Next Article in Journal
Noise Evaluation of Coated Polymer Gears
Previous Article in Journal
Study on the Mechanical Properties and Energy Absorbing Capability of Polyurethane Microcellular Elastomers under Different Compressive Strain Rates
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Coating Materials to Increase the Stability of Liposomes

by
Diana Pasarin
1,
Andra-Ionela Ghizdareanu
1,2,*,
Cristina Emanuela Enascuta
1,*,
Catalin Bogdan Matei
1,
Catalin Bilbie
3,
Luciana Paraschiv-Palada
3 and
Petronela-Andreea Veres
3
1
Institutul National de Cercetare-Dezvoltare pentru Chimie si Petrochimie, ICECHIM, 202 Splaiul Independentei, 060021 Bucuresti, Romania
2
Facultatea de Stiinta si Ingineria Materialelor, Universitatea Politehnica din Bucuresti, 313 Splaiul Independentei, 060042 Bucharest, Romania
3
Expergo Business Network SRL, 6 Radu Calomfirescu, 030216 Bucuresti, Romania
*
Authors to whom correspondence should be addressed.
Polymers 2023, 15(3), 782; https://doi.org/10.3390/polym15030782
Submission received: 25 January 2023 / Revised: 31 January 2023 / Accepted: 2 February 2023 / Published: 3 February 2023

Abstract

:
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. The aim of this review is to provide current knowledge regarding physical and chemical factors that influence stability, coating materials for liposomes with applications in the pharmaceutical and food fields to increase the stability of liposomes containing various sensitive compounds, and absorption of the liposomes and commercial liposomal products obtained through various technologies available on the market.

Graphical Abstract

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.
Firm Christian Dior SA (Paris, France) launched into the commercial market Capture anti-aging cream, and Laboratories RoC (Istanbul, Turkey) launched Myosphere—the first emulsion with the inclusion of liposomes and the first liposomal facial cream for men [26].
In food supplement formulations, liposomes can provide protection and the controlled release of sensitive active substances.
The encapsulation of various active compounds leads to the enrichment and strengthening of various food supplement formulations [27]. In the food industry, liposomes are used in various applications, such as enhancing the bioavailability of nutritional components, providing antimicrobial activity of the encapsulated ingredients, increasing intestinal absorption, improving flavors, and extending shelf life. There are very few food commercial applications of liposomes. They have been applied in different food matrices, namely dairy, meat, beverages, chocolate, and candy [28,29].
Liposomes are ideal delivery systems, but their use is limited by their short circulation half-life and their susceptibility to oxidation and hydrolysis. Their physical stability can be affected by gastric juice’s low pH, bile salts that solvate the liposomes, and enzymatic hydrolysis resulting in their breakage and release of the loaded molecules in the GT. There are many methods to increase the stability of liposomes, such as the application of a protective coating, changing the liposomal membrane structure, the addition of cryoprotectants before liposome lyophilization, and the use of surfactants or various special polymer gels [9,30,31,32]. Common cryoprotectants used to improve the stability of liposomes during lyophilization include dimethyl sulfoxide, glycerol, sugars and disaccharides, and polyampholytes. These materials help protect the integrity of liposomes during low-temperature storage by preventing the formation of harmful ice crystals [33].
To improve the stability of liposomes against the hostile environment of the GT, it is effective to modify their surface by the layer-by-layer electrostatic deposition method.
A variety of polymers, such as chitosan, pectin, alginate, etc., have been applied to coat the surface of liposomes with an impact on their functionality, namely (1) extending the circulation time, which improves the transport and release of active substances; (2) the reduction in oxygen exposure and implicitly the decrease in lipid oxidation due to the thick polymer layer; and (3) the decrease in the leakage of loaded components [34].
This review aims to provide current knowledge regarding coating materials for liposomes with applications in the pharmaceutical and food fields and their influence on the stability and absorption of liposomes.

2. Physical and Chemical Factors Influencing Liposome Stability after Being Obtained

The main problem with the application of liposomes at the industrial level is their low physical and chemical stability due to fragile phospholipid membranes and their peroxidation. Physical degradation can sometimes be due to changes in the structure of the liposomes.
The stability of the liposome structure influences the controlled release of active compounds, both in the bloodstream and in the intestinal mucosa, depending on the route of liposome administration (injectable or oral). Liposome stability is influenced by physical factors (storage temperature and light) and chemical factors (lipid peroxidation and variations in pH).

2.1. Physical Factors

Storage temperature is an important factor that influences the stability of liposomes. Studies have shown that liposomes with different compounds lose their stability at high temperatures. The range of experimental temperatures varied between −150 °C and 50 °C, and the testing time was between 24 h and 90 days [35,36]. Thermal liposomal stability depends on both the encapsulated active compound and the liposomal membrane structure. The thermal instability of peptide-loaded liposomes can limit their incorporation in thermally processed food [37].
The photostability of liposomes has been tested by exposing them to various sources of light (UV-A, UV-B, and UV-C) or radiation. The most common source used was sunlight exposure under different conditions for a testing period from 4 h to 6 months [38,39].
Details regarding the influence of physical factors on the stability of the liposomes are given in Table 1.
As shown in Table 1, the liposomes were degraded by almost 36% by irradiation with various UV rays (UV-A, UV-B, UV-C), and the stability of liposomes under direct natural sunlight decreased by 25%.
The photostability of liposomes is reduced or degraded due to the breakdown of lipids caused by exposure to direct sunlight, and fluorescent or ultraviolet lights that can cause the oxidation of lipids and other compounds. Photostability can alter the structure of liposomes by affecting the interaction between their hydrophobic and hydrophilic regions. This alteration can influence the permeability, stability, and other properties of the liposome.

2.2. Chemical Factors

The chemical stability of liposomes can be influenced by the oxidation process of the phospholipid components (lipid peroxidation), which can occur as a result of the obtaining process (purification), sterilization, or even storage conditions [49]. Lipid peroxidation refers to the oxidative degeneration of lipids, characterized by forming free radicals in the lipid tails, such as cyclic peroxides and hydroperoxides [50]. The free radicals from fatty acids function as intrinsic compounds, unsaturated fatty acids being more susceptible than saturated fatty acids to this process [51].
During the obtaining process, oxidation can occur in different stages, depending on the method used. The methods that involve the use of organic solvents (ethanol, methanol, chloroform, ether, and methylene chloride) facilitate the process of molecular dispersion of lipids, and encapsulation is thus conducted with high efficiency. The destabilization and initiation of the oxidation process are influenced by the temperature at which residual organic solvents are removed in the final stage [52,53].
Since the most common route of administration of various compounds encapsulated in liposomes is oral, they must be sterilized by various processes including steam heating (autoclaving), ultraviolet and gamma ionizing irradiation, chemicals, and filtration methods. All of these methods can lead to the oxidation of the liposomal membrane [54]. For example, steam heating results in phase structural transitions (degradation and/or leakage of the encapsulated compounds) as well as oxidation of the phospholipidic component due to the high temperatures at which it is achieved (T > 121 °C) [55]. Gamma ionizing irradiation uses a high energy ionizing power and has a strong penetration capacity. This type of sterilization can destabilize the liposomal membrane in several ways: peroxidation of lipids (unsaturated lipids), hydrolysis or lipid fragmentation components, and pH changes in the solution [56].
The stability of liposomes under storage conditions, regarding pH variations, is crucial for the delivery of pH-sensitive compounds. Liposomes are usually obtained in a neutral buffer solution so that the active compounds are also in a neutral medium after incorporation into the liposomes [57]. Experiments were performed in which the liposomes were stored under different conditions of pH 2.5–10.5 for 30 min to 30 days to determine the optimal storage pH range to maintain long-term stability. The studies regarding the influence of pH on liposome stability are given in Table 2.
As shown in Table 2, the stability of liposomes decreases by 50% when the pH of the solution is acidic and by 20% when the pH increases above neutral conditions. The pH of the liposomal solution can affect the stability, size, and permeability of the liposomes as well as the release rate of the substances embedded in the liposomes.
The researchers have shown that the stability of liposomes is maintained if certain conditions of obtaining and storage are respected. The liposomal solution pH must be neutral; the production and sterilization methods used have to avoid the use of organic solvents and lipid peroxidation processes. The liposomes obtained must be kept refrigerated, at a neutral pH, and protected from direct sunlight.
The liposome membranes can be protected from the process of lipid peroxidation or this type of reaction can be minimized when liposomes are kept under inert gases such as nitrogen or argon (there is practically minimal exposure to oxygen) or if the liposomes are lyophilized before storage. Companies such as LipoCellTech™ (Soest, The Netherlands) and LivOn labs (United States of America) maintain the storage stability of their liposomal commercial products by the lyophilization process or by maintaining the liposomes in a semiliquid form with the addition in the composition of thickeners and/or emulsifier agents (xanthan gum and Tween ™ 80).
Peroxidation can also be minimized by storing liposome formulations in light-resistant containers or by removing heavy metals that may be present as a result of the obtaining process [60]. In addition, the addition of cholesterol to obtain the liposomes leads to rigidifying of the phospholipid bilayers and thus inhibits lipid peroxidation induced by the addition of copper in the reaction medium [61].

