Next Article in Journal
Transport of Drugs Through Biological Barriers—An Asset or Risk
Previous Article in Journal
The Incorporation of CBD into Biodegradable DL-Lactide/Glycolide Copolymers Creates a Persistent Antibacterial Environment: An In Vitro Study on Streptococcus mutans and Staphylococcus aureus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Vesicular Carriers for Phytochemical Delivery: A Comprehensive Review of Techniques and Applications

1
Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman 4184, United Arab Emirates
2
Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman 346, United Arab Emirates
3
Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
4
Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science and Technology, Hisar 125001, India
5
Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
*
Author to whom correspondence should be addressed.
Pharmaceutics 2025, 17(4), 464; https://doi.org/10.3390/pharmaceutics17040464
Submission received: 24 February 2025 / Revised: 30 March 2025 / Accepted: 1 April 2025 / Published: 2 April 2025
(This article belongs to the Special Issue Novel Drug Delivery Systems for Natural Extracts)

Abstract

:
Natural substances, especially those derived from plants, exhibit a diverse range of therapeutic benefits, such as antioxidant, anti-inflammatory, anticancer, and antimicrobial effects. Nevertheless, their use in clinical settings is frequently impeded by inadequate solubility, limited bioavailability, and instability. Nanovesicular carriers, such as liposomes, niosomes, ethosomes, transferosomes, transethosomes, and cubosomes, have emerged as innovative phytochemical delivery systems to address these limitations. This review highlights recent developments in vesicular nanocarriers for phytochemical delivery, emphasizing preparation techniques, composition, therapeutic applications, and the future potential of these systems. Phytosomes, along with their key advantages and various preparation techniques, are extensively described. Various in vitro and in vivo characterization techniques utilized for evaluating these nanovesicular carriers are summarized. Completed clinical trials and patents granted for nanovesicles encapsulating phytochemicals designed for systemic delivery are tabulated. Phytochemical delivery via vesicular carriers faces challenges such as low stability, limited active loading, scalability issues, and high production costs. Additionally, immune clearance and regulatory hurdles hinder clinical application, requiring improved carrier design and formulation techniques.

1. Introduction

Natural products, particularly those derived from plants, owing to their diverse and significant biological activities have long been regarded as key resources for treating various diseases. Phytoconstituents are bioactive compounds naturally found in plants, responsible for their therapeutic properties and protective roles, and have been utilized in both traditional and modern medicine. Produced as secondary metabolites, they help plants resist environmental stressors, pathogens, and herbivores [1]. Phytochemicals, in their native form or as metabolites, provide health benefits by scavenging free radicals, chelating metals, inhibiting microtubule and microfilament assembly, and blocking protease activity [2].
Polyphenols, flavonoids, carotenoids, and allyl sulfide-containing compounds are renowned for their antioxidant and anti-inflammatory effects, aiding in the prevention and management of chronic diseases [3,4]. Isoflavones, particularly those derived from soy, function as phytoestrogens, mimicking hormonal activity and offering benefits in conditions like menopause [5]. Protease inhibitors and indole-containing compounds play crucial roles in enzyme regulation, contributing to cellular homeostasis and defense mechanisms. Proanthocyanidins serve as potent anti-infective agents, while saponins and capsaicin have been identified as modulators of DNA replication, indicating potential in cancer therapy [6]. Additionally, curcuminoids such as curcumin exhibit a wide spectrum of activities, including antitumor, antioxidant, and anti-inflammatory properties, making them valuable in the treatment of various ailments [7,8]. Natural compounds like piperine, caraway, curcumin, and genistein act as bioenhancers by increasing the bioavailability of drugs when taken orally [9]. Bioenhancers are agents that boost the therapeutic efficacy of a drug by increasing its concentration in biological fluids or systemic circulation, often by enhancing the absorption or bioavailability of the active compound [10]. Reducing therapeutic drug dosage can lower treatment costs, minimize side effects, and help prevent drug resistance in long-term diseases like cancer and infections [11]. Bioenhancers, such as piperine, can possess therapeutic properties, demonstrating effectiveness in treating diseases like diabetes, arthritis, cancer, and cardiovascular conditions [12]. Herbal bioenhancers hold promise in combating multifactorial diseases like cancer by overcoming resistance mechanisms and boosting the effectiveness of anticancer drugs [9]. In antimicrobial therapy, they offer hope against antibiotic resistance by enhancing the efficacy of antimicrobial agents [9]. This is achieved through mechanisms such as obstructing efflux pumps, preventing the formation of biofilm, modulating virulence factors, and modifying host defense mechanisms. The primary action of herbal bioenhancers is their ability to inhibit P-glycoprotein and metabolic enzymes such as cytochrome P450 enzymes and Uridine 5′-diphosphate-glucuronosyltransferase [13,14]. Thus, bioenhancers function by retaining the drug molecule within the cell without altering its metabolic activity. It was reported that piperine significantly enhances the bioavailability of the anticancer drug docetaxel by inhibiting the metabolizing enzyme CYP3A4, which led to an increase in the drug’s Cmax from 6808 ng/mL to 11,380 ng/mL [15]. Similarly, it was found that the bioavailability of tamoxifen was improved in rats pretreated with either curcumin or piperine [16]. Increased drug absorption, bypassing of hepatic first-pass metabolism, enhanced drug retention, and reduced multidrug resistance are the resulting effects of combining anticancer drugs with bioenhancers in the treatment of various cancers, leading to improved bioavailability. Moreover, various medicinal plants have been used for treating skin disorders [17].
Phytochemical delivery faces several significant challenges that limit their clinical translation and therapeutic potential. In addition to being broken down by enzymes in the gastrointestinal tract (GIT), the polarity of these molecules along with their size make it difficult for them to cross the blood–brain barrier (BBB), the inner endothelial lining of blood vessels, and the GIT and mucosa [18]. Furthermore, their clinical application is often limited by challenges such as low aqueous solubility, fast metabolism, low bioavailability, and elimination, instability and short duration of action [19]. This necessitates administering a higher dose of phytochemicals, which can potentially increase peripheral organ toxicity. Variable pharmacokinetics among individuals and extensive first-pass metabolism further contribute to inconsistent therapeutic outcomes. Moreover, conventional formulations often lack targeted delivery capabilities, reducing their effectiveness at the desired site of action. Collectively, these challenges underscore the need for advanced drug delivery strategies to enhance the clinical utility of phytochemicals.
The diverse therapeutic potential of phytochemicals has spurred significant interest in pharmaceutical technology, leading to the development of formulations that enhance their bioavailability and efficacy, thereby maximizing their health benefits [20]. To address the limitations associated with phytochemical delivery, a range of advanced drug delivery systems have been explored. The incorporation of these natural products into polymeric and lipid nanocarriers has emerged as a novel strategy to overcome these hurdles, offering enhanced delivery and efficacy [21]. Nanocarriers have several advantages, including protection from environmental degradation, enhanced solubility, and improved bioavailability. Incorporating stabilizers such as cholesterol and employing lyophilization technique have been shown to enhance the stability of advanced vesicular formulations, including transethosomes [22]. By providing an increased surface area, nanocarriers facilitate better absorption and penetration through biological barriers. Additionally, they provide sustained as well as controlled drug release, thus maintaining therapeutic levels over an extended time while minimizing systemic side effects through site-specific delivery [23]. Functionalized nanocarriers have further advanced the field by incorporating surface modifications and ligand conjugations can further improve site-specific delivery and cellular uptake [24]. Surface modification approaches, including PEGylation, have been utilized to enhance the stability and prolong the circulation time of nanosystems. By forming a steric barrier around nanoparticles, PEGylation minimizes protein adsorption and reduces recognition by macrophages, thus improving their pharmacokinetic profile [25]. These innovations enhance stability, entrapment efficiency, and therapeutic efficacy while addressing toxicity concerns [26]. Stimuli-responsive nanocarriers represent the next frontier, offering precise, adaptive drug delivery tailored to the needs of specific therapeutic applications [27]. Lipid–polymer hybrid nanoparticles are promising drug delivery systems that combine the biocompatibility of lipid nanosystems with the structural stability of polymers. They enable the co-delivery of amphiphilic molecules and have been effectively used to transport both phytochemicals and chemotherapeutic agents for enhanced cytotoxicity against cancer. By targeting tumor sites and enabling controlled drug release, hybrid nanocarriers improve therapeutic efficacy and chemo-sensitization of cancer cells [28]. Targeting ligands such as folic acid enhance cellular uptake through receptor-mediated endocytosis. Folic acid, which selectively binds to folate receptors, which are overexpressed in many tumors, helps reduce off-target effects on normal tissues. Ligand-functionalized pH-sensitive NPs were developed for co-delivery of carboplatin and paclitaxel in cervical cancer treatment [29]. These nanoparticles exhibited a uniform size (~170 nm), high cellular uptake (66.7%), and strong cell inhibition (23%). They also demonstrated pH-responsive drug release and a synergistic therapeutic effect of both drugs against cervical cancer cells.
Encapsulation of phytochemicals also helps bypass first-pass metabolism and improve systemic circulation time. Nanocarriers enhance the ability of natural products to cross the blood–brain barrier and facilitate their uptake into cancer cells via endocytosis, improving therapeutic potential. Scalable production of vesicular nanocarriers has advanced through technologies like microfluidics, high-pressure homogenization, continuous manufacturing, and bioreactor systems. These methods enhance control, uniformity, and practical yield, addressing limitations in large-scale production and supporting the clinical translation of nanocarrier-based drug delivery systems [30,31,32]. This integration of natural products into nanocarriers not only addresses their intrinsic limitations but also represents an important leap in drug delivery technology, paving the way for safer, more effective, and targeted treatments.
The aim of this review was to highlight both phyto-phospholipid complexes and advanced vesicular delivery systems such as liposomes, invasomes, niosomes, bilosomes, transfersomes, ethosomes, transethosomes, and cubosomes that have been extensively studied for enhancing the bioavailability, stability, and therapeutic effectiveness of phytochemicals. Vesicular carriers with limited evidence of application in phytochemical delivery or lacking relevance in current research trends were excluded to maintain focus and coherence. Although no strict exclusion criteria were applied, preference was given to systems supported by peer-reviewed publications, preclinical studies, clinical trials, or patent literature.

2. Bioavailability and Stability Challenges

Phytochemicals exhibit diverse physicochemical properties, which contribute to alteration in their solubility, bioavailability, and chemical stability profiles. The details of the most frequently investigated phytochemicals are summarized in Table 1.

2.1. Bioavailability

Applying the Lipinski rule of five to phytochemicals can help determine the drug likeness of these natural compounds for product development, similar to active pharmaceutical ingredients. Many natural compounds, such as phytochemicals, may have larger molecular weights, which limit their solubility and permeability [33]. When they exceed this threshold, modifications or nanoformulations might be needed to enhance their delivery. Excessive hydrogen bond donors in natural compounds, such as hydroxyl or amine groups, can hinder permeability across biological membranes. Similarly, a high number of acceptors, such as oxygen or nitrogen atoms, can restrict passive diffusion, necessitating careful structural modifications or encapsulation strategies. A compound’s lipophilicity (LogP) determines its solubility and permeability. For instance, lipophilic compounds like terpenes often face challenges with water solubility, limiting their gut absorption. Strategies such as incorporating lipid nanosystems, including solid lipid nanoparticles (SLNs) or self-emulsifying systems, are commonly employed to enhance their bioavailability [34,35]. Moreover, decreased aqueous solubility often necessitates the use of solvent-based extraction methods for isolating natural compounds from medicinal plants. Oral delivery of bioactive phytochemicals faces challenges such as the viscosity of GI contents and dietary intake, leading to the exploration of alternative delivery forms to enhance bioavailability [36]. In the GIT, phytochemicals undergo enzymatic metabolism in different regions, which can either reduce their activity or improve bioavailability compared to the parent compound [37,38].
Bioefficacy of phytochemicals refers to the ability of these compounds to exert beneficial biological effects in the body after they have been absorbed and metabolized, which depends on their bioaccessibility and bioavailability [38]. It is assessed through short-term changes in biomarkers like plasma lipid levels, glucose, blood pressure, antioxidant activity, and liver function. Bioaccessibility of phytochemicals refers to the extent and rate at which the active ingredients in phytochemicals become available for absorption in the GIT after ingestion [37]. This is dependent on various factors such as the chemical nature, polarity, and solubility of the phytochemicals, and how they interact with food components. After absorption, small intestinal cells enable cellular uptake and efflux, while hepatic cells transform inactive precursors into active forms, crucial for bioaccessibility [18]. Additionally, the microbiota in the large intestine metabolizes unabsorbed phytochemicals, impacting bioefficacy.

2.2. Stability in Solid State

Phytochemicals in solid state and aqueous solutions are influenced by environmental, chemical, and physical factors that alter the stability of the compounds. The presence of moisture/humidity can accelerate hydrolysis, particularly in compounds with ester and amide linkages. Elevated temperatures can increase the rate of chemical reactions, leading to faster degradation due to increased collision frequency, as described by the Arrhenius equation. L-ascorbic acid stability is affected by oxygen, light, and temperature [39]. Phytochemicals, namely procyanidin B2, (-)epicatechin, chlorogenic acid, hyperoside, and isoquercetin, remained stable for six months at 4 °C but degraded rapidly at 40 °C [40].
The stability of phenolic compounds following extraction from plant matrices is critical, as they are prone to degradation when exposed to the external environment (oxygen or thermal stress) [41]. The conjugated system of double bonds in carotenoids is susceptible to isomerization and oxidation, particularly during thermal processing due to high temperatures and oxygen exposure [42]. UV or visible light can induce photochemical degradation, and the presence of oxygen can promote oxidative degradation, particularly in compounds sensitive to oxidation [43]. Surface pH changes (even in the solid state) can influence the stability of sensitive molecules [44]. Different polymorphic forms of drugs may exhibit varying stability profiles, with some forms being more prone to degradation [45]. Smaller particle sizes increase the surface area exposed to environmental factors, enhancing the degradation rate. Excipients in formulations may interact with phytochemicals, leading to instability through chemical or physical interactions [46]. Residual solvents, catalysts, or trace metals can trigger degradation pathways such as hydrolysis or oxidation [47].

2.3. Stability in Aqueous Solution

Factors influencing degradation in aqueous solutions include pH, where acidic or basic conditions can accelerate hydrolysis, oxidation, or ionization. The stability of phenolic compounds is strongly influenced by pH and their structural conformation. Flavan-3-ols are highly stable in acidic conditions but become unstable at a neutral pH [48]. Higher temperatures increase molecular mobility, enhancing degradation. UV or visible light can cause photochemical degradation, and dissolved oxygen promotes photooxidation, especially in unsaturated or phenolic compounds. It was reported that thymoquinone, a potent anticancer phytochemical, is highly soluble in aqueous solutions (500 µg/mL) but remains unstable, with its stability significantly influenced by pH and light, the latter being more critical [49]. This instability makes pure aqueous solutions unsuitable as pharmaceutical carriers for drugs, suggesting that a combination of organic and aqueous solvents is a more effective alternative [49]. High ionic strength affects solubility and stability, while the solvent composition influences reactivity and overall stability [50]. Complexation with metal ions can lead to degradation, and residual catalysts or impurities can accelerate processes like oxidation or hydrolysis [51]. Additionally, higher concentrations of phytochemicals may promote aggregation or precipitation, impacting stability [52].

