Next Article in Journal
Electrospun Scaffolds for Tissue Engineering: A Review
Next Article in Special Issue
Development of Water-Resistant Autohesive Strength of Polyethylene Plates with Photografting of Alkyl (Meth)Acrylates
Previous Article in Journal
Oxo-Additives for Polyolefin Degradation: Kinetics and Mechanism
Previous Article in Special Issue
Agar-Agar and Chitosan as Precursors in the Synthesis of Functional Film for Foods: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Polyphenol-Loaded Polymeric Matrixes as Potential Biopharmaceuticals against Cancer

by
Manuel Adrian Picos-Salas
1,
Melissa García-Carrasco
1,
José Basilio Heredia
1,
Luis Angel Cabanillas-Bojórquez
1,2,
Nayely Leyva-López
1,2 and
Erick Paul Gutiérrez-Grijalva
3,*
1
Laboratory of Functional Foods and Nutraceuticals, Centro de Investigación en Alimentación y Desarrollo, A.C., Carretera a Eldorado Km. 5.5, Col. Campo El Diez, Culiacan CP 80110, Mexico
2
Posdoc CONAHCYT-Centro de Investigación en Alimentación y Desarrollo, A.C., Carretera a Eldorado Km. 5.5, Col. Campo El Diez, Culiacan CP 80110, Mexico
3
Catedras CONAHCYT-Centro de Investigación en Alimentación y Desarrollo, A.C., Carretera a Eldorado Km. 5.5, Col. Campo El Diez, Culiacan CP 80110, Mexico
*
Author to whom correspondence should be addressed.
Macromol 2023, 3(3), 507-523; https://doi.org/10.3390/macromol3030030
Submission received: 15 May 2023 / Revised: 11 July 2023 / Accepted: 1 August 2023 / Published: 3 August 2023
(This article belongs to the Special Issue Functionalization of Polymers for Advanced Applications)

Abstract

:
Polyphenols have attracted attention for their anti-inflammatory, antidiabetic, and anticancer properties. Due to the antioxidant and anti-inflammatory potential of these molecules, they are also proposed as a potential therapeutic tool to prevent complications of cancer and decrease the secondary effects of conventional chemotherapeutic drugs. Nonetheless, polyphenols such as flavonoids and phenolic acids have low bioavailability, as they are highly metabolized. Thus, administration strategies have been developed to enhance the anticancer properties of polyphenols. Most of these strategies involve different encapsulation techniques, such as nanoencapsulation, nanoemulsion, and the use of other polymeric matrixes. These techniques can increase the activity of these compounds after going through the gastrointestinal process and improve their solubility in an aqueous medium. This review comprises recent studies regarding encapsulation techniques to enhance the bioactivity of polyphenols against cancer and their current state in clinical studies. Overall, micro- and nanoencapsulation techniques with different polymers enhanced the anticancer properties of polyphenols by inhibiting tumor growth, modulating the expression of genes related to metastasis and angiogenesis, decreasing the expression of pro-inflammatory biomarkers.

Graphical Abstract

1. Introduction

Phenolic compounds, known by the general audience as “polyphenols”, are secondary metabolites from plants produced mainly as a defense mechanism against abiotic and biotic factors. These constituents also contribute to pollination by granting color to flowers and plants, attracting pollinators [1]. For their study, polyphenols can be grouped according to their chemical structure, number of phenol rings, and number and distribution of their -OH groups. The most common groups are (1) flavonoids: anthocyanins, chalcones, dihydrochalcones, dihydroflavonols, flavanols, flavanones, flavones, flavonols, isoflavonoids; (2) lignans, (3) non-phenolic compounds, (4) phenolic acids: hydroxybenzoic acids, hydroxycinnamic acids, hydroxyphenylacetic acids, hydroxyphenylpropanoic acids, hydroxyphenylpentanoic acids, (5) stilbenes, and (6) other polyphenols (Figure 1) [2,3,4]. Polyphenols are of interest in the nutraceutical and pharmaceutical industries because of their antioxidant, anti-inflammatory, and anticancer properties.
Cancer is a group of diseases that begins when abnormal cells grow uncontrollably and can start in nearly all tissues and organs. Cancer is the second leading cause of death worldwide, with around 9.6 million deaths in 2018. According to the International Agency for Research on Cancer of the World Health Organization, the cancers that caused most deaths in 2020 were lung, colorectum, liver, stomach, breast, esophagus, pancreas, and prostate cancer (Figure 2).
Currently, there is evidence that a regular intake of food sources rich in polyphenols can decrease cancer incidence and prevent disease onset. In vitro studies have shown that many polyphenols have antiproliferative activity through several mechanisms of action involved in signaling cascades in the different cancer development stages [6,7]. Furthermore, in vivo studies have also shown that polyphenols can exert an anticancer potential also at this level. Nonetheless, one of the problems dealing with polyphenols is their low bioavailability because the xenobiotic metabolism highly metabolizes them. In this sense, encapsulation strategies using polymeric matrixes have been developed to increase the bioavailability of polyphenols. This review summarizes recent studies on encapsulation strategies and the available in vivo evidence of their anticancer potential.

2. Materials and Methods

This work was compiled based on recent literature (2012–2023) from the Scopus, Google Scholar, and Web of Science databases to identify relevant studies on this topic. To identify relevant literature, we combined the keywords polyphenols, flavonoids, phenolic acids, and phenolic compounds with the following keywords: microencapsulation, nanoencapsulation, nanoemulsions, and cancer, anticancer, and antiproliferative. In addition, we decided to include only literature in the English language. Finally, we grouped 91 research papers for this manuscript.

3. Anticancer Properties of Polyphenols

3.1. In Vivo Studies

Different studies in mice and rats showed the potential anticancer properties of polyphenols (Table 1). They can act at different levels, including by apoptosis induction of tumor cells in different ways. In this sense, this type of cell death can be induced by ellagic acid and galangin via increased caspases-3 expression and activity in retinoblastoma tumors and lymphomas [8,9], as well as by an increase in Bad and a decrease in Bcl-2 gene expression by vanillic acid [10]. In addition, it was observed that polyphenols decrease metastasis by inhibiting the migration and invasion of cancer cells; among them, eriodictyol and luteolin showed this effect [11,12]. Furthermore, taxifolin decreased epithelial–mesenchymal transition, an early event of metastasis [13].
In addition, signaling pathways such as PI3K/Akt and MAPK/ERK are related to cell proliferation, which could be downregulated by caffeic acid and caffeic acid phenylpropyl ester in induced colon cancer [14].
On the other hand, inflammation and oxidative stress are two physiological processes which have been linked to cancer [15]; in this context, naringenin had an anticancer effect in the lungs by decreasing the expression of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) (a bridge between inflammation and cancer), which decreased the expression of cytokines such as interleukin (IL)-6, IL-8, and tumor necrosis factor (TNF-α); at the same time, this flavanone could increase the antioxidant enzymes and non-enzymatic antioxidants, exerting a cytoprotective function against induced lung cancer [16]. Moreover, taxifolin increased the antioxidant response by the upregulation of the nuclear factor erythroid 2-related factor 2 (Nrf2) and decreased inflammation by downregulation of the NF-κB and Wnt/β-catenin signaling pathways in induced colon cancer [17]; also, Nrf2 upregulation in induced hepatic cancer by vanillic acid was detected [10].
Table 1. Effect of polyphenols supplementation against cancer in rats and mice models.
Table 1. Effect of polyphenols supplementation against cancer in rats and mice models.
CompoundDoseModelEffectReference
Caffeic acid50 nmol/kgMice with human colon cancer xenograftsInhibition of tumor growth via downregulation of PI3K/Akt and MAPK/ERK signaling[14]
Caffeic acid phenylpropyl ester50 nmol/kgMice with human colon cancer xenograftsInhibition of tumor growth via downregulation of PI3K/Akt and MAPK/ERK signaling[14]
Chlorogenic acid20–40 mg/kgMice with breast cancer xenograftsDecrease in tumor growth and inhibition of metastasis via an increase in CD4+ and CD8+ cells in the spleen[18]
Ellagic acid40 mg/kgMice with human bladder cancer xenograftsDecrease in tumor growth rate, infiltrative behavior, and tumor-associated angiogenesis.[19]
80 mg/kgMice with induced lymphomaInduction of apoptosis via an increase in caspase-3 expression and activity and PKCs activity and a decrease in LDH-A activity and expression in ascites fluid[8]
Eriodictyol60 mg/kgMice with mammary cancer xenograftsDecrease in tumor growth and progression and in lung metastasis[11]
Galangin25–50 mg/kgMice with human retinoblastoma xenograftsDecrease in tumor growth via a decrease in Akt signaling pathway and increase in caspase-3 level[9]
Luteolin1.2 mg/gMice with AOM/DMH-induced colon cancerDecrease in LDH levels and in iNOS and COX-2 expression in colon tissue[20]
100 mg/kgMice with human epithelial xenograftDecrease in migration and invasion[12]
Naringenin50 mg/kgMice with benzo(a)pyrene induced lung cancerDownregulation of CYP1A1, PCNA, and NF-κB expression; decrease in lipid peroxidation, TNF-α, IL-6 and IL-1β; increase in antioxidant enzymes activity in lung tissue[16]
Quercetin30 mg/kgMice with AOM/dextran sodium sulfate-induced colorectal cancerDecrease in tumor growth and proliferation via a decrease in inflammation and ROS[21]
25–50 mg/kgRats with DMH-induced colon cancerDecrease in tumor incidence and multiplicity; downregulation of the Wnt signaling pathway in colon tissue[22]
Taxifolin4 µg/kgMice with DMH-induced colon cancerUpregulation of the Nrf2 signaling pathway, downregulation of the NF-κB and Wnt signaling pathways in colon tissue[17]
1 mg/kgMice with human lung cancer xenograftDecrease in tumor size via inhibition of PI3K and TCF4 signaling and by decreasing epithelial–mesenchymal transition[13]
Vanillic acid75 mg/kgRats with DMH-induced hepatic cancerUpregulation of the Nrf2 signaling pathway; induction of apoptosis via an increase in Bad and Caspase-3 genes expression and decrease in Bcl-2 gene expression; decrease in proliferation via a decrease in Cyclin D1 gene expression in hepatic tissue[10]
On the other hand, studies on different populations have shown that the intake of polyphenols is associated with a reduced cancer risk (Table 2). In this context, the concentration in the serum of flavonoids such as kaempferol and naringenin was associated with a lower breast cancer risk [23]; similarly, anthocyanidins and flavan-3-ols caused similar effects with synergism with butyl benzyl phthalate [24]. Also, the phenolic acids ferulic acid and caffeic acid reduced the risk to develop prostate cancer [25], and bladder cancer risk showed an inverse association with flavonols and lignans intake [26]. Interestingly, a study demonstrated an association between thyroid cancer risk and the intake of flavonoids and phenolic acid, but only in patients with a BMI ≥ 25, which can be attributed to the anti-inflammatory activity of these compounds, as obesity is a low-grade inflammation disease [27]. However, some studies indicated no association between polyphenols consumption and decrease in cancer risk, including colorectal, pancreatic, and epithelial ovarian cancer [28,29,30,31]. In contrast, a study in the Iranian population showed a lower colorectal cancer risk associated with polyphenols consumption, indicating variation between populations [32].

