Herbal drug‑based nanotherapy for hepatocellular carcinoma: Quercetin‑contained nanocarrier as a multipurpose therapeutic agent against hepatocellular carcinoma (Review)
- Authors:
- Published online on: December 9, 2024 https://doi.org/10.3892/br.2024.1907
- Article Number: 29
-
Copyright: © Batsukh et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
1. Introduction
Cancer remains a leading cause of morbidity and mortality globally, with liver cancer, particularly hepatocellular carcinoma (HCC), accounting for significant incidence and mortality rates. The Global Cancer Observatory (GLOBOCAN) reported 19.96 million new cancer cases and 9.74 million cancer-related mortalities in 2022, highlighting the urgent need for improved early diagnosis, treatment and prevention strategies (1,2).
HCC is the most prevalent form of primary liver cancer, representing ~75% of cases. Current chemotherapeutic agents, such as gemcitabine and cisplatin, show limited efficacy due to the chemoresistance from the cancer, often linked to overexpression of ATP-binding cassette transporter proteins and detoxification enzymes in liver cells. This resistance contributes to low response rates and severe adverse effects from traditional chemotherapy, including hair loss, nausea and fatigue (3-6). These are mainly being explained with non-targeted chemotherapeutics. Hence, reducing the chemotherapeutics-related adverse side effects has emerged as a crucial issue to improve the anti-cancer therapeutic efficacy.
At present, pharmaceutical engineering in the field of medicine has emerged to develop new tumor-targeted drug delivery systems and multi-purpose drug delivery systems to enhance the therapeutic efficacy of chemotherapy against cancer (7,8). As regards the tumor-specific drug delivery systems, a number of attempts have been tried to create near perfect nanocarriers against cancer, which may have some specific capabilities such as tumor targeted without nonspecific cellular invasion, tumor sensitive without adverse side effects and ability to moderate multidrug resistance mechanisms (9,10).
Traditional herbal medicine has been recognized as one of important treatments against liver cancer for years. So far, a number of herbal plants such as Cinnamomum species (11), Curcuma species (12), Artemisia species (13), Glycyrrhiza uralensis (14), Equisetum arvense (15), Rheum undulatum (16) and Scutellaria baicalensis (17) have been shown to have anti-proliferative effects on HCC. However, traditional herbal medicine has some drawbacks, including direct anti-proliferative effects on normal and cancer cells and producing herbal medicine-related adverse side effects such as allergic reactions, asthmas, rashes, vomiting and diarrhea. In recent years, traditional herbal medicine has been intensively studied to investigate its anticancer activity in the field of cancer pharmacology. It has been considered to develop new drug delivery systems to improve traditional herbal medicine anticancer efficacy. A number of biologically active substances such as quercetin (18) and sesquiterpene lactone (19) from traditional herbal medicine have been extensively researched.
The present study aimed to describe the anticancer mechanisms and inhibition capacity against multidrug resistance mechanism of quercetin-containing multipurpose nanocarriers and their targeting potential for HCC.
2. Quercetin
A number of medicinal plants, vegetables and fruits, including Moringa oleifera, Ficus religuisa, Styphnolobium japonicum (20), Polygonum hydropiper (21), Polygonum aviculare (22), citrus, apple, broccoli, onions and chili pepper, produce abundant amount of quercetin as a metabolite in their life cycle, and it plays important roles in some physiological processes, growth and developmental stages such as photosynthesis, antioxidant system and seed germination (23). Quercetin is a flavonoid compound with a chemical structure of 3, 3', 4', 5, 7-pentahydroxyflavone (24). In recent years, quercetin has repeatedly been declared as an effective agent against cancer, an effective inhibitor of P-glycoprotein (P-gp) and well-known protector of chemotherapy-exposed cells (25). The quercetin effects on numerous types of cancers are associated with several mechanisms such as anti-proliferative effect, inhibition of angiogenesis, reactive oxygen species (ROS) induction in cancerous cells and apoptosis induction.
3. Quercetin-related mechanisms against HCC
Effects on cell growth and proliferation
A number of studies have attempted to explain the anticancer mechanism of quercetin against HCC. Anticancer chemotherapeutics have mainly been designed to inhibit the cancer cell cycle in their various stages (26). Cell growth and proliferation are biologically explained by cell cycle staged with growth 1 (G1), synthesis (S), growth 2 (G2) and mitosis (M), which are regulated via cyclin-dependent kinases (CDK/cyclin) and signal transduction pathways. The quercetin mechanism is related to the downregulation of cyclin B1, D, CDK1 and CDK2 by activating protein 27 (p27) and protein 21, which triggers cell cycle arrest in M, G1, S and G2 phases (Fig. 1A) (27,28).
Quercetin-related mechanisms against HCC have been reported to be primarily associated with downregulation of Wnt/β-catenin signaling pathway (Fig. 1B), enhancing Bcl-2-associated X protein (BAX), caspase family, extracellular signal-regulated kinase (ERK) and AMP-activated protein kinase/mammalian target of rapamycin pathway, suppressing B-cell lymphoma-2 (Bcl-2), epidermal growth factor receptor, dihydrotestosterone and phosphatidylinositol 3-kinase/protein kinase B pathway (Fig. 1C) (29-31). Moreover, it is reported that mitogen-activated protein kinase pathway and ataxia telangiectasia mutated kinase-mediated signaling pathway are induced due to quercetin exposure (Fig. 1D) (32,33).
Effects on apoptosis induction
A potential role of quercetin has been reported in the regulation and induction of apoptosis in cancer cells. It is known that quercetin has the proapoptotic effect which contributes to the upregulation of tumor protein 53 gene (34,35) and suppression of Bcl-2 protein (Fig. 2A). Moreover, quercetin has a potential to increase the expression of proapoptotic proteins such as Bax and cleaved caspase-3 and 9, which regulate negatively antiapoptotic proteins such as Bcl-2 and myeloid leukemia 1 (36-38). This apoptotic pathway highlights the potential of quercetin as a therapeutic agent in cancer treatment by promoting the elimination of malignant cells through the activation of intrinsic apoptotic mechanisms.
Effects on oxidative stress
Another mechanism of action of quercetin against HCC is inhibition of oxidative stress which plays a significant role in fighting cancer cells (Fig. 2B). The mechanism of the quercetin is mainly attributed to the upregulated expression of the antioxidant response element pathway, nuclear factor (erythroid-derived 2) gene and thioredoxin system. The upregulation of these mechanisms is triggered by quercetin radicals-mediated pro-oxidative state due to peroxidase-catalyzed oxidation in the cytoplasm (39-41).