3. 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 [62]. 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 3.
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 [123].
Following the use of various saccharides (chitosan, alginate, pectin, starch, and others) and their derivatives as coatings, researchers observed improvements in the structural and physical-chemical properties of liposomes as well as an increase in the biochemical stability related to biological stimuli, such as pH, osmotic pressure, ionic strength, and temperature. A prolonged release of active substances has been observed with increased bioavailability and biostability for saccharide-coated liposomes compared to uncoated liposomes [57,68,73].
Chitosan is one of the most exploited polymers in biomedical science for drug delivery, gene delivery, and tissue engineering. Its main advantages are its mucoadhesive capacity, stability of labile drugs in the GT, bioavailability, and the controlled release of the drug [124].
The use of chitosan as a coating material has also several limitations; as it increases the size of the vesicles and the viscosity in the liposomal solution, it requires a low pH for its solubility (it is insoluble at the physiological pH of 7.4), and it is also difficult to remove excess chitosan from the liposome surface [125,126,127,128,129,130].
Physical-chemical changes in the structure of chitosan (i.e., molecular weight, crosslinking with anions, dextran, sulfates, tripolyphosphate, or covalent binding of functional groups) can contribute to overcoming limitations and extend its use in many applications. Interest in the structural changes in chitosan has led to the development, in recent years, of groups of derivatives with improved chemical, biological, and functional properties. The solubility of chitosan was increased by the introduction of alkyl (hydroxypropyl or carboxymethyl) groups into its structure. The trimethylation of the primary amino groups of chitosan improved its mucoadhesive properties and reduced trans-epithelial electrical resistance, while thiolation enhanced the gelling properties and permeability [124,131].
Among the recent applications of chitosan derivatives, we mention a few. Thiolated chitosan liposomes loaded with insulin, administered orally, have been tested under in vitro and in vivo conditions, with promising results in intestinal epithelial mucosal biodistribution and bioavailability [132]. Trimethyl chitosan nanoparticles in combination with fucoidan (hypoglycemic) inhibited α-glucosidase activity [133]. Chitosan crosslinking with sodium tripolyphosphate liposomes containing carvedilol exhibited a better bio-accessibility and antihypertensive effect [134].
The main challenges in the future development of chitosan-based liposomes (chitosomes) are safety and site-specific drug targeting.
Very rigid xanthan gum promotes the static stability of liposomes by resisting the Brownian motion of the lipid droplets, as an additional benefit, while the flexible guar gum prevents contact between the liposome vesicles. Their synergistic effects have been shown to improve the stability of the liposomal system during long-term storage. The use of gums as coating materials has the disadvantage of obtaining a viscous system, requiring more attention to the concentration of polysaccharides because, at high gum concentrations, the gel network could be damaged and, consequently, there may be an agglomeration of liposome vesicles and leakage of the bioactive substance [51,135]. Xanthan gum, as an anionic dietary fiber, can inhibit lipid oxidation by transferring its ability to bind iron ions (the process occurs faster at a pH of 3.5 than at a pH of 7, which may be due to the increased solubility of iron under acidic conditions), but a significant increase in the rate of lipid aggregation was observed under gastrointestinal conditions [136].
Alginate-coated liposomes have improved stability in the gastric environment, but in some cases, the interactions between them significantly alter the permeability of the membrane; calcium ions seem to induce a higher drug leakage than sodium ions [137].
Polymers are widely used as a coating material because they are more stable than phospholipids, and their properties can be imprinted on liposomes by controlled synthesis. The advantage of using polymers is their intercalation in the lipid secondary layer due to their varied block structure and because they reduce the predisposition of lipids to oxidative degradation [138]. Polymers are preferred as liposome coatings because of their improved mucoadhesive properties that can prolong the retention and enhance the adhesion and penetration of drugs through the gastrointestinal barriers [139]. For example, liposomes coated with charged polymers, such as poly-l-lysine or poly-l-arginine, are electrostatically repellent and stable in colloidal systems; the coating also serves as a barrier that controls the release rate of active compounds. However, a strong polymer–liposome interaction can cause leakage of the encapsulated active compounds, while a weak attachment can cause the polymer to detach after injection, leading to a shorter circulation time [140].
Recent research pursuing the modification and coating of liposome membranes by using film-forming polymers (PEG-polyethylene glycol, Eudragit EPO, poly (L-lysine), and others) has led to increased stability in the organism, showing properties of precise targeting and delayed release of drugs. PEG has been the most widely used coating material for liposomes carrying antitumor drugs due to its hydrophilic properties, molecular flexibility, and neutrality, which can prolong circulation time and reduce reticuloendothelial system (RES) clearance [141].
However, recent studies have shown that PEGylated products may cause an immune response (both intravenous and oral), such as hypersensitivity, cytoplasmic vacuolation, accelerated blood clearance, and antibody production under certain conditions. Recently, several potential safety issues have been raised from the repeated administration of these types of liposomal products because the use of high molecular weight PEG, which human enzymes cannot effectively degrade, leads to its accumulation in the body.
The use of lower molecular weight PEG has various toxic side effects. Liposomal drug formulations and their storage stability may be influenced by PEG’s instability under conditions of exogenous stress, such as heat, radiation, or mechanical forces [142,143,144,145].
A new kind of PEG derivative, which has two ends grafted with cholesterol (cholesterol–PEG–cholesterol, CPC), was reported by Wang, et al. [146]. This derivative reduces the uptake by phagocyte cells and extends the circulation time. Sadzuka, et al. [147] showed that a reduction in nanoparticle opsonization can be achieved by combining PEG with different-sized chains that modify the conformation of the polymer on the surface of the liposomes.
Many polymers have been proposed to stabilize liposomes as an alternative to PEGylation: polyglycerol-modified liposomes, the use of super-hydrophilic zwitterionic polymers such as poly (carboxy betaine), and a triblock non-ionic surfactant (Pluronic F127), largely used as a food and drug additive [148,149,150].
Lane, et al. [151] hypothesized that glycosaminoglycan (GAG) heparosan (HEP; [-4-GlcA-β1-4-GlcNAc-α1-] n), a natural polysaccharide, may serve as a PEG alternative for coating liposomes. The coated drug-carrying liposomes with HEP are protected from the mononuclear phagocyte system, extending liposome circulation time and potentially avoiding immune-mediated clearance.
Liposomes coated with natural macromolecules (natural biodegradable proteins), which are recognized by the body, can change the shield surface and the layers of the liposome membrane, having the same binding way, being a good alternative for the replacement of PEG coating materials [152].
Proteins (albumin, gelatin, silk fibroin (SF), and others) used as liposome coatings and drug-binding molecules improved the long blood retention of the active compounds and sustained permeation along with a high capacity of controlled targeting [96,100,101].
By combining groups of coating materials (chitosan–pectin, pectin–whey protein, PEG–chitosan, and others) the physical-chemical properties such as solubility, thermal protection, resistance to oxidative stress, and adequate cell protection have been improved [106,107,111,116].
Liposomes coated with dimethyl amino methyl-dextran, silica, and ceramics showed better skin permeation and oxygen protection along with increased photovoltaic stability and anti-interference capacity [85,119,120,122].
Challenges in using coating materials to improve liposome stability in drug delivery include controlling the uniformity of the coating, optimizing the surface area-to-volume ratio, and ensuring that the coating is compatible with the lipid components of the liposomes. This can be complicated due to the variability and complexity of natural lipids. In addition, certain coating materials may cause toxicity or other undesirable effects, which is a potential problem when they are used in pharmaceutical applications. Therefore, experimental methods are required to ensure that the coating materials provide the desired stability.

4. 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 [153].
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 [154].
The general coating materials for liposomes with applications in the food industry and their properties are illustrated in Table 4.
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 [82,155,156].
Some researchers [108] 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 [161,162].
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 [163]. 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 [154,163]. 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 [93,94]. 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 [108,113,158,159].
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.

5. 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 [164].
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 [165].
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 [34,166,167]. 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 [168].
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) [169].
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 [170,171].
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 [172].
The role of chitosan in liposome digestion and absorption is still controversial. Some authors [172,173] 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 [174].
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 [175,176].
The polymer coating is a promising way to modify the surface characteristics of the vesicle’s stability to improve its applicability [177].
The liposome content is delivered in the cell by four mechanisms [178]. 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 [179,180]. 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 [181].
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 [168,182]. 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 [183].
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 [184].
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 [185].
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 [186].
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 [187].
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 [188,189].
Therefore, instead of studying only one component, different pharmacokinetic parameters of both components, i.e., the drug and the carrier, should be used. Several coated liposome formulations are being investigated in various stages of clinical trials for medical applications; recently, several coated liposome formulations have been used in ongoing clinical trials. These clinical trials and their applications are listed in Table 5.
As the clinical trials conducted with coated liposomes are at different stages, it can be stated that each study mainly investigates the occurrence of adverse effects after the administration of liposomal formulations. According to the ADME literature studies, events such as low oral bioavailability, unfavorable clearance of nanoparticles by RES, excretion of the drug via urine in parenteral administration, and entrapment and elimination of circulating carriers by opsonization are the typical obstacles encountered in many routes of administration and the first processes analyzed.
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.

6. Patent Applications and Patents on Coated Liposomes

There are recent patent applications related to coated liposomes, but not all of them have been granted as patents. In addition, most of these patents relate to pharmaceuticals and not to the food industry, as different legal requirements apply to patenting a drug delivery system. Patents on food delivery systems are less common and often require additional evidence of the efficacy of such systems before they can be granted. Some of the recent patent applications and patents involving coated liposomes are listed in Table 6.
It is anticipated that patents regarding coated liposomes with applications in the food industry will be published in the future due to the lack of current representation in this sector.

