Skip to main content

Anti-cancer effect of nano-encapsulated boswellic acids, curcumin and naringenin against HepG-2 cell line

Abstract

Background

liver cancer is one of the most common cancers in the world. So far, there is no gold standard treatment for hepatocellular carcinoma. We conducted this in vitro study to assess the effect of three natural products: Boswellic acids, curcumin and naringin versus corresponding nanoparticles (NPs) on Hep G2 cells proliferation.

Methods

Boswellic acid, curcumin, naringin-loaded NPs were prepared using nanoprecipitation method. Human liver (HepG2) cell line was cultured in Dulbecco’s modified Eagle’s medium (DMEM). The cell growth inhibition and cytotoxicity were evaluated by MTT assay.

Results

Boswellic acid, curcumin, naringin were able to inhibit HepG2 cells proliferation. IC50 at 24 h, 48 h showed significant lower values in NPs versus Free herbs. IC50 values of free Boswellic acids and NPs at 24 h were (24.60 ± 1.89 and 7.78 ± 0.54, P < 0.001), at 48 h were (22.45 ± 1.13 and 5.58 ± 0.27, P < 0.001) respectively. IC50 values of free curcumin and NPs at 24 h were (5.89 ± 0.8 and 3.46 ± 0.23, P < 0.05), at 48 h were (5.57 ± 0.94 and 2.51 ± 0.11, P < 0.05), respectively. For free and naringenin NPs, IC50 values at 24 h were (14.57 ± 1.78 and 7.25 ± 0.17, P < 0.01), at 48 h were (11.37 ± 1.45 and 5.21 ± 0.18, P < 0.01) respectively.

Conclusion

Boswellic acid, curcumin, naringin and their nanoprecipitation prepared nanoparticles suppressed Hep G2 cells proliferation.

Peer Review reports

Background

Liver cancer is ranked as the fifth most common and the second death causing cancer in the world [1]. Egypt is facing a doubling incidence rate in the last years [2]. The majority of primary liver cancers are hepatocellular carcinoma (HCC) [3]. Chronic viral hepatitis (B and C), alcohol intake and aflatoxin exposure are the most frequent causes of HCC [1]. Persistent hepatic inflammation, hepatocytes necrosis and regeneration together with fibrosis add to the pathogenesis of HCC [4]. Where there is an imbalance between activation and inactivation of pro-oncogenes and tumor suppressor genes [5]. Hence, HCC treatment should involve different targeted approaches [6]. The goal of HCC therapeutic modalities is to prolong survival with better life quality. Surgical resection, transplantation, ablation, trans-arterial chemoembolization [7,8,9] and the tyrosine-kinase inhibitors sorafenib [10, 11] Lenvatinib [12] and regorafenib [13] are modalities with proven survival benefit. Arterial embolization without chemotherapy, external radiotherapy [14] and radio-embolization have shown anti-tumor activity [15,16,17] but no definitive proof of survival benefit has been found [18]. Systemic chemotherapy carries toxic side effects with no survival benefit. Agents such as tamoxifen, octreotide, and antiandrogens are completely ineffective [19]. 70% of cases are suffering from tumor recurrence in 5 years [20] Up till now, there is no proven recurrence preventing agents [21]. Complementary and alternative medicine (CAM) is commonly practiced worldwide especially among cancer patients in western countries [22, 23]. Boswellia serrata (B. serrata) exerts its action against HCC through different ways, suppression of topoisomerase I & II by a caspase-8 dependent pathway [24,25,26] acting on ribosomal protein production [27, 28] and inhibition of angiogenesis [29]. Poor oral bioavailability of Boswellia is managed by providing it with a standardized meal [30], addition of anionic drugs to improve uptake [31], lecithin delivery form (Phytosome R); nanoparticle delivery systems and synthetic derivatization [32,33,34]. Curcumin is a natural yellow lipid-soluble compound from the plant Curcuma Longa [35]. It has antioxidant, anti-inflammatory, anti-carcinogenic, and anti-angiogenic properties [36, 37]. Curcumin inhibited hypoxia-inducible factor-1α, the protein which participates in proliferation, migration, and invasiveness of cell cancer [38, 39]. Induction of apoptosis and suppression of cell proliferation is another cytotoxic mechanism of curcumin through suppression of cell membrane axon enrollment, target transcription gene and nuclear Beta-catenin aggregation [40,41,42]. Metabolic instability, insufficient absorption, and bioavailability all interfere with curcumin pharmacological effect [43]. Curcumin formulations and synthetic analogs have been tried to overcome these obstacles via its combination with polymeric micelles or nanoparticle-based encapsulation which achieved more stability and bioavailability with more potent anti-cancer effect than free curcumin [44, 45]. Naringenin is a flavonoid [46]. Being a lipophilic, facilitates its intestinal absorption through passive diffusion into enterocytes [47]. Naringenin exhibits cytotoxic effect against HCC through induction of the endogenous antioxidant system, inhibition of nuclear factor kappa B (NF-κB), vascular endothelial growth factor (VEGF) and matrix metalloproteinase (MMPs) [48,49,50,51]. Furthermore, naringenin up regulated p53 in a dose-dependent manner with subsequent cell cycle arrest in Hep G2 cells [52]. Naringenin attenuates two-pore channels (TPC2) activities and hence, VEGF mediated angiogenesis through interruption of intracellular calcium [53]. Moreover, anti-invasive and anti-migratory action of naringenin were clarified in 12-O-tetradecanoylphorbol-13-acetate (TPA)-activated hepatoma cells by the downregulation of protein kinase C (PKC), epidermal growth factor (EGF), mitogen activated protein kinase (MAPK) and phosphoinositide 3 – kinase L protein kinase B (PI3K/Akt) signaling pathways, and NF-κB, activator protein 1 (AP-1) and matrix metalloproteinase (MMP9) activities [54]. Nanoencapsulation through nanoparticle formation is currently used to increase the oral bioavailability and pharmacokinetic properties of many herbal and non-herbal drugs for the treatment of different diseases, especially cancer treatments [55,56,57,58]. These novel nanoparticulate drug delivery systems enhance the biological effects and pharmacokinetics properties of boswellic acids. Therefore, represents a promising method for improving the beneficial therapeutic effects of boswellic acids [59]. Nanoncapsulated curcumin exhibited superior bioactive properties in the aqueous medium, exerting antioxidant and anticancer effects against HCC [60]. The efficiency of Naringenin in treating hepatic cancer was increased by using a nanoparticle drug delivery approach. The enhanced oral bioavailability, and sustained action of entrapped Naringenin, showed better efficacy for the treatment of liver carcinoma [61].

