Innovative Delivery Systems Loaded with Plant Bioactive Ingredients: Formulation Approaches and Applications
Abstract
:1. Introduction
2. Challenges in the Use of Plant Bioactive Ingredients
2.1. Solubility
2.2. Bioavailability
2.3. Stability
2.4. Release
3. Organic-Based Delivery Systems
3.1. Lipid-Based Delivery Systems
3.1.1. Vesicular Systems
Liposomes
- First generation of liposomes. These are the oldest developed, conventional liposomes that consist mainly of natural phospholipids and, in some cases, cholesterol. Despite the fact that they demonstrate a series of issues, such as increased uptake by the reticuloendothelial system (RES) and physicochemical and chemical degradation [43], they are very common delivery systems, also for plant ingredients [44,45].
- Second generation of liposomes. The second generation includes more recent developments such as stealth and stimuli-responsive liposomes. Stealth liposomes are coated by polymers for the modification of size and charge. Polyethylene glycol-covered (PEGylated) liposomes improve the stability and reduce the probability of RES uptake, increasing the blood half-life of the system. Stealth liposomes with interesting properties have been developed in order to encapsulate resveratrol [46] and curcumin [47]. Stimuli-responsive liposomes are able to release their content depending on external triggering mechanisms, such as pH or temperature change, thus being more targeted than conventional ones. Resveratrol and curcumin have also been incorporated into pH-sensitive systems [48,49].
- Third generation of liposomes. These systems bear a ligand (enzyme, antibody, vitamin, etc.) that leads to targeted transportation of the incorporated molecule(s) due to affinity mechanisms. Upon careful design, this can lead to accumulation of liposomes and targeted release at the desired site [50]. Galangin-loaded liposomes have been designed to target liver tissue [51], while a curcumin liposomal system has been developed to target cancer cells [52].
- Fourth generation of liposomes (or theranostic liposomes) combine several strategies to achieve site-specific delivery and, at the same time, imaging [53]. Their main advantage is the multifunctionality—being diagnostic and therapeutic agents at the same time. For the moment in what concerns plant extracts, the literature is very limited. A good case study is the one by Wang et al. [54], who developed a magnetic targeting liposomal nanocarrier, loaded with resveratrol, that with the aid of an external magnetic field can cross the blood–brain barrier and could prove helpful for the treatment of cerebral disease.
Transfersomes, Ethosomes, Phytosomes and Niosomes
3.1.2. Νon-Vesicular Systems
Solid Lipid Nanoparticles
Nanostructured Lipid Carriers
3.2. Protein-Based Delivery Systems
3.3. Carbohydrate-Based Delivery Systems
3.4. Polymeric Systems
3.4.1. Polymer-Based Nanoparticles
3.4.2. Micelles
3.4.3. Dendrimers
3.4.4. Polymeric Nanoparticles and Nanogels
3.4.5. Nanocapsules
3.4.6. Nanospheres
3.4.7. Nanofibers
3.4.8. Polymersomes
3.5. Nanoemulsions
4. Inorganic-Based Delivery Systems
5. Other Delivery Approaches
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- McClements, D.J. Nanoparticle- and Microparticle-Based Delivery Systems: Encapsulation, Protection and Release of Active Compounds; CRC Press: Boca Raton, FL, USA, 2014; ISBN 9781482233162. [Google Scholar]
- Bao, C.; Jiang, P.; Chai, J.; Jiang, Y.; Li, D.; Bao, W.; Liu, B.; Liu, B.; Norde, W.; Li, Y. The delivery of sensitive food bioactive ingredients: Absorption mechanisms, influencing factors, encapsulation techniques and evaluation models. Food Res. Int. 2019, 120, 130–140. [Google Scholar] [CrossRef]
- Rahman, H.S.; Othman, H.H.; Hammadi, N.I.; Yeap, S.K.; Amin, K.M.; Samad, N.A.; Alitheen, N.B. Novel drug delivery systems for loading of natural plant extracts and their biomedical applications. Int. J. Nanomed. 2020, 15, 2439–2483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zuidam, N.J.; Nedovic, V.A. Encapsulation Technologies for Active Food Ingredients and Food Processing; Springer: New York, NY, USA, 2010; ISBN 9781441910073. [Google Scholar]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McClements, D.J. Advances in nanoparticle and microparticle delivery systems for increasing the dispersibility, stability, and bioactivity of phytochemicals. Biotechnol. Adv. 2020, 38, 107287. [Google Scholar] [CrossRef]
- Ahmad, R.; Srivastava, S.; Ghosh, S.; Khare, S.K. Phytochemical delivery through nanocarriers: A review. Coll. Surf. B Biointerfaces 2021, 197, 111389. [Google Scholar] [CrossRef] [PubMed]
- Qian, C.; Decker, E.A.; Xiao, H.; McClements, D.J. Nanoemulsion delivery systems: Influence of carrier oil on β-carotene bioaccessibility. Food Chem. 2012, 135, 1440–1447. [Google Scholar] [CrossRef] [PubMed]
- Zou, L.Q.; Liu, W.; Liu, W.L.; Liang, R.H.; Li, T.; Liu, C.M.; Cao, Y.L.; Niu, J.; Liu, Z. Characterization and bioavailability of tea polyphenol nanoliposome prepared by combining an ethanol injection method with dynamic high-pressure microfluidization. J. Agric. Food Chem. 2014, 62, 934–941. [Google Scholar] [CrossRef]
- Dang, S.; Gupta, S.; Bansal, R.; Ali, J.; Gabrani, R. Nano-encapsulation of a natural polyphenol, green tea catechins: Way to preserve. In Free Radicals in Human Health and Disease; Rani, V., Yadav, U., Eds.; Springer: Chennai, India, 2015; pp. 397–415. ISBN 978-81-322-2035-0. [Google Scholar]
- Isacchi, Β.; Galeotti, N.; Bergonzi, M.C.; Ghelardini, C.; Bilia, A.R.; Vincieri, F.F. Pharmacological in vivo test to evaluate the bioavailability of some St John’s Wort innovative oral preparations. Phyther. Res. 2009, 23, 197–205. [Google Scholar] [CrossRef]
- Bilia, A.R.; Piazzini, V.; Asprea, M.; Risaliti, L.; Vanti, G.; Bergonzi, M.C. Plants extracts loaded in nanocarriers: An emergent formulating approach. Nat. Prod. Commun. 2018, 13, 1157–1160. [Google Scholar] [CrossRef] [Green Version]
- Gunasekaran, T.; Haile, T.; Nigusse, T.; Dhanaraju, M.D. Nanotechnology: An effective tool for enhancing bioavailability and bioactivity of phytomedicine. Asian Pac. J. Trop. Biomed. 2014, 4, S1–S7. [Google Scholar] [CrossRef] [Green Version]
- Li, Z.; Yao, L.; Li, J.; Zhang, W.; Wu, X.; Liu, Y.; Lin, M.; Su, W.; Li, Y.; Liang, D. Celastrol nanoparticles inhibit corneal neovascularization induced by suturing in rats. Int. J. Nanomed. 2012, 7, 1163–1173. [Google Scholar] [CrossRef] [Green Version]
- Deepa, V.; Sridhar, R.; Goparaju, A.; Neelakanta Reddy, P.; Balakrishna Murthy, P. Nanoemulsified ethanolic extract of Pyllanthus amarus Schum & Thonn ameliorates CCl4 induced hepatotoxicity in Wistar rats. Indian J. Exp. Biol. 2012, 50, 785–794. [Google Scholar] [PubMed]
- Yen, C.C.; Chen, Y.C.; Wu, M.T.; Wang, C.C.; Wu, Y.T. Nanoemulsion as a strategy for improving the oral bioavailability and anti-inflammatory activity of andrographolide. Int. J. Nanomed. 2018, 13, 669–680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yuan, Y.; Gao, Y.; Zhao, J.; Mao, L. Characterization and stability evaluation of β-carotene nanoemulsions prepared by high pressure homogenization under various emulsifying conditions. Food Res. Int. 2008, 41, 61–68. [Google Scholar] [CrossRef]
- Nair, A.; Haponiuk, J.T.; Thomas, S.; Gopi, S. Natural carbon-based quantum dots and their applications in drug delivery: A review. Biomed. Pharmacother. 2020, 132, 110834. [Google Scholar] [CrossRef]
- Serhan, M.; Sprowls, M.; Jackemeyer, D.; Long, M.; Perez, I.D.; Maret, W.; Tao, N.; Forzani, E. Total iron measurement in human serum with a smartphone. In Proceedings of the 2019 AIChE Annual Meeting, Orlando, FL, USA, 10–15 November 2019. [Google Scholar]
- Thakur, L.; Ghodasra, U.; Patel, N.; Dabhi, M. Novel approaches for stability improvement in natural medicines. Pharmacogn. Rev. 2011, 5, 48–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saraf, S. Applications of novel drug delivery system for herbal formulations. Fitoterapia 2010, 81, 680–689. [Google Scholar] [CrossRef]
- Isacchi, B.; Bergonzi, M.C.; Iacopi, R.; Ghelardini, C.; Galeotti, N.; Bilia, A.R. Liposomal formulation to increase stability and prolong antineuropathic activity of verbascoside. Planta Med. 2017, 83, 412–419. [Google Scholar] [CrossRef] [PubMed]
- Devi, V.; Jain, N.; Valli, K. Importance of novel drug delivery systems in herbal medicines. Pharmacogn. Rev. 2010, 4, 27–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Natarajan, J.V.; Nugraha, C.; Ng, X.W.; Venkatraman, S. Sustained-release from nanocarriers: A review. J. Control. Release 2014, 193, 122–138. [Google Scholar] [CrossRef]
- Lu, C.; Lu, Y.; Chen, J.; Zhang, W.; Wu, W. Synchronized and sustained release of multiple components in silymarin from erodible glyceryl monostearate matrix system. Eur. J. Pharm. Biopharm. 2007, 66, 210–219. [Google Scholar] [CrossRef] [PubMed]
- Hasan, M.; Elkhoury, K.; Kahn, C.J.F.; Arab-Tehrany, E.; Linder, M. Preparation, characterization, and release kinetics of chitosan-coated nanoliposomes encapsulating curcumin in simulated environments. Molecules 2019, 24, 2023. [Google Scholar] [CrossRef] [Green Version]
- Ba, K.; Wei, X.Q. Construction a long-circulating delivery system of liposomal curcumin by coating albumin. ACS Omega 2020, 5, 16502–16509. [Google Scholar] [CrossRef]
- Md Saari, N.H.; Chua, L.S.; Hasham, R.; Yuliati, L. Curcumin-loaded nanoemulsion for better cellular permeation. Sci. Pharm. 2020, 88, 44. [Google Scholar] [CrossRef]
- Salem, H.F.; Kharshoum, R.M.; Abou-Taleb, H.A.; Naguib, D.M. Nanosized nasal emulgel of resveratrol: Preparation, optimization, in vitro evaluation and in vivo pharmacokinetic study. Drug Dev. Ind. Pharm. 2019, 45, 1624–1634. [Google Scholar] [CrossRef]
- Perinelli, D.R.; Palmieri, G.F.; Cespi, M.; Bonacucina, G. Encapsulation of flavours and fragrances into polymeric capsules and cyclodextrins inclusion complexes: An update. Molecules 2020, 25, 5878. [Google Scholar] [CrossRef]
- Tramón, C. Modeling the controlled release of essential oils from a polymer matrix-A special case. Ind. Crops Prod. 2014, 61, 23–30. [Google Scholar] [CrossRef]
- Dadwal, V.; Bhatt, S.; Joshi, R.; Gupta, M. Development and characterization of controlled released polyphenol rich micro-encapsulate of Murraya koenigii bark extract. J. Food Process. Preserv. 2020, 44, 1–12. [Google Scholar] [CrossRef]
- Shukla, T.; Upmanyu, N.; Prakash Pandey, S.; Gosh, D. Lipid Nanocarriers; Elsevier Inc.: Amsterdam, The Netherlands, 2018; ISBN 9780128136874. [Google Scholar]
- Chen, C.C.; Tsai, T.H.; Huang, Z.R.; Fang, J.Y. Effects of lipophilic emulsifiers on the oral administration of lovastatin from nanostructured lipid carriers: Physicochemical characterization and pharmacokinetics. Eur. J. Pharm. Biopharm. 2010, 74, 474–482. [Google Scholar] [CrossRef] [PubMed]
- Leitgeb, M.; Knez, Z.; Primozic, M. Sustainable technologies for liposome preparation. J. Supercrit. Fluids 2020, 165, 104984. [Google Scholar] [CrossRef]
- Pathak, K.; Keshri, L.; Shah, M. Lipid nanocarriers: Influence of lipids on product development and pharmacokinetics. Crit. Rev. Ther. Drug Carrier Syst. 2011, 28, 357–393. [Google Scholar] [CrossRef]
- Anu Bhushani, J.; Anandharamakrishnan, C. Electrospinning and electrospraying techniques: Potential food based applications. Trends Food Sci. Technol. 2014, 38, 21–33. [Google Scholar] [CrossRef]
- Jain, S.; Jain, V.; Mahajan, S.C. Lipid based vesicular drug delivery systems. Adv. Pharm. 2014, 2014, 574673. [Google Scholar] [CrossRef] [Green Version]
- Biju, S.S.; Talegaonkaρ, S.; Mishra, P.R.; Kharshoum, R.M. Vesicular systems: An overview. Indian J. Pharm. Sci. 2006, 68, 141–153. [Google Scholar] [CrossRef] [Green Version]
- Has, C.; Sunthar, P. A comprehensive review on recent preparation techniques of liposomes. J. Liposome Res. 2020, 30, 336–365. [Google Scholar] [CrossRef] [PubMed]
- Akbarzadeh, A.; Rezaei-sadabady, R.; Davaran, S.; Joo, S.W.; Zarghami, N. Liposome: Classification, preparation, and applications. Nanoscale Res. Lett. 2013, 8, 102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Islam Shishir, M.R.; Karim, N.; Gowd, V.; Zheng, X.; Chen, W. Liposomal delivery of natural product: A promising approach in health research. Trends Food Sci. Technol. 2019, 85, 177–200. [Google Scholar] [CrossRef]
- Sharma, D.; Ali, A.A.E.; Trivedi, L.R. An updated review on: Liposomes as drug delivery system. Pharmatutor 2018, 6, 50. [Google Scholar] [CrossRef]
- Olusanya, T.O.B.; Ahmad, R.R.H.; Ibegbu, D.M.; Smith, J.R.; Elkordy, A.A. Liposomal drug delivery systems and anticancer drugs. Molecules 2018, 23, 907. [Google Scholar] [CrossRef] [Green Version]
- Guldiken, B.; Gibis, M.; Boyacioglu, D.; Capanoglu, E.; Weiss, J. Physical and chemical stability of anthocyanin-rich black carrot extract-loaded liposomes during storage. Food Res. Int. 2018, 108, 491–497. [Google Scholar] [CrossRef] [PubMed]
- Ethemoglu, M.S.; Seker, F.B.; Akkaya, H.; Kilic, E.; Aslan, I.; Erdogan, C.S.; Yilmaz, B. Anticonvulsant activity of resveratrol-loaded liposomes in vivo. Neuroscience 2017, 357, 12–19. [Google Scholar] [CrossRef]
- Caddeo, C.; Pucci, L.; Gabriele, M.; Carbone, C.; Fernàndez-Busquets, X.; Valenti, D.; Pons, R.; Vassallo, A.; Fadda, A.M.; Manconi, M. Stability, biocompatibility and antioxidant activity of PEG-modified liposomes containing resveratrol. Int. J. Pharm. 2018, 538, 40–47. [Google Scholar] [CrossRef] [PubMed]
- Cuomo, F.; Cofelice, M.; Venditti, F.; Ceglie, A.; Miguel, M.; Lindman, B.; Lopez, F. In-vitro digestion of curcumin loaded chitosan-coated liposomes. Coll. Surf. B Biointerfaces 2018, 168, 29–34. [Google Scholar] [CrossRef]
- Caddeo, C.; Pons, R.; Carbone, C.; Fernàndez-Busquets, X.; Cardia, M.C.; Maccioni, A.M.; Fadda, A.M.; Manconi, M. Physico-chemical characterization of succinyl chitosan-stabilized liposomes for the oral co-delivery of quercetin and resveratrol. Carbohydr. Polym. 2017, 157, 1853–1861. [Google Scholar] [CrossRef]
- Seong, J.S.; Yun, M.E.; Park, S.N. Surfactant-stable and pH-sensitive liposomes coated with N-succinyl-chitosan and chitooligosaccharide for delivery of quercetin. Carbohydr. Polym. 2018, 181, 659–667. [Google Scholar] [CrossRef]
- Sercombe, L.; Veerati, T.; Moheimani, F.; Wu, S.Y.; Sood, A.K.; Hua, S. Advances and challenges of liposome assisted drug delivery. Front. Pharmacol. 2015, 6, 286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, J.; Wang, Q.; Li, H.; Zhang, H.; Zhu, Y.; Omari-Siaw, E.; Sun, C.; Wei, Q.; Deng, W.; Yu, J.; et al. Galangin-loaded, liver targeting liposomes: Optimization and hepatoprotective efficacy. J. Drug Deliv. Sci. Technol. 2018, 46, 339–347. [Google Scholar] [CrossRef]
- Kangarlou, S.; Ramezanpour, S.; Balalaie, S.; Roudbar Mohammadi, S.; Haririan, I. Curcumin-loaded nanoliposomes linked to homing peptides for integrin targeting and neuropilin-1- mediated internalization. Pharm. Biol. 2017, 55, 277–285. [Google Scholar] [CrossRef]
- Lee, W.; Im, H.J. Theranostics Based on Liposome: Looking Back and Forward. Nucl. Med. Mol. Imaging 2019, 53, 242–246. [Google Scholar] [CrossRef] [PubMed]
- Wang, M.; Li, L.; Zhang, X.; Liu, Y.; Zhu, R.; Liu, L.; Fang, Y.; Gao, Z.; Gao, D. Magnetic resveratrol liposomes as a new theranostic platform for magnetic resonance imaging guided Parkinson’s disease targeting therapy. ACS Sustain. Chem. Eng. 2018, 6, 17124–17133. [Google Scholar] [CrossRef]
- Huang, M.; Su, E.; Zheng, F.; Tan, C. Encapsulation of flavonoids in liposomal delivery systems: The case of quercetin, kaempferol and luteolin. Food Funct. 2017, 8, 3198–3208. [Google Scholar] [CrossRef] [PubMed]
- Anand David, A.V.; Arulmoli, R.; Parasuraman, S. Overviews of biological importance of quercetin: A bioactive flavonoid. Pharmacogn. Rev. 2016, 10, 84–89. [Google Scholar] [CrossRef] [Green Version]
- Bonechi, C.; Donati, A.; Tamasi, G.; Leone, G.; Consumi, M.; Rossi, C.; Lamponi, S.; Magnani, A. Protective effect of quercetin and rutin encapsulated liposomes on induced oxidative stress. Biophys. Chem. 2018, 233, 55–63. [Google Scholar] [CrossRef]