3. Functional Role of Nanoparticles in Phytochemical Delivery Systems

Nanoparticles are highly effective drug delivery carriers, owing to their submicron particle size and distinctive physicochemical properties that facilitate targeted tissue delivery [53,54]. These versatile carriers are available as particulate, soluble, or target-specific recognition moieties, which can encapsulate a wide variety of drugs. They are fabricated using diverse polymers and lipids utilizing a range of manufacturing techniques [18]. The composition of the carrier and the entrapping agent plays an important role in determining permeability, release rate, mucoadhesion, and targeting efficiency. Nanocarriers are typically formulated as aqueous dispersions or incorporated into gel or film matrices.
Vesicular nanocarriers with organized bilayer structures enable the effective encapsulation and delivery of hydrophilic and lipophilic drugs. These vesicular systems are used to encapsulate and deliver various phytoconstituents with enhanced efficiency [55,56,57]. They offer several advantages, including controlled release, targeted delivery, improved stability, and biocompatibility, making them suitable for various administration routes. A comparison of various nanovesicular carriers, summarizing their composition, advantages, limitations, key attributes, and stability are presented in Table 2 and illustrated in Figure 1. However, these are subject to challenges such as limited drug-loading capacity, high production costs, stability issues, and scalability constraints persist. Recent advances, including stimuli-responsive vesicles, and polymer–lipid hybrid systems have significantly improved their functionality and therapeutic potential [58,59]. These innovations continue to position vesicular systems as an efficient nanocarrier in modern pharmaceutical research for achieving better patient outcomes.

3.1. Phytosomes

Phytosome® or phyto-phospholipid complexes are well-defined systems formed by the association of phytochemicals (usually polyphenols) with phospholipids, typically phosphatidylcholine, in a defined stoichiometric ratio. This complex results in improved solubility, stability, skin/mucosal permeation, and bioavailability of the active compound. Nanophytosomes are phyto-phospholipid complexes under 200 nm in size, designed to enhance absorption and biological activity, with composition similar to regular phytosomes but improved delivery due to their nanoscale size. The formation of phytosomes involves interaction between the hydrophilic phytoconstituent and the phospholipid through hydrogen bonding and van der Waals forces. Among all phytochemicals, only those containing an active hydrogen atom (e.g., -COOH, -OH, -NH2, -NH), such as polyphenols, can be incorporated into a phytosome structure [60]. This active hydrogen atom enables hydrogen bond formation between the phytochemicals and the hydrophilic groups of the amphiphilic phospholipids [61]. It was found that hydroxyl groups present in polyphenols can efficiently interact with the nitrate and phosphate groups present in phospholipids. The binding results in the formation of a lipid-compatible molecular complex, where the hydrophilic head of the phospholipid binds to the phytoconstituent, and the lipophilic tail interacts with cell membranes [62]. Structurally, phytosomes resemble biological cell membranes due to the presence of phospholipids, which facilitate the absorption of active compounds [63,64]. Phytosomes are amphiphilic with the ability to encapsulate hydrophilic and lipophilic phytochemicals, providing stability and protection. Phytosomes are preferred for transporting poorly water-soluble flavonoids, such as curcumin, polyphenol, and quercetin, that often struggle to cross lipid-rich biological membranes [65,66]. Phospholipid complexes of curcumin achieve higher bioavailability of the parent compound compared to uncomplexed curcumin [66]. Indena® has developed a wide range of phytosome-based formulations aimed at enhancing the bioavailability and therapeutic efficacy of various phytochemicals. Notable products include Siliphos® (from Silybum marianum) [67] for hepatoprotective and antioxidant effects, Hawthorn phytosome (Crataegus flavonoids) with cardioprotective and antihypertensive properties, and Oleaselect™ Phytosome (from Olea europaea) for its anti-inflammatory and antihyperlipidemic benefits. Polinacea™ phytosome (Echinacea angustifolia) [68] and Ginseng phytosome (Panax ginseng) support immunomodulation [69], while Ubiqsome® phytosome delivers CoQ10 for mitochondrial and antioxidant activity [70]. Quercefit™ [71] and Greenselect® [72] target antioxidant and metabolic health, whereas Vazguard™ (bergamot extract) aids in regulating cholesterol and glucose levels. Other formulations, such as Casperome® (Boswellia serrata) [73], curcumin phytosome [74], Leucoselect® (grape seed) [75], and Virtiva® (Ginkgo biloba) [76], contribute to anti-inflammatory, joint, cardiovascular, and cognitive health. Additional products like Centevita® (Centella asiatica) [77], Soyselect® (soybean extract) [78], and Visnadex (Amni visnaga) [79] further extend applications to skin health, hormone balance, and microcirculation. These innovations reflect Indena’s role in developing clinically relevant, plant-based vesicular delivery systems.
A variety of solvents have been used as reaction mediums to formulate phyto-phospholipid complexes. Aprotic solvents, such as methylene chloride and ethyl acetate, do not form hydrogen bonds, while protic solvents like ethanol and methanol contain hydrogen atoms attached to electronegative atoms, making them more effective in yielding complexes with lower residues. Ethanol is commonly used for its high efficiency, while some studies also utilize mixed solvent systems, such as dichloromethane and methanol or water and diethyl ether, to dissolve phospholipids separately from the drug or extract [80,81].

3.2. Liposomes

Liposomes are spherical, bilayered vesicles, composed of phospholipids and cholesterol, capable of delivering hydrophilic drugs (within their aqueous core) and hydrophobic drugs (within their lipid bilayer). Due to their structural versatility and biocompatibility, liposomes have become a prominent carrier system in drug delivery [82]. They offer advantages such as biocompatibility, targeted delivery, improved drug stability, and reduced systemic toxicity, but face challenges like short circulation time, high production costs, and stability issues. Recent advances include PEGylated “stealth” liposomes, stimuli responsive designs, liposomal vaccines, and functionalized liposomes for active targeting. Liposomes are commonly categorized based on their structural characteristics, composition, and method of preparation [83]. Liposomes are administered via oral, parenteral, pulmonary and ocular routes, with each offering distinct advantages for drug delivery.
Liposomes play a pivotal role in the delivery of phytochemicals, which are often limited by low bioavailability, poor water solubility, and rapid metabolism [84]. By encapsulating phytoconstituents, such as curcumin, quercetin, betulinic acid, artemisinin, or resveratrol, liposomes enhance their stability, solubility, and targeted delivery to the desired site of action. They also improve the therapeutic efficacy of these compounds by ensuring sustained release and protection from degradation. Liposomes formulated with dioleoylphosphatidylcholine (DOPC) significantly improve curcumin’s solubility and stability. However, curcumin is quickly released in the presence of serum or cells, limiting the formulation’s effectiveness in vivo [85]. Quercetin, a potent natural antioxidant, was encapsulated in Eudragit-coated liposomes designed for targeted intestinal delivery. The Eudragit coating protected the liposomes from gastric conditions and enhanced their stability in simulated gastrointestinal fluids. Importantly, quercetin’s antioxidant activity remained intact, effectively inhibiting DPPH radicals and reducing reactive oxygen species (ROS) levels in human intestinal cells [86]. PEGylated liposomes enhance drug delivery by prolonging circulation time, reducing immune clearance, and improving drug accumulation at target sites, especially tumors. This results in increased stability, bioavailability, and therapeutic efficacy. PEGylated liposomes loaded with betulinic acid (BA) were developed to enhance drug delivery performance. These liposomes demonstrated a more sustained drug release over 72 h (89%) compared to conventional BA liposomes (78%). In vivo studies showed significantly greater tumor growth inhibition (64.3%) with the PEGylated formulation, highlighting its improved delivery efficiency, prolonged release, and enhanced anticancer activity over free BA or standard liposomes [87]. A liposome-based formulation containing soy phosphatidylcholine and cholesterol in a 3:1 ratio, combined with a biosurfactant (10% MEL-A) and 1% betulinic acid (BA), significantly inhibited HepG-2 cell proliferation, demonstrating strong anticancer activity. Additionally, it improved the solubility, stability, and bioavailability of the active compounds and has already been introduced into clinical use [88]. Dihydroartemisinin was encapsulated in both conventional and long-circulating liposomes and evaluated for cytotoxicity against MCF-7 breast cancer cells. Both formulations showed suitable size for parenteral use and achieved high encapsulation efficiency (~70%). They remained physically and chemically stable during storage and in the presence of albumin. Flow cytometry revealed efficient cellular uptake, with conventional liposomes showing greater internalization than long-circulating ones. Cytotoxicity studies showed IC50 values of 12.1 μM for free dihydroartemisinin, and 48.2 μM and 77.0 μM for the conventional and long-circulating liposomes, respectively [89]. Liposomes serve as an effective delivery system for resveratrol, as they help maintain its active trans-form by preventing conversion to the less effective cis-form. Resveratrol tends to localize at the liposome surface, allowing for controlled, sustained release that avoids cellular membrane overload and reduces cytotoxicity. This enables the use of drug concentrations that would otherwise be toxic in free form. Additionally, the liposomal bilayers enhance phytochemicals’ long-term stability and support the activation of cellular defense mechanisms [90,91]. Recent studies have highlighted liposomal formulations as effective systems for enhancing the bioactivity of herbal extracts and phytochemicals in treating chronic diseases, including cancer, inflammation, eye and skin disease, malaria, osteosarcomas and neurodegenerative disorders [92,93,94].
Liposomes are among the most effective lipid-based entrapment systems due to their ability to use natural materials, encapsulate phytochemicals with varying solubilities, and protect ingredients from oxidation by metal ions, free radicals, and enzymes [95]. They are well known for enhancing the cutaneous delivery of molecules of various sizes and have been recently utilized for skin delivery of phytochemicals due to their high loading efficiency, carrier capacity, and compatibility with different skin layers. The term ‘phospholipid vesicles’ is a broader term encompassing any vesicular system composed of phospholipids, such as liposomes, transferosomes, ethosomes, cubosomes, and phytosomes. Not all phospholipid vesicles are phytosomes, but all phytosomes fall under this general category. Phyto-phospholipid complex formulations are extensively studied for addressing bioavailability challenges by forming molecular complexes with phytochemicals, rather than encapsulating them in phospholipid vesicles [96].
The potential of phospholipid vesicles as carriers for topical and transdermal delivery of phytochemicals is described in a recent review [97]. Modified liposomes, such as glycerosomes, utilize glycerol as a partial penetration enhancer to improve the skin bioavailability of encapsulated phytochemicals, including extracts and oils, compared to traditional liposomes [98]. The moisturizing effect of glycerol disrupts the ordered structure of the stratum corneum, facilitating the accumulation of bioactive phytochemicals in various skin layers. Glycerosomes modified with maltodextrin and polymers, like gelatin forming gluglycerosomes, demonstrated improved stability, viscosity, and effectiveness in delivering Hypericum scruglii extract to wound sites [99]. Additionally, glycerosomes have been utilized to encapsulate Thymus capitatus oil for treating oral cavity diseases [100] and Rosmarinus officinalis extract to enhance the stability of antioxidant polyphenols [95]. Modifications with sodium hyaluronate to create gly-hyalurosomes, loaded with Citrus limon var. pompia extract, effectively prevented oxidative damage to keratinocytes and fibroblasts while supporting their viability [101]. Table 3 depicts liposomes encapsulating selected phytochemicals, including their preparation techniques, composition, and key features.

3.3. Invasomes

Invasomes are modified liposomes composed of phospholipids, ethanol, terpenes, and water, and may also include lysophosphatidylcholine. Frequently, phospholipids such as phosphatidylcholine and egg or soy lecithin form the bilayer of the vesicle; lysophosphatidylcholine acts as an edge activator, enhancing vesicle flexibility; meanwhile, terpenes, such as eugenol, thymol, limonene, citral, cineole, nerolidol, fenchone, anethole, geraniol, and linalool, along with ethanol, serve as penetration enhancers and contribute to the fluidity of the vesicle bilayer [127]. Other phospholipids, such as phosphatidylserine, phosphatidic acid, phosphatidylglycerol, and phosphatidylinositol, are less commonly used and typically depend on specific formulation requirements. Since these phospholipids are similar to those found in mammalian cells, they can adhere to the cell surface either nonspecifically or through specific interactions with cell surface receptors. The internalization of liposomes by target cells typically occurs via endocytosis. Other mechanisms include cell vesicle fusion and lipid exchange cell surface receptors. Ethanol and terpenes enhance invasome performance for transdermal drug delivery by disrupting the tightly packed lipid structure of the stratum corneum. Ethanol fluidizes the lipid bilayer, increases vesicle flexibility, and prevents aggregation by providing a negative surface charge. Terpenes act as penetration enhancers by breaking lipid packing in the outermost layer, improving drug partitioning, and opening polar pathways for deeper penetration. Both components facilitate vesicle deformation during skin permeation without rupture or drug loss. The effectiveness of terpenes depends on properties like molecular size, lipophilicity, and degree of unsaturation, and they are recognized as safe by the US FDA [127,128]. Membrane flexibility can promote vesicle aggregation and the formation of multilamellar onion-like structures. However, flexible vesicles are more sensitive to temperature changes, leading to reduced stability during storage. Terpenes such as limonene increased membrane rigidity of the vesicles, possibly due to competitive interactions with cholesterol.
Polar phospholipid-based invasomes, with their adaptability to low water content conditions and ultra flexibility, penetrate deeper skin layers by leveraging the transepidermal osmotic gradient and elastomechanics, similar to transferosomes. While effective for dermal and transdermal drug delivery, formulation challenges like oxidative degradation, hydrolysis, and leakage of encapsulated bioactives must be addressed [128]. To improve patient compliance and prolong drug retention, these carriers are often converted into invasome gels using gelling agents such as carbomers (Carbopol 934, 940), poloxamer 188, and HPMC K4M. Capsaicin is primarily found in plants of the Capsicum genus, which belongs to the Solanaceae family. Capsaicin is widely used in the pharmaceutical field for pain management, including neuropathic and musculoskeletal pain, due to its ability to desensitize TRPV1 receptors. It also exhibits anti-inflammatory, antimicrobial, and anticancer properties, with potential applications in weight management and respiratory conditions. Additionally, capsaicin is used in dermatology for relieving itchiness and inflammation in conditions like psoriasis. An investigation was carried out to prepare capsaicin-loaded (0.15% w/w) invasomes, which exhibited a size < 100 nm, a narrow size distribution (PDI ~0.01–0.30), and a slightly negative zeta potential < −20 mV [129]. Capsaicin-loaded invasomes demonstrated enhanced skin permeability compared to conventional liposomes and a commercial ethanolic solution (0.15%).
A study developed curcumin-loaded terpenoid invasomal nanovesicles using a mechanical dispersion process with eucalyptol as a permeation enhancer [130]. Transmission electron microscopy revealed spherical vesicles with a size of 461.57 ± 1.38 nm and an EE of 80.54 ± 0.38%. The in vitro release followed the Higuchi kinetic model, and ex vivo studies showed that the optimized formulation achieved two and a half times higher curcumin permeation across pig ear skin compared to a curcumin solution, with a flux of 179.44 ± 0.26 µg/cm2/h. Berberine-loaded invasomes formulated with 2% Carbopol gel exhibited superior analgesic and antiarthritic activity compared to the standard gel (Omnigel, 0.1%), providing prolonged effects [131]. Radiographic analysis of hind paw rats in the complete Freund’s adjuvant model revealed bone resorption, reduced joint gaps, and significant connective tissue expansion following treatment with berberine-loaded invasomal gel. Histology of ankle joints further confirmed the gel’s enhanced penetration capability and its profound therapeutic effects. Curcumin-loaded invasomes were developed using a factorial design to assess the impact of terpene type and concentration on formulation properties [132]. The optimized invasomal formulation, with an EE of 85.84 ± 0.56% and vesicle size of 302.33 ± 1.53 nm, was incorporated into a topical gel. This invasomal gel exhibited three times higher permeation flux than a plain gel. In vivo studies in psoriatic BALB/c mice showed that the invasomal gel significantly accelerated and improved recovery compared to conventional curcumin gel. In another attempt, the delivery of dapsone using invasomes for acne therapy was investigated in rats. The results here signify greater skin retention by the developed carrier [133]. Similarly, Castangia et al. developed santosomes, vesicles formulated with Santolina insularis essential oil, propylene glycol and phosphatidylcholine. Santosomes, loaded with the wound healing protein phycocyanin, demonstrated enhanced drug delivery and wound healing potential due to the synergistic action of terpenes and phospholipids in the essential oil [134].

