Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 4684 2023-05-03 07:36:12 |
2 format correct Meta information modification 4684 2023-05-04 03:02:22 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Pasarin, D.; Ghizdareanu, A.; Enascuta, C.E.; Matei, C.B.; Bilbie, C.; Paraschiv-Palada, L.; Veres, P. Coating Materials to Increase the Stability of Liposomes. Encyclopedia. Available online: https://encyclopedia.pub/entry/43693 (accessed on 28 August 2024).
Pasarin D, Ghizdareanu A, Enascuta CE, Matei CB, Bilbie C, Paraschiv-Palada L, et al. Coating Materials to Increase the Stability of Liposomes. Encyclopedia. Available at: https://encyclopedia.pub/entry/43693. Accessed August 28, 2024.
Pasarin, Diana, Andra-Ionela Ghizdareanu, Cristina Emanuela Enascuta, Catalin Bogdan Matei, Catalin Bilbie, Luciana Paraschiv-Palada, Petronela-Andreea Veres. "Coating Materials to Increase the Stability of Liposomes" Encyclopedia, https://encyclopedia.pub/entry/43693 (accessed August 28, 2024).
Pasarin, D., Ghizdareanu, A., Enascuta, C.E., Matei, C.B., Bilbie, C., Paraschiv-Palada, L., & Veres, P. (2023, May 03). Coating Materials to Increase the Stability of Liposomes. In Encyclopedia. https://encyclopedia.pub/entry/43693
Pasarin, Diana, et al. "Coating Materials to Increase the Stability of Liposomes." Encyclopedia. Web. 03 May, 2023.
Coating Materials to Increase the Stability of Liposomes
Edit

Liposomes carry various compounds with applications in pharmaceutical, food, and cosmetic fields, and the administration route is especially parenteral, oral, or transdermal. Liposomes are used to preserve and release the internal components, thus maintaining the properties of the compounds, the stability and shelf life of the encapsulated products, and their functional benefits. The main problem in obtaining liposomes at the industrial level is their low stability due to fragile phospholipid membranes. To increase the stability of liposomes, phospholipid bilayers have been modified or different coating materials have been developed and studied, both for liposomes with applications in the pharmaceutical field and liposomes in the food field. In the cosmetic field, liposomes need no additional coating because the liposomal formulation is intended to have a fast penetration into the skin.

coating drug food liposomes stability

1. Introduction

Liposomes can be defined as artificial spherical nanostructures that consist of one or more phospholipid bilayer membranes [1][2][3].
They are the most commercialized active substance delivery systems with a prime encapsulation capacity and therapeutic index, biocompatibility, biodegradability, flexibility, and safety due to their structural similarity to cell membranes (lipid bilayer) [4]. The liposome structures consisting of phospholipids come from various sources of lecithin (soy, egg yolk, sunflower, rice beans, and rape seed). The main phospholipids used are phosphatidylcholines, phosphatidylethanolamines, phosphatidylserines, and phosphatidylglycerols. These lipids are amphiphilic because they have a group of polar heads and a lipophilic tail, making them suitable for use in obtaining liposomes [5][6].
The aqueous core of liposomes is enclosed by the phospholipid bilayer. The core encapsulates the water-soluble drugs, and the hydrophobic domain entraps insoluble compounds. Due to their structure, their stability, bioactivity, and bioavailability in the human body are increased [7][8]. Liposomal nanostructures can have one or more layer membranes.
The number of layers influences the liposome size from 20 nm to 1000 nm [9]. Based on size and lamellarity, liposomes are classified into several categories: small unilamellar vesicles (20–100 nm) [10], large unilamellar vesicles (>100 nm) [11][12] giant unilamellar vesicles (>1000 nm) [13][14], oligolamellar vesicles (100–1000 nm) [15][16], multilamellar large vesicles (>500 nm) [17][18] and multivesicular vesicles (>1000 nm) [19].
The methods used to obtain liposomes can be grouped into conventional and modern methods. The conventional methods used are thin-film hydration, ethanol/ether injection, reverse phase evaporation (detergent depletion, microfluidic channel, heating, membrane extrusion, high shear homogenization, and sonication), and the proliposome method. The modern methods used are freeze drying, dual asymmetric centrifugation, and supercritical fluid methods (supercritical anti-solvent, supercritical CO2 reverse phase evaporation process, the rapid expansion of a supercritical solution, depressurization of an expanded liquid organic solution suspension, and super-critical assisted liposome formation) [20].
It is recommended that the encapsulation of the active ingredients in liposomes be conducted by appropriate methods to protect them both from the food processing conditions and the action of the gastrointestinal tract (GT) juices to ensure targeted, controlled release. Different categories of active substances can be encapsulated in liposomes, with applications in the pharmaceutical, cosmetic, and food industries, such as vitamins, enzymes, antimicrobial polypeptides, essential oils, phenolic compounds, minerals, antioxidants, food additives, flavors, fatty acids, drugs, toxins, proteins (peptides), antigens, and nucleotides [21][22][23][24].
The use of the liposome-based delivery system in the pharmaceutical field is predominant because the therapeutic effect is achieved by reducing the dose and frequency of administration of the encapsulated compounds [25]. Currently, there are various liposomal products on the market with applications in the pharmaceutical and nutraceutical fields, such as Abhope, Liposomal Vegan D3 K2 Magnesium, VENTUS liposomal Omega 3, LVC5, and others In cosmetics, there are many commercial nano-liposomal products.
Liposomes and niosomes are particularly interesting due to the phospholipid component that has a high-esterified essential fatty acid content. Phosphatidylcholine is preferred as an essential component in obtaining liposome membranes because of its conditioning and softening properties. Unilamellar liposomes are most often used in the cosmetic field, and they do not need an additional coating because they must have an easy and fast penetration into the skin. Studies have shown that liposomes are suitable for delivering active compounds due to their compatibility with the skin and their extended and slow dermal release.

2. Coating Materials for Liposomes with Applications in Drug Delivery

Liposomes used as nanocarriers with amphiphilic character can deliver different drugs by their encapsulation. In the bloodstream, they are treated as foreign objects and are destroyed by immune system cells by phagocytosis [26]. Protecting liposomes from the rapid reaction of the immune system and increasing their stability against low pHs in the stomach represent challenges.
To improve and achieve a good biological interaction (with blood and tissues) while using liposomes as carrier drugs, studies have been conducted to improve liposomes’ surface. Liposomes were coated with film-forming compounds to improve the stability of their membranes.
The most common materials used for coating and the change in the surface of liposomes are found in the following classes of compounds: saccharides and their derivatives, polymers, and proteins. The general coating materials for liposomes encapsulating drugs and their properties are illustrated in Table 1.
Table 1. Coating materials for liposomes as drug delivery means.
Saccharides and their derivatives tend to be more physically and chemically stable, resulting in site-vesicle structures specific to their biological environments. In the coating process of liposomes using saccharides, the greatest influence on the permeability, fluidity, and integrity of the membrane is given by the mechanisms (adsorption, coagulation, and bridging) and by the method of binding the saccharide to the lipid bilayer of the surface [89].