- Hao, J.; Guo, B.; Yu, S.; Zhang, W.; Zhang, D.; Wang, J.; Wang, Y. Encapsulation of the flavonoid quercetin with chitosan-coated nano-liposomes. LWT Food Sci. Technol. 2017, 85, 37–44. [Google Scholar] [CrossRef]
- Hu, J.; Wang, J.; Wang, G.; Yao, Z.; Dang, X. Pharmacokinetics and antitumor efficacy of DSPE-PEG2000 polymeric liposomes loaded with quercetin and temozolomide: Analysis of their effectiveness in enhancing the chemosensitization of drug-resistant glioma cells. Int. J. Mol. Med. 2016, 37, 690–702. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Shi, M.; Ma, B.; Niu, R.; Zhang, H.; Kun, L. Antitumor activity and safety evaluation of nanaparticle-based delivery of quercetin through intravenous administration in mice. Mater. Sci. Eng. C 2017, 77, 803–810. [Google Scholar] [CrossRef] [PubMed]
- Basnet, P.; Hussain, H.; Tho, I.; Skalko-Basnet, N. Liposomal delivery system enhances anti-inflammatory properties of curcumin. J. Pharm. Sci. 2012, 101, 598–609. [Google Scholar] [CrossRef]
- Zhao, Y.Z.; Lu, C.T.; Zhang, Y.; Xiao, J.; Zhao, Y.P.; Tian, J.L.; Xu, Y.Y.; Feng, Z.G.; Xu, C.Y. Selection of high efficient transdermal lipid vesicle for curcumin skin delivery. Int. J. Pharm. 2013, 454, 302–309. [Google Scholar] [CrossRef]
- Kianvash, N.; Bahador, A.; Pourhajibagher, M.; Ghafari, H.; Nikoui, V.; Rezayat, S.M.; Dehpour, A.R.; Partoazar, A. Evaluation of propylene glycol nanoliposomes containing curcumin on burn wound model in rat: Biocompatibility, wound healing, and anti-bacterial effects. Drug Deliv. Transl. Res. 2017, 7, 654–663. [Google Scholar] [CrossRef]
- Li, W.; Zhou, M.; Xu, N.; Hu, Y.; Wang, C.; Li, D.; Liu, L.; Li, D. Comparative analysis of protective effects of curcumin, curcumin-β-cyclodextrin nanoparticle and nanoliposomal curcumin on unsymmetrical dimethyl hydrazine poisoning in mice. Bioengineered 2016, 7, 334–341. [Google Scholar] [CrossRef]
- Chen, Y.; Wu, Q.; Zhang, Z.; Yuan, L.; Liu, X.; Zhou, L. Preparation of curcumin-loaded liposomes and evaluation of their skin permeation and pharmacodynamics. Molecules 2012, 17, 5972–5987. [Google Scholar] [CrossRef]
- Karewicz, A.; Bielska, D.; Loboda, A.; Gzyl-Malcher, B.; Bednar, J.; Jozkowicz, A.; Dulak, J.; Nowakowska, M. Curcumin-containing liposomes stabilized by thin layers of chitosan derivatives. Coll. Surf. B Biointerfaces 2013, 109, 307–316. [Google Scholar] [CrossRef]
- Apiratikul, N.; Penglong, T.; Suksen, K.; Svasti, S.; Chairoungdua, A.; Yingyongnarongkul, B. In vitro delivery of curcumin with cholesterol-based cationic liposomes. Russ. J. Bioorg. Chem. 2013, 39, 444–450. [Google Scholar] [CrossRef]
- Lin, Y.L.; Liu, Y.K.; Tsai, N.M.; Hsieh, J.H.; Chen, C.H.; Lin, C.M.; Liao, K.W. A Lipo-PEG-PEI complex for encapsulating curcumin that enhances its antitumor effects on curcumin-sensitive and curcumin-resistance cells. Nanomed. Nanotechnol. Biol. Med. 2012, 8, 318–327. [Google Scholar] [CrossRef] [PubMed]
- Rahman, S.; Cao, S.; Steadman, K.J.; Wei, M.; Parekh, H.S. Native and ß-cyclodextrin-enclosed curcumin: Entrapment within liposomes and their in vitro cytotoxicity in lung and colon cancer. Drug Deliv. 2012, 19, 346–353. [Google Scholar] [CrossRef]
- Hu, D.; Xu, Y.; Xie, J.; Sun, C.; Zheng, X.; Chen, W. Systematic evaluation of phenolic compounds and protective capacity of a new mulberry cultivar J33 against palmitic acid-induced lipotoxicity using a simulated digestion method. Food Chem. 2018, 258, 43–50. [Google Scholar] [CrossRef]
- Zhao, M.; Zhao, M.; Fu, C.; Yu, Y.; Fu, A. Targeted therapy of intracranial glioma model mice with curcumin nanoliposomes. Int. J. Nanomed. 2018, 13, 1601–1610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shaito, A.; Posadino, A.M.; Younes, N.; Hasan, H.; Halabi, S.; Alhababi, D.; Al-Mohannadi, A.; Abdel-Rahman, W.M.; Eid, A.H.; Nasrallah, G.K.; et al. Potential adverse effects of resveratrol: A literature review. Int. J. Mol. Sci. 2020, 21, 2084. [Google Scholar] [CrossRef] [Green Version]
- Jhaveri, A.; Deshpande, P.; Pattni, B.; Torchilin, V. Transferrin-targeted, resveratrol-loaded liposomes for the treatment of glioblastoma. J. Control. Release 2018, 277, 89–101. [Google Scholar] [CrossRef] [PubMed]
- Jøraholmen, M.W.; Škalko-Basnet, N.; Acharya, G.; Basnet, P. Resveratrol-loaded liposomes for topical treatment of the vaginal inflammation and infections. Eur. J. Pharm. Sci. 2015, 79, 112–121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ahamed, M.B.K.; Aisha, A.F.A.; Nassar, Z.D.; Siddiqui, J.M.; Ismail, Z.; Omari, S.M.S.; Parish, C.R.; Majid, A.M.S.A. Cat’s whiskers tea (orthosiphon stamineus) extract inhibits growth of colon tumor in nude mice and angiogenesis in endothelial cells via suppressing VEGFR phosphorylation. Nutr. Cancer 2012, 64, 89–99. [Google Scholar] [CrossRef]
- Arafat, O.M.; Tham, S.Y.; Sadikun, A.; Zhari, I.; Haughton, P.J.; Asmawi, M.Z. Studies on diuretic and hypouricemic effects of Orthosiphon stamineus methanol extracts in rats. J. Ethnopharmacol. 2008, 118, 354–360. [Google Scholar] [CrossRef]
- Alshawsh, M.A.; Abdulla, M.A.; Ismail, S.; Amin, Z.A. Hepatoprotective effects of Orthosiphon stamineus extract on thioacetamide-induced liver cirrhosis in rats. Evid. Based Complement. Altern. Med. 2011, 2011, 103039. [Google Scholar] [CrossRef] [Green Version]
- Aisha, A.F.A.; Majid, A.M.S.A.; Ismail, Z. Preparation and characterization of nano liposomes of Orthosiphon stamineus ethanolic extract in soybean phospholipids. BMC Biotechnol. 2014, 14, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Upputuri, R.T.P.; Mandal, A.K.A. Sustained release of green tea polyphenols from liposomal nanoparticles; Release kinetics and mathematical modelling. Iran. J. Biotechnol. 2017, 15, 277–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rashidinejad, A.; Birch, E.J.; Sun-Waterhouse, D.; Everett, D.W. Effect of liposomal encapsulation on the recovery and antioxidant properties of green tea catechins incorporated into a hard low-fat cheese following in vitro simulated gastrointestinal digestion. Food Bioprod. Process. 2016, 100, 238–245. [Google Scholar] [CrossRef]
- Gülseren, I.; Guri, A.; Corredig, M. Encapsulation of tea polyphenols in nanoliposomes prepared with milk phospholipids and their effect on the viability of HT-29 human carcinoma cells. Food Dig. 2012, 3, 36–45. [Google Scholar] [CrossRef]
- Zhang, J.; Han, X.; Li, X.; Luo, Y.; Zhao, H.; Yang, M.; Ni, B.; Liao, Z. Core-shell hybrid liposomal vesicles loaded with panax notoginsenoside: Preparation, characterization and protective effects on global cerebral ischemia/reperfusion injury and acute myocardial ischemia in rats. Int. J. Nanomed. 2012, 7, 4299–4310. [Google Scholar] [CrossRef] [Green Version]
- Xiong, J.; Guo, J.; Huang, L.; Meng, B.; Ping, Q. Self-micelle formation and the incorporation of lipid in the formulation affect the intestinal absorption of Panax notoginseng. Int. J. Pharm. 2008, 360, 191–196. [Google Scholar] [CrossRef]
- Gibis, M.; Zeeb, B.; Weiss, J. Formation, characterization, and stability of encapsulated hibiscus extract in multilayered liposomes. Food Hydrocoll. 2014, 38, 28–39. [Google Scholar] [CrossRef]
- Plangsombat, N.; Rungsardthong, K.; Kongkaneramit, L.; Waranuch, N.; Sarisuta, N. Anti-inflammatory activity of liposomes of Asparagus racemosus root extracts prepared by various methods. Exp. Ther. Med. 2016, 12, 2790–2796. [Google Scholar] [CrossRef] [Green Version]
- Manconi, M.; Marongiu, F.; Castangia, I.; Manca, M.L.; Caddeo, C.; Tuberoso, C.I.G.; D’hallewin, G.; Bacchetta, G.; Fadda, A.M. Polymer-associated liposomes for the oral delivery of grape pomace extract. Coll. Surf. B Biointerfaces 2016, 146, 910–917. [Google Scholar] [CrossRef] [PubMed]
- Marín, D.; Alemán, A.; Sánchez-Faure, A.; Montero, P.; Gómez-Guillén, M.C. Freeze-dried phosphatidylcholine liposomes encapsulating various antioxidant extracts from natural waste as functional ingredients in surimi gels. Food Chem. 2018, 245, 525–535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saber, F.R.; Abdelbary, G.A.; Salama, M.M.; Saleh, D.O.; Fathy, M.M.; Soliman, F.M. UPLC/QTOF/MS profiling of two Psidium species and the in-vivo hepatoprotective activity of their nano-formulated liposomes. Food Res. Int. 2018, 105, 1029–1038. [Google Scholar] [CrossRef] [PubMed]
- Pinilla, C.M.B.; Brandelli, A. Antimicrobial activity of nanoliposomes co-encapsulating nisin and garlic extract against Gram-positive and Gram-negative bacteria in milk. Innov. Food Sci. Emerg. Technol. 2016, 36, 287–293. [Google Scholar] [CrossRef]
- Khosravi-Darani, K.; Khoosfi, M.E.; Hosseini, H. Encapsulation of zataria multiflora boiss. Essential oil in liposome: Antibacterial activity against E. Coli O157:H7 in broth media and minced beef. J. Food Saf. 2016, 36, 515–523. [Google Scholar] [CrossRef]
- Gortzi, O.; Lalas, S.; Chinou, I.; Tsaknis, J. Evaluation of the antimicrobial and antioxidant activities of Origanum dictamnus extracts before and after encapsulation in liposomes. Molecules 2007, 12, 932–945. [Google Scholar] [CrossRef]
- Pinna, R.; Filigheddu, E.; Juliano, C.; Palmieri, A.; Manconi, M.; D’hallewin, G.; Petretto, G.; Maioli, M.; Caddeo, C.; Manca, M.L.; et al. Antimicrobial effect of thymus capitatus and citrus limon var. Pompia as raw extracts and nanovesicles. Pharmaceutics 2019, 11, 234. [Google Scholar] [CrossRef] [Green Version]
- Bilia, A.R.; Bergonzi, M.C.; Guccione, C.; Manconi, M.; Fadda, A.M.; Sinico, C. Vesicles and micelles: Two versatile vectors for the delivery of natural products. J. Drug Deliv. Sci. Technol. 2016, 32, 241–255. [Google Scholar] [CrossRef]
- Fernández-García, R.; Lalatsa, A.; Statts, L.; Bolás-Fernández, F.; Ballesteros, M.P.; Serrano, D.R. Transferosomes as nanocarriers for drugs across the skin: Quality by design from lab to industrial scale. Int. J. Pharm. 2020, 573, 118817. [Google Scholar] [CrossRef]
- Kumar, A. Transferosome: A recent approach for transdermal drug delivery. J. Drug Deliv. Ther. 2018, 8, 100–104. [Google Scholar] [CrossRef] [Green Version]
- Ramezani, V.; Honarvar, M.; Seyedabadi, M.; Karimollah, A.; Ranjbar, A.M.; Hashemi, M. Formulation and optimization of transfersome containing minoxidil and caffeine. J. Drug Deliv. Sci. Technol. 2018, 44, 129–135. [Google Scholar] [CrossRef]
- Jangdey, M.S.; Gupta, A.; Saraf, S.; Saraf, S. Development and optimization of apigenin-loaded transfersomal system for skin cancer delivery: In vitro evaluation. Artif. Cells Nanomed. Biotechnol. 2017, 45, 1452–1462. [Google Scholar] [CrossRef] [Green Version]
- Niu, X.Q.; Zhang, D.P.; Bian, Q.; Feng, X.F.; Li, H.; Rao, Y.F.; Shen, Y.M.; Geng, F.N.; Yuan, A.R.; Ying, X.Y.; et al. Mechanism investigation of ethosomes transdermal permeation. Int. J. Pharm. X 2019, 1, 100027. [Google Scholar] [CrossRef]
- Hallan, S.S.; Sguizzato, M.; Mariani, P.; Cortesi, R.; Huang, N.; Simelière, F.; Marchetti, N.; Drechsler, M.; Ruzgas, T.; Esposito, E. Design and characterization of ethosomes for transdermal delivery of caffeic acid. Pharmaceutics 2020, 12, 740. [Google Scholar] [CrossRef] [PubMed]
- Choi, J.H.; Cho, S.H.; Yun, J.J.; Yu, Y.B.; Cho, C.W. Ethosomes and transfersomes for topical delivery of ginsenoside Rh1 from red ginseng: Characterization and in vitro evaluation. J. Nanosci. Nanotechnol. 2015, 15, 5660–5662. [Google Scholar] [CrossRef] [PubMed]
- Lu, M.; Qiu, Q.; Luo, X.; Liu, X.; Sun, J.; Wang, C.; Lin, X.; Deng, Y.; Song, Y. Phyto-phospholipid complexes (phytosomes): A novel strategy to improve the bioavailability of active constituents. Asian J. Pharm. Sci. 2019, 14, 265–274. [Google Scholar] [CrossRef]
- Singh, R.P.; Gangadharappa, H.V.; Mruthunjaya, K. Phytosome loaded novel herbal drug delivery system: A review. Int. Res. J. Pharm. 2016, 7, 15–21. [Google Scholar] [CrossRef]
- Bhardwaj, P.; Tripathi, P.; Gupta, R.; Pandey, S. Niosomes: A review on niosomal research in the last decade. J. Drug Deliv. Sci. Technol. 2020, 56, 101581. [Google Scholar] [CrossRef]
- Gharbavi, M.; Amani, J.; Kheiri-Manjili, H.; Danafar, H.; Sharafi, A. Niosome: A Promising Nanocarrier for Natural Drug Delivery through Blood-Brain Barrier. Adv. Pharmacol. Sci. 2018, 2018. [Google Scholar] [CrossRef] [PubMed]
- Raeiszadeh, M.; Pardakhty, A.; Sharififar, F.; Mehrabani, M.; Nejatmehrab-Kermani, H.; Mehrabani, M. Phytoniosome: A novel drug delivery for myrtle extract. Iran. J. Pharm. Res. 2018, 17, 804–817. [Google Scholar] [CrossRef] [PubMed]
- Agarwal, S.; Mohamed, M.S.; Raveendran, S.; Rochani, A.K.; Maekawa, T.; Kumar, D.S. Formulation, characterization and evaluation of morusin loaded niosomes for potentiation of anticancer therapy. RSC Adv. 2018, 8, 32621–32636. [Google Scholar] [CrossRef] [Green Version]
- Barani, M.; Mirzaei, M.; Torkzadeh-Mahani, M.; Nematollahi, M.H. Lawsone-loaded Niosome and its antitumor activity in MCF-7 breast Cancer cell line: A Nano-herbal treatment for Cancer. DARU J. Pharm. Sci. 2018, 26, 11–17. [Google Scholar] [CrossRef]
- Un, R.N.; Barlas, F.B.; Yavuz, M.; Ag Seleci, D.; Seleci, M.; Gumus, Z.P.; Guler, E.; Demir, B.; Can, M.; Coskunol, H.; et al. Phyto-niosomes: In vitro assessment of the novel nanovesicles containing marigold extract. Int. J. Polym. Mater. Polym. Biomater. 2015, 64, 927–937. [Google Scholar] [CrossRef]
- Xu, Y.Q.; Chen, W.R.; Tsosie, J.K.; Xie, X.; Li, P.; Wan, J.B.; He, C.W.; Chen, M.W. Niosome encapsulation of curcumin: Characterization and cytotoxic effect on ovarian cancer cells. J. Nanomater. 2016, 2016. [Google Scholar] [CrossRef] [Green Version]
- Akbari, J.; Saeedi, M.; Enayatifard, R.; Morteza-Semnani, K.; Hassan Hashemi, S.M.; Babaei, A.; Rahimnia, S.M.; Rostamkalaei, S.S.; Nokhodchi, A. Curcumin Niosomes (curcusomes) as an alternative to conventional vehicles: A potential for efficient dermal delivery. J. Drug Deliv. Sci. Technol. 2020, 60, 102035. [Google Scholar] [CrossRef]
- Sadeghi Ghadi, Z.; Ebrahimnejad, P. Curcumin entrapped hyaluronan containing niosomes: Preparation, characterisation and in vitro/in vivo evaluation. J. Microencapsul. 2019, 36, 169–179. [Google Scholar] [CrossRef]
- García-Díaz, M.; Patiño, B.; Vázquez, C.; Gil-Serna, J. A novel niosome-encapsulated essential oil formulation to prevent aspergillus flavus growth and aflatoxin contamination of maize grains during storage. Toxins 2019, 11, 646. [Google Scholar] [CrossRef] [Green Version]
- Eid, R.K.; Essa, E.A.; El Maghraby, G.M. Essential oils in niosomes for enhanced transdermal delivery of felodipine. Pharm. Dev. Technol. 2019, 24, 157–165. [Google Scholar] [CrossRef]
- Raeiszadeh, M.; Pardakhty, A.; Sharififar, F.; Farsinejad, A.; Mehrebani, M.; Hosseini-Nave, H.; Mehrabani, M. Development, physicochemical characterization, and antimicrobial evaluation of niosomal myrtle essential oil. Res. Pharm. Sci. 2018, 13, 250–261. [Google Scholar] [CrossRef]
- Kumar, S.; Michael, I.J.; Park, J.; Granick, S.; Cho, Y.K. Cloaked exosomes: Biocompatible, durable, and degradable encapsulation. Small 2018, 14, 1802052. [Google Scholar] [CrossRef]
- Patil, S.M.; Sawant, S.S.; Kunda, N.K. Exosomes as drug delivery systems: A brief overview and progress update. Eur. J. Pharm. Biopharm. 2020, 154, 259–269. [Google Scholar] [CrossRef] [PubMed]
- Dou, G.; Tian, R.; Liu, X.; Yuan, P.; Ye, Q.; Liu, J.; Liu, S.; Zhou, J.; Deng, Z.; Chen, X.; et al. Chimeric apoptotic bodies functionalized with natural membrane and modular delivery system for inflammation modulation. Sci. Adv. 2020, 6, eaba2987. [Google Scholar] [CrossRef]
- Vashisht, M.; Sunita, R.P.; Onteru, S.K.; Singh, D. Curcumin Primed Exosomes Reverses LPS-Induced Pro-Inflammatory Gene Expression in Buffalo Granulosa Cells; Illumina, Inc.: San Diego, CA, USA, 2018; Volume 119, ISBN 9118422500. [Google Scholar]