3.4. Niosomes

Niosomes are self-assembled vesicles of nonionic surfactants in an aqueous phase, mostly in the presence of cholesterol or other stabilizing agents. These vesicles are structurally similar to liposomes but are more stable and cost-effective, making them a potential drug delivery system. Niosomes can encapsulate both hydrophilic and hydrophobic drugs, enhancing their stability, bioavailability, and therapeutic efficacy [135,136,137]. Niosomes are osmotically responsive, biocompatible, biodegradable, nontoxic, and possess immune-suppressive properties [138]. Niosomes are widely used in various drug delivery applications, such as cancer therapy, and gene and vaccine delivery. They have been explored as versatile drug carriers suitable for various administration routes, including parenteral, oral, dermal, ophthalmic, transdermal, and pulmonary applications.
Niosomes offer a promising delivery system for phytochemicals with low solubility in bodily fluids. Different types of niosomes have been developed to address the limitations of phytoconstituents effectively [139]. A niosome is known as a drug depot because it has the potential to transfer the drug in a controlled or sustained manner to the targeted site. The potential of this vesicle in oral delivery of Fumaria officinalis for antineuropathic and anti-inflammatory effects has been evaluated [140]. The bioguided fractionation, as well as phytochemical analysis, identified the alkaloid rich fraction as the major active ingredient. In vivo studies of the optimized formulation (Nio-2) indicate a decrease in TNF-α and Interleukin 6 while enhancing the anti-inflammatory factor IL-10 and ameliorating oxidative stress. The outcome of this investigation confirms the potential of an alkaloid-rich fraction of Fumaria officinalis as a new oral therapeutic agent in treating various inflammatory conditions including neuropathic pain. Another study reported the enhancement in solubility and bioavailability of morusin, a flavonoid having anticancer properties when formulated as niosomes [141]. The developed formulation exhibited ideal pharmaceutical characteristics, showed higher cytocompatibility, improved aqueous dispersibility, controlled release, and improved efficacy in four cancer cell lines tested. A study reported in 2020 developed niosomes containing hedera helix extract prepared using common thin film method [142]. The composition has cholesterol (30%) and span 60 (70%) which entrapped the hedera helix extract. The developed formulation exhibits good extract entrapment (~95%), nanosize (~132 nm), and strong negative surface charge with good colloidal stability (−41 mV). The sustained release of extract from niosomes was noticed for 2 days in both cancer and normal conditions. The MTT assay indicates the extract-loaded niosomes were more toxic to the HT-29 cell line.
PEGylated nanosystems offer numerous advantages, making them highly effective in drug delivery. Additionally, PEGylated nanoparticles are adaptable to various administration routes and can integrate multiple functionalities, such as imaging agents and targeting moieties, making them a versatile platform for advanced therapeutic applications. PEGylated niosomes loaded with artemisinin and metformin, prepared using the thin film hydration (TFH) method, were recently investigated [143]. The optimized formulation demonstrated nanosize (256 nm), higher encapsulation efficiency (95%), and appropriate PDI (0.202). PEGylation significantly reduced niosome size due to its hydrophilic properties. In vitro studies revealed dose-dependent toxicity of both free and niosomal formulations, effectively inhibiting the growth of A549 lung cancer cells. RT-PCR results further confirmed that the niosomes exhibited enhanced antiproliferative effects by suppressing antiapoptotic genes and promoting apoptotic gene expression. Table 4 provides an overview of niosomes loaded with selected phytochemicals, detailing their composition, preparation methods, and key characteristics.

3.5. Bilosomes

Bilosomes are advanced vesicular carriers made of lipid bilayers stabilized with bile salts, offering enhanced stability and bioavailability for encapsulated compounds. These carriers are designed to withstand the harsh GI environment, including acidic pH and enzymatic degradation, which makes them highly effective for delivering sensitive phytochemicals, such as flavonoids, terpenoids, and polyphenols. The enhanced flexibility of their structure improves transdermal delivery, enabling more effective penetration through the stratum corneum and deeper layers of epidermis and dermis. Moreover, this vesicle enables oral vaccine administration and helps reduce dosing frequency [159]. Due to their potential for controlled and targeted release, they are positioned as valuable tools in the development of functional foods and therapies for chronic diseases [160]. However, they also have limitations, including potential variability in encapsulation efficiency, scalability challenges in manufacturing, and the need for detailed toxicity and long-term safety studies. Despite these challenges, bilosomes remain a promising delivery system for overcoming the inherent limitations of phytochemical bioavailability and stability [161]. It has been reported that bilosomes play a significant role in immunization against infections and diseases by utilizing antigens instead of whole pathogens, enhancing safety [162]. While most bilosome research focuses on oral immunization, recent studies have explored their potential in phytochemical delivery [161]. Notable examples include the use of pharmacologically active polysaccharides from Enteromorpha intestinalis and tripterine from Tripterygium wilfordii for drug delivery. In vivo studies showed longer residence time, improved cellular uptake, and enhanced bioavailability of these compounds [163,164].
Surface coating of bilosomes enhances their stability, functionality, and efficacy in drug delivery [165,166]. It protects the vesicle from degradation in harsh environments, such as the GIT, and ensures prolonged shelf life. Coatings improve mucoadhesion, allowing for increased residence time and better drug absorption, while also providing controlled and sustained drug release. They enhance biocompatibility, reduce immune response, and offer protection against oxidative stress, ensuring the integrity of encapsulated drugs. Furthermore, surface coatings facilitate targeted delivery by enabling functionalization with ligands or antibodies and improve encapsulation efficiency by minimizing drug leakage. These advantages make coated bilosomes particularly effective for oral, ocular, and parenteral drug delivery applications. Chitosan is widely used as a coating agent for nanovesicles, leveraging electrostatic interactions between the negatively charged nanovesicle surface and the positively charged amino groups of chitosan. This coating enhances the stability of nanovesicles, prolongs drug release, increases cellular uptake, and prevents drug leakage [167]. Additionally, chitosan improves mucosal barrier permeability across various tissues, including ocular, intestinal, nasal, and skin barriers [168]. A positive charge on bilosomes facilitates interaction with the negatively charged stratum corneum, making it a promising candidate for delivering berberine to deeper skin layers effectively. Berberine-loaded, chitosan-coated bilosomes were evaluated for their anti-inflammatory efficacy in rheumatoid arthritis management in an animal model [169]. The optimized formulation, containing sodium deoxycholate, had a mean particle size of 202.3 nm with an EE of 83.8%, and zeta potential value of 30.8 mV. It demonstrated a delayed release profile in vitro, enhanced skin permeability ex vivo, and caused no skin irritation upon histological examination. In a carrageenan-induced paw edema model in rats the topical application of developed bilosome gel significantly reduced paw swelling to 24.4% after 12 h, outperforming other treatments.
In another investigation, apigenin-loaded bilosomes exhibited spherical, nanosized vesicles (~210 nm to 430 nm), good PDI (<0.5), negative charge (−15 to −29 mV), and high encapsulation efficiency (~70% to 80%) [170]. The chitosan-coated optimized formulation (F2C1) resulted in an increased vesicle size (~300 nm), positive zeta potential (+17 mV), enhanced encapsulation efficiency (~90%), and improved drug release (~70%). The chitosan coating enhanced permeation and mucoadhesion (p < 0.05), as chitosan facilitates the opening of tight junctions, improving drug delivery. The formulation showed greater antimicrobial activity and higher efficacy against cancer cell lines, indicating its potential as an alternative to conventional delivery systems.
PEGylated bilosomes represent a novel approach by improving the stability, bioavailability, and therapeutic efficacy of poorly soluble phytochemicals. PEGylation provides a protective hydrophilic layer that prevents degradation, prolongs circulation time by reducing immune recognition, and enables controlled release of phytochemicals [171]. PEGylation also improves solubility, absorption, and targeting through passive and active mechanisms. Additionally, it enhances mucoadhesion and GI retention for oral delivery and supports better penetration for parenteral, topical, and ocular applications. Composed of lipids, surfactants, bile salts, and polyethylene glycol, this vesicle shares structural similarities with liposomes but offers additional benefits, including protection against enzymatic degradation in the GIT. They address limitations of traditional liposomes, such as encapsulation efficiency, stability, vesicle lysis, and scalability challenges. Bile salts like sodium deoxycholate serve as nontoxic, biocompatible solubilizing agents and are known to enhance permeation [172]. Additionally, PEGylated bilosomes provide high drug-loading capacity and enable controlled, extended drug release. Biochanin A-loaded PEGylated bilosomes was successfully formulated using the TFH method and optimized through Box–Behnken design [173]. The optimized Biochanin A-loaded PEGylated bilosomes demonstrated key properties (particle size; 216 ± 6.62 nm, EE; 80.54 ± 1.02%, PDI; 0.231, and zeta potential; −15.4 mV). The formulation exhibited prolonged drug release (88.23 ± 3.54% over 24 h) and high ex vivo intestinal permeation (56.97 ± 2.76% in 6 h). It achieved 4.70-fold higher bioavailability and significantly greater anti-inflammatory activity at all time points compared to pure drug dispersion.
Surface engineering of bilosomes is crucial to enhance their functionality and target specific delivery. Hyaluronic acid, a key extracellular matrix component and glycosaminoglycan in synovial fluid protects articular cartilage and regulates inflammation, cellular migration, and angiogenesis through various receptors. Hyaluronic acid-functionalized bilosomes have been developed for targeted delivery of tripterine, an anti-inflammatory phytomedicine, to inflamed joints via ligand–receptor interactions [164]. The formulation was developed using the TFH method with a cationic lipid and subsequent coating with hyaluronan. The optimized formulation was characterized by particle size (118.5 nm), EE (99.56%), and structural morphology. Studies on in vitro drug release, hemocompatibility, and cellular uptake confirmed excellent performance, with developed bilosomes achieving prolonged circulation time and a significant increase in intraarthritic bioavailability (799.9% compared to drug solution). In vivo evaluations demonstrated superior antiarthritic efficacy and inflammation resolution compared to uncoated vesicles.
A novel method for functionalizing phosphatidylcholine/cholesterol-based vesicular systems with sodium cholate hydrate (biosurfactant) and Pluronic P123 (triblock copolymer) was developed by [165]. Bilosomes were prepared using TFH combined with sonication, extrusion, and homogenization. Optimization studies evaluated nanocarrier size, charge, morphology, and stability, identifying a promising formulation for co-loading hydrophilic (methylene blue) and hydrophobic (curcumin) compounds. The high encapsulation efficiency highlights their potential for applications in drug delivery, anticancer therapies, and diagnostics. Another study effectively demonstrated the potential of bilosomes containing sulphated polysaccharide protein complexes of Enteromorpha intestinalis for oral therapy in hepatocellular carcinoma [163]. Treatment with optimized formulation (EH-Bilo-2) showed a substantial reduction in various biomarkers related to cancer, signifying the anticancer and antiangiogenic properties of developed bilosomes.
Photodynamic therapy, known for its high efficacy, safety, and noninvasive nature, has received significant interest as a therapeutic option for various cancer treatments. This approach is approved by the FDA for treating basal and squamous cell carcinomas as well as certain esophageal and lung cancers; it uses photosensitizing agents and laser radiation to generate reactive oxygen species (ROS) that kill cancer cells, damage tumor blood vessels, and activate immune responses. However, its effectiveness in metastatic cancers like melanoma is limited by defense mechanisms, melanin interference, and reduced photosensitizer activity [174]. Recent research has focused on plant-derived bioactive compounds, such as curcumin, as complementary agents in this minimally invasive treatment. These compounds enhance treatment by boosting ROS generation, inducing apoptosis, inhibiting angiogenesis and metastasis, suppressing cell proliferation, and modulating critical molecular pathways (e.g., MAPK, microRNA, p21). Self-assembled bilosomes loaded with curcumin, stabilized with L-α-phosphatidylcholine, sodium cholate, Pluronic® P123, and cholesterol, were developed as a nanoplatform for photodynamic therapy [175]. These bilosomes co-encapsulated methylene blue (classical photosensitizer) and curcumin, as confirmed through spectroscopic techniques. In vitro studies demonstrated the stability, biocompatibility, and enhanced phototoxicity of the bilosomes’ formulation on melanoma (Me45) and epithelial (A375) cell lines, with selective uptake in tumor cells. Notably, the nanoplatform reduced Me45 melanoma cell viability to below 20% post-irradiation, outperforming non-encapsulated agents. Bilosomes encapsulating selected phytochemicals are presented in Table 5, along with details of their composition, preparation methods, and key characteristics.

3.6. Transferosomes

Transferosomes are ultra-deformable vesicular carriers composed of phospholipids, and edge activators (e.g., surfactants like Tween 80 or Span 80), which provide elasticity to the vesicles [184]. They are widely employed in transdermal drug delivery, particularly for systemic therapy and targeting deeper tissues. Unlike conventional liposomes, which are limited by the rigidity of their lipid bilayer, transferosomes incorporate edge activators that impart high flexibility, allowing them to traverse narrow intercellular spaces in the stratum corneum and penetrate via transfollicular routes [185]. This enhanced deformability contributes to improved skin permeation and deeper tissue penetration. Transferosomes are especially effective in delivering phytochemicals, as they enhance the stability, bioavailability, and permeability of these compounds while protecting them from degradation [186]. Additionally, they offer benefits, such as targeted delivery, sustained release, and suitability, for both hydrophilic and lipophilic drugs through a noninvasive route [187]. However, their application is associated with certain limitations, including formulation complexity, stability concerns, limited drug-loading capacity, and high production costs. Despite these challenges, transferosomes represent a promising strategy to improve the therapeutic efficacy of phytochemicals, particularly if scalability and stability issues are adequately addressed.
A recent study developed transferosomes incorporating Rhodomyrtus tomentosa leaf extract using L-α-phosphatidylcholine, cholesterol and sodium deoxycholate as edge activators [188]. Extract-loaded transferosomes formulated as a gel exhibited 81.90% EE, spherical vesicles (405.3 nm), low polydispersity index (0.16), and high zeta potential (−61.62 mV). They contained significant phenolic (15.65 μg gallic acid equivalent/g extract) and flavonoid (43.13 μg QE/g extract) levels. Extract-loaded transferosomes demonstrated greater antimicrobial activity (minimum inhibitory and bactericidal concentrations: 8–1024 μg/mL), strong antioxidant activity, and moderate tyrosinase inhibition (IC50: 245.32 μg/mL). Biocompatibility tests showed IC50 values of 7.05 and 4.73 μg/mL for L929 fibroblast and vero cells, respectively, and significantly reduced nitric oxide production by 6.78–88.25%. Stability studies confirmed durability under freeze-thaw conditions, with no significant changes in sedimentation or pH.
Transferosomes are often stabilized by incorporating hydrophilic polymers, which interact with the bilayer surfaces to create sterically stabilized vesicles in dispersion. Jabuticaba extract, rich in polyphenols, flavonoids, anthocyanins, and gallic acid, demonstrates strong antioxidant activity and promising biological efficacy [189]. The extract, incorporated into transferosomes modified with polymers (hydroxyethyl cellulose or sodium hyaluronate), demonstrated a larger particle size and greater storage stability compared to transferosomes without polymers [190]. The extract-loaded vesicles outperformed a reference solution in mitigating hydrogen peroxide toxicity and accelerating wound healing in cell monolayers, particularly with polymer enriched formulations. Table 6 presents transferosomes, ethosomes and transethosomes loaded with selected phytochemicals, providing details on their composition, preparation methods, and key features.