3.2. Clinical Studies

The effect of polyphenols supplementation has been studied on various cancers, including bladder, colorectal, oral, and prostate cancer (Table 3). For example, the compound curcumin has been studied for both oral cancer and familial adenomatous polyps (a condition that leads to colorectal cancer); in this sense, curcumin decreased interleukin content in the saliva and induced the attraction of T cells to the tumor microenvironment [34]. In contrast, no effect was observed on the mean size and number of polyps in the lower intestinal tract [35]. On the other hand, studies indicated no beneficial effect of epigallocatechin gallate against colorectal and prostate cancer [36,37]; in contrast, cranberry fruit powder rich in phenolic compounds was able to reduce the serum level of prostate-specific antigen in patients after prostate cancer removal, serving as a prophylactic agent against the recurrence of this disorder [38]. Furthermore, a ginger extract with 5% of gingerols decreased the proliferation of crypts in patients with a high risk of colorectal cancer [39].
These studies indicate a possible beneficial effect of different kinds of polyphenols against various cancers; however, their limitations should be considered, including a small sample size and the acute administration of polyphenols in some studies. Thus, more solid and robust clinical studies are necessary, considering the high variety of polyphenols and types of cancer, as well as other strategies to improve polyphenols bioaccessibility and bioavailability.

4. Polymeric Matrixes to Encapsulate Polyphenols

Polyphenolic compounds are secondary metabolites found more frequently in fruits and plants; several beneficial health effects have been attributed to them, and there is also the possibility of using them as adjuvants or biopharmaceuticals to treat chronic degenerative diseases such as cancer. However, due to the low solubility and low bioavailability that they present, in recent years, different alternatives have been sought to increase both their solubility in an aqueous medium and their bioavailability, as well as to increase their protection in the medium in which they are dissolved, since the majority of these compounds are susceptible to changes caused by light, temperature, pH, etc. [41]. That is why different alternatives have been proposed to encapsulate them by various techniques such as spray drying, nanoemulsion, coacervation, films, and others, which help these compounds enhance their activity in some cases. In Table 4, we can observe some of the advantages and disadvantages of encapsulated vs. non-encapsulated phenolic compounds, emphasizing that different amounts of these compounds may encapsulated using the different techniques, due to their chemical and/or physicochemical nature. For this reason, most researchers try not to use encapsulation methods that can damage or deteriorate the phenolic compounds [42,43,44,45,46].
Furthermore, the variety of functional groups in these compounds means that they may establish different covalent or non-covalent interactions and coordinate with diverse metals (See Figure 3).
Chitosan, being a natural cationic polysaccharide, shows high biocompatibility, low toxicity and biodegradability. Cyclodextrins are among the most widely used oligomers of natural origin for the encapsulation of active ingredients of natural origin and have been modeled for the encapsulation of different synthetic and natural active compounds, such as polyphenolic compounds [47]. Due to the polycationic characteristics that these matrices usually have, they usually contain groups that are capable of ionizing to establish an electrostatic interaction with free -OH groups (hydrogen bonds) or can produce esterification reactions with carbonyl groups to generate covalent bonds, as well as coordinate with different metals; so, a high content of these groups (-OH) in polyphenolic compounds could help these compounds to encapsulate in a greater proportion [47,48,49,50,51,52].
For this reason, the formulation of capsules with synthetic polymers and biopolymers with polycationic characteristics such as chitosan, albumin, whey protein, poly(d,l-lactide-co-glycolide (PGLA), among others, is normally used [53,54,55]. The following Table 5 provides a summary of some of the polymers used for the encapsulation of phenolic compounds on the nanometric scale (1–1000 nm).
For microencapsulation (1–1000 μM), biopolymers such as maltodextrin, gum arabic, B-cyclodextrin, modified chitosan (water soluble), sodium alginate, whey protein, among others, are commonly used. The techniques used to prepare these microcapsules vary and include spay drying, lyophilization, extrusion, freeze–drying, emulsion, etc. Unlike nanoencapsulation, non-covalent interactions predominate in microencapsulation (hydrogen bonds, electrostatics, etc.) [73,74,75,76,77]. In the following Table 6 some examples of microencapsulation are shown.