- Wu, H.; Zhou, J.; Zeng, C.; Wu, D.; Mu, Z.; Chen, B.; Xie, Y.; Ye, Y.; Liu, J. Curcumin increases exosomal TCF21 thus suppressing exosomeinduced lung cancer. Oncotarget 2016, 7, 87081–87090. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jang, J.Y.; Lee, J.K.; Jeon, Y.K.; Kim, C.W. Exosome derived from epigallocatechin gallate treated breast cancer cells suppresses tumor growth by inhibiting tumor-associated macrophage infiltration and M2 polarization. BMC Cancer 2013, 13, 421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kalani, A.; Kamat, P.K.; Chaturvedi, P.; Tyagi, S.C.; Tyagi, N. Curcumin-primed exosomes mitigate endothelial cell dysfunction during hyperhomocysteinemia. Life Sci. 2014, 107, 1–7. [Google Scholar] [CrossRef] [Green Version]
- Taverna, S.; Giallombardo, M.; Pucci, M.; Flugy, A.; Manno, M.; Raccosta, S.; Rolfo, C.; De Leo, G.; Alessandro, R. Curcumin inhibits in vitro and in vivo chronic myelogenous leukemia cells growth: A possible role for exosomal disposal of miR-21. Oncotarget 2015, 6, 21918–21933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Taverna, S.; Fontana, S.; Monteleone, F.; Pucci, M.; Saieva, L.; De Caro, V.; Cardinale, V.G.; Giallombardo, M.; Vicario, E.; Rolfo, C.; et al. Curcumin modulates chronic myelogenous leukemia exosomes composition and affects angiogenic phenotype, via exosomal miR-21. Oncotarget 2016, 7, 30420–30439. [Google Scholar] [CrossRef]
- Kalani, A.; Chaturvedi, P.; Kamat, P.K.; Maldonado, C.; Bauer, P.; Joshua, I.G.; Tyagi, S.C.; Tyagi, N. Curcumin-loaded embryonic stem cell exosomes restored neurovascular unit following ischemia-reperfusion injury. Int. J. Biochem. Cell Biol. 2016, 79, 360–369. [Google Scholar] [CrossRef] [Green Version]
- Aqil, F.; Munagala, R.; Jeyabalan, J.; Agrawal, A.K.; Gupta, R. Exosomes for the enhanced tissue bioavailability and efficacy of curcumin. AAPS J. 2017, 19, 1691–1702. [Google Scholar] [CrossRef]
- Vashisht, M.; Rani, P.; Onteru, S.K.; Singh, D. Curcumin encapsulated in milk exosomes resists human digestion and possesses enhanced intestinal permeability in vitro. Appl. Biochem. Biotechnol. 2017, 183, 993–1007. [Google Scholar] [CrossRef]
- Kim, G.Y.; Lee, Y.; Ha, J.; Han, S.; Lee, M. Engineering exosomes for pulmonary delivery of peptides and drugs to inflammatory lung cells by inhalation. J. Control. Release 2021, 330, 684–695. [Google Scholar] [CrossRef]
- Munagala, R.; Aqil, F.; Jeyabalan, J.; Agrawal, A.K.; Mudd, A.M.; Kyakulaga, A.H.; Singh, I.P.; Vadhanam, M.V.; Gupta, R.C. Exosomal formulation of anthocyanidins against multiple cancer types. Cancer Lett. 2017, 393, 94–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aqil, F.; Jeyabalan, J.; Agrawal, A.K.; Kyakulaga, A.H.; Munagala, R.; Parker, L.; Gupta, R.C. Exosomal delivery of berry anthocyanidins for the management of ovarian cancer. Food Funct. 2017, 8, 4100–4107. [Google Scholar] [CrossRef]
- Pucek, A.; Tokarek, B.; Waglewska, E.; Bazylińska, U. Recent advances in the structural design of photosensitive agent formulations using “soft” colloidal nanocarriers. Pharmaceutics 2020, 12, 587. [Google Scholar] [CrossRef] [PubMed]
- Ganesan, P.; Narayanasamy, D. Lipid nanoparticles: Different preparation techniques, characterization, hurdles, and strategies for the production of solid lipid nanoparticles and nanostructured lipid carriers for oral drug delivery. Sustain. Chem. Pharm. 2017, 6, 37–56. [Google Scholar] [CrossRef]
- Scioli Montoto, S.; Muraca, G.; Ruiz, M.E. Solid lipid nanoparticles for drug delivery: Pharmacological and biopharmaceutical aspects. Front. Mol. Biosci. 2020, 7, 587997. [Google Scholar] [CrossRef]
- Mishra, V.; Bansal, K.K.; Verma, A.; Yadav, N.; Thakur, S.; Sudhakar, K.; Rosenholm, J.M. Solid lipid nanoparticles: Emerging colloidal nano drug delivery systems. Pharmaceutics 2018, 10, 191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Borges, A.; de Freitas, V.; Mateus, N.; Fernandes, I.; Oliveira, J. Solid lipid nanoparticles as carriers of natural phenolic compounds. Antioxidants 2020, 9, 998. [Google Scholar] [CrossRef] [PubMed]
- Silva, A.M.; Martins-Gomes, C.; Fangueiro, J.F.; Andreani, T.; Souto, E.B. Comparison of antiproliferative effect of epigallocatechin gallate when loaded into cationic solid lipid nanoparticles against different cell lines. Pharm. Dev. Technol. 2019, 24, 1243–1249. [Google Scholar] [CrossRef] [Green Version]
- Shtay, R.; Keppler, J.K.; Schrader, K.; Schwarz, K. Encapsulation of (−)-epigallocatechin-3-gallate (EGCG) in solid lipid nanoparticles for food applications. J. Food Eng. 2019, 244, 91–100. [Google Scholar] [CrossRef]
- Ramesh, N.; Mandal, A.K.A. Pharmacokinetic, toxicokinetic, and bioavailability studies of epigallocatechin-3-gallate loaded solid lipid nanoparticle in rat model. Drug Dev. Ind. Pharm. 2019, 45, 1506–1514. [Google Scholar] [CrossRef]
- Ban, C.; Jo, M.; Park, Y.H.; Kim, J.H.; Han, J.Y.; Lee, K.W.; Kweon, D.H.; Choi, Y.J. Enhancing the oral bioavailability of curcumin using solid lipid nanoparticles. Food Chem. 2020, 302, 125328. [Google Scholar] [CrossRef] [PubMed]
- Gupta, T.; Singh, J.; Kaur, S.; Sandhu, S.; Singh, G.; Kaur, I.P. Enhancing bioavailability and stability of curcumin using solid lipid nanoparticles (CLEN): A covenant for its effectiveness. Front. Bioeng. Biotechnol. 2020, 8, 879. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Zhao, X.; Ma, Y.; Zhai, G.; Li, L.; Lou, H. Enhancement of gastrointestinal absorption of quercetin by solid lipid nanoparticles. J. Control. Release 2009, 133, 238–244. [Google Scholar] [CrossRef]
- Pandita, D.; Kumar, S.; Poonia, N.; Lather, V. Solid lipid nanoparticles enhance oral bioavailability of resveratrol, a natural polyphenol. Food Res. Int. 2014, 62, 1165–1174. [Google Scholar] [CrossRef]
- Zhang, L.; Zhu, K.; Zeng, H.; Zhang, J.; Pu, Y.; Wang, Z.; Zhang, T.; Wang, B. Resveratrol solid lipid nanoparticles to trigger credible inhibition of doxorubicin cardiotoxicity. Int. J. Nanomed. 2019, 14, 6061–6071. [Google Scholar] [CrossRef] [Green Version]
- Soldati, P.P.; Polonini, H.C.; Paes, C.Q.; Restrepob, J.A.S.; Creczynksi-Pasa, T.B.; Chaves, M.G.A.M.; Brandão, M.A.F.; Pittella, F.; Raposo, N.R.B. Controlled release of resveratrol from lipid nanoparticles improves antioxidant effect. IFAC PapersOnLine 2018, 51, 16–21. [Google Scholar] [CrossRef]
- Mohseni, R.; ArabSadeghabadi, Z.; Ziamajidi, N.; Abbasalipourkabir, R.; RezaeiFarimani, A. Oral Administration of resveratrol-loaded solid lipid nanoparticle improves insulin resistance through targeting expression of SNARE proteins in adipose and muscle tissue in rats with type 2 diabetes. Nanoscale Res. Lett. 2019, 14, 227. [Google Scholar] [CrossRef]
- Souto, E.B.; Zielinska, A.; Souto, S.B.; Durazzo, A.; Lucarini, M.; Santini, A.; Silva, A.M.; Atanasov, A.G.; Marques, C.; Andrade, L.N.; et al. (+)-limonene 1,2-epoxide-loaded slns: Evaluation of drug release, antioxidant activity, and cytotoxicity in an HaCaT cell line. Int. J. Mol. Sci. 2020, 21, 1449. [Google Scholar] [CrossRef] [Green Version]
- Badawi, N.; El-Say, K.; Attia, D.; El-Nabarawi, M.; Elmazar, M.; Teaima, M. Development of pomegranate extract-loaded solid lipid nanoparticles: Quality by design approach to screen the variables affecting the quality attributes and characterization. ACS Omega 2020, 5, 21712–21721. [Google Scholar] [CrossRef] [PubMed]
- Lammari, N.; Louaer, O.; Meniai, A.H.; Elaissari, A. Encapsulation of essential oils via nanoprecipitation process: Overview, progress, challenges and prospects. Pharmaceutics 2020, 12, 431. [Google Scholar] [CrossRef]
- Nasseri, M.; Golmohammadzadeh, S.; Arouiee, H.; Jaafari, M.R.; Neamati, H. Antifungal activity of zataria multiflora essential oil-loaded solid lipid nanoparticles in-vitro condition. Iran. J. Basic Med. Sci. 2016, 19, 1231–1237. [Google Scholar] [CrossRef]
- Zhao, Y.; Chang, Y.X.; Hu, X.; Liu, C.Y.; Quan, L.H.; Liao, Y.H. Solid lipid nanoparticles for sustained pulmonary delivery of Yuxingcao essential oil: Preparation, characterization and in vivo evaluation. Int. J. Pharm. 2017, 516, 364–371. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Ciftci, O.N. Development of free-flowing peppermint essential oil-loaded hollow solid lipid micro- and nanoparticles via atomization with carbon dioxide. Food Res. Int. 2016, 87, 83–91. [Google Scholar] [CrossRef]
- Chauhan, I.; Yasir, M.; Verma, M.; Singh, A.P. Nanostructured lipid carriers: A groundbreaking approach for transdermal drug delivery. Adv. Pharm Bull. 2020, 10, 150–165. [Google Scholar] [CrossRef]
- Piazzini, V.; Lemmi, B.; D’Ambrosio, M.; Cinci, L.; Luceri, C.; Bilia, A.R.; Bergonzi, M.C. Nanostructured lipid carriers as promising delivery systems for plant extracts: The case of silymarin. Appl. Sci. 2018, 8, 1163. [Google Scholar] [CrossRef] [Green Version]
- Bhise, K.; Kashaw, S.K.; Sau, S.; Iyer, A.K. Nanostructured lipid carriers employing polyphenols as promising anticancer agents: Quality by design (QbD) approach. Int. J. Pharm. 2017, 526, 506–515. [Google Scholar] [CrossRef] [PubMed]
- Kumar, P.; Sharma, G.; Kumar, R.; Singh, B.; Malik, R.; Katare, O.P.; Raza, K. Promises of a biocompatible nanocarrier in improved brain delivery of quercetin: Biochemical, pharmacokinetic and biodistribution evidences. Int. J. Pharm. 2016, 515, 307–314. [Google Scholar] [CrossRef] [PubMed]
- Kamel, A.E.; Fadel, M.; Louis, D. Curcumin-loaded nanostructured lipid carriers prepared using peceolTM and olive oil in photodynamic therapy: Development and application in breast cancer cell line. Int. J. Nanomed. 2019, 14, 5073–5085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sadati Behbahani, E.; Ghaedi, M.; Abbaspour, M.; Rostamizadeh, K.; Dashtian, K. Curcumin loaded nanostructured lipid carriers: In vitro digestion and release studies. Polyhedron 2019, 164, 113–122. [Google Scholar] [CrossRef]
- Rashidzadeh, H.; Salimi, M.; Sadighian, S.; Rostamizadeh, K.; Ramazani, A. In vivo antiplasmodial activity of curcumin-loaded nanostructured lipid carriers. Curr. Drug Deliv. 2019, 16, 923–930. [Google Scholar] [CrossRef]
- Selvaraj, K.; Yoo, B.K. Curcumin-loaded nanostructured lipid carrier modified with partially hydrolyzed ginsenoside. AAPS PharmSciTech 2019, 20, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Pimentel-Moral, S.; Teixeira, M.C.; Fernandes, A.R.; Borrás-Linares, I.; Arráez-Román, D.; Martínez-Férez, A.; Segura-Carretero, A.; Souto, E.B. Polyphenols-enriched Hibiscus sabdariffa extract-loaded nanostructured lipid carriers (NLC): Optimization by multi-response surface methodology. J. Drug Deliv. Sci. Technol. 2019, 49, 660–667. [Google Scholar] [CrossRef]
- Bashiri, S.; Ghanbarzadeh, B.; Ayaseh, A.; Dehghannya, J.; Ehsani, A.; Ozyurt, H. Essential oil-loaded nanostructured lipid carriers: The effects of liquid lipid type on the physicochemical properties in beverage models. Food Biosci. 2020, 35, 100526. [Google Scholar] [CrossRef]
- Ghodrati, M.; Farahpour, M.R.; Hamishehkar, H. Encapsulation of Peppermint essential oil in nanostructured lipid carriers: In-vitro antibacterial activity and accelerative effect on infected wound healing. Coll. Surf. A Physicochem. Eng. Asp. 2019, 564, 161–169. [Google Scholar] [CrossRef]
- Vieira, R.; Severino, P.; Nalone, L.A.; Souto, S.B.; Silva, A.M.; Lucarini, M.; Durazzo, A.; Santini, A.; Souto, E.B. Sucupira oil-loaded nanostructured lipid carriers (NLC): Lipid screening, factorial design, release profile, and cytotoxicity. Molecules 2020, 25, 685. [Google Scholar] [CrossRef] [Green Version]
- Avadhani, K.S.; Manikkath, J.; Tiwari, M.; Chandrasekhar, M.; Godavarthi, A.; Vidya, S.M.; Hariharapura, R.C.; Kalthur, G.; Udupa, N.; Mutalik, S. Skin delivery of epigallocatechin-3-gallate (EGCG) and hyaluronic acid loaded nano-transfersomes for antioxidant and anti-aging effects in UV radiation induced skin damage. Drug Deliv. 2017, 24, 61–74. [Google Scholar] [CrossRef] [Green Version]
- Arsić, I.; Tadić, V.; Vlaović, D.; Homåek, I.; Vesić, S.; Isailović, G.; Vuleta, G. Preparation of novel apigenin-enriched, liposomal and non-liposomal, antiinflammatory topical formulations as substitutes for corticosteroid therapy. Phyther. Res. 2011, 25, 228–233. [Google Scholar] [CrossRef]
- Al Shaal, L.; Shegokar, R.; Müller, R.H. Production and characterization of antioxidant apigenin nanocrystals as a novel UV skin protective formulation. Int. J. Pharm. 2011, 420, 133–140. [Google Scholar] [CrossRef]
- Fathi, M.; Donsi, F.; McClements, D.J. Protein-based delivery systems for the nanoencapsulation of food ingredients. Compr. Rev. Food Sci. Food Saf. 2018, 17, 920–936. [Google Scholar] [CrossRef] [Green Version]
- Rather, S.A.; Masoodi, F.A.; Rather, J.A.; Akhter, R.; Ganaie, T.A. Protein-based delivery systems. In Food Hydrocolloids as Encapsulating Agents in Delivery Systems; Adil, G., Masoodi, F., Shah, U., Shah, A., Eds.; CRC Press: Boca Raton, FL, USA, 2019; pp. 159–176. ISBN 9781138600140. [Google Scholar]
- Malekhosseini, P.; Alami, M.; Khomeiri, M.; Esteghlal, S.; Nekoei, A.R.; Hosseini, S.M.H. Development of casein-based nanoencapsulation systems for delivery of epigallocatechin gallate and folic acid. Food Sci. Nutr. 2019, 7, 519–527. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Jarunglumlert, T.; Nakagawa, K.; Adachi, S. Influence of aggregate structure of casein on the encapsulation efficiency of β-carotene entrapped via hydrophobic interaction. Food Struct. 2015, 5, 42–50. [Google Scholar] [CrossRef]
- Loewen, A.; Chan, B.; Li-Chan, E.C.Y. Optimization of vitamins A and D3 loading in re-assembled casein micelles and effect of loading on stability of vitamin D3 during storage. Food Chem. 2018, 240, 472–481. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Ha, H.K.; Rankin, S.A.; Lee, M.R.; Lee, W.J. Development and characterization of whey protein-based nano-delivery systems: A review. Molecules 2019, 24, 3254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alavi, F.; Emam-Djomeh, Z.; Yarmand, M.S.; Salami, M.; Momen, S.; Moosavi-Movahedi, A.A. Cold gelation of curcumin loaded whey protein aggregates mixed with k-carrageenan: Impact of gel microstructure on the gastrointestinal fate of curcumin. Food Hydrocoll. 2018, 85, 267–280. [Google Scholar] [CrossRef]
- Mohammadian, M.; Salami, M.; Momen, S.; Alavi, F.; Emam-Djomeh, Z.; Moosavi-Movahedi, A.A. Enhancing the aqueous solubility of curcumin at acidic condition through the complexation with whey protein nanofibrils. Food Hydrocoll. 2019, 87, 902–914. [Google Scholar] [CrossRef]
- Taha, S.; El-Sherbiny, I.; Enomoto, T.; Salem, A.; Nagai, E.; Askar, A.; Abady, G.; Abdel-Hamid, M. Improving the functional activities of curcumin using milk proteins as nanocarriers. Foods 2020, 9, 986. [Google Scholar] [CrossRef]
- Zhao, C.; Shen, X.; Guo, M. Stability of lutein encapsulated whey protein nano-emulsion during storage. PLoS ONE 2018, 13, e0192511. [Google Scholar] [CrossRef]
- Goyal, A.; Sharma, V.; Upadhyay, N.; Singh, A.K.; Arora, S.; Lal, D.; Sabikhi, L. Development of stable flaxseed oil emulsions as a potential delivery system of ω-3 fatty acids. J. Food Sci. Technol. 2015, 52, 4256–4265. [Google Scholar] [CrossRef] [Green Version]
- Hu, Y.; Kou, G.; Chen, Q.; Li, Y.; Zhou, Z. Protection and delivery of mandarin (Citrus reticulata Blanco) peel extracts by encapsulation of whey protein concentrate nanoparticles. LWT 2019, 99, 24–33. [Google Scholar] [CrossRef]
- Do Carmo, E.L.; Teodoro, R.A.R.; Félix, P.H.C.; de Fernandes, R.V.B.; de Oliveira, É.R.; Veiga, T.R.L.A.; Borges, S.V.; Botrel, D.A. Stability of spray-dried beetroot extract using oligosaccharides and whey proteins. Food Chem. 2018, 249, 51–59. [Google Scholar] [CrossRef]
- Kavas, G.; Kavas, N.; Saygili, D. The effects of thyme and clove essential oil fortified edible films on the physical, chemical and microbiological characteristics of kashar cheese. J. Food Qual. 2015, 38, 405–412. [Google Scholar] [CrossRef] [Green Version]
- Seydim, A.C.; Sarikus-Tutal, G.; Sogut, E. Effect of whey protein edible films containing plant essential oils on microbial inactivation of sliced Kasar cheese. Food Packag. Shelf Life 2020, 26, 100567. [Google Scholar] [CrossRef]
- Deng, L.; Kang, X.; Liu, Y.; Feng, F.; Zhang, H. Effects of surfactants on the formation of gelatin nanofibres for controlled release of curcumin. Food Chem. 2017, 231, 70–77. [Google Scholar] [CrossRef] [PubMed]
- Nejat, H.; Rabiee, M.; Varshochian, R.; Tahriri, M.; Jazayeri, H.E.; Rajadas, J.; Ye, H.; Cui, Z.; Tayebi, L. Preparation and characterization of cardamom extract-loaded gelatin nanoparticles as effective targeted drug delivery system to treat glioblastoma. React. Funct. Polym. 2017, 120, 46–56. [Google Scholar] [CrossRef]
- Zhang, H.; Liang, Y.; Li, X.; Kang, H. Effect of chitosan-gelatin coating containing nano-encapsulated tarragon essential oil on the preservation of pork slices. Meat Sci. 2020, 166, 108137. [Google Scholar] [CrossRef]
- Tavassoli-Kafrani, E.; Goli, S.A.H.; Fathi, M. Encapsulation of orange essential oil using cross-linked electrospun gelatin nanofibers. Food Bioprocess. Technol. 2018, 11, 427–434. [Google Scholar] [CrossRef]
- Wan, Z.L.; Guo, J.; Yang, X.Q. Plant protein-based delivery systems for bioactive ingredients in foods. Food Funct. 2015, 6, 2876–2889. [Google Scholar] [CrossRef] [PubMed]
- Tang, C.H. Nanostructured soy proteins: Fabrication and applications as delivery systems for bioactives—A review. Food Hydrocoll. 2019, 91, 92–116. [Google Scholar] [CrossRef]
- Gaber Ahmed, G.H.; Fernández-González, A.; Díaz García, M.E. Nano-encapsulation of grape and apple pomace phenolic extract in chitosan and soy protein via nanoemulsification. Food Hydrocoll. 2020, 108, 105806. [Google Scholar] [CrossRef]
- Pujara, N.; Jambhrunkar, S.; Wong, K.Y.; McGuckin, M.; Popat, A. Enhanced colloidal stability, solubility and rapid dissolution of resveratrol by nanocomplexation with soy protein isolate. J. Coll. Interface Sci. 2017, 488, 303–308. [Google Scholar] [CrossRef] [Green Version]
- Chen, F.P.; Li, B.S.; Tang, C.H. Nanocomplexation between curcumin and soy protein isolate: Influence on curcumin stability/bioaccessibility and in vitro protein digestibility. J. Agric. Food Chem. 2015, 63, 3559–3569. [Google Scholar] [CrossRef] [PubMed]
- Yi, J.; Lam, T.I.; Yokoyama, W.; Cheng, L.W.; Zhong, F. Beta-carotene encapsulated in food protein nanoparticles reduces peroxyl radical oxidation in Caco-2 cells. Food Hydrocoll. 2015, 43, 31–40. [Google Scholar] [CrossRef]
- Luo, Y.; Wang, Q. Zein-based micro- and nano-particles for drug and nutrient delivery: A review. J. Appl. Polym. Sci. 2014, 131, 40696. [Google Scholar] [CrossRef]
- Li, H.; Wang, D.; Liu, C.; Zhu, J.; Fan, M.; Sun, X.; Wang, T.; Xu, Y.; Cao, Y. Fabrication of stable zein nanoparticles coated with soluble soybean polysaccharide for encapsulation of quercetin. Food Hydrocoll. 2019, 87, 342–351. [Google Scholar] [CrossRef]
- Zou, L.; Zheng, B.; Zhang, R.; Zhang, Z.; Liu, W.; Liu, C.; Xiao, H.; McClements, D.J. Enhancing the bioaccessibility of hydrophobic bioactive agents using mixed colloidal dispersions: Curcumin-loaded zein nanoparticles plus digestible lipid nanoparticles. Food Res. Int. 2016, 81, 74–82. [Google Scholar] [CrossRef] [Green Version]
- Yuan, Y.; Li, H.; Zhu, J.; Liu, C.; Sun, X.; Wang, D.; Xu, Y. Fabrication and characterization of zein nanoparticles by dextran sulfate coating as vehicles for delivery of curcumin. Int. J. Biol. Macromol. 2020, 151, 1074–1083. [Google Scholar] [CrossRef]
- Wang, H.; Hao, L.; Wang, P.; Chen, M.; Jiang, S.; Jiang, S. Release kinetics and antibacterial activity of curcumin loaded zein fibers. Food Hydrocoll. 2017, 63, 437–446. [Google Scholar] [CrossRef]
- Cheng, C.J.; Ferruzzi, M.; Jones, O.G. Fate of lutein-containing zein nanoparticles following simulated gastric and intestinal digestion. Food Hydrocoll. 2019, 87, 229–236. [Google Scholar] [CrossRef]
- Chuacharoen, T.; Sabliov, C.M. Stability and controlled release of lutein loaded in zein nanoparticles with and without lecithin and pluronic F127 surfactants. Coll. Surf. A Physicochem. Eng. Asp. 2016, 503, 11–18. [Google Scholar] [CrossRef] [Green Version]
- Pinheiro Bruni, G.; dos Santos Acunha, T.; de Oliveira, J.P.; Martins Fonseca, L.; Tavares da Silva, F.; Martins Guimarães, V.; da Rosa Zavareze, E. Electrospun protein fibers loaded with yerba mate extract for bioactive release in food packaging. J. Sci. Food Agric. 2020, 100, 3341–3350. [Google Scholar] [CrossRef]
- Heydari-Majd, M.; Rezaeinia, H.; Shadan, M.R.; Ghorani, B.; Tucker, N. Enrichment of zein nanofibre assemblies for therapeutic delivery of Barije (Ferula gummosa Boiss) essential oil. J. Drug Deliv. Sci. Technol. 2019, 54, 101290. [Google Scholar] [CrossRef]
- Guo, Q.; Shu, X.; Hu, Y.; Su, J.; Chen, S.; Decker, E.A.; Gao, Y. Formulated protein-polysaccharide-surfactant ternary complexes for co-encapsulation of curcumin and resveratrol: Characterization, stability and in vitro digestibility. Food Hydrocoll. 2021, 111, 106265. [Google Scholar] [CrossRef]
- Guo, Q.; Su, J.; Shu, X.; Yuan, F.; Mao, L.; Gao, Y. Development of high methoxyl pectin-surfactant-pea protein isolate ternary complexes: Fabrication, characterization and delivery of resveratrol. Food Chem. 2020, 321, 126706. [Google Scholar] [CrossRef]
- Fan, Y.; Zeng, X.; Yi, J.; Zhang, Y. Fabrication of pea protein nanoparticles with calcium-induced cross-linking for the stabilization and delivery of antioxidative resveratrol. Int. J. Biol. Macromol. 2020, 152, 189–198. [Google Scholar] [CrossRef] [PubMed]
- Cuevas-Bernardino, J.C.; Leyva-Gutierrez, F.M.A.; Vernon-Carter, E.J.; Lobato-Calleros, C.; Román-Guerrero, A.; Davidov-Pardo, G. Formation of biopolymer complexes composed of pea protein and mesquite gum—Impact of quercetin addition on their physical and chemical stability. Food Hydrocoll. 2018, 77, 736–745. [Google Scholar] [CrossRef]
- Jiang, S.; Yildiz, G.; Ding, J.; Andrade, J.; Rababahb, T.M.; Almajwalc, A.; Abulmeatyc, M.M.; Feng, H. Pea Protein nanoemulsion and nanocomplex as carriers for protection of cholecalciferol (vitamin D3). Food Bioprocess. Technol. 2019, 12, 1031–1040. [Google Scholar] [CrossRef]
- Benito-Román, O.; Sanz, T.; Beltrán, S. Microencapsulation of rice bran oil using pea protein and maltodextrin mixtures as wall material. Heliyon 2020, 6, e03615. [Google Scholar] [CrossRef]
- Can Karaca, A. Encapsulation of black pepper seed oil using maltodextrin and pea protein. Food Sci. Technol. Int. 2020, 26, 369–378. [Google Scholar] [CrossRef]
- Jansen-Alves, C.; Fernandes, K.F.; Crizel-Cardozo, M.M.; Krumreich, F.D.; Borges, C.D.; Zambiazi, R.C. Microencapsulation of propolis in protein matrix using spray drying for application in food systems. Food Bioprocess. Technol. 2018, 11, 1422–1436. [Google Scholar] [CrossRef]
- Yildiz, G.; Ding, J.; Gaur, S.; Andrade, J.; Engeseth, N.E.; Feng, H. Microencapsulation of docosahexaenoic acid (DHA) with four wall materials including pea protein-modified starch complex. Int. J. Biol. Macromol. 2018, 114, 935–941. [Google Scholar] [CrossRef] [PubMed]
- Costa, A.M.M.; Nunes, J.C.; Lima, B.N.B.; Pedrosa, C.; Calado, V.; Torres, A.G.; Pierucci, A.P.T.R. Effective stabilization of CLA by microencapsulation in pea protein. Food Chem. 2015, 168, 157–166. [Google Scholar] [CrossRef] [Green Version]
- Ho, K.K.H.Y.; Schroën, K.; San Martín-González, M.F.; Berton-Carabin, C.C. Physicochemical stability of lycopene-loaded emulsions stabilized by plant or dairy proteins. Food Struct. 2017, 12, 34–42. [Google Scholar] [CrossRef]
- Fathi, M.; Martín, Á.; McClements, D.J. Nanoencapsulation of food ingredients using carbohydrate based delivery systems. Trends Food Sci. Technol. 2014, 39, 18–39. [Google Scholar] [CrossRef]
- Khaleel Basha, S.; Syed Muzammil, M.; Dhandayuthabani, R.; Sugantha Kumari, V. Polysaccharides as excipient in drug delivery system. Mater. Today Proc. 2019, 36, 280–289. [Google Scholar] [CrossRef]
- Rostamabadi, H.; Falsafi, S.R.; Jafari, S.M. Starch-based nanocarriers as cutting-edge natural cargos for nutraceutical delivery. Trends Food Sci. Technol. 2019, 88, 397–415. [Google Scholar] [CrossRef]
- Cai, R.; Yuan, Y.; Cui, L.; Wang, Z.; Yue, T. Cyclodextrin-assisted extraction of phenolic compounds: Current research and future prospects. Trends Food Sci. Technol. 2018, 79, 19–27. [Google Scholar] [CrossRef]
- Diamanti, A.C.; Igoumenidis, P.E.; Mourtzinos, I.; Yannakopoulou, K.; Karathanos, V.T. Green extraction of polyphenols from whole pomegranate fruit using cyclodextrins. Food Chem. 2017, 214, 61–66. [Google Scholar] [CrossRef] [PubMed]
- Korompokis, K.; Igoumenidis, P.E.; Mourtzinos, I.; Karathanos, V.T. Green extraction and simultaneous inclusion complex formation of Sideritis scardica polyphenols. Int. Food Res. J. 2017, 24, 1233–1238. [Google Scholar]
- Kyriakidou, K.; Mourtzinos, I.; Biliaderis, C.G.; Makris, D.P. Optimization of a green extraction/inclusion complex formation process to recover antioxidant polyphenols from oak acorn husks (Quercus robur) using aqueous 2-hydroxypropyl-β-cyclodextrin/glycerol mixtures. Environments 2016, 3, 3. [Google Scholar] [CrossRef] [Green Version]
- Akbari-Alavijeh, S.; Shaddel, R.; Jafari, S.M. Encapsulation of food bioactives and nutraceuticals by various chitosan-based nanocarriers. Food Hydrocoll. 2020, 105, 105774. [Google Scholar] [CrossRef]
- Rehman, A.; Ahmad, T.; Aadil, R.M.; Spotti, M.J.; Bakry, A.M.; Khan, I.M.; Zhao, L.; Riaz, T.; Tong, Q. Pectin polymers as wall materials for the nano-encapsulation of bioactive compounds. Trends Food Sci. Technol. 2019, 90, 35–46. [Google Scholar] [CrossRef]
- Kosaraju, S.L. Colon targeted delivery systems: Review of polysaccharides for encapsulation and delivery. Crit. Rev. Food Sci. Nutr. 2005, 45, 251–258. [Google Scholar] [CrossRef]
- Shishir, M.R.I.; Xie, L.; Sun, C.; Zheng, X.; Chen, W. Advances in micro and nano-encapsulation of bioactive compounds using biopolymer and lipid-based transporters. Trends Food Sci. Technol. 2018, 78, 34–60. [Google Scholar] [CrossRef]
- Taheri, A.; Jafari, S.M. Gum-based nanocarriers for the protection and delivery of food bioactive compounds. Adv. Coll. Interface Sci. 2019, 269, 277–295. [Google Scholar] [CrossRef]
- Jouybar, A.; Seyedjafari, E.; Ardeshirylajimi, A.; Zandi-Karimi, A.; Feizi, N.; Khani, M.M.; Pousti, I. Enhanced skin regeneration by herbal extract-coated poly-L-lactic acid nanofibrous scaffold. Artif. Organs 2017, 41, E296–E307. [Google Scholar] [CrossRef]
- Pijpers, I.A.B.; Abdelmohsen, L.K.E.A.; Xia, Y.; Cao, S.; Williams, D.S.; Meng, F.; Hest, J.C.M.; Zhong, Z. Adaptive polymersome and micelle morphologies in anticancer nanomedicine: From design rationale to fabrication and proof-of-concept studies. Adv. Ther. 2018, 1, 1800068. [Google Scholar] [CrossRef]
- Ahmad, M.; Gani, A. Ultrasonicated resveratrol loaded starch nanocapsules: Characterization, bioactivity and release behaviour under in-vitro digestion. Carbohydr. Polym. 2021, 251, 117111. [Google Scholar] [CrossRef]
- Ahmad, M.; Mudgil, P.; Gani, A.; Hamed, F.; Masoodi, F.A.; Maqsood, S. Nano-encapsulation of catechin in starch nanoparticles: Characterization, release behavior and bioactivity retention during simulated in-vitro digestion. Food Chem. 2019, 270, 95–104. [Google Scholar] [CrossRef]
- Farrag, Y.; Ide, W.; Montero, B.; Rico, M.; Rodríguez-Llamazares, S.; Barral, L.; Bouza, R. Preparation of starch nanoparticles loaded with quercetin using nanoprecipitation technique. Int. J. Biol. Macromol. 2018, 114, 426–433. [Google Scholar] [CrossRef] [PubMed]
- Hasanvand, E.; Fathi, M.; Bassiri, A. Production and characterization of vitamin D3 loaded starch nanoparticles: Effect of amylose to amylopectin ratio and sonication parameters. J. Food Sci. Technol. 2018, 55, 1314–1324. [Google Scholar] [CrossRef] [PubMed]
- Gómez-Aldapa, C.A.; Castro-Rosas, J.; Rangel-Vargas, E.; Navarro-Cortez, R.O.; Cabrera-Canales, Z.E.; Díaz-Batalla, L.; Martínez-Bustos, F.; Guzmán-Ortiz, F.A.; Falfan-Cortes, R.N. A modified Achira (Canna indica L.) starch as a wall material for the encapsulation of Hibiscus sabdariffa extract using spray drying. Food Res. Int. 2019, 119, 547–553. [Google Scholar] [CrossRef]
- Das, A.B.; Goud, V.V.; Das, C. Microencapsulation of anthocyanin extract from purple rice bran using modified rice starch and its effect on rice dough rheology. Int. J. Biol. Macromol. 2019, 124, 573–581. [Google Scholar] [CrossRef]
- Wang, T.; Wu, C.; Fan, G.; Li, T.; Gong, H.; Cao, F. Ginkgo biloba extracts-loaded starch nano-spheres: Preparation, characterization, and in vitro release kinetics. Int. J. Biol. Macromol. 2018, 106, 148–157. [Google Scholar] [CrossRef] [PubMed]
- Arya, P.; Raghav, N. In-vitro studies of Curcumin-β-cyclodextrin inclusion complex as sustained release system. J. Mol. Struct. 2021, 1228, 129774. [Google Scholar] [CrossRef]
- Duarte, A.; Martinho, A.; Luís, Â.; Figueiras, A.; Oleastro, M.; Domingues, F.C.; Silva, F. Resveratrol encapsulation with methyl-β-cyclodextrin for antibacterial and antioxidant delivery applications. LWT 2015, 63, 1254–1260. [Google Scholar] [CrossRef]
- Ahmad, M.; Ashraf, B.; Gani, A.; Gani, A. Microencapsulation of saffron anthocyanins using β glucan and β cyclodextrin: Microcapsule characterization, release behaviour & antioxidant potential during in-vitro digestion. Int. J. Biol. Macromol. 2018, 109, 435–442. [Google Scholar] [CrossRef]
- Kyriakoudi, A.; Tsimidou, M.Z. Properties of encapsulated saffron extracts in maltodextrin using the Büchi B-90 nano spray-dryer. Food Chem. 2018, 266, 458–465. [Google Scholar] [CrossRef]
- Lourenço, S.C.; Moldão-Martins, M.; Alves, V.D. Microencapsulation of pineapple peel extract by spray drying using maltodextrin, inulin, and arabic gum as wall matrices. Foods 2020, 9, 718. [Google Scholar] [CrossRef]
- Nair, R.S.; Morris, A.; Billa, N.; Leong, C.O. An evaluation of curcumin-encapsulated chitosan nanoparticles for transdermal delivery. AAPS PharmSciTech 2019, 20, 69. [Google Scholar] [CrossRef]
- Torpol, K.; Sriwattana, S.; Sangsuwan, J.; Wiriyacharee, P.; Prinyawiwatkul, W. Optimising chitosan–pectin hydrogel beads containing combined garlic and holy basil essential oils and their application as antimicrobial inhibitor. Int. J. Food Sci. Technol. 2019, 54, 2064–2074. [Google Scholar] [CrossRef]
- Tan, C.; Xie, J.; Zhang, X.; Cai, J.; Xia, S. Polysaccharide-based nanoparticles by chitosan and gum arabic polyelectrolyte complexation as carriers for curcumin. Food Hydrocoll. 2016, 57, 236–245. [Google Scholar] [CrossRef]
- Jamil, B.; Abbasi, R.; Abbasi, S.; Imran, M.; Khan, S.U.; Ihsan, A.; Javed, S.; Bokhari, H. Encapsulation of cardamom essential oil in chitosan nano-composites: In-vitro efficacy on antibiotic-resistant bacterial pathogens and cytotoxicity studies. Front. Microbiol. 2016, 7, 1580. [Google Scholar] [CrossRef] [PubMed]
- Sotelo-Boyás, M.E.; Correa-Pacheco, Z.N.; Bautista-Baños, S.; Corona-Rangel, M.L. Physicochemical characterization of chitosan nanoparticles and nanocapsules incorporated with lime essential oil and their antibacterial activity against food-borne pathogens. LWT Food Sci. Technol. 2017, 77, 15–20. [Google Scholar] [CrossRef]
- Shetta, A.; Kegere, J.; Mamdouh, W. Comparative study of encapsulated peppermint and green tea essential oils in chitosan nanoparticles: Encapsulation, thermal stability, in-vitro release, antioxidant and antibacterial activities. Int. J. Biol. Macromol. 2019, 126, 731–742. [Google Scholar] [CrossRef]
- Ashrafi, B.; Rashidipour, M.; Marzban, A.; Soroush, S.; Azadpour, M.; Delfani, S.; Ramak, P. Mentha piperita essential oils loaded in a chitosan nanogel with inhibitory effect on biofilm formation against S. mutans on the dental surface. Carbohydr. Polym. 2019, 212, 142–149. [Google Scholar] [CrossRef] [PubMed]
- Huang, X.; Dai, Y.; Cai, J.; Zhong, N.; Xiao, H.; McClements, D.J.; Hu, K. Resveratrol encapsulation in core-shell biopolymer nanoparticles: Impact on antioxidant and anticancer activities. Food Hydrocoll. 2017, 64, 157–165. [Google Scholar] [CrossRef] [Green Version]
- Ghasemi, S.; Jafari, S.M.; Assadpour, E.; Khomeiri, M. Nanoencapsulation of D-limonene within nanocarriers produced by pectin-whey protein complexes. Food Hydrocoll. 2018, 77, 152–162. [Google Scholar] [CrossRef]
- Veneranda, M.; Hu, Q.; Wang, T.; Luo, Y.; Castro, K.; Madariaga, J.M. Formation and characterization of zein-caseinate-pectin complex nanoparticles for encapsulation of eugenol. LWT Food Sci. Technol. 2018, 89, 596–603. [Google Scholar] [CrossRef]
