Resveratrol Encapsulation as an Anticancer Agent: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Resveratrol is a polyphenolic compound that has gained considerable attention in the past decade due to its multifaceted therapeutic potential, including anti-inflammatory and anticancer properties. However, its anticancer efficacy is impeded by low water solubility, dose-limiting toxicity, low bioavailability, and rapid hepatic metabolism. To overcome these hurdles, various nanoparticles such as organic and inorganic nanoparticles, liposomes, polymeric nanoparticles, dendrimers, solid lipid nanoparticles, gold nanoparticles, zinc oxide nanoparticles, zeolitic imidazolate frameworks, carbon nanotubes, bioactive glass nanoparticles, and mesoporous nanoparticles were employed to deliver resveratrol, enhancing its water solubility, bioavailability, and efficacy against various types of cancer.

  • anticancer properties
  • bioavailability
  • nanoparticles
  • polyphenolic compound
  • resveratrol encapsulation

1. Introduction

Cancer represents a significant global health challenge, standing as the second most common and prevalent cause of mortality. According to the World Health Organization (WHO), 10 million people died in 2020, a figure projected to increase by 70% in the next two decades [1][2]. The estimated worldwide economic cost of cancer is USD 25.2 trillion for 2020 to 2050 [3]. Conventional treatments include chemotherapy, immunotherapy, radiotherapy, and surgery [4][5]. However, modalities such as chemotherapy and radiotherapy encounter challenges in the form of radioresistance and chemoresistance. To overcome these limitations, exploring innovative therapeutic molecules and advancing drug delivery systems is imperative to effectively treat various cancer forms [6][7].
Plant-derived sources present an alternative reservoir of bioactive compounds with potential applications of therapeutic or prophylactic agents against various diseases [8][9][10][11][12][13][14][15]. To date, 350,000 vascular plant species are known, and new species are discovered yearly [16]. It is still a broad and understudied area of research with many prospects for new therapeutic development. However, active substances can be extracted and serve as valuable resources for medicinal applications and as building blocks for synthetic and semi-synthetic substances [17]. Among the diverse array of phytochemicals, encompassing terpenes, alkaloids, essential oils, flavonoids, gums, and a range of primary and secondary metabolic components, discernible medicinal effects have been identified [18][19]. A notable statistic underscores the significance of natural origins, indicating that 51% of the 1211 newly approved small-molecule drugs worldwide between 1981 and 2014 were derived from natural products [20]. Polyphenols have been known to have various preventive effects on different conditions such as diabetes, cardiovascular disease, neurodegenerative disorder, and obesity [21][22][23][24][25][26][27][28][29], and have been extensively studied to determine their anticancer potential and incorporate them into cancer treatment modalities like chemotherapy and targeted therapy [30][31][32][33].
Resveratrol (RSV), a stilbenoid polyphenolic compound, has emerged as a promising anticancer agent. However, its therapeutic potential is hindered due to its pharmacokinetic properties, such as chemical instability (due to oxidation and photosensitivity), low water solubility, low bioavailability, rapid metabolism, and elimination [34][35][36][37]. Many attempts have been made to overcome these hurdles using nanoparticles. The encapsulation of resveratrol in nanoparticles increases its absorption, bioavailability, and sustained release [38][39][40][41][42][43][44]. Cutting-edge nanoparticle technology has revolutionized engineers’ and scientists’ approaches to various fields of study. Nanoparticles are advancing the development of novel drug delivery systems, material engineering, and diagnostic sciences [45][46][47][48].

2. Resveratrol

Resveratrol (RSV) (3,5,4′-trihydroxystilbene) is a well-known naturally occurring polyphenolic compound present in various types of plants such as legumes, blueberries, cranberries, grapes, eucalyptus, and various grasses [49][50]. RSV is a secondary metabolite in different plant families, such as Gnetaceae, Dipterocarpaceae, Leguminosae, and Cyperaceae. Moreover, plants produce RSV in response to pathogen attacks, UV radiation, damage, stress, and exposure to ozone [51]. RSV can be modified into various structures, e.g., pterostilbene, 4,4′-hydroxy-trans-stilbene, monoalkoxy, dialkoxy derivatives, and trans 3,4′,5-trimethoxystilbene [52]. RSV has been confirmed to have many health benefits, such as antiviral, antioxidant, anti-inflammatory, neurological, and heart-disease-prevention properties [53][54][55][56]. RSV also enhances the antiviral activity of various drugs, such as zidovudine, zalcitabine, and didanosine [57][58][59]. The anticancer properties of RSV against multiple malignancies were first described by Jang et al. in 1997 [60].

2.1. The Structure and Physical Properties of Resveratrol

Resveratrol is a hydrophobic compound characterized by a molecular weight of 228.25 g/mol and a melting point of 254 °C [61]. The solubility of RSV is 30 µg/mL in water. RSV is soluble in polar compounds, especially dimethyl sulfoxide (DMSO) and ethanol [61][62]. It exists in two isoforms, i.e., trans-RSV and cis-RSV, depicted in Figure 1. The trans-isomer is more stable and predominant, with more therapeutic properties [63]. Cytotoxicity studies on pancreatic cancer, breast cancer, small-cell lung carcinoma, colon cancer, and prostate cancer cell lines revealed that trans-resveratrol possesses slightly more potent cytotoxic properties than the cis-isomer, attributed to its better bioavailability and biodistribution [64][65][66]. Upon exposure to sunlight or UV radiation at 254 nm or 366 nm, trans-RSV converts into cis-RSV and vice versa. Trans-RSV is more thermo- and photo-stable than cis-RSV. Trans-RSV remains stable in neutral aqueous buffers for 42 h and 28 days at acidic pH; however, cis-RSV remains stable at neutral pH [67][68]. Additionally, RSV becomes unstable when exposed to high humidity and prolonged exposure to light [69]. Additionally, there is evidence that RSV oxidizes into quinines and semiquinones, which cause cell damage [70].
Figure 1. The structural formula for resveratrol isomers.

2.2. Metabolism of Resveratrol

RSV has a half-life of 8 to 14 min in plasma after oral treatment, and its plasma concentrations are often low, sometimes not detectable at all [71]. Upon oral administration, RSV is absorbed by enterocytes, which undergo sulfate conjugation and glucuronidation in the liver and intestine, leading to the formation of trans-resveratrol-3-sulfate and trans-resveratrol-3-O-glucuronide metabolites [36]. Meanwhile, a small quantity of free RSV remains in the blood circulation after being absorbed by plasma proteins like albumin, blood cells, and lipoproteins [72]. Primarily, RSV is administered orally; different levels of free RSV can be detected in urine, ranging from negligible quantities to 17%. The sulfated form of cis-RSV-4′-sulfate is more prevalent than the glucuronidated form. In addition, several studies have found that a minor amount of RSV metabolites are excreted in human feces [73][74].