5. Studies with Polyphenol-Loaded Polymeric Matrixes with Anticancer Properties

We aimed to analyze information on the evidence of the pharmaceutical potential of polyphenols in polymeric matrixes. This section aims to summarize the recent information on in vivo studies using nano- or microparticles loaded with purified polyphenols (alone or in combination). So far, our literature research found few reports that we will mention. Encapsulation of polyphenols is being studied to increase the effectiveness of anti-cancer drugs. For instance, in a melanoma skin cancer mice model, treatment with nanoparticles containing co-encapsulated curcumin and chrysin in poly lactic-co-glycolic acid (PLGA) at the two concentrations of 15 mg/kg each and 30 mg/kg each showed better inhibition of tumor growth than treatment with the single encapsulated compounds. In addition, the treatment with the co-encapsulated polyphenols also decreased the expression of the MMP-9, MMP-2, and TERT genes, which are involved in metastasis and angiogenesis [81].
Hesperedin-conjugated gold nanoparticles in Wistar albino rats with diethylnitrosamine-induced hepatocarcinogenesis decreased the expression of the pro-inflammatory biomarkers TNF-α and NF-κB in the treated animals, which are involved in cancer proliferation. Moreover, the treatment with encapsulated hesperedin also decreased the levels of the proliferative marker proliferating cell nuclear antigen [82].
Baohuoside I (50 mg/kg) micelles mixed with lecithin and Solutol HS 15 in nude mice showed antitumor activity against non-small cell lung cancer by inhibiting tumor growth. The authors mentioned that the combination of bahuaoside I and Solutol could improve the cytotoxicity against this type of cancer cells due to the drug’s reduced efflux by inhibition of glycoprotein P.
Chitosane–sodium alginate microencapsulated quercetin (15 mg/100 g diet) with Bifidobacterium bifidum and Lactobacillus gasseri (107 CFU/100 g diet each) supplemented to C57BL/6J mice (ApcMin/+ mice model for gut cancer) significantly reduced the number of aberrant crypt foci by 45–57% and the number of adenomas by 60–80%. Moreover, after 73 days, the treatment modulated the regulation of the Wnt/β-catenin signaling pathway (polyp formation and colorectal cancer development). Therefore, it was hypothesized that the action against colorectal cancer of the co-encapsulation was partially mediated by the anti-inflammatory properties of quercetin and by the suppression of colonic mucosa cellular proliferation by the probiotics’ effect [80].
A study by Khan et al. [83] on a naringenin- (D2) loaded PLGA–doxorubicin nanoparticles treatment in Sprague-Dawley rats showed improved naringenin bioavailability and its distribution mainly in the liver. Furthermore, the author showed that the nanoencapsulated particles had antitumor properties in nude female mice with ectopic MCF-7-induced tumors, as the treatment decreased the tumor size from 2220 mm3 in the control animals to 75.78 mm3 in the treated animals.
Das et al. [84] formulated nanoparticles with apigenin and poly(lactic-co-glycolide) and administered them to Sprague Dawley rats with skin cancer induced by UVB irradiation and the carcinogen BaP. Oral and topical nanoencapsulated apigenin delayed the tumor onset and tumor multiplicity. The study also showed that the treatment decreased the damage in the epidermis and dermis, which could be related to the delayed tumor formation. Further analysis showed that apigenin administration reduced the expression levels of PCNA, upregulated the expression of apaf-1, caspase 9, and modulated the bax/bcl-2 ratio.
A study by Siddiqui et al. [85] evaluated the anticancer properties of epigallocatechin-3-gallate-encapsulated chitosan nanoparticles administered orally, five times a week, in athymic male nude mice implanted with Mel 928 cells (human melanoma cell xenograft model). It was reported that the nanoencapsulated flavanol inhibited the growth of melanoma tumors in the treated mice, without toxicity. Furthermore, one of the possible mechanisms of action by which this happened might involve the observed increased levels of pro-apoptotic Bax and the decreased anti-apoptotic Bcl-2 levels. Also, it was observed that the treatment modulated cyclin-dependent kinases such as CDK 4 and CDK 6 and decreased the expression levels of Ki-67 and PCNA, involved in tumor progression and cell proliferation.
Calcium phosphate-nanoencapsulated resveratrol was tested in a Swiss albino mouse model of skin cancer using 0.5 and 1 µg of nanoresveratrol; 30 min after treatment, the animals received topical 7,12-dimethylbenz[a]anthracene (DMBA)-initiated/12-O-tetradecanoylphorbol-13-acetate (TPA) to promote two-stage skin cancer. The calcium phosphate had a nanoencapsulation efficiency of 85% [86]. Moreover, edema in mice decreased using 10 µg of resveratrol alone, but this effect was observed with 0.5 µg of nanoencapsulated nanoresveratrol, which was attributed to the better stability of nanoencapsulated resveratrol and higher antioxidant and its anti-inflammatory activity; this also resulted in lower incidence and burden of the skin tumors. In both cases, calcium phosphate-nanoencapsulated resveratrol showed better results than non-encapsulated resveratrol.
In Table 7, we show a summarized revision of cases in which the anticancer properties of encapsulated polyphenols were evaluated using in vivo models.
In general, polyphenol delivery with polymers enhanced their anticancer properties by inducing cell cycle arrest, inducing apoptosis, enhancing antioxidant defense enzyme activity (such as superoxide dismutase activity), inhibiting cell proliferation, metastasis, and angiogenesis [87].
There are some studies that delt with polymer-loaded and -encapsulated polyphenols that evaluated their anticancer potential in vivo. However, preclinical and clinical studies are scarce. Further research on the pharmaceutical properties of these molecules should be carried out. Clinical studies are needed to evaluate their potential as pharmaceutical agents for the treatment or coadjuvant treatment of cancer.

6. Conclusions

Studies have shown that the encapsulation of polyphenols can lead to several pharmaceutical advantages, such as increased bioavailability and distribution to tissues and organs of polyphenols, decreased side effects of the currently used chemotherapy agents, prevention of drug cellular efflux (thus, increasing drug bioavailability), and can modulate the expression levels of biomarkers related to cell proliferation, tumor growth, and progression. With the biopharmaceutical perspective in mind, further studies should focus on evaluating the anticancer mechanisms of polyphenols loaded in macro and nano polymeric matrixes. So far, few studies have focused on this subject. However, given the increasing prevalence and economic burden of cancer worldwide, more attention should be given to the potential of polyphenols as chemotherapeutic agents or as coadjuvants of the currently used chemotherapy agents.

Author Contributions

Conceptualization, E.P.G.-G.; methodology, E.P.G.-G. and M.A.P.-S.; writing—original draft preparation, M.A.P.-S., M.G.-C., L.A.C.-B., N.L.-L., E.P.G.-G.; writing—review and editing, E.P.G.-G. and J.B.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