7. Liposome Commercial Products Available on the Market

The purpose of encapsulating active compounds is to maximize their bioavailability and health benefits through controlled release. Therefore, there is a growing demand for smart delivery systems that allow controlled delivery at the right time and place; the only essential requirement is that this delivery system is produced depending on the route of administration.
The liposome products available on the market showed greater stability, biological efficacy, and health benefits due to their synergistic properties [190]. A short selection of commercial products containing different active compounds encapsulated in liposomes are illustrated in Table 7.
The encapsulation technologies used in the development of liposomal pharmaceuticals are stealth liposome technology, DepoFoam™ technology, heat-sensitive liposomes, and non-PEGylated liposome technologies. Only in stealth technology (PEGylation), the structure of the liposome membrane is modified or coated, and the polymer used is polyethylene glycol (PEG). This technology is the most common for the development of cancer therapy drugs (such as Doxil and OnivydeTM) because it makes it difficult for mononuclear phagocytes to detect the liposomes [191,192,193,194].
Liposomal food supplements or food products are obtained by technologies that are designed to protect the liposomes from heat, high pressure, or chemicals. Hypernatura and LivOn labs use LIPOCELLTECH™ or cold-process liposomes for the food liposomal supplements obtained. The liposomes obtained by these technologies do not use a coating material [195,196].
Currently, there are only two food products on the market, in the form of functional teas, which contain active ingredients encapsulated in liposomes, obtained by the technology described in the patent of the company Bio-Up Mimetic Technologies Inc. The liposomes have no coating, but the method of obtaining them has provided optimal stability and high efficiency from an economic point of view [197].

8. Conclusions

Liposomes, due to their biocompatibility and encapsulation capacity, are a promising delivery system for different compounds. The integrity of phospholipid membranes ensures the stability of liposomes. The rate and degree of peroxidation and hydrolysis of phospholipids are major factors that determine the shelf life and performance of liposomes for medical or food applications. The hydrolysis of phospholipids can especially affect cholesterol-free liposomes used, in particular, in thermosensitive liposome formulations [198]. Physical and chemical factors decrease the integrity of lipid bilayers, and the liposomes are destabilized.
The most used coating material for liposomes with pharma applications is PEG, but considering the accumulation effect in the body—due to the frequent use of liposomes coated with PEG—new materials are sought.
In the food field, saccharides, their derivatives, and proteins are frequently studied as liposomal coating materials for their nontoxicity and neutral characteristics.
Stability is important for liposomes because, depending on the degree of stability, the compounds transported by liposomes reach the site where absorption takes place. If stability is low, then a large part of the compound is degraded along the route, and if stability is very high, then the compound cannot be released. This makes stability crucial in achieving the purpose for which the compound has been encapsulated in liposomes.
However, most of the recent studies on liposomal surface modification, particularly for phospholipid modification, are related to the pharmaceutical, medical, and cosmetic industries. Research on the in vitro digestion stability or interaction mechanism of liposomes with surface modifications in food technology is very rare.

9. Outlook

In recent years, different types of liposomes coated with various materials have been developed to increase the stability, protection, and controlled/targeted release of active compounds. These liposomes can be used in pharmaceutical, food, and cosmetic applications to increase the bioavailability of nutrients. Better solutions are still needed to improve the quality and shelf life of liposomes containing functional compounds.
A big challenge for liposome production at the industrial level is to maintain their optimal stability both in terms of physical factors (maintaining optimal conditions for manufacturing and storage), but also in terms of chemical factors (oxidation reactions that may occur during the obtaining process). However, it must also be taken into account that the technological process of obtaining liposomes needs to be economically efficient with a low number of nontoxic chemical reagents used.
Another aspect to consider is that liposomes, regardless of the route of administration, have to be protected from the immune system’s rapid reaction and gain increased stability against the low pHs in the stomach for the targeted delivery of active compounds.
The number of pharma-liposomal products on the market is higher compared to liposomal food supplements and nutraceuticals. As for food products that contain liposomes, they are at the laboratory research level with a very small number of products on the market. The liposomal beverages on the market are obtained without thermal processing and are kept in refrigerated conditions (optimal conditions for maintaining the stability of the liposomes). An additional challenge is to diversify the range of liposome-containing foods, which are obtained through a production technology that involves thermal processes, high pressure, or additional sterilization, such as bars, breakfast cereals, and instant noodles.
In the future, it is targeted to achieve the development at the industrial level of solid food products containing liposomes and their commercialization as well as the increase in liposome stability through the development of new coating materials.