3. Coating Materials for Liposomes with Applications in the Food Industry

In the food industry, the application of liposomes mainly focuses on texture alteration and water retention improvement. In recent years, studies have been conducted on the encapsulation of food components using various technologies (LipoCellTech, Kerkplein, The Netherlands, stealth liposomes, or non-PEGylated liposome technology). Active ingredients must be formulated in such a way as to protect them against production technology and environmental conditions so that they can be safely delivered to the targeted organs and cells. The results of various studies have shown that the coating of liposomes with different types of compounds by creating a layer on the surface of the membrane and providing electrostatic repulsion has led to increased physical stability, resistance to mechanical stress, and a low release speed of charged compounds [90].
Different types of materials have been used in the coating of liposome carriers for food-active compounds. Two groups of compounds have been used most frequently due to their food-grade qualities: saccharides and their derivatives and proteins. These groups of compounds have been chosen for their biocompatibility, low or nontoxicity, and neutral organoleptic properties [91].
The general coating materials for liposomes with applications in the food industry and their properties are illustrated in Table 2.
Table 2. Coating materials for liposomes with applications in the food industry.
Saccharides and their derivatives studied as coatings for food ingredients entrapped in liposomes have been shown to improve the thermal, physical and chemical, functional, and structural stability of liposomes during storage, with better release during in vitro digestion [47][97][98].
Some researchers [73] synthesized vitamin C and introduced it in mandarin juice. The multi-layered liposomes were the result of depositing positive chitosan and negative sodium alginate on the surface of the anionic nanoliposomes. The coated structure of nanoliposomes modified the surface characteristics of these. After 90 days, the vitamin C was still protected, and no significant organoleptic changes were observed in the fortified samples.
Low methoxyl pectin (LMP) can be used as a macromolecular material to modify the surface of liposomes. The liposomes formed not only have a good particle size and potential, but also have better stability during long-term storage. The double layer is protected from oxidation and maintains a good active compound release efficiency. LMP-coated liposomes added to orange juice create a bridge with metal ions and form a network-like gel to establish the stability of the liposomes. The addition of pectin with a different degree of esterification leads to an increase in the particle size of the liposomes. This is mainly due to the adsorption of pectin on the surface of the liposomes. Some environmental factors, such as pH, ionic strength, and temperature, have a significant effect on the appearance, particle size, and flow rate of liposomes, but LMP as a coating material can have a protective effect [103][104].
Inulin-coated liposomes showed better stability in the presence of surfactants and electrolytes. The use of cationic inulin helps to create a physical barrier to prevent the aggregation and fusion/coalescence phenomena, while using long-chain inulin has been shown to increase liposome stability during storage and improve gastric viability [105]. Coating liposomes with lactose or inulin prevents their rapid dissolution in alcohol, providing a protective effect. In addition, because lactose is a molecule present on the surface of blood cells, the affinity avoids macrophages. Depending on the microstructural order and the molecular weight of the saccharide, the structure of the liposome changes; for example, inulin as a coating material confers longer release properties due to its higher molecular weight. Thus, the thickness of the liposomes is closely related to the molecular weight; the higher the molecular weight, the thicker and more resistant the liposomes, which last longer in the gastrointestinal tract [91][105]. The use of proteins as a coating material has led to the enhancement of thermal and light stability with an improvement in the active compound stability under gastric conditions. Whey protein isolates used as a coating material for astaxanthin entrapped in liposomes improved the bio-accessibility and protected the liposome membranes against alteration during in vitro digestion [58][59]. The use of chitosan in combination with other compounds (alginate) to obtain a liposome coating material led to an improvement in the antimicrobial and prolonged antioxidant activity of the encapsulated compounds and the improvement of thermal and light storage stability [73][78][100][101].
Challenges in using coating materials to improve the stability of liposomes when used in the food industry include the need to ensure that the coating material is safe for consumption, the cost and complexity of manufacturing, the consistency of liposomal properties during storage, and the difficulty in determining the optimal parameters for coating. In addition, some modifications to the liposome formulation may be required to ensure that the liposomes remain stable over a long period of time and can survive during storage, handling, and shipment.

4. The Influence of Polymer Coatings on the Absorption of Liposomes

The stages of digestion for liposomes as vehicles for drug or bio-compound delivery have been studied under in vitro conditions in simple mono-compartment to complex multi-compartment dynamic digestive systems. In addition, an artificial gastric digestive system with a 3D-printed shape was developed and validated to follow the food digestion mechanisms [106].
The oral administration of liposomes raises several challenges, namely susceptibility to physiological factors in the GT, poor permeability of liposomes across gastrointestinal epithelia (the main absorption barrier), and liposomal formulations (manufacturing). Under the action of physiological factors, liposomes composed of phospholipids and cholesterol lose their integrity (are unstable), and the active ingredients are released but not in the target cell or tissues [107].
The liposome delivery systems cross the GT and change until they reach the intestinal mucosa where absorption takes place. Only some of the ingested liposomes reach full form and are absorbed by the lymph pathway [4].
At the level of the stomach, acid-stable gastric lipase-initiated digestion takes place. This can slightly affect the structure of the liposomes because they have a lipid bilayer membrane and because cholesterol from the structure increases the rigidity of membranes. So, they decrease slightly in diameter due to the pressure difference between the inside and outside of the two sides of the liposomes [108][109][110]. When the simulated digestion time was extended to 120 min, aggregation of the liposomes was observed due to the reduction in the electrostatic repulsion force between the liposomes under the conditions of a low pH. The encapsulated compounds, such as betacyanins, lutein, and β-carotene, could be degraded slower or faster without affecting the liposomes by crossing the liposomal membrane and exposure to gastric acid [111].
Most of the liposome digestion takes place in the small intestine under the action of pancreatic enzymes (colipase-dependent pancreatic lipase acts on unhydrolyzed triacylglycerols from the stomach, pancreatic lipase-related protein 2 acts as phospholipase and galactolipase, carboxyl ester hydrolase, bile salt-stimulated lipase, hydrolyses cholesterol esters, triacylglycerols, monoacylglycerols, vitamin (A, E) esters, phospholipids carotenoid esters, galactolipids, and polyethylene glycol mono- and di-esters, and pancreatic phospholipase A2, involved in the digestion of phospholipids, catalyzes the hydrolysis of the sn-2 fatty acyl ester bond of 3-sn-glycerophospholipids) [112].
Bile salts mediate digestion in several ways: (i) they weaken the interfacial stresses between molecules by facilitating the action of phospholipase A2 and lipase on the liposomal lipid phase; (ii) they weaken the structure of the phospholipid bilayers by the insertion of bile salt molecules and the formation of channels, which make the membranes more susceptible to the lipolysis process by fluidizing them; (iii) bile salts facilitate the hydrolysis of phospholipids and the release of fatty acids by the adsorption of lipase to phospholipid bilayers; (iv) they increase lipolysis by eliminating the accumulation of fatty acids and increasing the accessibility of lipase; and (v) they aid in the solubilization and absorption of the lipolysis products by forming mixed micelles [113][114].
When biopolymers are deposited on the surface of liposomes, their properties in different GT fluids may change the ability of enzymes to act on the surface of the lipids, which could improve the digestive stability of the liposomes. Their digestion in the GT involves a complex set of physical-chemical and biochemical reactions that affect the uptake of hydrophilic and hydrophobic-loaded molecules [115].
