Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 5002 word(s) 5002 2021-01-12 10:09:37 |
2 format correct Meta information modification 5002 2021-10-12 05:39:05 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Fernández-Palanca, P.; Fondevila, F.; Méndez Blanco, C.; Tuñón, M.; González-Gallego, J.; Mauriz, J.L. Quercetin in Hepatocellular Carcinoma. Encyclopedia. Available online: https://encyclopedia.pub/entry/6425 (accessed on 27 April 2024).
Fernández-Palanca P, Fondevila F, Méndez Blanco C, Tuñón M, González-Gallego J, Mauriz JL. Quercetin in Hepatocellular Carcinoma. Encyclopedia. Available at: https://encyclopedia.pub/entry/6425. Accessed April 27, 2024.
Fernández-Palanca, Paula, Flavia Fondevila, Carolina Méndez Blanco, María Tuñón, Javier González-Gallego, Jose L Mauriz. "Quercetin in Hepatocellular Carcinoma" Encyclopedia, https://encyclopedia.pub/entry/6425 (accessed April 27, 2024).
Fernández-Palanca, P., Fondevila, F., Méndez Blanco, C., Tuñón, M., González-Gallego, J., & Mauriz, J.L. (2021, January 14). Quercetin in Hepatocellular Carcinoma. In Encyclopedia. https://encyclopedia.pub/entry/6425
Fernández-Palanca, Paula, et al. "Quercetin in Hepatocellular Carcinoma." Encyclopedia. Web. 14 January, 2021.
Quercetin in Hepatocellular Carcinoma
Edit

Quercetin is a flavonoid present in fruits, vegetables and plants with beneficial effects in several human disorders, including liver cancer. The antioxidant and anti-inflammatory properties make quercetin an interesting drug to be evaluated in hepatocarcinoma (HCC), the major primary liver tumor with a high mortality rate. Moreover, increasing number of studies reported a high variety of antitumor actions which places quercetin as a promising antitumor agent, not only as single treatment but also improving current therapeutic options against advanced HCC.

combined treatments encapsulation flavonoid hepatocarcinoma quercetin quercetin derivative

1. Introduction

Quercetin (3,3’,4’,5,7-pentahydroxy flavone) is one of the main components of the polyphenol family of flavonoids [1] and it is mostly present in fruits, vegetables and some plant-derived beverages, such as wine or tea [2]. This flavonoid has many beneficial properties on human health [2], being associated its biological activity with the presence of five hydroxyl groups on the ring structure [1]. A number of studies have investigated quercetin effects on cellular processes involved in different human pathologies [3][4]. Anti-inflammatory, antioxidant and anticancer activities are some of the mainly described quercetin mechanisms of action [1][2][5]. Besides, therapeutic potential of this flavonoid has been evaluated in a broad variety of human disorders, including diabetes, cardiovascular [3], neurodegenerative [3][4][6] and Alzheimer’s diseases [6]; and positive actions on blood vessel pressure, intestinal microbiota and kidney disfunction [5], among others, were also related to quercetin efficacy.

Liver injury is largely caused by obesity or metabolic syndrome, in addition to high alcohol consumption [5][7]. Hepatocyte damage eventually contributes to the development of liver disorders including steatosis, alcoholic and non-alcoholic steatohepatitis which could cause non-alcoholic fatty liver disease (NAFLD), liver inflammation and hepatic fibrosis [5][7]. Hepatic chronic damage often leads to progression to liver cirrhosis and, in most cases, to hepatocarcinoma (HCC) [5][7]. In addition to the aforementioned beneficial effects, quercetin has been shown to exert multiple hepatoprotective actions through lipid biogenesis modulation, mitochondrial biogenesis activation [8] and the increase of cellular antioxidants and insulin sensitivity [5]. As part of its hepatoprotective ability, this flavonoid has demonstrated to reduce oxidative stress and inflammatory response in liver damage caused by alcohol and different toxic compounds (e.g. ethanol, metals and pesticides) [9]. Generation of an inflammatory and fibrotic microenvironment are key mechanisms produced in chronic-injured liver by hepatic stellate cells, and quercetin is able to abrogate its activation and modulate its polarization, restraining liver cells alteration [10]. Along with this, regulation of liver cell pathways involved in cell proliferation, differentiation and extracellular matrix synthesis is associated with quercetin-derived positive effects in the prevention of NAFLD [11][12] and liver fibrosis [7]. Some studies have also proved its beneficial activities against liver cirrhosis development and pulmonary associated complications [13][14], what makes quercetin a promising agent for the improvement of the outcomes in liver pathologies therapy [9].

HCC is the most common primary liver cancer and the sixth tumor with higher incidence, ranking as the fourth deadliest neoplasm worldwide [15]. Liver damage caused by different etiologic agents, mainly hepatitis C and B virus (HCV and HBV, respectively), contributes to HCC development through the stages of liver fibrosis and cirrhosis, which can take from years to decades [15]. Its complex pathogenesis and molecular heterogeneity hinder HCC early diagnosis making curative treatments not possible [15]. In these cases, systemic therapy is used, being available two tyrosine kinase inhibitors (TKIs), sorafenib and lenvatinib, in the first-line setting for advanced HCC [16]. Regardless its effectiveness, liver cancer cells are able to develop sorafenib resistance after sustained administration [17], where several TKIs (regorafenib and cabozantinib) and monoclonal antibodies (nivolumab, pembrolizumab and ramucirumab) have been recently approved [16]. Considering toxicity and adverse reactions caused by these chemotherapeutic agents, some investigations have focused on the study of antitumor effects of natural compounds against HCC, such as resveratrol, curcumin and melatonin [18][19][20].

2. Antitumor properties of quercetin as single agent against HCC

2.1. Antiproliferative activity of quercetin

Quercetin antitumor effects have been described in different cancer types, including HCC [1], being its antiproliferative effect widely observed in several researches [21][22][23][24][25][26][27][28][29][30][31][32][33][34][35][36][37][38][39]. It has been described that quercetin-derived inhibition of liver cancer cell growth could be mediated by the disruption of different pathways, including protein kinase B (Akt)/mammalian target of rapamycin (mTOR) [22][23][28], mitogen-activated protein kinase kinase 1 (MEK1)/extracellular signal-regulated kinase 1/2 (ERK1/2) [26][32] and Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling routes [21][34]. Another lesser known molecular pathways have also been altered by quercetin, including Src homology domain 2 containing tyrosine phosphatase-1/2 (SHP-1/2), transcription factor specificity protein 1 (Sp1), phosphatidyl inositol 3 kinase (PI3K) and protein kinase C (PKC) routes [28][30][34]. These findings evidence the high number of mechanisms of actions that are associated with the antiproliferative activity of quercetin.

2.2. Cell cycle modulation by quercetin

Otherwise, though cell cycle regulation is a common mechanism found to be altered in HCC cells, publications that have evaluated the effects of quercetin in this process show contradictory results. As common feature, quercetin was able to arrest cell cycle, either in G0/G1 or G2/M phases; however, modulation of cell cycle markers such as p53, p21, p27 and cyclin D1 by quercetin differs among the investigations performed [21][24][27][30][35][36][37][38].

2.3. Apoptosis induction by quercetin

Apoptosis has been clearly established as one of the mechanisms of quercetin-induced cell death in HCC [21][23][27][28][29][30][36][37][38][39][40], as it was demonstrated by the increase in proapoptotic proteins expression, such as Bax and cleaved caspases-3 and -9 [21][23][26][28][29][30][37][38][39][40], and the opposite trend in the levels of antiapoptotic proteins, for instance Bcl-2 and Mcl-1 [23][26][29][30][36][37][40]. Autophagy is a self-recycle process by which damaged cell components are degraded, and it has been associated with either prosurvival or antitumor role depending on the tumor cell context [21][41]. In this case, two studies observed that quercetin treatment promoted autophagy in HCC cells, being related to apoptosis induction and suppression of tumor progression [21][23].

2.4. Antiangiogenic effects of quercetin

Angiogenesis and metastasis are tightly associated with HCC progression and represent very common targeting processes in cancer treatment [15]; however, there is a lack of studies evaluating quercetin effects on cell migration and invasion ability [21]. It was reported that this flavonoid regulated the expression of epithelial and mesenchymal markers in favor of first ones, abrogating epithelial-to-mesenchymal transition (EMT) and invasiveness of HCC cells [21]. Similarly, another study revealed a greater antiangiogenic activity by combination of quercetin and sorafenib in an in vivo HCC model [42], which suggest a promising role of quercetin not only as cytostatic agent but also in preventing recurrence-associated processes of angiogenesis and invasion.