Author Contributions

D.P., A.-I.G. and C.E.E. wrote the manuscript, participated in the conceptualization, and the review and editing of the manuscript. C.B.M., C.B., L.P.-P. and P.-A.V. performed the literature review. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant from the Ministry of Research, Innovation and Digitization, CNCS/CCCDI-UEFISCDI, project number PN-III-P3-3.5-EUK-2019-0169, within PNCDI III; this work was carried out through the PN 23.06 Core Program—ChemNewDeal within the National Plan for Research, Development and Innovation 2022-2027, developed with the support from Ministry of Research, Innovation, and Digitization, project no. PN 23.06.01.01.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  9. Subramanian, P. Lipid-based nanocarrier system for the effective delivery of nutraceuticals. Molecules 2021, 26, 5510. [Google Scholar] [CrossRef]
  10. Arpita, S.; Kumar, S.V. Liposomes–A Review. Int. J. Indig. Herbs Drugs 2020, 5, 1–6. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef] [PubMed]
  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. [Google Scholar] [CrossRef]
  17. Trucillo, P.; Reverchon, E. Production of PEG-coated liposomes using a continuous supercritical assisted process. J. Supercrit. Fluids 2021, 167, 105048. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef] [PubMed]
  26. Ahmad, A.; Ahsan, H. Lipid-based formulations in cosmeceuticals and biopharmaceuticals. Biomed. Dermatol. 2020, 4, 1–10. [Google Scholar] [CrossRef]
  27. Zarrabi, A.; Abadi, M.A.; Khorasani, S.; Mohammadabadi, M.R.; Jamshidi, A.; Torkaman, S.; Taghavi, E.; Mozafari, M.R.; Rasti, B. Nanoliposomes and tocosomes as multifunctional nanocarriers for the encapsulation of nutraceutical and dietary molecules. Molecules 2020, 25, 638. [Google Scholar] [CrossRef]
  28. Padilla, C.E.; Villanueva, F.J.E. Encapsulation of food active ingredients in liposomes. J. Nutr. Health. Food. Eng. 2018, 8, 238–239. [Google Scholar] [CrossRef]
  29. Liu, W.; Hou, Y.; Jin, Y.; Wang, Y.; Xu, X.; Han, J. Research progress on liposomes: Application in food, digestion behavior and absorption mechanism. Trends Food Sci. Technol. 2020, 104, 177–189. [Google Scholar] [CrossRef]
  30. 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]
  31. Lee, D.E.; Lew, M.G.; Woodbury, D.J. Vesicle fusion to planar membranes is enhanced by cholesterol and low temperature. Chem. Phys. Lipids 2013, 166, 45–54. [Google Scholar] [CrossRef]
  32. Sepúlveda, C.T.; Alemán, A.; Zapata, J.E.; Montero, M.P.; Gómez-Guillén, M.C. Characterization and storage stability of spray dried soy-rapeseed lecithin/trehalose liposomes loaded with a tilapia viscera hydrolysate. Innov. Food Sci. Emerg. Technol. 2021, 71, 102708. [Google Scholar] [CrossRef]
  33. Boafo, G.F.; Magar, K.T.; Ekpo, M.D.; Qian, W.; Tan, S.; Chen, C. The Role of Cryoprotective Agents in Liposome Stabilization and Preservation. Int. J. Mol. Sci. 2022, 23, 12487. [Google Scholar] [CrossRef]
  34. 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. [Google Scholar] [CrossRef]
  35. Sydykov, B.; Oldenhof, H.; Sieme, H.; Wolkers, W.F. Storage stability of liposomes stored at elevated subzero temperatures in DMSO/sucrose mixtures. PLoS ONE 2018, 13, e0199867. [Google Scholar] [CrossRef] [PubMed]
  36. Ioele, G.; Grande, F.; De Luca, M.; Occhiuzzi, M.A.; Garofalo, A.; Ragno, G. Photodegradation of anti-inflammatory drugs: Stability tests and lipid nanocarriers for their photoprotection. Molecules 2021, 26, 5989. [Google Scholar] [CrossRef] [PubMed]
  37. Mohan, A.; Rajendran, S.R.; He, Q.S.; Bazinet, L.; Udenigwe, C.C. Encapsulation of food protein hydrolysates and peptides: A review. RSC Adv. 2015, 5, 79270–79278. [Google Scholar] [CrossRef]
  38. Petrović, S.; Tačić, A.; Savić, S.; Nikolić, V.; Nikolić, L.; Savić, S. Sulfanilamide in solution and liposome vesicles; in vitro release and UV-stability studies. Saudi Pharm. J. 2017, 25, 1194–1200. [Google Scholar] [CrossRef]
  39. Huang, J.; Wang, Q.; Chu, L.; Xia, Q. Liposome-chitosan hydrogel bead delivery system for the encapsulation of linseed oil and quercetin: Preparation and in vitro characterization studies. LWT Food Sci. Technol. 2020, 117, 108615. [Google Scholar] [CrossRef]
  40. Lin, X.; Li, B.; Wen, J.; Wu, J.; Tang, D.; Yu, Y.; Xu, Y.; Xu, B. Storage Stability and In Vitro Bioaccessibility of Liposomal Betacyanins from Red Pitaya (Hylocereus polyrhizus). Molecules 2022, 27, 1193. [Google Scholar] [CrossRef]
  41. Pentak, D. In vitro spectroscopic study of piperine-encapsulated nanosize liposomes. Eur. Biophys. J. 2016, 45, 175–186. [Google Scholar] [CrossRef]
  42. Ghareaghajlou, N.; Hallaj-Nezhadi, S.; Ghasempour, Z. Nano-liposomal system based on lyophilization of monophase solution technique for encapsulating anthocyanin-rich extract from red cabbage. Dye. Pigment. 2022, 202, 110263. [Google Scholar] [CrossRef]
  43. Guldike, B.; Gibi, 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]
  44. Xia, H.; Tang, Y.; Huang, R.; Liang, J.; Ma, S.; Chen, D.; Feng, Y.; Lei, Y.; Zhang, Q.; Yang, Y.; et al. Nanoliposome Use to Improve the Stability of Phenylethyl Resorcinol and Serve as a Skin Penetration Enhancer for Skin Whitening. Coatings 2022, 12, 362. [Google Scholar] [CrossRef]
  45. Wu, Y.; Wang, K.; Liu, Q.; Liu, X.; Mou, B.; Lai, O.M.; Tan, C.P.; Cheong, L.Z. Selective antibacterial activities and storage stability of curcumin-loaded nanoliposomes prepared from bovine milk phospholipid and cholesterol. Food Chem. 2022, 367, 130700. [Google Scholar] [CrossRef] [PubMed]
  46. Almurshedi, A.S.; Radwan, M.; Omar, S.; Alaiya, A.A.; Badran, M.M.; Elsaghire, H.; Saleem, Y.I.; Hutcheon, G.A. A novel pH-sensitive liposome to trigger delivery of afatinib to cancer cells: Impact on lung cancer therapy. J. Mol. Liq. 2018, 259, 154–166. [Google Scholar] [CrossRef]
  47. Tai, K.; Rappolt, M.; Mao, L.; Gao, Y.; Li, X.; Yuan, F. The stabilization and release performances of curcumin-loaded liposomes coated by high and low molecular weight chitosan. Food Hydrocoll. 2020, 99, 105355. [Google Scholar] [CrossRef]
  48. Alkhatib, D.; Zelai, N. Preparation, characterization and stability of silver sulfadiazine nanoliposomes. Trop. J. Pharm. Res. 2021, 20, 665–671. [Google Scholar] [CrossRef]
  49. Lombardo, D.; Kiselev, M.A. Methods of Liposomes Preparation: Formation and Control Factors of Versatile Nanocarriers for Biomedical and Nanomedicine Application. Pharmaceutics 2022, 14, 543. [Google Scholar] [CrossRef]
  50. Rems, L.; Viano, M.; Kasimova, M.A.; Miklavčič, D.; Tarek, M. The contribution of lipid peroxidation to membrane permeability in electropermeabilization: A molecular dynamics study. Bioelectrochemistry 2019, 125, 46–57. [Google Scholar] [CrossRef]
  51. Tan, C.; Wang, J.; Sun, B. Biopolymer-liposome hybrid systems for controlled delivery of bioactive compounds: Recent advances. Biotechnol. Adv. 2021, 48, 107727. [Google Scholar] [CrossRef]
  52. Shi, N.Q.; Qi, X.R. Preparation of Drug Liposomes by Reverse-Phase Evaporation. In Liposome-Based Drug Delivery Systems; Lu, W.L., Qi, X.R., Eds.; Biomaterial Engineering; Springer: Berlin, Germany, 2021; pp. 37–46. [Google Scholar] [CrossRef]
  53. Liu, C.; Liu, Y.Y.; Chang, Q.; Shu, Q.; Shen, N.; Wang, H.; Xie, Y.; Deng, X. Pressure-Controlled Encapsulation of Graphene Quantum Dots into Liposomes by the Reverse-Phase Evaporation Method. Langmuir 2021, 37, 14096–14104. [Google Scholar] [CrossRef]
  54. Delma, K.L.; Lechanteur, A.; Evrard, B.; Semdé, R.; Piel, G. Sterilization methods of liposomes: Drawbacks of conventional methods and perspectives. Int. J. Pharm. 2021, 597, 120271. [Google Scholar] [CrossRef]
  55. Qi, N.; Tang, X.; Lin, X.; Gu, P.; Cai, C.; Xu, H.; Zhang, Y. Sterilization stability of vesicular phospholipid gels loaded with cytarabine for brain implant. Int. J. Pharm. 2012, 427, 234–241. [Google Scholar] [CrossRef]
  56. Sakar, F.; Özer, A.Y.; Erdogan, S.; Ekizoglu, M.; Kart, D.; Özalp, M.; Colak, S.; Zencir, Y. Nano drug delivery systems and gamma radiation sterilization. Pharm. Dev. Technol. 2017, 22, 775–784. [Google Scholar] [CrossRef] [PubMed]
  57. 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. [Google Scholar] [CrossRef] [PubMed]
  58. Rehman, A.U.; Omran, Z.; Anton, H.; Mély, Y.; Akram, S.; Vandamme, T.F.; Anton, N. Development of doxorubicin hydrochloride loaded pH-sensitive liposomes: Investigation on the impact of chemical nature of lipids and liposome composition on pH-sensitivity. Eur. J. Pharm. Biopharm. 2018, 133, 331–338. [Google Scholar] [CrossRef] [PubMed]
  59. Shao, X.R.; Wei, X.Q.; Zhang, S.; Fu, N.; Lin, Y.F.; Cai, X.X.; Peng, Q. Effects of micro-environmental pH of liposome on chemical stability of loaded drug. Nanoscale Res. Lett. 2017, 12, 1–8. [Google Scholar] [CrossRef]
  60. Franzé, S.; Selmin, F.; Samaritani, E.; Minghetti, P.; Cilurzo, F. Lyophilization of liposomal formulations: Still necessary, still challenging. Pharmaceutics 2018, 10, 139. [Google Scholar] [CrossRef] [PubMed]
  61. Schnitzer, E.; Pinchuk, I.; Bor, A.; Leikin-Frenkel, A.; Lichtenberg, D. Oxidation of liposomal cholesterol and its effect on phospholipid peroxidation. Chem. Phys. Lipids. 2007, 146, 43–53. [Google Scholar] [CrossRef] [PubMed]
  62. 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. [Google Scholar] [CrossRef]
  63. 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. [Google Scholar] [CrossRef]
  64. 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. [Google Scholar] [CrossRef]
  65. 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. [Google Scholar] [CrossRef]
  66. 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. [Google Scholar]
  67. 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. [Google Scholar] [CrossRef] [PubMed]
  68. Tamaddon, L.; Mohamadi, N.; Bavarsad, N. Preparation and Characterization of Mucoadhesive Loratadine Nanoliposomes for Intranasal Administration. Turk. J. Pharm. Sci. 2021, 18, 492. [Google Scholar] [CrossRef] [PubMed]