2.3. Mechanism of Action of Resveratrol against Cancer

RSV is known for its anticancerous properties, mediating apoptosis, cell growth, metastasis, and angiogenesis, as illustrated in Figure 2 [75][76][77][78][79][80][81][82]. Its mechanism involves reducing angiogenesis and increasing apoptosis through the inhibition of vascular endothelial growth factor (VEGF) expression by downregulating hypoxia-inducible factor 1 (HIF-1) [76][77]. RSV promotes apoptosis by arresting the cell cycle at G0/G1 by upregulating the expression of cyclin-dependent kinase (CDK) inhibitors p21 and p27. It also upregulates the expression of cyclin D1, CDK 4, and CDK6 [83].
Figure 2. Schematic representation of resveratrol’s anticancer effects.
RSV causes the apoptosis of cancerous cells by downregulating HER2/neu expression [84]. RSV also inhibits cancer-promoting molecular pathways, such as nuclear factor kappa B (NF-kB), PI3K/AKT/mTOR, and STAT3 [85]. AKT serine/threonine kinase is an oncogene protein involved in cell survival, apoptosis, proliferation, and growth. It is involved in the phosphoinositide 3-kinase (PI3K)/AKT signaling pathway activated by inflammation, DNA damage, and growth factors [86]. AKT overexpression has been observed in various cancers [87][88]. RSV also inhibits NF-kB-regulated genes such as VEGF, B cell lymphoma protein-2 (Bcl-2), B-cell lymphoma-extra-large (Bcl-xL), and matrix metalloproteinase (MMP). As NF-kB activates, it alters caspase activity and increases antiapoptotic gene expression, promoting cell proliferation and protecting cells from apoptosis [89].
Moreover, RSV also activates the p53 kinase mediated by MAPKs (mitogen-activated protein kinases) [90]. RSV induces the activation of Apaf-1, which involves activating cytochrome C-dependent caspase and triggers a cascade of apoptosis events [91]. RSV inhibits the activity of the cyclooxygenase enzyme (COX) which converts arachidonic acid to prostaglandin, an inflammatory factor that induces tumor cell proliferation [92][93]. RSV also activates the SIRT1 enzyme, which deacetylates histone and non-histone proteins. SIRT1 regulates inflammation, cell cycle defects, and metabolic control [94][95]. In vitro research demonstrates RSV’s efficacy at reducing cell proliferation and promoting apoptosis by downregulating molecular targets, including p-Akt, cyclin D1, the mammalian target of rapamycin, and androgen receptor (AR) protein [96].
Despite its therapeutic potential, RSV has low gastrointestinal (GI) absorption because of its low water solubility and quick metabolism, and the degradation of RSV by oxidative enzymes [97]. Moreover, cancer cells can develop resistance to chemotherapeutic drugs due to mutations known as multidrug resistance (MDR). Other carcinogenesis processes lead to MDR, including pathways leading to apoptosis, DNA damage response, downstream signaling pathways, changes in drug efflux attributable to modifications in proteins involved in drug transfer from the cell membrane, changes in enzymes involved in drug processing and metabolism, changes in the composition of the cell membrane, cancer stem cells (CSCs), epithelial–mesenchymal transition, and changes in the tumor environment [98].
A potential solution to these problems involves the development of nanoparticles capable of carrying resveratrol, ensuring targeted delivery without inducing toxicity [97].
Critical considerations in nanoparticle design, crucial for an effective drug delivery system, include the following:
  • Nanoparticles should enhance free molecules’ specificity, efficacy, therapeutic index, and tolerability [99].
  • Nanoparticles must be non-toxic, non-immunogenic, non-thrombogenic, and biodegradable [100].
  • They should also protect and maintain active compounds’ structural integrity and enhance their bioavailability [40][101][102].

3. Application of Nanoparticles to Improve the Therapeutic Potential of Resveratrol for Cancer

In nanotechnology, a particle is often categorized according to its physical diameter: ultrafine particles typically have a physical diameter between 1 and 100 nm in at least one dimension [40]. Drugs can be conjugated to nanoparticle surfaces through covalent or ionic bonds, structural absorption, or encapsulation inside nanoparticle cores [103]. These nanoparticle-based formulations can increase absorption, bioavailability, and chemical integrity, enhance permeability and retention effect (EPR) across the biological membrane, and ensure the optimal dosage of drugs reaches the cancer target cells [104][105]. Biodistribution studies have demonstrated that RSV-loaded nanoparticles or RSV-conjugated nanoparticles have a much longer circulation time than free RSV [106][107][108][109]. Nanoparticles protect RSV from rapid metabolism and elimination, resulting in sustained blood levels. According to pharmacokinetic studies, nanoencapsulated RSV has an extended half-life [110]. Pharmacokinetic studies have also revealed that RSV bioavailability is increased when delivered in nanoformulations. Nanoparticles improve RSV absorption in the gastrointestinal tract and protect first-pass metabolism in the liver [111][112][113]. Biodistribution studies revealed that nanoparticles enhanced RSV accumulation in tumor tissues compared to free RSV due to enhanced permeability and retention (EPR) in tumor vasculature. Nanoformulations provide controlled and sustained RSV release. The controlled release of RSV contributes to prolonged therapeutic effects and decreases the need for frequent dosing [114][115].
Various strategies, e.g., triggered drug release and stimuli-responsive approaches, have been used in RSV nanoformulation to release RSV in response to specific internal or external triggers. These strategies aim to increase therapeutic efficacy and precise control, and minimize the side effects of drug release. The common stimuli-responsive approaches are pH-responsive release, enzyme-responsive release, temperature-responsive release, redox-responsive release, light-responsive release, magnetic-responsive release, and ultrasound-responsive release [116].
In pH-sensitive nanoparticles, RSV is released in response to acidic pH conditions in tumor microenvironments. This pH-triggered release enhances drug delivery to cancer cells while minimizing release in normal tissues [117]. Thermosensitive nanoparticles loaded with RSV release the drug in response to local temperature changes. Hyperthermia treatment can trigger drug release [118]. The enzyme-triggered release of RSV can be achieved by conjugating enzyme-cleavable compounds in the nanoparticle structure. The presence of disease-specific enzymes, such as matrix metalloproteinases in tumors, releases RSV [119]. Redox-responsive nanoparticles release RSV in response to elevated reactive oxygen species (ROS) at the tumor site. Incorporating photo-responsive materials into resveratrol nanoparticles allows light-induced drug release [120]. Resveratrol-loaded magnetic nanoparticles can be guided to specific target sites using external magnets. Magnetic fields induce drug release at the desired location [121]. Ultrasound-responsive nanoparticles loaded with resveratrol can be triggered to release the drug at the target site using focused ultrasound waves, providing spatial and temporal control. These stimuli-activated approaches offer precise control over drug-release kinetics, improving resveratrol’s therapeutic index and minimizing systemic side effects. The trigger mechanism’s choice depends on the target tissue’s specific characteristics and the desired therapeutic outcome [122].
Nanoparticles can broadly be categorized into organic and inorganic types, with recent extensive studies focusing on organic particles. Specifically, liposomes, polymersomes, polymer constructions, and micelles are used for imaging and drug and gene delivery methods. Although inorganic nanoparticles exhibit highly material- and size-dependent physicochemical characteristics, incomparable with conventional lipid- or polymer-based NPs, they have also attracted researchers’ interest in recent years [123]. Various nanoformulations, including liposomes, metallic nanoparticles, solid lipid nanoparticles, micelles, polymeric nanoparticles, and inorganic nanoparticles, are illustrated in Figure 3 and Figure 4.
Figure 3. A variety of resveratrol-loaded organic nanoparticles.
Figure 4. Various types of resveratrol-loaded/conjugated inorganic nanoparticles.
Several attempts have been made to develop nanotechnology-based strategies and increase bioavailability and effectiveness in various cancer models [40][41][42][43][44][124]. In an initial attempt at encapsulating RSV, chitosan NPs were used. The study conducted by Yao et al. showed that sustained release of RSV, with resveratrol-loaded nanoparticles at lower concentrations, caused an increased percentage of cell death compared to an equivalent dose of free resveratrol [125].