- Zhou, M.; Wang, T.; Hu, Q.; Luo, Y. Low density lipoprotein/pectin complex nanogels as potential oral delivery vehicles for curcumin. Food Hydrocoll. 2016, 57, 20–29. [Google Scholar] [CrossRef] [Green Version]
- Guo, Q.; Su, J.; Shu, X.; Yuan, F.; Mao, L.; Liu, J.; Gao, Y. Production and characterization of pea protein isolate-pectin complexes for delivery of curcumin: Effect of esterified degree of pectin. Food Hydrocoll. 2020, 105, 105777. [Google Scholar] [CrossRef]
- Hu, Y.; Zhang, W.; Ke, Z.; Li, Y.; Zhou, Z. In vitro release and antioxidant activity of Satsuma mandarin (Citrus reticulata Blanco cv. unshiu) peel flavonoids encapsulated by pectin nanoparticles. Int. J. Food Sci. Technol. 2017, 52, 2362–2373. [Google Scholar] [CrossRef]
- Mohammadi, A.; Jafari, S.M.; Assadpour, E.; Faridi Esfanjani, A. Nano-encapsulation of olive leaf phenolic compounds through WPC-pectin complexes and evaluating their release rate. Int. J. Biol. Macromol. 2016, 82, 816–822. [Google Scholar] [CrossRef]
- Esfanjani, A.F.; Jafari, S.M.; Assadpoor, E.; Mohammadi, A. Nano-encapsulation of saffron extract through double-layered multiple emulsions of pectin and whey protein concentrate. J. Food Eng. 2015, 165, 149–155. [Google Scholar] [CrossRef]
- Zhou, Y.; Sun, S.; Bei, W.; Zahi, M.R.; Yuan, Q.; Liang, H. Preparation and antimicrobial activity of oregano essential oil Pickering emulsion stabilized by cellulose nanocrystals. Int. J. Biol. Macromol. 2018, 112, 7–13. [Google Scholar] [CrossRef] [PubMed]
- Razavi, M.S.; Golmohammadi, A.; Nematollahzadeh, A.; Fiori, F.; Rovera, C.; Farris, S. Preparation of cinnamon essential oil emulsion by bacterial cellulose nanocrystals and fish gelatin. Food Hydrocoll. 2020, 109, 106111. [Google Scholar] [CrossRef]
- Criado, P.; Fraschini, C.; Jamshidian, M.; Salmieri, S.; Desjardins, N.; Sahraoui, A.; Lacroix, M. Effect of cellulose nanocrystals on thyme essential oil release from alginate beads: Study of antimicrobial activity against Listeria innocua and ground meat shelf life in combination with gamma irradiation. Cellulose 2019, 26, 5247–5265. [Google Scholar] [CrossRef]
- Kasiri, N.; Fathi, M. Entrapment of peppermint oil using cellulose nanocrystals. Cellulose 2018, 25, 319–329. [Google Scholar] [CrossRef]
- De Frates, K.; Markiewicz, T.; Gallo, P.; Rack, A.; Weyhmiller, A.; Jarmusik, B.; Hu, X. Protein polymer-based nanoparticles: Fabrication and medical applications. Int. J. Mol. Sci. 2018, 19, 1717. [Google Scholar] [CrossRef] [Green Version]
- Nagavarma, B.V.N.; Yadav, H.K.S.; Ayaz, A.; Vasudha, L.S.; Shivakumar, H.G. Different techniques for preparation of polymeric nanoparticles—A review. Asian J. Pharm. Clin. Res. 2012, 5, 16–23. [Google Scholar]
- Suriya Prabha, A.; Dorothy, R.; Jancirani, S.; Rajendran, S.; Singh, G.; Senthil Kumaran, S. Recent advances in the study of toxicity of polymer-based nanomaterials. In Nanotoxicity—Prevention and Antibacterial Applications of Nanomaterials; Elsevier: Amsterdam, The Netherlands, 2020; pp. 143–165. ISBN 9780128199435. [Google Scholar]
- Torres, M.P.; Wilson-Welder, J.H.; Lopac, S.K.; Phanse, Y.; Carrillo-Conde, B.; Ramer-Tait, A.E.; Bellaire, B.H.; Wannemuehler, M.J.; Narasimhan, B. Polyanhydride microparticles enhance dendritic cell antigen presentation and activation. Acta Biomater. 2011, 7, 2857–2864. [Google Scholar] [CrossRef] [Green Version]
- Zielińska, A.; Carreiró, F.; Oliveira, A.M.O.; Neves, A.; Pires, B.; Venkatesh, D.N.; Durazzo, A.; Lucarini, M.; Eder, P.; Silva, A.; et al. Polymeric nanoparticles: Production, characterization, toxicology and ecotoxicology. Molecules 2020, 25, 3731. [Google Scholar] [CrossRef]
- Abdelghany, S.; Parumasivam, T.; Pang, A.; Roediger, B.; Tang, P.; Jahn, K.; Britton, W.J.; Chan, H.K. Alginate modified-PLGA nanoparticles entrapping amikacin and moxifloxacin as a novel host-directed therapy for multidrug-resistant tuberculosis. J. Drug Deliv. Sci. Technol. 2019, 52, 642–651. [Google Scholar] [CrossRef]
- Bertoni, S.; Passerini, N.; Albertini, B. Nanomaterials for Oral Drug Administration; Elsevier Inc.: Amsterdam, The Netherlands, 2020; ISBN 9780128180389. [Google Scholar]
- Mohanraj, V.J.; Chen, Y. Nanoparticles—A review. Trop. J. Pharm. Res. 2007, 5, 561–573. [Google Scholar] [CrossRef] [Green Version]
- George, A.; Shah, P.A.; Shrivastav, P.S. Natural biodegradable polymers based nano-formulations for drug delivery: A review. Int. J. Pharm. 2019, 561, 244–264. [Google Scholar] [CrossRef]
- Soppimath, K.S.; Aminabhavi, T.M.; Kulkarni, A.R.; Rudzinski, W.E. Biodegradable polymeric nanoparticles as drug delivery devices. J. Control. Release 2001, 70, 1–20. [Google Scholar] [CrossRef]
- Bhatia, S. Natural Polymer Drug Delivery Systems; Springer: Cham, Switzerland, 2016; ISBN 9783319411286. [Google Scholar]
- Andreani, T.; Fangueiro, J.; Jose, S.; Santini, A.; Silva, A.; Souto, E. Hydrophilic polymers for modified-release nanoparticles: A review of mathematical modelling for pharmacokinetic analysis. Curr. Pharm. Des. 2015, 21, 3090–3096. [Google Scholar] [CrossRef] [PubMed]
- Jawahar, N.; Meyyanathan, S. Polymeric nanoparticles for drug delivery and targeting: A comprehensive review. Int. J. Health Allied Sci. 2012, 1, 217. [Google Scholar] [CrossRef]
- Amgoth, C.; Phan, C.; Banavoth, M.; Rompivalasa, S.; Tang, G. Polymer properties: Functionalization and surface modified nanoparticles. In Role of Novel Drug Delivery Vehicles in Nanobiomedicine; IntechOpen: London, UK, 2020. [Google Scholar] [CrossRef] [Green Version]
- Pinto Reis, C.; Neufeld, R.J.; Ribeiro, A.J.; Veiga, F. Nanoencapsulation I. Methods for preparation of drug-loaded polymeric nanoparticles. Nanomed. Nanotechnol. Biol. Med. 2006, 2, 8–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hernández-Giottonini, K.Y.; Rodríguez-Córdova, R.J.; Gutiérrez-Valenzuela, C.A.; Peñuñuri-Miranda, O.; Zavala-Rivera, P.; Guerrero-Germán, P.; Lucero-Acuña, A. PLGA nanoparticle preparations by emulsification and nanoprecipitation techniques: Effects of formulation parameters. RSC Adv. 2020, 10, 4218–4231. [Google Scholar] [CrossRef] [Green Version]
- Bennet, D.; Kim, S. Polymer nanoparticles for smart drug delivery. In Application of Nanotechnology in Drug Delivery; IntechOpen: London, UK, 2014. [Google Scholar] [CrossRef] [Green Version]
- Desgouilles, S.; Vauthier, C.; Bazile, D.; Vacus, J.; Grossiord, J.L.; Veillard, M.; Couvreur, P. The design of nanoparticles obtained by solvent evaporation: A comprehensive study. Langmuir 2003, 19, 9504–9510. [Google Scholar] [CrossRef]
- Jose, S.; Sowmya, S.; Cinu, T.A.; Aleykutty, N.A.; Thomas, S.; Souto, E.B. Surface modified PLGA nanoparticles for brain targeting of Bacoside-A. Eur. J. Pharm. Sci. 2014, 63, 29–35. [Google Scholar] [CrossRef] [PubMed]
- Vieira, R.; Souto, S.B.; Sánchez-López, E.; Machado, A.L.; Severino, P.; Jose, S.; Santini, A.; Fortuna, A.; García, M.L.; Silva, A.M.; et al. Sugar-lowering drugs for type 2 diabetes mellitus and metabolic syndrome—Review of classical and new compounds: Part-I. Pharmaceuticals 2019, 12, 152. [Google Scholar] [CrossRef] [Green Version]
- Bohrey, S.; Chourasiya, V.; Pandey, A. Polymeric nanoparticles containing diazepam: Preparation, optimization, characterization, in-vitro drug release and release kinetic study. Nano Converg. 2016, 3, 3–9. [Google Scholar] [CrossRef] [Green Version]
- Vauthier, C.; Ponchel, G. Methods for the Preparation of Nanoparticles by Polymerization. In Polymer Nanoparticles for Nanomedicines; Vauthier, C., Ponchel, G., Eds.; Springer: Cham, Switzerland, 2016; ISBN 9783319414195. [Google Scholar]
- Sharma, N.; Madan, P.; Lin, S. Effect of process and formulation variables on the preparation of parenteral paclitaxel-loaded biodegradable polymeric nanoparticles: A co-surfactant study. Asian J. Pharm. Sci. 2016, 11, 404–416. [Google Scholar] [CrossRef] [Green Version]
- Szczęch, M.; Szczepanowicz, K. Polymeric core-shell nanoparticles prepared by spontaneous emulsification solvent evaporation and functionalized by the layer-by-layer method. Nanomaterials 2020, 10, 496. [Google Scholar] [CrossRef] [Green Version]
- Kumar, S.; Dilbaghi, N.; Saharan, R.; Bhanjana, G. Nanotechnology as emerging tool for enhancing solubility of poorly water-soluble drugs. Bionanoscience 2012, 2, 227–250. [Google Scholar] [CrossRef]
- Guterres, S.S.; Alves, M.P.; Pohlmann, A.R. Polymeric nanoparticles, nanospheres and nanocapsules, for cutaneous applications. Drug Target. Insights 2007, 2, 117739280700200. [Google Scholar] [CrossRef] [Green Version]
- Quintanar-Guerrero, D.; Allémann, E.; Doelker, E.; Fessi, H. Preparation and characterization of nanocapsules from preformed polymers by a new process based on emulsification-diffusion technique. Pharm. Res. 1998, 15, 1056–1062. [Google Scholar] [CrossRef]
- Wang, Y.; Li, P.; Tran, T.T.D.; Zhang, J.; Kong, L. Manufacturing techniques and surface engineering of polymer based nanoparticles for targeted drug delivery to cancer. Nanomaterials 2016, 6, 26. [Google Scholar] [CrossRef] [Green Version]
- Mahalingam, M.; Krishnamoorthy, K. Selection of a suitable method for the preparation of polymeric nanoparticles: Multi-criteria decision making approach. Adv. Pharm. Bull. 2015, 5, 57–67. [Google Scholar] [CrossRef]
- Silva, A.M.; Alvarado, H.L.; Abrego, G.; Martins-Gomes, C.; Garduño-Ramirez, M.L.; García, M.L.; Calpena, A.C.; Souto, E.B. In vitro cytotoxicity of oleanolic/ursolic acids-loaded in PLGA nanoparticles in different cell lines. Pharmaceutics 2019, 11, 362. [Google Scholar] [CrossRef] [Green Version]
- Carbone, C.; Martins-Gomes, C.; Pepe, V.; Silva, A.M.; Musumeci, T.; Puglisi, G.; Furneri, P.M.; Souto, E.B. Repurposing itraconazole to the benefit of skin cancer treatment: A combined azole-DDAB nanoencapsulation strategy. Coll. Surf. B Biointerfaces 2018, 167, 337–344. [Google Scholar] [CrossRef] [PubMed]
- Doktorovova, S.; Souto, E.B.; Silva, A.M. Nanotoxicology applied to solid lipid nanoparticles and nanostructured lipid carriers—A systematic review of in vitro data. Eur. J. Pharm. Biopharm. 2014, 87, 1–18. [Google Scholar] [CrossRef]
- Liang, H.F.; Chen, C.T.; Chen, S.C.; Kulkarni, A.R.; Chiu, Y.L.; Chen, M.C.; Sung, H.W. Paclitaxel-loaded poly(γ-glutamic acid)-poly(lactide) nanoparticles as a targeted drug delivery system for the treatment of liver cancer. Biomaterials 2006, 27, 2051–2059. [Google Scholar] [CrossRef] [PubMed]
- Venditti, I. Morphologies and functionalities of polymeric nanocarriers as chemical tools for drug delivery: A review. J. King Saud Univ. Sci. 2019, 31, 398–411. [Google Scholar] [CrossRef]
- Wu, H.; Yu, T.; Tian, Y.; Wang, Y.; Zhao, R.; Mao, S. Enhanced liver-targeting via coadministration of 10-Hydroxycamptothecin polymeric micelles with vinegar baked Radix Bupleuri. Phytomedicine 2018, 44, 1–8. [Google Scholar] [CrossRef]
- Su, Y.; Huang, N.; Chen, D.; Zhang, L.; Dong, X.; Sun, Y.; Zhu, X.; Zhang, F.; Gao, J.; Wang, Y.; et al. Sucessful in vivo hyperthermal therapy toward breast cancer by cchinese medicine shikonin-loaded thermosensitive micelle. Int. J. Nanomed. 2017, 12, 4019–4035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Anantaworasakul, P.; Okonogi, S. Encapsulation of Sesbania grandiflora extract in polymeric micelles to enhance its solubility, stability, and antibacterial activity. J. Microencapsul. 2017, 34, 73–81. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Tomalia, D.A.; Christensen, J.B.; Boas, U. Dendrimers, Dendrons, and Dendritic Polymers; Cambridge University Press: Cambridge, UK, 2012. [Google Scholar] [CrossRef]
- Nowacek, A.; Gendelman, H.E. NanoART, neuroAIDS and CNS drug delivery. Nanomedicine 2009, 4, 557–574. [Google Scholar] [CrossRef] [Green Version]
- Wong, H.L.; Wu, X.Y.; Bendayan, R. Nanotechnological advances for the delivery of CNS therapeutics. Adv. Drug Deliv. Rev. 2012, 64, 686–700. [Google Scholar] [CrossRef] [PubMed]
- Menjoge, A.R.; Kannan, R.M.; Tomalia, D.A. Dendrimer-based drug and imaging conjugates: Design considerations for nanomedical applications. Drug Discov. Today 2010, 15, 171–185. [Google Scholar] [CrossRef]
- Santos, A.; Veiga, F.; Figueiras, A. Dendrimers as pharmaceutical excipients: Synthesis, properties, toxicity and biomedical applications. Materials 2020, 13, 65. [Google Scholar] [CrossRef] [Green Version]
- Bugno, J.; Hsu, H.J.; Hong, S. Tweaking dendrimers and dendritic nanoparticles for controlled nano-bio interactions: Potential nanocarriers for improved cancer targeting. J. Drug Target. 2015, 23, 642–650. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Xu, X.; Zhang, Y.; Zhang, Y.; Zhu, Y.; Shi, J.; Sun, Y.; Huang, Q. Encapsulation of curcumin within poly(amidoamine) dendrimers for delivery to cancer cells. J. Mater. Sci. Mater. Med. 2013, 24, 2137–2144. [Google Scholar] [CrossRef] [PubMed]
- Falconieri, M.C.; Adamo, M.; Monasterolo, C.; Bergonzi, M.C.; Coronnello, M.; Bilia, A.R. New dendrimer-based nanoparticles enhance curcumin solubility. Planta Med. 2017, 83, 420–425. [Google Scholar] [CrossRef]
- Elmi, T.; Shafiee Ardestani, M.; Hajialiani, F.; Motevalian, M.; Mohamadi, M.; Sadeghi, S.; Zamani, Z.; Tabatabaie, F. Novel chloroquine loaded curcumin based anionic linear globular dendrimer G2: A metabolomics study on Plasmodium falciparum in vitro using 1H NMR spectroscopy. Parasitology 2020, 147, 747–759. [Google Scholar] [CrossRef] [PubMed]
- Diaz, C.; Guzmán, J.; Jiménez, V.A.; Alderete, J.B. Partially PEGylated PAMAM dendrimers as solubility enhancers of Silybin. Pharm. Dev. Technol. 2018, 23, 689–696. [Google Scholar] [CrossRef] [PubMed]
- Yesil-Celiktas, O.; Pala, C.; Cetin-Uyanikgil, E.O.; Sevimli-Gur, C. Synthesis of silica-PAMAM dendrimer nanoparticles as promising carriers in Neuro blastoma cells. Anal. Biochem. 2017, 519, 1–7. [Google Scholar] [CrossRef] [PubMed]
- Qu, W.J.; Li, H.F.; Su, Y.Y.; Dong, Z.Q.; Ge, Y.R. Absorption enhancing effects and safety of PAMAM dendrimers on liquiritin. Zhongguo Zhong Yao Za Zhi 2017, 42, 1766–1771. [Google Scholar] [CrossRef]
- Alfei, S.; Oliveri, P.; Malegori, C. Assessment of the efficiency of a nanospherical gallic acid Dendrimer for long-term preservation of essential oils: An integrated chemometric-assisted FTIR study. ChemistrySelect 2019, 4, 8891–8901. [Google Scholar] [CrossRef]
- Thanh, V.M.; Bui, L.M.; Bach, L.G.; Nguyen, N.T.; Le Thi, H.; Thi, T.T.H. Origanum majorana L. essential oil-associated polymeric nano dendrimer for antifungal activity against Phytophthora infestans. Materials 2019, 12, 1446. [Google Scholar] [CrossRef] [Green Version]
- Maes, C.D.; Brostaux, Y.; Bouquillon, S.; Fauconnier, M.L. Use of new glycerol-based dendrimers for essential oils encapsulation: Optimization of stirring time and rate using a plackett-burman design and a surface. Foods 2021, 10, 207. [Google Scholar] [CrossRef]
- De Angelis, R.; Venditti, I.; Fratoddi, I.; De Matteis, F.; Prosposito, P.; Cacciotti, I.; D’Amico, L.; Nanni, F.; Yadav, A.; Casalboni, M.; et al. From nanospheres to microribbons: Self-assembled Eosin Y doped PMMA nanoparticles as photonic crystals. J. Coll. Interface Sci. 2014, 414, 24–32. [Google Scholar] [CrossRef]
- Almeida, K.B.; Araujo, J.L.; Cavalcanti, J.F.; Romanos, M.T.V.; Mourão, S.C.; Amaral, A.C.F.; Falcão, D.Q. In vitro release and anti-herpetic activity of Cymbopogon citratus volatile oil-loaded nanogel. Rev. Bras. Farmacogn. 2018, 28, 495–502. [Google Scholar] [CrossRef]
- Mora-Huertas, C.E.; Fessi, H.; Elaissari, A. Polymer-based nanocapsules for drug delivery. Int. J. Pharm. 2010, 385, 113–142. [Google Scholar] [CrossRef]
- Chen, Y.; Lin, X.; Park, H.; Greever, R. Study of artemisinin nanocapsules as anticancer drug delivery systems. Nanomed. Nanotechnol. Biol. Med. 2009, 5, 316–322. [Google Scholar] [CrossRef]
- Bonifácio, B.V.; da Silva, P.B.; dos Ramos, M.A.S.; Negri, K.M.S.; Bauab, T.M.; Chorilli, M. Nanotechnology-based drug delivery systems and herbal medicines: A review. Int. J. Nanomed. 2014, 9, 1. [Google Scholar]