The role of chitosan in liposome digestion and absorption is still controversial. Some authors [115][116] report that the polycationic nature of chitosan prolongs the retention time through the intestinal mucosa. The explanation is that mucin, an anionic glycosylated protein negatively charged at the mouth pH, covers the chitosan-coated liposomes, offering further protection to the loaded active molecules during the other digestion phases. It takes place in the electrostatic interaction between the amine group (NH3+) of chitosan and the carboxylate (COO) or sulfonate (SO3) group of mucins. In addition, the adhesion of chitosan-coated liposomes to the mucosal membranes, negatively charged, enhances; so, the bioactive compounds are more available for absorption and the half-time of clearance increases.
The mechanism responsible for the permeation is based on the positive charges of this polysaccharide, which structurally reorganizes (opens) the tight junctions of the mucosal cell membrane proteins, facilitating the paracellular transport of hydrophilic macromolecules. Chitosan’s molecular weight and degree of deacetylation influence the increase in membrane permeability. Thus, a high degree of deacetylation and a high molecular mass contribute to the increase in the chitosan charge density, which leads to the increase in epithelial permeability and implicitly to the increase in drug transportation [117].
Highly methoxylated pectin is a widely used liposomal coating because it increases the stability of liposomes during storage and adheres to the intestinal epithelium without influencing membrane permeability [4].
Coating liposomes with PEG increases their intravenous circulation time and increases the stability of liposomes at the intestinal level through a mechanism of adhesion to the mucus of intestinal epithelia. The adhesion mechanism of positively charged mucoadhesive polymers is based on the ionic interaction between them and negative compounds from the mucus layer [118][119].
The polymer coating is a promising way to modify the surface characteristics of the vesicle’s stability to improve its applicability [120].
The liposome content is delivered in the cell by four mechanisms [121]. The first mechanism is the adsorption of liposomes on cells which can be specific—through specific receptors on the cell membrane and liposomes—and nonspecific, realized through attractive forces. The second mechanism represents the exchange of lipids between the cell membrane and the liposomal membrane due to their similarity. The third mechanism is endocytosis (for large particles by phagocytosis and receptor-dependent internalization by pinocytosis). The fourth mechanism is the fusion between the plasma and liposomal membranes. The liposomal content is delivered directly into the cell [122][123]. The liposome membrane is broken, and the encapsulated active compounds are released; these can be internalized into the cell in three ways: simple diffusion, facilitated diffusion, and active transport [124].
The cell uptake of liposomal oral or injectable products can be influenced by the liposomes’ size and surface charge. Experiments with liposomal formulations with surface charge and varied lipid compositions have shown that anionic or neutral liposomes are efficiently absorbed by monocyte-derived DCs [111][125]. Depending on the size of the liposomes, they follow different pathways. Studies show that liposomes between 40.6 nm and 276.6 nm in diameter are up-taken by Caco-2 cells [126].
From a pharmacokinetic perspective, the main goals of liposome drug delivery systems are improved in vivo drug release profiles, including enhanced drug absorption, targeted drug delivery, a modified metabolic pattern, a prolonged residence time of the drug in the body (e.g., in the bloodstream), and delayed and/or reduced renal excretion of the drug. From the initial stages of liposome system design through to the final clinical evaluations, absorption, distribution, metabolism, and excretion (ADME) must be considered to accurately understand the pharmacokinetic properties of this drug delivery system. In terms of ADME affecting the pharmacokinetic behavior of the drugs, for liposome delivery systems, the lipid bilayers serve as barriers between aqueous compartments and distribution compartments [127].
A quantitative explanation of the in vivo conditions under which a drug dose leads to therapeutic or side effects is provided by pharmacokinetics. For this purpose, the drug concentrations in the biophase and/or toxic phase must be considered. The concentration–time curves of drugs serve as the basis for pharmacokinetic research, which in turn serves as the starting point for estimating pharmacokinetic parameters with the help of corresponding mathematical models [128].
These factors should provide a quantitative link between biological concentrations and drug effects. In this situation, liposomes as drug carriers can be used as “pharmacokinetic modifiers” to achieve predetermined spatial and/or temporal targeted drug delivery. Recently, coated liposomes have been developed for the targeted delivery of therapeutic drugs to increase oral drug bioavailability, solubilize drugs for intravascular delivery, maintain the effects of drugs or genes in target tissues, reduce the potential for toxic effects or adverse reactions, and/or improve the stability of therapeutic drugs against enzymatic hydrolysis or other particular nutrients, peptides, and nucleic acids [129].
Liposomes, due to their subcellular size, can penetrate the tissue through the capillary walls and cross epithelial tissues and are usually taken up by cells. A therapeutic concentration must be achieved in the target tissues by modulating the physicochemical properties of the liposomes, as unfavorable exposure of nontarget tissues to these drugs can potentially lead to adverse effects.
Various characterization experiments are often performed during the development of these nanocarriers, mainly in vitro and in vivo tests, to optimize the drug delivery of liposome systems. Particle size, shape, chemical composition, surface hydrophilicity, polarity, drug release profile, and other physicochemical characteristics are used in in vitro studies to provide an indirect measurement of the drug delivery capabilities of different compounds [130].
On the other hand, successful in vitro tests are followed by in vivo studies to test the liposome drug carriers for efficacy in a living, intact organism or in specific organs or tissues. The produced drug nanocarriers are often subjected to two different types of in vivo experiments: pharmacodynamic tests on the pharmacological effects and pharmacokinetic studies for the expected effects of the particle and/or the drug associated with the particle.
Coated and uncoated liposome drug carriers often consist of a large number of individual parts that interact as integrated systems in a special structure. Compared to the free drug, these different components, especially the therapeutic portion (drug), have different ADME properties (absorption, distribution, metabolism, and excretion). Therefore, the localization of the drug and coated liposome drug carriers in the biological system is a problem in ADME-related animal studies with coated liposome drug carriers. in vivo studies in which each component is independently tracked may not be sufficient to determine the therapeutic efficacy and toxicity of coated liposome carriers [131][132].
Coating liposomes with certain materials can alter the ADME of the drug they are designed to deliver. Advantages of coating liposomes include increased protection of the drug from degradation, improved permeation through cell membranes, and the ability to control the release of the drug at a specific rate. The main disadvantages of coating liposomes are the impairment of pharmacokinetics, bioavailability, and the biological half-life of the drug, the difficulty in determining the optimal parameters for coating, and the possible toxic effects of the coating material. Toxicity depends on the material used, but general potential toxic effects include inflammation and irritation of skin and tissues, an increase in antigenicity, an increased risk of sensitization and allergic reactions, and disruption of cell membrane barrier functions.

References

  1. Jacob, S.; Nair, A.B.; Shah, J.; Gupta, S.; Boddu, S.H.; Sreeharsha, N. Lipid Nanoparticles as a Promising Drug Delivery Carrier for Topical Ocular Therapy—An Overview on Recent Advances. Pharmaceutics 2022, 14, 533.
  2. Kim, T.; Lee, J.; Jo, Y.J.; Choi, M.J. Application of Liposome Encapsulating Lactobacillus curvatus Extract in Cosmetic Emulsion Lotion. Materials 2021, 14, 7571.