  69. 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. [Google Scholar] [CrossRef]
  70. 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. [Google Scholar] [CrossRef]
  71. 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. [Google Scholar] [CrossRef]
  72. 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. [Google Scholar] [CrossRef]
  73. 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. [Google Scholar] [CrossRef]
  74. 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. [Google Scholar] [CrossRef]
  75. 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. [Google Scholar] [CrossRef]
  76. 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. [Google Scholar] [CrossRef] [PubMed]
  77. 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. [Google Scholar] [CrossRef] [PubMed]
  78. 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. [Google Scholar] [CrossRef] [PubMed]
  79. 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. [Google Scholar] [CrossRef]
  80. 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. [Google Scholar] [CrossRef]
  81. 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. [Google Scholar] [CrossRef]
  82. 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. [Google Scholar] [CrossRef]
  83. 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. [Google Scholar] [CrossRef]
  84. 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. [Google Scholar] [CrossRef]
  85. 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. [Google Scholar] [CrossRef]
  86. 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. [Google Scholar] [CrossRef] [PubMed]
  87. 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. [Google Scholar] [CrossRef] [PubMed]
  88. 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. [Google Scholar] [CrossRef]
  89. 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).
  90. 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. [Google Scholar]
  91. 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. [Google Scholar] [CrossRef] [PubMed]
  92. 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. [Google Scholar] [CrossRef]
  93. 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. [Google Scholar] [CrossRef]
  94. 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. [Google Scholar] [CrossRef]
  95. 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. [Google Scholar] [CrossRef]
  96. 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. [Google Scholar] [CrossRef]
  97. 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. [Google Scholar] [CrossRef] [PubMed]
  98. 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. [Google Scholar] [CrossRef] [PubMed]
  99. 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. [Google Scholar] [CrossRef]
  100. 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. [Google Scholar] [CrossRef] [PubMed]
  101. 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. [Google Scholar] [CrossRef]
  102. 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. [Google Scholar] [CrossRef]
  103. 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. [Google Scholar] [CrossRef]
  104. 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. [Google Scholar] [CrossRef]
  105. 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. [Google Scholar] [CrossRef]
  106. 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. [Google Scholar] [CrossRef]
  107. 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. [Google Scholar] [CrossRef]
  108. 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. [Google Scholar] [CrossRef]
  109. 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. [Google Scholar] [CrossRef] [PubMed]
  110. 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. [Google Scholar] [CrossRef]
  111. 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. [Google Scholar] [CrossRef]
  112. Islam, N.; Ferro, V. Recent advances in chitosan-based nanoparticulate pulmonary drug delivery. Nanoscale 2016, 8, 14341–14358. [Google Scholar] [CrossRef]
  113. 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. [Google Scholar] [CrossRef]
  114. 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. [Google Scholar] [CrossRef]
  115. 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. [Google Scholar] [CrossRef] [PubMed]
  116. 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. [Google Scholar] [CrossRef] [PubMed]
  117. 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. [Google Scholar] [CrossRef]
  118. 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. [Google Scholar] [CrossRef]
  119. 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. [Google Scholar] [CrossRef] [PubMed]
  120. 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. [Google Scholar] [CrossRef]
  121. 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. [Google Scholar] [CrossRef]
  122. 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. [Google Scholar] [CrossRef]
  123. 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. [Google Scholar] [CrossRef]
  124. Mikušová, V.; Mikuš, P. Advances in chitosan-based nanoparticles for drug delivery. Int. J. Mol. Sci. 2021, 22, 9652. [Google Scholar] [CrossRef]
  125. Jøraholmen, M.W.; Bhargava, A.; Julin, K.; Johannessen, M.; Škalko-Basnet, N. The antimicrobial properties of chitosan can be tailored by formulation. Mar. Drugs 2020, 18, 96. [Google Scholar] [CrossRef]
  126. 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. [Google Scholar] [CrossRef]
  127. 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. [Google Scholar] [CrossRef]
  128. 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. [Google Scholar] [CrossRef]
  129. 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]
  130. 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. [Google Scholar] [CrossRef]
  131. Kumar, S.; Deepak, V.; Kumari, M.; Dutta, P.K. Antibacterial activity of diisocyanate-modified chitosan for biomedical applications. Int. J. Biol. Macromol. 2016, 84, 349–353. [Google Scholar] [CrossRef] [PubMed]
  132. Sudhakar, S.; Chandran, S.V.; Selvamurugan, N.; Nazeer, R.A. Biodistribution and pharmacokinetics of thiolated chitosan nanoparticles for oral delivery of insulin in vivo. Int. J. Biol. Macromol. 2020, 150, 281–288. [Google Scholar] [CrossRef]
  133. Tsai, L.C.; Chen, C.H.; Lin, C.W.; Ho, Y.C.; Mi, F.L. Development of multifunctional nanoparticles self-assembled from trimethyl chitosan and fucoidan for enhanced oral delivery of insulin. Int. J. Biol. Macromol. 2019, 126, 141–150. [Google Scholar] [CrossRef]
  134. Sharma, M.; Sharma, R.; Jain, D.K.; Saraf, A. Enhancement of oral bioavailability of poorly water-soluble carvedilol by chitosan nanoparticles: Optimization and pharmacokinetic study. Int. J. Biol. Macromol. 2019, 135, 246–260. [Google Scholar] [CrossRef]
  135. Chaves, M.A.; Filho, P.L.O.; Jange, C.G.; Sinigaglia-Coimbra, R.; Oliveira, C.L.P.; Pinho, S.C. Structural characterization of multilamellar liposomes coencapsulating curcumin and vitamin D3. Colloids Surf. A Physicochem. Eng. Asp. 2018, 549, 112–121. [Google Scholar] [CrossRef]
  136. Qiu, C.; Zhao, M.; Decker, E.A.; McClements, D.J.; Qiu, C.; Zhao, M.; Decker, E.A.; McClements, D.J. Influence of anionic dietary fibers (xanthan gum and pectin) on oxidative stability and lipid digestibility of wheat protein-stabilized fish oil-in-water emulsion. Food Res. Int. 2015, 74, 131–139. [Google Scholar] [CrossRef]
  137. Grijalvo, S.; Mayr, J.; Eritja, R.; Díaz, D.D. Biodegradable liposome-encapsulated hydrogels for biomedical applications: A marriage of convenience. Biomater. Sci. 2016, 4, 555–574. [Google Scholar] [CrossRef]
  138. De Leo, V.; Milano, F.; Agostiano, A.; Catucci, L. Recent advancements in polymer/liposome assembly for drug delivery: From surface modifications to hybrid vesicles. Polymers 2021, 13, 1027. [Google Scholar] [CrossRef] [PubMed]
  139. Sahatsapan, N.; Pamornpathomkul, B.; Rojanarata, T.; Ngawhirunpat, T.; Poonkhum, R.; Opanasopit, P.; Patrojanasophon, P. Feasibility of mucoadhesive chitosan maleimide-coated liposomes for improved buccal delivery of a protein drug. J. Drug Deliv. Sci. Tec. 2022, 69, 103173. [Google Scholar] [CrossRef]
  140. Cao, Y.; Dong, X.; Chen, X. Polymer-Modified Liposomes for Drug Delivery: From Fundamentals to Applications. Pharmaceutics 2022, 14, 778. [Google Scholar] [CrossRef] [PubMed]
  141. Torchilin, V.P.; Narula, J.; Khaw, B.A.; Trubetskoy, V.S. PEG-modified liposomes for gamma-and magnetic resonance imaging. In Stealth Liposomes; Lasic, D.D., Martin, F.J., Eds.; CRC Press: Boca Raton, FL, USA, 2018; pp. 247–260. [Google Scholar]
  142. Mastrotto, F.; Brazzale, C.; Bellato, F.; De Martin, S.; Grange, G.; Mahmoudzadeh, M.; Caliceti, P. In vitro and in vivo behavior of liposomes decorated with PEGs with different chemical features. Mol. Pharm. 2019, 17, 472–487. [Google Scholar] [CrossRef]
  143. Abbina, S.; Parambath, A. PEGylation and its alternatives: A summary. In Engineering of Biomaterials for Drug Delivery Systems: Beyond Polyethylene Glycol; Parambath, A., Ed.; Woodhead Publishing Series in Biomaterials; Woodhead: Duxford, UK, 2018; pp. 363–376. [Google Scholar] [CrossRef]
  144. Hatakeyama, H.; Akita, H.; Harashima, H. The polyethyleneglycol dilemma: Advantage and disadvantage of PEGylation of liposomes for systemic genes and nucleic acids delivery to tumors. Biol. Pharm. Bull. 2013, 36, 892–899. [Google Scholar] [CrossRef]
  145. Knop, K.; Hoogenboom, R.; Fischer, D.; Schubert, U.S. Poly (ethylene glycol) in drug delivery: Pros and cons as well as potential alternatives. Angew. Chem. Int. Ed. 2010, 49, 6288–6308. [Google Scholar] [CrossRef]
  146. Wang, Y.; Wang, J.; Sun, M.; Zhang, J.; Bi, Y. Coating liposomes with ring-like PEG: The synthesis and stealth effect of cholesterol–PEG–cholesterol. Mater. Adv. 2022, 3, 2417–2424. [Google Scholar] [CrossRef]