This entry is adapted from the peer-reviewed paper 10.3390/ph17010126

References

  1. Sleeman, K.E.; de Brito, M.; Etkind, S.; Nkhoma, K.; Guo, P.; Higginson, I.J.; Gomes, B.; Harding, R. The Escalating Global Burden of Serious Health-Related Suffering: Projections to 2060 by World Regions, Age Groups, and Health Conditions. Lancet Glob. Health 2019, 7, e883–e892.
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249.
  3. Chen, S.; Cao, Z.; Prettner, K.; Kuhn, M.; Yang, J.; Jiao, L.; Wang, Z.; Li, W.; Geldsetzer, P.; Bärnighausen, T.; et al. Estimates and Projections of the Global Economic Cost of 29 Cancers in 204 Countries and Territories from 2020 to 2050. JAMA Oncol. 2023, 9, 465.
  4. Mathan, S.V.; Rajput, M.; Singh, R.P. CHAPTER 14—Chemotherapy and Radiation Therapy for Cancer. In Understanding Cancer; Jain, B., Pandey, S., Eds.; Academic Press: New York, NY, USA, 2022; pp. 217–236. ISBN 978-0-323-99883-3.
  5. Baskar, R.; Lee, K.A.; Yeo, R.; Yeoh, K.-W. Cancer and Radiation Therapy: Current Advances and Future Directions. Int. J. Med. Sci. 2012, 9, 193–199.
  6. Senapati, S.; Mahanta, A.K.; Kumar, S.; Maiti, P. Controlled Drug Delivery Vehicles for Cancer Treatment and Their Performance. Signal Transduct. Target. Ther. 2018, 3, 7.
  7. Miller, K.D.; Siegel, R.L.; Lin, C.C.; Mariotto, A.B.; Kramer, J.L.; Rowland, J.H.; Stein, K.D.; Alteri, R.; Jemal, A. Cancer Treatment and Survivorship Statistics, 2016. CA Cancer J. Clin. 2016, 66, 271–289.
  8. Nagori, K.; Nakhate, K.T.; Yadav, K.; Ajazuddin; Pradhan, M. Unlocking the Therapeutic Potential of Medicinal Plants for Alzheimer’s Disease: Preclinical to Clinical Trial Insights. Future Pharmacol. 2023, 3, 877–907.
  9. Singh Purewal, S.; Punia Bangar, S.; Kaur, P. (Eds.) Recent Advances in Citrus Fruits; Springer International Publishing: Cham, Switzerland, 2023; ISBN 978-3-031-37533-0.
  10. El-Saadony, M.T.; Zabermawi, N.M.; Zabermawi, N.M.; Burollus, M.A.; Shafi, M.E.; Alagawany, M.; Yehia, N.; Askar, A.M.; Alsafy, S.A.; Noreldin, A.E. Nutritional Aspects and Health Benefits of Bioactive Plant Compounds against Infectious Diseases: A Review. Food Rev. Int. 2023, 39, 2138–2160.
  11. Adetuyi, B.O.; Odelade, K.A.; Odine, G.O.; Adetuyi, O.A.; Omowumi, S.O.; Ogunlana, O.O.; Egbuna, C. Neurorestorative Potential of Medicinal Plants and Their Phytochemicals. In Phytochemical Drug Discovery for Central Nervous System Disorders; Wiley: Hoboken, NJ, USA, 2023; pp. 291–310. ISBN 978-1-119-79412-7.
  12. Čižmárová, B.; Hubková, B.; Tomečková, V.; Birková, A. Flavonoids as Promising Natural Compounds in the Prevention and Treatment of Selected Skin Diseases. Int. J. Mol. Sci. 2023, 24, 6324.
  13. Gorain, B.; Karmakar, V.; Sarkar, B.; Dwivedi, M.; Leong, J.T.L.; Toh, J.H.; Seah, E.; Ling, K.Y.; Chen, K.Y.; Choudhury, H.; et al. Biomacromolecule-Based Nanocarrier Strategies to Deliver Plant-Derived Bioactive Components for Cancer Treatment: A Recent Review. Int. J. Biol. Macromol. 2023, 253, 126623.
  14. Abdallah, E.M.; Alhatlani, B.Y.; de Paula Menezes, R.; Martins, C.H. Back to Nature: Medicinal Plants as Promising Sources for Antibacterial Drugs in the Post-Antibiotic Era. Plants 2023, 12, 3077.
  15. Islam, M.R.; Jony, M.H.; Thufa, G.K.; Akash, S.; Dhar, P.S.; Rahman, M.M.; Afroz, T.; Ahmed, M.; Hemeg, H.A.; Rauf, A.; et al. A Clinical Study and Future Prospects for Bioactive Compounds and Semi-Synthetic Molecules in the Therapies for Huntington’s Disease. Mol. Neurobiol. 2023.
  16. Gosline, G.; Bidault, E.; van der Burgt, X.; Cahen, D.; Challen, G.; Condé, N.; Couch, C.; Couvreur, T.L.; Dagallier, L.-P.M.; Darbyshire, I. A Taxonomically-Verified and Vouchered Checklist of the Vascular Plants of the Republic of Guinea. Sci. Data 2023, 10, 327.
  17. Schultz, F.; Garbe, L. How to Approach a Study in Ethnopharmacology? Providing an Example of the Different Research Stages for Newcomers to the Field Today. Pharmacol. Res. Perspect. 2023, 11, e01109.
  18. Cadoná, F.C.; Dantas, R.F.; de Mello, G.H.; Silva, F.P., Jr. Natural Products Targeting into Cancer Hallmarks: An Update on Caffeine, Theobromine, and (+)-Catechin. Crit. Rev. Food Sci. Nutr. 2022, 62, 7222–7241.
  19. Priya, S.; Satheeshkumar, P. Natural Products from Plants: Recent Developments in Phytochemicals, Phytopharmaceuticals, and Plant-Based Neutraceuticals as Anticancer Agents. Funct. Preserv. Prop. Phytochem. 2020, 145–163.
  20. Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803.