E.P.G.G. would like to thank the “Cátedras CONACYT Project #397: Fortalecimiento del grupo de investigación del potencial nutracéutico de plantas medicinales”.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Quideau, S.; Deffieux, D.; Douat-Casassus, C.; Pouységu, L. Plant Polyphenols: Chemical Properties, Biological Activities, and Synthesis. Angew. Chem. Int. Ed. 2011, 50, 586–621. [Google Scholar] [CrossRef]
  2. Neveu, V.; Perez-Jiménez, J.; Vos, F.; Crespy, V.; du Chaffaut, L.; Mennen, L.; Knox, C.; Eisner, R.; Cruz, J.; Wishart, D.; et al. Phenol-Explorer: An online comprehensive database on polyphenol contents in foods. Database 2010, 2010, bap024. [Google Scholar] [CrossRef]
  3. Rothwell, J.A.; Perez-Jimenez, J.; Neveu, V.; Medina-Remón, A.; M’Hiri, N.; García-Lobato, P.; Manach, C.; Knox, C.; Eisner, R.; Wishart, D.S.; et al. Phenol-Explorer 3.0: A major update of the Phenol-Explorer database to incorporate data on the effects of food processing on polyphenol content. Database 2013, 2013, bat070. [Google Scholar] [CrossRef]
  4. Rothwell, J.A.; Urpi-Sarda, M.; Boto-Ordoñez, M.; Knox, C.; Llorach, R.; Eisner, R.; Cruz, J.; Neveu, V.; Wishart, D.; Manach, C.; et al. Phenol-Explorer 2.0: A major update of the Phenol-Explorer database integrating data on polyphenol metabolism and pharmacokinetics in humans and experimental animals. Database 2012, 2012, bas031. [Google Scholar] [CrossRef]
  5. Ferlay, J.; Ervik, M.; Lam, F.; Colombet, M.; Mery, L.; Piñeros, M.; Znaor, A.; Soerjomataram, I.; Bray, F. Global Cancer Observatory: Cancer Today. International Agency for Research on Cancer: Lyon, France. Available online: https://gco.iarc.fr/today (accessed on 11 May 2023).
  6. Chen, A.Y.; Chen, Y.C. A review of the dietary flavonoid, kaempferol on human health and cancer chemoprevention. Food Chem. 2013, 138, 2099–2107. [Google Scholar] [CrossRef] [Green Version]
  7. Naksuriya, O.; Okonogi, S.; Schiffelers, R.M.; Hennink, W.E. Curcumin nanoformulations: A review of pharmaceutical properties and preclinical studies and clinical data related to cancer treatment. Biomaterials 2014, 35, 3365–3383. [Google Scholar] [CrossRef] [PubMed]
  8. Mishra, S.; Vinayak, M. Ellagic Acid Induces Novel and Atypical PKC Isoforms and Promotes Caspase-3 Dependent Apoptosis by Blocking Energy Metabolism. Nutr. Cancer 2014, 66, 675–681. [Google Scholar] [CrossRef] [PubMed]
  9. Zou, W.-W.; Xu, S.-P. Galangin inhibits the cell progression and induces cell apoptosis through activating PTEN and Caspase-3 pathways in retinoblastoma. Biomed. Pharmacother. 2018, 97, 851–863. [Google Scholar] [CrossRef] [PubMed]
  10. Punvittayagul, C.; Chariyakornkul, A.; Jarukamjorn, K.; Wongpoomchai, R. Protective Role of Vanillic Acid against Diethylnitrosamine- and 1,2-Dimethylhydrazine-Induced Hepatocarcinogenesis in Rats. Molecules 2021, 26, 2718. [Google Scholar] [CrossRef]
  11. Debnath, S.; Sarkar, A.; Mukherjee, D.D.; Ray, S.; Mahata, B.; Mahata, T.; Parida, P.K.; Das, T.; Mukhopadhyay, R.; Ghosh, Z.; et al. Eriodictyol mediated selective targeting of the TNFR1/FADD/TRADD axis in cancer cells induce apoptosis and inhibit tumor progression and metastasis. Transl. Oncol. 2022, 21, 101433. [Google Scholar] [CrossRef]
  12. Yao, Y.; Rao, C.; Zheng, G.; Wang, S. Luteolin suppresses colorectal cancer cell metastasis via regulation of the miR-384/pleiotrophin axis. Oncol. Rep. 2019, 42, 131–141. [Google Scholar] [CrossRef]
  13. Wang, R.; Zhu, X.; Wang, Q.; Li, X.; Wang, E.; Zhao, Q.; Wang, Q.; Cao, H. The anti-tumor effect of taxifolin on lung cancer via suppressing stemness and epithelial-mesenchymal transition in vitro and oncogenesis in nude mice. Ann. Transl. Med. 2020, 8, 590. [Google Scholar] [CrossRef] [PubMed]
  14. Chiang, E.-P.I.; Tsai, S.-Y.; Kuo, Y.-H.; Pai, M.-H.; Chiu, H.-L.; Rodriguez, R.L.; Tang, F.-Y. Caffeic Acid Derivatives Inhibit the Growth of Colon Cancer: Involvement of the PI3-K/Akt and AMPK Signaling Pathways. PLoS ONE 2014, 9, e99631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Vaidya, F.U.; Chhipa, A.S.; Sagar, N.; Pathak, C. Oxidative Stress and Inflammation Can Fuel Cancer; Springer: Singapore, 2020; pp. 229–258. [Google Scholar]
  16. Bodduluru, L.N.; Kasala, E.R.; Madhana, R.M.; Barua, C.C.; Hussain, M.I.; Haloi, P.; Borah, P. Naringenin ameliorates inflammation and cell proliferation in benzo(a)pyrene induced pulmonary carcinogenesis by modulating CYP1A1, NFκB and PCNA expression. Int. Immunopharmacol. 2016, 30, 102–110. [Google Scholar] [CrossRef] [PubMed]
  17. Manigandan, K.; Manimaran, D.; Jayaraj, R.L.; Elangovan, N.; Dhivya, V.; Kaphle, A. Taxifolin curbs NF-κB-mediated Wnt/β-catenin signaling via up-regulating Nrf2 pathway in experimental colon carcinogenesis. Biochimie 2015, 119, 103–112. [Google Scholar] [CrossRef]
  18. Zeng, A.; Liang, X.; Zhu, S.; Liu, C.; Wang, S.; Zhang, Q.; Zhao, J.; Song, L. Chlorogenic acid induces apoptosis, inhibits metastasis and improves antitumor immunity in breast cancer via the NF-κB signaling pathway. Oncol. Rep. 2020, 45, 717–727. [Google Scholar] [CrossRef]
  19. Ceci, C.; Tentori, L.; Atzori, M.; Lacal, P.; Bonanno, E.; Scimeca, M.; Cicconi, R.; Mattei, M.; De Martino, M.; Vespasiani, G.; et al. Ellagic Acid Inhibits Bladder Cancer Invasiveness and In Vivo Tumor Growth. Nutrients 2016, 8, 744. [Google Scholar] [CrossRef] [Green Version]
  20. Pandurangan, A.K.; Sadagopan, S.K.A.; Dharmalingam, P.; Ganapasam, S. Luteolin, a bioflavonoid inhibits azoxymethane-induced colon carcinogenesis: Involvement of iNOS and COX-2. Pharmacogn. Mag. 2014, 10, 306–310. [Google Scholar] [CrossRef] [Green Version]
  21. Lin, R.; Piao, M.; Song, Y.; Liu, C. Quercetin Suppresses AOM/DSS-Induced Colon Carcinogenesis through Its Anti-Inflammation Effects in Mice. J. Immunol. Res. 2020, 2020, 9242601. [Google Scholar] [CrossRef]
  22. Shree, A.; Islam, J.; Sultana, S. Quercetin ameliorates reactive oxygen species generation, inflammation, mucus depletion, goblet disintegration, and tumor multiplicity in colon cancer: Probable role of adenomatous polyposis coli, β-catenin. Phytother. Res. 2021, 35, 2171–2184. [Google Scholar] [CrossRef]
  23. Feng, X.-L.; Zhan, X.-X.; Zuo, L.-S.-Y.; Mo, X.-F.; Zhang, X.; Liu, K.-Y.; Li, L.; Zhang, C.-X. Associations between serum concentration of flavonoids and breast cancer risk among Chinese women. Eur. J. Nutr. 2021, 60, 1347–1362. [Google Scholar] [CrossRef] [PubMed]
  24. Mérida-Ortega, Á.; Hernández-Alcaraz, C.; Hernández-Ramírez, R.U.; García-Martínez, A.; Trejo-Valdivia, B.; Salinas-Rodríguez, A.; Svensson, K.; Cebrián, M.E.; Franco-Marina, F.; López-Carrillo, L. Phthalate exposure, flavonoid consumption and breast cancer risk among Mexican women. Environ. Int. 2016, 96, 167–172. [Google Scholar] [CrossRef] [PubMed]
  25. Russo, G.; Campisi, D.; Di Mauro, M.; Regis, F.; Reale, G.; Marranzano, M.; Ragusa, R.; Solinas, T.; Madonia, M.; Cimino, S.; et al. Dietary Consumption of Phenolic Acids and Prostate Cancer: A Case-Control Study in Sicily, Southern Italy. Molecules 2017, 22, 2159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Zamora-Ros, R.; Sacerdote, C.; Ricceri, F.; Weiderpass, E.; Roswall, N.; Buckland, G.; St-Jules, D.E.; Overvad, K.; Kyrø, C.; Fagherazzi, G.; et al. Flavonoid and lignan intake in relation to bladder cancer risk in the European Prospective Investigation into Cancer and Nutrition (EPIC) study. Br. J. Cancer 2014, 111, 1870–1880. [Google Scholar] [CrossRef] [Green Version]
  27. Zamora-Ros, R.; Cayssials, V.; Franceschi, S.; Kyrø, C.; Weiderpass, E.; Hennings, J.; Sandström, M.; Tjønneland, A.; Olsen, A.; Overvad, K.; et al. Polyphenol intake and differentiated thyroid cancer risk in the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort. Int. J. Cancer 2020, 146, 1841–1850. [Google Scholar] [CrossRef]
  28. Nimptsch, K.; Zhang, X.H.; Cassidy, A.; Song, M.Y.; O’Reilly, E.J.; Lin, J.H.; Pischon, T.; Rimm, E.B.; Willett, W.C.; Fuchs, C.S.; et al. Habitual intake of flavonoid subclasses and risk of colorectal cancer in 2 large prospective cohorts. Am. J. Clin. Nutr. 2016, 103, 184–191. [Google Scholar] [CrossRef] [Green Version]
  29. Zamora-Ros, R.; Barupal, D.K.; Rothwell, J.A.; Jenab, M.; Fedirko, V.; Romieu, I.; Aleksandrova, K.; Overvad, K.; Kyrø, C.; Tjønneland, A.; et al. Dietary flavonoid intake and colorectal cancer risk in the European prospective investigation into cancer and nutrition (EPIC) cohort. Int. J. Cancer 2017, 140, 1836–1844. [Google Scholar] [CrossRef] [Green Version]
  30. Londoño, C.; Cayssials, V.; De Villasante, I.; Crous-Bou, M.; Scalbert, A.; Weiderpass, E.; Agudo, A.; Tjønneland, A.; Olsen, A.; Overvad, K.; et al. Polyphenol Intake and Epithelial Ovarian Cancer Risk in the European Prospective Investigation into Cancer and Nutrition (EPIC) Study. Antioxidants 2021, 10, 1249. [Google Scholar] [CrossRef]
  31. Molina-Montes, E.; Sánchez, M.J.; Zamora-Ros, R.; Bueno-De-Mesquita, H.B.; Wark, P.A.; Obon-Santacana, M.; Kühn, T.; Katzke, V.; Travis, R.C.; Ye, W.; et al. Flavonoid and lignan intake and pancreatic cancer risk in the European prospective investigation into cancer and nutrition cohort. Int. J. Cancer 2016, 139, 1480–1492. [Google Scholar] [CrossRef]
  32. Bahrami, A.; Jafari, S.; Rafiei, P.; Beigrezaei, S.; Sadeghi, A.; Hekmatdoost, A.; Rashidkhani, B.; Hejazi, E. Dietary intake of polyphenols and risk of colorectal cancer and adenoma–A case-control study from Iran. Complement. Ther. Med. 2019, 45, 269–274. [Google Scholar] [CrossRef]
  33. Zhou, Y.; Gu, K.; Zhou, F. Dietary Flavonoid Intake and Cancer Mortality: A Population-Based Cohort Study. Nutrients 2023, 15, 976. [Google Scholar] [CrossRef] [PubMed]
  34. Basak, S.K.; Bera, A.; Yoon, A.J.; Morselli, M.; Jeong, C.; Tosevska, A.; Dong, T.S.; Eklund, M.; Russ, E.; Nasser, H.; et al. A randomized, phase 1, placebo-controlled trial of APG-157 in oral cancer demonstrates systemic absorption and an inhibitory effect on cytokines and tumor-associated microbes. Cancer 2020, 126, 1668–1682. [Google Scholar] [CrossRef] [PubMed]
  35. Cruz-Correa, M.; Hylind, L.M.; Marrero, J.H.; Zahurak, M.L.; Murray-Stewart, T.; Casero, R.A.; Montgomery, E.A.; Iacobuzio-Donahue, C.; Brosens, L.A.; Offerhaus, G.J.; et al. Efficacy and Safety of Curcumin in Treatment of Intestinal Adenomas in Patients With Familial Adenomatous Polyposis. Gastroenterology 2018, 155, 668–673. [Google Scholar] [CrossRef]
  36. Zhang, Z.; Garzotto, M.; Beer, T.M.; Thuillier, P.; Lieberman, S.; Mori, M.; Stoller, W.A.; Farris, P.E.; Shannon, J. Effects of ω-3 Fatty Acids and Catechins on Fatty Acid Synthase in the Prostate: A Randomized Controlled Trial. Nutr. Cancer 2016, 68, 1309–1319. [Google Scholar] [CrossRef] [Green Version]
  37. Sinicrope, F.A.; Viggiano, T.R.; Buttar, N.S.; Song, L.M.W.K.; Schroeder, K.W.; Kraichely, R.E.; Larson, M.V.; Sedlack, R.E.; Kisiel, J.B.; Gostout, C.J.; et al. Randomized Phase II Trial of Polyphenon E versus Placebo in Patients at High Risk of Recurrent Colonic Neoplasia. Cancer Prev. Res. 2021, 14, 573–580. [Google Scholar] [CrossRef] [PubMed]
  38. Student, V.; Vidlar, A.; Bouchal, J.; Vrbkova, J.; Kolar, Z.; Kral, M.; Kosina, P.; Vostalova, J. Cranberry intervention in patients with prostate cancer prior to radical prostatectomy. Clinical, pathological and laboratory findings. Biomed. Pap. 2016, 160, 559–565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Citronberg, J.; Bostick, R.; Ahearn, T.; Turgeon, D.K.; Ruffin, M.T.; Djuric, Z.; Sen, A.; Brenner, D.E.; Zick, S.M. Effects of Ginger Supplementation on Cell-Cycle Biomarkers in the Normal-Appearing Colonic Mucosa of Patients at Increased Risk for Colorectal Cancer: Results from a Pilot, Randomized, and Controlled Trial. Cancer Prev. Res. 2013, 6, 271–281. [Google Scholar] [CrossRef] [Green Version]
  40. Messing, E.; Gee, J.R.; Saltzstein, D.R.; Kim, K.; Disant’Agnese, A.; Kolesar, J.; Harris, L.; Faerber, A.; Havighurst, T.; Young, J.M.; et al. A Phase 2 Cancer Chemoprevention Biomarker Trial of Isoflavone G-2535 (Genistein) in Presurgical Bladder Cancer Patients. Cancer Prev. Res. 2012, 5, 621–630. [Google Scholar] [CrossRef] [Green Version]
  41. Pilvenyte, G.; Ratautaite, V.; Boguzaite, R.; Ramanavicius, A.; Viter, R.; Ramanavicius, S. Molecularly Imprinted Polymers for the Determination of Cancer Biomarkers. Int. J. Mol. Sci. 2023, 24, 4105. [Google Scholar] [CrossRef]
  42. Yin, Z.; Zheng, T.; Ho, C.-T.; Huang, Q.; Wu, Q.; Zhang, M. Improving the stability and bioavailability of tea polyphenols by encapsulations: A review. Food Sci. Hum. Wellness 2022, 11, 537–556. [Google Scholar] [CrossRef]
  43. Garcia-Carrasco, M.; Picos-Corrales, L.A.; Gutiérrez-Grijalva, E.P.; Angulo-Escalante, M.A.; Licea-Claverie, A.; Heredia, J.B. Loading and Release of Phenolic Compounds Present in Mexican Oregano (Lippia graveolens) in Different Chitosan Bio-Polymeric Cationic Matrixes. Polymers 2022, 14, 3609. [Google Scholar] [CrossRef] [PubMed]
  44. Pimentel-Moral, S.; Teixeira, M.C.; Fernandes, A.R.; Arráez-Román, D.; Martínez-Férez, A.; Segura-Carretero, A.; Souto, E.B. Lipid nanocarriers for the loading of polyphenols—A comprehensive review. Adv. Colloid Interface Sci. 2018, 260, 85–94. [Google Scholar] [CrossRef] [PubMed]
  45. Manzanares-Guevara, L.A.; Licea-Claverie, A.; Oroz-Parra, I.; Bernaldez-Sarabia, J.; Diaz-Castillo, F.; Licea-Navarro, A.F. Smart Nanoformulation Based on Stimuli-Responsive Nanogels and Curcumin: Promising Therapy against Colon Cancer. ACS Omega 2020, 5, 9171–9184. [Google Scholar] [CrossRef] [Green Version]
  46. Hu, B.; Liu, X.; Zhang, C.; Zeng, X. Food macromolecule based nanodelivery systems for enhancing the bioavailability of polyphenols. J. Food Drug Anal. 2017, 25, 3–15. [Google Scholar] [CrossRef]
  47. Tang, H.-Y.; Fang, Z.; Ng, K. Dietary fiber-based colon-targeted delivery systems for polyphenols. Trends Food Sci. Technol. 2020, 100, 333–348. [Google Scholar] [CrossRef]
  48. Song, J.; Feng, H.; Wu, M.; Chen, L.; Xia, W.; Zhang, W. Development of a bioactive chitosan HPMC-based membrane with tea polyphenols encapsulated in β-cyclodextrin as an effective enhancement. Mater. Today Commun. 2021, 27, 102324. [Google Scholar] [CrossRef]
  49. Zhang, X.; Wang, C.; Qi, Z.; Zhao, R.; Wang, C.; Zhang, T. Pea protein based nanocarriers for lipophilic polyphenols: Spectroscopic analysis, characterization, chemical stability, antioxidant and molecular docking. Food Res. Int. 2022, 160, 111713. [Google Scholar] [CrossRef]
  50. Cheng, H.; Fang, Z.; Wusigale; Bakry, A.M.; Chen, Y.; Liang, L. Complexation of trans- and cis-resveratrol with bovine serum albumin, β-lactoglobulin or α-lactalbumin. Food Hydrocoll. 2018, 81, 242–252. [Google Scholar] [CrossRef]
  51. Costa, M.; Sezgin-Bayindir, Z.; Losada-Barreiro, S.; Paiva-Martins, F.; Saso, L.; Bravo-Díaz, C. Polyphenols as Antioxidants for Extending Food Shelf-Life and in the Prevention of Health Diseases: Encapsulation and Interfacial Phenomena. Biomedicines 2021, 9, 1909. [Google Scholar] [CrossRef]
  52. López de Dicastillo, C.; Piña, C.; Garrido, L.; Arancibia, C.; Galotto, M.J. Enhancing Thermal Stability and Bioaccesibility of Açaí Fruit Polyphenols through Electrohydrodynamic Encapsulation into Zein Electrosprayed Particles. Antioxidants 2019, 8, 464. [Google Scholar] [CrossRef] [Green Version]
  53. Dai, Q.; Geng, H.; Yu, Q.; Hao, J.; Cui, J. Polyphenol-Based Particles for Theranostics. Theranostics 2019, 9, 3170–3190. [Google Scholar] [CrossRef] [PubMed]
  54. Brito, J.; Hlushko, H.; Abbott, A.; Aliakseyeu, A.; Hlushko, R.; Sukhishvili, S.A. Integrating Antioxidant Functionality into Polymer Materials: Fundamentals, Strategies, and Applications. ACS Appl. Mater. Interfaces 2021, 13, 41372–41395. [Google Scholar] [CrossRef] [PubMed]
  55. Rahaiee, S.; Assadpour, E.; Faridi Esfanjani, A.; Silva, A.S.; Jafari, S.M. Application of nano/microencapsulated phenolic compounds against cancer. Adv. Colloid Interface Sci. 2020, 279, 102153. [Google Scholar] [CrossRef] [PubMed]
  56. Piazzini, V.; Vasarri, M.; Degl’Innocenti, D.; Guastini, A.; Barletta, E.; Salvatici, M.C.; Bergonzi, M.C. Comparison of Chitosan Nanoparticles and Soluplus Micelles to Optimize the Bioactivity of Posidonia oceanica Extract on Human Neuroblastoma Cell Migration. Pharmaceutics 2019, 11, 655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Mostafa, M.M.; Amin, M.M.; Zakaria, M.Y.; Hussein, M.A.; Shamaa, M.M.; Abd El-Halim, S.M. Chitosan Surface-Modified PLGA Nanoparticles Loaded with Cranberry Powder Extract as a Potential Oral Delivery Platform for Targeting Colon Cancer Cells. Pharmaceutics 2023, 15, 606. [Google Scholar] [CrossRef]
  58. Choi, S.M.; Pham, V.C.; Lee, S.; Kim, J.A. Metabolism of Diterpenoids Derived from the Bark of Cinnamomum cassia in Human Liver Microsomes. Pharmaceutics 2021, 13, 1316. [Google Scholar] [CrossRef]
  59. Atallah, M.A.; Sallam, M.A.; Abdelmoneem, M.A.; Teleb, M.; Elkhodairy, K.A.; Bekhit, A.A.; Khafaga, A.F.; Noreldin, A.E.; Elzoghby, A.O.; Khattab, S.N. Green self-assembled lactoferrin carboxymethyl cellulose nanogels for synergistic chemo/herbal breast cancer therapy. Colloids Surf. B Biointerfaces 2022, 217, 112657. [Google Scholar] [CrossRef]
  60. Levit, S.L.; Yang, H.; Tang, C. Rapid Self-Assembly of Polymer Nanoparticles for Synergistic Codelivery of Paclitaxel and Lapatinib via Flash NanoPrecipitation. Nanomaterials 2020, 10, 561. [Google Scholar] [CrossRef] [Green Version]
  61. Hong, S.; Dia, V.P.; Baek, S.J.; Zhong, Q. Nanoencapsulation of apigenin with whey protein isolate: Physicochemical properties, in vitro activity against colorectal cancer cells, and bioavailability. LWT 2022, 154, 112751. [Google Scholar] [CrossRef]
  62. Suárez-García, S.; Esposito, T.V.F.; Neufeld-Peters, J.; Bergamo, M.; Yang, H.; Saatchi, K.; Schaffer, P.; Häfeli, U.O.; Ruiz-Molina, D.; Rodríguez-Rodríguez, C.; et al. Hybrid Metal–Phenol Nanoparticles with Polydopamine-like Coating for PET/SPECT/CT Imaging. ACS Appl. Mater. Interfaces 2021, 13, 10705–10718. [Google Scholar] [CrossRef] [PubMed]
  63. Aljabali, A.A.A.; Bakshi, H.A.; Hakkim, F.L.; Haggag, Y.A.; Al-Batanyeh, K.M.; Al Zoubi, M.S.; Al-Trad, B.; Nasef, M.M.; Satija, S.; Mehta, M.; et al. Albumin Nano-Encapsulation of Piceatannol Enhances Its Anticancer Potential in Colon Cancer Via Downregulation of Nuclear p65 and HIF-1α. Cancers 2020, 12, 113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Ghayour, N.; Hosseini, S.M.H.; Eskandari, M.H.; Esteghlal, S.; Nekoei, A.-R.; Hashemi Gahruie, H.; Tatar, M.; Naghibalhossaini, F. Nanoencapsulation of quercetin and curcumin in casein-based delivery systems. Food Hydrocoll. 2019, 87, 394–403. [Google Scholar] [CrossRef]
  65. Peñalva, R.; Morales, J.; González-Navarro, C.J.; Larrañeta, E.; Quincoces, G.; Peñuelas, I.; Irache, J.M. Increased Oral Bioavailability of Resveratrol by Its Encapsulation in Casein Nanoparticles. Int. J. Mol. Sci. 2018, 19, 2816. [Google Scholar] [CrossRef] [Green Version]
  66. Mendez-Pfeiffer, P.; Juarez, J.; Hernandez, J.; Taboada, P.; Virués, C.; Alday, E.; Valencia, D.; Velazquez, C. Polymeric nanoparticles for the delivery of Sonoran desert propolis: Synthesis, characterization and antiproliferative activity on cancer cells. Colloids Surf. B: Biointerfaces 2022, 215, 112475. [Google Scholar] [CrossRef] [PubMed]
  67. Ahmed, R.; Tariq, M.; Ahmad, I.S.; Fouly, H.; Fakhari, A.; Hasan, A.; Kushad, M. Poly(lactic-co<-glycolic acid) Nanoparticles Loaded with Callistemon citrinus Phenolics Exhibited Anticancer Properties against Three Breast Cancer Cell Lines. J. Food Qual. 2019, 2019, 2638481. [Google Scholar] [CrossRef] [Green Version]
  68. Sankaran, V.; Murali, S.K.; Ghidan, A.; Antary, T.A.; David, E. Oral cancer preventive potential of Polydatin: A nanoencapsulation approach. J. Phytol. 2020, 12, 109–116. [Google Scholar] [CrossRef]
  69. Han, Y.; Dong, Z.; Wang, C.; Li, Q.; Hao, Y.; Yang, Z.; Zhu, W.; Zhang, Y.; Liu, Z.; Feng, L. Ferrous ions doped calcium carbonate nanoparticles potentiate chemotherapy by inducing ferroptosis. J. Control. Release 2022, 348, 346–356. [Google Scholar] [CrossRef]
  70. Ma, Z.; Gao, X.; Raza, F.; Zafar, H.; Huang, G.; Yang, Y.; Shi, F.; Wang, D.; He, X. Design of GSH-Responsive Curcumin Nanomicelles for Oesophageal Cancer Therapy. Pharmaceutics 2022, 14, 1802. [Google Scholar] [CrossRef]
  71. Yadav, K.; Bajaj, R.K.; Mandal, S.; Mann, B. Encapsulation of grape seed extract phenolics using whey protein concentrate, maltodextrin and gum arabica blends. J. Food Sci. Technol. 2020, 57, 426–434. [Google Scholar] [CrossRef]
  72. Parashar, P.; Rathor, M.; Dwivedi, M.; Saraf, S.A. Hyaluronic Acid Decorated Naringenin Nanoparticles: Appraisal of Chemopreventive and Curative Potential for Lung Cancer. Pharmaceutics 2018, 10, 33. [Google Scholar] [CrossRef] [Green Version]
  73. Pudziuvelyte, L.; Marksa, M.; Sosnowska, K.; Winnicka, K.; Morkuniene, R.; Bernatoniene, J. Freeze-Drying Technique for Microencapsulation of Elsholtzia ciliata Ethanolic Extract Using Different Coating Materials. Molecules 2020, 25, 2237. [Google Scholar] [CrossRef]
  74. Hussain, S.A.; Hameed, A.; Nazir, Y.; Naz, T.; Wu, Y.; Suleria, H.A.; Song, Y. Microencapsulation and the Characterization of Polyherbal Formulation (PHF) Rich in Natural Polyphenolic Compounds. Nutrients 2018, 10, 843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Ralaivao, M.; Lucas, J.; Rocha, F.; Estevinho, B.N. Food-Grade Microencapsulation Systems to Improve Protection of the Epigallocatechin Gallate. Foods 2022, 11, 1990. [Google Scholar] [CrossRef] [PubMed]
  76. Bannikova, A.; Zyainitdinov, D.; Evteev, A.; Drevko, Y.; Evdokimov, I. Microencapsulation of polyphenols and xylooligosaccharides from oat bran in whey protein-maltodextrin complex coacervates: In-vitro evaluation and controlled release. Bioact. Carbohydr. Diet. Fibre 2020, 23, 100236. [Google Scholar] [CrossRef]
  77. Lee, Y.-K.; Chang, Y.H. Microencapsulation of a maca leaf polyphenol extract in mixture of maltodextrin and neutral polysaccharides extracted from maca roots. Int. J. Biol. Macromol. 2020, 150, 546–558. [Google Scholar] [CrossRef]
  78. Fathi, F.; Ebrahimi, S.N.; Pereira, D.M.; Estevinho, B.N.; Rocha, F. Preliminary studies of microencapsulation and anticancer activity of polyphenols extract from Punica granatum peels. Can. J. Chem. Eng. 2022, 100, 3240–3252. [Google Scholar] [CrossRef]
  79. Farrag, A.; El-Messery, T.M.; El-Said, M.M.; Soliman, T.N.; El-Din, H.M.F. Research Article Microencapsulation of Grape Phenolic Compounds Using Whey Proteins as a Carrier Vehicle. J. Biol. Sci. 2018, 18, 373–380. [Google Scholar] [CrossRef] [Green Version]
  80. Benito, I.; Encío, I.J.; Milagro, F.I.; Alfaro, M.; Martínez-Peñuela, A.; Barajas, M.; Marzo, F. Microencapsulated Bifidobacterium bifidum and Lactobacillus gasseri in Combination with Quercetin Inhibit Colorectal Cancer Development in ApcMin/+ Mice. Int. J. Mol. Sci. 2021, 22, 4906. [Google Scholar] [CrossRef] [PubMed]
  81. Tavakoli, F.; Jahanban-Esfahlan, R.; Seidi, K.; Jabbari, M.; Behzadi, R.; Pilehvar-Soltanahmadi, Y.; Zarghami, N. Effects of nano-encapsulated curcumin-chrysin on telomerase, MMPs and TIMPs gene expression in mouse B16F10 melanoma tumour model. Artif. Cells Nanomed. Biotechnol. 2018, 46, 75–86. [Google Scholar] [CrossRef] [Green Version]
  82. Krishnan, G.; Subramaniyan, J.; Chengalvarayan Subramani, P.; Muralidharan, B.; Thiruvengadam, D. Hesperetin conjugated PEGylated gold nanoparticles exploring the potential role in anti-inflammation and anti-proliferation during diethylnitrosamine-induced hepatocarcinogenesis in rats. Asian J. Pharm. Sci. 2017, 12, 442–455. [Google Scholar] [CrossRef]
  83. Khan, I.; Sarkar, B.; Joshi, G.; Nakhate, K.T.; Ajazuddin; Mantha, A.K.; Kumar, R.; Kaul, A.; Chaturvedi, S.; Mishra, A.K.; et al. Biodegradable nanoparticulate co-delivery of flavonoid and doxorubicin: Mechanistic exploration and evaluation of anticancer effect in vitro and in vivo. Biomater. Biosyst. 2021, 3, 100022. [Google Scholar] [CrossRef]
  84. Das, S.; Das, J.; Samadder, A.; Paul, A.; Khuda-Bukhsh, A.R. Efficacy of PLGA-loaded apigenin nanoparticles in Benzo[a]pyrene and ultraviolet-B induced skin cancer of mice: Mitochondria mediated apoptotic signalling cascades. Food Chem. Toxicol. 2013, 62, 670–680. [Google Scholar] [CrossRef] [PubMed]
  85. Siddiqui, I.A.; Bharali, D.J.; Nihal, M.; Adhami, V.M.; Khan, N.; Chamcheu, J.C.; Khan, M.I.; Shabana, S.; Mousa, S.A.; Mukhtar, H. Excellent anti-proliferative and pro-apoptotic effects of (-)-epigallocatechin-3-gallate encapsulated in chitosan nanoparticles on human melanoma cell growth both in vitro and in vivo. Nanomed. Nanotechnol. Biol. Med. 2014, 10, 1619–1626. [Google Scholar] [CrossRef] [PubMed]
  86. Arora, R.; Samim, M. Evaluation of anti-inflammatory and anti-cancer activity of calcium phosphate encapsulated Resveratrol in mouse skin cancer model. Biomed. Pharmacol. J. 2021, 14, 113–122. [Google Scholar] [CrossRef]
  87. Baksi, R.; Singh, D.P.; Borse, S.P.; Rana, R.; Sharma, V.; Nivsarkar, M. In vitro and in vivo anticancer efficacy potential of Quercetin loaded polymeric nanoparticles. Biomed. Pharmacother. 2018, 106, 1513–1526. [Google Scholar] [CrossRef] [PubMed]
  88. Eskandari, Z.; Bahadori, F.; Yenigun, V.B.; Demiray, M.; Eroğlu, M.S.; Kocyigit, A.; Oner, E.T. Levan enhanced the NF-κB suppression activity of an oral nano PLGA-curcumin formulation in breast cancer treatment. Int. J. Biol. Macromol. 2021, 189, 223–231. [Google Scholar] [CrossRef]
  89. Song, Y.Y.; Yuan, Y.; Shi, X.; Che, Y.Y. Improved drug delivery and anti-tumor efficacy of combinatorial liposomal formulation of genistein and plumbagin by targeting Glut1 and Akt3 proteins in mice bearing prostate tumor. Colloids Surf. B Biointerfaces 2020, 190, 110966. [Google Scholar] [CrossRef] [PubMed]
  90. Huo, M.; Wang, H.; Zhang, Y.; Cai, H.; Zhang, P.; Li, L.; Zhou, J.; Yin, T. Co-delivery of silybin and paclitaxel by dextran-based nanoparticles for effective anti-tumor treatment through chemotherapy sensitization and microenvironment modulation. J. Control. Release 2020, 321, 198–210. [Google Scholar] [CrossRef] [PubMed]
  91. Chen, S.; Fan, J.X.; Zheng, D.W.; Liu, F.; Zeng, X.; Yan, G.P.; Zhang, X.Z. A multi-functional drug delivery system based on polyphenols for efficient tumor inhibition and metastasis prevention. Biomater. Sci. 2020, 8, 702–711. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structures of known polyphenols: (a) anthocyanins (cyanidin), (b) dihydrochalcones (phloretin), (c) flavanols (catechin), (d) flavanones (naringenin), (e) flavones (apigenin), (f) flavonols (quercetin).
Figure 1. Chemical structures of known polyphenols: (a) anthocyanins (cyanidin), (b) dihydrochalcones (phloretin), (c) flavanols (catechin), (d) flavanones (naringenin), (e) flavones (apigenin), (f) flavonols (quercetin).
Macromol 03 00030 g001
Figure 2. Estimated number of prevalent cases worldwide (5 years) in 2020 for both sexes of all ages. Graphic was made by the authors from data of the International Agency for Cancer Research database [5].
Figure 2. Estimated number of prevalent cases worldwide (5 years) in 2020 for both sexes of all ages. Graphic was made by the authors from data of the International Agency for Cancer Research database [5].
Macromol 03 00030 g002
Figure 3. Types of interactions of polyphenols with different polymers.
Figure 3. Types of interactions of polyphenols with different polymers.
Macromol 03 00030 g003
Table 2. Studies of the association between polyphenols intake and cancer risk.
Table 2. Studies of the association between polyphenols intake and cancer risk.
Compound(s)Cancer TypePopulationSubjects (Age)EffectReference
Flavonols and lignansBladder477,312 European subjects35–70 yearsInverse association between flavonols and lignans intake and bladder cancer risk[26]
Flavonols, isorhamnetin, kaempferol, flavanones and naringeninBreast877 Chinese women with breast cancer,
792 control subjects
25–70 yearsThe concentration of the flavonoids in the serum was associated with a lower breast cancer risk[23]
Anthocyanidins and flavan-3-olsBreast233 Mexican women with breast cancer,
221 control subjects
>18 (mean 53) yearsHigher intake of flavonoids reduced the risk of breast cancer, synergistically working with butyl benzyl phthalate[24]
Flavonols, flavones, flavanones,
flavan-3-ols, and anthocyanins
Colorectal51,528 US male health professionals and
121,701 US female nurses
Men: 40–75 years
Women: 30–55 years
No decrease in colorectal cancer was detected[28]
FlavonoidsColorectal521,448 European subjects (with exceptions)35–70 yearsNo association between flavonoids intake and colorectal cancer was found[29]
Phenolic acids, hydroxycinnamic acids, flavonols, and stilbenesColorectal and colorectal adenoma129 Iranian subjects with colorectal cancer, 130 with colorectal adenoma, and 240 controls30–79 yearsHigher intake of phenolic acids, hydroxycinnamic acids, and flavonols was associated with a decrease in colorectal cancer risk.
Higher intake of stilbenes was associated with a lower colorectal adenoma risk.
[32]
PolyphenolsEpithelial ovarian cancer309,129 European women35–70 yearsNo association between polyphenols intake and endothelial ovarian cancer was found[30]
Naringenin, peonidin, and catechinGeneral14,029 US subjects>18Inverse association between flavonoids intake and cancer mortality[33]
Flavonoids and lignansPancreatic477,309 European subjects25–70
years
No association between flavonoids and lignans intake and pancreatic cancer was found[31]
Caffeic acid and ferulic acidProstate118 Italian prostate cancer subjects,
22 controls
Mean age: 69.13 yearsHigh intake of phenolic acids may be associated with a decrease in prostate cancer risk[25]
Polyphenols and phenolic acidsThyroid476,108 European subjects35–70 yearsInverse association between polyphenols and phenolic acids intake and thyroid cancer risk in patients with BMI ≥ 25[27]
Table 3. Clinical studies of polyphenols against different types of cancer.
Table 3. Clinical studies of polyphenols against different types of cancer.
CancerCompoundDoseSubjectsEffectReference
BladderGenistein300 or 600 mg59 subjects with urothelial bladder cancerInhibition of bladder cancer growth by inhibiting the phosphorylation of the epidermal growth factor receptor[40]
Colorectal Ginger (Zingiber officinale) extract with 5% of gingerols2 g 21 healthy subjects with a high risk of colorectal cancerDecrease in the proliferation of crypts[39]
Epigallocatechin gallate780 mg32 subjects with rectal aberrant crypt fociNo difference in the number of the rectal aberrant crypt foci[37]
Familial adenomatous polyposisCurcumin3000 mg44 subjects with familial adenomatous polyposisNo difference in the mean number or size of polyps[35]
Oral Curcuma longa phenolic extract100 or 200 mg12 oral cancer patients
13 normal subjects
Decrease in IL-1β, IL-6, and IL-8 content in the saliva.
Increased gene expression related to differentiation and T cell recruitment to the tumor microenvironment.
[34]
ProstateCranberry fruit powder1500 mg62 subjects with prostate cancerDecrease in serum prostate-specific antigen [38]
Epigallocatechin gallate600 mg43 subjects with a prior negative biopsy, but suspicious No difference in fatty acid synthase or antigen Ki-76[36]
Table 4. Comparison of the advantages and disadvantages of encapsulated and non-encapsulated phenolic compounds.
Table 4. Comparison of the advantages and disadvantages of encapsulated and non-encapsulated phenolic compounds.
EncapsulatedNon-Encapsulated
  • Increased solubility in aqueous medium