2.5. Other antitumor actions of quercetin

In addition to the described properties, quercetin has shown to act as an antioxidant agent in human HCC models [24][28][43], although some investigations denoted contradictory results [29]. In addition to the already mentioned processes, several studies have determined effects of quercetin in different specific mechanisms [26][32][44][45][46]. These include inhibition of chymotrypsin-like activity of the proteasome, involved in proteasomal regulation of cancer signaling pathways [26][32]; rise of intracellular labile zinc, which has second messenger molecule activities in tumor cells [46]; and modulation of microRNAs expression [45]. Curiously, another study published in 2018 employed quercetin to analyze adequacy of the cellular antioxidant (CAA) assay in HepG2 cells in order to determine the antioxidant activity of extracts from tree nuts [44]. Besides, beneficial effects of quercetin against HCC were evaluated focusing on the sensitization of chemoresistant liver cancer cells and its hepatoprotective effects [25][31][47][48].

Altogether, the wide variety of antitumor effects of quercetin along with its demonstrated efficacy against HCC cells, set this flavonoid as a promising therapeutic agent in the treatment of HCC.

3. Encapsulation strategy for the improvement of quercetin effects in HCC

Drug delivery systems have emerged as a novel mechanism of targeting cancer progression, enhancing drug efficacy through its encapsulation [49]. In this line, nanomedicine has developed numerous nanoparticles employing either organic- or inorganic-based nanocarriers [50]. Mesoporous silica nanoparticles (MSNs) conjugated with folic acid (FA) were designed to improve antitumor activity of quercetin. As inorganic-based carriers, gold-nanoparticles are drug delivery systems commonly chosen in nanomedicine [50]. Different publications synthesized poly(DL-lactide-co-glycolide) (PLGA)-loaded gold-quercetin nanoparticles leading to higher antiproliferative actions of quercetin in different HCC cell lines and in a xenograft tumor mouse model [49][51]. Otherwise, changes in cell cycle modulation were also observed after quercetin-nanocapsules administration and apoptosis induction was incremented in both in vitro and in vivo experiments [49][51]. In addition to cell proliferation and apoptosis, PLGA nanoparticles carrying quercetin generated morphologic alterations and inhibited migration ability of HCC cells [49][51]. Likewise, another group of researchers demonstrated greater cell death stimulation by encapsulating quercetin into PLGA nano-prototypes decorated with chitosan (CS) and polyethylene glycol (PEG) in HepG2 cells [52].

Some studies have also evaluated lipid-based formulations as quercetin-encapsulation strategies [53][54]. Solid lipid nanoparticles (SLNs) containing three sterol variables (cholesterol, stigmastanol and stigmasterol) were designed and evaluated, rising quercetin-derived cell viability reduction [54]. Similarly, the employment of methoxy-poly(ethylene glycol)-b-oligo(e-caprolactone), mPEG750-b-OCL-Bz micelles to co-encapsulate quercetin and superparamagnetic iron oxide nanoparticles (SPIONs) denoted lower proliferative capacity, morphological changes and cell cycle arrest in G0/G1 phase in an HCC model [53].

Even though few studies analyzed quercetin nanoencapsulation as a drug delivery system in liver cancer cells, it has arisen as a novel therapeutic approach which could improve quercetin properties against HCC progression by specifically targeting tumor and increasing drug cellular uptake.

4. Synergistic effects through quercetin combination against HCC 

Although a great number of antitumor properties of quercetin have been established in HCC treatment mainly in cell line but also in animal models, some researchers have focused on searching for synergistic combinations with this flavonoid with the aim of improving its effectiveness against liver cancer [33][34][35][40][36][37][38][39][55][56]. Enhanced properties with the well-stablished first-line drug sorafenib was demonstrated in several HCC cell lines by reducing its half-maximal inhibitory concentration (IC50) [36] and improving its tumor suppression activity [33]. Similarly, quercetin was able to raise antiproliferative action of several molecules, such as interferon-a (IFN-a) [34]; an oncolytic adenovirus expressing tumor necrosis factor-related apoptosis inducing ligand (ZD55-TRAIL) [56]; two derivatives of the organic compound maleic anhydride (3’5’-dimaleamylbenzoic acid and 3’5’-dimaleimylbenzoic acid) [35]; the chemotherapeutic drugs celecoxib [40], 5-fluorouracil (5-FU) [37] and cisplatin [38]; and the CDK inhibitor roscovitine [39]. Results reported quercetin-derived alterations in cell morphology when combined with roscovitine [39] as well as growth inhibition and cell cycle arrest after cisplatin and two maleic anhydride derivatives co-administration [35][38]. A higher quercetin-associated apoptosis was also observed when administered with the following compounds, ZD55-TRAIL [56], two maleic anhydride derivatives [35], celecoxib [40], cisplatin [38] and roscovitine [39].

Regulation of oxidative stress and ROS production by quercetin was also altered after addition of 3’5’-dimaleamylbenzoic acid and 3’5’-dimaleimylbenzoic acid, showing an increase in the reactive oxygen species (ROS) accumulation [35]. On the other hand, effects of quercetin and dasatinib combination in cell senescence were assessed in HCC cells, which showed inability to prevent doxorubicin-induced senescence [55].

It should be mentioned that two independent studies put together both quercetin combination and encapsulation strategies and evaluated its cytotoxic actions both in vitro and in vivo models [42][57]. Lactoferrin shell-oily core nanocapsules coupled with lactobionic acid (LA) or glycyrrhetinic acid (GA) were designed for targeted delivery of both quercetin and sorafenib, showing greater antitumor effects in HepG2 cell line and HCC-bearing mice [42]. Similar results were described with arginine-glycineaspartic acid (RGD)-modified lipid-coated nanoparticles loaded with quercetin and sorafenib using HepG2 cells and a mouse model of HCC [57].

Globally, co-treatment strategy of quercetin with different compounds may enhance its effectiveness by raising mainly its antiproliferative and proapoptotic effects and leading to improve quercetin single-therapy properties against HCC.

5. Effects of quercetin derivatives as treatment in HCC

Quercetin glycosides are one of the main quercetin naturally occurring forms, what makes them interesting compounds for cancer treatment [58]. This led several researchers to analyze effects of quercetin derivatives in different tumors, including HCC [43][58][59][60]. In this line, quercetin-3-O-glucoside (Q3G) and six long chain fatty acid esters of Q3G (stearic acid ester, oleic acid ester, linoleic acid ester, alpha-linoleic acid ester, eicosapentaenoic acid ester and docosahexanoic acid ester) displayed antiproliferative actions, cell cycle arrest and apoptosis induction in in vitro models of HCC [58][60]. On the other hand, 3,4-dihydroxyphenylacetic acid (DOPAC), a catabolite of some quercetin glycosides produced by colonic microflora, has been shown to modulate oxidative stress enzymes and to protect against acetaldehyde damage [59]. In the same way, both 3’-O-methyl quercetin (3’MQ) and quercetin-3-O-glucuronide (Q3GA) exhibited antioxidant properties by reverting ethanol-derived ROS accumulation [43].

Bioactive compounds derived from quercetin have been shown to abrogate cancer progression in liver cancer cells; nonetheless, a greater number of studies would be needed to search for more quercetin derivatives and study underlaying mechanisms of its antitumor action against HCC cells.

Main findings here described reporting quercetin effects in both in vitro and in vivo models are summarized in Table 1 and Table 2, respectively.

Table 1. Basic characteristics of in vitro studies using quercetin in single, encapsulated, combined or derived forms in HCC.