Quercetin can reduce the ROS level in the cytoplasm by inhibiting the p66hc-mediated Ras-related C3 botulinum toxin substrate 1 activation to decrease the cell migration and proliferation (42).
Quercetin can undergo oxidation, leading to formation of reactive intermediates, specifically quercetin-semiquinone and quercetin-quinone. These radicals can interact with cellular components, contributing to an increase in overall ROS levels within cancer cells (43).
Notably, it has been reported that quercetin generates ROS in HepG2 by overexpressing the p53-indcible gene 3 and its product of oxidoreductase. The oxidoreductase enzyme catalyzes the quercetin and generate ROS with HepG2 cell lines (44).
4. Angiogenesis-related inhibition
The progression stage of HCC is also associated with the angiogenesis and metastasis. Thus, the angiogenesis inhibition may be a targeting option for HCC treatment. The mechanism of quercetin against angiogenesis is related to the vascular endothelial growth factor receptor (VEGF-R) 2 targeting, which regulates AKT/mTOR/P70S6K signaling pathway (Fig. 2C) (45). Igura et al (46) first reported in 2001 that quercetin inhibited angiogenesis and then could be used as an efficient antitumor drug (47).
5. Quercetin nanocarriers for HCC
In recent years, several nanocarriers for quercetin have been designed to use its benefits for HCC treatment (Table SI).
Ren et al (48) synthesized quercetin nanoparticles by using gold nanoparticles, quercetin solution and poly (DL-lactide-co-glycolide) PLGA to treat liver cancer. As a result, the quercetin nanoparticles efficiently suppressed liver cancer cell growth and colony formation by upregulating p27 and downregulating c-Myc, cyclin-D1, CDK1, matrix metallopeptidase 7 and β-catenin within the cancer cells. Another possible mechanism was also revealed in that the quercetin nanoparticle markedly induced the apoptosis of liver cancer cells via activating cytochrome c/caspase signaling. The Akt/ERK1/2 signaling pathway was also inactivated by the quercetin nanoparticles, which suppressed liver cancer cell growth. It was also reported that the quercetin nanoparticles suppressed the transcriptional factor AP-2 alpha/human telomerase reverse transcriptase signaling pathway within the liver cancer cells. These results showed that the quercetin nanoparticles can be a multipurpose nanocarrier against liver cancer cells (48).
Guan et al (49) designed a quercetin-loaded nanoparticle by using poly (lactic-co-glycolic acid)-d-α-tocopheryl polyethylene glycol 1000 succinate (PLGA and TPGS1000) for the targeted treatment of liver cancer. Their research results proved that the quercetin by liver cancer cell uptake can be increased by using PLGA and TPGS1000 polymers and the cytotoxicity of liver cancer was efficiently increased by the designed nanoparticles. Also, they observed that quercetin nanoparticles markedly promoted cell apoptosis. The study suggested that quercetin nanoparticles had a higher suppressive effect on tumor growth by inducing the cell apoptosis and can be a potential tool to treat liver cancer.
Quercetin-loaded superparamagnetic polymeric micelles were prepared by a film hydration method to enhance the treatment efficacy against HCC and improve the disease monitoring. The designed quercetin-loaded micelles increased the cytotoxicity and apoptosis for the liver cancer cells. The main mechanism of action of the quercetin-loaded superparamagnetic polymeric micelles was explained by the higher accumulation of the quercetin nanoparticles within the targeted liver cancer cells due to the extrinsic magnetic field. Hence, Srisa-nga et al (50) suggested that co-delivery of quercetin and superparamagnetic iron oxide nanoparticles may increase the therapeutic efficacy against HCC and decrease the side effects-related to the chemotherapeutics.
A targeted lipid coated nanoparticle for hepatocellular carcinoma treatment was designed by using RGD peptide to conduct a co-delivery of sorafenib and quercetin (51). The RGD peptide has extensively applied as a targeting agent for cancer treatment due to its capability of targeting the tumor-associated blood vessels. As a result of the study, the liver cancer cell uptake of nanoparticles was increased up to 71.5% and the cytotoxicity was detected higher than control groups. Thus, it was suggested that RGD peptide modification may be more significant for liver cancer targeting and the combination of quercetin and sorafenib was more effective against HCC. Sorafenib is recognized as an effective multikinase inhibitor which effectively suppresses tumor cell proliferation and growth by downregulating VEGFR and platelet-derived growth factor receptor signaling pathways (52). It has now been reported that HCC cells are becoming resistant for sorafenib (53). Therefore, the sorafenib combination therapy with quercetin and an active targeting agent might be a more powerful therapeutic tool against HCC.
Quercetin encapsulated biodegradable plasmonic nanoparticles were obtained by preparing liposomes and loading quercetin into the liposomes. Gold coating was conducted to obtain the gold-coated liposomes. As a consequence of the photothermal therapy, the obtained quercetin-loaded and gold-coated liposomes had an efficient effect in inducing the apoptotic death of cancer cells by suppressing heat shock protein 70 (Hsp70) expression and also had a marked effect to trigger the disorganization of the microtubules network and DNA damage. A combination therapy of quercetin and photothermal therapy was a significant against cancer (54).
Varshosaz et al (55) fabricated a solid lipid nanoparticle with loaded quercetin and checked the cellular uptake of the nanoparticles. HepG2 cells markedly internalized the quercetin-loaded solid lipid nanoparticles compared with control cells. The viability of HepG2 cells was also significantly decreased. The research team suggested that all sterol-contained solid lipid nanoparticles had good inhibitory effects for cancer cells.
6. Quercetin effect on reversing multidrug resistance for anticancer treatment
Although anticancer drugs have efficient therapeutic effects by suppressing the growth or killing the cancer cells, a number of drawbacks or adverse effects triggered by the anticancer drugs have been reported and cause treatment failure.
Anticancer drug resistance in HCC is one of critical issues in clinical medicine for several reasons, such as limited treatment options and mechanisms of anticancer drug resistance. HCC has historically had limited treatment options, including surgery, locoregional and systemic therapies (56). Main chemotherapeutics against liver cancer are gemcitabine, cisplatin, doxorubicin, etoposide and methotrexate and their treatment response rate is comparably low due to drug resistance mechanisms (5,57,58). Anticancer drug resistance leads to treatment failure and therefore is becoming imperative to discuss and address. HCC can exhibit various mechanisms of resistance to anticancer drugs, including alterations in drug metabolism, changes in tumor microenvironment and genetic mutations in genes which encodes the transporter proteins and detoxification enzymes (59).