  147. Sadzuka, Y.; Nakade, A.; Hirama, R.; Miyagishima, A.; Nozawa, Y.; Hirota, S.; Sonobe, T. Effects of mixed polyethyleneglycol modification on fixed aqueous layer thickness and antitumor activity of doxorubicin containing liposome. Int. J. Pharm. 2002, 238, 171–180. [Google Scholar] [CrossRef]
  148. Lila, A.S.A.; Nawata, K.; Shimizu, T.; Ishida, T.; Kiwada, H. Use of polyglycerol (PG), instead of polyethylene glycol (PEG), prevents induction of the accelerated blood clearance phenomenon against long-circulating liposomes upon repeated administration. Int. J. Pharm. 2013, 456, 235–242. [Google Scholar] [CrossRef]
  149. Lin, W.; Kampf, N.; Goldberg, R.; Driver, M.J. Poly-phosphocholinated liposomes form stable superlubrication vectors. Langmuir 2019, 35, 6048–6054. [Google Scholar] [CrossRef]
  150. de Morais, F.A.P.; Gonçalves, R.S.; Braga, G.; Calori, I.R.; Pereira, P.C.S.; Batistela, V.R.; Caetano, W.; Hioka, N. Stable dipalmitoylphosphatidylcholine liposomes coated with an F127 copolymer for hypericin loading and delivery. ACS Appl. Nano. Mater. 2020, 3, 4530–4541. [Google Scholar] [CrossRef]
  151. Lane, R.S.; Haller, F.M.; Chavaroche, A.A.; Almond, A.; DeAngelis, P.L. Heparosan-coated liposomes for drug delivery. Glycobiology 2017, 27, 1062–1074. [Google Scholar] [CrossRef] [PubMed]
  152. Gaber, M.; Medhat, W.; Hany, M.; Saher, N.; Fang, J.Y.; Elzoghby, A. Protein-lipid nanohybrids as emerging platforms for drug and gene delivery: Challenges and outcomes. J. Control. Release 2017, 254, 75–91. [Google Scholar] [CrossRef] [PubMed]
  153. 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. [Google Scholar] [CrossRef]
  154. 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. [Google Scholar] [CrossRef]
  155. 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. [Google Scholar] [CrossRef]
  156. 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. [Google Scholar] [CrossRef]
  157. 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. [Google Scholar] [CrossRef]
  158. 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. [Google Scholar] [CrossRef]
  159. 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. [Google Scholar] [CrossRef]
  160. 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. [Google Scholar] [CrossRef]
  161. 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. [Google Scholar] [CrossRef]
  162. 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. [Google Scholar] [CrossRef]
  163. 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. [Google Scholar] [CrossRef] [PubMed]
  164. 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. [Google Scholar] [CrossRef] [PubMed]
  165. Wu, W.; Lu, Y.; Qi, J. Oral delivery of liposomes. Ther. Deliv. 2015, 6, 1239–1241. [Google Scholar] [CrossRef] [PubMed]
  166. 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. [Google Scholar] [CrossRef]
  167. 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. [Google Scholar] [CrossRef]
  168. 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. [Google Scholar] [CrossRef]
  169. 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. [Google Scholar] [CrossRef]
  170. 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. [Google Scholar] [CrossRef]
  171. 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. [Google Scholar] [CrossRef]
  172. 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. [Google Scholar] [CrossRef] [PubMed]
  173. Liu, J.; Zhang, J.; Xia, W. Hypocholesterolaemic effects of different chitosan samples in vitro and in vivo. Food. Chem. 2008, 107, 419–425. [Google Scholar] [CrossRef]
  174. Garg, U.; Chauhan, S.; Nagaich, U.; Jain, N. Current advances in chitosan nanoparticles-based drug delivery and targeting. Adv. Pharm. Bull. 2019, 9, 195. [Google Scholar] [CrossRef] [PubMed]
  175. 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. [Google Scholar] [CrossRef]
  176. 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. [Google Scholar] [CrossRef]
  177. 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. [Google Scholar] [CrossRef]
  178. 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. [Google Scholar] [CrossRef]
  179. Wang, J.; Byrne, J.D.; Napier, M.E.; DeSimone, J.M. More effective nanomedicines through particle design. Small 2011, 7, 1919–1931. [Google Scholar] [CrossRef]
  180. 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. [Google Scholar] [CrossRef]
  181. 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. [Google Scholar] [CrossRef] [PubMed]
  182. 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. [Google Scholar] [CrossRef] [PubMed]
  183. 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. [Google Scholar] [CrossRef] [PubMed]
  184. 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. [Google Scholar] [CrossRef]
  185. Li, M.; Al-Jamal, K.T.; Kostarelos, K.; Reineke, J. Physiologically based pharmacokinetic modeling of nanoparticles. ACS Nano 2010, 4, 6303–6317. [Google Scholar] [CrossRef]
  186. Oberdörster, G. Safety assessment for nanotechnology and nanomedicine: Concepts of nanotoxicology. J. Intern. Med. 2010, 267, 89–105. [Google Scholar] [CrossRef]
  187. 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. [Google Scholar] [CrossRef]
  188. Moghimi, S.M.; Hunter, A.C.; Murray, J.C. Long-circulating and target-specific nanoparticles: Theory to practice. Pharmacol. Rev. 2001, 53, 283–318. [Google Scholar]
  189. 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]
  190. Bulbake, U.; Doppalapudi, S.; Kommineni, N.; Khan, W. Liposomal formulations in clinical use: An updated review. Pharmaceutics 2017, 9, 12. [Google Scholar] [CrossRef]
  191. Shen, Z.; Fisher, A.; Liu, W.K.; Li, Y. PEGylated “stealth” nanoparticles and liposomes. In Engineering of Biomaterials for Drug Delivery Systems; Parambath, A., Ed.; Woodhead Publishing: Sawston, UK, 2018; pp. 1–26. [Google Scholar] [CrossRef]
  192. Salehi, B.; Mishra, A.P.; Nigam, M.; Kobarfard, F.; Jave, Z.; Rajabi, S.; Sharifi-Rad, J. Multivesicular liposome (Depofoam) in human diseases. Iran. J. Pharm. Res. 2020, 19, 9. [Google Scholar] [CrossRef] [PubMed]
  193. Motamarry, A.; Asemani, D.; Haemmerich, D. Thermosensitive liposomes. In Liposome; IntechOpen Limited: London, UK, 2017; pp. 187–212. [Google Scholar] [CrossRef]
  194. Barenholz, Y.C. Doxil®—The first FDA-approved nano-drug: Lessons learned. J. Control. Release 2012, 160, 117–134. [Google Scholar] [CrossRef] [PubMed]
  195. Zhang, Y. U.S. Method of Making Liposomes, Liposome Compositions Made by the Methods, and Methods of Using the Same. U.S. Patent 10016389, 10 July 2018. [Google Scholar]
  196. Lipocelltech™–Next Generation Liposomes, The Netherlands. Available online: https://lipocelltech.com/#production (accessed on 15 April 2022).
  197. Crispin, E.G. Re-Oiled, and Hyper-Oiled Lecithin Carrier Vehicles. U.S. Patent 20220071198A1, 4 December 2017. [Google Scholar]
  198. Chiu, G.N.; Abraham, S.A.; Ickenstein, L.M.; Karlsson, G.; Edwards, K.; Bally, M.B. Encapsulation of doxorubicin into thermosensitive liposomes via complexation with the transition metal manganese. J. Control. Release 2005, 104, 271–288. [Google Scholar] [CrossRef] [PubMed]
Table 1. Influence of physical factors on liposome stability.
Table 1. Influence of physical factors on liposome stability.
Factors
Range/Value
Stability MeasurementsTesting TimeActive Compounds Encapsulated in LiposomesBibliography
Temperature
4 °C, 25 °C, 37 °C75.54%, 67.57%, 20.28% retention rates21 daysBetacyanin[40]
20–47 °C3.47–4.33% release20 daysPiperine[41]
−150 °C, −80 °C, −25 °C
(Liquid nitrogen)
76–90%, 75–88%, 37% retention90 daysCarboxyfluorescein[35]
4 °C, 25 °C14.25%, 40.04%
loss in nanoliposomes
21 daysRed cabbage anthocyanins[42]
Room temperature30–90% degradation of extract21 daysBlack carrot extract[43]
4 °C, 25 °C96.81%, 8.82% encapsulation efficiency 90 days Phenylethyl resorcinol[44]
4 °C, 50 °C89.43% retention rate30 daysCurcumin [45]
4 °C, 25 °C90%, 80% encapsulation efficiency30 daysAfatinib[46]
4 °C95% change rate of vesicle size21 daysCurcumin[47]
Light
UV-A (12.86 W/m2), UV-B (15 W/m2), UV-C (14.29 W/m2) irradiationInstability to UV irradiation: UV-A < UV-B < UV-C; 25.841%, 32.881%, 35.678% degradation for small unilamellar vesicles liposomes4 hFulfonamides, sulfamethoxypyridazine, sulfachloropyridazine[38]
UV light irradiance (0.35 W/m2)88% retention6 hCurcumin[47]
Natural sunlight75.21% retention rate180 days Quercetin[39]
Direct sunlight;
sun UV irradiation through window glass; outdoors under the shade
Completely degraded24 daysSilver sulfadiazine[48]
UVA irradiation100–36.5% degradation 48 hPhenylethyl resorcinol[44]
Fluorescent lamp (20 Watt) or dark49.44% improved storage stability30 daysCurcumin[45]
D65/ID65 emission Standard (artificial daylight fluorescent lamp, indoor indirect daylight)Not available30 daysAnti-inflammatory drugs[36]
Table 2. Influence of pH on liposome stability.
Table 2. Influence of pH on liposome stability.
pH ValueStability MeasurementsTesting TimeActive Compounds Encapsulated in LiposomesBibliography
3, 4.3, 5, 750%, 80%, 80%, 80 retention rates21 daysBetacyanin[48]
5.5, 6.0, 6.4, 7.4Not available30 minDoxorubicin hydrochloride[58]
5.5, 7.4Cationic liposomes 63.6%, nontargeting liposomes 28.1%, pH-sensitive liposomes 35.6%—release rate in neutral pH 7.44 h for acidic conditions, 24 h for alkaline conditionsAfatinib[46]
2.5, 5.0, 7.455%, 43%, 34% encapsulation efficiency post-incubation6 hCurcumin[59]
4.5, 7.4, 10.5Completely degraded at day 18 acidic, neutral conditions, degraded at day 30 alkaline conditions30 daysSilver sulfadiazine[48]
5, 6, 7, 8, 9>80% acidic conditions retention rate,
<50% neutral and alkaline conditions retention rate
30 daysCurcumin[45]
Table 3. Coating materials for liposomes as drug delivery means.
Table 3. Coating materials for liposomes as drug delivery means.