  3. Lee, J.; Yang, J.H.; Kim, K.S.; Park, G.D. Effect of Liposome Storage of Cyanocobalamin on Its Degradation by Ascorbic Acid. Food Suppl. Biomater. Health 2021, 1, e7.
  4. Nguyen, T.X.; Huang, L.; Liu, L.; Abdalla, A.M.E.; Gauthier, M.; Yang, G. Chitosan-coated nano-liposomes for the oral delivery of berberine hydrochloride. J. Mater. Chem. B 2014, 2, 7149–7159.
  5. Marsanasco, M.; Del Valle Alonso, S. Why produce food-bioactive compounds to generate functional grade foods? In Functional Foods-Phytochemicals and Health Promoting Potential; Muhammad, S.A., Muhammad, H.A., Eds.; IntechOpen: London, UK, 2021.
  6. Van Hoogevest, P.; Wendel, A. The use of natural and synthetic phospholipids as pharmaceutical excipients. Eur. J. Lipid. Sci. Technol. 2014, 116, 1088–1107.
  7. Nakhaei, P.; Margiana, R.; Bokov, D.O.; Abdelbasset, W.K.; Kouhbanani, M.A.J.; Varma, R.S. Liposomes: Structure, biomedical applications, and stability parameters with emphasis on cholesterol. Front. Bioeng. Biotechnol. 2021, 9, 748.
  8. Karunaratne, D.N.; Pamunuwa, G.K.; Nicholas, I.H.; Ariyarathna, I.R. Science of Spices and Culinary Herbs-Latest Laboratory. In Pre-Clinical, and Clinical Studies; Atta-ur-Rahman, Choudhary, M.I., Yousuf, S., Eds.; Bentham Science Publishers: Sharjah, United Arab Emirates, 2019; Volume 1, pp. 104–147.
  9. Subramanian, P. Lipid-based nanocarrier system for the effective delivery of nutraceuticals. Molecules 2021, 26, 5510.
  10. Arpita, S.; Kumar, S.V. Liposomes–A Review. Int. J. Indig. Herbs Drugs 2020, 5, 1–6.
  11. Paliwal, R.; Paliwal, S.R.; Kenwat, R.; Kurmi, B.D.; Sahu, M.K. Solid lipid nanoparticles: A review on recent perspectives and patents. Expert. Opin. Ther. Pat. 2020, 30, 179–194.
  12. Ajeeshkumar, K.K.; Aneesh, P.A.; Raju, N.; Suseela, M.; Ravishankar, C.N.; Benjakul, S. Advancements in liposome technology: Preparation techniques and applications in food, functional foods, and bioactive delivery: A review. Compr. Rev. Food. Sci. Food Saf. 2021, 20, 1280–1306.
  13. Lazuardi, M.; Suharjomo, S.; Chien, C.H.; He, J.L.; Sugihartuti, R.; Maslachah, L. Encapsulation of progesterone-like compounds in 10% liposome increases their concentration in rats administered an injectable dosage form of these compounds. Kafkas Univ. Veter-Fak. Derg. 2022, 28, 27–34.
  14. de Freitas, C.F.; Calori, I.R.; Tessaro, A.L.; Caetano, W.; Hioka, N. Rapid formation of small unilamellar vesicles (suv) through low-frequency sonication: An innovative approach. Colloid. Surf. B 2019, 181, 837–844.
  15. Subramani, T.; Ganapathyswamy, H. An overview of liposomal nano-encapsulation techniques and its applications in food and nutraceutical. J. Food. Sci. Technol. 2020, 57, 3545–3555.
  16. Sheoran, R.; Khokra, S.L.; Chawla, V.; Dureja, H. Recent patents, formulation techniques, classification and characterization of liposomes. Recent Pat. Nanotechnol. 2019, 13, 17–27.
  17. Trucillo, P.; Reverchon, E. Production of PEG-coated liposomes using a continuous supercritical assisted process. J. Supercrit. Fluids 2021, 167, 105048.
  18. Rashidinejad, A.; Jafari, S.M. Nanoencapsulation of bioactive food ingredients. In Handbook of Food Nanotechnology; Jafari, S.M., Ed.; Academic Press: Cambridge, MA, USA, 2020; pp. 279–344.
  19. Saudagar, R.B.; Saokar, S. Anti-inflammatory natural compounds from herbal and marine origin. J. Drug Deliv. Ther. 2019, 9, 669–672. Available online: https://jddtonline.info/index.php/jddt/article/view/2906 (accessed on 15 April 2022).
  20. Andra, V.V.S.N.L.; Bhatraju, L.V.K.P.; Ruddaraju, L.K. A comprehensive review on novel liposomal methodologies, commercial formulations, clinical trials and patents. BioNanoScience 2022, 12, 274–291.
  21. Pamunuwa, G.K.; Karunaratne, D. Liposomal Delivery of Plant Bioactives Enhances Potency in Food Systems: A Review. J. Food Qual. 2022, 2022, 1–11.
  22. Emami, S.; Azadmard-Damirchi, S.; Peighambardoust, S.H.; Valizadeh, H.; Hesari, J. Liposomes as carrier vehicles for functional compounds in food sector. J. Exp. Nanosci. 2016, 11, 737–759.
  23. Mehnath, S.; Das, A.K.; Verma, S.K.; Jeyaraj, M. Biosynthesized/green-synthesized nanomaterials as potential vehicles for delivery of antibiotics/drugs. In Comprehensive Analytical Chemistry; Sandeep, K.V., Ashok, K.D., Eds.; Elsevier: Amsterdam, The Netherlands, 2021; Volume 94, pp. 363–432.
  24. Liu, P.; Chen, G.; Zhang, J. A review of liposomes as a drug delivery system: Current status of approved products, regulatory environments, and future perspectives. Molecules 2022, 27, 1372.
  25. Pentak, D.; Ploch-Jankowska, A.; Zięba, A.; Kozik, V. The Advances and Challenges of Liposome-Assisted Drug Release in the Presence of Serum Albumin Molecules: The Influence of Surrounding pH. Materials 2022, 15, 1586.
  26. Weber, C.; Voig, M.; Simo, J.; Danner, A.K.; Frey, H.; Mailänder, V.; Helm, M.; Morsbach, S.; Landfester, K. Functionalization of liposomes with hydrophilic polymers results in macrophage uptake independent of the protein corona. Biomacromolecules 2019, 20, 2989–2999.
  27. Li, X.; Tang, C.; Salama, M.; Xia, M.; Huang, X.; Sheng, L.; Cai, Z. Encapsulation efficiency and oral delivery stability of chitosan–liposome-encapsulated immunoglobulin Y. J. Food. Sci. 2022, 87, 1708–1720.
  28. Assadpour, S.; Akhtari, J.; Shiran, M.R. Pharmacokinetics study of chitosan-coated liposomes containing sumatriptan in the treatment of migraine. Casp. J. Intern. Med. 2022, 13, 90.
  29. Kari, N.; Shishir, M.R.I.; Li, Y.; Zineb, O.Y.; Mo, J.; Tangpong, J.; Chen, W. Pelargonidin-3-O-Glucoside Encapsulated Pectin-Chitosan-Nanoliposomes Recovers Palmitic Acid-Induced Hepatocytes Injury. Antioxidants 2022, 11, 623.