During chemotherapy, the cancer cells may gain the capability to reduce the efficacy of chemotherapeutics by overexpressing P-gp and other proteins on the membrane of the cancer cells (Fig. 3) (60-62).
One of frequent causes to trigger the multidrug resistance of the anticancer drugs is P-gp (MDR1) which is encoded by the ATP-binding cassette sub family B member 1 (ABCB1) gene (63) and has the ability to efflux the anticancer drugs from the tumor cells. To reverse the P-gp-associated multidrug resistance of the cancer cells, a number of attempts have been conducted in last two decades and several inhibitors of the P-gp efflux pump discovered, such as amiodarone, verapamil, diosmin sesquiterpene, quercetin, naringin, biochanin and silymarin (64-68).
Multidrug resistance-associated protein 2 (MRP2) is another efflux transporter, primarily in the removal of organic anions and drug conjugates from cells. It is responsible for anticancer drug resistance (69). Quercetin has been shown to modulate MRP2 activity, especially suppressing the efflux mechanism of the transporter in cancer cells. Therefore, it leads to an increased accumulation of anticancer drugs inside cancer cells, which could enhance their anticancer effects (69).
Glutathione S-transferase (GST) is a family of enzymes involved in the detoxification process of chemical compounds. The enzyme binds to drugs and xenobiotics via glutathione and facilitates drug excretion. Quercetin may suppress the expression and activity of the enzyme, which can decrease the detoxification processes of chemotherapeutics and their efficacy (70). The mechanism of quercetin on GST enzyme may be attributed to suppressing GST activation through competitive or non-competitive inhibition (71).
Cytochrome P450 3A4 (CYP3A4) is a major enzyme in the liver which metabolizes numerous drugs, including a number of chemotherapeutics. It has been reported that quercetin can suppress its anticancer drug efficacy (72).
Among the inhibitors of P-gp, quercetin has attracted more attention from researchers to suppress the growth of cancer cells by blocking the cellular signaling pathway and reverse the multidrug resistance during the chemotherapy (73). The mechanism of the action of quercetin to inhibit P-gp is related to blocking the function of P-gp efflux pump by binding the interfaces of intracellular helices and nucleotide-binding domains (74).
7. Quercetin nanocarriers for P-gp inhibition
So far, quercetin has been used as a promising inhibitor for the design of nanocarriers with the ability to reduce anticancer drug resistance in HCC treatment (Table SII).
In a study, quercetin-chitosan micelles were prepared by the ultrasound method and doxorubicin was entrapped into the micelles. Then, in vitro cytotoxicity, cellular uptake and inhibition of P-gp efflux were tested for the DOX-entrapped quercetin-chitosan micelles. Quercetin and quercetin-chitosan micelles had comparably effective inhibition of the P-gp efflux pump and produced a marked long-lasting cytotoxicity on cancer cells. These results suggested that the quercetin-chitosan micelles were a promising multipurpose nanocarrier for oral delivery of anticancer drugs (75).
Guo et al (76) designed a polymeric nanoparticle to attenuate the drug resistance mechanisms during cancer therapy by co-delivering paclitaxel and quercetin. The polymeric nanoparticle was prepared by a sonication method and characterized by checking its cellular uptake, in vivo biodistribution and antitumor efficacy. P-gp inhibition assay for the nanoparticle was conducted to evaluate its effect on drug resistance mechanism of anticancer drugs. As a result of the polymeric nanoparticle, an enhanced tumor-suppressing effect was observed due to decrease of P-gp expression in the cancer cells.
In another study, a polyethylene glycolated liposome was developed to conduct a codelivery of adriamycin and quercetin to reverse cancer therapy-related drug resistance. Adriamycin is known to be a substrate of P-gp and effluxes from the tumor cells during anticancer therapy. In the study, film-ultrasound technique with ammonium sulfate transmembrane gradient method was used to prepare the adriamycin and quercetin co-loaded liposome and pharmacokinetic study, cell toxicity study and pharmacodynamic study were also conducted. The relative tumor volume was decreased and P-gp was effectively suppressed (77).
An amphiphilic carboxymethyl chitosan-quercetin conjugate has been obtained by using a self-assembly technique to improve the oral delivery of paclitaxel. Paclitaxel is known as a substrate of P-gp and therefore leads to therapeutic failure due to chemotherapeutic efflux mechanism in the tumor cells. For this purpose, quercetin was applied as an inhibitor of P-gp to increase the capability of therapeutic efficacy of the drug. In this study, the antitumor efficacy of the nanocarrier was investigated in in vivo xenograft model and the inhibition of P-gp was evaluated by western blot analysis. As a result, paclitaxel-loaded carboxymethyl chitosan-quercetin polymeric micelles displayed an improved downregulation effect on P-gp (78).
Another team developed quercetin-loaded benzoylated methoxy-poly (ethylene glycol)-b-oligo (ε-caprolactone) polymeric micelles to overcome the drug resistance issues for cancer therapy. They used P-gp overexpressed leukemia cells (K562/ADR) and checked the effect of quercetin on P-gp function to reverse the multidrug resistance. According to the results, quercetin-loaded micelles were effective in reducing the overexpression of P-gp on K562/ADR cells (79).
Another nanomicelle was reported in 2018. Chitosan, quercetin and citraconic anhydride was used to design a pH-responsive nanomicelle to enhance the intracellular bioavailability of doxorubicin for cancer treatment. In the study, a self-assembly technique was used to obtain the nanomicelle and P-gp inhibition assay was performed for quercetin function on the efflux pump. The nanomicelle had an improved inhibitory effect on the P-gp efflux pump and obvious enhancement of suppressing effect on MCF-7/ADR cells (80).
Multiwalled carbon nanotubes-based nanoconstructs have been designed to enhance the therapeutic efficacy of drugs against multidrug resistant cancers. Using this nanocarrier, a combination of N-desmethyl tamoxifen and quercetin was delivered into MDA-MB-231 cells and indirectly checked the inhibition of P-gp. In this study, an increased biodistribution, biocompatibility and marked cellular uptake of the drug-loaded nanocarriers was observed. These results were explained by quercetin-related P-gp inhibition (81).