  • Higher content of bioavailable phenolic compounds after the gastrointestinal process
  • Increased activity of encapsulated compounds after the gastrointestinal process
  • Low content of compounds within the capsule, depending on the size and chemical characteristics of the particle and the compounds.
Ionization and/or loss of phenolic compounds throughout the gastrointestinal process
Low solubility in aqueous medium, high solubility in organic media frequently unfit for human consumption.
Decreased activity of polyphenolic compounds after the gastrointestinal process
Table 5. Summary of polymers used for the encapsulation of phenolic compounds on the nanometric scale.
Table 5. Summary of polymers used for the encapsulation of phenolic compounds on the nanometric scale.
Wall MaterialsInteractionTechnique of EncapsulationPhenolic CompoundApplicationReference
Chitosan–PEGMA ChitosanHydrogen bondDesolvation method
Nanoemulsion
Lippia graveolens
(ethanolic extract)
Posidonia oceanica extract
-
Inhibitory activity against neuroblastoma cell migration.
[43,56]
Chitosan–poly (d,l-lactide-co-glycolide)Hydrogen bondDouble emulsion solvent evaporationCranberry powder extractsCytotoxicity of colon cancer cells (HT-29)[57]
β-CyclodextrinHydrogen bondCoacervation
Desolvation method
Curcumin
Resveratrol and Oxyresveratrol
Transdermal delivery (melanoma treatment)
Antiproliferative effect in prostate cancer cells
[58]
Carboxymethyl cellulose–lactoferrinHydrogen bondDesolvation methodHidroxypropyl-beta-cyclodetrin–Polyphenol honokiolInhibition of tumor growth in in vivo studies (EAT)[59]
α-tocopherol and polystyrene block–polyethylene glycolCovalent and chelation Flash nanoprecipitationTannic acid, PaclitaxelAgainst OVCA-432 ovarian cancer cells[60,61]
In(III) and Cu(III)Covalent Catechol 3,4-dihydroxycinnamicContrast Agent[62]
AlbuminHydrogen bondDesolvation methodPiceatannolDown-regulation of p65 and HIF-1 (proteins associated with different types of cancer)[63]
CaseinHydrogen bondDesolvation method
Coacervation (Spray drying)
Quercetin and Curcumin
Resveratrol
Against MCF-7 breast cancer cells
Increased bioavailability of resveratrol in plasma
[64,65]
Whey proteinHydrogen bondpH-cycling treatmentApigeninAgainst colorectal cancer cells HTC-116 and HT-29[61]
Poly-(lactide-co-glycolide) acid (PLGA)Hydrogen bondNanoprecipitation
Emulsion (O/W)
Sonoran desert propolis
Callistemon citrinus Phenolics and berberine
Polydatin
Antiproliferative activity
Antiproliferative effect in breast cancer cells (MCF-7,
MCF-10A, and MDA-MB 231)
Decreasing lipid peroxidation activity in hamster induced with oral cancer
[66,67,68]
PGFCaCO3-PEGChelation-Gallic acidSuppressed 4T1 tumor growth[69]
Retinoic acids and hyaluronic acidHydrogen bondDialysisCurcuminPresent sensibility to GSH and promote the liberation of curcumin in esophageal cancer cells (ECA-109)[70]
SoluplusHydrogen bondDesolvation methodPosidonia oceanica ExtractInhibitory activity against neuroblastoma cell migration[56]
Whey protein–maltodextrin/Arabic gumHydrogen bondCoacervationGrape seed extractCombination of the different polymers improved microencapsulation[71]
Poly(ε-caprolactam)–hyaluronic acidHydrogen bondNanoprecipitationNaringeninAntiproliferation of lung cancer cells[72]
Table 6. Summary of some examples of polyphenol microencapsulation.
Table 6. Summary of some examples of polyphenol microencapsulation.
Wall MaterialsInteractionTechnique of EncapsulationPhenolic CompoundApplicationReferences
Maltodextrin/modified chitosan/pectinHydrogen bondSpay dryingPunica granatum peels extractThe cytotoxicity was improved when the extract was encapsulated in AGS (human gastric adenocarcinoma) and A549 (human lung carcinoma) cell lines[78]
Whey proteinHydrogen bondEmulsionGrape PhenolicAfter digestion, the activity of the polyphenols was stabilized[79]
MaltodextrinHydrogen bondSpray dryingGrape phenolicsAfter digestion, the activity and the bioaccessibility of the polyphenols increased[79]
Sodium alginateHydrogen bondExtrusionBifidobacterium bifidum and Lactobacillus gasseri in combination with quercetinCombining probiotics and quercetin resulted in a more effective protection and prevented hepatomegaly. Mechanistically, the results suggested that microencapsulated probiotics in combination with quercetin could exert the inhibition of the canonical Wnt/β-catenin signaling pathway in the colon [80]
Table 7. In vivo studies on the anticancer properties of encapsulated polyphenols.
Table 7. In vivo studies on the anticancer properties of encapsulated polyphenols.
Wall AgentEncapsulated PolyphenolTreatmentResultReferences
Chitosan, nanoencapsulationQuercetinC57BL6 mice used to examine anti-tumor activity. Tumors were induced in immunocompromised mice, using human xenografts obtained with A549 and MDA-MB-468 cells. Chitosan–quercetin nanoencapsulation increased superoxide dismutase activity in treated mice.Tumor regression (62.86% and 49.96% volume reduction for A549 and MDA-MB-468 cells, respectively) was observed after treatment with chitosan-nanoencapsulated quercetin, as a better anticancer agent than quercetin alone.[87]
Whey protein isolate, nanoencapsulationApigeninMale and female C57BL/6J mice were administered nanoencapsulated apigenin at 50 mg/kgChitosan nanoencapsulation of apigenin improved the latter’s bioavailability with respect to free apigenin.
Nanoencapsulation improved the absorption of apigenin
[61]
Poly(lactide-co-glycolide) and levanCurcuminThe nanoformulation consisted of 10 mg of curcumin, 50 mg of poly(lactide-co-glycolide), and 80 mg of levan. Intraperitoneal administration of the formulation for 17 daysThe nanoformulation of poly(lactide-co-glycolide) and levan with curcumin enhanced curcumin accumulation at the tumor site. The treatment also inhibited NF-kB.[88]
LiposomesPlumbagin and genistein (10:1 ratio)Xenografted tumors by subcutaneous injection of PC3 or LNCaP prostate cancer cells in female athymic nude mice. Liposomes (1.5 mg genistein/kg and 15 mg plumbagin/kg bodyweight) were administered intravenously for 18 days.Treatment with genistein and plumbagin-loaded liposomes decreased by nearly 80% the tumor growth.[89]
Dextran–deoxycholic acid amphiphilic polymerSilybin (co-encapsulated with paclitaxel)Subcutaneous inoculation of A549 cells in BALB/c nude mice. The nanoencapsulated paclitaxel and silybin (7 mg/kg and 10 mg/kg bodyweight, respectively) were administered through the tail vein.Co-encapsulated paclitaxel/silybin enhanced blood circulation from 1 to 5 h.Tthe improved blood circulation also enhanced the tumor accumulation of the compounds and caused tumor growth suppression[90]
Epigallocatechin-3-gallate–iron nanoparticles with doxorubicinEpigallocatechin-3-gallateIntravenous injection in 4T1 tumor-bearing BALB/c mice with epigallocatechin-3-gallate–iron nanoparticles with doxorubicin. Treatment decreased tumor volume and caused necrosis in tumors, which was associated with tumor growth inhibition.[91]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Picos-Salas, M.A.; García-Carrasco, M.; Heredia, J.B.; Cabanillas-Bojórquez, L.A.; Leyva-López, N.; Gutiérrez-Grijalva, E.P. Polyphenol-Loaded Polymeric Matrixes as Potential Biopharmaceuticals against Cancer. Macromol 2023, 3, 507-523. https://doi.org/10.3390/macromol3030030

AMA Style

Picos-Salas MA, García-Carrasco M, Heredia JB, Cabanillas-Bojórquez LA, Leyva-López N, Gutiérrez-Grijalva EP. Polyphenol-Loaded Polymeric Matrixes as Potential Biopharmaceuticals against Cancer. Macromol. 2023; 3(3):507-523. https://doi.org/10.3390/macromol3030030

Chicago/Turabian Style

Picos-Salas, Manuel Adrian, Melissa García-Carrasco, José Basilio Heredia, Luis Angel Cabanillas-Bojórquez, Nayely Leyva-López, and Erick Paul Gutiérrez-Grijalva. 2023. "Polyphenol-Loaded Polymeric Matrixes as Potential Biopharmaceuticals against Cancer" Macromol 3, no. 3: 507-523. https://doi.org/10.3390/macromol3030030

Article Metrics

Back to TopTop