Materials and methods

This in vitro study was conducted in Tanta University to assess the effect of free Boswellic acids, Curcumin, naringin, and their corresponding nanoprecipitation prepared nanoparticles on Hep G2 cells proliferation.

Natural products

Boswellic acids, curcumin, [62,63,64] and naringin [65] were previously isolated in the laboratory of Prof. Dr. Farid Badria (Liver Research Lab, Fab-Lab, Mansoura, university, Mansoura 35,516 Egypt) (Flow charts 1–3, respectively).

Chemicals and reagents

PLGA (Poly lactic-co-glycolic acid) (Resomer® RG 503 H, 50:50 lactic: glycolic ratio MW34 kDa), Polyvinyl alcohol (PVA, 87–89% degree of hydrolysis, M. wt 31,000–50,000), Tween-80, Phosphate-buffered saline (PBS), Acetone, Ethanol Dichloromethane (DCM) and Acetonitrile (HPLC grade) were purchased from Sigma-Aldrich Chemical Co. (St. Louis, MO, USA). All chemicals and reagents were of analytical/HPLC grade. Milli-Q® water was used throughout the study.

Chemicals and reagents

PLGA (Poly lactic-co-glycolic acid) (Resomer® RG 503 H, 50:50 lactic: glycolic ratio MW34 kDa), Polyvinyl alcohol (PVA, 87–89% degree of hydrolysis, M. wt 31,000–50,000), Tween-80, Phosphate-buffered saline (PBS), Acetone, Ethanol Dichloromethane (DCM) and Acetonitrile (HPLC grade) were purchased from Sigma-Aldrich Chemical Co. (St. Louis, MO, USA). All chemicals and reagents were of analytical/HPLC grade. Milli-Q® water was used throughout the study.

Preparation of drugs-loaded nanoparticles (NPs)

Boswellic acids, curcumin, naringin-loaded NPs were prepared using two different preparation techniques. The first was single oil in water emulsion/solvent evaporation technique (F1) [66]. All drugs (5 mg from each drug) and PLGA (150 mg) were dissolved in 3.5 ml of DCM, once dissolution was complete, the organic solution was added using a syringe to 25 ml of aqueous solution of 1% w/v PVA and sonicated using an ultrasonic homogenizer equipped with a 3.2 mm probe (Cole-Parmer, 4710 series, United States). The final emulsion was stirred overnight to assist in the removal of residual organic solvents. The nanospheres collected by centrifugation were washed three times with ultra-pure water and sucrose solutions. NPs were lyophilized for 72 h using a freeze-dryer and stored at 4 °C until further use. The other preparation method was nanoprecipitation (F2) [67]. Polymer and the three drugs were dissolved in a mixture of acetone/ethanol 3:1 v/v. The drug solution was added to an aqueous solution of 0.5% v/v tween-80 under continuous stirring using a magnetic stirrer for 4 h. The nanospheres were collected using the same procedures, lyophilized, and stored as previously mentioned.

Physicochemical characterization, particle size, polydispersity index (PDI), and zeta potential (ZP)

Particle size and PDI were obtained by dynamic light scattering technique, while ZP was determined by measurement of the electrophoretic mobility. Each sample was diluted in purified water at 25 ℃. Determinations were performed in triplicate using a Zeta Sizer Nano-ZS (Malvern Instruments Ltd., Malvern, UK).

Morphological characterization

The morphology of drug-loaded NPs was observed using scanning electron microscopy (FEI Quanta 400 FEG, FEI). A sample of drugs-loaded PLGA NPs was mounted on carbon tape and sputter-coated with gold under a vacuum in an argon atmosphere before microscopic analysis.

Determination of drug entrapment efficiency (%EE)

Drug encapsulation was measured using the indirect method after collecting the supernatant. The non-encapsulated drug in the supernatant was measured by the High-performance liquid chromatography (HPLC) technique. The HPLC analysis was performed using a C18 column (250 mm×4.6 mm, 5 μm; Waters Corporation, Milford, MA, USA). The mobile phase of curcumin was acetonitrile and water (50:50 v/v) acidified with 2% acetic acid at a flow rate of 1 mL min− 1 and UV detection at 425 nm [68]. The mobile phase of naringenin was methanol: water (70:30) acidified by orthophosphoric acid with a flow rate of 1 mL min− 1 and ultraviolet detection at 289 nm [69]. The mobile phase used for boswellic acid was acetonitrile and water (85:15) acidified with (0.1% acetic acid) and flow rate of 0.5 mL min− 1 and UV detection at 254 nm [70]. The %EE was calculated using the following equation:

\(\%EE=\frac{Total\ Drug\ added-Free\ un-entrapped\ drug}{Total\ Drug\ added}\ \times100\)

Stability tests

The freshly prepared freeze-dried NPs were stored in a stable chamber at 25 °C for 3 months. Periodically, a sample was collected to measure particle size, PDI, and ZP. The evaluation was performed in triplicate for each analysis.

Drug release studies

In vitro, drug release test was assessed by direct dialysis method. Drug-loaded PLGA NPs were respectively placed in a 3 000 Da dialysis bag and were incubated in 50 mL of phosphate-buffered saline medium (pH 7.4) [55, 71]. The system was maintained at 37 ± 2 °C, under stirring at 100 ± 10 rpm. Samples were withdrawn at pre-determined intervals and replaced by the same volume of fresh buffer solution. The concentrations of each drug in samples were analyzed by HPLC as described above.

Herbal nanoparticles codelivery for HPG2 cells cell culture and exposure of NPs

Human liver (HepG2) cells (VACSERA Company, Egypt) were cultured in Dulbecco’s modified Eagle’s medium (DMEM) (Invitrogen, CA, USA) containing 10% fetal bovine serum (Invitrogen), 100 U/mL penicillin, and 100 µg/mL streptomycin (Invitrogen) with the supply of 5% CO2 at 37 °C. At 75-85% confluence, cells were harvested and further subcultured for biochemical studies. Cells were seeded in a 96-well tissue culture plate (4.5 × 104) cells in 100 µl of culture medium. Cells were allowed for 24 h to attach on the surface of the culture plate before exposure to free herbs or codelivery NPs [72]. Dry powder of NPs has been suspended in DMEM at IC 50 of 1 mg/mL. The stock solution was further diluted to different concentrations required for cytotoxicity experiments. The various concentrations of NPs were sonicated at room temperature for 30 min at 40 W to avoid agglomeration before exposure to cells. Cells not exposed to NPs served as a control for each experiment [73].

