikawa H. Curcumin: the Indian solid gold. In: Aggarwal BB, Surh YJ, Shishodia S, editors. The Molecular Targets and Therapeutic Uses of Curcumin in Health and Disease. Boston: Springer; 2007. pp. 1–75. [DOI] [PubMed]
Wei ZQ, Zhang YH, Ke CZ, Chen HX, Ren P, He YL, et al. Curcumin inhibits hepatitis B virus infection by down-regulating cccDNA-bound histone acetylation. World J Gastroenterol. 2017;23:625260. [DOI] [PubMed] [PMC]
Lim T, Lee S, Huang Z, Lim DY, Chen H, Jung SK, et al. Curcumin suppresses proliferation of colon cancer cells by targeting CDK2. Cancer Prev Res (Phila). 2014;7:46674. [DOI] [PubMed] [PMC]
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. [DOI] [PubMed] [PMC]
Xu MX, Zhao L, Deng C, Yang L, Wang Y, Guo T, et al. Curcumin suppresses proliferation and induces apoptosis of human hepatocellular carcinoma cells via the wnt signaling pathway. Int J Oncol. 2013;43:19519. [DOI] [PubMed]
Zhang J, Li J, Shi Z, Yang Y, Xie X, Lee SM, et al. pH-sensitive polymeric nanoparticles for co-delivery of doxorubicin and curcumin to treat cancer via enhanced pro-apoptotic and anti-angiogenic activities. Acta Biomater. 2017;58:34964. [DOI] [PubMed]
Kim JH, Yi Y, Kim M, Cho JY. Role of ginsenosides, the main active components of Panax ginseng, in inflammatory responses and diseases. J Ginseng Res. 2017;41:43543. [DOI] [PubMed] [PMC]
Lu Z, Xu H, Yu X, Wang Y, Huang L, Jin X, et al. 20(S)-Protopanaxadiol induces apoptosis in human hepatoblastoma HepG2 cells by downregulating the protein kinase B signaling pathway. Exp Ther Med. 2018;15:127784. [DOI] [PubMed] [PMC]
Yang Z, Zhao T, Liu H, Zhang L. Ginsenoside Rh2 inhibits hepatocellular carcinoma through β-catenin and autophagy. Sci Rep. 2016;6:19383. [DOI] [PubMed] [PMC]
Yang L, Zhang X, Li K, Li A, Yang W, Yang R, et al. Protopanaxadiol inhibits epithelial-mesenchymal transition of hepatocellular carcinoma by targeting STAT3 pathway. Cell Death Dis. 2019;10:630. [DOI] [PubMed] [PMC]
Pu Z, Ge F, Wang Y, Jiang Z, Zhu S, Qin S, et al. Ginsenoside-Rg3 inhibits the proliferation and invasion of hepatoma carcinoma cells via regulating long non-coding RNA HOX antisense intergenic. Bioengineered. 2021;12:2398409. [DOI] [PubMed] [PMC]
Zhang Y, Sheng Z, Xiao J, Li Y, Huang J, Jia J, et al. Advances in the roles of glycyrrhizic acid in cancer therapy. Front Pharmacol. 2023;14:1265172. Erratum in: Front Pharmacol. 2023;14:1345663. [DOI] [PubMed] [PMC]
Mittal A, Nagpal M, Vashistha VK. Recent Advances in the Pharmacological Activities of Glycyrrhizin, Glycyrrhetinic Acid, and Their Analogs. Rev Bras Farmacogn. 2023;33:115469. [DOI]
Cai S, Bi Z, Bai Y, Zhang H, Zhai D, Xiao C, et al. Glycyrrhizic Acid-Induced Differentiation Repressed Stemness in Hepatocellular Carcinoma by Targeting c-Jun N-Terminal Kinase 1. Front Oncol. 2020;9:1431. [DOI] [PubMed] [PMC]
Tsai J, Pan P, Hsu F, Chung J, Chiang I. Glycyrrhizic Acid Modulates Apoptosis through Extrinsic/Intrinsic Pathways and Inhibits Protein Kinase B- and Extracellular Signal-Regulated Kinase-Mediated Metastatic Potential in Hepatocellular Carcinoma In Vitro and In Vivo. Am J Chin Med. 2020;48:22344. [DOI] [PubMed]
Speciale A, Muscarà C, Molonia MS, Cristani M, Cimino F, Saija A. Recent Advances in Glycyrrhetinic Acid-Functionalized Biomaterials for Liver Cancer-Targeting Therapy. Molecules. 2022;27:1775. [DOI] [PubMed] [PMC]
Almeleebia TM, Alsayari A, Wahab S. Pharmacological and Clinical Efficacy of Picrorhiza kurroa and Its Secondary Metabolites: A Comprehensive Review. Molecules. 2022;27:8316. [DOI] [PubMed] [PMC]
Bhatnagar A. A review on chemical constituents and biological activities of the genus picrorhiza (scrophulariace). Int J Curr Pharm Res. 2021;13:1827. [DOI]
Lou C, Zhu Z, Xu X, Zhu R, Sheng Y, Zhao H. Picroside II, an iridoid glycoside from Picrorhiza kurroa, suppresses tumor migration, invasion, and angiogenesis in vitro and in vivo. Biomed Pharmacother. 2019;120:109494. [DOI] [PubMed]
Shen B, Zhao C, Chen C, Li Z, Li Y, Tian Y, et al. Picroside II Protects Rat Lung and A549 Cell Against LPS-Induced Inflammation by the NF-κB Pathway. Inflammation. 2017;40:75261. [DOI] [PubMed]
Ma C, Shi A. Picroside II prevents inflammation injury in mice with diabetic nephropathy via TLR4/NF-κB pathway. Qual Assur Saf Crop. 2021;13:3843. [DOI]
Bijak M. Silybin, a Major Bioactive Component of Milk Thistle (Silybum marianum L. Gaernt.)-Chemistry, Bioavailability, and Metabolism. Molecules. 2017;22:1942. [DOI] [PubMed] [PMC]
Ray PP, Islam MA, Islam MS, Han A, Geng P, Aziz MA, et al. A comprehensive evaluation of the therapeutic potential of silibinin: a ray of hope in cancer treatment. Front Pharmacol. 2024;15:1349745. [DOI] [PubMed] [PMC]
Li W, Wang H. Inhibitory effects of Silibinin combined with doxorubicin in hepatocellular carcinoma; an in vivo study. J BUON. 2016;21:91724. [PubMed]
Gu HR, Park SC, Choi SJ, Lee JC, Kim YC, Han CJ, et al. Combined treatment with silibinin and either sorafenib or gefitinib enhances their growth-inhibiting effects in hepatocellular carcinoma cells. Clin Mol Hepatol. 2015;21:4959. [DOI] [PubMed] [PMC]
Koltai T, Fliegel L. Role of Silymarin in Cancer Treatment: Facts, Hypotheses, and Questions. J Evid Based Integr Med. 2022;27:2515690X211068826. [DOI] [PubMed] [PMC]
Sharifi-Rad J, Cristina Cirone Silva N, Jantwal A, Bhatt DI, Sharopov F, Cho WC, et al. Therapeutic Potential of Allicin-Rich Garlic Preparations: Emphasis on Clinical Evidence toward Upcoming Drugs Formulation. Appl Sci. 2019;9:5555. [DOI]
Talib WH, Baban MM, Azzam AO, Issa JJ, Ali AY, AlSuwais AK, et al. Allicin and Cancer Hallmarks. Molecules. 2024;29:1320. [DOI] [PubMed] [PMC]
Talib WH, Atawneh S, Shakhatreh AN, Shakhatreh GN, Rasheed aljarrah IS, Hamed RA, et al. Anticancer potential of garlic bioactive constituents: Allicin, Z-ajoene, and organosulfur compounds. Pharmacia. 2024;71:123. [DOI]
Antony ML, Singh SV. Molecular mechanisms and targets of cancer chemoprevention by garlic-derived bioactive compound diallyl trisulfide. Indian J Exp Biol. 2011;49:80516. [PubMed] [PMC]
Zhou Y, Li X, Luo W, Zhu J, Zhao J, Wang M, et al. Allicin in Digestive System Cancer: From Biological Effects to Clinical Treatment. Front Pharmacol. 2022;13:903259. [DOI] [PubMed] [PMC]
Zhang X, Zhu Y, Duan W, Feng C, He X. Allicin induces apoptosis of the MGC-803 human gastric carcinoma cell line through the p38 mitogen-activated protein kinase/caspase-3 signaling pathway. Mol Med Rep. 2015;11:275560. [DOI] [PubMed]
Guterres AN, Villanueva J. Targeting telomerase for cancer therapy. Oncogene. 2020;39:581124. [DOI] [PubMed] [PMC]
Sun L, Wang X. Effects of allicin on both telomerase activity and apoptosis in gastric cancer SGC-7901 cells. World J Gastroenterol. 2003;9:19304. [DOI] [PubMed] [PMC]
Arreola R, Quintero-Fabián S, López-Roa RI, Flores-Gutiérrez EO, Reyes-Grajeda JP, Carrera-Quintanar L, et al. Immunomodulation and anti-inflammatory effects of garlic compounds. J Immunol Res. 2015;2015:401630. [DOI] [PubMed] [PMC]
Gao Z, Li X, Feng S, Chen J, Song K, Shi Y, et al. Autophagy suppression facilitates macrophage M2 polarization via increased instability of NF-κB pathway in hepatocellular carcinoma. Int Immunopharmacol. 2023;123:110685. [DOI] [PubMed]
Kong S, Li J, Pan X, Zhao C, Li Y. Allicin regulates Sestrin2 ubiquitination to affect macrophage autophagy and senescence, thus inhibiting the growth of hepatoma cells. Tissue Cell. 2024;88:102398. [DOI] [PubMed]
Chen H, Zhu B, Zhao L, Liu Y, Zhao F, Feng J, et al. Allicin Inhibits Proliferation and Invasion in Vitro and in Vivo via SHP-1-Mediated STAT3 Signaling in Cholangiocarcinoma. Cell Physiol Biochem. 2018;47:64153. [DOI] [PubMed]
Lv Q, Xia Q, Li J, Wang Z. Allicin suppresses growth and metastasis of gastric carcinoma: the key role of microRNA-383-5p-mediated inhibition of ERBB4 signaling. Biosci Biotechnol Biochem. 2020;84:19972004. [DOI] [PubMed]
Shi G, Li X, Wang W, Hou L, Yin L, Wang L. Allicin Overcomes Doxorubicin Resistance of Breast Cancer Cells by Targeting the Nrf2 Pathway. Cell Biochem Biophys. 2024;82:65967. [DOI] [PubMed]
Maitisha G, Aimaiti M, An Z, Li X. Allicin induces cell cycle arrest and apoptosis of breast cancer cells in vitro via modulating the p53 pathway. Mol Biol Rep. 2021;48:726172. [DOI] [PubMed]
Sun S, Liu X, Wei X, Zhang S, Wang W. Diallyl trisulfide induces pro-apoptotic autophagy via the AMPK/SIRT1 signalling pathway in human hepatocellular carcinoma HepG2 cell line. Food Nutr Res. 2023;66. [DOI] [PubMed] [PMC]
Guan F, Ding Y, He Y, Li L, Yang X, Wang C, et al. Involvement of adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1 in diallyl trisulfide-induced cytotoxicity in hepatocellular carcinoma cells. Korean J Physiol Pharmacol. 2022;26:45768. [DOI] [PubMed] [PMC]