Coating MaterialType of Encapsulated DrugsThe Advantage of the Coating Material UsedBibliography
Saccharides and their derivatives
ChitosanImmunoglobulin/mupirocin/Curcumin/sumatriptan/mucoadhesive loratadine/pelargonidin-3 -O-glucoside
-
Increased bioactivity, bioavailability, and biostability during the oral digestion of the drug
-
Increased the capability of delaying the release of the drug in gastrointestinal simulated digestion after obtaining a high encapsulation efficiency in the liposome for the lowest particle size
-
Improved the physical properties of the liposomes
[63,64,65,66,67,68]
Poly-galacturonic acidNisin
-
Controlled release of antimicrobial peptides
-
Increased the encapsulation efficiency and thermal stability of the liposomes
[69,70,71]
Sodium alginateCalcitonin/Perilla Oil
-
Increased the resistance of the membrane structure, the bioavailability, and the prolonged release of the drug
-
Provided good chemical stability and in vitro release behavior of the active compound
[72,73]
Calcium alginateOxaliplatin/Acid Folic/Ampicillin/Metformin
-
Prolonged the release of the drug and enhanced the control of liposome dimension distribution
-
Increased entrapment efficiency and enhanced stability of liposomes
[74,75]
PectinResveratrol/Amoxicillin/Tagitinine C/Phlorizin
-
Improved the long-term release of drugs and system stability
-
Presented small-sized liposomes with a slightly slower release
-
Improved immobilization, encapsulation efficiency, as well as physical storage stability of liposomes
[57,76,77]
StarchFasudil
-
Increased the storage stability and in vitro release of the active compound
-
Enhanced the uptake of liposomes and had a high entrapment efficiency
[78]
Guar gumHydrophobic bioactive compounds/Vitamin D3
-
Improved the liposome membrane stability (reduced membrane degradation after simulated digestion)
-
Increased thermal and light stability of the liposomes with a prolonged storage stability
[79,80]
Xanthan gumDioctadecyl dimethylammonium bromide
-
Improved the release profile of the low-stability drug
[81]
Cationic inulinBetaine/carvone
-
Improved the thermal, physical, and oxidative stability of liposomes; provided in vitro sustained release of the active compound
[82]
GalactomannanAscorbic acid
-
Presented good encapsulation efficiency and improved the stability of the drug
-
Sustained the release of the active compound under gastrointestinal pH conditions
[83]
Hyaluronic acidAntitumoral drug delivery/anti-melanoma agents (dacarbazine and eugenol)
-
Reduced immune response and improved the pharmacokinetics of the drug
-
Presented a good stealth property in blood circulation and improved stability in plasma
[84]
Diethylaminomethyl-dextran Drug
-
Improved the stability of liposomes and enhanced skin permeation
[85]
Hydroxypropyl methylcelluloseSildenafil
-
Provided greater bio-adhesion and higher entrapment efficiencies for the liposomes
-
Prolonged the drug release
[86]
Polymer/Copolymer
Polyethylene glycol PEGVitexin
-
Increased the stability of long drug carriers
-
Increased the drug entrapment efficiency and obtained a delayed release
[17,87,88]
Poly (hydroxyethyl-l-asparagine)Antitumoral drugs
-
Enhanced the stability and the bioavailability of the drug
-
Improved long circulation properties and the precise targeting of drug
[89,90]
Poly(L-lysine) and poly (L-glutamic acid)Recommended for drug formulations
-
Increased liposome stability in the biological environment
[91]
Eudragit EPOCurcumin
-
Prolonged the release of the drug and improved the bioavailability upon oral administration
-
Improved the in vivo activity and enhanced the stability in the gastrointestinal tract of the active compound
[92]
Proteins
Whey proteinAstaxanthin, iron
-
Improved the physical properties of liposomes: higher thermal stability, monodisperse distribution, and high encapsulation efficiency
[93,94,95]
AlbuminVancomycin/paclitaxel/ellagic acid
-
Used as a drug-binding molecule to improve the long blood retention and biocompatibility
-
Increased the long release of the drug, showing superior deliverability for substances, high stability, effective loading content, and high capacity of controlled targeting
[96,97,98]
ZeinIndocyanine green
-
Improved the chemical stability and storage stability of liposomes
[99]
Silk fibroin (SF)Ibuprofen
-
Sustained ocular drug release and in vitro corneal permeation
[100]
GelatineArginyl-glycyl-aspartic acid/aspartic acid/Lactobacilli rhamnoses/Amphotericin B
-
Increased the efficiency of drug delivery
-
Obtained small-sized liposomes, physically stable, with high drug encapsulation efficiency
[101,102]
Collagen Dexamethasone
-
High potential for use as drug delivery systems for implant substances that can induce bone and cartilage differentiation
[103]
Combinations between groups of materials
Chitosan–gelatineω-3 PUFA/BSA
-
Enhanced the physical and chemical stability of liposomes
-
Improved the release of the active compound
[104,105]
Chitosan–sodium alginateInactivated PR8 Influenza virus/cationic liposomes/resveratrol
-
Improved the long release of the virus and improved resistance during digestion
-
Improved liposome structures
[106,107,108]
Chitosan–pectinNeohesperidin/norfloxacin
-
Increased resistance during digestion of liposomes
-
Improved the liposomes’ physical and chemical stability
[109,110]
Chitosan–arabic gum5I-1H-indole (5ID)
-
Improved the solubility and sustained the release of the antifungal drug
[111]
Chitosan–xanthan gumPulmonary drugs
-
Improved physical and chemical properties and storage stability of liposomes
-
Prolonged the release of the active compound
[112]
Chitosan–zeinPulicaria gnaphadoles (Vent) Boiss
-
Prolonged the antimicrobial and antioxidant activity of the active compound
-
Controlled release of the active compound
[113]
PEG–chitosanDoxorubicin/Stearoyl spermine
-
Increased stability and prolonged the release of the drug
[114]
Pectin–whey proteinNegatively and positively charged liposomes
-
Improved the physical and chemical stability of liposomes
-
Protected the liposomes during gastrointestinal digestion
[115]
Pectin–polygalacturonic acidNisin
-
Sustained the release of the active compound and improved the physical and chemical properties
[71]
Whey protein, xanthan gum, tragacanth, arabic gum, and sodium alginateGingerol
-
Provided proper cell protection against oxidative stress
-
Increased the stability of the liposomes during storage
-
The liposomes maintained their structures in acidic and neutral media
[116]
Other
Genipin (glycoside, cross-linker)Flaxseed oil/perilla oil/tannic acid
-
Increased the bioavailability and prolonged the release of the drug
-
Improved physical and chemical properties, long-term stability, and in vitro release of the active compound
-
Obtained small-sized liposomes with high encapsulation efficiency and improved biocompatibility
[51,72,117,118]
SilicaEpirubicin-hydrochloride/Alfa-choriogonadotropin
-
Used for the potential oxygen protection of liposomes
-
Enhanced the stability of drug-loaded vesicles in the gastrointestinal tract and the photovoltaic stability
[119,120]
CalciumcarbonateDrug
-
Increased stability and reduced leakage due to the continuity of smooth and uniformly coated liposomes
[121]
CeramicCholesterol
-
Good stability and anti-interference ability with a good practical application for liposomes
[122]
Table 4. Coating materials for liposomes with applications in the food industry.
Table 4. Coating materials for liposomes with applications in the food industry.
Coating MaterialType of Encapsulated Food-Grade Active Compounds The Advantage of the Coating Material UsedBibliography
Saccharides and their derivatives
ChitosanFish-derived peptide/glutathione/caffeic acid/flavonoids/quercetin/sour cherry extract/Morus nigra waste extract/caffeine
-
Enhanced the thermal stability and maintained the antioxidant activity of the peptide fractions
-
Protected the synergistic antioxidant effect of the active compounds
-
Improved stability and the antioxidant and anti-proliferative activities with reduced syneresis were protected
-
Improved bio-accessibility and sustained the release of active compounds in the digestive system
[126,127,128,129,130]
Sodium alginateVitamin C
-
Increased the microbiological stability of liposomes
-
Enhanced the controlled release property
[83]
Calcium alginateEnzymes
-
Improved the functional properties of the liposome carriers due to increased surface area
[155]
PectinEchinacosides and verbascoside
-
Provided an improved release in vitro digestion
[156]
InulinWithout active compound
-
Showed improved thermal, physical, and oxidative stability of liposomes
[157]
LactoseQuercetin
-
Improved conservation and release of antioxidants
[154]
Proteins
Whey proteinAstaxanthin
-
Improved thermal and light stability of liposomes
-
Improved the bio-accessibility of the active compounds and the stability under gastric conditions
[68,69]
Combinations between groups of materials
Chitosan–sodium alginatePolyelectrolyte
-
Improved physical and in vitro digestion stability of the liposomes
[83]
Chitosan–pectinHibiscus extract
-
Improved physical and in vitro digestion stability of the liposomes
[158]
Chitosan–zeinPulicaria gnaphalodes (Vent) extract/quercetagetin
-
Prolonged antimicrobial and antioxidant activity of the encapsulated compound
-
Improved light, thermal, and storage stability of liposomes
-
Enhanced the controlled release of active compounds in digestion
[113,159]
Pectin–whey proteinAntimicrobial peptides
-
Protected the liposomes against gastrointestinal digestion
[90]
Others
Poly (L-lysine)Bacteriophage/bioactive peptides
-
Improved chemical and thermal stability of liposomes
-
Improved antimicrobial properties of the active compound
[160]
Table 5. Recent clinical trials with coated liposomes.
Table 5. Recent clinical trials with coated liposomes.