- Ibili, H.; Dasdemir, M.; Çankaya, İ.T.; Orhan, M.; Güneşoğlu, C.; Arabacı Anul, S. Investigation of poly(lactic acid) nanocapsules containing the plant extract via coaxial electrospraying method for functional nonwoven applications. J. Ind. Text. 2021. [Google Scholar] [CrossRef]
- Souza, C.F.; Baldissera, M.D.; Cossetin, L.F.; Dalla Lana, D.F.; Monteiro, S.G. Achyrocline satureioides essential oil loaded in nanocapsules ameliorate the antioxidant/oxidant status in heart of rats infected with Trypanosoma evansi. Microb. Pathog. 2017, 105, 30–36. [Google Scholar] [CrossRef] [PubMed]
- Martínez Rivas, C.J.; Tarhini, M.; Badri, W.; Miladi, K.; Greige-Gerges, H.; Nazari, Q.A.; Galindo Rodríguez, S.A.; Román, R.Á.; Fessi, H.; Elaissari, A. Nanoprecipitation process: From encapsulation to drug delivery. Int. J. Pharm. 2017, 532, 66–81. [Google Scholar] [CrossRef]
- Baldissera, M.D.; Souza, C.F.; Boligon, A.A.; Grando, T.H.; De Sá, M.F.; Da Silva, A.S.; Stefani, L.M.; Baldisserotto, B.; Monteiro, S.G. Solving the challenge of the blood-brain barrier to treat infections caused by Trypanosoma evansi: Evaluation of nerolidol-loaded nanospheres in mice. Parasitology 2017, 144, 1543–1550. [Google Scholar] [CrossRef]
- Pereira, K.D.C.; Quintela, E.D.; Da Silva, D.J.; Do Nascimento, V.A.; Da Rocha, D.V.M.; Arrudae Silva, J.F.; Forim, M.R.; Silva, F.G.; Cazal, C.D.M. Characterization of nanospheres containing Zanthoxylum riedelianum fruit essential oil and their insecticidal and deterrent activities against Bemisia tabaci (Hemiptera: Aleyrodidae). Molecules 2018, 23, 2052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Holz, J.P.; Bottene, M.K.; Jahno, V.D.; Einloft, S.; Ligabue, R. Menthol-loaded PLGA micro and nanospheres: Synthesis, characterization and degradation in artificial saliva. Mater. Res. 2018, 21, 17–24. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Feng, N. Nanocarriers for the delivery of active ingredients and fractions extracted from natural products used in traditional Chinese medicine (TCM). Adv. Coll. Interface Sci. 2015, 221, 60–76. [Google Scholar] [CrossRef] [PubMed]
- Wagh, A.; Singh, J.; Qian, S.; Law, B. A short circulating peptide nanofiber as a carrier for tumoral delivery. Nanomed. Nanotechnol. Biol. Med. 2013, 9, 449–457. [Google Scholar] [CrossRef]
- Wang, H.; Wei, J.; Yang, C.; Zhao, H.; Li, D.; Yin, Z.; Yang, Z. The inhibition of tumor growth and metastasis by self-assembled nanofibers of taxol. Biomaterials 2012, 33, 5848–5853. [Google Scholar] [CrossRef]
- Wang, J.; Tian, L.; He, L.; Chen, N.; Ramakrishna, S.; So, K.F.; Mo, X. Lycium barbarum polysaccharide encapsulated Poly lactic-co-glycolic acid Nanofibers: Cost effective herbal medicine for potential application in peripheral nerve tissue engineering. Sci. Rep. 2018, 8, 8669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Parvathi, K.; Krishnan, A.G.; Anitha, A.; Jayakumar, R.; Nair, M.B. Poly(L-lactic acid) nanofibers containing Cissus quadrangularis induced osteogenic differentiation in vitro. Int. J. Biol. Macromol. 2018, 110, 514–521. [Google Scholar] [CrossRef]
- Gupta, P.K.; Jaiswal, A.K.; Asthana, S.; Dube, A.; Mishra, P.R. Antigen Presenting Cells Targeting and Stimulation Potential of Lipoteichoic Acid Functionalized Lipo-Polymerosome: A Chemo-Immunotherapeutic Approach against Intracellular Infectious Disease. Biomacromolecules 2015, 16, 1073–1087. [Google Scholar] [CrossRef] [PubMed]
- Goyal, K.; Konar, A.; Kumar, B.S.H.; Koul, V. Lactoferrin-conjugated pH and redox-sensitive polymersomes based on PEG-S-S-PLA-PCL-OH boost delivery of bacosides to the brain. Nanoscale 2018, 10, 17781–17798. [Google Scholar] [CrossRef] [PubMed]
- Jia, T.T.; Sun, Z.G.; Lu, Y.; Gao, J.; Zou, H.; Xie, F.Y.; Zhang, G.Q.; Xu, H.; Sun, D.X.; Yu, Y.; et al. A dual brain-targeting curcumin-loaded polymersomes ameliorated cognitive dysfunction in intrahippocampal amyloid-β1-42-injected mice. Int. J. Nanomed. 2016, 11, 3765–3775. [Google Scholar] [CrossRef] [Green Version]
- Aswathanarayan, J.B.; Vittal, R.R. Nanoemulsions and their potential applications in food industry. Front. Sustain. Food Syst. 2019, 3, 1–21. [Google Scholar] [CrossRef] [Green Version]
- Halnor, V.; Pande, V.; Borawake, D.D.; Nagare, H. Nanoemulsion: A Novel Platform for Drug Delivery System. J. Mater. Sci. Nanotechnol. 2018, 6, 104. [Google Scholar]
- Moradi, S.; Barati, A. Essential oils nanoemulsions: Preparation, characterization and study of antibacterial activity against Escherichia coli. Int. J. Nanosci. Nanotechnol. 2019, 15, 199–210. [Google Scholar]
- Das, A.K.; Nanda, P.K.; Bandyopadhyay, S.; Banerjee, R.; Biswas, S.; McClements, D.J. Application of nanoemulsion-based approaches for improving the quality and safety of muscle foods: A comprehensive review. Compr. Rev. Food Sci. Food Saf. 2020, 19, 2677–2700. [Google Scholar] [CrossRef]
- Rao, J.; Chen, B.; McClements, D.J. Improving the Efficacy of Essential Oils as Antimicrobials in Foods: Mechanisms of Action. Annu. Rev. Food Sci. Technol. 2019, 10, 365–387. [Google Scholar] [CrossRef]
- Anton, N.; Vandamme, T.F. The universality of low-energy nano-emulsification. Int. J. Pharm. 2009, 377, 142–147. [Google Scholar] [CrossRef]
- Harwansh, R.K.; Deshmukh, R.; Rahman, M.A. Nanoemulsion: Promising nanocarrier system for delivery of herbal bioactives. J. Drug Deliv. Sci. Technol. 2019, 51, 224–233. [Google Scholar] [CrossRef]
- Komaiko, J.S.; Mcclements, D.J. Formation of food-grade nanoemulsions using low-energy preparation methods: A review of available methods. Compr. Rev. Food Sci. Food Saf. 2016, 15, 331–352. [Google Scholar] [CrossRef] [PubMed]
- Jaiswal, M.; Dudhe, R.; Sharma, P.K. Nanoemulsion: An advanced mode of drug delivery system. 3 Biotech 2015, 5, 123–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qi, J.; Zhuang, J.; Wu, W.; Lu, Y.; Song, Y.; Zhang, Z.; Jia, J.; Ping, Q. Enhanced effect and mechanism of water-in-oil microemulsion as an oral delivery system of hydroxysafflor yellow A. Int. J. Nanomed. 2011, 6, 985–991. [Google Scholar] [CrossRef] [Green Version]
- Censi, R.; Martena, V.; Hoti, E.; Malaj, L.; Di Martino, P. Permeation and skin retention of quercetin from microemulsions containing Transcutol® P. Drug Dev. Ind. Pharm. 2012, 38, 1128–1133. [Google Scholar] [CrossRef]
- Bose, S.; Du, Y.; Takhistov, P.; Michniak-Kohn, B. Formulation optimization and topical delivery of quercetin from solid lipid based nanosystems. Int. J. Pharm. 2013, 441, 56–66. [Google Scholar] [CrossRef]
- Tran, T.H.; Guo, Y.; Song, D.; Bruno, R.S.; Lu, X. Quercetin-containing self-nanoemulsifying drug delivery system for improving oral bioavailability. J. Pharm. Sci. 2014, 103, 840–852. [Google Scholar] [CrossRef]
- Guo, C.Y.; Yang, C.F.; Li, Q.L.; Tan, Q.; Xi, Y.W.; Liu, W.N.; Zhai, G.X. Development of a Quercetin-loaded nanostructured lipid carrier formulation for topical delivery. Int. J. Pharm. 2012, 430, 292–298. [Google Scholar] [CrossRef]
- Zeng, Z.; Zhou, G.; Wang, X.; Huang, E.Z.; Zhan, X.; Liu, J.; Wang, S.; Wang, A.; Li, H.; Pei, X.; et al. Preparation, characterization and relative bioavailability of oral elemene o/w microemulsion. Int. J. Nanomed. 2010, 5, 567–572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, T.; Dong, D.; Lu, D.; Wang, S.; Wu, B. Cremophor EL-based nanoemulsion enhances transcellular permeation of emodin through glucuronidation reduction in UGT1A1-overexpressing MDCKII cells. Int. J. Pharm. 2016, 501, 190–198. [Google Scholar] [CrossRef]
- Harwansh, R.K.; Mukherjee, P.K.; Kar, A.; Bahadur, S.; Al-Dhabi, N.A.; Duraipandiyan, V. Enhancement of photoprotection potential of catechin loaded nanoemulsion gel against UVA induced oxidative stress. J. Photochem. Photobiol. B Biol. 2016, 160, 318–329. [Google Scholar] [CrossRef]
- Harwansh, R.K.; Mukherjee, P.K.; Biswas, S. Nanoemulsion as a novel carrier system for improvement of betulinic acid oral bioavailability and hepatoprotective activity. J. Mol. Liq. 2017, 237, 361–371. [Google Scholar] [CrossRef]
- Nazari-Vanani, R.; Moezi, L.; Heli, H. In vivo evaluation of a self-nanoemulsifying drug delivery system for curcumin. Biomed. Pharmacother. 2017, 88, 715–720. [Google Scholar] [CrossRef] [PubMed]
- Gao, T.; Wei, Z.; Zhao, Y. In vitro and in vivo anti-tumor characterizations of β-elemene-loaded nanoemulsion. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 513. [Google Scholar] [CrossRef]
- Bhargava, K.; Conti, D.S.; da Rocha, S.R.P.; Zhang, Y. Application of an oregano oil nanoemulsion to the control of foodborne bacteria on fresh lettuce. Food Microbiol. 2015, 47, 69–73. [Google Scholar] [CrossRef]
- Ghosh, V.; Mukherjee, A.; Chandrasekaran, N. Ultrasonic emulsification of food-grade nanoemulsion formulation and evaluation of its bactericidal activity. Ultrason. Sonochem. 2013, 20, 338–344. [Google Scholar] [CrossRef]
- Wang, X.; Wang, Y.W.; Huang, Q. Enhancing stability and oral bioavailability of polyphenols using nanoemulsions. ACS Symp. Ser. 2009, 1007, 198–212. [Google Scholar] [CrossRef]
- Rathore, S.; Mukim, M.; Sharma, P.; Devi, S.; Chandra Nagar, J.; Khalid, M. Curcumin: A review for health benefits. Int. J. Res. Rev. 2020, 7, 1. [Google Scholar]
- Sari, T.P.; Mann, B.; Kumar, R.; Singh, R.R.B.; Sharma, R.; Bhardwaj, M.; Athira, S. Preparation and characterization of nanoemulsion encapsulating curcumin. Food Hydrocoll. 2015, 43, 540–546. [Google Scholar] [CrossRef]
- Artiga-Artigas, M.; Lanjari-Pérez, Y.; Martín-Belloso, O. Curcumin-loaded nanoemulsions stability as affected by the nature and concentration of surfactant. Food Chem. 2018, 266, 466–474. [Google Scholar] [CrossRef] [PubMed]
- Ma, P.; Zeng, Q.; Tai, K.; He, X.; Yao, Y.; Hong, X.; Yuan, F. Development of stable curcumin nanoemulsions: Effects of emulsifier type and surfactant-to-oil ratios. J. Food Sci. Technol. 2018, 55, 3485–3497. [Google Scholar] [CrossRef] [PubMed]
- Kharat, M.; Aberg, J.; Dai, T.; McClements, D.J. Comparison of emulsion and nanoemulsion delivery systems: The chemical stability of curcumin decreases as oil droplet size decreases. J. Agric. Food Chem. 2020, 68, 9205–9212. [Google Scholar] [CrossRef] [PubMed]
- Kumar, R.; Kaur, K.; Uppal, S.; Mehta, S.K. Ultrasound processed nanoemulsion: A comparative approach between resveratrol and resveratrol cyclodextrin inclusion complex to study its binding interactions, antioxidant activity and UV light stability. Ultrason. Sonochem. 2017, 37, 478–489. [Google Scholar] [CrossRef] [PubMed]
- McClements, D.J.; Decker, E.A.; Weiss, J. Emulsion-based delivery systems for lipophilic bioactive components. J. Food Sci. 2007, 72, 109–124. [Google Scholar] [CrossRef] [PubMed]
- Chuanxun, Y.; Xueru, Z.; Risheng, J. Development and characterization of phytosterol nanoemulsions and self-microemulsifying drug delivery systems. bioRxiv 2019, 585166. [Google Scholar] [CrossRef]
- Acevedo-Estupiñan, M.V.; Gutierrez-Lopez, G.F.; Cano-Sarmiento, C.; Parra-Escudero, C.O.; Rodriguez-Estrada, M.T.; Garcia-Varela, R.; García, H.S. Stability and characterization of O/W free phytosterols nanoemulsions formulated with an enzymatically modified emulsifier. LWT 2019, 107, 151–157. [Google Scholar] [CrossRef]
- Borba, C.M.; Tavares, M.N.; Macedo, L.P.; Araújo, G.S.; Furlong, E.B.; Dora, C.L.; Burkert, J.F.M. Physical and chemical stability of β-carotene nanoemulsions during storage and thermal process. Food Res. Int. 2019, 121, 229–237. [Google Scholar] [CrossRef]
- Qian, C.; Decker, E.A.; Xiao, H.; McClements, D.J. Physical and chemical stability of β-carotene-enriched nanoemulsions: Influence of pH, ionic strength, temperature, and emulsifier type. Food Chem. 2012, 132, 1221–1229. [Google Scholar] [CrossRef]
- Teixeira, M.C.; Severino, P.; Andreani, T.; Boonme, P.; Santini, A.; Silva, A.M.; Souto, E.B. D-α-tocopherol nanoemulsions: Size properties, rheological behavior, surface tension, osmolarity and cytotoxicity. Saudi Pharm. J. 2017, 25, 231–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pangeni, R.; Kang, S.W.; Oak, M.; Park, E.Y.; Park, J.W. Oral delivery of quercetin in oil-in-water nanoemulsion: In vitro characterization and in vivo anti-obesity efficacy in mice. J. Funct. Foods 2017, 38, 571–581. [Google Scholar] [CrossRef]
- de Carli, C.; Moraes-Lovison, M.; Pinho, S.C. Production, physicochemical stability of quercetin-loaded nanoemulsions and evaluation of antioxidant activity in spreadable chicken pâtés. LWT 2018, 98, 154–161. [Google Scholar] [CrossRef]
- Vaz, G.R.; Clementino, A.; Bidone, J.; Villetti, M.A.; Falkembach, M.; Batista, M.; Barros, P.; Sonvico, F.; Dora, C. Curcumin and quercetin-loaded nanoemulsions: Physicochemical compatibility study and validation of a simultaneous quantification method. Nanomaterials 2020, 10, 1650. [Google Scholar] [CrossRef]
- Iqbal, R.; Mehmood, Z.; Baig, A.; Khalid, N. Formulation and characterization of food grade O/W nanoemulsions encapsulating quercetin and curcumin: Insights on enhancing solubility characteristics. Food Bioprod. Process. 2020, 123, 304–311. [Google Scholar] [CrossRef]
- Son, H.Y.; Lee, M.S.; Chang, E.; Kim, S.Y.; Kang, B.; Ko, H.; Kim, I.H.; Zhong, Q.; Jo, Y.H.; Kim, C.T.; et al. Formulation and characterization of quercetin-loaded oil in water nanoemulsion and evaluation of hypocholesterolemic activity in rats. Nutrients 2019, 11, 244. [Google Scholar] [CrossRef] [Green Version]
- Das, S.S.; Verma, P.R.P.; Singh, S.K. Screening and preparation of quercetin doped nanoemulsion: Characterizations, antioxidant and anti-bacterial activities. LWT 2020, 124, 109141. [Google Scholar] [CrossRef]
- Oliveira, A.E.M.F.M.; Duarte, J.L.; Amado, J.R.R.; Cruz, R.A.S.; Rocha, C.F.; Souto, R.N.P.; Ferreira, R.M.A.; Santos, K.; Da Conceição, E.C.; De Oliveira, L.A.R.; et al. Development of α larvicidal nanoemulsion with pterodon emarginatus Vogel oil. PLoS ONE 2016, 11, e0145835. [Google Scholar] [CrossRef] [PubMed]
- Mulia, K.; Putri, G.A.; Krisanti, E. Encapsulation of mangosteen extract in virgin coconut oil based nanoemulsions: Preparation and characterization for topical formulation. Mater. Sci. Forum 2018, 929, 234–242. [Google Scholar] [CrossRef]
- Ghazy, O.A.; Fouad, M.T.; Saleh, H.H.; Kholif, A.E.; Morsy, T.A. Ultrasound-assisted preparation of anise extract nanoemulsion and its bioactivity against different pathogenic bacteria. Food Chem. 2021, 341, 128259. [Google Scholar] [CrossRef]
- Chen, B.H.; Inbaraj, B.S. Nanoemulsion and nanoliposome based strategies for improving anthocyanin stability and bioavailability. Nutrients 2019, 11, 1052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Devaraj, A.; Mahalingam, G. Nanoemulsion formulation as an effective therapeutic drug delivery system in diabetes mellitus. Nanotechnol. Life Sci. 2020, 317–329. [Google Scholar] [CrossRef]
- Vallet-Regí, M.; Colilla, M.; Izquierdo-Barba, I.; Manzano, M. Mesoporous silica nanoparticles for drug delivery: Current insights. Molecules 2018, 23, 47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dahoumane, S.A.; Jeffryes, C.; Mechouet, M.; Agathos, S.N. Biosynthesis of inorganic nanoparticles: A fresh look at the control of shape, size and composition. Bioengineering 2017, 4, 14. [Google Scholar] [CrossRef] [Green Version]
- Md Ishak, N.A.I.; Kamarudin, S.K.; Timmiati, S.N. Green synthesis of metal and metal oxide nanoparticles via plant extracts: An overview. Mater. Res. Express 2019, 6, 112004. [Google Scholar] [CrossRef]
- El-Seedi, H.R.; El-Shabasy, R.M.; Khalifa, S.A.M.; Saeed, A.; Shah, A.; Shah, R.; Iftikhar, F.J.; Abdel-Daim, M.M.; Omri, A.; Hajrahand, N.H.; et al. Metal nanoparticles fabricated by green chemistry using natural extracts: Biosynthesis, mechanisms, and applications. RSC Adv. 2019, 9, 24539–24559. [Google Scholar] [CrossRef] [Green Version]
- Iravani, S. Green synthesis of metal nanoparticles using plants. Green Chem. 2011, 13, 2638–2650. [Google Scholar] [CrossRef]
- Yadi, M.; Mostafavi, E.; Saleh, B.; Davaran, S.; Aliyeva, I.; Khalilov, R.; Nikzamir, M.; Nikzamir, N.; Akbarzadeh, A.; Panahi, Y.; et al. Current developments in green synthesis of metallic nanoparticles using plant extracts: A review. Artif. Cells Nanomed. Biotechnol. 2018, 46, S336–S343. [Google Scholar] [CrossRef] [Green Version]