  21. Rudzińska, A.; Juchaniuk, P.; Oberda, J.; Wiśniewska, J.; Wojdan, W.; Szklener, K.; Mańdziuk, S. Phytochemicals in Cancer Treatment and Cancer Prevention—Review on Epidemiological Data and Clinical Trials. Nutrients 2023, 15, 1896.
  22. Bernitsa, S.; Dayan, R.; Stephanou, A.; Tzvetanova, I.D.; Patrikios, I.S. Natural Biomolecules and Derivatives as Anticancer Immunomodulatory Agents. Front. Immunol. 2023, 13, 1070367.
  23. Jang, J.-H.; Lee, T.-J. Mechanisms of Phytochemicals in Anti-Inflammatory and Anti-Cancer. Int. J. Mol. Sci. 2023, 24, 7863.
  24. Majrashi, T.A.; Alshehri, S.A.; Alsayari, A.; Muhsinah, A.B.; Alrouji, M.; Alshahrani, A.M.; Shamsi, A.; Atiya, A. Insight into the Biological Roles and Mechanisms of Phytochemicals in Different Types of Cancer: Targeting Cancer Therapeutics. Nutrients 2023, 15, 1704.
  25. Sohel, M.; Aktar, S.; Biswas, P.; Amin, M.A.; Hossain, M.A.; Ahmed, N.; Mim, M.I.H.; Islam, F.; Mamun, A.A. Exploring the Anti-cancer Potential of Dietary Phytochemicals for the Patients with Breast Cancer: A Comprehensive Review. Cancer Med. 2023, 12, 14556–14583.
  26. Dogra, A.; Kumar, J. Biosynthesis of Anticancer Phytochemical Compounds and Their Chemistry. Front. Pharmacol. 2023, 14, 1136779.
  27. Gahtori, R.; Tripathi, A.H.; Kumari, A.; Negi, N.; Paliwal, A.; Tripathi, P.; Joshi, P.; Rai, R.C.; Upadhyay, S.K. Anticancer Plant-Derivatives: Deciphering Their Oncopreventive and Therapeutic Potential in Molecular Terms. Future J. Pharm. Sci. 2023, 9, 14.
  28. Liang, Z.; Xu, Y.; Zhang, Y.; Zhang, X.; Song, J.; Jin, J.; Qian, H. Anticancer Applications of Phytochemicals in Gastric Cancer: Effects and Molecular Mechanism. Front. Pharmacol. 2023, 13, 1078090.
  29. Mandal, M.K.; Mohammad, M.; Parvin, S.I.; Islam, M.M.; Gazi, H.A.R. A Short Review on Anticancer Phytochemicals. Pharmacogn. Rev. 2023, 17, 11–23.
  30. Curti, V.; Di Lorenzo, A.; Dacrema, M.; Xiao, J.; Nabavi, S.M.; Daglia, M. In Vitro Polyphenol Effects on Apoptosis: An Update of Literature Data. Semin. Cancer Biol. 2017, 46, 119–131.
  31. Cook, M.T.; Mafuvadze, B.; Besch-Williford, C.; Ellersieck, M.R.; Goyette, S.; Hyder, S.M. Luteolin Suppresses Development of Medroxyprogesterone Acetate-Accelerated 7,12-Dimethylbenz(a)Anthracene-Induced Mammary Tumors in Sprague-Dawley Rats. Oncol. Rep. 2016, 35, 825–832.
  32. Briguglio, G.; Costa, C.; Pollicino, M.; Giambò, F.; Catania, S.; Fenga, C. Polyphenols in Cancer Prevention: New Insights (Review). Int. J. Funct. Nutr. 2020, 1, 9.
  33. Ali, M.; Benfante, V.; Stefano, A.; Yezzi, A.; Di Raimondo, D.; Tuttolomondo, A.; Comelli, A. Anti-Arthritic and Anti-Cancer Activities of Polyphenols: A Review of the Most Recent In Vitro Assays. Life 2023, 13, 361.
  34. Jurczyk, M.; Kasperczyk, J.; Wrześniok, D.; Beberok, A.; Jelonek, K. Nanoparticles Loaded with Docetaxel and Resveratrol as an Advanced Tool for Cancer Therapy. Biomedicines 2022, 10, 1187.
  35. Ren, B.; Kwah, M.X.-Y.; Liu, C.; Ma, Z.; Shanmugam, M.K.; Ding, L.; Xiang, X.; Ho, P.C.-L.; Wang, L.; Ong, P.S.; et al. Resveratrol for Cancer Therapy: Challenges and Future Perspectives. Cancer Lett. 2021, 515, 63–72.
  36. Walle, T. Bioavailability of Resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 9–15.
  37. Rauf, A.; Imran, M.; Butt, M.S.; Nadeem, M.; Peters, D.G.; Mubarak, M.S. Resveratrol as an Anti-Cancer Agent: A Review. Crit. Rev. Food Sci. Nutr. 2018, 58, 1428–1447.
  38. Bozorgi, A.; Haghighi, Z.; Khazaei, M.R.; Bozorgi, M.; Khazaei, M. The Anti-Cancer Effect of Chitosan/Resveratrol Polymeric Nanocomplex against Triple-Negative Breast Cancer; an in Vitro Assessment. IET Nanobiotechnol. 2023, 17, 91–102.
  39. Muller, A.G.; Sarker, S.D.; Fatokun, A.A.; Hutcheon, G.A. Formulation of Resveratrol into PGA-Co-PDL Nanoparticles Increases Its Cytotoxic Potency against Lung Cancer Cells. RPS Pharm. Pharmacol. Rep. 2023, 2, rqac007.
  40. Gagliardi, A.; Giuliano, E.; Venkateswararao, E.; Fresta, M.; Bulotta, S.; Awasthi, V.; Cosco, D. Biodegradable Polymeric Nanoparticles for Drug Delivery to Solid Tumors. Front. Pharmacol. 2021, 12, 601626.
  41. Neves, A.R.; Queiroz, J.F.; Reis, S. Brain-Targeted Delivery of Resveratrol Using Solid Lipid Nanoparticles Functionalized with Apolipoprotein E. J. Nanobiotechnol. 2016, 14, 27.
  42. Mohan, A.; Narayanan, S.; Sethuraman, S.; Krishnan, U.M. Novel Resveratrol and 5-Fluorouracil Coencapsulated in PEGylated Nanoliposomes Improve Chemotherapeutic Efficacy of Combination against Head and Neck Squamous Cell Carcinoma. BioMed Res. Int. 2014, 2014, 424239.