Qian et al (82) designed a core-shell micelle to deliver paclitaxel for treatment of multi drug resistant breast cancer. In this study, quercetin was also chosen as an inhibitor of P-gp. The characteristics and antitumor efficacy of the micelles were checked and the effect of quercetin on P-gp efflux pump was evaluated in MDA-MB-231/MDR1 cells. According to the results, the micelles enhanced the cellular uptake of antitumor drug by downregulating P-gp expression in MDA-MB-231/MDR1 cells.
8. Challenges and considerations
Major drawbacks of the anticancer therapeutics such as adverse effects and multidrug resistance cause failure in cancer treatment (83). A number of different strategies have been employed to overcome the drawbacks for effective cancer treatment. Herbal drug-based nanomedicine has recently gained attention for cancer treatment due to its biosafety, biodegradability, promising therapeutic efficacy and ability of combating anticancer drug resistance (84).
The field of pharmaceutical engineering has produced more data to develop a favorable therapeutic tool for cancer therapy (85).
Targeted drug delivery systems, which are methods of delivering drugs to specific parts of a diseased body, have been extensively researched in cancer nanotechnology. They have several benefits as drug delivery systems. They not only significantly improve the specificity of nanocarriers to tumor cells or diseased organs but also reduce the side effects of anticancer drugs in comparison with traditional chemotherapy (86). As for liver targeted delivery, receptor-mediated drug delivery systems such as asialoglycoprotein receptor (ASGPR), folate receptor and retinol binding protein receptor-mediated targeted delivery systems have been widely recognized as the most effective tools in treating HCC (87,88). Hepatic carcinoma cells are able to recognize galactose and N-acetylgalactosamine-terminated glycoproteins through ASGPR located on their surfaces. In recent years, a number of researches have been confirming that galactosylated nanoparticles (NPs) exhibit high hepatocyte specificity in vitro as well as in vivo for anticancer drug delivery (89).
At present, nanomedicines provide a great contribution to delivering high concentrations of chemotherapeutic drugs and MDR inhibitors to tumor cells, in which biodegradable, biocompatible nanomaterials and target-specific ligands play more important roles.
Quercetin is now a well-known anticancer agent with many purposes but it needs further improvement on its pharmacology and clinical applications (90). There are a number of drawbacks of quercetin-related chemotherapy including less absorption in gastrointestinal tract, easy metabolization by enzymes, instability, poor solubility, low hydrophobicity and lack of specificity to the target site (91).
To overcome these challenges, researchers have been developing quercetin-loaded nanocarriers, which can enhance the therapeutic potential of quercetin, especially against drug-resistant HCC. Quercetin-loaded nanocarriers have enhanced bioavailability, targeted delivery and the possibility of overcoming drug resistance. Quercetin-loaded nanocarriers can improve the solubility and stability of quercetin, increasing its bioavailability in the bloodstream and allowing for more effective concentrations to reach tumor sites. Quercetin-loaded nanocarriers can be designed to target cancer cells specifically, minimizing damage to healthy tissues and enhancing the therapeutic effect on HCC cells. This targeting can reduce the overall side effects of treatment. Quercetin has been shown to modulate various pathways involved in drug resistance, such as inhibition of drug efflux pumps, induction of apoptosis and modulation of signaling pathways. As for the synergistic effect of quercetin, it can be used in combination with other anticancer drugs and quercetin can enhance their effectiveness and help overcome resistance mechanisms.
Hence, quercetin-supported nanocarriers with good therapeutic efficacy and cancer-targeting ability, as well as capacity of attenuating multidrug resistance have emerged as a promising approach to enhance the bioavailability and therapeutic efficacy of quercetin for cancer therapy. In further studies, addressing these aforementioned weaknesses of quercetin is now a major challenge to develop an effective nanocarrier with quercetin against HCC.
9. Conclusion
Based on these well-studied considerations, the present review demonstrated that quercetin has multiple antitumor properties. There are some limitations, but advances in pharmaceutical engineering may mitigate these. Consequently, comprehensive research is essential to develop effective nanocarriers for quercetin targeting HCC.
Supplementary Material
Summary of quercetin nanoparticles designed for hepatocellular carcinoma treatment.
Summary of quercetin nanocarriers designed for P-gp inhibition.
Acknowledgements
Not applicable.
Funding
Funding: The present study was supported by an internal grant from Science Technology Foundation, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia (grant no. STF-MNUMS-200138).
Availability of data and materials
Not applicable.
Authors' contributions
AT performed the conception and design of the review article and provided administrative support. TB collected the information and performed the interpretation. The two authors read and approved the final manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Morgan E, Soerjomataram I, Rumgay H, Coleman HG, Thrift AP, Vignat J, Laversanne M, Ferlay J and Arnold M: The global landscape of esophageal squamous cell carcinoma and esophageal adenocarcinoma incidence and mortality in 2020 and projections to 2040: New estimates from GLOBOCAN 2020. Gastroenterology. 163:649–658.e2. 2022.PubMed/NCBI View Article : Google Scholar | |