Paul R, Prasad M, Sah NK. Anticancer biology of Azadirachta indica L (neem): a mini review. Cancer Biol Ther. 2011;12:46776. [DOI] [PubMed]
Mehmetoglu-Gurbuz T, Lakshmanaswamy R, Perez K, Sandoval M, Jimenez CA, Rocha J, et al. Nimbolide Inhibits SOD2 to Control Pancreatic Ductal Adenocarcinoma Growth and Metastasis. Antioxidants (Basel). 2023;12:1791. [DOI] [PubMed] [PMC]
Liu J, Hou C, Lin F, Tsao Y, Hou S. Nimbolide Induces ROS-Regulated Apoptosis and Inhibits Cell Migration in Osteosarcoma. Int J Mol Sci. 2015;16:2340524. [DOI] [PubMed] [PMC]
Teng Y, Huang Y, Yu H, Wu C, Yan Q, Wang Y, et al. Nimbolide targeting SIRT1 mitigates intervertebral disc degeneration by reprogramming cholesterol metabolism and inhibiting inflammatory signaling. Acta Pharm Sin B. 2023;13:226980. [DOI] [PubMed] [PMC]
Xia D, Chen D, Cai T, Zhu L, Lin Y, Yu S, et al. Nimbolide attenuated the inflammation in the liver of autoimmune hepatitis’s mice through regulation of HDAC3. Toxicol Appl Pharmacol. 2022;434:115795. [DOI] [PubMed]
Ram AK, Vairappan B, Srinivas BH. Nimbolide inhibits tumor growth by restoring hepatic tight junction protein expression and reduced inflammation in an experimental hepatocarcinogenesis. World J Gastroenterol. 2020;26:713152. [DOI] [PubMed] [PMC]
Zhang J, Ahn KS, Kim C, Shanmugam MK, Siveen KS, Arfuso F, et al. Nimbolide-Induced Oxidative Stress Abrogates STAT3 Signaling Cascade and Inhibits Tumor Growth in Transgenic Adenocarcinoma of Mouse Prostate Model. Antioxid Redox Signal. 2016;24:57589. [DOI] [PubMed]
Sophia J, Kowshik J, Dwivedi A, Bhutia SK, Manavathi B, Mishra R, et al. Nimbolide, a neem limonoid inhibits cytoprotective autophagy to activate apoptosis via modulation of the PI3K/Akt/GSK-3β signalling pathway in oral cancer. Cell Death Dis. 2018;9:1087. [DOI] [PubMed] [PMC]
Arumugam A, Subramani R, Lakshmanaswamy R. Involvement of actin cytoskeletal modifications in the inhibition of triple-negative breast cancer growth and metastasis by nimbolide. Mol Ther Oncolytics. 2021;20:596606. [DOI] [PubMed] [PMC]
Sekino Y, Teishima J. Molecular mechanisms of docetaxel resistance in prostate cancer. Cancer Drug Resist. 2020;3:67685. [DOI] [PubMed] [PMC]
Zhang J, Jung YY, Mohan CD, Deivasigamani A, Chinnathambi A, Alharbi SA, et al. Nimbolide enhances the antitumor effect of docetaxel via abrogation of the NF-κB signaling pathway in prostate cancer preclinical models. Biochim Biophys Acta Mol Cell Res. 2022;1869:119344. [DOI] [PubMed]
Semenescu I, Avram S, Similie D, Minda D, Diaconeasa Z, Muntean D, et al. Phytochemical, Antioxidant, Antimicrobial and Safety Profile of Glycyrrhiza glabra L. Extract Obtained from Romania. Plants (Basel). 2024;13:3265. [DOI] [PubMed] [PMC]
Cite this Article
Export Citation
Nagar MK, Sudha D, Vairappan B. The role of medicinal plants in the management of hepatocellular carcinoma and its metastasis. Explor Dig Dis. 2025;4:100588. https://doi.org/10.37349/edd.2025.100588
Article Metrics

View: 175

Download: 12

Times Cited: 0