Cell viability assay

The Cell growth inhibition and cytotoxicity of free herbs and their codelivery NPs on HepG2 cells were evaluated by MTT assay. The HepG2 cells were treated with different concentrations (1, 3, 5, 7, 9 µg/mL) of free herbs or codelivery NPs for 24 and 48 h. Cell viability was assessed by MTT assay. Live cells could reduce MTT in blue formazan products dissolved in a solvent, and absorbance was recorded at 570 nm employing a microplate reader (Synergy-HT, Biotek, Winooski, VT, USA). Based on the MTT cell viability assay results, the inhibitory concentration (IC 50) values were calculated by GraphPad Prism 8 software (GraphPad S).

Statistical analysis

All analyses were performed using GraphPad Prism Ver. 6.0 (GraphPad Software Inc. San Diego, CA). All data were expressed as mean ± SEM with statistical significance indicated when P < 0.05. Statistical comparisons between control and treatment groups were determined using one-way ANOVA with Tukey’s post hoc tests.

Results

Effect of preparation technique on the physio-chemical properties of drug-loaded NPs

Nanoprecipitation method (F2) prepared polymeric NPs were lower in size and higher in %EE (P < 0.05) compared to O/W single emulsion solvent evaporation technique (F1) prepared NPs (Fig. 1A). The size distribution of NPs produced by nanoprecipitation is more uniform and homogenous than the other technique. There was a significant decrease in PDI values (P < 0.05) (Fig. 1B). zeta potential of both types of NPs exhibited non-significant change (P ˃ 0.05) (Fig. 1C). A sharp increase in %E.E of naringenin and Boswellia (P < 0.01) was observed in case of NP prepared by nanoprecipitation compared to the other NPs. (Fig. 1D). A significant increase in initial burst release (P < 0.05) after 24 h was observed in nanoprecipitation prepared NPs. Initial bursts of 47%, 50% and 62% were observed for curcumin, naringenin and Boswellia versus 36%, 38% and 42% from NPs prepared through emulsion formation, respectively ( Fig. 2). Stability results for three months showed that F2 exhibited a non-significant increase in NP size, PDI also a non-significant decrease in zeta potential was observed which confirmed high storage stability of these lyophilized NPs in vitro (Table 1). From the above results, F2 was superior to F1 consequently, it was chosen for further in vitro studies.

Fig. 1
figure 1

Effect of preparation technique on the physio-chemical properties of drug-loaded nanoparticles. (A) polymeric NPs sizes. The figure shows polymeric nanoparticles which were prepared by nanoprecipitation method (F2) were significantly lower in size compared to drug-loaded NP formulations fabricated by O/W single emulsion solvent evaporation technique (F1) (B) polydispersity index (PDI) values. The figure shows the size distribution of NPs produced by nanoprecipitation is more uniform and homogenous compared to those prepared by other techniques with significant decrease in polydispersity index (PDI) values. (C) zeta potential of both types of NPs. The figure shows zeta potential of both types of NPs exhibited non-significant change. (D) drug entrapment efficiency (%EE). The figure shows a sharp increase in %E.E of naringenin and Boswellia was observed in case of NP prepared by nanoprecipitation compared to other NPs. However, %E.E of Curcumin showed non-significant increase

Fig. 2
figure 2

Release profile of both types of nanoparticles. The figure shows a significant increase in initial burst release after 24 h was observed in case of NPs prepared by nanoprecipitation compared to other NPs prepared by emulsion technique

Table 1 In Vitro Stability Results of Drug-loaded Nanoparticles (NPs) of Nanoprecipitation Method

Scanning electron microscopy

The drug-loaded NP (F2) were of spherical shape with a size distribution in approximate agreement with light scattering data. The smooth NP surface was free from void and pores (Fig. 3).

Fig. 3
figure 3

Scanning electron microscopy, a micrograph of the drug-loaded nanoparticles of precipitation method. The figure shows a micrograph of the drug-loaded NP (F2). NP were of spherical shape with a size distribution in approximate agreement with light scattering data. The smooth NP surface was free from void and pores

Invitro cytotoxicity assay

Boswellic acids, curcumin and naringenin inhibited proliferation of HepG2 cells with significantly lower IC50 values in NPs (Table 2) .

Table 2 Half Maximal Inhibitory Concentration (IC50) of Boswellic acids, Curcumin, Naringenin and Nanoparticles