  30. Katual, M.K.; Gogna, S.; Singh, G. Advancements in Treatment of Laceration by Chitosan Coated Flexible Liposomes of Mupirocin: A current prospective. Sea 2022, 4, 5.
  31. Salehi, S.; Nourbakhsh, M.S.; Yousefpour, M.; Rajabzadeh, G.; Sahab-Negah, S. Chitosan-coated niosome as an efficient curcumin carrier to cross the blood–brain barrier: An animal study. J. Lipos. Res. 2022, 32, 284–292.
  32. Tamaddon, L.; Mohamadi, N.; Bavarsad, N. Preparation and Characterization of Mucoadhesive Loratadine Nanoliposomes for Intranasal Administration. Turk. J. Pharm. Sci. 2021, 18, 492.
  33. Lopes, N.A.; Mertins, O.; Pinilla, C.M.B.; Brandelli, A. Nisin induces lamellar to cubic liquid-crystalline transition in pectin and polygalacturonic acid liposomes. Food Hydrocoll. 2021, 112, 106320.
  34. Lopes, N.A.; Pinilla, C.M.B.; Brandelli, A. Antimicrobial activity of lysozyme-nisin co-encapsulated in liposomes coated with polysaccharides. Food Hydrocoll. 2019, 93, 1–9.
  35. Lopes, N.A.; Pinilla, C.M.B.; Brandelli, A. Pectin and polygalacturonic acid-coated liposomes as novel delivery system for nisin: Preparation, characterization and release behavior. Food Hydrocoll. 2017, 70, 1–7.
  36. Ghaleshahi, A.Z.; Rajabzadeh, G. The influence of sodium alginate and genipin on physico-chemical properties and stability of WPI coated liposomes. Food Res. Int. 2020, 130, 108966.
  37. Ghaleshahi, A.Z.; Rajabzadeh, G.; Ezzatpanah, H. Influence of Sodium Alginate and Genipin on Stability of Chitosome Containing Perilla Oil in Model and Real Drink. Eur. J. Lipid. Sci. Technol. 2020, 122, 1900358.
  38. Trucillo, P.; Cardea, S.; Baldino, L.; Reverchon, E. Production of liposomes loaded alginate aerogels using two supercritical CO2 assisted techniques. J. CO2 Util. 2020, 39, 101161.
  39. Maestrelli, F.; Mura, P.; González-Rodríguez, M.L.; Cózar-Bernal, M.J.; Rabasco, A.M.; Mannelli, L.D.C.; Ghelardini, C. Calcium alginate microspheres containing metformin hydrochloride niosomes and chitosomes aimed for oral therapy of type 2 diabetes mellitus. Int. J. Pharm. 2017, 530, 430–439.
  40. Shao, P.; Wang, P.; Niu, B.; Kang, J. Environmental stress stability of pectin-stabilized resveratrol liposomes with different degree of esterification. Int. J. Biol. Macromol. 2018, 119, 53–59.
  41. Gottesmann, M.; Goycoolea, F.M.; Steinbacher, T.; Menogni, T.; Hensel, A. Smart drug delivery against Helicobacter pylori: Pectin-coated, mucoadhesive liposomes with antiadhesive activity and antibiotic cargo. Appl. Microbiol. 2020, 104, 5943–5957.
  42. Tran, T.T.T.; Tran, V.H.; Lam, T.T. Encapsulation of tagitinin C in liposomes coated by Tithonia diversifolia pectin. J. Microencapsul. 2019, 36, 53–61.
  43. Iacob, A.T.; Lupascu, F.G.; Apotrosoaei, M.; Vasinc, I.M.; Tauser, R.G.; Lupascu, D.; Giusca, S.E.; Caruntu, I.-D.; Profire, L. Recent biomedical approaches for chitosan-based materials as drug delivery nanocarriers. Pharmaceutics 2021, 13, 587.
  44. Pu, C.; Tang, W.; Li, X.; Li, M.; Sun, Q. Stability enhancement efficiency of surface decoration on curcumin-loaded liposomes: Comparison of guar gum and its cationic counterpart. Food Hydrocoll. 2019, 87, 29–37.
  45. Barba, A.A.; Bochicchio, S.; Bertoncin, P.; Lamberti, G.; Dalmoro, A. Coating of nanolipid structures by a novel simil-microfluidic technique: Experimental and theoretical approaches. Coatings 2019, 9, 491.
  46. Kaminski, G.A.; Sierakowski, M.R.; Pontarolo, R.; Dos Santos, L.A.; de Freitas, R.A. Layer-by-layer polysaccharide-coated liposomes for sustained delivery of epidermal growth factor. Carbohyd. Polym. 2016, 140, 129–135.
  47. Amjadi, S.; Almasi, H.; Hamishehkar, H.; Khaledabad, M.A.; Lim, L.T. Coating of betanin and carvone Co-loaded nanoliposomes with synthesized cationic inulin: A strategy for enhancing the stability and bioavailability. Food. Chem. 2022, 373, 131403.
  48. Joseph, A.; Kumar, D.; Balakrishnan, A.; Shanmughan, P.; Maliakel, B.; Krishnakumar, I.M. Surface-engineered liposomal particles of calcium ascorbate with fenugreek galactomannan enhanced the oral bioavailability of ascorbic acid: A randomized, double-blinded, 3-sequence, crossover study. Rsc. Adv. 2021, 11, 38161–38171.
  49. Kari, O.K.; Tavakol, S.; Parkkila, P.; Baan, S.; Savolaine, R.; Ruoslaht, T.; Johansson, N.G.; Ndika, J.; Alenius, H.; Viitala, T.; et al. Light-Activated Liposomes Coated with Hyaluronic Acid as a Potential Drug Delivery System. Pharmaceutics 2020, 12, 763.
  50. Menon, P.; Teo, Y.Y.; Misra, M. Effect of diethylaminoethyl-dextran coated liposomes on the rheological properties of carbopol gel. Appl. Rheol. 2018, 28, 1–6.
  51. Refai, H.; Hassan, D.; Abdelmonem, R. Development and characterization of polymer-coated liposomes for vaginal delivery of sildenafil citrate. Drug. Deliv. 2017, 24, 278–288.
  52. Farooq, A.; Iqbal, A.; Rana, N.F.; Fatima, M.; Maryam, T.; Batool, F.; Rehman, Z.; Menaa, F.; Azhar, S.; Nawaz, A.; et al. A Novel Sprague-Dawley Rat Model Presents Improved NASH/NAFLD Symptoms with PEG Coated Vitexin Liposomes. Int. J. Mol. Sci. 2022, 23, 3131.
  53. Nunes, S.S.; Fernandes, R.S.; Cavalcante, C.H.; da Costa César, I.; Leite, E.A.; Lopes, S.C.A.; Ferretti, A.; Rubello, D.; Townsend, D.M.; de Oliveira, M.C.; et al. Influence of PEG coating on the biodistribution and tumor accumulation of pH-sensitive liposomes. Drug. Deliv. Transl. Res. 2019, 9, 123–130.
  54. Haseena, M. Scholar: National School of Leadership, 8. 2019. Available online: https://jconsortium.com/index.php/scholar/article/view/23 (accessed on 15 April 2022).
  55. Nazeer, N.; Panicker, J.T.; Rajalekshmi, S.M.; Shaiju, S.D.A. A Review on Surface Modified Sterically Stabilized Liposomes. Int. J. Innov. Sci. Res. Technol. 2019, 4, 795–801.
  56. Hermal, F.; Frisch, B.; Specht, A.; Bourel-Bonnet, L.; Heurtault, B. Development and characterization of layer-by-layer coated liposomes with poly (L-lysine) and poly (L-glutamic acid) to increase their resistance in biological media. Int. J. Pharm. 2020, 586, 119568.
  57. Martí Coma-Cros, E.; Biosca, A.; Lantero, E.; Manca, M.L.; Caddeo, C.; Gutiérrez, L.; Ramírez, M.; Borgheti-Cardoso, L.N.; Manconi, M.; Fernàndez-Busquets, X. Antimalarial activity of orally administered curcumin incorporated in Eudragit®-containing liposomes. Int J. Mol. Sci. 2018, 19, 1361.
  58. Pan, L.; Li, H.; Hou, L.; Chang, Z.; Li, Y.; Li, X. Gastrointestinal digestive fate of whey protein isolate coated liposomes loading astaxanthin: Lipolysis, release, and bioaccessibility. Food Biosci. 2022, 45, 101464.
  59. Pan, L.; Zhang, X.; Fan, X.; Li, H.; Xu, B.; Li, X. Whey protein isolate coated liposomes as novel carrier systems for astaxanthin. Eur. J. Lipid. Sci. Technol. 2020, 122, 1900325.
  60. Yi, X.; Zheng, Q.; Pan, M.H.; Chiou, Y.S.; Li, Z.; Li, L.; Chen, Y.; Hu, J.; Duan, S.; Wei, S.; et al. Liposomal vesicles-protein interaction: Influences of iron liposomes on emulsifying properties of whey protein. Food Hydrocoll. 2019, 89, 602–612.
  61. Taguchi, K.; Okamoto, Y.; Matsumoto, K.; Otagiri, M.; Chuang, V.T.G. When albumin meets liposomes: A feasible drug carrier for biomedical applications. Pharmaceuticals 2021, 14, 296.
  62. Li, M.; Du, C.; Guo, N.; Teng, Y.; Meng, X.; Sun, H.; Li, S.; Yu, P.; Galons, H. Composition design and medical application of liposomes. Eur. J. Med. Chem. 2019, 164, 640–653.
  63. Sato, H.; Nakhaei, E.; Kawano, T.; Murata, M.; Kishimura, A.; Mori, T.; Katayama, Y. Ligand-mediated coating of liposomes with human serum albumin. Langmuir 2018, 34, 2324–2331.
  64. Lee, E.H.; Lee, M.K.; Lim, S.J. Enhanced stability of indocyanine green by encapsulation in zein-phosphatidylcholine hybrid nanoparticles for use in the phototherapy of cancer. Pharmaceutics 2021, 13, 305.
  65. Dong, Y.; Dong, P.; Huang, D.; Mei, L.; Xia, Y.; Wang, Z.; Pan, X.; Li, G.; Wu, C. Fabrication and characterization of silk fibroin-coated liposomes for ocular drug delivery. Eur. J. Pharm. Biopharm. 2015, 91, 82–90.
  66. Battogtokh, G.; Joo, Y.; Abuzar, S.M.; Park, H.; Hwang, S.J. Gelatin Coating for the Improvement of Stability and Cell Uptake of Hydrophobic Drug-Containing Liposomes. Molecules 2022, 27, 1041.
  67. Hosseini, S.F.; Ansari, B.; Gharsallaoui, A. Polyelectrolytes-stabilized liposomes for efficient encapsulation of Lactobacillus rhamnosus and improvement of its survivability under adverse conditions. Food Chem. 2022, 372, 131358.
  68. Barrera, Y.A.B.; Husteden, C.; Alherz, J.; Fuhrmann, B.; Wölk, C.; Groth, T. Extracellular matrix-inspired surface coatings functionalized with dexamethasone-loaded liposomes to induce osteo-and chondrogenic differentiation of multipotent stem cells. Mater. Sci. Eng. C 2021, 131, 112516.
  69. Dutta, S.; Moses, J.A.; Anandharamakrishnan, C. Biomedical and food applications of biopolymer-based liposome. In Biopolymer-Based Formulations; Kunal, P., Indranil, B., Preetam, S., Doman, K., Win-Ping, D., Navneet, K.D., Kaustav, M., Eds.; Elsevier: Amsterdam, The Netherlands, 2020; pp. 167–192.
  70. Hosseini, S.F.; Soofi, M.; Rezaei, M. Enhanced physicochemical stability of ω-3 PUFAs concentrates-loaded nanoliposomes decorated by chitosan/gelatin blend coatings. Food. Chem. 2021, 345, 128865.
  71. Mosafer, J.; Sabbaghi, A.H.; Badiee, A.; Dehghan, S.; Tafaghodi, M. Preparation, characterization and in vivo evaluation of alginate-coated chitosan and trimethylchitosan nanoparticles loaded with PR8 influenza virus for nasal immunization. Asian J. Pharm. Sci. 2019, 14, 216–221.
  72. Parchen, G.P.; Jacumazo, J.; Koop, H.S.; Biscaia, S.M.P.; Trindade, E.S.; Silveira, J.L.M.; de Freitas, R.A. Modulation of epidermal growth factor release by biopolymer-coated liposomes. J. Pharm. Sci. 2020, 109, 2294–2301.
  73. Liu, W.; Tian, M.; Kong, Y.; Lu, J.; Li, N.; Han, J. Multilayered vitamin C nanoliposomes by self-assembly of alginate and chitosan: Long-term stability and feasibility application in mandarin juice. LWT Food Sci. Technol. 2017, 75, 608–615.
  74. Karim, N.; Shishir, M.R.I.; Chen, W. Surface decoration of neohesperidin-loaded nanoliposome using chitosan and pectin for improving stability and controlled release. Int. J. Biol. Macromol. 2020, 164, 2903–2914.
  75. Ribeiro, L.N.D.M.; de Paula, E.; Rossi, D.A.; Monteiro, G.P.; Júnior, E.C.V.; Silva, R.R.; Fonseca, B.B. Hybrid pectin-liposome formulation against multi-resistant bacterial strains. Pharmaceutics 2020, 12, 769.
  76. Raj, V.; Raorane, C.J.; Lee, J.H.; Lee, J. Appraisal of chitosan-gum Arabic-coated bipobiopolymericocarriers for efficient dye removal and eradication of the plant pathogen Botrytis cinerea. ACS Appl. Mater. Inter. 2021, 13, 47354–47370.
  77. Islam, N.; Ferro, V. Recent advances in chitosan-based nanoparticulate pulmonary drug delivery. Nanoscale 2016, 8, 14341–14358.
  78. Mehdizadeh, A.; Shahidi, S.A.; Shariatifar, N.; Shiran, M.; Ghorbani-HasanSaraei, A. Evaluation of chitosan-zein coating containing free and nano-encapsulated Pulicaria gnaphalodes (Vent.) Boiss. extract on quality attributes of rainbow trout. J. Aquat. Food. Prod. Technol. 2021, 30, 62–75.