  43. Khatun, M.; Choudhury, S.; Liu, B.; Lemmens, P.; Pal, S.K.; Mazumder, S. Resveratrol–ZnO Nanohybrid Enhanced Anti-Cancerous Effect in Ovarian Cancer Cells through ROS. RSC Adv. 2016, 6, 105607–105617.
  44. Aras, A.; Khokhar, A.R.; Qureshi, M.Z.; Silva, M.F.; Sobczak-Kupiec, A.; Pineda, E.A.G.; Hechenleitner, A.A.W.; Farooqi, A.A. Targeting Cancer with Nano-Bullets: Curcumin, EGCG, Resveratrol and Quercetin on Flying Carpets. Asian Pac. J. Cancer Prev. 2014, 15, 3865–3871.
  45. Khan, I.; Saeed, K.; Khan, I. Nanoparticles: Properties, Applications and Toxicities. Arab. J. Chem. 2019, 12, 908–931.
  46. Mekuye, B.; Abera, B. Nanomaterials: An Overview of Synthesis, Classification, Characterization, and Applications. Nano Sel. 2023, 4, 486–501.
  47. Singh, V.; Yadav, S.S.; Chauhan, V.; Shukla, S.; Vishnolia, K.K. Applications of Nanoparticles in Various Fields: In Advances in Medical Technologies and Clinical Practice; Yadav, D., Bansal, A., Bhatia, M., Hooda, M., Morato, J., Eds.; IGI Global: Hershey, PA, USA, 2021; pp. 221–236. ISBN 978-1-79986-527-8.
  48. Kumari, S.; Raturi, S.; Kulshrestha, S.; Chauhan, K.; Dhingra, S.; András, K.; Thu, K.; Khargotra, R.; Singh, T. A Comprehensive Review on Various Techniques Used for Synthesizing Nanoparticles. J. Mater. Res. Technol. 2023, 27, 1739–1763.
  49. Park, E.-J.; Pezzuto, J.M. The Pharmacology of Resveratrol in Animals and Humans. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2015, 1852, 1071–1113.
  50. Tellone, E.; Galtieri, A.; Russo, A.; Giardina, B.; Ficarra, S. Resveratrol: A Focus on Several Neurodegenerative Diseases. Oxidative Med. Cell. Longev. 2015, 2015, 392169.
  51. Morales, M.; Barcelo, A.; Pedreño, M. Plant Stilbenes: Recent Advances in Their Chemistry and Biology. Adv. Plant Physiol. 2000, 3, 39–70.
  52. Bostanghadiri, N.; Pormohammad, A.; Chirani, A.S.; Pouriran, R.; Erfanimanesh, S.; Hashemi, A. Comprehensive Review on the Antimicrobial Potency of the Plant Polyphenol Resveratrol. Biomed. Pharmacother. 2017, 95, 1588–1595.
  53. Salehi, B.; Mishra, A.P.; Nigam, M.; Sener, B.; Kilic, M.; Sharifi-Rad, M.; Fokou, P.V.; Martins, N.; Sharifi-Rad, J. Resveratrol: A Double-Edged Sword in Health Benefits. Biomedicines 2018, 6, 91.
  54. Jojima, K.; Tanaka, A.; Node, K. Resveratrol Supplementation: A Therapeutic Potential for Cardiac Remodeling in Hypertensive Heart Disease. Hypertens. Res. 2023, 46, 1596–1598.
  55. Chen, X.; Song, X.; Zhao, X.; Zhang, Y.; Wang, Y.; Jia, R.; Zou, Y.; Li, L.; Yin, Z. Insights into the Anti-Inflammatory and Antiviral Mechanisms of Resveratrol. Mediat. Inflamm. 2022, 2022, 7138756.
  56. Andrade, S.; Ramalho, M.J.; Pereira, M.D.C.; Loureiro, J.A. Resveratrol Brain Delivery for Neurological Disorders Prevention and Treatment. Front. Pharmacol. 2018, 9, 1261.
  57. Clouser, C.L.; Chauhan, J.; Bess, M.A.; Van Oploo, J.L.; Zhou, D.; Dimick-Gray, S.; Mansky, L.M.; Patterson, S.E. Anti-HIV-1 Activity of Resveratrol Derivatives and Synergistic Inhibition of HIV-1 by the Combination of Resveratrol and Decitabine. Bioorganic Med. Chem. Lett. 2012, 22, 6642–6646.
  58. Abba, Y.; Hassim, H.; Hamzah, H.; Noordin, M.M. Antiviral Activity of Resveratrol against Human and Animal Viruses. Adv. Virol. 2015, 2015, 184241.
  59. Heredia, A.; Davis, C.; Redfield, R. Synergistic Inhibition of HIV-1 in Activated and Resting Peripheral Blood Mononuclear Cells, Monocyte-Derived Macrophages, and Selected Drug-Resistant Isolates with Nucleoside Analogues Combined with a Natural Product, Resveratrol. JAIDS J. Acquir. Immune Defic. Syndr. 2000, 25, 246–255.
  60. Jang, M.; Cai, L.; Udeani, G.O.; Slowing, K.V.; Thomas, C.F.; Beecher, C.W.; Fong, H.H.; Farnsworth, N.R.; Kinghorn, A.D.; Mehta, R.G. Cancer Chemopreventive Activity of Resveratrol, a Natural Product Derived from Grapes. Science 1997, 275, 218–220.
  61. Robinson, K.; Mock, C.; Liang, D. Pre-Formulation Studies of Resveratrol. Drug Dev. Ind. Pharm. 2015, 41, 1464–1469.
  62. Amri, A.; Chaumeil, J.; Sfar, S.; Charrueau, C. Administration of Resveratrol: What Formulation Solutions to Bioavailability Limitations? J. Control. Release 2012, 158, 182–193.
  63. Perrone, D.; Fuggetta, M.P.; Ardito, F.; Cottarelli, A.; De Filippis, A.; Ravagnan, G.; De Maria, S.; Lo Muzio, L. Resveratrol (3,5,4′-Trihydroxystilbene) and Its Properties in Oral Diseases. Exp. Ther. Med. 2017, 14, 3–9.