3.7. Ethosomes

Ethosomes, a third-generation vesicular system, are particularly notable due to their makeup with phospholipids and ethanol, which improves drug solubility and stability [215]. These structures consist of phospholipids (20–45%), water, and significant amounts of short-chain alcohols such as ethanol (20–45%) and isopropyl alcohol or propylene glycol (up to 15%) [216]. Their enhanced delivery capability, compared to vesicles like liposomes, is largely attributed to ethanol [216]. Ethanol, a known permeation enhancer, increases lipid fluidity and reduces the density of the lipid multilayer, a phenomenon called the ethanol effect. This is followed by the ethosome effect, where the nanovesicles penetrate and fuse with skin lipids, opening new pathways and releasing bioactive compounds into the skin’s deeper layers [215].
Pluronic F127 (Poloxamer 407), a nonionic triblock copolymer composed of a hydrophobic polypropylene oxide core flanked by hydrophilic polyethylene oxide segments, can self-assemble into micelles or form ordered aggregates depending on its concentration [217]. The modified novel plurethosomes, combining Pluronic F127 with phosphatidylcholine and ethanol, have been shown to significantly interact with lipid bilayers, aided by polypropylene oxide blocks, thereby enhancing the permeation of associated drugs. Mangiferin-loaded plurethosomes formed multilamellar vesicles that demonstrated particle sizes ranging from 200 to 550 nm, influenced by phosphatidylcholine concentration and the presence of polysorbate or Pluronic [218]. The biphasic release profile of plurethosomes highlighted their superior drug retention capability compared to unilamellar vesicles like transethosomes.
These carriers improve the delivery of both hydrophilic and lipophilic drugs without triggering adverse effects. Similarly, deformable vesicles, like transferosomes and transethosomes, incorporate penetration enhancers, enabling them to traverse biological barriers more effectively [22]. Despite their advantages, vesicular systems face challenges such as scalability, potential aggregation, and the need for precise formulation techniques to ensure long-term stability. However, their unique attributes, including targeted delivery, controlled drug release, and reduced systemic side effects, make them highly advantageous for various applications. These include the delivery of small molecules, proteins, vaccines, and even gene therapies, showcasing their versatility in addressing diverse therapeutic needs [219].

3.8. Transethosomes

Transethosomes are an advanced version of ethosomes, typically composed of ethanol, an edge activator (surfactant), phospholipids, and water, along with optional stabilizers (e.g., cholesterol), buffering agents, penetration enhancers, and imaging dyes, merging the properties of transferosomes and ethosomes [220]. Integrating surfactants with phosphatidylcholine in ethosome formulations enhances membrane flexibility and significantly improves their potential for transdermal delivery. This unique combination enhances biocompatibility, vesicle flexibility, and encapsulation efficiency compared to individual transferosomes and ethosomes [221]. Their particle size typically varies from 50 to 350 nm, mainly depending on the physicochemical characteristics of the drug [222].
Transethosomal gels were developed to overcome the low viscosity and retention of transethosomes, enhancing the skin delivery of herbal extracts and phytochemicals. Transethosomes gels loaded with extracts such as ginger, curcumin, and black cumin seed demonstrate enhanced antioxidant, anti-inflammatory, and anti-infective properties [223]. Recent research also highlights the incorporation of plant bioactives, like apigenin, glycyrrhizic acid, sinapic acid, hesperidin, hexatriacontane, mangiferin, catechin, and quercetin, into transethosome vesicles, resulting in improved bioavailability, stability, and solubility [223]. Transethosomes loaded with bioactive compounds from plants, such as Carissa carandas L. extract, or metabolites from plants, such as Chenopodium murale [208], have demonstrated effectiveness in therapeutic areas including antifungal, antibacterial, rheumatoid arthritis, antihyperlipidemic, anti-inflammatory, and cardiovascular treatments. These advancements highlight significant progress in transdermal delivery systems, offering promising applications for managing and treating various diseases [224]. Transethosomes offer a dual drug delivery system, enabling simultaneous transdermal and systemic administration, and are capable of encapsulating drugs with varying molecular weights. They address challenges like poor solubility, low bioavailability, and limited skin permeability, as seen in ginger extract [211] and catechins [225]. Their small particle size and elastic nature enhance skin penetration, making them effective for bioactive agents and large peptides, as demonstrated with palmitoyl pentapeptide [226]. This carrier has shown improvement in bioavailability, therapeutic efficacy, and patient compliance while minimizing the adverse effects of mangiferin [227]. However, challenges such as agglomeration, coagulation, and product loss during media transfer require careful preparation to ensure stability and efficiency.
Transethosomes overcome the stratum corneum barrier by leveraging their deformable and elastic vesicles, which allow them to penetrate deeply through the SC without rupturing, enhancing drug delivery in transdermal systems. Phospholipid ethanol interaction enhances drug permeation by disrupting the skin’s lipid bilayer and increasing fluidity, enabling flexible transethosome vesicles to penetrate deeper skin layers [224]. This carrier utilizes two primary mechanisms to enhance skin penetration: hydrotaxis and elastomechanics. Hydrotaxis drives the carrier along the skin’s hydration gradient, promoting lipid adhesion and membrane fusion for effective entry. Elastomechanics increases their flexibility upon dehydration, allowing the carrier to navigate tight skin junctions and penetrate deeply into the epidermis or dermis, improving drug delivery efficiency [228].
Encapsulation within transethosomes protects sensitive phytochemicals from environmental degradation, such as oxidation, light, and heat, resulting in improved stability and extended shelf life. The gel matrix complements this system by providing viscosity and bioadhesive properties, ensuring prolonged skin residence time and localized drug delivery while reducing systemic side effects [223]. Moreover, transethosomes can accommodate both hydrophilic and lipophilic phytochemicals, increasing formulation versatility. The non-greasy, cooling nature of gels further enhances patient compliance, making the formulation user-friendly. However, challenges exist in this approach. The formulation process utilizes typical methods, such as hot homogenization, cold methods, the thin-film lipid hydration method, and the mechanical dispersion technique, but may also involve advanced techniques like sonication or extrusion, which can increase production costs. Stability concerns may still arise due to potential interactions between phytochemicals, phospholipids, and gel excipients, affecting efficacy. Additionally, transethosomes have a limited drug-loading capacity, and certain excipients may irritate sensitive skin. Scaling up production to ensure consistent vesicle size and uniformity can also pose difficulties. Despite these limitations, careful optimization of materials and processes can maximize the benefits of transethosomal gel formulations for delivering phytochemicals effectively.

3.9. Cubosomes

Cubosomes are nanostructured lipid-based carriers with a cubic phase geometry, characterized by a highly ordered honeycomb-like internal structure formed through the self-assembly of amphiphilic lipids in water. Stabilization is achieved via electrostatic interactions or steric factors, leading to the development of nonlamellar phases [229]. Cubosomes are unique nanomaterials made from unsaturated monoglycerides like glycerol monooleate and phytantriol, forming bicontinuous cubic liquid crystalline structures [230]. Their lipid-rich core enables high loading of hydrophobic compounds, while their partially water-exposed surface allows effective delivery of hydrophilic molecules. Glycerol monooleate is widely used as a matrix for cubosome fabrication due to its nontoxic, biodegradable, and biocompatible properties [231]. This monoglyceride can form various structures upon contact with water, including reversed micellar phases and liquid crystalline phases (lamellar, reversed hexagonal, and cubic), depending on the preparation temperature and concentration [232]. Phytantriol is a stable, biocompatible, and biodegradable alternative to glycerol monooleate, offering higher resistance to esterase degradation [233]. It forms an inverse cubic structure in water at room temperature and transitions to a reverse hexagonal phase at elevated temperatures (40–60 °C), making it ideal for bicontinuous cubic crystalline phases.
Few studies have explored tailoring cubosome membranes for specific applications like drug delivery, imaging agents, and biosensors. One approach involves modifying the curvature of lipidic systems by introducing substances such as cholesterol (30 mol%), sucrose stearate (20 mol%), or octyl glucoside (10 mol%) to lipid bilayers, which alters the curvature elasticity, packing efficiency, and water channel diameter, potentially leading to phase transitions [234]. The incorporation of poloxamer into the cubosomes led to an increase in their complex viscosity, suggesting the formation of a tighter network structure that enhanced resistance to erosion in the prepared cubosomes [230].
Nanosized liquid lyotropic crystalline particles enable the loading of hydrophilic, lipophilic, and amphipathic compounds, offering enhanced solubility, stability, and bioavailability. Cubosomes play a critical role in delivering drugs by providing controlled and sustained release, enhancing cell permeation, improving absorption, and enabling targeted delivery through surface modifications [235]. In phytochemical delivery, cubosomes overcome challenges such as poor solubility, low stability, and limited bioavailability of plant-derived compounds. They enhance solubility, protect phytochemicals from degradation caused by environmental factors, and provide sustained release for prolonged therapeutic effects. Additionally, their biocompatibility and ability to transport across biological membranes make them highly effective for phytochemical delivery. However, cubosomes’ applications face challenges such as complex preparation methods, difficulties in scaling up production, sensitivity to environmental conditions, and potential cytotoxicity in certain formulations. Encapsulation efficiency can also vary depending on the physicochemical properties of specific phytochemicals. Despite these limitations, cubosomes hold significant potential as innovative carriers for phytochemical delivery, addressing key limitations of conventional formulations. The potential of this carrier in delivering capsaicin for topical and transdermal delivery was reported [236]. Two formulations were developed using glycerol monooleate and phytantriol and have shown sustained release as well as skin targeting. Another study reported the wound healing efficacy of β-sitosterol, a phytosterol, when formulated into cubosomes [237]. Additionally, the cubosome formulation of Ruta graveolens extract has demonstrated promising results in allergic asthma [238]. Table 7 summarizes various cubosome-loaded phytochemicals, highlighting their composition, preparation methods, and key features.

4. Preparation Methods

Phytosomes are prepared using various techniques that enhance the bioavailability and stability of plant-derived bioactive compounds. These methods include the rotary evaporator, anti-solvent precipitation, cosolvency, salting out, supercritical fluid, and freeze-drying, each with specific advantages and limitations. Conventional methods for liposome preparation include techniques such as gentle hydration of a phospholipid film, electroformation, coalescence of small vesicles, solvent injection, and detergent dialysis [246]. Other methods include reverse phase evaporation, the solvent spherule method, and size reduction of large liposome vesicles (e.g., ultrasonication, extrusion). In addition, there are novel methods such as microfluidic techniques (like micro hydrodynamic focusing, microfluidic droplets, pulsed jet flow microfluidics, lipid film hydration in microtubes), supercritical fluids, modified electroformation, freeze-drying of double emulsions, the membrane contactor method, hydration of phospholipids deposited on nanostructured materials, liposome formation by curvature tuning, and biomimetic reactions for vesicular self-assembly [247]. Vesicular nanocarriers are prepared using diverse techniques depending on their composition and intended application. Invasomes are typically produced by TFH followed by sonication or extrusion to enhance skin penetration [248]. Niosomes utilize methods like THF, ether injection, or microfluidization, forming bilayers from nonionic surfactants and cholesterol [249]. Bilosomes, incorporating bile salts for improved gastrointestinal stability, are prepared via TFH and further processed through sonication or extrusion [165,250]. Transferosomes involve TFH with edge activators and may use ultrasonic dispersion or ethanol injection to produce ultra-deformable vesicles [251]. Ethosomes, prepared by hot or cold methods with ethanol and phospholipids, often include sonication or extrusion to achieve optimal vesicle size [252]. Transethosomes combine features of ethosomes and transferosomes, using similar methods to enhance skin delivery [221,253]. Cubosomes are prepared either by dispersing bulk cubic phases (top-down) or by self-assembly from lipid solutions (bottom-up), typically stabilized with polymers like Pluronic F127 [229]. Table 8 provides a comprehensive overview of the preparation techniques, commonly used excipients, key features and stability characteristics of various vesicular nanocarriers used for the delivery of phytochemicals.

4.1. Thin Film Hydration

The TFH technique is one of the most widely used and classical methods for the preparation of vesicular carriers. In this method, lipids or surfactants and other excipients are first dissolved in a volatile organic solvent, typically chloroform or a chloroform–methanol mixture. The solvent is then evaporated under reduced pressure using a rotary evaporator, forming a thin, dry lipid film on the walls of a round-bottom flask. This film is subsequently hydrated with a buffered aqueous phase under continuous agitation, leading to the spontaneous formation of multilamellar vesicles. The resulting vesicles can be further processed using sonication or extrusion to achieve the desired size and lamellarity. This technique is appreciated for its simplicity, versatility, and ability to encapsulate both hydrophilic and lipophilic drugs. The phytosomes are prepared by mixing a stoichiometric ratio of phytochemicals and phospholipids in an appropriate organic solvent using the same procedure. TFH is a simple and widely used method but requires precise temperature control to prevent the degradation of sensitive phytochemicals. The solvent evaporation method was used to make complexes of oleanolic acid–phospholipid [254], while a rapid solvent evaporation method followed by a self-assembly technique was used to prepare berberine phospholipid complexes for the development of a berberine drug delivery system with higher efficiency [255]. Nano-phytosomes are phyto-phospholipid complexes, typically under 200 nm in size, designed to enhance absorption and biological activity. They have a composition similar to regular phytosomes or phyto-phospholipid complexes but offer improved delivery due to their nanoscale size. A stable colloidal phytosome delivery system was developed to enhance the antioxidant properties of pomegranate peel extract by utilizing phosphatidylcholine and gamma oryzanol through the TFH/sonication method [256]. The resulting homogeneous spherical nano-phytosomes at a ratio of 8:2:2% w/w (extract: phosphatidylcholine: gamma oryzanol) exhibited nano size (60.61 ± 0.81 nm), negative zeta potential (−32.24 ± 0.84 mV), acceptable polydispersity index (PDI, 0.19 ± 0.01), loading (19.13 ± 0.30%), and encapsulation (95.66 ± 1.52%). In a different study, the phytosome prepared using the TFH method with egg yolk phosphatidylcholine demonstrated greater encapsulation efficiency (98.27%) and better solubility (89.15%) [257]. It also exhibited good total phenolic content (114.44 mg gallic acid equivalent/g), ferric reducing antioxidant power (15.77 mmol Trolox/g), and radical scavenging activity (9.49 mmol Trolox/g). A phyto-phospholipid complex formulation encapsulating Calendula officinalis extract and gold nanoparticles was made using the TFH method with extrusion. The prepared vesicle with low particle size (under 100 nm) exhibited good encapsulation efficiency for both quercetin and chlorogenic acid [258]. They demonstrated notable wound healing and antioxidant activities compared to free components and plain liposomes. Cellular interactions were confirmed using fluorescence microscopy with Texas Red-labeled vesicles.

4.2. Anti-Solvent Precipitation Technique

The anti-solvent precipitation technique involves dissolving the phyto-phospholipid complex in a solvent, followed by precipitation with a nonsolvent, offering high encapsulation efficiency and cost-effectiveness. However, it may lead to incomplete precipitation and solvent residues that can affect product purity. Phytosome nanosuspensions for silybin–phospholipid complexes were successfully developed using the traditional solvent evaporation/antisolvent precipitation method to address potential formulation challenges of silybin such as low bioavailability due to poor solubility [259]. The complexes were prepared through the formation of weak intermolecular interactions between the polar head of the phospholipids and the hydroxyl group of silybin. In another study, hydroalcoholic Murraya koenigii extract and soy lecithin were dissolved in alcohol, while cholesterol was dissolved in dichloromethane [260]. The mixture was subsequently heated to 60 °C, concentrated, and hexane was added as an antisolvent to induce precipitation. Similarly, a phyto-phospholipid complex consisting of orange peel and liquorice extracts was refluxed with dichloromethane, followed by evaporation and precipitation with n-hexane [261]. Additionally, phytosomes incorporating Bombax ceiba extract [262] and Allium cepa phospholipid complexes [263] were prepared by this method.