Discussion

Hepatocellular carcinoma (HCC) is considered one of the main causes of morbidity and mortality all over the world [74]. Phytochemicals have been utilized in the treatment of variable chronic health problems [75, 76]. There are multiple anti-cancer drugs from natural sources such as vinca alkaloids, vinblastine (VLB), and vincristine (VCR) isolated from the Madagascar periwinkle, Catharanthus roseus G. Don. (Apocynaceae), etoposide (VM 26) and teniposide (VP 16–213) from Podophyllum species (Podophyllaceae), Paclitaxel initially was isolated from the bark of Taxus brevifolia Nutt. (Taxaceae) [77, 78]. As an attempt to introduce some natural agents for HCC treatment, we evaluated the effect of free Boswellic acids, curcumin, and naringenin and nanoparticles on Hep G2 cell proliferation. Single emulsion solvent evaporation technique was evaluated against nanoprecipitation for the entrapment of Boswellic acids, curcumin, and naringenin into PLGA NPs. Generally, the single emulsion process is most suited for water-insoluble drugs such as steroids, while the double emulsion process is considered ideal to encapsulate water-soluble drugs such as peptides [79, 80]. Although the emulsion solvent evaporation technique is the most used method to prepare PLGA NPs, this method is affected by the payload’s physical and chemical properties [81, 82]. In the present study, drug-loaded NPs (F2) prepared by nanoprecipitation resulted in a significant decrease in nano-size and PDI meanwhile a significant increase in %E.E compared to NPs prepared by emulsion/evaporation method (F1). The nanoprecipitation technique was employed instead of the single emulsification/solvent evaporation technique to optimize the physicochemical characteristics of the prepared drug-loaded nanoparticles such as the size, PDI, and %EE [67, 83]. This can be attributed to the ability of these drugs to diffuse through the organic solvent after the emulsification/solvent evaporation step to the water phase leading to inefficient encapsulation of these drugs in the polymeric matrix. On the other hand, the nanoprecipitation technique seems more efficient in increasing the entrapment of all drugs inside the core of the PLGA polymeric matrix. Therefore, the nanoprecipitation method decreased the size and PDI of the prepared NPs and increased all drugs encapsulation, especially for these hydrophobic drugs. The release profile of F2 showed significantly higher initial and higher total amounts of drugs released. The effects of nanoparticles physicochemical properties, such as size, shape and drug encapsulation, on their biological efficacy have been studied extensively. However, the effect of drug entrapment and their controlled drug release on NP efficacy and toxicity has not been thoroughly evaluated both in vitro and in vivo [83]. Our study aims to highlight the importance of nanoencapsulation of three hydrophobic drugs (boswellic acids, curcumin, and naringenin) and their controlled release in vitro compared to these free drugs. The in vitro cytotoxic activity of the drug-loaded NPs was significantly higher than free drugs due to the advantages of cellular interaction between the drug-loaded NPs compared to free drugs. Both free and herbal nanoparticles induced apoptosis in Hep G2 cells with significantly lower IC50 values in nanoparticles (NPs). Boswellia anti-neoplastic action was proved in variable human cancer cell lines and in HepG2 cell line as well [27, 84,85,86]. Among fractions and volatile oil, Boswellia petroleum ether and methanol extracts were superior with IC50 values of 1.58 and 5.82 µg/mL at 48 h, respectively in comparison to doxorubicin with an IC50 of 4.68 µg/mL at 48 h [87]. Moreover, Boswellia extract inhibited Hep G2 as a monotherapy (IC50 value of 21.21 ± 0.92 µg/mL) and exhibited a synergistic effect in combination with doxorubicin in another study [88]. Curcumin, an active ingredient of Curcuma longa possesses multiple chemotherapeutic activities in hepatic, gastric, colon, ovarian, breast and cervical cell cancer [89, 90]. Furthermore, curcumin was superior to other anti-cancer drugs in initiating apoptosis with less insult to normal cells [91, 92]. Change of morphology and inhibition of Hep G2 cell line was induced by curcumin with IC50 of 17.5 ± 3.2 µM. This effect was mediated by interruption in mitochondrial membrane potential and intracellular free Ca2 + level [93]. In a concentration of 50 µmol/L at 24 h, curcumin induced apoptosis in hepatocellular carcinoma in rats by initiation of reactive oxygen species without any harm to normal hepatocytes in a dose and time dependent manner [91]. In another study, Nano curcumin exerted a preventive and therapeutic effect in diethyl nitrosamine (DEN) induced hepatocellular carcinoma (HCC) in rats [94]. Naringenin has a potent effect against multiple cancer cells [52]. Among multiple flavonoids in citrus seeds extract, cytotoxic activity of naringenin on HepG2 cells was revealed. It provoked apoptosis with IC50 value of 172.00 ± 10.39 µM 24 h [95]. Naringenin significantly reduced Hep G2 cells proliferation in dose-dependent manner after 24 h of incubation with IC50 of 100 µM. Furthermore, it increased p53, Bax, caspase-3 and cytochrome C levels in dose dependent manner and decreased the expression of Bcl-2 in comparison to control [52]. This study points out the importance of Boswellic acids, curcumin and naringenin NPs against hepatocellular carcinoma but because of the financial limitation the anti-cancer mechanism was not explained. In fact, the use of NPs has the advantage to augment herbal effect, improve permeability, decrease clearance and provide herbal retention in hepatocellular carcinoma to reach an optimal efficacy [96]. The promising results encourage more research for new herbal formulations and mechanism interpretation.

Conclusion

Free and nano-encapsulated boswellic acids, curcumin, and naringenin possessed anti-cancer potential by inhibition of HepG2 cell line proliferation. The nanoprecipitation-prepared particles exerted significantly lower IC50 values. This anti-cancer effect carries hope for patients with hepatocellular carcinoma as a safe, and effective therapeutic agent.

Data Availability

The datasets generated and/or analyzed during the current study are not publicly available due to institutional policy but are available from the corresponding author on reasonable request.

References

  1. Akinyemiju T, Abera S, Ahmed M, Alam N, Alemayohu MA, Allen C, et al. The burden of primary liver cancer and underlying etiologies from 1990 to 2015 at the global, regional, and national level: results from the global burden of disease study 2015. JAMA Oncol. 2017;3(12):1683–91.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Anwar WA, Khaled HM, Amra HA, El-Nezami H, Loffredo CA. Changing pattern of hepatocellular carcinoma (HCC) and its risk factors in Egypt: possibilities for prevention. Mutat research/reviews Mutat Res. 2008;659(1–2):176–84.

    Article  CAS  Google Scholar 

  3. El-Serag HB. Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology. 2012;142(6):1264–73. e1.

    Article  PubMed  Google Scholar 

  4. Schulze K, Nault J-C, Villanueva A. Genetic profiling of hepatocellular carcinoma using next-generation sequencing. J Hepatol. 2016;65(5):1031–42.

    Article  CAS  PubMed  Google Scholar 

  5. Alqahtani A, Khan Z, Alloghbi A, Said Ahmed S, Ashraf T, Hammouda MM. Hepatocellular carcinoma: molecular mechanisms and targeted therapies. Medicina. 2019;55(9):526.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Yoon SK. Molecular mechanism of hepatocellular carcinoma. Hepatoma Res. 2018;4(8):42.

    Article  Google Scholar 

  7. Llovet JM, Real MI, Montaña X, Planas R, Coll S, Aponte J, et al. Arterial embolisation or chemoembolisation versus symptomatic treatment in patients with unresectable hepatocellular carcinoma: a randomised controlled trial. Lancet (London England). 2002;359(9319):1734–9.

    Article  PubMed  Google Scholar 

  8. Lo C-M, Ngan H, Tso W-K, Liu C-L, Lam C-M, Poon RT-P, et al. Randomized controlled trial of transarterial lipiodol chemoembolization for unresectable hepatocellular carcinoma. Hepatology. 2002;35(5):1164–71.

    Article  CAS  PubMed  Google Scholar 

  9. Llovet JM, Bruix J. Systematic review of randomized trials for unresectable hepatocellular carcinoma: chemoembolization improves survival. Hepatology. 2003;37(2):429–42.

    Article  CAS  PubMed  Google Scholar 

  10. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359(4):378–90.

    Article  CAS  PubMed  Google Scholar 

  11. Cheng AL, Kang YK, Chen Z, Tsao CJ, Qin S, Kim JS, et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 2009;10(1):25–34.