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024.PubMed/NCBI View Article : Google Scholar | |
Mattiuzzi C and Lippi G: Current cancer epidemiology. J Epidemiol Glob Health. 9:217–222. 2019.PubMed/NCBI View Article : Google Scholar | |
Sayiner M, Golabi P and Younossi ZM: Disease burden of hepatocellular carcinoma: A global Dig Dis. Sci. 64:910–917. 2019.PubMed/NCBI View Article : Google Scholar | |
Joyce H, McCann A, Clynes M and Larkin A: Influence of multidrug resistance and drug transport proteins on chemotherapy drug metabolism. Expert Opin Drug Metab Toxicol. 11:795–809. 2015.PubMed/NCBI View Article : Google Scholar | |
Lundqvist EÅ, Fujiwara K and Seoud M: Principles of chemotherapy. Int J Gynaecol Obstet. 131 (Suppl 2):S146–S149. 2015.PubMed/NCBI View Article : Google Scholar | |
Wu H, Wei M, Xu Y, Li Y, Zhai X, Su P, Ma Q and Zhang H: PDA-based drug delivery nanosystems: A potential approach for glioma treatment. Int J Nanomedicine. 17:3751–3775. 2022.PubMed/NCBI View Article : Google Scholar | |
Ding Y, Li W, Zhang F, Liu Z, Zanjanizadeh Ezazi N, Liu D and Santos HA: Electrospun fibrous architectures for drug delivery, tissue engineering and cancer therapy. Adv Funct Mater. 29(1802852)2019. | |
Ali I, Nadeem Lone M, Suhail M, Danish Mukhtar S and Asnin L: Advances in nanocarriers for anticancer drugs delivery. Curr Med Chem. 23:2159–2187. 2016.PubMed/NCBI View Article : Google Scholar | |
Sun Y, Su J, Liu G, Chen J, Zhang X, Zhang R, Jiang M and Qiu M: Advances of blood cell-based drug delivery systems. Eur J Pharm Sci. 96:115–128. 2017.PubMed/NCBI View Article : Google Scholar | |
Upadhyay RK: Therapeutic and pharmaceutical potential of Cinnamomum tamala. Res Rev Pharm Pharm Sci. 6:18–28. 2017. | |
Sultana S, Munir N, Mahmood Z, Riaz M, Akram M, Rebezov M, Kuderinova N, Moldabayeva Z, Shariati MA, Rauf A and Rengasamy KRR: Molecular targets for the management of cancer using Curcuma longa Linn. phytoconstituents: A review. Biomed Pharmacother. 135(111078)2021.PubMed/NCBI View Article : Google Scholar | |
Taleghani A, Emami SA and Tayarani-Najaran Z: Artemisia: A promising plant for the treatment of cancer. Bioorg Med Chem. 28(115180)2020.PubMed/NCBI View Article : Google Scholar | |
Ji S, Li Z, Song W, Wang Y, Liang W, Li K, Tang S, Wang Q, Qiao X, Zhou D, et al: Bioactive constituents of Glycyrrhiza uralensis (Licorice): Discovery of the effective components of a traditional herbal medicine. J Nat Prod. 79:281–292. 2016.PubMed/NCBI View Article : Google Scholar | |
Makia R, Al-Sammarrae K, Al-Halbosiy M and Al-Mashhadani M: In vitro cytotoxic activity of total flavonoid from Equisetum arvense extract. Rep Biochem Mol Biol. 11:487–492. 2022.PubMed/NCBI View Article : Google Scholar | |
Lee D, Park S, Choi S, Kim SH and Kang KS: In vitro estrogenic and breast cancer inhibitory activities of chemical constituents isolated from Rheum undulatum L. Molecules. 23(1215)2018.PubMed/NCBI View Article : Google Scholar | |
Cheng CS, Chen J, Tan HY, Wang N, Chen Z and Feng Y: Scutellaria baicalensis and cancer treatment: Recent progress and perspectives in biomedical and clinical studies. Am J Chin Med. 46:25–54. 2018.PubMed/NCBI View Article : Google Scholar | |
Men K, Duan X, Wei XW, Gou ML, Huang MJ, Chen LJ, Qian ZY and Wei YQ: Nanoparticle-delivered quercetin for cancer therapy. Anticancer Agents Med Chem. 14:826–832. 2014.PubMed/NCBI View Article : Google Scholar | |
An T, Yin H, Lu Y and Liu F: The emerging potential of parthenolide nanoformulations in tumor therapy. Drug Des Devel Ther. 16:1255–1272. 2022.PubMed/NCBI View Article : Google Scholar | |
Dang X, Cho S, Wang H, Seok WJ, Ha JH and Kim IH: Quercetin extracted from Sophora japonica flower improves growth performance, nutrient digestibility, cecal microbiota, organ indexes, and breast quality in broiler chicks. Anim Biosci. 35:577–586. 2022.PubMed/NCBI View Article : Google Scholar | |
Lekar AV, Borisenko SN, Vetrova EV, Sushkova SN and Borisenko NI: Extraction of quercetin from Polygonum hydropiper L. by subcritical water. Am J Agric Biol Sci. 9:1–5. 2014. | |
Yang HH, Hwangbo K, Zheng MS, Cho JH, Son JK, Kim HY, Baek SH, Choi HC, Park SY and Kim JR: Quercetin-3-O-β-D-glucuronide isolated from Polygonum aviculare inhibits cellular senescence in human primary cells. Arch Pharm Res. 37:1219–1233. 2014.PubMed/NCBI View Article : Google Scholar | |
Anand David AV, Arulmoli R and Parasuraman S: Overviews of Biological importance of quercetin: A bioactive flavonoid. Pharmacogn Rev. 10:84–89. 2016.PubMed/NCBI View Article : Google Scholar | |
Michala AS and Pritsa A: Quercetin: A molecule of great biochemical and clinical value and its beneficial effect on diabetes and cancer. Diseases. 10(37)2022.PubMed/NCBI View Article : Google Scholar | |
Wang SY, Duan KM, Li Y, Mei Y, Sheng H, Liu H, Mei X, Ouyang W, Zhou HH and Liu ZQ: Effect of quercetin on P-glycoprotein transport ability in Chinese healthy subjects. Eur J Clin Nutr. 67:390–394. 2013.PubMed/NCBI View Article : Google Scholar | |
Davoodvandi A, Shabani Varkani M, Clark CCT and Jafarnejad S: Quercetin as an anticancer agent: Focus on esophageal cancer. J Food Biochem. 44(e13374)2020.PubMed/NCBI View Article : Google Scholar | |