4.3. Cosolvency Method

The cosolvency method is a widely used technique for preparing phyto-phospholipid complexes, involving the dissolution of plant extracts and phospholipids in compatible solvents, such as methanol, to prepare a uniform mixture. The extract is added dropwise to the phospholipid solution with continuous stirring to form the phytosomal complex [264]. This method is ideal for poorly soluble compounds and offers several advantages, including simplicity, cost-effectiveness, and the ability to achieve high encapsulation efficiency with controlled particle size. However, its limitations include the potential for solvent residues, the need for careful solvent selection to avoid degradation of sensitive compounds, and scalability challenges for large-scale production. A nano-phytosome in thermogel containing Glycine max (soybean) extract was developed and evaluated for its antiobesity effects on body weight, adipose tissue size, and lipid profile [78]. Three preparation methods, cosolvency, solvent evaporation, and salting out, were employed. The optimized formulation based on the cosolvency method demonstrated high encapsulation efficiency (>99%), particle sizes ranging from 51.66 to 650.67 nm, and drug release rates of 77.61% to 99.78%. Fourier-transform infrared and TEM analyses confirmed successful phytosome formation. In vivo studies showed significant reductions in body weight, adipose tissue mass, and lipid profile, highlighting its potential as an antiobesity treatment.

4.4. Salting-Out Method

The salting out technique involves adding a salt solution to reduce the solubility of the phyto-phospholipid complex, causing precipitation. It is cost-effective, minimizes organic solvent use, and is easy to scale, but may require optimization to avoid salt residue affecting purity. In a reported method, diosmin and soy phosphatidylcholine complexes were prepared using three methods: solvent evaporation, salting out, and lyophilization [265]. According to the solvent evaporation method, the drug and phospholipid were dissolved in a methanol–dioxane mixture, refluxed, and the solvent was evaporated; diosmin and lipids were added to a mixture of dimethyl sulfoxide, dehydrated ethanol, and chloroform, stirred until they were dissolved, and then precipitated with n-hexane.

4.5. Freeze-Drying Technique

The freeze-drying method involves freezing the phyto-phospholipid mixture and sublimating the solvent under vacuum to produce a dry phytosome powder, resulting in highly stable, lightweight, and porous formulations that are suitable for thermolabile compounds [266]. Using the lyophilization technique, both ingredients were mixed with dimethyl sulfoxide and t-butyl alcohol, stirred, and then lyophilized at −70 °C and −40 °C. The final complex had a yield of 90.4% and was stored in a desiccator at 4 °C. In another attempt, kaempferol phospholipid complexes were prepared using the lyophilization method [267].

4.6. Supercritical Fluid Technique

The supercritical fluid technique is an advanced method for preparing phytosomes, utilizing carbon dioxide under controlled conditions to produce thermally stable and high purity formulations with uniform particle sizes [62]. The process involves dissolving phytoconstituents and phospholipids in fluid, leading to phytosome formation upon controlled pressure or temperature reduction. Key advantages include ecofriendliness, enhanced bioavailability, and scalability, while limitations involve high costs, technical expertise, and safety concerns. Additionally, certain phytoconstituents may exhibit poor solubility in this fluid, necessitating the use of co-solvents that can complicate the process, and batch-to-batch consistency remains a challenge during scale-up. Development and evaluation of puerarin phospholipid complex microparticles using the supercritical fluids method have been described [268]. The effects of various processing variables, such as temperature, solution concentration, pressure, flow rate of supercritical carbon dioxide, and the drug solution on lipid complex particle characteristics were assessed. The complex obtained using this method consisted of amorphous, partially agglomerated spheres with particle sizes around 1 μm.

5. In Vitro and In Vivo Tests for Vesicular Carriers

In vitro tests simulate biological conditions to predict carrier behavior and optimize formulations before animal or human studies [269,270]. Table 9 presents frequently used in vitro and in vivo tests for vesicular carriers employed in various phytochemical delivery applications. Drug release studies, cell-based (Caco-2 cell permeability assay, parallel artificial membrane permeability assay), intestinal (Everted gut sac assay, Ussing chamber assay), blood–brain barrier (human brain microvascular endothelial cells (hCMEC/D3), skin based (Franz-diffusion cell assay), and endothelial cell-based (Electric cell-substrate impedance sensing) permeability tests, particle size, and zeta potential analyses assess release kinetics, absorption potential, stability, and colloidal properties. Additional tests, like antioxidant assays such as 2,2-Diphenyl-1-picrylhydrazyl (DPPH), 2,2′-Azino-bis(3-ethylbenzothiazoline-6-sulfonic acid (ABTS), Ferric Reducing Antioxidant Power (FRAP), Oxygen Radical Absorbance Capacity (ORAC), Ferrous Ion Chelating (FIC), Cupric Reducing Antioxidant Capacity (CUPRAC) and anti-inflammatory assays including enzyme-based (Cyclooxygenase inhibition assay, lipoxygenase inhibition assay, inducible nitric oxide synthase inhibition assay), cytokine-based (Tumor necrosis factor-alpha (TNF-α) inhibition assay, interleukin inhibition assays), cell-based (RAW 264.7 macrophage assay, peripheral blood mononuclear cell assay), oxidative stress-based (Reactive oxygen species (ROS) inhibition assay, lipid peroxidation assay), in vivo animal model (Carrageenan-induced paw edema test, cotton pellet granuloma assay, acetic acid-induced vascular permeability assay), and nuclear factor kappa B inhibition assay, measure the protective effects of phytochemicals, while photostability, thermal and oxidative stability tests evaluate degradation under environmental stress. Immunomodulatory cell-based (Lymphocyte proliferation, toll-like receptor activation), delayed-type hypersensitivity or Cyclophosphamide-induced immunosuppression animal models evaluate the ability of phytochemicals to either enhance or suppress the immune system. These in vitro evaluations provide crucial preliminary data on release, bioactivity, and stability. In vivo experiments, conversely, are performed using animal models to evaluate pharmacokinetics, distribution within the body, and effectiveness of treatments. Pharmacokinetic studies measure drug absorption, distribution, metabolism, and elimination through parameters like Cmax, Tmax, AUC, and half-life. Biodistribution studies track drug accumulation in target organs, while therapeutic efficacy tests evaluate specific biological effects, including anticancer, neuroprotective, and cardioprotective activities. Specialized tests, such as antiviral (cytopathic effect reduction assay, Quantitative PCR assay), antimicrobial (Kirby-Bauer test, time-kill kinetics assay, live/dead staining (SYTO9/PI), in vivo animal models (Murine sepsis model, wound infection model), and ex vivo and biofilm assays, antidiabetic (Streptozotocin (STZ) or Alloxan induced diabetic animal model), and hepatoprotective (Carbon tetrachloride or Paracetamol induced liver toxicity animal model), cardioprotective (Ischemia-reperfusion (I/R) injury animal model, myocardial infarction (MI) animal model), neuroprotective (Stroke, Alzheimer’s disease, Parkinson’s disease, traumatic brain injury animal model, Morris water maze test, au and β-Amyloid (Aβ) ELISA) provide insights into the carrier’s potential in specific disease models. Cell viability, necrosis and apoptosis tests such as 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, annexin V/PI staining, caspase-3/7, terminal dUTP nick end-labeling assays are widely used in phytochemical research to evaluate the biological activity, cytotoxicity, and therapeutic potential of plant-derived compounds. Together, these in vivo tests confirm the formulation’s safety, efficacy, and drug targeting capabilities. Overall, both types of evaluations are critical for ensuring that phytochemical-loaded vesicular carriers deliver therapeutic compounds effectively and maintain stability for optimal clinical outcomes.

6. Clinical Trials and Patents

Clinical trials and patents on phytochemicals emphasize their transformative role in modern drug delivery and therapeutic applications. In clinical research, various phytochemicals have demonstrated promising outcomes for conditions ranging from cancer to metabolic and cardiovascular diseases. For example, liposomal curcumin has progressed to Phase IIb trials (NCT02138955) for treating locally advanced metastatic cancer, aiming to improve pharmacokinetics and safety profiles through enhanced bioavailability. Similarly, ursolic acid (NCT02337933), betulinic acid (NCT03904511), and lutein (NCT01534533) are being investigated for metabolic syndrome, psychological stress, and carotid atherosclerosis, respectively, highlighting their potential for treating chronic and lifestyle-related disorders. Furthermore, compounds like thymoquinone and ginsenoside Rg3 have shown potential in reducing cancer recurrence and managing vascular dementia. Clinical trials on phytochemicals in vesicular carriers are limited, despite preclinical evidence supporting their potential to enhance bioavailability and therapeutic efficacy. The lack of registered trials highlights the need for further research to bridge the gap between preclinical findings and clinical applications.
Patents offer complementary insights by presenting innovations that optimize the delivery and therapeutic effectiveness of phytochemicals (Table 10). For instance, a recent invention features a brain-targeting liposome incorporating a paeonol-ozagrel conjugate disclosed in patent CN107823649B, designed to cross the BBB and provide prolonged drug circulation for cerebral conditions [295]. Another patent (CN105534907A) describes a macrophage membrane-based nano-delivery system with betulinic acid capable of inhibiting cancer proliferation by targeting specific cellular pathways. Other notable developments include liposomal systems for neurodegenerative diseases using echinacoside and synergistic cancer treatments combining thymoquinone and taxane. Additionally, formulations like allicin liposomes offer enhanced antimicrobial, protozoal, and tumor fighting capabilities, broadening the application scope of phytochemicals. Together these clinical and patented innovations underscore the strategic integration of phytochemicals in nanovesicular delivery systems, addressing critical issues like drug solubility, stability, and targeted delivery. These advances are poised to improve therapeutic efficacy, reduce side effects, and expand the range of diseases treatable by naturally derived compounds. This dual pathway of clinical validation and technological innovation fosters the translation of phytochemicals from research to practical, patient-centered treatments.

7. Future Perspectives, Challenges and Conclusions

The future of phytochemical delivery through various vesicular carriers discussed here offers immense potential for improving the therapeutic efficacy and bioavailability of plant-derived compounds. These carriers provide targeted drug delivery, enhanced solubility, and prolonged circulation times, addressing key limitations of conventional formulations. For instance, ethosomes and transethosomes have shown promise in transdermal delivery due to their flexibility and ability to penetrate deep into the skin. Liposomes, composed of lipid bilayer vesicles, can encapsulate both hydrophilic and lipophilic phytochemicals, enhancing solubility, stability, and controlled release [296]. Nonionic surfactant-based niosomes offer greater stability than liposomes, enhancing the transdermal and oral bioavailability of phytochemicals. Cubosomes, with their unique cubic structure, offer high drug-loading capacity and sustained release, making them suitable for chronic conditions [297]. Phytosomes, a specialized carrier, form complexes with phytochemicals such as flavonoids and polyphenols to enhance absorption and stability, particularly for poorly water-soluble compounds like curcumin and quercetin, which are widely used in nutraceutical formulations [186]. Similarly, nanophytosomes represent a recent advancement in lipid-based vesicles, characterized by their smaller size, designed to further enhance the delivery of plant-derived nutraceuticals. Nanotechnology improves the delivery and efficacy of phytotherapeutics and photosensitizers in photodynamic therapy by enabling their simultaneous encapsulation in biocompatible nanostructures. This approach overcomes pharmacokinetic challenges typically encountered in combination therapy, enhancing selective tumor accumulation and synergistic therapeutic effects through the enhanced permeability and retention effect [298]. Hybrid vesicular systems integrate different vesicle types to enhance the stability, encapsulation efficiency, and bioavailability of phytochemicals while overcoming individual limitations. A liposome–niosome hybrid system developed for curcumin demonstrated improved encapsulation, stability in biological fluids, and enhanced bioavailability compared to conventional vesicles [299]. Stimuli-responsive vesicles, which release phytochemicals in response to specific stimuli such as pH, temperature, or enzymes, are being investigated for targeted therapy [27]. Surface-modified vesicles functionalized with ligands such as folic acid and peptides are being investigated for the active targeting of phytochemicals to specific cells or tissues, enhancing therapeutic precision and efficacy [297]. Nano-vesicular carriers, such as liposomes, niosomes, and polymersomes, are designed to withstand gastric degradation and enhance intestinal absorption, improving the oral bioavailability of phytochemicals. Recent advancements, including chitosan-based nanoparticles, have further optimized solubility, stability, and bioactivity for more effective phytochemical delivery [33]. However, several challenges remain in optimizing these delivery systems. Stability during storage, large-scale production, and regulatory approval for these complex nanostructures pose significant hurdles. The encapsulation efficiency and controlled release of phytochemicals can also vary depending on the carrier type and preparation technique. Additionally, the high cost of some vesicular systems, along with the need for advanced characterization methods to ensure quality and reproducibility, can limit widespread applications. Low encapsulation efficiency in nanocarriers limits the therapeutic potential of phytochemicals in pharmacotherapy. Enhancing phytochemical loading through composition changes or layer-by-layer designs, along with surface modifications like ligand attachment or tumor-penetrating peptides, can improve targeting and penetration across biological barriers. Future research should focus on designing biomimetic, stimuli-responsive nanostructures to optimize targeted phytochemical delivery in disease treatment. Research must focus on developing scalable, cost-effective manufacturing processes and enhancing carrier stability while maintaining phytochemical efficacy. Collaborative efforts between research institutions and the pharmaceutical industry, alongside advancements in nanotechnology and regulatory frameworks, will be crucial to fully harness the therapeutic potential of phytochemicals through these innovative delivery platforms. Innovative preparation techniques, surface modifications, and hybrid systems play a crucial role in enhancing the clinical applicability of phytochemical delivery. Ongoing research and technological advancements will further optimize these nanocarriers, ensuring their continued relevance in treating chronic and complex diseases while improving patient outcomes.

Author Contributions

Conceptualization, S.J. and A.B.N.; literature review, S.J., F.S.K., S.H.S.B., R.R. and A.B.N.; formal analysis, S.J., F.S.K., S.H.S.B., R.R. and A.B.N.; data curation, S.J., F.S.K., S.H.S.B., R.R. and A.B.N.; writing—original draft preparation, S.J., F.S.K., S.H.S.B. and R.R.; writing—review and editing, S.J. and A.B.N. All authors have read and agreed to the published version of the manuscript.

Funding

This article did not receive any funding and the APC was funded by Gulf Medical University, Ajman, United Arab Emirates.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study is contained within this article.