    Article  CAS  PubMed  Google Scholar 

  12. Kudo M, Finn RS, Qin S, Han K-H, Ikeda K, Piscaglia F, et al. Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3 non-inferiority trial. The Lancet. 2018;391(10126):1163–73.

    Article  CAS  Google Scholar 

  13. Bruix J, Qin S, Merle P, Granito A, Huang Y-H, Bodoky G, et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet. 2017;389(10064):56–66.

    Article  CAS  Google Scholar 

  14. Kalogeridi M-A, Zygogianni A, Kyrgias G, Kouvaris J, Chatziioannou S, Kelekis N, et al. Role of radiotherapy in the management of hepatocellular carcinoma: a systematic review. World J Hepatol. 2015;7(1):101.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Sangro B, Carpanese L, Cianni R, Golfieri R, Gasparini D, Ezziddin S, et al. Survival after yttrium-90 resin microsphere radioembolization of hepatocellular carcinoma across Barcelona clinic liver cancer stages: a european evaluation. Hepatology. 2011;54(3):868–78.

    Article  PubMed  Google Scholar 

  16. Mazzaferro V, Sposito C, Bhoori S, Romito R, Chiesa C, Morosi C, et al. Yttrium-90 radioembolization for intermediate‐advanced hepatocellular carcinoma: a phase 2 study. Hepatology. 2013;57(5):1826–37.

    Article  CAS  PubMed  Google Scholar 

  17. Salem R, Gordon AC, Mouli S, Hickey R, Kallini J, Gabr A, et al. Y90 radioembolization significantly prolongs time to progression compared with chemoembolization in patients with hepatocellular carcinoma. Gastroenterology. 2016;151(6):1155–63. e2.

    Article  PubMed  Google Scholar 

  18. Vilgrain V, Pereira H, Assenat E, Guiu B, Ilonca AD, Pageaux G-P, et al. Efficacy and safety of selective internal radiotherapy with yttrium-90 resin microspheres compared with sorafenib in locally advanced and inoperable hepatocellular carcinoma (SARAH): an open-label randomised controlled phase 3 trial. Lancet Oncol. 2017;18(12):1624–36.

    Article  CAS  PubMed  Google Scholar 

  19. Ikeda M, Mitsunaga S, Ohno I, Hashimoto Y, Takahashi H, Watanabe K, et al. Systemic chemotherapy for advanced hepatocellular carcinoma: past, present, and future. Diseases. 2015;3(4):360–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hasegawa K, Kokudo N, Makuuchi M, Izumi N, Ichida T, Kudo M, et al. Comparison of resection and ablation for hepatocellular carcinoma: a cohort study based on a japanese nationwide survey. J Hepatol. 2013;58(4):724–9.

    Article  PubMed  Google Scholar 

  21. Lu L-C, Cheng A-L, Poon RT, editors. Recent advances in the prevention of hepatocellular carcinoma recurrence. Seminars in liver disease. Thieme Medical Publishers; 2014.

  22. Nahin RL. Costs of complementary and alternative medicine (CAM) and frequency of visits to CAM practitioners: US 2007. Diane Publishing; 2010.

  23. Zhai X-f, Chen Z, Li B, Shen F, Fan J, Zhou W-p, et al. Traditional herbal medicine in preventing recurrence after resection of small hepatocellular carcinoma: a multicenter randomized controlled trial. J Integr Med. 2013;11(2):90–100.

    Article  PubMed  Google Scholar 

  24. Huang MT, Badmaev V, Ding Y, Liu Y, Xie JG, Ho CT. Anti-tumor and anti‐carcinogenic activities of triterpenoid, β‐boswellic acid. BioFactors. 2000;13(1–4):225–30.

    Article  CAS  PubMed  Google Scholar 

  25. Xia L, Chen D, Han R, Fang Q, Waxman S, Jing Y. Boswellic acid acetate induces apoptosis through caspase-mediated pathways in myeloid leukemia cells. Mol Cancer Ther. 2005;4(3):381–8.

    Article  CAS  PubMed  Google Scholar 

  26. Suhail MM, Wu W, Cao A, Mondalek FG, Fung K-M, Shih P-T, et al. Boswellia sacra essential oil induces tumor cell-specific apoptosis and suppresses tumor aggressiveness in cultured human breast cancer cells. BMC Complement Altern Med. 2011;11:1–14.

    Article  Google Scholar 

  27. Shao Y, Ho C-T, Chin C-K, Badmaev V, Ma W, Huang M-T. Inhibitory activity of boswellic acids from Boswellia serrata against human leukemia HL-60 cells in culture. Planta Med. 1998;64(04):328–31.

    Article  CAS  PubMed  Google Scholar 

  28. Hoernlein R, Orlikowsky T, Zehrer C, Niethammer D, Sailer E, Simmet T, et al. Acetyl-11-keto-β-boswellic acid induces apoptosis in HL-60 and CCRF-CEM cells and inhibits topoisomerase I. J Pharmacol Exp Ther. 1999;288(2):613–9.

    CAS  PubMed  Google Scholar 

  29. Pang X, Yi Z, Zhang X, Sung B, Qu W, Lian X, et al. Acetyl-11-keto-β-boswellic acid inhibits prostate tumor growth by suppressing vascular endothelial growth factor receptor 2–mediated angiogenesis. Cancer Res. 2009;69(14):5893–900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Skarke C, Kuczka K, Tausch L, Werz O, Rossmanith T, Barrett JS, et al. Increased bioavailability of 11-keto-β-boswellic acid following single oral dose frankincense extract administration after a standardized meal in healthy male volunteers: modeling and simulation considerations for evaluating drug exposures. J Clin Pharmacol. 2012;52(10):1592–600.

    Article  CAS  PubMed  Google Scholar 

  31. Krüger P, Kanzer J, Hummel J, Fricker G, Schubert-Zsilavecz M, Abdel-Tawab M. Permeation of Boswellia extract in the Caco-2 model and possible interactions of its constituents KBA and AKBA with OATP1B3 and MRP2. Eur J Pharm Sci. 2009;36(2–3):275–84.

    Article  PubMed  Google Scholar 

  32. Hüsch J, Bohnet J, Fricker G, Skarke C, Artaria C, Appendino G, et al. Enhanced absorption of boswellic acids by a lecithin delivery form (Phytosome®) of Boswellia extract. Fitoterapia. 2013;84:89–98.