  79. Deygen, I.M.; Seidl, C.; Kölmel, D.K.; Bednarek, C.; Heissler, S.; Kudryashova, E.V.; Bräse, S.; Schepers, U. Novel prodrug of doxorubicin modified by stearoylspermine encapsulated into PEG-chitosan-stabilized liposomes. Langmuir 2016, 32, 10861–10869.
  80. Gomaa, A.I.; Martinent, C.; Hammami, R.; Fliss, I.; Subirade, M. Dual coating of liposomes as encapsulating matrix of antimicrobial peptides: Development and characterization. Front. Chem. 2017, 5, 103.
  81. Rezvani, M.; Manca, M.L.; Muntoni, A.; De Gioannis, G.; Pedraz, J.L.; Gutierrez, G.; Manconi, M. From process effluents to intestinal health promotion: Developing biopolymer-whey liposomes loaded with gingerol to heal intestinal wounds and neutralize oxidative stress. Int. J. Pharm. 2022, 613, 121389.
  82. Tan, C.; Wang, J.; Sun, B. Biopolymer-liposome hybrid systems for controlled delivery of bioactive compounds: Recent advances. Biotechnol. Adv. 2021, 48, 107727.
  83. Luo, R.; Lin, M.; Zhang, C.; Shi, J.; Zhang, S.; Chen, Q.; Gao, F. Genipin-crosslinked human serum albumin coating using a tannic acid layer for enhanced oral administration of curcumin in the treatment of ulcerative colitis. Food Chem. 2020, 330, 127241.
  84. Zamani-Ghaleshahi, A.; Rajabzadeh, G.; Ezzatpanah, H.; Ghavami, M. Biopolymer coated nanoliposome as enhanced carrier system of perilla oil. Food. Biophys. 2020, 15, 273–287.
  85. Shah, V.; Jobanputra, A.; Saxena, B.; Nivsarkar, M. Development and Characterization of Saturated Fatty Acid-Engineered, Silica-Coated Lipid Vesicular System for Effective Oral Delivery of Alfa-Choriogonadotropin. AAPS PharmSciTech 2021, 22, 1–16.
  86. Pham, X.H.; Park, S.M.; Ham, K.M.; Kyeong, S.; Son, B.S.; Kim, J.; Hahm, E.; Kim, Y.H.; Bock, S.; Kim, W.; et al. Synthesis and application of silica-coated quantum dots in biomedicine. Int. J. Mol. Sci. 2021, 22, 10116.
  87. Bewernitz, M.A.; Lovett, A.C.; Gower, L.B. Liquid–Solid Core-Shell Microcapsules of Calcium Carbonate Coated Emulsions and Liposomes. Appl. Sci. 2020, 10, 8551.
  88. Wu, S.; Jiang, M.; Mao, H.; Zhao, N.; He, D.; Chen, Q.; Liu, D.; Zhang, W.; Song, X.M. A sensitive cholesterol electrochemical biosensor based on biomimetic cerasome and graphene quantum dots. Anal. Bioanal. Chem. 2022, 414, 3593–3603.
  89. Hasan, M.; Messaoud, G.B.; Michaux, F.; Tamayol, A.; Kahn, C.J.; Belhaj, N.; Linder, M.; Arab-Tehrany, E. Chitosan-coated liposomes encapsulating curcumin: Study of lipid–polysaccharide interactions and nanovesicle behavior. Rsc. Adv. 2016, 6, 45290–45304.
  90. Li, S.; Moosa, B.A.; Croissant, J.G.; Khashab, N.M. Electrostatic Assembly/Disassembly of Nanoscaled Colloidosomes for Light-Triggered Cargo Release. Angew. Chem. Int. Ed. 2015, 127, 6908–6912.
  91. Román-Aguirre, M.; Leyva-Porras, C.; Cruz-Alcantar, P.; Aguilar-Elguézabal, A.; Saavedra-Leos, M.Z. Comparison of Polysaccharides as Coatings for Quercetin-Loaded Liposomes (QLL) and Their Effect as Antioxidants on Radical Scavenging Activity. Polymers 2020, 12, 2793.
  92. Ramezanzade, L.; Hosseini, S.F.; Akbari-Adergani, B.; Yaghmur, A. Cross-linked chitosan-coated liposomes for encapsulation of fish-derived peptide. LWT Food Sci. Technol. 2021, 150, 112057.
  93. Seyedabadi, M.M.; Rostami, H.; Jafari, S.M.; Fathi, M. Development and characterization of chitosan-coated nanoliposomes for encapsulation of caffeine. Food Biosci. 2021, 40, 100857.
  94. Ran, L.; Chi, Y.; Huang, Y.; He, Q.; Ren, Y. Synergistic antioxidant effect of glutathione and edible phenolic acids and improvement of the activity protection by coencapsulation into chitosan-coated liposomes. LWT Food Sci. Technol. 2020, 127, 109409.
  95. 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.
  96. Akgün, D.; Gültekin-Özgüven, M.; Yücetepe, A.; Altin, G.; Gibis, M.; Weiss, J.; Özçelik, B. Stirred-type yoghurt incorporated with sour cherry extract in chitosan-coated liposomes. Food Hydrocoll. 2020, 101, 105532.
  97. Mohammadi, A.; Jafari, S.M.; Mahoonak, A.S.; Ghorbani, M. Liposomal/nanoliposomal encapsulation of food-relevant enzymes and their application in the food industry. Food Bioprocess. Technol. 2021, 14, 23–38.
  98. Šeremet, D.; Vugrinec, K.; Petrović, P.; Butorac, A.; Kuzmić, S.; Cebin, A.V.; Mandura, A.; Lovrić, M.; Pjanović, R.; Komes, D. Formulation and characterization of liposomal encapsulated systems of bioactive ingredients from traditional plant mountain germander (Teucrium montanum L.) for incorporation into coffee drinks. Food Chem. 2022, 370, 131257.
  99. Amjadi, S.; Almasi, H.; Hamishehkar, H.; Khaledabad, M.A.; Lim, L.T. Cationic inulin as a new surface decoration hydrocolloid for improving the stability of liposomal nanocarriers. Colloid. Surface. B 2022, 213, 112401.
  100. Gibis, M.; Rahn, N.; Weiss, J. Physical and oxidative stability of uncoated and chitosan-coated liposomes containing grape seed extract. Pharmaceutics 2013, 5, 421–433.
  101. Chen, S.; Ma, X.; Han, Y.; Wei, Y.; Guo, Q.; Yang, S.; Zhang, Y.; Liao, W.; Gao, Y. Effect of chitosan molecular weight on zein-chitosan nanocomplexes: Formation, characterization, and the delivery of quercetagetin. Int. J. Biol. Macromol. 2020, 164, 2215–2223.
  102. Lin, L.; Zhu, Y.; Cui, H. Inactivation of Escherichia coli O157: H7 treated by poly-L-lysine-coated bacteriophages liposomes in pork. J. Food. Safety. 2018, 38, e12535.
  103. Li, Y.; Zhao, H.; Duan, L.R.; Li, H.; Yang, Q.; Tu, H.H.; Cao, W.; Wang, S.W. Preparation, characterization and evaluation of bufalin liposomes coated with citrus pectin. Colloids Surf. A Physicochem. Eng. Asp. 2014, 444, 54–62.