  64. Pettit, G.R.; Grealish, M.P.; Jung, M.K.; Hamel, E.; Pettit, R.K.; Chapuis, J.-C.; Schmidt, J.M. Antineoplastic Agents. 465. Structural Modification of Resveratrol: Sodium Resverastatin Phosphate. J. Med. Chem. 2002, 45, 2534–2542.
  65. Navarro-Orcajada, S.; Conesa, I.; Vidal-Sánchez, F.J.; Matencio, A.; Albaladejo-Maricó, L.; García-Carmona, F.; López-Nicolás, J.M. Stilbenes: Characterization, Bioactivity, Encapsulation and Structural Modifications. A Review of Their Current Limitations and Promising Approaches. Crit. Rev. Food Sci. Nutr. 2023, 63, 7269–7287.
  66. Jeon, D.; Jo, M.; Lee, Y.; Park, S.-H.; Phan, H.T.L.; Nam, J.H.; Namkung, W. Inhibition of ANO1 by Cis-and Trans-Resveratrol and Their Anticancer Activity in Human Prostate Cancer PC-3 Cells. Int. J. Mol. Sci. 2023, 24, 1186.
  67. Santos, A.C.; Pereira, I.; Magalhães, M.; Pereira-Silva, M.; Caldas, M.; Ferreira, L.; Figueiras, A.; Ribeiro, A.J.; Veiga, F. Targeting Cancer Via Resveratrol-Loaded Nanoparticles Administration: Focusing on In Vivo Evidence. AAPS J. 2019, 21, 57.
  68. Dariya, B.; Girish, B.P.; Merchant, N.; Srilatha, M.; Nagaraju, G.P. Resveratrol: Biology, Metabolism, and Detrimental Role on the Tumor Microenvironment of Colorectal Cancer. Nutr. Rev. 2023, nuad133.
  69. Santos, A.C.; Veiga, F.; Ribeiro, A.J. New Delivery Systems to Improve the Bioavailability of Resveratrol. Expert Opin. Drug Deliv. 2011, 8, 973–990.
  70. Plauth, A.; Geikowski, A.; Cichon, S.; Wowro, S.J.; Liedgens, L.; Rousseau, M.; Weidner, C.; Fuhr, L.; Kliem, M.; Jenkins, G.; et al. Hormetic Shifting of Redox Environment by Pro-Oxidative Resveratrol Protects Cells against Stress. Free Radic. Biol. Med. 2016, 99, 608–622.
  71. Singh, G.; Pai, R.S. Optimized PLGA Nanoparticle Platform for Orally Dosed Trans-Resveratrol with Enhanced Bioavailability Potential. Expert Opin. Drug Deliv. 2014, 11, 647–659.
  72. Albuquerque, B.; Costa, M.S.; Peça, I.N.; Cardoso, M.M. Production of Double-walled Nanoparticles Containing Meloxicam. Polym. Eng. Sci. 2013, 53, 146–152.
  73. Wenzel, E.; Somoza, V. Metabolism and Bioavailability of Trans-resveratrol. Mol. Nutr. Food Res. 2005, 49, 472–481.
  74. Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E.; Walle, U.K. High Absorption but Very Low Bioavailability of Oral Resveratrol in Humans. Drug Metab. Dispos. 2004, 32, 1377–1382.
  75. Almeida, T.C.; Melo, A.S.; Lima, A.P.B.; Branquinho, R.T.; da Silva, G.N. Resveratrol Induces the Production of Reactive Oxygen Species, Interferes with the Cell Cycle, and Inhibits the Cell Migration of Bladder Tumour Cells with Different TP53 Status. Nat. Prod. Res. 2023, 37, 3838–3843.
  76. Yousef, M.; Vlachogiannis, I.; Tsiani, E. Effects of Resveratrol against Lung Cancer: In Vitro and In Vivo Studies. Nutrients 2017, 9, 1231.
  77. Bishayee, A.; Petit, D.M.; Samtani, K. Angioprevention Is Implicated in Resveratrol Chemoprevention of Experimental Hepatocarcinogenesis. J Carcinog. Mutagen. 2010, 1, 102.
  78. Bhattacharjee, J.; Kirby, M.; Softic, S.; Miles, L.; Salazar-Gonzalez, R.-M.; Shivakumar, P.; Kohli, R. Hepatic Natural Killer T-Cell and CD8+ T-Cell Signatures in Mice with Nonalcoholic Steatohepatitis: Bhattacharjee, Kirby; et al. Hepatol. Commun. 2017, 1, 299–310.
  79. Arablou, T.; Aryaeian, N.; Khodaverdi, S.; Kolahdouz-Mohammadi, R.; Moradi, Z.; Rashidi, N.; Delbandi, A.-A. The Effects of Resveratrol on the Expression of VEGF, TGF-β, and MMP-9 in Endometrial Stromal Cells of Women with Endometriosis. Sci. Rep. 2021, 11, 6054.
  80. Pradhan, R.; Paul, S.; Das, B.; Sinha, S.; Dash, S.R.; Mandal, M.; Kundu, C.N. Resveratrol Nanoparticle Attenuates Metastasis and Angiogenesis by Deregulating Inflammatory Cytokines through Inhibition of CAFs in Oral Cancer by CXCL-12/IL-6-Dependent Pathway. J. Nutr. Biochem. 2023, 113, 109257.
  81. Gołąbek-Grenda, A.; Kaczmarek, M.; Juzwa, W.; Olejnik, A. Natural Resveratrol Analogs Differentially Target Endometriotic Cells into Apoptosis Pathways. Sci. Rep. 2023, 13, 11468.
  82. Khayat, M.T.; Zarka, M.A.; El-Telbany, D.; Farag, A.; El-Halawany, A.M.; Kutbi, H.I.; Elkhatib, W.F.; Noreddin, A.M.; Khayyat, A.N.; El-Telbany, R.F.A.; et al. Intensification of Resveratrol Cytotoxicity, pro-Apoptosis, Oxidant Potentials in Human Colorectal Carcinoma HCT-116 Cells Using Zein Nanoparticles. Sci. Rep. 2022, 12, 15235.
  83. Singh, S.K.; Banerjee, S.; Acosta, E.P.; Lillard, J.W.; Singh, R. Resveratrol Induces Cell Cycle Arrest and Apoptosis with Docetaxel in Prostate Cancer Cells via a P53/p21WAF1/CIP1 and p27KIP1 Pathway. Oncotarget 2017, 8, 17216–17228.
  84. Catania, A.; Barrajón-Catalán, E.; Nicolosi, S.; Cicirata, F.; Micol, V. Immunoliposome Encapsulation Increases Cytotoxic Activity and Selectivity of Curcumin and Resveratrol against HER2 Overexpressing Human Breast Cancer Cells. Breast Cancer Res. Treat. 2013, 141, 55–65.
  85. Varoni, E.M.; Lo Faro, A.F.; Sharifi-Rad, J.; Iriti, M. Anticancer Molecular Mechanisms of Resveratrol. Front. Nutr. 2016, 3, 8.
  86. Li, D.; Wang, G.; Jin, G.; Yao, K.; Zhao, Z.; Bie, L.; Guo, Y.; Li, N.; Deng, W.; Chen, X.; et al. Resveratrol Suppresses Colon Cancer Growth by Targeting the AKT/STAT3 Signaling Pathway. Int. J. Mol. Med. 2019, 43, 630–640.
  87. Ghafouri-Fard, S.; Bahroudi, Z.; Shoorei, H.; Hussen, B.M.; Talebi, S.F.; Baig, S.G.; Taheri, M.; Ayatollahi, S.A. Disease-Associated Regulation of Gene Expression by Resveratrol: Special Focus on the PI3K/AKT Signaling Pathway. Cancer Cell Int. 2022, 22, 298.
  88. Song, M.; Bode, A.M.; Dong, Z.; Lee, M.-H. AKT as a Therapeutic Target for Cancer. Cancer Res. 2019, 79, 1019–1031.
  89. Verzella, D.; Pescatore, A.; Capece, D.; Vecchiotti, D.; Ursini, M.V.; Franzoso, G.; Alesse, E.; Zazzeroni, F. Life, Death, and Autophagy in Cancer: NF-κB Turns up Everywhere. Cell Death Dis. 2020, 11, 210.
  90. Lin, H.-Y.; Tang, H.-Y.; Davis, F.B.; Davis, P.J. Resveratrol and Apoptosis: Resveratrol-Induced Apoptosis. Ann. N. Y. Acad. Sci. 2011, 1215, 79–88.
  91. Chin, Y.-T.; Hsieh, M.-T.; Yang, S.-H.; Tsai, P.-W.; Wang, S.-H.; Wang, C.-C.; Lee, Y.-S.; Cheng, G.-Y.; HuangFu, W.-C.; London, D.; et al. Anti-Proliferative and Gene Expression Actions of Resveratrol in Breast Cancer Cells in Vitro. Oncotarget 2014, 5, 12891–12907.
  92. Tang, H.-Y.; Shih, A.; Cao, H.J.; Davis, F.B.; Davis, P.J.; Lin, H.-Y. Resveratrol-Induced Cyclooxygenase-2 Facilitates P53-Dependent Apoptosis in Human Breast Cancer Cells. Mol. Cancer Ther. 2006, 5, 2034–2042.
  93. Ko, J.-H.; Sethi, G.; Um, J.-Y.; Shanmugam, M.K.; Arfuso, F.; Kumar, A.P.; Bishayee, A.; Ahn, K.S. The Role of Resveratrol in Cancer Therapy. Int. J. Mol. Sci. 2017, 18, 2589.
  94. Yang, X.; Jiang, T.; Wang, Y.; Guo, L. The Role and Mechanism of SIRT1 in Resveratrol-Regulated Osteoblast Autophagy in Osteoporosis Rats. Sci. Rep. 2019, 9, 18424.
  95. Berman, A.Y.; Motechin, R.A.; Wiesenfeld, M.Y.; Holz, M.K. The Therapeutic Potential of Resveratrol: A Review of Clinical Trials. npj Precis. Oncol. 2017, 1, 35.
  96. Narayanan, N.K.; Nargi, D.; Randolph, C.; Narayanan, B.A. Liposome Encapsulation of Curcumin and Resveratrol in Combination Reduces Prostate Cancer Incidence in PTEN Knockout Mice. Int. J. Cancer 2009, 125, 1–8.
  97. Annaji, M.; Poudel, I.; Boddu, S.H.S.; Arnold, R.D.; Tiwari, A.K.; Babu, R.J. Resveratrol-loaded Nanomedicines for Cancer Applications. Cancer Rep. 2021, 4, e1353.
  98. Sarvari, P.; Sarvari, P. Advances in Nanoparticle-Based Drug Delivery in Cancer Treatment. Glob. Transl. Med. 2023, 2, 0394.
  99. Chenthamara, D.; Subramaniam, S.; Ramakrishnan, S.G.; Krishnaswamy, S.; Essa, M.M.; Lin, F.-H.; Qoronfleh, M.W. Therapeutic Efficacy of Nanoparticles and Routes of Administration. Biomater. Res. 2019, 23, 20.
  100. Sharma, A.; Madhunapantula, S.V.; Robertson, G.P. Toxicological Considerations When Creating Nanoparticle-Based Drugs and Drug Delivery Systems. Expert Opin. Drug Metab. Toxicol. 2012, 8, 47–69.
  101. Jin, C.; Wang, K.; Oppong-Gyebi, A.; Hu, J. Application of Nanotechnology in Cancer Diagnosis and Therapy—A Mini-Review. Int. J. Med. Sci. 2020, 17, 2964–2973.
  102. Martínez-Ballesta, M.; Gil-Izquierdo, Á.; García-Viguera, C.; Domínguez-Perles, R. Nanoparticles and Controlled Delivery for Bioactive Compounds: Outlining Challenges for New “Smart-Foods” for Health. Foods 2018, 7, 72.
  103. Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; Rodriguez-Torres, M.D.P.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano Based Drug Delivery Systems: Recent Developments and Future Prospects 10 Technology 1007 Nanotechnology 03 Chemical Sciences 0306 Physical Chemistry (Incl. Structural) 03 Chemical Sciences 0303 Macromolecular and Materials Chemistry 11 Medical and Health Sciences 1115 Pharmacology and Pharmaceutical Sciences 09 Engineering 0903 Biomedical Engineering Prof Ueli Aebi, Prof Peter Gehr. J. Nanobiotechnol. 2018, 16, 71.
  104. Khushnud, T.; Mousa, S.A. Potential Role of Naturally Derived Polyphenols and Their Nanotechnology Delivery in Cancer. Mol. Biotechnol. 2013, 55, 78–86.
  105. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering Precision Nanoparticles for Drug Delivery. Nat. Rev. Drug Discov. 2021, 20, 101–124.
  106. Jose, S.; Anju, S.S.; Cinu, T.A.; Aleykutty, N.A.; Thomas, S.; Souto, E.B. In Vivo Pharmacokinetics and Biodistribution of Resveratrol-Loaded Solid Lipid Nanoparticles for Brain Delivery. Int. J. Pharm. 2014, 474, 6–13.
  107. Xin, Y.; Liu, T.; Yang, C. Development of PLGA-Lipid Nanoparticles with Covalently Conjugated Indocyanine Green as a Versatile Nanoplatform for Tumor-Targeted Imaging and Drug Delivery. Int. J. Nanomed. 2016, 11, 5807–5821.
  108. Chu, K.S.; Schorzman, A.N.; Finniss, M.C.; Bowerman, C.J.; Peng, L.; Luft, J.C.; Madden, A.J.; Wang, A.Z.; Zamboni, W.C.; DeSimone, J.M. Nanoparticle Drug Loading as a Design Parameter to Improve Docetaxel Pharmacokinetics and Efficacy. Biomaterials 2013, 34, 8424–8429.
  109. Cavalcante de Freitas, P.G.; Rodrigues Arruda, B.; Araújo Mendes, M.G.; Barroso de Freitas, J.V.; da Silva, M.E.; Sampaio, T.L.; Petrilli, R.; Eloy, J.O. Resveratrol-Loaded Polymeric Nanoparticles: The Effects of D-α-Tocopheryl Polyethylene Glycol 1000 Succinate (TPGS) on Physicochemical and Biological Properties against Breast Cancer In Vitro and In Vivo. Cancers 2023, 15, 2802.
  110. Chimento, A.; De Amicis, F.; Sirianni, R.; Sinicropi, M.; Puoci, F.; Casaburi, I.; Saturnino, C.; Pezzi, V. Progress to Improve Oral Bioavailability and Beneficial Effects of Resveratrol. IJMS 2019, 20, 1381.
  111. Paiva-Santos, A.C.; Veiga, F.; Sequeira, J.; Fortuna, A.; Falcão, A.; Pereira, I.; Pattekari, P.; Ribeiro, C.; Ribeiro, A. First-Time Orally Administered Resveratrol-Loaded Layer-by-Layer Nanoparticles to Rats—A Pharmacokinetics Study. Analyst 2019, 144, 2062–2079.
  112. 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.
  113. Augustin, M.A.; Sanguansri, L.; Lockett, T. Nano- and Micro-Encapsulated Systems for Enhancing the Delivery of Resveratrol. Ann. N. Y. Acad. Sci. 2013, 1290, 107–112.
  114. Intagliata, S.; Modica, M.N.; Santagati, L.M.; Montenegro, L. Strategies to Improve Resveratrol Systemic and Topical Bioavailability: An Update. Antioxidants 2019, 8, 244.
  115. Vijayakumar, M.R.; Vajanthri, K.Y.; Balavigneswaran, C.K.; Mahto, S.K.; Mishra, N.; Muthu, M.S.; Singh, S. Pharmacokinetics, Biodistribution, in Vitro Cytotoxicity and Biocompatibility of Vitamin E TPGS Coated Trans Resveratrol Liposomes. Colloids Surf. B Biointerfaces 2016, 145, 479–491.
  116. Tian, H.; Zhang, T.; Qin, S.; Huang, Z.; Zhou, L.; Shi, J.; Nice, E.C.; Xie, N.; Huang, C.; Shen, Z. Enhancing the Therapeutic Efficacy of Nanoparticles for Cancer Treatment Using Versatile Targeted Strategies. J. Hematol. Oncol. 2022, 15, 132.
  117. AlSawaftah, N.M.; Awad, N.S.; Pitt, W.G.; Husseini, G.A. pH-Responsive Nanocarriers in Cancer Therapy. Polymers 2022, 14, 936.
  118. Amin, M.; Huang, W.; Seynhaeve, A.L.B.; Ten Hagen, T.L.M. Hyperthermia and Temperature-Sensitive Nanomaterials for Spatiotemporal Drug Delivery to Solid Tumors. Pharmaceutics 2020, 12, 1007.
  119. Hu, Q.; Katti, P.S.; Gu, Z. Enzyme-Responsive Nanomaterials for Controlled Drug Delivery. Nanoscale 2014, 6, 12273–12286.
  120. Yang, Y.; Sun, W. Recent Advances in Redox-Responsive Nanoparticles for Combined Cancer Therapy. Nanoscale Adv. 2022, 4, 3504–3516.
  121. Liu, J.F.; Jang, B.; Issadore, D.; Tsourkas, A. Use of Magnetic Fields and Nanoparticles to Trigger Drug Release and Improve Tumor Targeting. WIREs Nanomed. Nanobiotechnol. 2019, 11, e1571.
  122. Awad, N.S.; Paul, V.; AlSawaftah, N.M.; Ter Haar, G.; Allen, T.M.; Pitt, W.G.; Husseini, G.A. Ultrasound-Responsive Nanocarriers in Cancer Treatment: A Review. ACS Pharmacol. Transl. Sci. 2021, 4, 589–612.
  123. Khalid, K.; Tan, X.; Mohd Zaid, H.F.; Tao, Y.; Lye Chew, C.; Chu, D.-T.; Lam, M.K.; Ho, Y.-C.; Lim, J.W.; Chin Wei, L. Advanced in Developmental Organic and Inorganic Nanomaterial: A Review. Bioengineered 2020, 11, 328–355.
  124. Sanna, V.; Siddiqui, I.A.; 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. 2013, 10, 3871–3881.
  125. Yao, Q.; Hou, S.-X.; He, W.-L.; Feng, J.-L.; Wang, X.-C.; Fei, H.-X.; Chen, Z.-H. Study on the preparation of resveratrol chitosan nanoparticles with free amino groups on the surface. Zhongguo Zhong Yao Za Zhi 2006, 31, 205–208.
More
This entry is offline, you can click here to edit this entry!
Video Production Service