    Article  PubMed  Google Scholar 

  33. Aqil F, Munagala R, Jeyabalan J, Vadhanam MV. Bioavailability of phytochemicals and its enhancement by drug delivery systems. Cancer Lett. 2013;334(1):133–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Wanga S, Sua R, Niea S, Suna M, Zhanga J, Wub D, et al. Application of nanotechnology in improving bioavailability and bioactivity of dietderived phytochemicals. J Nutr Biochem. 2014;25:363–76.

    Article  Google Scholar 

  35. Aggarwal BB, Surh Y-J, Shishodia S. The molecular targets and therapeutic uses of curcumin in health and disease. Springer Science & Business Media; 2007.

  36. Hasima N, Aggarwal BB. Cancer-linked targets modulated by curcumin. Int J Biochem Mol Biol. 2012;3(4):328.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Mimeault M, Batra SK. Potential applications of curcumin and its novel synthetic analogs and nanotechnology-based formulations in cancer prevention and therapy. Chin Med. 2011;6(1):1–19.

    Article  Google Scholar 

  38. Duan W, Chang Y, Li R, Xu Q, Lei J, Yin C, et al. Curcumin inhibits hypoxia inducible factor–1α–induced epithelial–mesenchymal transition in HepG2 hepatocellular carcinoma cells. Mol Med Rep. 2014;10(5):2505–10.

    Article  CAS  PubMed  Google Scholar 

  39. Li Y, Shi X, Zhang J, Zhang X, Martin RC. Hepatic protection and anticancer activity of curcuma: a potential chemopreventive strategy against hepatocellular carcinoma. Int J Oncol. 2014;44(2):505–13.

    Article  CAS  PubMed  Google Scholar 

  40. Xu MX, Zhao L, Deng C, Yang L, Wang Y, Guo T, et al. Curcumin suppresses proliferation and induces apoptosis of human hepatocellular carcinoma cells via the wnt signaling pathway. Int J Oncol. 2013;43(6):1951–9.

    Article  CAS  PubMed  Google Scholar 

  41. Huang L-Z, Wang J, Lu F-T, Yang F-C, Chen X, Hong X, et al. Mechanism study on anti-proliferative effects of curcumol in human hepatocarcinoma HepG2 cells. Zhongguo Zhong yao za zhi = Zhongguo zhongyao zazhi = China. J Chin materia Med. 2013;38(11):1812–5.

    CAS  Google Scholar 

  42. Chen J, Wang F-L, Chen W-D. Modulation of apoptosis-related cell signalling pathways by curcumin as a strategy to inhibit tumor progression. Mol Biol Rep. 2014;41:4583–94.

    Article  CAS  PubMed  Google Scholar 

  43. Itokawa H, Shi Q, Akiyama T, Morris-Natschke SL, Lee K-H. Recent advances in the investigation of curcuminoids. Chin Med. 2008;3:1–13.

    Article  Google Scholar 

  44. Yallapu MM, Maher DM, Sundram V, Bell MC, Jaggi M, Chauhan SC. Curcumin induces chemo/radio-sensitization in ovarian cancer cells and curcumin nanoparticles inhibit ovarian cancer cell growth. J ovarian Res. 2010;3:1–12.

    Article  Google Scholar 

  45. Mulik RS, Mönkkönen J, Juvonen RO, Mahadik KR, Paradkar AR. Transferrin mediated solid lipid nanoparticles containing curcumin: enhanced in vitro anticancer activity by induction of apoptosis. Int J Pharm. 2010;398(1–2):190–203.

    Article  CAS  PubMed  Google Scholar 

  46. Bredsdorff L, Nielsen ILF, Rasmussen SE, Cornett C, Barron D, Bouisset F, et al. Absorption, conjugation and excretion of the flavanones, naringenin and hesperetin from α-rhamnosidase-treated orange juice in human subjects. Br J Nutr. 2010;103(11):1602–9.

    Article  CAS  PubMed  Google Scholar 

  47. Chabane MN, Ahmad AA, Peluso J, Muller CD, Ubeaud-Séquier G. Quercetin and naringenin transport across human intestinal Caco-2 cells. J Pharm Pharmacol. 2009;61(11):1473–83.

    Article  CAS  Google Scholar 

  48. Heim KE, Tagliaferro AR, Bobilya DJ. Flavonoid antioxidants: chemistry, metabolism and structure-activity relationships. J Nutr Biochem. 2002;13(10):572–84.

    Article  CAS  PubMed  Google Scholar 

  49. Rice-Evans CA, Miller NJ, Paganga G. Structure-antioxidant activity relationships of flavonoids and phenolic acids. Free Radic Biol Med. 1996;20(7):933–56.

    Article  CAS  PubMed  Google Scholar 

  50. Subramanian P, Arul D. Expression of concern: attenuation of NDEA-induced hepatocarcinogenesis by naringenin in rats. Cell Biochem Funct. 2013;31(6):511–7.

    Article  CAS  PubMed  Google Scholar 

  51. Arul D, Subramanian P. Inhibitory effect of naringenin (citrus flavonone) on N-nitrosodiethylamine induced hepatocarcinogenesis in rats. Biochem Biophys Res Commun. 2013;434(2):203–9.

    Article  CAS  PubMed  Google Scholar 

  52. Arul D, Subramanian P. Naringenin (citrus flavonone) induces growth inhibition, cell cycle arrest and apoptosis in human hepatocellular carcinoma cells. Pathol Oncol Res. 2013;19:763–70.

    Article  CAS  PubMed  Google Scholar 

  53. Pafumi I, Festa M, Papacci F, Lagostena L, Giunta C, Gutla V, et al. Naringenin impairs two-pore channel 2 activity and inhibits VEGF-induced angiogenesis. Sci Rep. 2017;7(1):1–11.

    Article  CAS  Google Scholar 

  54. Yen H-R, Liu C-J, Yeh C-C. Naringenin suppresses TPA-induced tumor invasion by suppressing multiple signal transduction pathways in human hepatocellular carcinoma cells. Chemico-Biol Interact. 2015;235:1–9.

    Article  CAS  Google Scholar 

  55. Haggag Y, Elshikh M, El-Tanani M, Bannat IM, McCarron P, Tambuwala MM. Nanoencapsulation of sophorolipids in PEGylated poly (lactide-co-glycolide) as a novel approach to target colon carcinoma in the murine model. Drug Delivery and Translational Research. 2020;10:1353–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Zewail MB, El-Gizawy SA, Osman MA, Haggag YA. Preparation and in vitro characterization of a novel self-nano emulsifying drug delivery system for a fixed-dose combination of candesartan cilexetil and hydrochlorothiazide. J Drug Deliv Sci Technol. 2021;61:102320.