First author, Year of publication

Quercetin administration strategy

Cell line

General effects

Molecular alterations

Wu, 2019 [21]

Quercetin

LM3 cells

Cell viability reduction

Apoptosis induction

Cell cycle arrest at S and G2/M phases

Autophagy induction

Cell migration and invasion suppression

Morphological changes

­ ↑ Early stage apoptotic cells

↓ PCNA mRNA and protein levels

­ ↑ Bax mRNA and protein levels

↓ Colony formation

­ ↑ Fluorescence in TUNEL staining

↓ G0/G1 phase cells and ­↓ S and G2/M phase cells

↓ Cyclin B1 protein expression

­ ↑ E-cadherin and ↓ vimentin and MMP-9 mRNA and protein levels

↓ N-cadherin protein expression

↓ Invasiveness (Transwell invasion assay)

↓ Migrating cells (Wound-healing assay)

­ ↑ LC3 mRNA and protein levels

­ ↑ Beclin1 protein expression

↓ p62 mRNA and protein levels

↓ p-STAT3 protein expression

­ ↑ LC3 protein levels decreased by IL-6

↓ PCNA and MMP-9 protein levels enhanced by IL-6

↓ Wound healing speed

Wu, 2019 [22]

Quercetin

SMMC-7721, BEL-7402 HCC cells

LO2 normal liver cells

Proliferation suppression of HCC cell lines

No cytotoxic for normal hepatic cells

Glycolysis inhibition

↓ Glucose uptake and lactate production

↑ 2-DG-derived cell growth inhibition

↓ HK2 mRNA and protein expression

↓ p-Akt/Akt and p-mTOR/mTOR rates

Ji, 2019 [23]

Quercetin

SMMC-7721 and HepG2 HCC cells

LO2 normal hepatic cells

Cell growth inhibition in HCC cell lines

Absence of antiproliferation effect in LO2 cells

Induction of autophagy

Apoptosis increase

↑ In both tumor cell lines:

­ ↑ LC3A/LC3B-II and Beclin1 protein levels

↓ p62 protein expression

In SMMC-7721: ↑ Autophagosomes and autolysosomes

In all cell lines:

↓ p-Akt, p-mTOR, p-p70S6K and p-4EBP1 protein levels

­ ↑ p-JNK, p-ERK1/2 and p-p38 MAPK protein expression

­ ↑ Apoptotic cells percentage

­ ↑ Bax and cleaved caspase-3 protein levels

↓ Bcl-2 protein expression

Jeon, 2019 [24]

Quercetin

HepG2, HuH7, PLC/PRF-5 and Hep3B cells

Proliferation inhibition (in HepG2, PLC/PRF-5 and Hep3B cells)

ROS levels reduction (in HepG2 cells)

Morphological alterations

Only in HepG2 cell line:

­ ↑ p53 and HO-1 protein expression

↓ Cyclin A and CHK1 protein levels

No variation in cyclin E and SOD1 protein expression

Chen, 2018 [25]

Quercetin

BEL-7402 HCC cells

Multidrug resistant cell line BEL/5-FU

Increase of 5-FU, MMC and ADR chemosensitivity in BEL/5-FU cells

Only in BEL/5-FU cell line:

↓ IC50 of 5-FU, MMC and ADR

↓ ABCB1, ABCC1 and ABCC2 mRNA levels

­ ↑ Rh123 accumulation

Inhibition of ABCC2 function

­ ↑ ADR accumulation

In both cell lines:

↓ FZD7, b-catenin (nuclear and cytoplasmic), ABCB1, ABCC1 and ABCC2 mRNA and protein expression

Ding, 2018 [26]

Quercetin

HepG2 HCC cell line

Decrease of cell viability

Apoptosis induction

Inhibition of chymotrypsin-like activity

­ ↑ TUNEL-positive cells

­ ↑ Cleaved caspase-3, cleaved PARP and Bax protein expression

↓ Bcl-2 protein levels

↓ Chymotrypsin-like activity

No changes in trypsin-like and caspase-like activities

­ ↑ p-p38 MAPK and JNK protein expression

↓ p-ERK1/2 protein levels

↓ Protein expression of b1, b2 and b5 proteasomal subunits

Kellet, 2018 [44]

Quercetin

HepG2 cells

Antioxidant activity

­ ↑ CAA unit dose dependent

Shaalan, 2018 [45]

Quercetin

HuH7 cell line

-

­ ↑ miR-1275 mRNA levels

↓ IGF2BP1 and IGF2BP3 mRNA expression

Pi, 2016 [27]

Quercetin

HepG2 cells

Suppression of cell proliferation

Cell cycle arrest at G2/M phase

Apoptosis increase

Disruption of mitochondrial membrane potential

Morphological alterations

Changes in surface ultrastructure

­ ↑ G2/M cell population

­ ↑ Early apoptotic, late apoptotic and necrotic cells

↓ Fluorescence signal of Rh123

­ ↑ F-actin filaments aggregation in apoptotic cells

­ ↑ Particle size on HepG2 membrane

­ ↑ Surface root-mean-squared and surface average roughness

­ ↑ Cell stiffness and Young’s modulus

Maurya, 2015 [28]

Quercetin

HepG2 cells

Antiproliferative activity

Morphological changes

↓ ROS generation and PKC activity

↓ p-p85α, p-PKC, PKCα, COX-2 protein levels

­ ↑ p53 protein expression and Bax mRNA levels

Zhang, 2015 [29]

Quercetin

HepG2 cells

Cell viability inhibition

Induction of apoptosis

↑ Chromatin condensation and nuclei fragmentation into oligonucleosomes

­ ↑ PIG3 mRNA and protein expression

­ ↑ Early apoptotic cells

­ ↑ ROS accumulation

↓ Mitochondrial membrane potential

↓ Mitochondrial cytochrome c and Bcl-2 protein expression

­ ↑ Cytosolic cytochrome c, Bax and activated caspases -9 and -3

Lee, 2015 [30]

Quercetin

HepG2 cells

Decrease in cell viability

Apoptosis induction

­ ↑ Nuclear condensation and fragmentation

­ ↑ Early and late apoptotic cells

↓ Sp1 mRNA and protein levels

­ ↑ p21, p27, Bax, cleaved caspase-3 and cleaved PARP protein expression

↓ Cyclin D1, Mcl-1, survivin and Bcl-xL protein levels

Dabbagh-Bazarbachi, 2014 [46]

Quercetin

Mouse hepatoma Hepa 1-6 cell line

Augmented cytoplasmic labile zinc

High ionophore activity

­ ↑ FluoZin-3-detectable zinc

­ ↑ Fluorescence signal of FluoZin-3

Kozics, 2011 [31]

Quercetin

HepG2 cells

Reduction of cell proliferation

↓ B(a)P-induced micronuclei formation and DNA damage

Oliva, 2011 [47]

Quercetin

Cederbaum’s CYP2E1 overexpressing HepG2 cell line

Decrease of ethanol-derived oxidative stress

↓ MDA, 4-HNE and carbonyl protein levels augmented by ethanol

↓ Ethanol-induced glutathione peroxidase 4 and SOD2 mRNA expression

↓ Gadd45b mRNA levels

­ ↑ Nrf2 protein levels reduced by ethanol

Choi, 2010 [48]

Quercetin

HepG2 cells

Reduction of the AFB1 antiproliferative effect

↓ ROS accumulation generated by AFB1

­ ↑ AFB1-reduced GSH levels

Granado-Serrano, 2010 [32]

Quercetin

HepG2 cells

Cell proliferation suppression

↓ NF-kB and p65 nuclear translocation, NF-kB DNA-binding activity

↓ p-IkBa and IKKa protein expression

↓ Chymotrypsin-like activity

No changes in trypsin-like activity

­ ↑ DNA-binding activity of AP-1

­ ↑ Nuclear c-Jun levels

Srisa-nga, 2019 [53]

Quercetin encapsulation

(Quercetin-SPION-loaded micelles)

HepG2.2.15 cell line

Suppression of cell growth

Morphological alterations

­ ↑ G0/G1 and ↓ G2/M phase cells

AbouAitah, 2018 [50]

Quercetin encapsulation (FA-conjugated MSNs)

HepG2 cells

Increased antiproliferative activity

­ ↑ Antioxidant effect

­ ↑ Inhibition of ABTS.+ radical formation

Abd-Rabou, 2017 [52]

Quercetin encapsulation (CS and PEG-decorated PLGA nano-prototypes)

HepG2 cells

Cell viability reduction

Apoptosis induction

↓ Quercetin IC50

­ ↑ Late apoptotic and necrotic cells

Ren, 2017 [49]

Quercetin encapsulation (PLGA-loaded gold-quercetin nanoparticles)

MHCC97H, Hep3B, HCCLM3 and BEL-7402 HCC cell lines

Decreased cell proliferation

Only in MHCC97H cell line:

Morphological alterations

Reduction of cell migration ability

Apoptosis increase

Only in MHCC97H line:

↓ Colony formation

­ ↑ Cell-to-cell adhesions and ↓ filopodia generation and cell spreading

↓ Migrating cells

­ ↑ P-27 protein levels

↓ c-Myc, cyclin D1, CDK1, MMP-7 and b-catenin protein expression

­ ↑ Apoptotic cell number

­ ↑ Cleaved caspases -9 and -3 protein levels

­ ↑ Cytochrome c release to cytoplasm

↓ hTERT and AP-2β mRNA and protein expression

↓ hTERT promoter-binding activity of AP-2β

↓ COX-2 protein expression

↓ Binding activity of p50 on COX-2 promoter

↓ p-IKKa and p-IkBa protein levels

­ ↑ NF-kB and p50 cytoplasm translocation from nuclei

↓ p-Akt and p-ERK1/2 protein levels

Bishayee, 2015 [51]

Quercetin encapsulation (PLGA-loaded gold-quercetin nanoparticles)

HepG2 cells

Inhibition of cell proliferation

Growth rate reduction

Apoptosis stimulation

Morphological changes

Alteration of B-conformation of DNA

↓ p-Akt protein expression

­ ↑ sub G-phase cells and ↓ S-phase cells

­ ↑ p21 protein levels

↓ CDK1 and cyclin D1 protein expression

↓ HDAC activity and HDAC1/2 protein levels

­ ↑ ROS formation

­ ↑ rac-1 activity and later returned to basal levels

↑ Depolarization of mitochondrial membrane

↑ Bax translocation to the mitochondrial outer membrane

­ ↑ Cytochrome c release to cytosol

↑ Generation of DNA damage

↓ Mcl-1, Bcl-2 and Bcl-xL protein levels

­ ↑ Apaf1, caspases -9 and -3, and cleaved PARP protein expression

Varshosaz, 2013 [54]

Quercetin encapsulation (SLNs containing cholesterol, stigmastanol or stigmasterol)

HepG2 cells

Cell growth inhibition (the highest with cholesterol)

-

Kovacovicova, 2018 [55]

Quercetin combined with dasatinib

HepG2 and HuH7 cell lines

No senolytic activity exhibited

No effects in β-galactosidase activity

No protein expression alteration of the senescence markers p16 and γH2A.X

Bahman, 2018 [33]

Quercetin

HepG2 and Hep3B cells

Antiproliferative effect

-

Quercetin combined with sorafenib

Suppression of cell proliferation

-

Zou, 2018 [56]

Quercetin combined with ZD55-TRAIL

SMMC-7721, HepG2 and HuH7 cell lines

Decrease of cell proliferation

Apoptosis induction

­ ↑ Apoptotic bodies, nuclear fragmentation and chromatin condensation

­ ↑ Cleaved caspases -9 and -3, cleaved PARP, Bid and Bax protein expression

↓ Bcl-2 and FLIP protein levels

↓ IκBα, p65 and p50 protein expression

Igbe, 2017 [34]

Quercetin

HepG2 and HuH7 HCC cell lines

Inhibition of cell viability

↓ SHP-1 and SHP-2 protein expression in HepG2 cells

Quercetin combined with IFN-a

Increased cell growth inhibition in both HCC cell lines

Only in HepG2 cell line:

↓ SHP-2 protein expression

­ ↑ p-STAT1, p-Jak1 and p-Tyk2 protein levels

­ ↑ ISRE reporter expression

­ ↑ 2’,5’-OAS and PKR mRNA levels

↓ Colony formation

↓ Cyclin D1 protein expression

Carrasco-Torres, 2017 [35]

Quercetin

HuH7 and HepG2 HCC cells

Antiproliferative effect

Cell cycle arrest at G0/G1 phase

­ ↑ G0/G1 cell population

↓ ROS levels and oxidized glutathione levels

­ ↑ Reduced glutathione and GSH/GSSG index

­ ↑ Nuclear condensation

­ ↑ Pro-caspase-9 and cleaved caspases -9 and -3 protein expression

Quercetin combined with 3’5’-dimaleamylbenzoic acid or 3’5’-dimaleimylbenzoic acid

Cell viability reduction

Cell cycle arrest at S phase

Antioxidant activity

Apoptosis induction

In both cell lines:

↓ G2/M-phase and ­↑ S-phase populations

↓ Reduced and oxidized glutathione levels and GSH/GSSG index in both cell lines (maleic anhydride derivative + quercetin)

­ ↑ Nuclear condensation, degradation of actin and DNA

­ ↑ Pyknotic nuclei number and TUNEL-positive cells

­ ↑ Pro-caspase-9 and cleaved caspases -9 and -3 protein expression

In HuH7 line: ↓ ROS levels

In HepG2 line:

↓ ROS levels (quercetin + maleic anhydride derivative)

­ ↑ ROS levels (maleic anhydride derivative + quercetin)

­ ↑ Reduced glutathione levels and de novo glutathione synthesis (quercetin + maleic anhydride derivative)

Yu, 2017 [40]

Quercetin combined with celecoxib

HepG2 and HuH7 cell lines

Antiproliferative effect

Apoptosis induction

­ ↑ DNA fragmentation

­ ↑ Bax protein expression

↓ Bcl-2 protein levels

Brito, 2016 [36]

Quercetin

HepG2, HuH7 and Hep3B2.1-7 HCC cell lines

Inhibition of cell growth and survival

Apoptosis increase

Cell cycle arrest

­ ↑ Apoptotic and necrotic cells

­ ↑ Bax/Bcl-2 ratio

­ ↑ G0/G1 and G2/M cell population in HepG2 and HuH7

↓ S phase cells in all cell lines

↓ p53 protein expression in HepG2 and HuH7 cells

­ ↑ DNA damage

­ ↑ Membrane expression of GLUT-1

↓ Cytoplasmic expression of GLUT-1 in HepG2 and HuH7 cells

18F-FDG uptake

Quercetin combined with sorafenib

Decrease in sorafenib IC50

-

Dai, 2016 [37]

Quercetin

HepG2 and SMMC-7721 HCC cells

Suppression of cell proliferation

Cell cycle arrest at G0/G1 phase

Apoptosis increase

­ ↑ G0/G1 phase and ↓ S phase cell population

­ ↑ Bax and Bad protein expression

↓ Bcl-2 and survivin protein levels

Quercetin combined with 5-FU

Rise of 5-FU antiproliferative effects

Higher apoptotic activity

-

Zhao, 2014 [38]

Quercetin

HepG2 cells

Inhibition of cell survival

Apoptosis induction

G1-phase arrest of cell cycle

­ ↑ Cleaved caspase-3 and cleaved PARP protein levels

­ ↑ p21, p53 and p16 protein expression

­ ↑ G1-phase cells and ↓ S-phase cells

­ ↑ sub-G1 cell population

Quercetin combined with cisplatin

Increased growth inhibitory action

Greater apoptotic effects

­ ↑ Cleaved caspase-3 and cleaved PARP protein levels

­ ↑ p21 and p53 protein levels

Sharma, 2011 [39]

Quercetin

HepG2 and Hep3B cell lines

Reduced cell survival

Morphological changes

Apoptosis induction

­ ↑ Apoptotic bodies

­ ↑ p53 protein expression in HepG2 cells

↓ Pro-caspase-9 and ↑­ caspase-9 protein levels in HepG2 cells

Quercetin combined with roscovitine

Augmented cell proliferation inhibition

Morphological alterations

Apoptosis stimulation

↓ Cell density

­ ↑ Floating cells number and apoptotic bodies

↓ p-Akt, Bcl-2 and pro-caspases -9 and -3 protein expression

↓ Bcl-2/Bax ratio and ↑ Caspases -9 and -3 protein levels

Abdelmoneem, 2019 [42]

Co-encapsulated quercetin and sorafenib (LF-coated, LA/LF-coated or GA/LF-coated nanocapsules)

HepG2 cells

Higher antitumoral efficacy of quercetin and sorafenib

Cell viability suppression

↓ IC50 of quercetin and sorafenib

↓ Combination index

­ ↑ Dose reduction index of quercetin and sorafenib

­ ↑ Cellular uptake of both drugs

Wang, 2016 [57]

Co-encapsulated quercetin and sorafenib (RGD-modified lipid-coated nanoparticles)

HepG2 cells

Reduced cell proliferation

↓ IC50 of quercetin and sorafenib

Lee, 2017 [43]

Quercetin

HepG2 cells

Reduced antiproliferative action of ethanol

Antioxidant activity

Reversal of ethanol effects:

↓ ROS formation

↓ MDA levels

­ ↑ GSH, SOD and CAT levels

­ ↑ HO-1 and nuclear Nrf2 protein expression

­ ↑ AP-1 activity

3’MQ

Lower ethanol-induced cell viability inhibition

Antioxidant activity

Reversal of ethanol effects:

↓ ROS formation

­ ↑ SOD and CAT levels

­ ↑ HO-1 and nuclear Nrf2 protein expression

­ ↑ AP-1 activity

Q3GA

Reversion of proliferation suppression induced by ethanol

Antioxidant activity

Reversal of ethanol effects:

↓ ROS formation and ­ GSH, SOD and CAT levels

­ ↑ HO-1 and nuclear Nrf2 protein expression

­ ↑ AP-1 activity

Liu, 2017 [59]

DOPAC

Mouse hepatoma Hepa1c1c7 cell line

Reduced acetaldehyde-derived cell growth inhibition

­ ↑ ALDH activity

­ ↑ ALDH1A1, ALDH2 and ALDH3A1 mRNA and protein levels

­ ↑ Nrf2 and AhR total and nuclear protein expression

↓ NF-kB nuclear expression

Sudan, 2015 [60]

Six Q3G esters:

Stearic acid ester

Oleic acid ester

Linoleic acid ester

Alpha-linoleic acid ester

Eicosapentaenoic acid ester

Docosahexanoic acid ester

HepG2 HCC cells and normal hepatocytes

Higher cell viability of normal hepatocytes

In HepG2 cells:

Cell proliferation decrease

Morphology changes

Apoptosis induction

Activity as catalytic inhibitor by DNA relaxation activity blockade

In HepG2 cells:

↓ HepG2 cell number

­ ↑ DNA fragmentation

­ ↑ Caspase-3 activity

­ ↑ S and G2/M cell population

↓ G0/G1-phase cells

No stabilization of topoisomerase II cleavage complexes and no formation of single linear DNA

­ ↑ Supercoiled DNA intensity

Sudan, 2014 [58]

Q3G

HepG2 cell line

Cell growth suppression

S-phase arrest of cell cycle

Morphology alterations

Apoptosis induction

Catalytic inhibitor action by DNA relaxation activity inhibition

­ ↑ S-phase and ↓ G0/G1 cell percentage

­ ↑ DNA fragmentation

­ ↑ Caspase-3 activity

­ ↑ Apoptotic and necrotic cells

No stabilization of topoisomerase II cleavage complexes and no formation of single linear DNA

­ ↑ Supercoiled DNA intensity

18F-FDG: fluorine-18 fluorodeoxy-glucose; 2’5’-OAS: 2’5’ oligoadenylate synthetase; 2-DG: 2-deoxy-D-glucose; 3’MQ: 3’-O-methyl quercetin; 4-HNE: 4-hydroxynonenal; 4EBP1: eukaryotic translation initiation factor 4E-binding protein 1; 5-FU: 5-fluorouracil; ABCB1: ATP-binding cassette subfamily B member 1; ABCC1: ATP-binding cassette subfamily C member 1; ABCC2: ATP-binding cassette subfamily C member 2; ABTS.+: radical cations of 2,2’-azino-bis(3-ethyl-benzothiazoline-6-sulphonic acid) diammonium salt; ADR: doxorubicin; AFB1: aflatoxin B1; AhR: aryl hydrocarbon receptor; Akt: protein kinase B; ALDH: aldehyde dehydrogenase; ALDH1A1: aldehyde dehydrogenase 1 member A1; ALDH2: aldehyde dehydrogenase 2; ALDH3A1: aldehyde dehydrogenase 3 member A1; AP-1: transcription factor AP-1; Apaf1: apoptotic protease-activating factor 1; B(a)P: benzo[a]pyrene; Bad: Bcl-2-associated agonist of cell death; Bax: Bcl-2-associated X; Bcl-xL: Bcl-2-like protein 1; Bid: BH3-interacting domain death agonist; CAA: cellular antioxidant activity; CAT: catalase; CDK1: cyclin-dependent kinase 1; CHK1: checkpoint kinase 1; COX-2: cyclooxygenase-2; CS: chitosan; DOPAC: 3,4-dihydroxyphenylacetic acid; ERK1/2: extracellular signal-regulated kinase 1/2; FA: folic acid; FLIP: FLICE-like inhibitory protein; FZD7: Frizzled homolog protein 7; GA: glycyrrhetinic acid; Gadd45b: growth arrest and DNA damage-inducible protein GADD45 beta; GLUT-1: glucose transporter type 1; GSH: glutathione; GSSG: oxidized glutathione; HCC: hepatocarcinoma; HDAC: histone deacetylase; HK2: hexokinase-2; HO-1: heme oxygenase-1; hTERT: telomerase reverse transcriptase; IC50: half-maximal inhibitory concentration; IFN-a: interferon-a; IGF2BP1: insulin-like growth factor-2 binding protein 1; IGF2BP3: insulin-like growth factor-2 binding protein 3; IkBa: nuclear factor-kB inhibitor a; IKKa: inhibitor of nuclear factor-kB kinase subunit a; IL-6: interleukin 6; ISRE: interferon-sensitive response element; Jak1: Janus kinase 1; JNK: c-Jun N-terminal kinase; LA: lactobionic acid; LC3: microtubule-associated protein 1 light chain 3; LC3A: microtubule-associated protein 1A/1B light chain 3A; LC3B-II: microtubule-associated protein 1A/1B light chain 3B; LF: lactoferrin; MDA: malondialdehyde; Mcl-1: induced myeloid leukemia cell differentiation protein; MMC: mitomycin; MMP-7: matrix metalloproteinase-7; MMP-9: matrix metalloproteinase-9; MSNs: mesoporous silica nanoparticles; mTOR: mammalian target of rapamycin; NF-kB: nuclear factor-kB; Nrf2: nuclear factor erythroid 2-related factor 2; p38 MAPK: mitogen-activated protein kinase p38; p62: sequestosome-1; p70S6K: ribosomal protein S6 kinase beta-1; PARP: poly(ADP-ribose) polymerase; PCNA: proliferating cell nuclear antigen; PEG: polyethylene glycol; PIG3: p53-inducible gene 3; PKC: protein kinase C; PKR: RNA-activated protein kinase; PLGA: poly(DL-lactide-co-glycolide); Q3G: quercetin-3-O-glucoside; Q3GA: quercetin-3-O-glucuronide; RGD: arginine-glycineaspartic acid; Rh123: rhodamine 123; ROS: reactive oxygen species; SHP-1: Src homology domain 2 tyrosine phosphatase-1; SHP-2: Src homology domain 2 containing tyrosine phosphatase-2; SLNs: solid lipid nanoparticles; SOD: superoxide dismutase; SOD1: superoxide dismutase 1; SOD2: superoxide dismutase 2; Sp1: specificity protein 1; SPION: superparamagnetic iron oxide nanoparticles; STAT1: signal transducer and activator of transcription 1; STAT3: signal transducer and activator of transcription 3; TUNEL: terminal deoxynucleotidyl transferase dUTP nick end labeling; Tyk2: non-receptor tyrosine-protein kinase 2; ZD55-TRAIL: oncolytic adenovirus expressing tumor necrosis factor-related apoptosis inducing ligand.

 

Table 2. Basic characteristics of in vivo studies using quercetin in single, encapsulated, combined or derived forms in HCC.

Wu, 2019 [21]

Quercetin

Nude mice subcutaneously injected with LM3 HCC cells

Tumor growth inhibition

↓ Tumor volume (70% vs control)

↓ Mouse weight and tumor volume

­ ↑ Necrosis

­ ↑ TUNEL-positive cells

↓ PCNA protein levels

­ ↑ Bax and Beclin1 protein levels

Wu, 2019 [22]

Quercetin

SMMC-7721 xenograft mouse model

Tumor growth inhibition

↓ Tumor size

↓ HK2 and Ki67 protein expression

↓ p-Akt and p-mTOR protein levels

Ji, 2019 [23]

Quercetin

Nude mice subcutaneously injected with SMMC-7221 HCC cells

Suppression of tumor growth

Apoptosis and autophagy induction

↓ Tumor weight and volume

­ ↑ LC3A/LC3B and ↓ p62 protein levels

­ ↑ Necrosis and TUNEL staining

­ ↑ Bax and cleaved caspase-3 protein levels

↓ Bcl-2 protein expression

Ren, 2017 [49]

Quercetin encapsulation (PLGA-loaded gold-quercetin nanoparticles)

MHCC97H xenograft mouse model

Suppression of tumor growth and progression

Apoptosis increase

↓ Tumor weight and volume

↓ AP-2β and COX-2 protein levels

­ ↑ TUNEL-positive cells

↓ Cleaved caspases -9 and -3, cytoplasmic cytochrome c, p-IKKα, p-IκBα, p-NF-kB, p50, hTERT, p-Akt, Raf, and p-ERK1/2 protein expression

Kovacovicova, 2018 [55]

Quercetin combined with dasatinib

Mice subcutaneously injected with HuH7 cells

Absence of tumor growth inhibition

-

Zou, 2018 [56]

Quercetin combined with ZD55-TRAIL

HuH7 xenograft mouse model

Tumor growth inhibition

↓ Tumor volume

Dai, 2016 [37]

Quercetin

Nude mice subcutaneously injected with HepG2 HCC cells

Decreased tumor progression

↓ Tumor volume

Quercetin combined with 5-FU

Higher tumor growth inhibition

↓ Tumor volume

Abdelmoneem, 2019 [42]

Co-encapsulated quercetin and sorafenib (LF-coated, LA/LF-coated or GA/LF-coated nanocapsules)

DEN-induced HCC in a rat model

Antiangiogenic activity

Apoptosis induction

Liver weight reduction

↓ NF-kB and TNF-α mRNA expression

↓ VEGF and Ki67 protein expression

­ ↑ Caspase-3 protein expression

↓ ALT levels by LF-coated nanocapsules

↓ ALT, AST and RLW levels by LA/LF-coated and GA/LF-coated nanoparticles

Improved histological features

Wang, 2016 [57]

Co-encapsulated quercetin and sorafenib (RGD-modified lipid-coated nanoparticles)

HepG2 xenograft mouse model

Tumor progression suppression

↓ Tumor volume

5-FU: 5-fluorouracil; Akt: protein kinase B; ALT: alanine aminotransferase; AST: aspartate aminotransferase; Bax: Bcl-2-associated X; COX-2: cyclooxygenase-2; DEN: diethylnitrosamine; ERK1/2: extracellular signal-regulated kinase 1/2; GA: glycyrrhetinic acid; HCC: hepatocarcinoma; HK2: hexokinase-2; hTERT: telomerase reverse transcriptase; IkBa: nuclear factor-kB inhibitor a; IKKa: inhibitor of nuclear factor-kB kinase subunit a; LA: lactobionic acid; LC3A: microtubule-associated protein 1A/1B light chain 3A; LC3B: microtubule-associated protein 1A/1B light chain 3B; LF: lactoferrin; mTOR: mammalian target of rapamycin; NF-kB: nuclear factor-kB; p62: sequestosome-1; PCNA: proliferating cell nuclear antigen; PLGA: poly(DL-lactide-co-glycolide); RGD: arginine-glycineaspartic acid; RLW: relative liver weight; TNF-a: tumor necrosis factor-a ; TUNEL: terminal deoxynucleotidyl transferase dUTP nick end labeling; VEGF: vascular endothelial growth factor; ZD55-TRAIL: oncolytic adenovirus expressing tumor necrosis factor-related apoptosis inducing ligand.

 

6. Conclusion

In conclusion, quercetin seems to have a clear antiproliferative and proapoptotic effect against HCC, and likely a modulating role on tumor cell cycle progression which needs to be deeper investigated. This flavonoid seems to have antitumoral efficacy through the alteration of a great variety of cellular processes and signaling pathways, though more studies are required to further elucidate its mechanisms of action against HCC. Arising strategies of combination and drug-delivery systems may improve such cancer inhibition properties and, along with emergent use of quercetin derivatives with anticancer efficacy, let enlarging therapeutic options for HCC patients.

 

The entry is from 10.3390/nu11122875 

References

  1. Fazlullah Khan; Kamal Niaz; Faheem Maqbool; Fatima Ismail Hassan; Mohammad Abdollahi; Kalyan C. Nagulapalli Venkata; Seyed Mohammad Nabavi; Anupam Bishayee; Molecular Targets Underlying the Anticancer Effects of Quercetin: An Update. Nutrients 2016, 8, 529, 10.3390/nu8090529.
  2. Agnes W. Boots; Guido R.M.M. Haenen; Aalt Bast; Health effects of quercetin: From antioxidant to nutraceutical. European Journal of Pharmacology 2008, 585, 325-337, 10.1016/j.ejphar.2008.03.008.
  3. Oboh, G.; Ademosun, A.O.; Ogunsuyi, O.B. Quercetin and its role in chronic diseases. Adv. Exp. Med. Biol.
  4. Elumalai, P.; Lakshmi, S. Role of quercetin benefits in neurodegeneration. Adv. Neurobiol. 2016, 12, 229–245.
  5. Kyuichi Kawabata; Rie Mukai; Akari Ishisaka; Quercetin and related polyphenols: new insights and implications for their bioactivity and bioavailability. Food & Function 2015, 6, 1399-1417, 10.1039/c4fo01178c.
  6. Fatemeh Babaei; Mohammadreza Mirzababaei; Marjan Nassiri‐Asl; Quercetin in Food: Possible Mechanisms of Its Effect on Memory. Journal of Food Science 2018, 83, 2280-2287, 10.1111/1750-3841.14317.
  7. MinKyung Bae; Young-Ki Park; Ji-Young Lee; Food components with antifibrotic activity and implications in prevention of liver disease. The Journal of Nutritional Biochemistry 2018, 55, 1-11, 10.1016/j.jnutbio.2017.11.003.
  8. Alessandra Ferramosca; Mariangela Di Giacomo; Vincenzo Zara; Antioxidant dietary approach in treatment of fatty liver: New insights and updates. World Journal of Gastroenterology 2017, 23, 4146-4157, 10.3748/wjg.v23.i23.4146.
  9. Selvaraj Miltonprabu; Michał Tomczyk; Krystyna Skalicka‐Woźniak; Luca Rastrelli; Maria Daglia; Seyed Mohammad Nabavi; Seyed Moayed Alavian; Seyed Mohammad Nabavi; Hepatoprotective effect of quercetin: From chemistry to medicine. Food and Chemical Toxicology 2017, 108, 365-374, 10.1016/j.fct.2016.08.034.
  10. Xi Li; Qianwen Jin; Qunyan Yao; Beili Xu; Lixin Li; Shuncai Zhang; Chuan-Tao Tu; The Flavonoid Quercetin Ameliorates Liver Inflammation and Fibrosis by Regulating Hepatic Macrophages Activation and Polarization in Mice. Frontiers in Pharmacology 2018, 9, 72, 10.3389/fphar.2018.00072.
  11. Sandra Pisonero-Vaquero; Ángel Martínez-Ferreras; María Victoria García-Mediavilla; Susana Martínez-Flórez; Jose Carlos Fernandez-Checa; Marta Benet; José Luis Olcoz; Ramiro Jover; Javier González-Gallego; Sonia Sánchez-Campos; et al. Quercetin ameliorates dysregulation of lipid metabolism genes via the PI3K/AKT pathway in a diet-induced mouse model of nonalcoholic fatty liver disease. Molecular Nutrition & Food Research 2015, 59, 879-893, 10.1002/mnfr.201400913.
  12. David Porras; Esther Nistal; Susana Martínez-Florez; Sandra Pisonero-Vaquero; José Luis Olcoz; Ramiro Jover; Javier González-Gallego; María Victoria García-Mediavilla; Sonia Sánchez-Campos; Protective effect of quercetin on high-fat diet-induced non-alcoholic fatty liver disease in mice is mediated by modulating intestinal microbiota imbalance and related gut-liver axis activation. Free Radical Biology and Medicine 2017, 102, 188-202, 10.1016/j.freeradbiomed.2016.11.037.
  13. Javier González-Gallego; María J. Cuevas; Rafael Vercelino; María J. Tuñón; Norma P. Marroni; Javier González-Gallego; Quercetin Administration Ameliorates Pulmonary Complications of Cirrhosis in Rats. The Journal of Nutrition 2009, 139, 1339-1346, 10.3945/jn.109.105353.
  14. Sael Casas-Grajales; Luis F. Vázquez-Flores; Erika Ramos-Tovar; Erika Hernández-Aquino; Rosa E. Flores-Beltrán; Carlos M. Cerda-García-Rojas; Javier Camacho; Mineko Shibayama; Víctor Tsutsumi; Pablo Muriel; et al. Quercetin reverses experimental cirrhosis by immunomodulation of the proinflammatory and profibrotic processes. Fundamental & Clinical Pharmacology 2017, 31, 610-624, 10.1111/fcp.12315.
  15. A. Forner; María Reig; Jordi Bruix; Hepatocellular carcinoma. The Lancet 2018, 391, 1301-1314, 10.1016/s0140-6736(18)30010-2.
  16. Flavia Fondevila; Carolina Méndez-Blanco; Paula Fernández-Palanca; Javier González-Gallego; José Luis Mauriz; Anti-tumoral activity of single and combined regorafenib treatments in preclinical models of liver and gastrointestinal cancers. Experimental & Molecular Medicine 2019, 51, 1-15, 10.1038/s12276-019-0308-1.
  17. Carolina Méndez-Blanco; Flavia Fondevila; Andrés García-Palomo; Javier González-Gallego; José Luis Mauriz; Sorafenib resistance in hepatocarcinoma: role of hypoxia-inducible factors. Experimental & Molecular Medicine 2018, 50, 1-9, 10.1038/s12276-018-0159-1.
  18. Divya Rawat; Somi Shrivastava; Rayees Ahmad Naik; Saurabh Kumar Chhonker; Aditi Mehrotra; Raj Kumar Koiri; An Overview of Natural Plant Products in the Treatment of Hepatocellular Carcinoma. Anti-Cancer Agents in Medicinal Chemistry 2019, 18, 1838-1859, 10.2174/1871520618666180604085612.
  19. Néstor Prieto-Domínguez; Raquel Ordóñez; Anna Fernández; Carolina Méndez-Blanco; Anna Baulies; Carmen Garcia-Ruiz; José C. Fernández-Checa; José L. Mauriz; Javier González-Gallego; Melatonin-induced increase in sensitivity of human hepatocellular carcinoma cells to sorafenib is associated with reactive oxygen species production and mitophagy. Journal of Pineal Research 2016, 61, 396-407, 10.1111/jpi.12358.
  20. Néstor Prieto-Domínguez; Carolina Méndez-Blanco; Sara Carbajo-Pescador; Flavia Fondevila; Andrés García-Palomo; Javier González-Gallego; José Luis Mauriz; Melatonin enhances sorafenib actions in human hepatocarcinoma cells by inhibiting mTORC1/p70S6K/HIF-1α and hypoxia-mediated mitophagy. Oncotarget 2017, 8, 91402-91414, 10.18632/oncotarget.20592.
  21. Liwei Wu; Jingjing Li; Tong Liu; Sainan Li; Jiao Feng; Qiang Yu; Jie Zhang; Jiaojiao Chen; Yuting Zhou; Jie Ji; et al.Kan ChenYuqing MaoFan WangWeiqi DaiXiaoMing FanJianye WuChuanyong Guo Quercetin shows anti‐tumor effect in hepatocellular carcinoma LM3 cells by abrogating JAK2/STAT3 signaling pathway. Cancer Medicine 2019, 8, 4806-4820, 10.1002/cam4.2388.
  22. Hongyan Wu; Lanlan Pan; Cuixiang Gao; Hongtao Xu; Yanping Li; Lihu Zhang; Linwei Ma; Li Meng; Xiu‐Lan Sun; Hongbing Qin; et al. Quercetin Inhibits the Proliferation of Glycolysis-Addicted HCC Cells by Reducing Hexokinase 2 and Akt-mTOR Pathway. Molecules 2019, 24, 1993, 10.3390/molecules24101993.
  23. Yi Ji; Liu Li; Yan-Xia Ma; Wen-Ting Li; Heng-Zhou Zhu; Mian-Hua Wu; Jin-Rong Zhou; Quercetin inhibits growth of hepatocellular carcinoma by apoptosis induction in part via autophagy stimulation in mice. The Journal of Nutritional Biochemistry 2019, 69, 108-119, 10.1016/j.jnutbio.2019.03.018.
  24. Ji-Sook Jeon; Sora Kwon; Kiwon Ban; Young- Kwon Hong; Curie Ahn; Jung-Suk Sung; Inho Choi; Regulation of the Intracellular ROS Level Is Critical for the Antiproliferative Effect of Quercetin in the Hepatocellular Carcinoma Cell Line HepG2. Nutrition and Cancer 2019, 71, 861-869, 10.1080/01635581.2018.1559929.
  25. Zhaolin Chen; Cheng Huang; Taotao Ma; Ling Jiang; Liqin Tang; Tianlu Shi; Shantang Zhang; Lei Zhang; Pengli Zhu; Jun Li; et al.Aizong Shen Reversal effect of quercetin on multidrug resistance via FZD7/β-catenin pathway in hepatocellular carcinoma cells. Phytomedicine 2018, 43, 37-45, 10.1016/j.phymed.2018.03.040.
  26. Youming Ding; Xiaoyan Chen; Bin Wang; Bin Yu; Jianhui Ge; Xiaokang Shi; Yu Bin; Quercetin suppresses the chymotrypsin-like activity of proteasome via inhibition of MEK1/ERK1/2 signaling pathway in hepatocellular carcinoma HepG2 cells. Canadian Journal of Physiology and Pharmacology 2018, 96, 521-526, 10.1139/cjpp-2017-0655.
  27. Jiang Pi; Baole Li; LvYing Tu; Haiyan Zhu; Hua Jin; Fen Yang; Haihua Bai; Huaihong Cai; Jiye Cai; Investigation of quercetin-induced HepG2 cell apoptosis-associated cellular biophysical alterations by atomic force microscopy. Scanning 2015, 38, 100-112, 10.1002/sca.21245.
  28. Akhilendra Kumar Maurya; Manjula Vinayak; Anticarcinogenic action of quercetin by downregulation of phosphatidylinositol 3-kinase (PI3K) and protein kinase C (PKC) via induction of p53 in hepatocellular carcinoma (HepG2) cell line. Molecular Biology Reports 2015, 42, 1419-1429, 10.1007/s11033-015-3921-7.
  29. Qiang Zhang; Guangdong Cheng; HongBin Qiu; Liling Zhu; Zhongjuan Ren; Wei Zhao; Tao Zhang; Lei Liu; The p53-inducible gene 3 involved in flavonoid-induced cytotoxicity through the reactive oxygen species-mediated mitochondrial apoptotic pathway in human hepatoma cells. Food & Function 2015, 6, 1518-1525, 10.1039/c5fo00142k.
  30. Ra Ham Lee; Jin Hyoung Cho; Young-Joo Jeon; Woong Bang; Jung-Jae Cho; Nag-Jin Choi; Kang Seok Seo; Jung-Hyun Shim; Jung-Il Chae; Quercetin Induces Antiproliferative Activity Against Human Hepatocellular Carcinoma (HepG2) Cells by Suppressing Specificity Protein 1 (Sp1). Drug Development Research 2015, 76, 9-16, 10.1002/ddr.21235.
  31. Katarína Kozics; Z. Valovicova; D. Slamenova; Structure of flavonoids influences the degree inhibition of Benzo(a)pyrene - induced DNA damage and micronuclei in HepG2 cells.. Neoplasma 2011, 58, 516-524, 10.4149/neo_2011_06_516.
  32. Ana Belén Granado-Serrano; María Angeles Martín; Laura Bravo Clemente; Luis Goya; Sonia Ramos; Quercetin Modulates NF-κ B and AP-1/JNK Pathways to Induce Cell Death in Human Hepatoma Cells. Nutrition and Cancer 2010, 62, 390-401, 10.1080/01635580903441196.
  33. Abdulmajeed A. Bahman; Mohamed Salah I. Abaza; Sarah I. Khoushiash; Rajaa J. Al‑Attiyah; Sequence‑dependent effect of sorafenib in combination with natural phenolic compounds on hepatic cancer cells and the possible mechanism of action. International Journal of Molecular Medicine 2018, 42, 1695-1715, 10.3892/ijmm.2018.3725.
  34. Ighodaro Igbe; Xiao-Fei Shen; Wei Jiao; Zhe Qiang; Teng Deng; Sheng Li; Wanli Liu; Han-Wei Liu; Guo-Lin Zhang; Fei Wang; et al. Dietary quercetin potentiates the antiproliferative effect of interferon-α in hepatocellular carcinoma cells through activation of JAK/STAT pathway signaling by inhibition of SHP2 phosphatase. Oncotarget 2017, 8, 113734-113748, 10.18632/oncotarget.22556.
  35. Gabriela Carrasco-Torres; Rafael Baltiérrez-Hoyos; Erik Andrade-Jorge; Saúl Villa-Treviño; José G. Trujillo-Ferrara; Verónica R. Vásquez-Garzón; Cytotoxicity, Oxidative Stress, Cell Cycle Arrest, and Mitochondrial Apoptosis after Combined Treatment of Hepatocarcinoma Cells with Maleic Anhydride Derivatives and Quercetin. Oxidative Medicine and Cellular Longevity 2017, 2017, 1-16, 10.1155/2017/2734976.
  36. Ana Filipa Brito; Marina Ribeiro; Ana Margarida Abrantes; Ana Catarina Mamede; Mafalda Laranjo; João Eduardo Casalta-Lopes; Ana Cristina Gonçalves; Ana Bela Sarmento-Ribeiro; José Guilherme Tralhão; Maria Filomena Botelho; et al. New Approach for Treatment of Primary Liver Tumors: The Role of Quercetin. Nutrition and Cancer 2016, 68, 250-266, 10.1080/01635581.2016.1145245.
  37. Wei Dai; Quangen Gao; Jianping Qiu; Jianmao Yuan; Guoliang Wu; Genhai Shen; Quercetin induces apoptosis and enhances 5-FU therapeutic efficacy in hepatocellular carcinoma. Tumor Biology 2015, 37, 6307-6313, 10.1007/s13277-015-4501-0.
  38. Ji-Ling Zhao; Jing Zhao; Hong-Jun Jiao; Synergistic Growth-Suppressive Effects of Quercetin and Cisplatin on HepG2 Human Hepatocellular Carcinoma Cells. Applied Biochemistry and Biotechnology 2013, 172, 784-791, 10.1007/s12010-013-0561-z.
  39. Aanchal Sharma; Manoj Kumar Bhat; Enhancement of Carboplatin- and Quercetin-Induced Cell Death by Roscovitine Is Akt Dependent and p53 Independent in Hepatoma Cells. Integrative Cancer Therapies 2011, 10, NP4-NP14, 10.1177/1534735411423922.
  40. Yu, C.-P.; Qiu, R.-G.; Shi, L.; Liang, J.; Celecoxib and quercetin induce apoptosis in human hepatocarcinoma. Biomedical Research 2017, 28, 3465-3470.
  41. Michael J. Houghton; Asimina Kerimi; Sarka Tumova; John P. Boyle; Gary Williamson; Quercetin preserves redox status and stimulates mitochondrial function in metabolically-stressed HepG2 cells. Free Radical Biology and Medicine 2018, 129, 296-309, 10.1016/j.freeradbiomed.2018.09.037.
  42. Mona A. Abdelmoneem; Manar A. Elnaggar; Ruwan S. Hammady; Shaza M. Kamel; Maged W. Helmy; Mohammad A. Abdulkader; Amira Zaky; Jia-You Fang; Kadria A. Elkhodairy; Ahmed O. Elzoghby; et al. Dual-Targeted Lactoferrin Shell-Oily Core Nanocapsules for Synergistic Targeted/Herbal Therapy of Hepatocellular Carcinoma. ACS Applied Materials & Interfaces 2019, 11, 26731-26744, 10.1021/acsami.9b10164.
  43. Yoo-Jung Lee; Song-Yi Beak; Inho Choi; Jung-Suk Sung; Quercetin and its metabolites protect hepatocytes against ethanol-induced oxidative stress by activation of Nrf2 and AP-1. Food Science and Biotechnology 2017, 27, 809-817, 10.1007/s10068-017-0287-8.
  44. Mary E. Kellett; Phillip Greenspan; Ronald B. Pegg; Modification of the cellular antioxidant activity (CAA) assay to study phenolic antioxidants in a Caco-2 cell line. Food Chemistry 2018, 244, 359-363, 10.1016/j.foodchem.2017.10.035.
  45. Yasmin M. Shaalan; H. Handoussa; R. A. Youness; R. A. Assal; A. H. El-Khatib; M. W. Linscheid; H. M. El Tayebi; A. I. Abdelaziz; Destabilizing the interplay between miR-1275 and IGF2BPs by Tamarix articulata and quercetin in hepatocellular carcinoma. Natural Product Research 2017, 32, 2217-2220, 10.1080/14786419.2017.1366478.
  46. Husam Dabbagh-Bazarbachi; Gael Clergeaud; Isabel M. Quesada; Mayreli Ortiz; Ciara K. O’Sullivan; Juan B. Fernández-Larrea; Zinc Ionophore Activity of Quercetin and Epigallocatechin-gallate: From Hepa 1-6 Cells to a Liposome Model. Journal of Agricultural and Food Chemistry 2014, 62, 8085-8093, 10.1021/jf5014633.
  47. Joan Oliva; Fawzia Bardag-Gorce; Brittany Tillman; Samuel French; Protective effect of quercetin, EGCG, catechin and betaine against oxidative stress induced by ethanol in vitro. Experimental and Molecular Pathology 2011, 90, 295-299, 10.1016/j.yexmp.2011.02.006.
  48. Kyung Cheol Choi; Won Tae Chung; Jung Kee Kwon; Jin Young Yu; Yang Soo Jang; S.-M. Park; Sung Yong Lee; J.-C. Lee; Inhibitory effects of quercetin on aflatoxin B1-induced hepatic damage in mice. Food and Chemical Toxicology 2010, 48, 2747-2753, 10.1016/j.fct.2010.07.001.
  49. Ke-Wei Ren; Ya-Hua Li; Gang Wu; Jian-Zhuang Ren; Hui-Bin Lu; Zong-Ming Li; Xin-Wei Han; Quercetin nanoparticles display antitumor activity via proliferation inhibition and apoptosis induction in liver cancer cells. International Journal of Oncology 2017, 50, 1299-1311, 10.3892/ijo.2017.3886.
  50. Khaled AbouAitah; Anna Swiderska-Sroda; Ahmed A. Farghali; Jacek Wojnarowicz; Agata Stefanek; Stanislaw Gierlotka; Agnieszka Opalinska; Abdou K. Allayeh; Tomasz Ciach; Witold Lojkowski; et al. Folic acid-conjugated mesoporous silica particles as nanocarriers of natural prodrugs for cancer targeting and antioxidant action. Oncotarget 2018, 9, 26466-26490, 10.18632/oncotarget.25470.
  51. Kausik Bishayee; Anisur Rahman Khuda-Bukhsh; And Sung-Oh Huh; PLGA-Loaded Gold-Nanoparticles Precipitated with Quercetin Downregulate HDAC-Akt Activities Controlling Proliferation and Activate p53-ROS Crosstalk to Induce Apoptosis in Hepatocarcinoma Cells. Molecules and Cells 2015, 38, 518-527, 10.14348/molcells.2015.2339.
  52. Ahmed A. Abd-Rabou; Hanaa H. Ahmed; CS-PEG decorated PLGA nano-prototype for delivery of bioactive compounds: A novel approach for induction of apoptosis in HepG2 cell line. Advances in Medical Sciences 2017, 62, 357-367, 10.1016/j.advms.2017.01.003.
  53. Korawith Srisa-Nga; Samlee Mankhetkorn; Siriporn Okonogi; Ruttiros Khonkarn; Delivery of Superparamagnetic Polymeric Micelles Loaded With Quercetin to Hepatocellular Carcinoma Cells.. Journal of Pharmaceutical Sciences 2018, 108, 996-1006, 10.1016/j.xphs.2018.08.008.
  54. Jaleh Varshosaz; Abbas Jafarian; Golnaz Salehi; Behzad Zolfaghari; Comparing different sterol containing solid lipid nanoparticles for targeted delivery of quercetin in hepatocellular carcinoma. Journal of Liposome Research 2013, 24, 191-203, 10.3109/08982104.2013.868476.
  55. Kristina Kovacovicova; Marianna Skolnaja; Mihkel Heinmaa; Martin Mistrik; Pille Pata; Illar Pata; Jiri Bartek; Manlio Vinciguerra; Senolytic Cocktail Dasatinib+Quercetin (D+Q) Does Not Enhance the Efficacy of Senescence-Inducing Chemotherapy in Liver Cancer. Frontiers in Oncology 2018, 8, 459, 10.3389/fonc.2018.00459.
  56. Hai Zou; Yong-Fa Zheng; Wei Ge; Shi-Bing Wang; Xiao-Zhou Mou; Synergistic Anti-tumour Effects of Quercetin and Oncolytic Adenovirus expressing TRAIL in Human Hepatocellular Carcinoma.. Scientific Reports 2018, 8, 2182, 10.1038/s41598-018-20213-7.
  57. Can Wang; Liang Su; Chengsheng Wu; Jianlin Wu; Chengbao Zhu; Guangying Yuan; RGD peptide targeted lipid-coated nanoparticles for combinatorial delivery of sorafenib and quercetin against hepatocellular carcinoma. Drug Development and Industrial Pharmacy 2016, 42, 1938-1944, 10.1080/03639045.2016.1185435.
  58. Sudhanshu Sudan; H. P. Vasantha Rupasinghe; Quercetin-3-O-glucoside induces human DNA topoisomerase II inhibition, cell cycle arrest and apoptosis in hepatocellular carcinoma cells.. Anticancer Research 2014, 34, 1691-1700.
  59. Yujia Liu; Ayuki Kurita; Sayaka Nakashima; Beiwei Zhu; Shintaro Munemasa; Toshiyuki Nakamura; Yoshiyuki Murata; Yoshimasa Nakamura; 3,4-Dihydroxyphenylacetic acid is a potential aldehyde dehydrogenase inducer in murine hepatoma Hepa1c1c7 cells. Bioscience, Biotechnology, and Biochemistry 2017, 81, 1978-1983, 10.1080/09168451.2017.1361809.
  60. Sudhanshu Sudan; H. P. Vasantha Rupasinghe; Antiproliferative activity of long chain acylated esters of quercetin-3-O-glucoside in hepatocellular carcinoma HepG2 cells. Experimental Biology and Medicine 2015, 240, 1452-1464, 10.1177/1535370215570828.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , ,
View Times: 731
Revisions: 2 times (View History)
Update Date: 12 Oct 2021
1000/1000