  104. Feng, S.; Sun, Y.; Wang, P.; Sun, P.; Ritzoulis, C.; Shao, P. Co-encapsulation of resveratrol and epigallocatechin gallate in low methoxyl pectin-coated liposomes with great stability in orange juice. Int. J. Food. Sci. Technol. 2020, 55, 1872–1880.
  105. Belhaj, N.; Arab-Tehrany, E.; Loing, E.; Bézivin, C. Skin delivery of hydrophilic molecules from liposomes and polysaccharide-coated liposomes. Int. J. Cosmet. Sci. 2017, 39, 435–441.
  106. Liu, W.; Fu, D.; Zhang, X.; Chai, J.; Tian, S.; Han, J. Development and validation of a new artificial gastric digestive system. J. Food Res. Int. 2019, 122, 183–190.
  107. Wu, W.; Lu, Y.; Qi, J. Oral delivery of liposomes. Ther. Deliv. 2015, 6, 1239–1241.
  108. Liu, W.; Ye, A.; Han, F.; Han, J. Advances and challenges in liposome digestion: Surface interaction, biological fate, and GIT modeling. Adv. Colloid Interface Sci. 2019, 263, 52–67.
  109. Zaeim, D.; Mulet-Cabero, A.I.; Read, S.A.; Liu, W.; Han, J.; Wilde, P.J. Effect of oil droplet size on the gastric digestion of milk protein emulsions using a semi-dynamic gastric model. Food Hydrocoll. 2022, 124, 107278.
  110. Infantes-Garcia, M.R.; Verkempinck, S.H.E.; Gonzalez-Fuentes, P.G.; Hendrickx, M.E.; Grauwet, T. Lipolysis products formation during in vitro gastric digestion is affected by the emulsion interfacial composition. Food Hydrocoll. 2021, 110, 106163.
  111. Liu, W.; Jin, Y.; Wilde, P.J.; Hou, Y.; Wang, Y.; Han, J. Mechanisms, physiology, and recent research progress of gastric emptying. Crit. Rev. Food Sci. Nutr. 2021, 61, 2742–2755.
  112. Salhi, A.; Carriere, F.; Grundy, M.M.L.; Aloulou, A. Enzymes involved in lipid digestion. In Bioaccessibility and Digestibility of Lipids from Food; Grundy, M.M.L., Wilde, P.J., Eds.; Springer International Publishing: Cham, Switzerland, 2003; pp. 3–28.
  113. Liu, W.; Kong, Y.; Ye, A.; Shen, P.; Dong, L.; Xu, X.; Han, J. Preparation, formation mechanism and in vitro dynamic digestion behavior of quercetin-loaded liposomes in hydrogels. Food Hydrocoll. 2020, 104, 105743.
  114. Macierzanka, A.; Torcello-Gómez, A.; Jungnickel, C.; Maldonado-Valderrama, J. Bile salts in digestion and transport of lipids. Adv. Colloid Interface Sci. 2019, 274, 102045.
  115. Cuomo, F.; Cofelice, M.; Venditti, F.; Ceglie, A.; Miguel, M.; Lindman, B.; Lopez, F. In-vitro digestion of curcumin loaded chitosan-coated liposomes. Colloids Surf. B Biointerfaces 2018, 168, 29–34.
  116. Liu, J.; Zhang, J.; Xia, W. Hypocholesterolaemic effects of different chitosan samples in vitro and in vivo. Food. Chem. 2008, 107, 419–425.
  117. Garg, U.; Chauhan, S.; Nagaich, U.; Jain, N. Current advances in chitosan nanoparticles-based drug delivery and targeting. Adv. Pharm. Bull. 2019, 9, 195.
  118. Han, H.K.; Shin, H.J.; Ha, D.H. Improved oral bioavailability of alendronate via the mucoadhesive liposomal delivery system. Eur. J. Pharm. Sci. 2012, 46, 500–507.
  119. Huang, A.; Makhlof, A.; Ping, Q.; Tozuka, Y.; Takeuchi, H. N-trimethyl chitosan-modified liposomes as carriers for oral delivery of salmon calcitonin. Drug. Deliv. 2011, 18, 562–569.
  120. Park, S.N.; Jo, N.R.; Jeon, S.H. Chitosan-coated liposomes for enhanced skin permeation of resveratrol. J. Ind. Eng. Chem. 2014, 20, 1481–1485.
  121. Ducat, E.; Evrard, B.; Peulen, O.; Piel, G. Cellular uptake of liposomes monitored by confocal microscopy and flow cytometry. J. Drug. Deliv. Sci. Tec. 2011, 21, 469–477.
  122. Wang, J.; Byrne, J.D.; Napier, M.E.; DeSimone, J.M. More effective nanomedicines through particle design. Small 2011, 7, 1919–1931.
  123. Nejdl, L.; Kudr, J.; Moulick, A.; Hegerova, D.; Ruttkay-Nedecky, B.; Gumulec, J.; Cihalova, K.; Smerkova, K.; Dostalova, S.; Krizkova, S.; et al. Platinum nanoparticles induce damage to DNA and inhibit DNA replication. PLoS ONE 2017, 12, e0180798.
  124. Rautio, J.; Kumpulainen, H.; Heimbach, T.; Oliyai, R.; Oh, D.; Järvinen, T.; Savolainen, J. Prodrugs: Design and clinical applications. Nat. Rev. Drug. Discov. 2008, 7, 255–270.
  125. Nagy, N.A.; Castenmiller, C.; Vigario, F.L.; Sparrius, R.; van Capel, T.M.; de Haas, A.M.; de Jong, E.C. Uptake kinetics of liposomal formulations of differing charge influences development of in vivo dendritic cell immunotherapy. J. Pharm. Sci. 2022, 111, 1081–1091.
  126. Andar, A.U.; Hood, R.R.; Vreeland, W.N.; DeVoe, D.L.; Swaan, P.W. Microfluidic preparation of liposomes to determine particle size influence on cellular uptake mechanisms. Pharm. Res. 2014, 31, 401–413.
  127. Hamidi, M.; Azadi, A.; Rafiei, P.; Ashrafi, H. A pharmacokinetic overview of nanotechnology-based drug delivery systems: An ADME-oriented approach. Crit. Rev. ™ Ther. Drug Carr. Syst. 2013, 30, 435–467.
  128. Li, M.; Al-Jamal, K.T.; Kostarelos, K.; Reineke, J. Physiologically based pharmacokinetic modeling of nanoparticles. ACS Nano 2010, 4, 6303–6317.
  129. Oberdörster, G. Safety assessment for nanotechnology and nanomedicine: Concepts of nanotoxicology. J. Intern. Med. 2010, 267, 89–105.
  130. Yang, R.S.; Chang, L.W.; Yang, C.S.; Lin, P. Pharmacokinetics and physiologically-based pharmacokinetic modeling of nanoparticles. J. Nanosci. Nanotechnol. 2010, 10, 8482–8490.
  131. Moghimi, S.M.; Hunter, A.C.; Murray, J.C. Long-circulating and target-specific nanoparticles: Theory to practice. Pharmacol. Rev. 2001, 53, 283–318.
  132. 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.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , ,
View Times: 336
Revisions: 2 times (View History)
Update Date: 04 May 2023
1000/1000
ScholarVision Creations