    Article  CAS  Google Scholar 

  57. Ibrahim B, Mady OY, Tambuwala MM, Haggag YA. pH-sensitive nanoparticles containing 5-fluorouracil and leucovorin as an improved anti-cancer option for colon cancer. Nanomedicine. 2022;17(6):367–81.

    Article  CAS  PubMed  Google Scholar 

  58. Salem MA, Manaa EG, Osama N, Aborehab NM, Ragab MF, Haggag YA, et al. Coriander (Coriandrum sativum L.) essential oil and oil-loaded nano-formulations as an anti-aging potentiality via TGFβ/SMAD pathway. Sci Rep. 2022;12(1):6578.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Nakhaei K, Bagheri-Hosseini S, Sabbaghzade N, Behmadi J, Boozari M. Boswellic Acid Nanoparticles: promising strategies for increasing Therapeutic Effects. Revista Brasileira de Farmacognosia; 2023.

  60. Dos Santos PDF, Francisco CRL, Coqueiro A, Leimann FV, Pinela J, Calhelha RC, et al. The nanoencapsulation of curcuminoids extracted from Curcuma longa L. and an evaluation of their cytotoxic, enzymatic, antioxidant and anti-inflammatory activities. Food Funct. 2019;10(2):573–82.

    Article  CAS  PubMed  Google Scholar 

  61. Wang W, Liu Q, Liang X, Kang Q, Wang Z. Protective role of naringin loaded solid nanoparticles against aflatoxin B1 induced hepatocellular carcinoma. Chemico-Biol Interact. 2022;351:109711.

    Article  CAS  Google Scholar 

  62. Houssen ME, Ragab A, Mesbah A, El-Samanoudy AZ, Othman G, Moustafa AF, et al. Natural anti-inflammatory products and leukotriene inhibitors as complementary therapy for bronchial asthma. Clin Biochem. 2010;43(10–11):887–90.

    Article  CAS  PubMed  Google Scholar 

  63. Badria FA, Mohammed EA, El-Badrawy MK, El-Desouky M. Natural leukotriene inhibitor from Boswellia: a potential new alternative for treating bronchial asthma. Altern Complement Ther. 2004;10(5):257–65.

    Article  Google Scholar 

  64. Badria FA, El-Farahaty T, Shabana AA, Hawas SA, El-Batoty MF. Boswellia–curcumin preparation for treating knee osteoarthritis: a clinical evaluation. Altern Complement Ther. 2002;8(6):341–8.

    Article  Google Scholar 

  65. Badria FA. A potent hepatoprotective agent from group fruit. Alexandria J Pharm Sci. 1994;8(3):165–9.

    CAS  Google Scholar 

  66. Khan MN, Haggag YA, Lane ME, McCarron PA, Tambuwala MM. Polymeric nano-encapsulation of curcumin enhances its anti-cancer activity in breast (MDA-MB231) and lung (A549) cancer cells through reduction in expression of HIF-1α and nuclear p65 (REL A). Curr Drug Deliv. 2018;15(2):286–95.

    Article  CAS  PubMed  Google Scholar 

  67. Fessi H, Puisieux F, Devissaguet JP, Ammoury N, Benita S. Nanocapsule formation by interfacial polymer deposition following solvent displacement. Int J Pharm. 1989;55(1):R1–R4.

    Article  CAS  Google Scholar 

  68. Fonseca-Santos B, Gremião MPD, Chorilli M. A simple reversed phase high-performance liquid chromatography (HPLC) method for determination of in situ gelling curcumin-loaded liquid crystals in in vitro performance tests. Arab J Chem. 2017;10(7):1029–37.

    Article  CAS  Google Scholar 

  69. Bhandari R, Kuhad A, Paliwal JK, Kuhad A. Development of a new, sensitive, and robust analytical and bio-analytical RP-HPLC method for in-vitro and in-vivo quantification of naringenin in polymeric nanocarriers. J Anal Sci Technol. 2019;10(1):1–14.

    Article  Google Scholar 

  70. Bairwa K, Jachak SM. Nanoparticle formulation of 11-keto-β-boswellic acid (KBA): anti-inflammatory activity and in vivo pharmacokinetics. Pharm Biol. 2016;54(12):2909–16.

    Article  CAS  PubMed  Google Scholar 

  71. Haggag YA, Ibrahim RR, Hafiz AA. Design, formulation and in vivo evaluation of novel honokiol-loaded PEGylated PLGA nanocapsules for treatment of breast cancer. Int J Nanomed. 2020:1625–42.

  72. Zhang K, Zhang L, Liu W, Ma X, Cen J, Sun Z, et al. In vitro expansion of primary human hepatocytes with efficient liver repopulation capacity. Cell Stem Cell. 2018;23(6):806–19. e4.

    Article  CAS  PubMed  Google Scholar 

  73. Ahmad N, Ahmad R, Alrasheed RA, Almatar HMA, Al-Ramadan AS, Buheazah TM, et al. A Chitosan-PLGA based catechin hydrate nanoparticles used in targeting of lungs and cancer treatment. Saudi J Biol Sci. 2020;27(9):2344–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Farazi PA, DePinho RA. Hepatocellular carcinoma pathogenesis: from genes to environment. Nat Rev Cancer. 2006;6(9):674–87.

    Article  CAS  PubMed  Google Scholar 

  75. Ullah MF, Khan MW. Food as medicine: potential therapeutic tendencies of plant derived polyphenolic compounds. Asian Pac J Cancer Prev. 2008;9(2):187–96.

    PubMed  Google Scholar 

  76. Moiseeva EP, Manson MM. Dietary chemopreventive phytochemicals: too little or too much? Cancer Prev Res. 2009;2(7):611–6.

    Article  CAS  Google Scholar 

  77. Cragg GM, Newman DJ. Plants as a source of anti-cancer agents. J Ethnopharmacol. 2005;100(1–2):72–9.

    Article  CAS  PubMed  Google Scholar 

  78. Zulkipli IN, David SR, Rajabalaya R, Idris A. Medicinal plants: a potential source of compounds for targeting cell division. Drug target insights. 2015;9:DTI.

    Article  Google Scholar 

  79. Haggag YA, Faheem AM. Evaluation of nano spray drying as a method for drying and formulation of therapeutic peptides and proteins. Frontiers Media SA; 2015. p. 140.