Vafadar A, Shabaninejad Z, Movahedpour A, Fallahi F, Taghavipour M, Ghasemi Y, Akbari M, Shafiee A, Hajighadimi S, Moradizarmehri S, et al: Quercetin and cancer: New insights into its therapeutic effects on ovarian cancer cells. Cell Biosci. 10(31)2020.PubMed/NCBI View Article : Google Scholar | |
Jeong JH, An JY, Kwon YT, Rhee JG and Lee YJ: Effects of low dose quercetin: Cancer cell-specific inhibition of cell cycle progression. J Cell Biochem. 106:73–82. 2009.PubMed/NCBI View Article : Google Scholar | |
Ganthala PD, Alavala S, Chella N, andugulapati SB, Bathini NB and Sistla R: Co-encapsulated nanoparticles of erlotinib and quercetin for targeting lung cancer through nuclear EGFR and PI3K/AKT inhibition. Colloids Surf B Biointerfaces. 211(112305)2022.PubMed/NCBI View Article : Google Scholar | |
Safi A, Heidarian E and Ahmadi R: Quercetin synergistically enhances the anticancer efficacy of docetaxel through induction of apoptosis and modulation of PI3K/AKT, MAPK/ERK and JAK/STAT3 signaling pathways in MDA-MB-231 breast cancer cell line. Int J Mol Cell Med. 10:11–22. 2021.PubMed/NCBI View Article : Google Scholar | |
Guo T, Wu C, Zhang J, Yu J, Li G, Jiang H, Zhang X, Yu R and Liu X: Dual blockade of EGFR and PI3K signaling pathways offers a therapeutic strategy for glioblastoma. Cell Commun Signal. 21(363)2023.PubMed/NCBI View Article : Google Scholar | |
Li Y, Han N, Hou P, Zhao FQ and Liu H: Roles of MAPK and Nrf2 signaling pathways in quercetin alleviating redox imbalance induced by hydrogen peroxide in mammary epithelial cells. Anim Nutr. 1(e1)2024. | |
Liu W, Chen D, Su J, Zheng R, Kong R, Zhu B, Dong H and Li Y: Quercetin induced HepG2 cells apoptosis through ATM/JNK/STAT3 signaling pathways. Biocell. 47:187–194. 2023. | |
Wang ZX, Ma J, Li XY, Wu Y, Shi H, Chen Y, Lu G, Shen HM, Lu GD and Zhou J: Quercetin induces p53-independent cancer cell death through lysosome activation by the transcription factor EB and reactive oxygen species-dependent ferroptosis. Br J Pharmacol. 178:1133–1148. 2021.PubMed/NCBI View Article : Google Scholar | |
Chan ST, Yang NC, Huang CS, Liao JW and Yeh SL: Quercetin enhances the antitumor activity of trichostatin A through upregulation of p53 protein expression in vitro and in vivo. PLoS One. 8(e54255)2013.PubMed/NCBI View Article : Google Scholar | |
Granado-Serrano AB, Martín MA, Bravo L, Goya L and Ramos S: Quercetin induces apoptosis via caspase activation, regulation of Bcl-2, and inhibition of PI-3-kinase/Akt and ERK pathways in a human hepatoma cell line (HepG2). J Nutr. 136:2715–2721. 2006.PubMed/NCBI View Article : Google Scholar | |
Lee DH, Szczepanski M and Lee YJ: Role of Bax in quercetin-induced apoptosis in human prostate cancer cells. Biochem Pharmacol. 75:2345–2355. 2008.PubMed/NCBI View Article : Google Scholar | |
Huang CF, Liu SH, Ho TJ, Lee KI, Fang KM, Lo WC, Liu JM, Wu CC and Su CC: Quercetin induces tongue squamous cell carcinoma cell apoptosis via the JNK activation-regulated ERK/GSK-3α/β-mediated mitochondria-dependent apoptotic signaling pathway. Oncol Lett. 23(78)2022.PubMed/NCBI View Article : Google Scholar | |
Xu D, Hu MJ, Wang YQ and Cui YL: Antioxidant activities of quercetin and its complexes for medicinal application. Molecules. 24(1123)2019.PubMed/NCBI View Article : Google Scholar | |
Lee YJ, Lee DM and Lee SH: Nrf2 expression and apoptosis in quercetin-treated malignant mesothelioma cells. Mol Cells. 38:416–425. 2015.PubMed/NCBI View Article : Google Scholar | |
Rashidi Z, Aleyasin A, Eslami M, Nekoonam S, Zendedel A, Bahramrezaie M and Amidi F: Quercetin protects human granulosa cells against oxidative stress via thioredoxin system. Reprod Biol. 19:245–254. 2019.PubMed/NCBI View Article : Google Scholar | |
Baba RA, Mir HA, Mokhdomi TA, Bhat HF, Ahmad A and Khanday FA: Quercetin suppresses ROS production and migration by specifically targeting Rac1 activation in gliomas. Front Pharmacol. 15(1318797)2024.PubMed/NCBI View Article : Google Scholar | |
Biswas P, Dey D, Biswas PK, Rahaman TI, Saha S, Parvez A, Khan DA, Lily NJ, Saha K, Sohel M, et al: A comprehensive analysis and anti-cancer activities of quercetin in ROS-mediated cancer and cancer stem cells. Int J Mol Sci. 23(11746)2022.PubMed/NCBI View Article : Google Scholar | |
Zhang Q, Cheng G, Qiu H, Zhu L, Ren Z, Zhao W, Zhang T and Liu L: The p53-inducible gene 3 involved in flavonoid-induced cytotoxicity through the reactive oxygen species-mediated mitochondrial apoptotic pathway in human hepatoma cells. Food Funct. 6:1518–1525. 2015.PubMed/NCBI View Article : Google Scholar | |
Pratheeshkumar P, Budhraja A, Son YO, Wang X, Zhang Z, Ding S, Wang L, Hitron A, Lee JC, Xu M, et al: Quercetin inhibits angiogenesis mediated human prostate tumor growth by targeting VEGFR-2 regulated AKT/mTOR/P70S6K signaling pathways. PLoS One. 7(e47516)2012.PubMed/NCBI View Article : Google Scholar | |
Igura K, Ohta T, Kuroda Y and Kaji K: Resveratrol and quercetin inhibit angiogenesis in vitro. Cancer Lett. 171:11–16. 2001.PubMed/NCBI View Article : Google Scholar | |
Okumo T, Furuta A, Kimura T, Yusa K, Asano K and Sunagawa M: Inhibition of angiogenic factor productions by quercetin in vitro and in vivo. Medicines (Basel). 8(22)2021.PubMed/NCBI View Article : Google Scholar | |
Ren KW, Li YH, Wu G, Ren JZ, Lu HB, Li ZM and Han XW: Quercetin nanoparticles display antitumor activity via proliferation inhibition and apoptosis induction in liver cancer cells. Int J Oncol. 50:1299–1311. 2017.PubMed/NCBI View Article : Google Scholar | |
Guan X, Gao M, Xu H, Zhang C, Liu H, Lv L, Deng S, Gao D and Tian Y: Quercetin-loaded poly (lactic-co-glycolic acid)-d-α-tocopheryl polyethylene glycol 1000 succinate nanoparticles for the targeted treatment of liver cancer. Drug Deliv. 23:3307–3318. 2016.PubMed/NCBI View Article : Google Scholar | |
Srisa-Nga K, Mankhetkorn S, Okonogi S and Khonkarn R: Delivery of superparamagnetic polymeric micelles loaded with quercetin to hepatocellular carcinoma cells. J Pharm Sci. 108:996–1006. 2019.PubMed/NCBI View Article : Google Scholar | |
Wang C, Su L, Wu C, Wu J, Zhu C and Yuan G: RGD peptide targeted lipid-coated nanoparticles for combinatorial delivery of sorafenib and quercetin against hepatocellular carcinoma. Drug Dev Ind Pharm. 42:1938–1944. 2016.PubMed/NCBI View Article : Google Scholar | |
Deng Q, Huang Y, Zeng J, Li X, Zheng X, Guo L, Shi J and Bai L: Recent advancements in the small-molecule drugs for hepatocellular carcinoma (HCC): Structure-activity relationships, pharmacological activities, and the clinical trials. Biomed Pharmacother. 179(117343)2024.PubMed/NCBI View Article : Google Scholar | |
Tang W, Chen Z, Zhang W, Cheng Y, Zhang B, Wu F, Wang Q, Wang S, Rong D, Reiter FP, et al: The mechanisms of sorafenib resistance in hepatocellular carcinoma: Theoretical basis and therapeutic aspects. Signal Transduct Target Ther. 5(87)2020.PubMed/NCBI View Article : Google Scholar | |
Pradhan A, Kumari A, Srivastava R and Panda D: Quercetin encapsulated biodegradable plasmonic nanoparticles for photothermal therapy of hepatocellular carcinoma cells. ACS Appl Bio Mater. 2:5727–5738. 2019.PubMed/NCBI View Article : Google Scholar | |
Varshosaz J, Jafarian A, Salehi G and Zolfaghari B: Comparing different sterol containing solid lipid nanoparticles for targeted delivery of quercetin in hepatocellular carcinoma. J Liposome Res. 24:191–203. 2014.PubMed/NCBI View Article : Google Scholar | |
Koulouris A, Tsagkaris C, Spyrou V, Pappa E, Troullinou A and Nikolaou M: Hepatocellular carcinoma: An overview of the changing landscape of treatment options. J Hepatocell Carcinoma. 8:387–401. 2021.PubMed/NCBI View Article : Google Scholar | |
Bukowski K, Kciuk M and Kontek R: Mechanisms of multidrug resistance in cancer chemotherapy. Int J Mol Sci. 21(3233)2020.PubMed/NCBI View Article : Google Scholar | |
Talib WH, Alsayed AR, Barakat M, Abu-Taha MI and Mahmod AI: Targeting drug chemo-resistance in cancer using natural products. Biomedicines. 9(1353)2021.PubMed/NCBI View Article : Google Scholar | |
Al Saihati HA and Rabaan AA: Cellular resistance mechanisms in cancer and the new approaches to overcome resistance mechanisms chemotherapy. Saudi Med J. 44:329–344. 2023.PubMed/NCBI View Article : Google Scholar | |
Halder J, Pradhan D, Kar B, Ghosh G and Rath G: Nanotherapeutics approaches to overcome P-glycoprotein-mediated multi-drug resistance in cancer. Nanomedicine. 40(102494)2022.PubMed/NCBI View Article : Google Scholar | |
Waghray D and Zhang Q: Inhibit or evade multidrug resistance P-glycoprotein in cancer treatment: Miniperspective. J Med Chem. 61:5108–5121. 2017.PubMed/NCBI View Article : Google Scholar | |
Li S, Zhao Q, Wang B, Yuan S, Wang X and Li K: Quercetin reversed MDR in breast cancer cells through down-regulating P-gp expression and eliminating cancer stem cells mediated by YB-1 nuclear translocation. Phytother Res. 32:1530–1536. 2018.PubMed/NCBI View Article : Google Scholar | |
Ganesan M, Kanimozhi G, Pradhapsingh B, Khan HA, Alhomida AS, Ekhzaimy A, Brindha GR and Prasad NR: Phytochemicals reverse P-glycoprotein mediated multidrug resistance via signal transduction pathways. Biomed Pharmacother. 139(111632)2021.PubMed/NCBI View Article : Google Scholar | |
Suroowan S, Abdallah HH and Mahomoodally MF: Herb-drug interactions and toxicity: Underscoring potential mechanisms and forecasting clinically relevant interactions induced by common phytoconstituents via data mining and computational approaches. Food Chem Toxicol. 156(112432)2021.PubMed/NCBI View Article : Google Scholar | |
Palmeira A, Sousa E, Vasconcelos MH and Pinto MM: Three decades of P-gp inhibitors: Skimming through several generations and scaffolds. Curr Med Chem. 19:1946–2025. 2012.PubMed/NCBI View Article : Google Scholar | |
Gurunath S, Nanjwade BK and Patil PA: Oral bioavailability and intestinal absorption of candesartan cilexetil: Role of naringin as P-glycoprotein inhibitor. Drug Dev Ind Pharm. 41:170–176. 2015.PubMed/NCBI View Article : Google Scholar | |
Di Sotto A, Irannejad H, Eufemi M, Mancinelli R, Abete L, Mammola CL, Altieri F, Mazzanti G and Di Giacomo S: Potentiation of low-dose doxorubicin cytotoxicity by affecting P-glycoprotein through caryophyllane sesquiterpenes in hepG2 cells: An in vitro and in silico study. Int J Mol Sci. 21(633)2020.PubMed/NCBI View Article : Google Scholar | |
Dewanjee S, Dua TK, Bhattacharjee N, Das A, Gangopadhyay M, Khanra R, Joardar S, Riaz M, Feo V and Zia-Ul-Haq M: Natural products as alternative choices for P-glycoprotein (P-gp) inhibition. Molecules. 22(871)2017.PubMed/NCBI View Article : Google Scholar | |
Mohos V, Fliszár-Nyúl E, Ungvári O, Kuffa K, Needs PW, Kroon PA, Telbisz Á, Özvegy-Laczka C and Poór M: Inhibitory effects of quercetin and its main methyl, sulfate, and glucuronic acid conjugates on cytochrome p450 enzymes, and on OATP, BCRP and MRP2 transporters. Nutrients. 12(2306)2020.PubMed/NCBI View Article : Google Scholar | |
Arinç E, Yilmaz D and Bozcaarmutlu A: Mechanism of inhibition of CYP1A1 and glutathione S-transferase activities in fish liver by quercetin, resveratrol, naringenin, hesperidin, and rutin. Nutr Cancer. 67:137–144. 2015.PubMed/NCBI View Article : Google Scholar | |
van Zanden JJ, Ben Hamman O, van Iersel ML, Boeren S, Cnubben NH, Lo Bello M, Vervoort J, van Bladeren PJ and Rietjens IM: Inhibition of human glutathione S-transferase P1-1 by the flavonoid quercetin. Chem Biol Interact. 145:139–148. 2003.PubMed/NCBI View Article : Google Scholar | |
Choi JS, Piao YJ and Kang KW: Effects of quercetin on the bioavailability of doxorubicin in rats: role of CYP3A4 and P-gp inhibition by quercetin. Arch Pharm Res. 34:607–613. 2011.PubMed/NCBI View Article : Google Scholar | |
Borska S, Sopel M, Chmielewska M, Zabel M and Dziegiel P: Quercetin as a potential modulator of P-glycoprotein expression and function in cells of human pancreatic carcinoma line resistant to daunorubicin. Molecules. 15:857–870. 2010.PubMed/NCBI View Article : Google Scholar | |
Singh A, Patel SK, Kumar P, Das KC, Verma D, Sharma R, Tripathi T, Giri R, Martins N and Garg N: Quercetin acts as a P-gp modulator via impeding signal transduction from nucleotide-binding domain to transmembrane domain. J Biomol Struct Dyn. 40:4507–4515. 2020.PubMed/NCBI View Article : Google Scholar | |
Mu Y, Fu Y, Li J, Yu X, Li Y, Wang Y, Wu X, Zhang K, Kong M, Feng C and Chen X: Multifunctional quercetin conjugated chitosan nano-micelles with P-gp inhibition and permeation enhancement of anticancer drug. Carbohydr Polym. 203:10–18. 2019.PubMed/NCBI View Article : Google Scholar | |
Guo Y, Liu S, Luo F, Tang D, Yang T, Yang X and Xie Y: A nanosized codelivery system based on intracellular stimuli-triggered dual-drug release for multilevel chemotherapy amplification in drug-resistant breast cancer. Pharmaceutics. 14(422)2022.PubMed/NCBI View Article : Google Scholar | |
Yu J, Chen H, Jiang L and Wang J, Dai J and Wang J: Codelivery of adriamycin and P-gp inhibitor quercetin using PEGylated liposomes to overcome cancer drug resistance. J Pharm Sci. 108:1788–1799. 2019.PubMed/NCBI View Article : Google Scholar | |
Wang X, Chen Y, Dahmani FZ, Yin L, Zhou J and Yao J: Amphiphilic carboxymethyl chitosan-quercetin conjugate with P-gp inhibitory properties for oral delivery of paclitaxel. Biomaterials. 35:7654–7665. 2014.PubMed/NCBI View Article : Google Scholar | |
Khonkarn R, Daowtak K and Okonogi S: Chemotherapeutic efficacy enhancement in P-gp-overexpressing cancer cells by flavonoid-loaded polymeric micelles. AAPS PharmSciTech. 21(121)2020.PubMed/NCBI View Article : Google Scholar | |
Mu Y, Wu G, Su C, Dong Y, Zhang K, Li J, Sun X, Li Y, Chen X and Feng C: pH-sensitive amphiphilic chitosan-quercetin conjugate for intracellular delivery of doxorubicin enhancement. Carbohydr Polym. 223(115072)2019.PubMed/NCBI View Article : Google Scholar | |
Kumar M, Sharma G, Misra C, Kumar R, Singh B, Katare OP and Raza K: N-desmethyl tamoxifen and quercetin-loaded multiwalled CNTs: A synergistic approach to overcome MDR in cancer cells. Mater Sci Eng C Mater Biol Appl. 89:274–282. 2018.PubMed/NCBI View Article : Google Scholar | |
Qian J, Liu S, Yang T, Xiao Y, Sun J, Zhao J, Zhang Z and Xie Y: Polyethyleneimine-tocopherol hydrogen succinate/hyaluronic acid-quercetin (PEI-TOS/HA-QU) core-shell micelles delivering paclitaxel for combinatorial treatment of MDR breast cancer. J Biomed Nanotechnol. 17:382–398. 2021.PubMed/NCBI View Article : Google Scholar | |
Dallavalle S, Dobričić V, Lazzarato L, Gazzano E, Machuqueiro M, Pajeva I, Tsakovska I, Zidar N and Fruttero R: Improvement of conventional anti-cancer drugs as new tools against multidrug resistant tumors. Drug Resist Updat. 50(100682)2020.PubMed/NCBI View Article : Google Scholar | |
Deshmukh R, Prajapati M and Harwansh RK: Management of colorectal cancer using nanocarriers-based drug delivery for herbal bioactives: Current and emerging approaches. Curr Pharm Biotechnol. 25:599–622. 2024.PubMed/NCBI View Article : Google Scholar | |
Kenchegowda M, Rahamathulla M, Hani U, Begum MY, Guruswamy S, Osmani RAM, Gowrav MP, Alshehri S, Ghoneim MM, Alshlowi A and Gowda DV: Smart nanocarriers as an emerging platform for cancer therapy: A review. Molecules. 27(146)2021.PubMed/NCBI View Article : Google Scholar | |
Tang L, Li J, Zhao Q, Pan T, Zhong H and Wang W: Advanced and innovative nano-systems for anticancer targeted drug delivery. Pharmaceutics. 13(1151)2021.PubMed/NCBI View Article : Google Scholar | |
Kumar V, Rahman M, Gahtori P, Al-Abbasi F, Anwar F and Kim HS: Current status and future directions of hepatocellular carcinoma-targeted nanoparticles and nanomedicine. Expert Opin Drug Deliv. 18:673–694. 2021.PubMed/NCBI View Article : Google Scholar | |
Mishra AK, Pandey M, Dewangan HK, Sl N and Sahoo PK: A comprehensive review on liver targeting: Emphasis on nanotechnology-based molecular targets and receptors mediated approaches. Curr Drug Targets. 23:1381–1405. 2022.PubMed/NCBI View Article : Google Scholar | |
Dutta R and Mahato RI: Recent advances in hepatocellular carcinoma therapy. Pharmacol Ther. 173:106–117. 2017.PubMed/NCBI View Article : Google Scholar | |
Aghababaei F and Hadidi M: Recent advances in potential health benefits of quercetin. Pharmaceuticals (Basel). 16(1020)2023.PubMed/NCBI View Article : Google Scholar | |
Manzoor MF, Hussain A, Sameen A, Sahar A, Khan S, Siddique R, Aadil RM and Xu B: Novel extraction, rapid assessment and bioavailability improvement of quercetin: A review. Ultrason Sonochem. 78(105686)2021.PubMed/NCBI View Article : Google Scholar |