- Marinheiro, D.; Ferreira, B.J.M.L.; Oskoei, P.; Oliveira, H.; Daniel-da-silva, A.L. Encapsulation and enhanced release of resveratrol from mesoporous silica nanoparticles for melanoma therapy. Materials 2021, 14, 1382. [Google Scholar] [CrossRef] [PubMed]
- Chaudhary, Z.; Subramaniam, S.; Khan, G.M.; Abeer, M.M.; Qu, Z.; Janjua, T.; Kumeria, T.; Batra, J.; Popat, A. Encapsulation and controlled release of resveratrol within functionalized mesoporous silica nanoparticles for prostate cancer therapy. Front. Bioeng. Biotechnol. 2019, 7, 225. [Google Scholar] [CrossRef]
- Elbialy, N.S.; Aboushoushah, S.F.; Sofi, B.F.; Noorwali, A. Multifunctional curcumin-loaded mesoporous silica nanoparticles for cancer chemoprevention and therapy. Microporous Mesoporous Mater. 2020, 291, 109540. [Google Scholar] [CrossRef]
- Wu, C.; Zhu, Y.; Wu, T.; Wang, L.; Yuan, Y.; Chen, J.; Hu, Y.; Pang, J. Enhanced functional properties of biopolymer film incorporated with curcurmin-loaded mesoporous silica nanoparticles for food packaging. Food Chem. 2019, 288, 139–145. [Google Scholar] [CrossRef] [PubMed]
- Melendez-Rodriguez, B.; Figueroa-Lopez, K.J.; Bernardos, A.; Martínez-Máñez, R.; Cabedo, L.; Torres-Giner, S.; Lagaron, J.M. Electrospun antimicrobial films of poly(3-hydroxybutyrate-co-3-hydroxyvalerate) containing eugenol essential oil encapsulated in mesoporous silica nanoparticles. Nanomaterials 2019, 9, 227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mai, Z.; Chen, J.; Hu, Y.; Liu, F.; Fu, B.; Zhang, H.; Dong, X.; Huang, W.; Zhou, W. Novel functional mesoporous silica nanoparticles loaded with Vitamin E acetate as smart platforms for pH responsive delivery with high bioactivity. J. Coll. Interface Sci. 2017, 508, 184–195. [Google Scholar] [CrossRef] [PubMed]
- Bravo Cadena, M.; Preston, G.M.; Van der Hoorn, R.A.L.; Townley, H.E.; Thompson, I.P. Species-specific antimicrobial activity of essential oils and enhancement by encapsulation in mesoporous silica nanoparticles. Ind. Crops Prod. 2018, 122, 582–590. [Google Scholar] [CrossRef]
- Mahmoudi, M.; Sant, S.; Wang, B.; Laurent, S.; Sen, T. Superparamagnetic iron oxide nanoparticles (SPIONs): Development, surface modification and applications in chemotherapy. Adv. Drug Deliv. Rev. 2011, 63, 24–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sallem, F.; Haji, R.; Vervandier-Fasseur, D.; Nury, T.; Maurizi, L.; Boudon, J.; Lizard, G.; Millot, N. Elaboration of trans-resveratrol derivative-loaded superparamagnetic iron oxide nanoparticles for glioma treatment. Nanomaterials 2019, 9, 287. [Google Scholar] [CrossRef] [Green Version]
- Khosravi, A.; Hasani, A.; Rahimi, K.; Aliaghaei, A.; Pirani, M.; Azad, N.; Ramezani, F.; Tamimi, A.; Behnam, P.; Raoofi, A.; et al. Ameliorating effects of curcumin-loaded superparamagnetic iron oxide nanoparticles (SPIONs) on the mouse testis exposed to the transient hyperthermia: A molecular and stereological study. Acta Histochem. 2020, 122, 151632. [Google Scholar] [CrossRef]
- Enteshari Najafabadi, R.; Kazemipour, N.; Esmaeili, A.; Beheshti, S.; Nazifi, S. Using superparamagnetic iron oxide nanoparticles to enhance bioavailability of quercetin in the intact rat brain. BMC Pharmacol. Toxicol. 2018, 19, 59. [Google Scholar] [CrossRef]
- McClements, D.J.; Xiao, H. Is nano safe in foods? Establishing the factors impacting the gastrointestinal fate and toxicity of organic and inorganic food-grade nanoparticles. npj Sci. Food 2017, 1, 6. [Google Scholar] [CrossRef]
- Kokila, T.; Ramesh, P.S.; Geetha, D. Biosynthesis of silver nanoparticles from Cavendish banana peel extract and its antibacterial and free radical scavenging assay: A novel biological approach. Appl. Nanosci. 2015, 5, 911–920. [Google Scholar] [CrossRef] [Green Version]
- Logaranjan, K.; Raiza, A.J.; Gopinath, S.C.B.; Chen, Y.; Pandian, K. Shape- and Size-Controlled Synthesis of Silver Nanoparticles Using Aloe vera Plant Extract and Their Antimicrobial Activity. Nanoscale Res. Lett. 2016, 11, 520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tippayawat, P.; Phromviyo, N.; Boueroy, P.; Chompoosor, A. Green synthesis of silver nanoparticles in aloe vera plant extract prepared by a hydrothermal method and their synergistic antibacterial activity. PeerJ 2016, 4, e2589. [Google Scholar] [CrossRef] [PubMed]
- Jayaprakash, N.; Vijaya, J.J.; Kaviyarasu, K.; Kombaiah, K.; Kennedy, L.J.; Ramalingam, R.J.; Munusamy, M.A.; Al-Lohedan, H.A. Green synthesis of Ag nanoparticles using Tamarind fruit extract for the antibacterial studies. J. Photochem. Photobiol. B Biol. 2017, 169, 178–185. [Google Scholar] [CrossRef] [PubMed]
- Premkumar, J.; Sudhakar, T.; Dhakal, A.; Shrestha, J.B.; Krishnakumar, S.; Balashanmugam, P. Synthesis of silver nanoparticles (AgNPs) from cinnamon against bacterial pathogens. Biocatal. Agric. Biotechnol. 2018, 15, 311–316. [Google Scholar] [CrossRef]
- Vélez, E.; Campillo, G.; Morales, G.; Hincapié, C.; Osorio, J.; Arnache, O. Silver nanoparticles obtained by aqueous or ethanolic aloe Vera extracts: An assessment of the antibacterial activity and mercury removal capability. J. Nanomater. 2018, 2018. [Google Scholar] [CrossRef] [Green Version]
- Haghparasti, Z.; Mahdavi Shahri, M. Green synthesis of water-soluble nontoxic inorganic polymer nanocomposites containing silver nanoparticles using white tea extract and assessment of their in vitro antioxidant and cytotoxicity activities. Mater. Sci. Eng. C 2018, 87, 139–148. [Google Scholar] [CrossRef]
- Govindan, L.; Anbazhagan, S.; Altemimi, A.B. Efficacy of antimicrobial and larvicidal activities of green synthesized silver nanoparticles using leaf extract of plumbago auriculata lam. Plants 2020, 9, 1577. [Google Scholar] [CrossRef] [PubMed]
- Alkhulaifi, M.M.; Alshehri, J.H.; Alwehaibi, M.A.; Awad, M.A.; Al-Enazi, N.M.; Aldosari, N.S.; Hatamleh, A.A.; Abdel- Raouf, N. Green synthesis of silver nanoparticles using Citrus limon peels and evaluation of their antibacterial and cytotoxic properties. Saudi J. Biol. Sci. 2020, 27, 3434–3441. [Google Scholar] [CrossRef]
- Jaiswal, S.; Mishra, P. Antimicrobial and antibiofilm activity of curcumin-silver nanoparticles with improved stability and selective toxicity to bacteria over mammalian cells. Med. Microbiol. Immunol. 2018, 207, 39–53. [Google Scholar] [CrossRef] [PubMed]
- Alsammarraie, F.K.; Wang, W.; Zhou, P.; Mustapha, A.; Lin, M. Green synthesis of silver nanoparticles using turmeric extracts and investigation of their antibacterial activities. Coll. Surf. B Biointerfaces 2018, 171, 398–405. [Google Scholar] [CrossRef]
- Khatoon, A.; Khan, F.; Ahmad, N.; Shaikh, S.; Rizvi, S.M.D.; Shakil, S.; Al-Qahtani, M.H.; Abuzenadah, A.M.; Tabrez, S.; Ahmed, A.B.F.; et al. Silver nanoparticles from leaf extract of Mentha piperita: Eco-friendly synthesis and effect on acetylcholinesterase activity. Life Sci. 2018, 209, 430–434. [Google Scholar] [CrossRef]
- Biswal, A.K.; Misra, P.K. Biosynthesis and characterization of silver nanoparticles for prospective application in food packaging and biomedical fields. Mater. Chem. Phys. 2020, 250, 123014. [Google Scholar] [CrossRef]
- Ozdal, Z.D.; Sahmetlioglu, E.; Narin, I.; Cumaoglu, A. Synthesis of gold and silver nanoparticles using flavonoid quercetin and their effects on lipopolysaccharide induced inflammatory response in microglial cells. 3 Biotech 2019, 9, 212. [Google Scholar] [CrossRef]
- Mata, R.; Bhaskaran, A.; Sadras, S.R. Green-synthesized gold nanoparticles from Plumeria alba flower extract to augment catalytic degradation of organic dyes and inhibit bacterial growth. Particuology 2016, 24, 78–86. [Google Scholar] [CrossRef]
- Mishra, P.; Ray, S.; Sinha, S.; Das, B.; Khan, M.I.; Behera, S.K.; Il Yun, S.; Tripathy, S.K.; Mishra, A. Facile bio-synthesis of gold nanoparticles by using extract of Hibiscus sabdariffa and evaluation of its cytotoxicity against U87 glioblastoma cells under hyperglycemic condition. Biochem. Eng. J. 2016, 105, 264–272. [Google Scholar] [CrossRef]
- Rodríguez-León, E.; Rodríguez-Vázquez, B.E.; Martínez-Higuera, A.; Rodríguez-Beas, C.; Larios-Rodríguez, E.; Navarro, R.E.; López-Esparza, R.; Iñiguez-Palomares, R.A. Synthesis of gold nanoparticles using mimosa tenuiflora extract, assessments of cytotoxicity, cellular uptake, and catalysis. Nanoscale Res. Lett. 2019, 14, 334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thipe, V.C.; Amiri, K.P.; Bloebaum, P.; Karikachery, A.R.; Khoobchandani, M.; Katti, K.K.; Jurisson, S.S.; Katti, K.V. Development of resveratrol-conjugated gold nanoparticles: Interrelationship of increased resveratrol corona on anti-tumor efficacy against breast, pancreatic and prostate cancers. Int. J. Nanomed. 2019, 14, 4413–4428. [Google Scholar] [CrossRef] [Green Version]
- Zangeneh, M.M.; Zangeneh, A. Novel green synthesis of Hibiscus sabdariffa flower extract conjugated gold nanoparticles with excellent anti-acute myeloid leukemia effect in comparison to daunorubicin in a leukemic rodent model. Appl. Organomet. Chem. 2020, 34, e5271. [Google Scholar] [CrossRef]
- Nasrollahzadeh, M.; Sajadi, S.M.; Maham, M. Green synthesis of palladium nanoparticles using Hippophae rhamnoides Linn leaf extract and their catalytic activity for the Suzuki-Miyaura coupling in water. J. Mol. Catal. A Chem. 2015, 396, 297–303. [Google Scholar] [CrossRef]
- Majumdar, R.; Tantayanon, S.; Bag, B.G. Synthesis of palladium nanoparticles with leaf extract of Chrysophyllum cainito (Star apple) and their applications as efficient catalyst for C–C coupling and reduction reactions. Int. Nano Lett. 2017, 7, 267–274. [Google Scholar] [CrossRef] [Green Version]
- Ahmadi, R.; Tanomand, A.; Kazeminava, F.; Kamounah, F.S.; Ayaseh, A.; Ganbarov, K.; Yousefi, M.; Katourani, A.; Yousefi, B.; Kafil, H.S. Fabrication and characterization of a titanium dioxide (TiO2) nanoparticles reinforced bio-nanocomposite containing miswak (salvadora persica L.) extract—The antimicrobial, thermo-physical and barrier properties. Int. J. Nanomed. 2019, 14, 3439–3454. [Google Scholar] [CrossRef] [Green Version]
- Santhoshkumar, J.; Kumar, S.V.; Rajeshkumar, S. Synthesis of zinc oxide nanoparticles using plant leaf extract against urinary tract infection pathogen. Resour. Technol. 2017, 3, 459–465. [Google Scholar] [CrossRef]
- Naseer, M.; Aslam, U.; Khalid, B.; Chen, B. Green route to synthesize Zinc Oxide Nanoparticles using leaf extracts of Cassia fistula and Melia azadarach and their antibacterial potential. Sci. Rep. 2020, 10, 44. [Google Scholar] [CrossRef]
- Alamdari, S.; Ghamsari, M.S.; Lee, C.; Han, W.; Park, H.H.; Tafreshi, M.J.; Afarideh, H.; Ara, M.H.M. Preparation and characterization of zinc oxide nanoparticles using leaf extract of sambucus ebulus. Appl. Sci. 2020, 10, 3620. [Google Scholar] [CrossRef]
- Selim, Y.A.; Azb, M.A.; Ragab, I.H.M.; Abd El-Azim, M. Green synthesis of zinc oxide nanoparticles using aqueous extract of deverra tortuosa and their cytotoxic activities. Sci. Rep. 2020, 10, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Chung, Y.H.; Cai, H.; Steinmetz, N.F. Viral nanoparticles for drug delivery, imaging, immunotherapy, and theranostic applications. Adv. Drug Deliv. Rev. 2020, 156, 214–235. [Google Scholar] [CrossRef]
- Shahgolzari, M.; Pazhouhandeh, M.; Milani, M.; Yari Khosroushahi, A.; Fiering, S. Plant viral nanoparticles for packaging and in vivo delivery of bioactive cargos. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2020, 12, e1629. [Google Scholar] [CrossRef]
- Gardikis, K.; Signorelli, M.; Ferrario, C.; Schiraldi, A.; Fortina, M.G.; Hatziantoniou, S.; Demetzos, C.; Fessas, D. Microbial biosensors to monitor the encapsulation effectiveness of doxorubicin in chimeric advanced drug delivery nano systems: A calorimetric approach. Int. J. Pharm. 2017, 516, 178–184. [Google Scholar] [CrossRef]
- Demetzos, C.; Pippa, N. Advanced drug delivery nanosystems (aDDnSs): A mini-review. Drug Deliv. 2014, 21, 250–257. [Google Scholar] [CrossRef]
Type of Lipidic Carrier | Encapsulated Material | Target of Encapsulation | Size of the Obtained Delivery System | Application | Reference |
---|---|---|---|---|---|
Liposome | Quercetin | Solubility | 107–139 nm | Oxidative stress and enhanced internalization by cells | [56] |
Liposome | Quercetin | Solubility | 75–150 nm | Antioxidant activity and stability | [55] |
Liposome | Curcumin | Solubility, stability and biocompatibility | 350–600 nm | Antioxidant activity and stability | [58] |
Liposome | Quercetin | Solubility | 50–300 nm | Anticancer and treatment of glioma | [59] |
Liposome | Curcumin | Solubility, stability and bioavailability | 100–200 nm | Anti-inflammatory activity, sustained-release properties and increased antioxidant activity | [60] |
Liposome | Curcumin | Solubility | 200 nm | Antioxidant activity and anti-inflammatory | [61] |
Liposome | Curcumin | Solubility | 182.4 ± 89.2 nm | Anti-inflammatory | [62] |
Liposome | Curcumin | Bioavailability | 147 ± 6 nm | Wound healing, antibacterial activity and biocompatibility | [63] |
Liposome | Curcumin | Solubility and bioavailability | 121.81 ± 9.78 nm | Hepatoprotective | [64] |
Liposome | Curcumin | Solubility and bioavailability | 82.37 ± 2.19–92.42 ± 4.56 nm | Anticancer (skin) | [65] |
Liposome | Curcumin | Bioavailability and stability | 51.75–140.35 nm | Anticancer (skin) | [66] |
Liposome | Curcumin | Bioavailability | >270 nm | Anticancer | [68] |
Liposome | Curcumin | Solubility and delivery | 420–600 nm | Anticancer (cytotoxicity in lung and colon cancer) | [69] |
PEGylated liposomes | Resveratrol | Stability and biocompatibility | 86 ± 2.7–171 ± 27.8 nm | Oxidative stress (in vitro and ex vivo) | [46] |
Liposome | Resveratrol | Bioavailability and solubility | 182.3 ± 12.1–211.2 ± 0.8 nm | Anticancer (brain) | [73] |
Liposome | Resveratrol | Solubility and bioavailability | 206 ± 10–225 ± 10nm | Antioxidant activity and anti-inflammatory | [74] |
Liposome | O. stamineus extract | Solubility | 152.5 ± 1.1 nm | Antioxidant activity | [78] |
Liposome | Green tea polyphenols | Stability, bioavailability and biotransformation | 64.5–252 nm | Antioxidant activity and controlled release | [79] |
Liposome (soy lecithin liposomes) | Green tea polyphenols (catechin and epigallocatechin gallate) | Stability and shelf- life | - a | Stability | [80] |
Liposome | Curcumin | Solubility and bioavailability | 45–130 nm | Anticancer (brain) | [71] |
Liposome | P. notoginseng saponins | Bioavailability, stability and in vitro release | 337.8 ± 40.2–117.1 ± 9.7 nm | Edema of brain and reduce the infarct volume | [82] |
Liposome | P. notoginseng saponins | Bioavailability | 40nm | Absorption from intestinal tract in rats | [83] |
Liposome | H. sabdariffa extract | Stability | 46 nm | Higher oxidative stability | [84] |
Transfersomes | Caffeine and minoxidil | Stability and release | - a | Alopecia | [96] |
Transfersomes | Apigenin | Stability and release | 35.41 nm | Skin cancer | [97] |
Transfersomes | Epigallocatechin-3-gallate (from C. sinensis) and hyaluronic | Solubility and stability | 101.2 ± 6.0 nm | Antioxidant and anti-aging properties (antioxidant and anti-aging effects in UV radiation induced skin damage) | [163] |
Ethosomes | Caffeic acid | Stability | 200 nm | Antioxidant | [99] |
Ethosomes | Ginsenoside from P. ginseng | Delivery | 108.5 to 322.9 nm | Enhanced skin permeation, retention and deposition in vitro | [100] |
Νiosomes | Herbal constituents | Solubility, bioavailability, controlled release and stability | - a | Blood–brain barrier targeted delivery | [104] |
Νiosomes | M. communis | Solubility and permeability | 5.3 ± 0.3 to 15.9 ± 2.2 μm | Antimicrobial activity | [105] |
Νiosomes | Flavonoid morusin | Solubility and controlled release | 400–500 nm (479 nm) | Antimicrobial activity | [106] |
Liposomes | Apigenin | Bioavailability | 304.10–361.46 nm | Anti-inflammatory | [164] |
Nanocrystals | Apigenin | Bioavailability | 439 ± 20 nm | Antioxidant activity | [165] |
Solid Lipid Nanoparticles | Epigallocatechin-3-gallate (EGCG) | Biocompatibility and toxicity | 144–134 nm | Antiproliferative effect | [135] |
NLC | Silymarin | Bioavailability, controlled release | 213.6 ± 16.0 nm | Used as model | [152] |
NLC and SLN | Quercetin | Bioavailability, loading efficiency | 67.46–74.61 nm | Brain cancer | [154] |
NLC | Curcumin | Cell penetration | 100–1250 nm | Breast cancer | [155] |
NLC | Curcumin | In vitro digestion, controlled release | 225.8 ± 2.3 nm | Used as model | [156] |
NLC | Curcumin | In vivo antiplasmodial activity, controlled release | 145 nm | Malaria | [157] |
NLC | Curcumin and partially hydrolyzed ginsenoside | Bioavailability, controlled release | 150–200 nm | Used as model | [158] |
NLC | H. sabdariffa extract | Bioavailability, encapsulation efficiency, stability | 470 ± 8–344 ± 12 nm | Used as model | [159] |
NLC | Cinnamon essential oil | Protection and stability | 100 ± 1–120 ± 10 nm | Food beverages | [160] |
NLC | Peppermint essential oil | Bioavailability, protection | 40–250 nm | Antimicrobial, wound healing | [161] |
NLC | Sucupira essential oil | Controlled release | 148.1 ± 1 nm | Diabetes mellitus | [162] |
Carbohydrate as Wall Material | Carbohydrate Origin and Characteristics | Core Material | Encapsulation Process | Type of the Obtained Delivery System | Morphological Characteristics of the Obtained Delivery System | Application | Reference |
---|---|---|---|---|---|---|---|
Starch | Starch from water chestnut seeds, horse chestnut seeds and lotus stem | Resveratrol | Ultrasonication method | Nanocapsules | 419, 797 and 691 nm, increased amorphous character |
| [227] |
Starch | Starch from horse chestnut, water chestnut and lotus stem | Catechin | Ultrasonication | Nanoparticles | 322.7, 559.2 and 615.6 nm |
| [228] |
Starch | Starch from pea, corn and potato | Quercetin (standard) | Nanoprecipitation | Nanoparticles | Non-uniformly shaped and nanofiber-like nanoparticles (500 nm) from pea, corn and potato starch, respectively |
| [229] |
Starch | High-amylose corn starch with 70% amylose and low-amylose potato starch | Vitamin D3 | Ultrasonication | Nanoparticles | 32.0–99.2 nm |
| [230] |
Starch | Modified (extruded) | H. sabdariffa extract | Spray drying | Microparticles | Oval or round, <10 μm |
| [231] |
Starch | Modified from rice starch | Anthocyanin extract from purple rice bran | Spray drying | Microparticles | Spherical, 6.4 μm |
| [232] |
Starch | Dafozhi, damaling and daguo starches (amylose contents of 33.5%, 26.7% and 29.8%, respectively) | G. biloba extracts | Nanoprecipitation | Nanospheres | Spherical, 255–396 nm |
| [233] |
β-Cyclodextrin | β-Cyclodextrin (purity 98%) | Curcumin | Inclusion complexation | Particles | 2–3 µm |
| [234] |
β-Cyclodextrin | Methylated-β-cyclodextrin, Mw = 1191 Da | Resveratrol | Inclusion complexation | Particles | Irregular shape |
| [235] |
β-Cyclodextrin with β-glucan | - a | Saffron anthocyanins | Spray drying | Microcapsules | Irregular shape, <124 µm |
| [236] |
Maltodextrin | Maltodextrin | Saffron aqueous extract | Nano-spray drying | Nanoparticles | Spherical, 1.5–4.2 µm |
| [237] |
Maltodextrin | Commercial maltodextrin, 4-7 DE | Pineapple peel hydroalcoholic extract | Spray drying | Microparticles | Spherical, 18.2 µm |
| [238] |
Chitosan | Low molecular weight chitosan | Curcumin | Ionic gelation | Nanoparticles | Spherical, 167.3–251.5 nm |
| [239] |
Chitosan and pectin | Low molecular weight chitosan from shrimp (deacetylation degree 94.87%) and commercial grade low-methoxy pectin from citrus peel (degree of esterification 2.9%) | Garlic and holy basil essential oils | Ionic gelation | Hydrogel beads | Globular, smooth bead surface, 1.65–2.86 mm |
| [240] |
Chitosan and gum Arabic | Deacetylation degree 93% | Curcumin | Polyelectrolyte complexation | Nanoparticles | Spherical and smooth, 250–290 nm |
| [241] |
Chitosan | Medium molecular weight chitosan (deacetylation degree 75–85%) | Cardamom essential oil | Ionic gelation | Nanoparticles | 50–100 nm |
| [242] |
Chitosan | Medium molecular weight chitosan (deacetylation degree 75–85%) | Lime essential oil | Nanoprecipitation | Nanoparticles | Spherical, 6.1 ± 0.4 nm |
| [243] |
Chitosan | Medium molecular weight chitosan (deacetylation degree 84.8%) | Peppermint and green tea essential oils | Emulsification-ionic gelation | Nanoparticles | Spherical, 20–60 nm |
| [244] |
Chitosan | Medium molecular weight chitosan (deacetylation degree 75–85%) | Mentha piperita essential oil | Sol-gel method | Nanogel | 567.1–575.6 nm |
| [245] |
Pectin and zein | Citrus peel pectin | Resveratrol | Antisolvent precipitation and electrostatic deposition | Nanoparticles | Spherical, 235 nm |
| [246] |
Pectin with whey protein concentrate | Citrus low-methoxyl pectin (DE 16–20%) | D-Limonene | Nanocomplex formation | Nanoparticles | Spherical, 100 nm |
| [247] |
Pectin, zein and sodium caseinate | Citrus peel pectin | Eugenol | Nanocomplex formation and nano-spray drying | Nanoparticles | Spherical, 140 nm |
| [248] |
Pectin and egg yolk low density lipoprotein | Citrus peel pectin | Curcumin | Heat-induced nanocomplex formation | Nanogels | Spherical, <60 nm |
| [249] |
Pectin and pea protein isolate | High-methoxyl citrus pectin (DE 90%), beet pectin (DE 62%), low-methoxyl citrus pectin (DE 29%), apple pectin (DE 78%) | Curcumin | Nanocomplex formation | Nanoparticles | Spherical, 559.2 ± 6.2 nm |
| [250] |
Pectin | Citrus pectin | Citrus peel flavonoids | Ionic gelation | Nanoparticles | Spherical, 271.5 ± 5.3 nm |
| [251] |
Pectin with whey protein concentrate (WPC) | Citrus high-methoxyl pectin (DE 71.1%) | Olive leaf extract | Double-layered emulsification | Nanoemulsions | 1443 nm |
| [252] |
Pectin with whey protein concentrate | Citrus high-methoxyl pectin (DE 71.1%) | Saffron extract | Double-layered emulsification and spray drying | Nanoparticles | Spherical, 482.3–536.3 nm | [253] | |
Cellulose | Microcrystalline cellulose | Origanum vulgare. essential oil | Ammonium persulfate hydrolysis | Cellulose nanocrystals | 1.2–2.9 µm |
| [254] |
Cellulose | Bacterial cellulose produced by Komagataeibacter sucrofermentans | Cinnamon essential oil | Emulsification | Cellulose nanocrystals | Spherical and rod-like, 350–550 nm |
| [255] |
Cellulose with alginate beads | Cellulose nanocrystals | Thyme essential oil | Emulsification | Cellulose nanocrystals | <200 nm |
| [256] |
Cellulose | Cellulose nanocrystals extracted from pistachio shells | Peppermint oil | Drop-wise addition of a peppermint oil ethanolic solution in cellulose nanocrystals suspension | Cellulose nanocrystals | Rod-like and spherical, 36.6–55.5 nm |
| [257] |
Type of Polymeric Carrier | Encapsulated Material | Target of Encapsulation | Size of the Obtained Delivery System | Application | Reference |
---|---|---|---|---|---|
Micelles | 10-Hydroxycamptothecin | Solubility, stability and controlled release | 340 nm | Inhibitory effect on the activity of glutathione S-transferase with enhanced pharmaco-kinetic and targeting in liver | [292] |
Micelles | Shikonin (from Lithospermum erythrorhizon) | Solubility, stability and controlled release | 53–98 nm | Targeting to breast cancer cells by temperature regulation | [293] |
Micelles | S. grandiflora extract | Solubility, stability and controlled release | 24.95 ± 0.34 nm | Antibacterial activity in an in vitro study against S. aureus. | [294] |
Micelles | P. oceanica extract | Bioavailability, solubility and stability | 252–55.74 nm | Anticancer properties as it inhibits the migration of cancer cells | [295] |
Dendrimers (PAMAM) | Curcumin (from C. longa) | Solubility and controlled release | -a | Better effect on the antiproliferative activity against lung cancer cells | [302] |
Dendrimers (PAMAM) | Curcumin | Bioavailability, solubility | ~150 nm | -a | [303] |
Dendrimer G2 | Curcumin | Solubility | 239 nm | Effective anti-Plasmodium compound—against malaria | [304] |
Dendrimers (PAMAM) | Silybin (from milk thistle plant) | Solubility, stability and controlled release | -a | Drug solubilization/inherent dendrimer cytotoxicity was reduced | [305] |
Dendrimers (PAMAM) | Black carrot anthocyanins (from D. carota plant) | Solubility, stability, biocompatibility and controlled release | 134.8 nm | Cytotoxicity against neuroblastoma cell line | [306] |
Dendrimers (PAMAM) | Liquiritin (from G. uralensis plant) | Solubility, stability and biocompatibility | - a | Permeability of intestinal absorption | [307] |
Dendrimers (PAMAM) | O. majorana essential oil | Solubility, stability and volatility | 20–30 nm | Action against the fungus P. infestans | [309] |
Dendrimers | C. zeylanicum and C. winterianus essential oil | Controlled release | - a | Biopesticides | [310] |
Nanoparticles | C. citratus | Controlled release | 217.1 ± 19.9 nm | In vitro anti-herpetic activity | [312] |
Nanocapsules (PLA) | P. europaea extract | Controlled release | 271.2 ± 13–1750 ± 305 nm | Antibacterial efficiency | [316] |
Nanocapsules | A. satureioides essential oil | - a | 235.9 nm | Oxidative stress | [317] |
Type of Nanoemulsion | Encapsulated Material | Target of Encapsulation | Size of the Obtained Delivery System | Application | Reference |
---|---|---|---|---|---|
W/O a | Hydroxysafflor yellow A | Bioavailability | 53.3 nm | Oral bioavailability | [339] |
O/W b | Emodin | Oral bioavailability | 116 ± 6.5 nm | Inhibition of UGT metabolism | [345] |
W/O a | Catechin | Bioavailability | 98.6 ± 1.01 nm | Photoprotection against UVA-induced oxidative stress | [346] |
W/O a and O/W b | Betulinic acid | Bioavailability and solubility | 150.3 ± 0.56 nm | Hepatoprotective and in vivo antioxidant efficacy activity | [347] |
O/W b | Curcumin | Oral bioavailability | 11.2 nm | Enhancement in Cmax | [348] |
W/O a | β-Elemene | Solubility | 52.68 nm | Antitumor activity | [349] |
O/W b | Quercetin | Bioavailability and solubility | 19.3 ± 0.17 nm | Contribute to preventing weight gain | [365] |
O/W/O | Quercetin | Bioavailability and solubility | 180–200 nm | (candidate for the treatment of obesity) | [366] |
O/W b | Curcumin and quercetin | Simultaneous drug administration and protection of the encapsulated compounds from degradation | 112.33 ± 1.51 nm | Protecting against lipid oxidation (chicken paté) | [367] |
O/W b | Curcumin and quercetin | Solubility, high encapsulation efficiency and long-term stability | 175.44 nm | Thermal stability, higher bioavailability and consequently drug effectiveness | [368] |
O/W b | Quercetin | Poor water solubility and high susceptibility to chemical degradation | 207–289 nm | Drug delivery system | [369] |
W/O a | Quercetin | Solubility | 38.9–266.67 nm | Antioxidant and antibacterial activity | [370] |
O/W b | Oregano oil | Solubility | 148 nm | Antimicrobial activity in food | [350] |
O/W b | Pterodon emarginatus | Solubility | 125 nm | Larvicidal property against Aedes aegypti | [371] |
O/W b | Garcinia mangostana extract | Bioavailability and solubility | 181 nm (167.3–222.0 nm) | - c | [372] |
O/W b | Pimpinella anisum essential oil | Solubility | 440 nm | Antimicrobial activity | [373] |
- c | Anthocyanin | Bioavailability and stability | - c | Antimicrobial activity | [374] |
- c | 2,4,6-triphenylaniline (TPA) | Stability and bioavailability | - c | Therapeutic drug delivery system in diabetes mellitus | [375] |
Inorganic Material | Core Material | Shape and Size of the Obtained Delivery System | Application | Reference |
---|---|---|---|---|
Silver | Cavendish banana peels | Spherical, crystalline, 55 nm | Antimicrobial activity against S. aureus, B. subtilis, E. coli and K. pneumonia | [394] |
Silver | A. vera | Octahedral, 5–50 nm | Antimicrobial activity against S. aureus, B. cereus, Micrococcus luteus, E. coli and K. pneumonia | [395] |
Silver | A. vera | Crystalline, 70–192 nm | Antibacterial activity against S. epidermidis and P. aeruginosa | [396] |
Silver | Tamarind fruit | Spherical, crystalline, 6–8 nm | Antibacterial activity against B. cereus, S. aureus, M. luteus, B. subtilis, Enterococcus sp., P. aeruginosa, Salmonella typhi, E. coli and K. pneumonia | [397] |
Silver | Cinnamon | Spherical, 50–70 nm | Antibacterial activity against S. aureus, E. coli, B. cereus and Pseudomonas species | [398] |
Silver | A. vera | Spherical, crystalline, <15 nm | Antibacterial activity against Kocuria varians and mercury removal capacity | [399] |
Silver | White tea leaves | Spherical, 19.8 nm | Antioxidant activity | [400] |
Silver | Plumbago auriculata | Spherical, hexagonal, <50 nm | Antimicrobial activity against S. aureus, E. coli, Klebsiella pneumoniae and Bacillus subtilis | [401] |
Silver | Citrus limon peels | Spherical, 59.7 nm | Antibacterial and cytotoxic activity | [402] |
Silver | Curcumin | Spherical, polycrystalline, 25–35 nm | Antibacterial activity against P. aeruginosa, E. coli, B. subtilis and S. aureus | [403] |
Silver | Turmeric extracts | Spherical and quasi-spherical, crystalline, 18 nm | Antimicrobial activity against E. coli O157:H7 and L. monocytogenes | [404] |
Silver | Mentha piperita | Spherical, 35 nm | Effect on the neurological enzyme acetylcholinesterase to predict its neurotoxicity | [405] |
Silver | Madhuca latifolia aqueous extract | Spherical, crystalline, 2–30 nm | Antioxidant and antibacterial activity against E. coli, S. aureus, L. monocytogenes, S. faecalis, S. typhimurium | [406] |
Silver and gold | Quercetin | Crystalline 53 and 27, respectively | Anti-neuroinflammatory activity on BV-2 microglial cells | [407] |
Gold | Plumeria alba flower | Spherical, 15.6–28 nm | Antibacterial activity against E. coli | [408] |
Gold | Hibiscus sabdariffa leaves | Spherical, crystalline, 10–60 nm | Cytotoxic activity against U87 glioblastoma cells under hyperglycemic condition | [409] |
Gold | Mimosa tenuiflora | Spherical, 20–200 nm | Cytotoxic activity and catalytic properties | [410] |
Gold | Resveratrol | Spherical, crystalline, 14.9–16.1 nm | Anticancer activity against human breast, pancreatic and prostate cancer cells | [411] |
Gold | Hibiscus sabdariffa flower | Spherical, crystalline, 15–45 nm | Anti-acute myeloid leukemia effect in a leukemic rodent model | [412] |
Palladium | Hippophae rhamnoides leaves | Spherical, crystalline, 10 nm | Catalytic activity for the Suzuki–Miyaura coupling in water | [413] |
Palladium | Chrysophyllum cainito | Crystalline, 169.2 nm | Catalytic activity for C–C coupling and reduction reactions | [414] |
Titanium dioxide | Salvadora persica aqueous ethanolic extract | Crystalline,19.8 nm | Antimicrobial activity against S. aureus and E. coli | [415] |
Zinc oxide | Passiflora caerulea | Spherical, 70 nm | Antibacterial activity against microbes that cause urinary tract infections (e.g., E. coli, Enterococcus sp., Streptococcus sp.) | [416] |
Zinc oxide | Cassia fistula and Melia azedarach | Spherical, 3–68 nm | Antimicrobial activity against S. aureus and E. coli | [417] |
Zinc oxide | Sambucus ebulus | Spherical, hexagonal, 17 nm | Antibacterial activity against B. cereus, S. aureus and E. coli | [418] |
Zinc oxide | Deverra tortuosa | - a 9.3–31.2 nm | In vitro cytotoxic activity against two cancer cell lines, i.e., human colon adenocarcinoma Caco-2 and human lung adenocarcinoma A549 | [419] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kyriakoudi, A.; Spanidi, E.; Mourtzinos, I.; Gardikis, K. Innovative Delivery Systems Loaded with Plant Bioactive Ingredients: Formulation Approaches and Applications. Plants 2021, 10, 1238. https://doi.org/10.3390/plants10061238
Kyriakoudi A, Spanidi E, Mourtzinos I, Gardikis K. Innovative Delivery Systems Loaded with Plant Bioactive Ingredients: Formulation Approaches and Applications. Plants. 2021; 10(6):1238. https://doi.org/10.3390/plants10061238
Chicago/Turabian StyleKyriakoudi, Anastasia, Eleni Spanidi, Ioannis Mourtzinos, and Konstantinos Gardikis. 2021. "Innovative Delivery Systems Loaded with Plant Bioactive Ingredients: Formulation Approaches and Applications" Plants 10, no. 6: 1238. https://doi.org/10.3390/plants10061238