  80. Haggag YA, Matchett KB, Dakir E-H, Buchanan P, Osman MA, Elgizawy SA, et al. Nano-encapsulation of a novel anti-Ran-GTPase peptide for blockade of regulator of chromosome condensation 1 (RCC1) function in MDA-MB-231 breast cancer cells. Int J Pharm. 2017;521(1–2):40–53.

    Article  CAS  PubMed  Google Scholar 

  81. Haggag Y, Abdel-Wahab Y, Ojo O, Osman M, El-Gizawy S, El-Tanani M, et al. Preparation and in vivo evaluation of insulin-loaded biodegradable nanoparticles prepared from diblock copolymers of PLGA and PEG. Int J Pharm. 2016;499(1–2):236–46.

    Article  CAS  PubMed  Google Scholar 

  82. Haggag YA, Faheem AM, Tambuwala MM, Osman MA, El-Gizawy SA, O’Hagan B, et al. Effect of poly (ethylene glycol) content and formulation parameters on particulate properties and intraperitoneal delivery of insulin from PLGA nanoparticles prepared using the double-emulsion evaporation procedure. Pharm Dev Technol. 2018;23(4):370–81.

    Article  CAS  PubMed  Google Scholar 

  83. Alshamsan A. Nanoprecipitation is more efficient than emulsion solvent evaporation method to encapsulate cucurbitacin I in PLGA nanoparticles. Saudi Pharm J. 2014;22(3):219–22.

    Article  PubMed  Google Scholar 

  84. Park YS, Lee JH, Bondar J, Harwalkar JA, Safayhi H, Golubic M. Cytotoxic action of acetyl-11-keto-β-boswellic acid (AKBA) on meningioma cells. Planta Med. 2002;68(05):397–401.

    Article  CAS  PubMed  Google Scholar 

  85. Shanmugam MK, Nguyen AH, Kumar AP, Tan BK, Sethi G. Targeted inhibition of tumor proliferation, survival, and metastasis by pentacyclic triterpenoids: potential role in prevention and therapy of cancer. Cancer Lett. 2012;320(2):158–70.

    Article  CAS  PubMed  Google Scholar 

  86. Liu J-J, Nilsson Ã, Oredsson S, Badmaev V, Zhao W-Z, Duan R-D. Boswellic acids trigger apoptosis via a pathway dependent on caspase-8 activation but independent on Fas/Fas ligand interaction in colon cancer HT-29 cells. Carcinogenesis. 2002;23(12):2087–93.

    Article  CAS  PubMed  Google Scholar 

  87. Ahmed HH, Abd-Rabou AA, Hassan AZ, Kotob SE. Phytochemical analysis and anti-cancer investigation of Boswellia serrata bioactive constituents in vitro. Asian Pac J Cancer Prev. 2015;16(16):7179–88.

    Article  PubMed  Google Scholar 

  88. Khan MA, Singh M, Khan MS, Najmi AK, Ahmad S. Caspase mediated synergistic effect of Boswellia serrata extract in combination with doxorubicin against human hepatocellular carcinoma. BioMed research international. 2014;2014.

  89. Song X, Zhang M, Dai E, Luo Y. Molecular targets of curcumin in breast cancer. Mol Med Rep. 2019;19(1):23–9.

    CAS  PubMed  Google Scholar 

  90. Aggarwal BB, Kumar A, Bharti AC. Anticancer potential of curcumin: preclinical and clinical studies. Anticancer Res. 2003;23(1/A):363–98.

    CAS  PubMed  Google Scholar 

  91. Syng-Ai C, Kumari AL, Khar A. Effect of curcumin on normal and tumor cells: role of glutathione and bcl-2. Mol Cancer Ther. 2004;3(9):1101–8.

    Article  CAS  PubMed  Google Scholar 

  92. Kunwar A, Barik A, Mishra B, Rathinasamy K, Pandey R, Priyadarsini K. Quantitative cellular uptake, localization and cytotoxicity of curcumin in normal and tumor cells. Biochim et Biophys Acta (BBA)-General Subj. 2008;1780(4):673–9.

    Article  CAS  Google Scholar 

  93. Wang M, Ruan Y, Chen Q, Li S, Wang Q, Cai J. Curcumin induced HepG2 cell apoptosis-associated mitochondrial membrane potential and intracellular free Ca2 + concentration. Eur J Pharmacol. 2011;650(1):41–7.

    Article  CAS  PubMed  Google Scholar 

  94. Hassan SK, Mousa AM, Eshak MG, Farrag A, Badawi A. Therapeutic and chemopreventive effects of nano curcumin against diethylnitrosamine induced hepatocellular carcinoma in rats. Int J Pharm Pharm Sci. 2014;6(3):54–62.

    Google Scholar 

  95. Banjerdpongchai R, Wudtiwai B, Khaw-On P, Rachakhom W, Duangnil N, Kongtawelert P. Hesperidin from Citrus seed induces human hepatocellular carcinoma HepG2 cell apoptosis via both mitochondrial and death receptor pathways. Tumor Biology. 2016;37:227–37.

    Article  CAS  PubMed  Google Scholar 

  96. Gad A, Kydd J, Piel B, Rai P. Targeting cancer using polymeric nanoparticle mediated combination chemotherapy. Int J Nanomed nanosurgery. 2016;2(3).

Download references

Acknowledgements

We would like to thank the staff members of cell culture unit of Tanta University Central laboratories as well as the researchers of Nanotechnology center, Kafr Elsheikh University for their kind support.

Funding

This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Open access funding provided by The Science, Technology & Innovation Funding Authority (STDF) in cooperation with The Egyptian Knowledge Bank (EKB).

Author information

Authors and Affiliations

Authors

Contributions

S.E. designed and supervised the study. Y. H. , S.S. and N.S. performed all the experiments. A.E., F.B. analyzed the spectral characteristics of the compound and statistical data were interpreted by M.I. All authors reviewed the manuscript.

Corresponding author

Correspondence to Sally Elnawasany.

Ethics declarations

Ethics approval and consent to participate

The study was approved by the Research Ethics Committee of the Faculty of Medicine, Tanta University. Approval code: 36264PR237623.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Elnawasany, S., Haggag, Y.A., Shalaby, S.M. et al. Anti-cancer effect of nano-encapsulated boswellic acids, curcumin and naringenin against HepG-2 cell line. BMC Complement Med Ther 23, 270 (2023). https://doi.org/10.1186/s12906-023-04096-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12906-023-04096-4

Keywords