DrugFormulation/Coating TechnologyUsed forExpected Results after the Clinical TrialPhaseClinicaltrials.gov Identifier
BupivacaineLiposomal bupivacaine/DepoFoam technologyOpioid use
pain, postoperative
colectomy
colorectal surgery
A substantially longer duration of action than normal bupivacaine (96 h versus 8–9 h, respectively)IVNCT03638635
2B3-101- (glutathione (GSH) pegylated liposomal doxorubicin hydrochloride formulation)PEG coatingMeningeal carcinomatosisCoating liposomes with PEG guarantees that they circulate for a longer period of time in plasmaIINCT01818713
Anti-EGFR immunoliposomes loaded with doxorubicinAttached are monoclonal antibodies or antibody fragments to the surface of liposomes (immunoliposomes, antibody-linked nanoparticles)Solid tumorsImmunoliposomes coated with antibodies bind more selectively to antigens expressed on target cells, and they are internalized more efficiently. Drug resistance can be overcome by such delivery systemsINCT01702129
Liposomal astaxanthinNot specifiedBioavailability of astaxanthin formulationsHuman crossover pharmacokinetic study pathways of astaxanthin in the bloodstreamNot applicableNCT02397811
DoxilNot specifiedBreast cancerAssess the effectiveness of using heat therapy in addition to the chemotherapy drug Doxil to treat recurrent breast cancer that has spread to the chest wall after mastectomyIINCT02192021
Table 6. Recent patent applications and patents on coated liposomes.
Table 6. Recent patent applications and patents on coated liposomes.
Patent Application/Patent, ID, Title, YearMaterial CoatingEffects on Stability
Ceftazidime combined powder injection and preparation method and product specification thereof, CN111840232A, 2020The ceftazidime and chitosan nanoparticles are each coated with vesicles, and the liposomes are then combined to create liposome-mixed nanoparticlesThe product purity is high, the selection range is expanded, the side effects are minimal, and the safety is high. The product obtained by the preparation process is of stable quality and good pharmacological effect.
Treatment of age-related macular degeneration, US2020262903A1, 2020The nanoparticles are coated with a drug targeting the vascular endothelial growth factor receptor (VEGFR), such as anti-VEGFR antibodies, anti-VEGFR aptamers, anti-VEGFR binding peptidesThe stability of the nanocomposite at elevated temperatures indicates the successful support of GOF for liposomes
A biodegradable nano-theranostic composite and process of preparation thereof, US2020237667A1, 2020Graphene oxide (GO) was deposited in the form of a thin film on both the inner and outer surfaces of the liposomesThe stability problem was solved by reinforcing the very fragile lipid membrane-based liposome wall with a dense inclusion of GO. This makes the wall very stable, even at a pH as low as 5 for several hours and at temperature as high as 50 ᵒC
DNA brick-assisted liposome sorting
US20210267894A1, 2021
Liposomes were coated with DNAThe stability of a liposome was improved and more functionalization was possible when a DNA coating was applied. DNA coatings have been useful because nucleases can easily remove them, and they are inert to most biochemical reagents.
Liposomes encapsulating anticancer drugs and use thereof in the treatment of malignant tumors
US20050100590A1, 2005
Liposomes were coated with a lipopeptide consisting of a lipid fragment, an active oligopeptide, and an oligopeptide spacer between the other two fragmentsThe addition of a negatively charged phospholipid favors the stability of the liposome solution and prevents the spontaneous aggregation of the liposomes
Liposome-based mucus-penetrating particles for mucosal delivery
US20170281541A1, 2017
Liposomes were coated with PEGPEG was used to increase the stability and solubility of liposomes with drugs, reduce toxicity, and prolong the half-life
Method of producing immunoliposomes and compositions thereof
US20090232730A1, 2009
Liposomes were coated with hyaluronan/hyaluronic acid or other glycosaminoglycansHyaluronan/hyaluronic acid provided protection against lyophilization and reconstitution so that only nanoscale liposomes covalently coated with hyaluronan/hyaluronic acid were structurally preserved
Liposomal formulations for delivery of nucleic acids
US10583084, 2020
Liposomes were coated with a glycosaminoglycan (hyaluronic acid)The coating materials improved the condensation and stability of the liposomes
Table 7. Liposome commercial products available on the market.
Table 7. Liposome commercial products available on the market.
Class of Active CompoundsActive Compounds EncapsulatedCommercial Name(Company/Country)Route of Administration
Pharmaceutics
Protease inhibitorAmprenavirAgenerase® (GlaxoSmithKline/United Kingdom)Oral product
Protease inhibitorRitonavirNorvir (Abbott laboratories/United States of America)Oral product
Protease inhibitorSaquinavirFortovase® (Hoffmann-La Roche lnc/Switzerland)Oral product
Antitumor antibioticDoxorubicinDoxil (Sequus Pharmaceuticals, Inc.,/United States of America), Evacet, Lipo-Dox, DC99® (Liposome Company NJ/United States of America)Injectable product
Antitumor antibioticDaunorubicinDaounoXome™ (NeXstar Pharmceuticals, Inc., Co/United States of America)Injectable product
Antitumor antibioticIrinotecanOnivydeTM (PharmaEngine/Taiwan)Injectable product
Anti-inflammatory drugIbuprofenIbunex (Phoenix Pharma Pvt. Ltd./India), Solufen® (Sanofi- Aventis/France)Oral product
AntihypertensiveAtorvastatinLipirex® (Sanofi-Aventis/France)Oral product
ImmunosuppressiveCyclosporineNeoral® (Novartis/Switzerland)Injectable product
Local anestheticLidocaineELA-Max (Ferndale Pharmaceuticals Ltd./United States of America)Oral product
AntifungalAmphotericin BAbhope (Abbott laboratories/United States of America), Ambilon (Celon Pharma Ltd./Poland), Abelcet (Liposome Company NJ/United States of America), AmBisome (Astellas Pharma Inc.,/United States of America, NeXstar Pharmceuticals, Inc., Co/United States of America), Amphocil (Sequus Pharmaceuticals, Inc.,/United States of America), Myocet® (GP-Pharm/Spain), Amphonex (Bharat serums & vaccines ltd/India)Injectable product
Photosensitizing agentsVerteporfinVisudyne® (Bausch & Lomb Incorporated/United States of America)Injectable product
Antimetabolite antineoplastic agentCytarabineDepoCyt® (Pacira Pharmaceuticals, Inc/United States of America)Injectable product
Chemotherapy drugCisplatinLipoplatin® (Regulon, Inc/Greece)Injectable product
OpioidMorphine sulfateDepoDur® (Skyepharma Production SAS/France)Injectable product
AntibioticAmikacinMiKasome® (NeXstar Pharmceuticals, Inc., Co/United States of America)Injectable product
Dietary/Food supplements and nutraceuticals
Vitamins and mineralsVitamin CLiposomal Vitamin C (Hypernatura®/Romania, WeightWorld/United Kingdom, NutriFlair/United States of America, Curesupport/Netherlands), Altrient C (LivOn Labs/United States of America), Vitamin C Liposomal (Actinovo Actinovo/Germany, Laboratoire Biocyte/France)Oral products
Vitamin D3Liposomal Vitamin D3 (Lipolife/United Kingdom, California Gold Nutrition/United States of America, Dr Mercola/United States of America), Mega-Liposomal Vitamin D3 (Aurora Nutrascience/Canada), Vitamin D3 Liposomal (Laboratoire Biocyte/France)
ZincUltraZin®liposomal zinc (Laboratoire Biocyte/France), Liposomal zinc (AbelaPharm/Serbia)
MineralsCal/Mag/Zinc Liposomal (Laboratoire Biocyte/France)
Multivitamins and mineralsLipozomal Vegan D3 K2 Magneziu (Hypernatura/Romania), Liposomal multivitamin (GymBeam/Germany), VENTUS liposomal Omega 3, Omega 6 (Life Spirit/United States of America), LVC5 (Lipolife/United Kingdom)
ProteinsCollagenCollagen liposomal (Actinovo/Germany, Healthydrops/North America), Collagen Zooki (YourZooki/United States of America)
Glutathione Liposomal Glutathione (Hypernatura, Lipolife), Glutathione Liposomal (ActiNovo/Germany, Laboratoire Biocyte/France), Altrient Glutathione (LivOn Labs/United States of America)
PolyphenolsResveratrolLiposomal Resveratrol (CureSupport, Lipolife/United Kingdom, Actinovo/Germany, Healthydrops/North America,), LLR1 Liposomal Resveratrol (Lipolife/United Kingdom)
QuercetinQuercetin Liposomal (Actinovo/Germany), Liposomal Bio Quercetin (DesBio PAO/Germany)
Antioxidant mixLiposomal NMN (Codeage/United States of America), HistX (Lipolife/United Kingdom)
Curcumin Micelle Liposomal Turmeric (Purality Health®/United States of America), Curcumin Lipozomal (Actinovo/Germany, Somavita®/United States of America), Liposomal curcumin (Hypernatura®/Romania, Lipolife/United Kingdom, Healthy Drops/United States of America), Liposomal Curcumin C3 LIPOSOL (SABINSA/United States of America)
Othersγ-Aminobutyric acid, melatonin, magnesiumLipozomal Sleep Formula (Hypernatura®/Romania)
MSM, glucosamine, Boswellia Lipozomal Joint Formula (Hypernatura®/Romania)
Food products
Functional teasPlant sterols, EGCG, choline, potassiumCholesterol Aid (Bio-Up Mimetic Technologies Codeage/United States of America)Oral product
Omega-3, CoQ10, choline, EGCGCardio Vitality (Bio-Up Mimetic Technologies Codeage/United States of America)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pasarin, D.; Ghizdareanu, A.-I.; Enascuta, C.E.; Matei, C.B.; Bilbie, C.; Paraschiv-Palada, L.; Veres, P.-A. Coating Materials to Increase the Stability of Liposomes. Polymers 2023, 15, 782. https://doi.org/10.3390/polym15030782

AMA Style

Pasarin D, Ghizdareanu A-I, Enascuta CE, Matei CB, Bilbie C, Paraschiv-Palada L, Veres P-A. Coating Materials to Increase the Stability of Liposomes. Polymers. 2023; 15(3):782. https://doi.org/10.3390/polym15030782

Chicago/Turabian Style

Pasarin, Diana, Andra-Ionela Ghizdareanu, Cristina Emanuela Enascuta, Catalin Bogdan Matei, Catalin Bilbie, Luciana Paraschiv-Palada, and Petronela-Andreea Veres. 2023. "Coating Materials to Increase the Stability of Liposomes" Polymers 15, no. 3: 782. https://doi.org/10.3390/polym15030782

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop