We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Nanoencapsulated dietary polyphenols for cancer prevention and treatment: successes and challenges

    Shams Tabrez

    *Author for correspondence:

    E-mail Address: shamstabrez1@gmail.com and stabrez@kau.edu.sa

    King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia

    Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia

    ,
    Nasimudeen R Jabir

    Department of Biochemistry, Centre for Research & Development, PRIST University, Vallam, Thanjavur, Tamil Nadu, 613403, India

    ,
    Vaqar Mustafa Adhami

    Department of Dermatology, University of Wisconsin-Madison, WI 53706, USA

    ,
    Mohammad Imran Khan

    Department of Biochemistry, King Abdulaziz University, Jeddah, 21589, Saudi Arabia

    ,
    Mohammed Moulay

    King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia

    Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia

    ,
    Mohammad Amjad Kamal

    King Fahd Medical Research Center, King Abdulaziz University, Jeddah, 21589, Saudi Arabia

    &
    Hasan Mukhtar

    Department of Dermatology, University of Wisconsin-Madison, WI 53706, USA

    Published Online:https://doi.org/10.2217/nnm-2019-0398

    Many dietary polyphenols have been investigated for their therapeutic potential either as single agents or in combinations. Despite the significant anticancer potential of these polyphenols in in vitro cell culture and in vivo animal models, their clinical applications have been limited because of challenges such as ineffective systemic delivery, stability and low bioavailability. Nanoencapsulation of these polyphenols could prolong circulation, improve localization, enhance efficacy and reduce the chances of multidrug resistance. This review summarized the use of various polyphenols especially epigallocatechin gallate, quercetin, curcumin and resveratrol as nanoformulations for cancer prevention and treatment. Despite some success, more research is warranted to design a nanoencapsulated combination of polyphenols, effective in in vitro, in vivo and human systems.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. You W, Henneberg M. Cancer incidence increasing globally: the role of relaxed natural selection. Evol. Appl. 11(2), 140–152 (2017).
    • 2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA: Cancer J. Clin. 68(1), 7–30 (2018).
    • 3. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: Cancer. J. Clin. 68(6), 394–424 (2018).
    • 4. Cao Y, Teng J, Selbo J. Amorphous solid dispersion of epigallocatechin gallate for enhanced physical stability and controlled release. Pharmaceuticals 10(4), 88 (2017).
    • 5. Siddiqui IA, Adhami VM, Bharali DJ et al. Introducing nanochemoprevention as a novel approach for cancer control: proof of principle with green tea polyphenol epigallocatechin-3-gallate. Cancer Res. 69(5), 1712–1716 (2009). • The encapsulation of epigallocatechin-3-gallate (EGCG) in poly(lactic acid)-poly(ethylene glycol) nanoparticles (NPs) showed tenfold dose advantage for exerting its proapoptotic and angiogenesis inhibitory effects in both in vitro and in vivo systems.
    • 6. Khan N, Bharali DJ, Adhami VM et al. Oral administration of naturally occurring chitosan-based nanoformulated green tea polyphenol EGCG effectively inhibits prostate cancer cell growth in a xenograft model. Carcinogenesis 35(2), 415–423 (2014).
    • 7. Tabrez S, Priyadarshini M, Urooj M et al. Cancer chemoprevention by polyphenols and their potential application as nanomedicine. J. Environ. Sci. Health. C. Environ. Carcinog. Ecotoxicol. Rev. 31(1), 67–98 (2013). • The different combination of nanopolyphenols has been suggested as cancer chemotherapeutic agents.
    • 8. Mashhadi Malekzadeh A, Ramazani A, Tabatabaei Rezaei SJ, Niknejad H. Design and construction of multifunctional hyperbranched polymers coated magnetite nanoparticles for both targeting magnetic resonance imaging and cancer therapy. J. Colloid. Interface. Sci. 490, 64–73 (2017).
    • 9. Meybodi NM, Mortazavian AM, Monfared AB, Sohrabvandi S, Meybodi FA. Phytochemicals in cancer prevention: a review of the evidence. Iran. J. Cancer Prev. 10(1), e7219 (2017).
    • 10. Nosrati N, Bakovic M, Paliyath G. Molecular mechanisms and pathways as targets for cancer prevention and progression with dietary compounds. Int. J. Mol. Sci. 18(10), e2050 (2017).
    • 11. Mileo AM, Miccadei S. Polyphenols as modulator of oxidative stress in cancer disease: new therapeutic strategies. Oxid. Med. Cell. Longev. 2016, 6475624 (2016).
    • 12. Abbas M, Saeed F, Anjum FM et al. Natural polyphenols: an overview. Int. J. Food. Prop. 20(8), 1689–1699 (2017).
    • 13. Khan N, Mukhtar H. Tea polyphenols in promotion of human health. Nutrients 11(1), 39 (2019). • The role of tea polyphenols in the prevention of cancer, diabetes, cardiovascular and neurological diseases have been summarized in this article.
    • 14. Rezaei SJT, Malekzadeh AM, Ramazani A, Niknejad H. pH-sensitive magnetite nanoparticles modified with hyperbranched polymers and folic acid for targeted imaging and therapy. Curr. Drug. Deliv. 16(9), 839–848 (2019).
    • 15. Munin A, Edwards-Lévy F. Encapsulation of natural polyphenolic compounds: a review. Pharmaceutics. 3(4), 793–829 (2011).
    • 16. Faraji L, Nadri H, Moradi A et al. Aminoalkyl-substituted flavonoids: synthesis, cholinesterase inhibition, β-amyloid aggregation, and neuroprotective study. Med. Chem. Res. 28(7), 974–983 (2019).
    • 17. Hokmabad VR, Soodabeh D, Marziyeh A, Effat A, Roya S, Ali R. Effect of incorporating elaeagnus angustifolia extract in PCL-PEG-PCL nanofibers for bone tissue engineering. Front. Chem. Sci. Eng. 13(1), 108–119 (2019).
    • 18. Oliveira LDLD, Carvalho MVD, Melo L. Health promoting and sensory properties of phenolic compounds in food. Rev. Ceres. 61, 764–779 (2014).
    • 19. Hosseinzadeh Z, Ramazani A, Razzaghi-Asl N. Anti-cancer nitrogen-containing heterocyclic compounds. Curr. Org. Chem. 22(23), 2256–2279 (2018).
    • 20. Hosseini F, Ramazani A, Mohammadi-Khanaposhtani M et al. Design, synthesis, and biological evaluation of novel 4-oxobenzo[d]1,2,3-triazin-benzylpyridinum derivatives as potent anti-Alzheimer agents. Bioorg. Med. Chem. 27(13), 2914–2922 (2019).
    • 21. Hosseinzadeh Z, Ramazani A. An overview of the chemistry and pharmacological potentials of furanones skeletons. Curr. Org. Chem. 23(14), 1581–1599 (2019).
    • 22. Conte R, De Luca I, De Luise A, Petillo O, Calarco A, Peluso G. New therapeutic potentials of nanosized phytomedicine. J. Nanosci. Nanotechnol. 16, 8176–8187 (2016).
    • 23. Jalili-Baleh L, Babaei E, Abdpour S et al. A review on flavonoid-based scaffolds as multi-target-directed ligands (MTDLs) for Alzheimer's disease. Eur. J. Med. Chem. 152, 570–589 (2018).
    • 24. Granja A, Pinheiro M, Reis S. Epigallocatechin gallate nanodelivery systems for cancer therapy. Nutrients 8(5), 307 (2016).
    • 25. Islam MT, Tabrez S, Jabir NR et al. An insight into the therapeutic potential of major coffee components. Curr. Drug. Metab. 19(6), 544–556 (2018).
    • 26. Jabir NR, Islam MT, Tabrez S et al. An insight towards anticancer potential of major coffee constituents. BioFactors 44(4), 315–326 (2018). • The anticancer potential of different coffee components have been summarized in this article.
    • 27. Kang H, Rha SY, Oh KW, Nam CM. Green tea consumption and stomach cancer risk: a meta-analysis. Epidemiol. Health. 32, e2010001 (2010).
    • 28. De Pace RCC, Liu X, Sun M et al. Anticancer activities of (-)-epigallocatechin-3-gallate encapsulated nanoliposomes in MCF7 breast cancer cells. J. Liposome. Res. 23(3), 187–196 (2013).
    • 29. Kwak TW, Park SB, Kim H-J, Jeong Y-IL, Kang DH. Anticancer activities of epigallocatechin-3-gallate against cholangiocarcinoma cells. Onco. Targets. Ther. 10, 137–144 (2016).
    • 30. Granja A, Frias I, Neves AR, Pinheiro M, Reis S. Therapeutic potential of epigallocatechin gallate nanodelivery systems. Biomed. Res. Int. 2017, 5813793 (2017).
    • 31. Sanna V, Singh CK, Jashari R et al. Targeted nanoparticles encapsulating (−)-epigallocatechin-3-gallate for prostate cancer prevention and therapy. Sci. Rep. 7, 41573 (2017).
    • 32. Siddiqui IA, Adhami VM, Ahmad N, Mukhtar H. Nanochemoprevention: sustained release of bioactive food components for cancer prevention. Nutr. Cancer. 62(7), 883–890 (2010). •• Provides a foundation for the use of nano-EGCG to enhance the bioavailability and its effectiveness. The perceived toxicity could also be minimized by nano-EGCG.
    • 33. Shutava TG, Balkundi SS, Vangala P et al. Layer by layer coated gelatin nanoparticles as a vehicle for delivery of natural polyphenols. ACS Nano 3(7), 1877–1885 (2009).
    • 34. Chen C-C, Hsieh D-S, Huang K-J et al. Improving anticancer efficacy of (–)-epigallocatechin-3-gallate gold nanoparticles in murine B16F10 melanoma cells. Drug. Des. Devel. Ther. 8, 459–474 (2014).
    • 35. Siddiqui IA, Bharali DJ, Nihal M et al. 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. Nanomedicine 10(8), 1619–1626 (2014). •• Excellent anti-proliferative and pro-apoptotic effects of (-)-EGCG encapsulated in chitosan nanoparticles (NPs) have been reported in both in vitro and in vivo model.
    • 36. Narayanan S, Mony U, Vijaykumar DK, Koyakutty M, Paul-Prasanth B, Menon D. Sequential release of epigallocatechin gallate and paclitaxel from PLGA-casein core/shell nanoparticles sensitizes drug-resistant breast cancer cells. Nanomedicine 11(6), 1399–1406 (2015).
    • 37. Shafiei SS, Solati-Hashjin M, Samadikuchaksaraei A, Kalantarinejad R, Asadi-Eydivand M, Abu Osman NA. Epigallocatechin gallate/layered double hydroxide nanohybrids: preparation, characterization, and in vitro anti-tumor study. PLoS ONE 10(8), e0136530 (2015).
    • 38. Li J, Wu S, Wu C et al. Versatile surface engineering of porous nanomaterials with bioinspired polyphenol coatings for targeted and controlled drug delivery. Nanoscale 8(16), 8600–8606 (2016).
    • 39. Zeng L, Yan J, Luo L, Ma M, Zhu H. Preparation and characterization of (−)-epigallocatechin-3-gallate (EGCG)-loaded nanoparticles and their inhibitory effects on human breast cancer MCF-7 cells. Sci. Rep. 7, 45521 (2017).
    • 40. Yuan X, He Y, Zhou G, Li X, Feng A, Zheng W. Target challenging-cancer drug delivery to gastric cancer tissues with a fucose graft epigallocatechin-3-gallate-gold particles nanocomposite approach. J. Photochem. Photobiol. 183, 147–153 (2018).
    • 41. Hajipour H, Hamishehkar H, Nazari Soltan Ahmad S, Barghi S, Maroufi NF, Taheri RA. Improved anticancer effects of epigallocatechin gallate using RGD-containing nanostructured lipid carriers. Artif. Cells. Nanomed. Biotechnol. 46(S1), 283–292 (2018).
    • 42. Velavan B, Divya T, Sureshkumar A, Sudhandiran G. Nano-chemotherapeutic efficacy of (–)-epigallocatechin 3-gallate mediating apoptosis in A549 cells: involvement of reactive oxygen species mediated Nrf2/Keap1signaling. Biochem. Biophys. Res. Commun. 503(3), 1723–1731 (2018).
    • 43. Li K, Xiao G, Richardson JJ et al. Targeted therapy against metastatic melanoma based on self-assembled metal-phenolic nanocomplexes comprised of green tea catechin. Adv. Sci. (Weinh.) 6(5), 1801688 (2019).
    • 44. Wang C, Sang H, Wang Y et al. Foe to friend: supramolecular nanomedicines consisting of natural polyphenols and bortezomib. Nano Lett. 18(11), 7045–7051 (2018).
    • 45. Liang C, Guo B, Wu H et al. Aptamer-functionalized lipid nanoparticles targeting osteoblasts as a novel RNA interference-based bone anabolic strategy. Nat. Med. 21(3), 288–294 (2015).
    • 46. Dowdy SF. Overcoming cellular barriers for RNA therapeutics. Nat. Biotechnol. 35(3), 222–229 (2017).
    • 47. Shen W, Wang Q, Shen Y et al. Green tea catechin dramatically promotes RNAi mediated by low-molecular-weight polymers. ACS. Cent. Sci. 4(10), 1326–1333 (2018).
    • 48. Liu C, Shen W, Li B, Li T, Chang H, Cheng Y. Natural polyphenols augment cytosolic protein delivery by a functional polymer. Chem. Mater. 31(6), 1956–1965 (2019).
    • 49. Cheng X, Li M, Wang H, Cheng Y. All-small-molecule dynamic covalent gels with antibacterial activity by boronate-tannic acid gelation. Chinese Chem. Lett. doi:10.1016/j.cclet.2019.07.013 (2019).
    • 50. Li M, Wang H, Hu J et al. Smart hydrogels with antibacterial properties built from all natural building blocks. Chem. Mater. 31(18), 7678–7685 (2019).
    • 51. Massi A, Bortolini O, Ragno D et al. Research progress in the modification of quercetin leading to anticancer agents. Molecules 22(8), e1270 (2017).
    • 52. Amiri A, Ramazani A, Jahanshahi M, Moghadamnia AA. Synthesis and evaluating of nanoporous molecularly imprinted polymers for extraction of quercetin as a bioactive component of medicinal plants. Iran. J. Chem. Chem. Eng. 35(4), 11–19 (2016).
    • 53. Hashemzaei M, Far AD, Yari A et al. Anticancer and apoptosis-inducing effects of quercetin in vitro and in vivo. Oncol. Rep. 38(2), 819–828 (2017).
    • 54. Amiri A, Ramazani A, Jahanshahi M, Moghadamnia AA. Synthesis of a nanostructured molecularly imprinted acrylic acid-based network copolymer as a solid sorbentforthe quercetinextraction. J. Nanostruct. 4(3), 277–283 (2014).
    • 55. Chuan L, Jia Z, Yu-Jiao Z et al. Biocompatible and biodegradable nanoparticles for enhancement of anti-cancer activities of phytochemicals. Chin. J. Nat. Med. 13(9), 641–652 (2015).
    • 56. Huo C, Wan SB, Lam WH et al. The challenge of developing green tea polyphenols as therapeutic agents. Inflammopharmacology 16(5), 248–252 (2008).
    • 57. Pool H, Mendoza S, Xiao H, McClements DJ. Encapsulation and release of hydrophobic bioactive components in nanoemulsion-based delivery systems: impact of physical form on quercetin bioaccessibility. Food. Funct. 4(1), 162–174 (2013).
    • 58. Bishayee K, Khuda-Bukhsh AR, Huh S-O. PLGA-loaded gold-nanoparticles precipitated with quercetin downregulate HDAC-Akt activities controlling proliferation and activate p53-ROS crosstalk to induce apoptosis in hepatocarcinoma cells. Mol. Cells 38(6), 518–527 (2015).
    • 59. Ren K-W, Li Y-H, Wu G et al. Quercetin nanoparticles display antitumor activity via proliferation inhibition and apoptosis induction in liver cancer cells. Int. J. Oncol. 50(4), 1299–1311 (2017).
    • 60. Penalva R, González-Navarro CJ, Gamazo C, Esparza I, Irache JM. Zein nanoparticles for oral delivery of quercetin: pharmacokinetic studies and preventive anti-inflammatory effects in a mouse model of endotoxemia. Nanomedicine 13(1), 103–110 (2017).
    • 61. Singh S, Kushwah V, Agrawal AK, Jain S. Insulin- and quercetin-loaded liquid crystalline nanoparticles: implications on oral bioavailability, antidiabetic and antioxidant efficacy. Nanomedicine. 13(5), 521–537 (2018).
    • 62. Nan W, Ding L, Chen H et al. Topical use of quercetin-loaded chitosan nanoparticles against ultraviolet B radiation. Front. Pharmacol. 9, e826 (2018).
    • 63. Yilmaz M, Karanastasis AA, Chatziathanasiadou MV et al. Inclusion of quercetin in gold nanoparticles decorated with supramolecular hosts amplifies its tumor targeting properties. ACS. Appl. Bio. Mater. 2(7), 2715–2725 (2019).
    • 64. Barbosa AI, Costa Lima SA, Reis S. Application of pH-responsive fucoidan/chitosan nanoparticles to improve oral quercetin delivery. Molecules 24(2), 346 (2019).
    • 65. Srisa-Nga K, Mankhetkorn S, Okonogi S, Khonkarn R. Delivery of superparamagnetic polymeric micelles loaded with quercetin to hepatocellular carcinoma cells. J. Pharm. Sci. 108(2), 996–1006 (2019).
    • 66. Gholibegloo E, Mortezazadeh T, Salehian F et al. Folic acid decorated magnetic nanosponge: an efficient nanosystem for targeted curcumin delivery and magnetic resonance imaging. J. Colloid. Interface. Sci. 556, 128–139 (2019).
    • 67. Gholibegloo E, Mortezazadeh T, Salehian F, Ramazani A, Amanlou M, Khoobi M. Improved curcumin loading, release, solubility and toxicity by tuning the molar ratio of cross-linker to β-cyclodextrin. Carbohydr. Polym. 213, 70–78 (2019).
    • 68. Hewlings SJ, Kalman DS. Curcumin: a review of its' effects on human health. Foods 6(10), e92 (2017).
    • 69. Tomeh MA, Hadianamrei R, Zhao X. A review of curcumin and its derivatives as anticancer agents. Int. J. Mol. Sci. 20(5), 1033 (2019).
    • 70. Khalil NM, Nascimento TCFD, Casa DM et al. Pharmacokinetics of curcumin-loaded PLGA and PLGA–PEG blend nanoparticles after oral administration in rats. Colloids. Surf. B Biointerfaces. 101, 353–360 (2013).
    • 71. Zhou S, Zhang S, Shen H et al. Curcumin inhibits cancer progression through regulating expression of microRNAs. Tumour. Biol. 39(2), doi:10.1177/1010428317691680 (2017).
    • 72. Liu G, Xiang T, Wu Q-F, Wang W-X. Curcumin suppresses the proliferation of gastric cancer cells by downregulating H19. Oncol. Lett. 12(6), 5156–5162 (2016).
    • 73. Zhang L, Yang G, Zhang R et al. Curcumin inhibits cell proliferation and motility via suppression of TROP2 in bladder cancer cells. Int. J. Oncol. 53(2), 515–526 (2018).
    • 74. Ghalandarlaki N, Alizadeh AM, Ashkani-Esfahani S. Nanotechnology-applied curcumin for different diseases therapy. Biomed. Res. Int. 2014, 394264 (2014).
    • 75. Shaikh J, Ankola DD, Beniwal V, Singh D, Kumar MNVR. Nanoparticle encapsulation improves oral bioavailability of curcumin by at least 9-fold when compared to curcumin administered with piperine as absorption enhancer. Eur. J. Pharm. Sci. 37(3), 223–230 (2009).
    • 76. Tsai Y-M, Chien C-F, Lin L-C, Tsai T-H. Curcumin and its nano-formulation: the kinetics of tissue distribution and blood–brain barrier penetration. Int. J. Pharm. 416(1), 331–338 (2011).
    • 77. Xie X, Tao Q, Zou Y et al. PLGA nanoparticles improve the oral bioavailability of curcumin in rats: characterizations and mechanisms. J. Agric. Food. Chem. 59(17), 9280–9289 (2011).
    • 78. Yallapu MM, Ebeling MC, Khan S et al. Novel curcumin-loaded magnetic nanoparticles for pancreatic cancer treatment. Mol. Cancer Ther. 12(8), 1471–1480 (2013).
    • 79. Udompornmongkol P, Chiang B-H. Curcumin-loaded polymeric nanoparticles for enhanced anti-colorectal cancer applications. J. Biomater. Appl. 30(5), 537–546 (2015).
    • 80. Thadakapally R, Aafreen A, Aukunuru J, Habibuddin M, Jogala S. Preparation and characterization of PEG-albumin-curcumin nanoparticles intended to treat breast cancer. Ind. J. Pharm. Sci. 78(1), 65 (2016).
    • 81. Kalashnikova I, Mazar J, Neal CJ et al. Nanoparticle delivery of curcumin induces cellular hypoxia and ROS-mediated apoptosis via modulation of Bcl-2/Bax in human neuroblastoma. Nanoscale 9(29), 10375–10387 (2017).
    • 82. Song W, Muthana M, Mukherjee J, Falconer RJ, Biggs CA, Zhao X. Magnetic-silk core–shell nanoparticles as potential carriers for targeted delivery of curcumin into human breast cancer cells. ACS Biomater. Sci. Eng. 3(6), 1027–1038 (2017).
    • 83. Samrot AV, Burman U, Philip SA, N S Chandrasekaran K. Synthesis of curcumin loaded polymeric nanoparticles from crab shell derived chitosan for drug delivery. Inform. Med. Unlocked. 10, 159–182 (2018).
    • 84. Vijayakurup V, Thulasidasan AT, G MS et al. Chitosan encapsulation enhances the bioavailability and tissue retention of curcumin and improves its efficacy in preventing B[a]P-induced lung carcinogenesis. Cancer Prev. Res. 12(4), 225–236 (2019).
    • 85. Harini L, Srivastava S, Gnanakumar GP et al. An ingenious non-spherical mesoporous silica nanoparticle cargo with curcumin induces mitochondria-mediated apoptosis in breast cancer (MCF-7) cells. Oncotarget 10(11), 1193–1208 (2019).
    • 86. Kim J-H, Park E-Y, Ha H-K et al. Resveratrol-loaded nanoparticles induce antioxidant activity against oxidative stress. Asian-Australas. J. Anim. Sci. 29(2), 288–298 (2016).
    • 87. Pangeni R, Sahni JK, Ali J, Sharma S, Baboota S. Resveratrol: review on therapeutic potential and recent advances in drug delivery. Expert Opin. Drug. Deliv. 11(8), 1285–1298 (2014).
    • 88. Santos AC, Pereira I, Magalhães M et al. Targeting cancer via resveratrol-loaded nanoparticles administration: focusing on in vivo evidence. AAPS J. 21(4), 57 (2019).
    • 89. Ruivo J, Francisco C, Oliveira R et al. The main potentialities of resveratrol for drug delivery systems. Braz. J. Pharm. 51(3), 499–513 (2015).
    • 90. Dai Z, Li Y, Quarles LD et al. Resveratrol enhances proliferation and osteoblastic differentiation in human mesenchymal stem cells via ER-dependent ERK1/2 activation. Phytomedicine 14(12), 806–814 (2007).
    • 91. Udenigwe CC, Ramprasath VR, Aluko RE, Jones PJH. Potential of resveratrol in anticancer and anti-inflammatory therapy. Nutr. Rev. 66(8), 445–454 (2008).
    • 92. Johnson WD, Morrissey RL, Usborne AL et al. Subchronic oral toxicity and cardiovascular safety pharmacology studies of resveratrol, a naturally occurring polyphenol with cancer preventive activity. Food. Chem. Toxicol. 49(12), 3319–3327 (2011).
    • 93. Neves AR, Lúcio M, Martins S, Lima JLC, Reis S. Novel resveratrol nanodelivery systems based on lipid nanoparticles to enhance its oral bioavailability. Int. J. Nanomedicine 8, 177–187 (2013).
    • 94. Siu FY, Ye S, Lin H, Li S. Galactosylated PLGA nanoparticles for the oral delivery of resveratrol: enhanced bioavailability and in vitro anti-inflammatory activity. Int. J. Nanomedicine 13, 4133–4144 (2018).
    • 95. Peñalva R, Morales J, González-Navarro CJ et al. Increased oral bioavailability of resveratrol by its encapsulation in casein nanoparticles. Int. J. Mol. Sci. 19(9), 2816 (2018).
    • 96. Breuss JM, Atanasov AG, Uhrin P. Resveratrol and its effects on the vascular system. Int. J. Mol. Sci. 20(7), 1523 (2019).
    • 97. Davidov-Pardo G, Mcclements DJ. Nutraceutical delivery systems: resveratrol encapsulation in grape seed oil nanoemulsions formed by spontaneous emulsification. Food. Chem. 167, 205–212 (2015).
    • 98. Bernkop-Schnürch A. Nanocarrier systems for oral drug delivery: do we really need them? Eur. J. Pharm. Sci. 49(2), 272–277 (2013).
    • 99. Teskač K, Kristl J. The evidence for solid lipid nanoparticles mediated cell uptake of resveratrol. Int. J. Pharm. 390(1), 61–69 (2010).
    • 100. Karthikeyan S, Hoti SL, Prasad NR. Resveratrol loaded gelatin nanoparticles synergistically inhibits cell cycle progression and constitutive NF-kappaB activation, and induces apoptosis in non-small cell lung cancer cells. Biomed. Pharmacother. 70, 274–282 (2015).
    • 101. Sanna V, Siddiqui IA, Sechi M, Mukhtar H. Resveratrol-loaded nanoparticles based on poly(epsilon-caprolactone) and poly(D,L-lactic-co-glycolic acid)-poly(ethylene glycol) blend for prostate cancer treatment. Mol. Pharm. 10(10), 3871–3881 (2013).
    • 102. Figueiró F, Bernardi A, Frozza RL et al. Resveratrol-loaded lipid-core nanocapsules treatment reduces in vitro and in vivo glioma growth. J. Biomed. Nanotechnol. 9(3), 516–526 (2013).
    • 103. Jung K-H, Lee JH, Park JW et al. Resveratrol-loaded polymeric nanoparticles suppress glucose metabolism and tumor growth in vitro and in vivo. Int. J. Pharm. 478(1), 251–257 (2015).
    • 104. Guo L, Peng Y, Li Y et al. Cell death pathway induced by resveratrol-bovine serum albumin nanoparticles in a human ovarian cell line. Oncol. Lett. 9(3), 1359–1363 (2015).
    • 105. Wang W, Zhang L, Chen T et al. Anticancer effects of resveratrol-loaded solid lipid nanoparticles on human breast cancer cells. Molecules. 22(11), 1814 (2017).
    • 106. Nassir AM, Shahzad N, Ibrahim IaA, Ahmad I, Md S, Ain MR. Resveratrol-loaded PLGA nanoparticles mediated programmed cell death in prostate cancer cells. Saudi. Pharm. J. 26(6), 876–885 (2018).
    • 107. Zhang D, Zhang J, Zeng J et al. Nano-gold loaded with resveratrol enhance the anti-hepatoma effect of resveratrol in vitro and in vivo. J. Biomed. Nanotechnol. 15(2), 288–300 (2019). •• The increase in antihepatoma potential by nanogold loaded resveratrol has been reported in vitro and in vivo systems.
    • 108. Niedzwiecki A, Roomi MW, Kalinovsky T, Rath M. Anticancer efficacy of polyphenols and their combinations. Nutrients 8(9), 552 (2016).
    • 109. Singh CK, Siddiqui IA, El-Abd S, Mukhtar H, Ahmad N. Combination chemoprevention with grape antioxidants. Mol. Nutr. Food Res. 60(6), 1406–1415 (2016).
    • 110. Singh CK, George J, Ahmad N. Resveratrol-based combinatorial strategies for cancer management. Ann. N.Y. Acad. Sci. 1290(1), 113–121 (2013).
    • 111. Sun X-D, Liu X-E, Huang D-S. Curcumin induces apoptosis of triple-negative breast cancer cells by inhibition of EGFR expression. Mol. Med. Rep. 6(6), 1267–1270 (2012).
    • 112. Du Q, Hu B, An H-M et al. Synergistic anticancer effects of curcumin and resveratrol in Hepa1-6 hepatocellular carcinoma cells. Oncol. Rep. 29(5), 1851–1858 (2013).
    • 113. Shindikar A, Singh A, Nobre M, Kirolikar S. Curcumin and resveratrol as promising natural remedies with nanomedicine approach for the effective treatment of triple negative breast cancer. J. Oncol. 2016, e9750785 (2016).
    • 114. Masuelli L, Stefano ED, Fantini M et al. Resveratrol potentiates the in vitro and in vivo anti-tumoral effects of curcumin in head and neck carcinomas. Oncotarget 5(21), 10745–10762 (2014).
    • 115. Sanna V, Pala N, Dessì G et al. Single-step green synthesis and characterization of gold-conjugated polyphenol nanoparticles with antioxidant and biological activities. Int. J. Nanomedicine 9, 4935–4951 (2014). •• The green synthesis and characterization of gold-conjugated polyphenol NPs highlighting better antioxidant and biological activities have been reported.