Acknowledgments

The authors are highly thankful to the College of Pharmacy, Gulf Medical University, Ajman, United Arab Emirates, for the support and encouragement.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ng, P.Q.; Ling, L.S.C.; Chellian, J.; Madheswaran, T.; Panneerselvam, J.; Kunnath, A.P.; Gupta, G.; Satija, S.; Mehta, M.; Hansbro, P.M.; et al. Applications of Nanocarriers as Drug Delivery Vehicles for Active Phytoconstituents. Curr. Pharm. Des. 2020, 26, 4580–4590. [Google Scholar] [CrossRef] [PubMed]
  2. Alfei, S.; Schito, A.M.; Zuccari, G. Nanotechnological Manipulation of Nutraceuticals and Phytochemicals for Healthy Purposes: Established Advantages vs. Still Undefined Risks. Polymers 2021, 13, 2262. [Google Scholar] [CrossRef] [PubMed]
  3. Rasmus, P.; Kozłowska, E. Antioxidant and Anti-Inflammatory Effects of Carotenoids in Mood Disorders: An Overview. Antioxidants 2023, 12, 676. [Google Scholar] [CrossRef] [PubMed]
  4. Ginwala, R.; Bhavsar, R.; Chigbu, D.I.; Jain, P.; Khan, Z.K. Potential Role of Flavonoids in Treating Chronic Inflammatory Diseases with a Special Focus on the Anti-Inflammatory Activity of Apigenin. Antioxidants 2019, 8, 35. [Google Scholar] [CrossRef]
  5. Gómez-Zorita, S.; González-Arceo, M.; Fernández-Quintela, A.; Eseberri, I.; Trepiana, J.; Portillo, M.P. Scientific Evidence Supporting the Beneficial Effects of Isoflavones on Human Health. Nutrients 2020, 12, 3853. [Google Scholar] [CrossRef]
  6. Rauf, A.; Imran, M.; Abu-Izneid, T.; Iahtisham Ul, H.; Patel, S.; Pan, X.; Naz, S.; Sanches Silva, A.; Saeed, F.; Rasul Suleria, H.A. Proanthocyanidins: A comprehensive review. Biomed. Pharmacother. 2019, 116, 108999. [Google Scholar] [CrossRef]
  7. Kunnumakkara, A.B.; Hegde, M.; Parama, D.; Girisa, S.; Kumar, A.; Daimary, U.D.; Garodia, P.; Yenisetti, S.C.; Oommen, O.V.; Aggarwal, B.B. Role of Turmeric and Curcumin in Prevention and Treatment of Chronic Diseases: Lessons Learned from Clinical Trials. ACS Pharmacol. Transl. Sci. 2023, 6, 447–518. [Google Scholar] [CrossRef]
  8. Jacob, S.; Kather, F.S.; Morsy, M.A.; Boddu, S.H.S.; Attimarad, M.; Shah, J.; Shinu, P.; Nair, A.B. Advances in Nanocarrier Systems for Overcoming Formulation Challenges of Curcumin: Current Insights. Nanomaterials 2024, 14, 672. [Google Scholar] [CrossRef]
  9. Unnikrishnan Meenakshi, D.; Narde, G.K.; Ahuja, A.; Akhtar, M.J.; Khan, S.A. Role of Natural Phytoconstituents as a Potential Bioenhancer of Anti-Cancer and Anti-Microbial Agents: Spotlight on the Mechanism of Action, Clinical Studies and Patents. Processes 2024, 12, 2060. [Google Scholar] [CrossRef]
  10. Thorat, S.S.; Gujar, K.N.; Karale, C.K. Bioenhancers from mother nature: An overview. Future J. Pharm. Sci. 2023, 9, 20. [Google Scholar] [CrossRef]
  11. Ara, N.; Sultana, T.; Bolleddu, R.; Venkatesh, S.; Kiran, A.R. A strategy to enhance bioavailability of drug candidates: Natural bioenhancers. Int. J. Pharm. Bio Med. Sci. 2021, 1, 10–14. [Google Scholar]
  12. Tripathi, A.K.; Ray, A.K.; Mishra, S.K. Molecular and pharmacological aspects of piperine as a potential molecule for disease prevention and management: Evidence from clinical trials. Beni-Suef Univ. J. Basic Appl. Sci. 2022, 11, 16. [Google Scholar] [CrossRef]
  13. Breedveld, P.; Beijnen, J.H.; Schellens, J.H. Use of P-glycoprotein and BCRP inhibitors to improve oral bioavailability and CNS penetration of anticancer drugs. Trends Pharmacol. Sci. 2006, 27, 17–24. [Google Scholar] [CrossRef] [PubMed]
  14. Kang, M.J.; Cho, J.Y.; Shim, B.H.; Kim, D.K.; Lee, J. Bioavailability enhancing activities of natural compounds from medicinal plants. J. Med. Plants Res. 2009, 3, 1204–1211. [Google Scholar]
  15. Makhov, P.; Golovine, K.; Canter, D.; Kutikov, A.; Simhan, J.; Corlew, M.M.; Uzzo, R.G.; Kolenko, V.M. Co-administration of piperine and docetaxel results in improved anti-tumor efficacy via inhibition of CYP3A4 activity. Prostate 2012, 72, 661–667. [Google Scholar] [CrossRef]
  16. Kumar, V.; Mittal, V.; Jalwal, P. Role of herbal bioenhancers (Piperine and Curcumin) on the oral bioavailability of Tamoxifen using experimental rats. J. Pharmacogn. Phytochem. 2015, 4, 133–136. [Google Scholar]
  17. Chakraborty, T.; Gupta, S.; Nair, A.; Chauhan, S.; Saini, V. Wound healing potential of insulin-loaded nanoemulsion with Aloe vera gel in diabetic rats. J. Drug Deliv. Sci. Technol. 2021, 64, 102601. [Google Scholar] [CrossRef]
  18. Ahmad, R.; Srivastava, S.; Ghosh, S.; Khare, S.K. Phytochemical delivery through nanocarriers: A review. Colloids Surf. B Biointerfaces 2021, 197, 111389. [Google Scholar] [CrossRef]
  19. Patra, J.K.; Das, G.; Fraceto, L.F.; Campos, E.V.R.; Rodriguez-Torres, M.D.P.; Acosta-Torres, L.S.; Diaz-Torres, L.A.; Grillo, R.; Swamy, M.K.; Sharma, S.; et al. Nano based drug delivery systems: Recent developments and future prospects. J. Nanobiotechnol. 2018, 16, 71. [Google Scholar] [CrossRef]
  20. Singh, V.K.; Arora, D.; Ansari, M.I.; Sharma, P.K. Phytochemicals based chemopreventive and chemotherapeutic strategies and modern technologies to overcome limitations for better clinical applications. Phytother. Res. 2019, 33, 3064–3089. [Google Scholar] [CrossRef]
  21. Dhiman, N.; Awasthi, R.; Sharma, B.; Kharkwal, H.; Kulkarni, G.T. Lipid Nanoparticles as Carriers for Bioactive Delivery. Front. Chem. 2021, 9, 580118. [Google Scholar] [CrossRef]
  22. Seenivasan, R.; Halagali, P.; Nayak, D.; Tippavajhala, V.K. Transethosomes: A Comprehensive Review of Ultra-Deformable Vesicular Systems for Enhanced Transdermal Drug Delivery. AAPS PharmSciTech 2025, 26, 41. [Google Scholar] [CrossRef]
  23. Santos, A.C.; Rodrigues, D.; Sequeira, J.A.D.; Pereira, I.; Simões, A.; Costa, D.; Peixoto, D.; Costa, G.; Veiga, F. Nanotechnological breakthroughs in the development of topical phytocompounds-based formulations. Int. J. Pharm. 2019, 572, 118787. [Google Scholar] [CrossRef]
  24. Nair, A.B.; Shah, J.; Al-Dhubiab, B.E.; Patel, S.S.; Morsy, M.A.; Patel, V.; Chavda, V.; Jacob, S.; Sreeharsha, N.; Shinu, P. Development of asialoglycoprotein receptor-targeted nanoparticles for selective delivery of gemcitabine to hepatocellular carcinoma. Molecules 2019, 24, 4566. [Google Scholar] [CrossRef]
  25. Shi, L.; Zhang, J.; Zhao, M.; Tang, S.; Cheng, X.; Zhang, W.; Li, W.; Liu, X.; Peng, H.; Wang, Q. Effects of polyethylene glycol on the surface of nanoparticles for targeted drug delivery. Nanoscale 2021, 13, 10748–10764. [Google Scholar] [CrossRef]
  26. Morsy, M.A.; Nair, A.B. Prevention of rat liver fibrosis by selective targeting of hepatic stellate cells using hesperidin carriers. Int. J. Pharm. 2018, 552, 241–250. [Google Scholar] [CrossRef] [PubMed]
  27. Zandieh, M.A.; Farahani, M.H.; Daryab, M.; Motahari, A.; Gholami, S.; Salmani, F.; Karimi, F.; Samaei, S.S.; Rezaee, A.; Rahmanian, P.; et al. Stimuli-responsive (nano)architectures for phytochemical delivery in cancer therapy. Biomed. Pharmacother. 2023, 166, 115283. [Google Scholar] [CrossRef]
  28. Jin, Y.; Wu, Z.; Li, C.; Zhou, W.; Shaw, J.P.; Baguley, B.C.; Liu, J.; Zhang, W. Optimization of Weight Ratio for DSPE-PEG/TPGS Hybrid Micelles to Improve Drug Retention and Tumor Penetration. Pharm. Res. 2018, 35, 13. [Google Scholar] [CrossRef]
  29. Wang, J. Combination Treatment of Cervical Cancer Using Folate-Decorated, pH-Sensitive, Carboplatin and Paclitaxel Co-Loaded Lipid-Polymer Hybrid Nanoparticles. Drug Des. Dev. Ther. 2020, 14, 823–832. [Google Scholar] [CrossRef]
  30. Seder, I.; Zheng, T.; Zhang, J.; Rojas, C.C.; Helalat, S.H.; Téllez, R.C.; Sun, Y. A Scalable Microfluidic Platform for Nanoparticle Formulation: For Exploratory- and Industrial-Level Scales. Nano Lett. 2024, 24, 5132–5138. [Google Scholar] [CrossRef]
  31. Lin, H.; Leng, J.; Fan, P.; Xu, Z.; Ruan, G. Scalable production of microscopic particles for biological delivery. Mater. Adv. 2023, 4, 2885–2908. [Google Scholar] [CrossRef]
  32. Ng, C.Y.; Kee, L.T.; Al-Masawa, M.E.; Lee, Q.H.; Subramaniam, T.; Kok, D.; Ng, M.H.; Law, J.X. Scalable Production of Extracellular Vesicles and Its Therapeutic Values: A Review. Int. J. Mol. Sci. 2022, 23, 7986. [Google Scholar] [CrossRef] [PubMed]
  33. Imam, S.S.; Alshehri, S.; Ghoneim, M.M.; Zafar, A.; Alsaidan, O.A.; Alruwaili, N.K.; Gilani, S.J.; Rizwanullah, M. Recent Advancement in Chitosan-Based Nanoparticles for Improved Oral Bioavailability and Bioactivity of Phytochemicals: Challenges and Perspectives. Polymers 2021, 13, 4036. [Google Scholar] [CrossRef] [PubMed]
  34. Ashfaq, R.; Rasul, A.; Asghar, S.; Kovács, A.; Berkó, S.; Budai-Szűcs, M. Lipid Nanoparticles: An Effective Tool to Improve the Bioavailability of Nutraceuticals. Int. J. Mol. Sci. 2023, 24, 5764. [Google Scholar] [CrossRef]
  35. Kamboj, S.; Bala, S.; Nair, A.B. Solid lipid nanoparticles: An effective lipid based technology for poorly water soluble drugs. Int. J. Pharm. Sci. Rev. Res. 2010, 5, 78–90. [Google Scholar]
  36. Afzal, O.; Rizwanullah, M.; Altamimi, A.S.A.; Alossaimi, M.A.; Kamal, M.; Ahmad, J. Harnessing natural polysaccharides-based nanoparticles for oral delivery of phytochemicals: Knocking down the barriers. J. Drug Deliv. Sci. Technol. 2023, 82, 104368. [Google Scholar] [CrossRef]
  37. Nicolescu, A.; Babotă, M.; Barros, L.; Rocchetti, G.; Lucini, L.; Tanase, C.; Mocan, A.; Bunea, C.I.; Crișan, G. Bioaccessibility and bioactive potential of different phytochemical classes from nutraceuticals and functional foods. Front. Nutr. 2023, 10, 1184535. [Google Scholar] [CrossRef]
  38. Hu, Y.; Lin, Q.; Zhao, H.; Li, X.; Sang, S.; McClements, D.J.; Long, J.; Jin, Z.; Wang, J.; Qiu, C. Bioaccessibility and bioavailability of phytochemicals: Influencing factors, improvements, and evaluations. Food Hydrocoll. 2023, 135, 108165. [Google Scholar] [CrossRef]
  39. Yin, X.; Chen, K.; Cheng, H.; Chen, X.; Feng, S.; Song, Y.; Liang, L. Chemical Stability of Ascorbic Acid Integrated into Commercial Products: A Review on Bioactivity and Delivery Technology. Antioxidants 2022, 11, 153. [Google Scholar] [CrossRef]
  40. Negi, P.S. Stability of phytochemicals at the point of sale. In Handbook of Plant Food Phytochemicals: Sources, Stability and Extraction; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2013; pp. 375–395. [Google Scholar]
  41. Mrázková, M.; Sumczynski, D.; Orsavová, J. Influence of Storage Conditions on Stability of Phenolic Compounds and Antioxidant Activity Values in Nutraceutical Mixtures with Edible Flowers as New Dietary Supplements. Antioxidants 2023, 12, 962. [Google Scholar] [CrossRef]
  42. Li, D.; Xiao, Y.; Zhang, Z.; Liu, C. Light-induced oxidation and isomerization of all-trans-β-cryptoxanthin in a model system. J. Photochem. Photobiol. B Biol. 2015, 142, 51–58. [Google Scholar] [CrossRef] [PubMed]
  43. Lu, B.; Zhao, Y. Photooxidation of phytochemicals in food and control: A review. Ann. N. Y. Acad. Sci. 2017, 1398, 72–82. [Google Scholar] [CrossRef] [PubMed]
  44. Yang, J.; He, X.; Zhao, D. Factors affecting phytochemical stability. In Handbook of Plant Food Phytochemicals: Sources, Stability and Extraction; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2013; pp. 332–374. [Google Scholar]
  45. Censi, R.; Di Martino, P. Polymorph Impact on the Bioavailability and Stability of Poorly Soluble Drugs. Molecules 2015, 20, 18759–18776. [Google Scholar] [CrossRef]
  46. Darji, M.A.; Lalge, R.M.; Marathe, S.P.; Mulay, T.D.; Fatima, T.; Alshammari, A.; Lee, H.K.; Repka, M.A.; Narasimha Murthy, S. Excipient Stability in Oral Solid Dosage Forms: A Review. AAPS PharmSciTech 2018, 19, 12–26. [Google Scholar] [CrossRef]
  47. González-González, O.; Ramirez, I.O.; Ramirez, B.I.; O’Connell, P.; Ballesteros, M.P.; Torrado, J.J.; Serrano, D.R. Drug Stability: ICH versus Accelerated Predictive Stability Studies. Pharmaceutics 2022, 14, 2324. [Google Scholar] [CrossRef]
  48. Odriozola-Serrano, I.; Nogueira, D.P.; Esparza, I.; Vaz, A.A.; Jiménez-Moreno, N.; Martín-Belloso, O.; Ancín-Azpilicueta, C. Stability and Bioaccessibility of Phenolic Compounds in Rosehip Extracts during In Vitro Digestion. Antioxidants 2023, 12, 1035. [Google Scholar] [CrossRef]
  49. Salmani, J.M.; Asghar, S.; Lv, H.; Zhou, J. Aqueous solubility and degradation kinetics of the phytochemical anticancer thymoquinone; probing the effects of solvents, pH and light. Molecules 2014, 19, 5925–5939. [Google Scholar] [CrossRef]
  50. Wang, X.; Liu, L.; Xia, S.; Muhoza, B.; Cai, J.; Zhang, X.; Duhoranimana, E.; Su, J. Sodium carboxymethyl cellulose modulates the stability of cinnamaldehyde-loaded liposomes at high ionic strength. Food Hydrocoll. 2019, 93, 10–18. [Google Scholar] [CrossRef]
  51. Gabrič, A.; Hodnik, Ž.; Pajk, S. Oxidation of Drugs during Drug Product Development: Problems and Solutions. Pharmaceutics 2022, 14, 325. [Google Scholar] [CrossRef]
  52. Zhou, F.; Peterson, T.; Fan, Z.; Wang, S. The Commonly Used Stabilizers for Phytochemical-Based Nanoparticles: Stabilization Effects, Mechanisms, and Applications. Nutrients 2023, 15, 3881. [Google Scholar] [CrossRef]
  53. Bakrim, S.; Aboulaghras, S.; El Menyiy, N.; El Omari, N.; Assaggaf, H.; Lee, L.H.; Montesano, D.; Gallo, M.; Zengin, G.; AlDhaheri, Y.; et al. Phytochemical Compounds and Nanoparticles as Phytochemical Delivery Systems for Alzheimer’s Disease Management. Molecules 2022, 27, 9043. [Google Scholar] [CrossRef] [PubMed]
  54. Melim, C.; Magalhães, M.; Santos, A.C.; Campos, E.J.; Cabral, C. Nanoparticles as phytochemical carriers for cancer treatment: News of the last decade. Expert Opin. Drug Deliv. 2022, 19, 179–197. [Google Scholar] [CrossRef] [PubMed]
  55. Pandey, R.; Bhairam, M.; Shukla, S.S.; Gidwani, B. Colloidal and vesicular delivery system for herbal bioactive constituents. DARU J. Pharm. Sci. 2021, 29, 415–438. [Google Scholar] [CrossRef]
  56. Nair, A.B.; Dalal, P.; Kadian, V.; Kumar, S.; Garg, M.; Rao, R.; Almuqbil, R.M.; Alnaim, A.S.; Aldhubiab, B.; Alqattan, F. Formulation Strategies for Enhancing Pharmaceutical and Nutraceutical Potential of Sesamol: A Natural Phenolic Bioactive. Plants 2023, 12, 1168. [Google Scholar] [CrossRef]
  57. Kotta, S.; Eshmawi, B.A.; Alahmadi, A.A.; Aldawsari, H.M.; Badr-Eldin, S.M.; Nair, A.B. Bioactivities, Biopharmaceutics, and Advanced Drug Delivery Systems of Magnolol. Indian J. Pharm. Educ. Res. 2024, 58, s1102–s1121. [Google Scholar]
  58. Lagoa, R.; Silva, J.; Rodrigues, J.R.; Bishayee, A. Advances in phytochemical delivery systems for improved anticancer activity. Biotechnol. Adv. 2020, 38, 107382. [Google Scholar] [CrossRef]
  59. Date, T.; Nimbalkar, V.; Kamat, J.; Mittal, A.; Mahato, R.I.; Chitkara, D. Lipid-polymer hybrid nanocarriers for delivering cancer therapeutics. J. Control. Release 2018, 271, 60–73. [Google Scholar] [CrossRef]
  60. Barani, M.; Sangiovanni, E.; Angarano, M.; Rajizadeh, M.A.; Mehrabani, M.; Piazza, S.; Gangadharappa, H.V.; Pardakhty, A.; Mehrbani, M.; Dell’Agli, M.; et al. Phytosomes as Innovative Delivery Systems for Phytochemicals: A Comprehensive Review of Literature. Int. J. Nanomed. 2021, 16, 6983–7022. [Google Scholar] [CrossRef]
  61. Pu, Y.; Zhang, X.; Zhang, Q.; Wang, B.; Chen, Y.; Zang, C.; Wang, Y.; Dong, T.T.; Zhang, T. 20(S)-Protopanaxadiol Phospholipid Complex: Process Optimization, Characterization, In Vitro Dissolution and Molecular Docking Studies. Molecules 2016, 21, 1396. [Google Scholar] [CrossRef]
  62. Lu, M.; Qiu, Q.; Luo, X.; Liu, X.; Sun, J.; Wang, C.; Lin, X.; Deng, Y.; Song, Y. Phyto-phospholipid complexes (phytosomes): A novel strategy to improve the bioavailability of active constituents. Asian J. Pharm. Sci. 2019, 14, 265–274. [Google Scholar] [CrossRef]
  63. Babazadeh, A.; Zeinali, M.; Hamishehkar, H. Nano-Phytosome: A Developing Platform for Herbal Anti-Cancer Agents in Cancer Therapy. Curr. Drug Targets 2018, 19, 170–180. [Google Scholar] [CrossRef] [PubMed]
  64. Kidd, P.M. Bioavailability and activity of phytosome complexes from botanical polyphenols: The silymarin, curcumin, green tea, and grape seed extracts. Altern. Med. Rev. 2009, 14, 226–246. [Google Scholar] [PubMed]
  65. Talebi, M.; Shahbazi, K.; Dakkali, M.S.; Akbari, M.; Almasi Ghale, R.; Hashemi, S.; Sashourpour, M.; Mojab, F.; Aminzadeh, S. Phytosomes: A promising nanocarrier system for enhanced bioavailability and therapeutic efficacy of herbal products. Phytomed. Plus 2025, 5, 100779. [Google Scholar] [CrossRef]
  66. Giori, A.; Franceschi, F. Phospholipid Complexes of Curcumin Having Improved Bioavailability. U.S. Patent 20090131373A1, 31 March 2020. [Google Scholar]
  67. Kidd, P.; Head, K. A review of the bioavailability and clinical efficacy of milk thistle phytosome: A silybin-phosphatidylcholine complex (Siliphos). Altern. Med. Rev. 2005, 10, 193–203. [Google Scholar]
  68. Li, F.; Yang, X.; Yang, Y.; Li, P.; Yang, Z.; Zhang, C. Phospholipid complex as an approach for bioavailability enhancement of echinacoside. Drug Dev. Ind. Pharm. 2015, 41, 1777–1784. [Google Scholar] [CrossRef]
  69. Kiefer, D.; Pantuso, T. Panax ginseng. Am. Fam. Physician 2003, 68, 1539–1542. [Google Scholar]
  70. Petrangolini, G.; Ronchi, M.; Frattini, E.; De Combarieu, E.; Allegrini, P.; Riva, A. A New Food-grade Coenzyme Q10 Formulation Improves Bioavailability: Single and Repeated Pharmacokinetic Studies in Healthy Volunteers. Curr. Drug Deliv. 2019, 16, 759–767. [Google Scholar] [CrossRef]
  71. Cesarone, M.R.; Belcaro, G.; Hu, S.; Dugall, M.; Hosoi, M.; Ledda, A.; Feragalli, B.; Maione, C.; Cotellese, R. Supplementary prevention and management of asthma with quercetin phytosome: A pilot registry. Minerva Medica 2019, 110, 524–529. [Google Scholar] [CrossRef]
  72. Di Pierro, F.; Menghi, A.B.; Barreca, A.; Lucarelli, M.; Calandrelli, A. Greenselect Phytosome as an adjunct to a low-calorie diet for treatment of obesity: A clinical trial. Altern. Med. Rev. 2009, 14, 154–160. [Google Scholar]
  73. Riva, A.; Allegrini, P.; Franceschi, F.; Togni, S.; Giacomelli, L.; Eggenhoffner, R. A novel boswellic acids delivery form (Casperome®) in the management of musculoskeletal disorders: A review. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 5258–5263. [Google Scholar] [CrossRef]
  74. Drobnic, F.; Riera, J.; Appendino, G.; Togni, S.; Franceschi, F.; Valle, X.; Pons, A.; Tur, J. Reduction of delayed onset muscle soreness by a novel curcumin delivery system (Meriva®): A randomised, placebo-controlled trial. J. Int. Soc. Sports Nutr. 2014, 11, 31. [Google Scholar] [CrossRef] [PubMed]
  75. Magrone, T.; Pugliese, V.; Fontana, S.; Jirillo, E. Human use of Leucoselect® Phytosome® with special reference to inflammatory-allergic pathologies in frail elderly patients. Curr. Pharm. Des. 2014, 20, 1011–1019. [Google Scholar] [CrossRef] [PubMed]
  76. Di Pierro, F.; Togni, S.; Franceschi, F.; Eghenhofner, R.; Giacomelli, L. Effects of standardized Ginkgo biloba extract complexed with phosphatidylserine (Virtiva®) on physiological response to prolonged, intense physical activity. Minerva Ortop. Traumatol. 2016, 67, 119–123. [Google Scholar]
  77. Sbrini, G.; Brivio, P.; Fumagalli, M.; Giavarini, F.; Caruso, D.; Racagni, G.; Dell’Agli, M.; Sangiovanni, E.; Calabrese, F. Centella asiatica L. Phytosome Improves Cognitive Performance by Promoting Bdnf Expression in Rat Prefrontal Cortex. Nutrients 2020, 12, 355. [Google Scholar] [CrossRef]
  78. El-Menshawe, S.F.; Ali, A.A.; Rabeh, M.A.; Khalil, N.M. Nanosized soy phytosome-based thermogel as topical anti-obesity formulation: An approach for acceptable level of evidence of an effective novel herbal weight loss product. Int. J. Nanomed. 2018, 13, 307–318. [Google Scholar] [CrossRef]
  79. Alam, M.A.; Al-Jenoobi, F.I.; Al-Mohizea, A.M. Commercially bioavailable proprietary technologies and their marketed products. Drug Discov. Today 2013, 18, 936–949. [Google Scholar] [CrossRef]
  80. Toma, L.; Deleanu, M.; Sanda, G.M.; Barbălată, T.; Niculescu, L.; Sima, A.V.; Stancu, C.S. Bioactive Compounds Formulated in Phytosomes Administered as Complementary Therapy for Metabolic Disorders. Int. J. Mol. Sci. 2024, 25, 4162. [Google Scholar] [CrossRef]
  81. Pottathil, S.; Nain, P.; Morsy, M.A.; Kaur, J.; Al-Dhubiab, B.E.; Jaiswal, S.; Nair, A.B. Mechanisms of antidiabetic activity of methanolic extract of Punica granatum leaves in nicotinamide/streptozotocin-induced type 2 diabetes in rats. Plants 2020, 9, 1609. [Google Scholar] [CrossRef]
  82. Gorain, B.; Al-Dhubiab, B.E.; Nair, A.; Kesharwani, P.; Pandey, M.; Choudhury, H. Multivesicular Liposome: A Lipid-based Drug Delivery System for Efficient Drug Delivery. Curr. Pharm. Des. 2021, 27, 4404–4415. [Google Scholar]
  83. Nsairat, H.; Khater, D.; Sayed, U.; Odeh, F.; Al Bawab, A.; Alshaer, W. Liposomes: Structure, composition, types, and clinical applications. Heliyon 2022, 8, e09394. [Google Scholar] [CrossRef]
  84. Gabizon, A.A.; Tahover, E.; Golan, T.; Geva, R.; Perets, R.; Amitay, Y.; Shmeeda, H.; Ohana, P. Pharmacokinetics of mitomycin-c lipidic prodrug entrapped in liposomes and clinical correlations in metastatic colorectal cancer patients. Investig. New Drugs 2020, 38, 1411–1420. [Google Scholar] [CrossRef] [PubMed]
  85. Kolter, M.; Wittmann, M.; Köll-Weber, M.; Süss, R. The suitability of liposomes for the delivery of hydrophobic drugs—A case study with curcumin. Eur. J. Pharm. Biopharm. 2019, 140, 20–28. [Google Scholar] [CrossRef] [PubMed]
  86. Caddeo, C.; Gabriele, M.; Fernàndez-Busquets, X.; Valenti, D.; Fadda, A.M.; Pucci, L.; Manconi, M. Antioxidant activity of quercetin in Eudragit-coated liposomes for intestinal delivery. Int. J. Pharm. 2019, 565, 64–69. [Google Scholar] [CrossRef] [PubMed]
  87. Liu, Y.; Gao, D.; Zhang, X.; Liu, Z.; Dai, K.; Ji, B.; Wang, Q.; Luo, L. Antitumor drug effect of betulinic acid mediated by polyethylene glycol modified liposomes. Mater. Sci. Eng. C Mater. Biol. Appl. 2016, 64, 124–132. [Google Scholar] [CrossRef]
  88. Shu, Q.; Wu, J.; Chen, Q. Synthesis, Characterization of Liposomes Modified with Biosurfactant MEL—A Loading Betulinic Acid and Its Anticancer Effect in HepG2 Cell. Molecules 2019, 24, 3939. [Google Scholar] [CrossRef]
  89. Righeschi, C.; Coronnello, M.; Mastrantoni, A.; Isacchi, B.; Bergonzi, M.C.; Mini, E.; Bilia, A.R. Strategy to provide a useful solution to effective delivery of dihydroartemisinin: Development, characterization and in vitro studies of liposomal formulations. Colloids Surf. B Biointerfaces 2014, 116, 121–127. [Google Scholar] [CrossRef]
  90. Caddeo, C.; Teskac, K.; Sinico, C.; Kristl, J. Effect of resveratrol incorporated in liposomes on proliferation and UV-B protection of cells. Int. J. Pharm. 2008, 363, 183–191. [Google Scholar] [CrossRef]
  91. Santos, A.C.; Veiga, F.; Ribeiro, A.J. New delivery systems to improve the bioavailability of resveratrol. Expert Opin. Drug Deliv. 2011, 8, 973–990. [Google Scholar] [CrossRef]
  92. Singh, M.; Devi, S.; Rana, V.S.; Mishra, B.B.; Kumar, J.; Ahluwalia, V. Delivery of phytochemicals by liposome cargos: Recent progress, challenges and opportunities. J. Microencapsul. 2019, 36, 215–235. [Google Scholar] [CrossRef]
  93. Chavda, V.P.; Vihol, D.; Mehta, B.; Shah, D.; Patel, M.; Vora, L.K.; Pereira-Silva, M.; Paiva-Santos, A.C. Phytochemical-loaded liposomes for anticancer therapy: An updated review. Nanomedicine 2022, 17, 547–568. [Google Scholar] [CrossRef]
  94. Dutt, Y.; Pandey, R.P.; Dutt, M.; Gupta, A.; Vibhuti, A.; Raj, V.S.; Chang, C.-M.; Priyadarshini, A. Liposomes and phytosomes: Nanocarrier systems and their applications for the delivery of phytoconstituents. Coord. Chem. Rev. 2023, 491, 215251. [Google Scholar] [CrossRef]
  95. Jahanfar, S.; Gahavami, M.; Khosravi-Darani, K.; Jahadi, M.; Mozafari, M.R. Entrapment of rosemary extract by liposomes formulated by Mozafari method: Physicochemical characterization and optimization. Heliyon 2021, 7, e08632. [Google Scholar] [CrossRef]
  96. Sahu, R.K.; Aboulthana, W.M.; Mehta, D.K. Phyto-Phospholipid Complexation as a Novel Drug Delivery System for Management of Cancer with Better Bioavailability: Current Perspectives and Future Prospects. Anticancer Agents Med. Chem. 2021, 21, 1403–1412. [Google Scholar] [CrossRef] [PubMed]
  97. Allaw, M.; Manca, M.L.; Castangia, I.; Manconi, M. From plants to phospholipid vesicles: A comprehensive review on the incorporation of phytochemicals into phospholipid vesicles designed for skin applications with special focus on scalability and in vitro and in vivo efficacy. J. Drug Deliv. Sci. Technol. 2022, 67, 103049. [Google Scholar] [CrossRef]
  98. Sharma, D.; Rani, A.; Singh, V.D.; Shah, P.; Sharma, S.; Kumar, S. Glycerosomes: Novel Nano-Vesicles for Efficient Delivery of Therapeutics. Recent Adv. Drug Deliv. Formul. 2023, 17, 173–182. [Google Scholar] [CrossRef]
  99. Allaw, M.; Manconi, M.; Aroffu, M.; Marongiu, F.; Porceddu, M.; Bacchetta, G.; Usach, I.; Rached, R.A.; Rajha, H.N.; Maroun, R.G.; et al. Extraction, Characterization and Incorporation of Hypericum scruglii Extract in Ad Hoc Formulated Phospholipid Vesicles Designed for the Treatment of Skin Diseases Connected with Oxidative Stress. Pharmaceutics 2020, 12, 1010. [Google Scholar] [CrossRef]
  100. Manconi, M.; Petretto, G.; D’Hallewin, G.; Escribano, E.; Milia, E.; Pinna, R.; Palmieri, A.; Firoznezhad, M.; Peris, J.E.; Usach, I.; et al. Thymus essential oil extraction, characterization and incorporation in phospholipid vesicles for the antioxidant/antibacterial treatment of oral cavity diseases. Colloids Surf. B Biointerfaces 2018, 171, 115–122. [Google Scholar] [CrossRef]
  101. Manconi, M.; Manca, M.L.; Marongiu, F.; Caddeo, C.; Castangia, I.; Petretto, G.L.; Pintore, G.; Sarais, G.; D’Hallewin, G.; Zaru, M.; et al. Chemical characterization of Citrus limon var. pompia and incorporation in phospholipid vesicles for skin delivery. Int. J. Pharm. 2016, 506, 449–457. [Google Scholar] [CrossRef]
  102. Elkholy, N.S.; Shafaa, M.W.; Mohammed, H.S. Biophysical characterization of lutein or beta carotene-loaded cationic liposomes. RSC Adv. 2020, 10, 32409–32422. [Google Scholar] [CrossRef]
  103. Franca, L.; Ferraz, M.; Barros, M.C.; Gibson, V.; Xavier-Júnior, F.H.; Magalhães, N.S.S.; Lira-Nogueira, M. ConA-Coated Liposomes as a System to Delivery β-Lapachone to Breast Cancer Cells. Anticancer Agents Med. Chem. 2022, 22, 968–977. [Google Scholar] [CrossRef]
  104. Lu, Q.; Lu, P.-M.; Piao, J.-H.; Xu, X.-L.; Chen, J.; Zhu, L.; Jiang, J.-G. Preparation and physicochemical characteristics of an allicin nanoliposome and its release behavior. LWT Food Sci. Technol. 2014, 57, 686–695. [Google Scholar] [CrossRef]
  105. Yu, Y.; Zu, C.; He, D.; Li, Y.; Chen, Q.; Chen, Q.; Wang, H.; Wang, R.; Chaurasiya, B.; Zaro, J.L.; et al. pH-dependent reversibly activatable cell-penetrating peptides improve the antitumor effect of artemisinin-loaded liposomes. J. Colloid Interface Sci. 2021, 586, 391–403. [Google Scholar] [CrossRef]
  106. Dubinin, M.V.; Semenova, A.A.; Ilzorkina, A.I.; Mikheeva, I.B.; Yashin, V.A.; Penkov, N.V.; Vydrina, V.A.; Ishmuratov, G.Y.; Sharapov, V.A.; Khoroshavina, E.I.; et al. Effect of betulin and betulonic acid on isolated rat liver mitochondria and liposomes. Biochim. Biophys. Acta (BBA) Biomembr. 2020, 1862, 183383. [Google Scholar] [CrossRef]
  107. Castangia, I.; Manca, M.L.; Razavi, S.H.; Nácher, A.; Díez-Sales, O.; Peris, J.E.; Allaw, M.; Terencio, M.C.; Usach, I.; Manconi, M. Canthaxanthin Biofabrication, Loading in Green Phospholipid Vesicles and Evaluation of In Vitro Protection of Cells and Promotion of Their Monolayer Regeneration. Biomedicines 2022, 10, 157. [Google Scholar] [CrossRef]
  108. Hong, S.-C.; Park, K.-M.; Hong, C.R.; Kim, J.-C.; Yang, S.-H.; Yu, H.-S.; Paik, H.-D.; Pan, C.-H.; Chang, P.-S. Microfluidic assembly of liposomes dual-loaded with catechin and curcumin for enhancing bioavailability. Colloids Surf. A Physicochem. Eng. Asp. 2020, 594, 124670. [Google Scholar] [CrossRef]
  109. Chen, W.; Cheng, F.; Swing, C.J.; Xia, S.; Zhang, X. Modulation effect of core-wall ratio on the stability and antibacterial activity of cinnamaldehyde liposomes. Chem. Phys. Lipids 2019, 223, 104790. [Google Scholar] [CrossRef]
  110. Ou, Z.; You, Y.; Yi, H.; Liu, X.; Tong, Y.; Liu, D.; Wang, J. Key Lipoprotein Receptor Targeted Echinacoside-Liposomes Effective Against Parkinson’s Disease in Mice Model. Int. J. Nanomed. 2024, 19, 8463–8483. [Google Scholar] [CrossRef]
  111. Chen, W.; Zou, M.; Ma, X.; Lv, R.; Ding, T.; Liu, D. Co-Encapsulation of EGCG and Quercetin in Liposomes for Optimum Antioxidant Activity. J. Food Sci. 2019, 84, 111–120. [Google Scholar] [CrossRef]
  112. Renault-Mahieux, M.; Vieillard, V.; Seguin, J.; Espeau, P.; Le, D.T.; Lai-Kuen, R.; Mignet, N.; Paul, M.; Andrieux, K. Co-Encapsulation of Fisetin and Cisplatin into Liposomes for Glioma Therapy: From Formulation to Cell Evaluation. Pharmaceutics 2021, 13, 970. [Google Scholar] [CrossRef]
  113. Song, Y.Y.; Yuan, Y.; Shi, X.; Che, Y.Y. Improved drug delivery and anti-tumor efficacy of combinatorial liposomal formulation of genistein and plumbagin by targeting Glut1 and Akt3 proteins in mice bearing prostate tumor. Colloids Surf. B Biointerfaces 2020, 190, 110966. [Google Scholar] [CrossRef]
  114. Zhang, X.; Chen, H.; Zhang, Y.; Huang, Q.; Feng, J.; Xing, H.; Fu, X.; Yan, X.; Zhang, Y.; Xu, Q.; et al. HA-DOPE-Modified Honokiol-Loaded Liposomes Targeted Therapy for Osteosarcoma. Int. J. Nanomed. 2022, 17, 5137–5151. [Google Scholar] [CrossRef]
  115. Zhu, J.; Hu, Q.; Shen, S. Enhanced antitumor efficacy and attenuated cardiotoxicity of doxorubicin in combination with lycopene liposomes. J. Liposome Res. 2020, 30, 37–44. [Google Scholar] [CrossRef] [PubMed]
  116. Shi, J.; Wu, Y.; Guo, S.; Zhang, H.; Chen, G.; Xu, X. The efficacy of anti-VEGF antibody-modified liposomes loaded with paeonol in the prevention and treatment of hypertrophic scars. Drug Dev. Ind. Pharm. 2019, 45, 439–455. [Google Scholar] [CrossRef] [PubMed]
  117. Imam, S.S.; Alshehri, S.; Altamimi, M.A.; Hussain, A.; Qamar, W.; Gilani, S.J.; Zafar, A.; Alruwaili, N.K.; Alanazi, S.; Almutairy, B.K. Formulation of Piperine-Chitosan-Coated Liposomes: Characterization and In Vitro Cytotoxic Evaluation. Molecules 2021, 26, 3281. [Google Scholar] [CrossRef]
  118. Wu, Z.; Wang, T.; Song, Y.; Lu, Y.; Chen, T.; Chen, P.; Hui, A.; Chen, Y.; Wang, H.; Zhang, W. Optimization on conditions of podophyllotoxin-loaded liposomes using response surface methodology and its activity on PC3 cells. J. Liposome Res. 2019, 29, 133–141. [Google Scholar] [CrossRef]
  119. Tang, L.; Li, K.; Zhang, Y.; Li, H.; Li, A.; Xu, Y.; Wei, B. Quercetin liposomes ameliorate streptozotocin-induced diabetic nephropathy in diabetic rats. Sci. Rep. 2020, 10, 2440. [Google Scholar] [CrossRef]
  120. Huang, M.; Liang, C.; Tan, C.; Huang, S.; Ying, R.; Wang, Y.; Wang, Z.; Zhang, Y. Liposome co-encapsulation as a strategy for the delivery of curcumin and resveratrol. Food Funct. 2019, 10, 6447–6458. [Google Scholar] [CrossRef]
  121. Peng, J.; Wang, Q.; Sun, R.; Zhang, K.; Chen, Y.; Gong, Z. Phospholipids of inhaled liposomes determine the in vivo fate and therapeutic effects of salvianolic acid B on idiopathic pulmonary fibrosis. J. Control. Release 2024, 371, 1–15. [Google Scholar] [CrossRef]
  122. Landucci, E.; Mazzantini, C.; Calvani, M.; Pellegrini-Giampietro, D.E.; Bergonzi, M.C. Evaluation of Conventional and Hyaluronic Acid-Coated Thymoquinone Liposomes in an In Vitro Model of Dry Eye. Pharmaceutics 2023, 15, 578. [Google Scholar] [CrossRef]
  123. Wang, B.; Xu, Q.; Zhou, C.; Lin, Y. Liposomes co-loaded with ursolic acid and ginsenoside Rg3 in the treatment of hepatocellular carcinoma. Acta Biochim. Pol. 2021, 68, 711–715. [Google Scholar] [CrossRef]
  124. Isacchi, B.; Bergonzi, M.C.; Iacopi, R.; Ghelardini, C.; Galeotti, N.; Bilia, A.R. Liposomal Formulation to Increase Stability and Prolong Antineuropathic Activity of Verbascoside. Planta Medica 2017, 83, 412–419. [Google Scholar] [CrossRef] [PubMed]
  125. Mao, W.; Wu, F.; Lee, R.J.; Lu, W.; Wang, J. Development of a stable single-vial liposomal formulation for vincristine. Int. J. Nanomed. 2019, 14, 4461–4474. [Google Scholar] [CrossRef]
  126. Tian, J.; Wang, L.; Wang, L.; Ke, X. A wogonin-loaded glycyrrhetinic acid-modified liposome for hepatic targeting with anti-tumor effects. Drug Deliv. 2014, 21, 553–559. [Google Scholar] [CrossRef]
  127. Kumar, B.; Pandey, M.; Aggarwal, R.; Sahoo, P.K. A comprehensive review on invasomal carriers incorporating natural terpenes for augmented transdermal delivery. Future J. Pharm. Sci. 2022, 8, 50. [Google Scholar] [CrossRef]
  128. Nangare, S.; Dugam, S. Smart invasome synthesis, characterizations, pharmaceutical applications, and pharmacokinetic perspective: A review. Future J. Pharm. Sci. 2020, 6, 123. [Google Scholar] [CrossRef]
  129. Duangjit, S.; Nimcharoenwan, T.; Chomya, N.; Locharoenrat, N.; Ngawhirunpat, T. Design and development of optimal invasomes for transdermal drug delivery using computer program. Asian J. Pharm. Sci. 2016, 11, 52–53. [Google Scholar] [CrossRef]
  130. Kumar, B.; Sahoo, P.K.; Manchanda, S. Development of eucalyptol enriched nano vesicles for better transdermal delivery of curcumin: Preparation, characterisation and ex vivo skin analysis. Nanomed. J. 2022, 9, 223–230. [Google Scholar] [CrossRef]
  131. Jain, S.; Tripathi, S.; Tripathi, P.K. Antiarthritic potential of berberine loaded invasomal gel. Phytomed. Plus 2022, 2, 100373. [Google Scholar] [CrossRef]
  132. Kumar, B.; Sahoo, P.K. Augmented Transdermal Delivery of Curcumin for the Effective Management of Plaque Psoriasis—Design, Formulation, Characterisation, and In Vivo Studies. AAPS PharmSciTech 2023, 24, 134. [Google Scholar] [CrossRef]
  133. El-Nabarawi, M.A.; Shamma, R.N.; Farouk, F.; Nasralla, S.M. Dapsone-Loaded Invasomes as a Potential Treatment of Acne: Preparation, Characterization, and In Vivo Skin Deposition Assay. AAPS PharmSciTech 2018, 19, 2174–2184. [Google Scholar] [CrossRef]
  134. Castangia, I.; Manca, M.L.; Caddeo, C.; Bacchetta, G.; Pons, R.; Demurtas, D.; Diez-Sales, O.; Fadda, A.M.; Manconi, M. Santosomes as natural and efficient carriers for the improvement of phycocyanin reepithelising ability in vitro and in vivo. Eur. J. Pharm. Biopharm. 2016, 103, 149–158. [Google Scholar] [CrossRef] [PubMed]
  135. Shinu, P.; Nair, A.B.; Kumari, B.; Jacob, S.; Kumar, M.; Tiwari, A.; Tiwari, V.; Venugopala, K.N.; Attimarad, M.; Nagaraja, S. Recent Advances and Appropriate use of Niosomes for the Treatment of Skin Cancer. Indian J. Pharm. Educ. Res. 2022, 56, 1–14. [Google Scholar]
  136. Shehata, T.M.; Nair, A.B.; Al-Dhubiab, B.E.; Shah, J.; Jacob, S.; Alhaider, I.A.; Attimarad, M.; Elsewedy, H.S.; Ibrahim, M.M. Vesicular emulgel based system for transdermal delivery of insulin: Factorial design and in vivo evaluation. Appl. Sci. 2020, 10, 5341. [Google Scholar] [CrossRef]
  137. Shah, J.; Nair, A.B.; Shah, H.; Jacob, S.; Shehata, T.M.; Morsy, M.A. Enhancement in antinociceptive and anti-inflammatory effects of tramadol by transdermal proniosome gel. Asian J. Pharm. Sci. 2020, 15, 786–796. [Google Scholar] [CrossRef]
  138. Momekova, D.B.; Gugleva, V.E.; Petrov, P.D. Nanoarchitectonics of Multifunctional Niosomes for Advanced Drug Delivery. ACS Omega 2021, 6, 33265–33273. [Google Scholar] [CrossRef]
  139. Han, H.S.; Koo, S.Y.; Choi, K.Y. Emerging nanoformulation strategies for phytocompounds and applications from drug delivery to phototherapy to imaging. Bioact. Mater. 2022, 14, 182–205. [Google Scholar] [CrossRef]
  140. Raafat, K.M.; El-Zahaby, S.A. Niosomes of active Fumaria officinalis phytochemicals: Antidiabetic, antineuropathic, anti-inflammatory, and possible mechanisms of action. Chin. Med. 2020, 15, 40. [Google Scholar] [CrossRef]
  141. Agarwal, S.; Mohamed, M.S.; Raveendran, S.; Rochani, A.K.; Maekawa, T.; Kumar, D.S. Formulation, characterization and evaluation of morusin loaded niosomes for potentiation of anticancer therapy. RSC Adv. 2018, 8, 32621–32636. [Google Scholar] [CrossRef]
  142. Parnian, F.; Hekmati-Moghaddam, S.H.; Majdizadeh, M.; Jebali, A.; Haghiralsadat, B.F. Fabrication of niosomal nano-carriers containing aqueous extract of hedera helix and comparison of toxicity of free extract and niosome extract on HT29 colorectal cancer cell line. J. Knowl. Health Basic Med. Sci. 2020, 15, 31–45. [Google Scholar] [CrossRef]
  143. Shahbazi, R.; Mirjafary, Z.; Zarghami, N.; Saeidian, H. Efficient PEGylated magnetic nanoniosomes for co-delivery of artemisinin and metformin: A new frontier in chemotherapeutic efficacy and cancer therapy. Sci. Rep. 2024, 14, 27380. [Google Scholar] [CrossRef]
  144. El-Banna, A.H.; Abo El-Ela, F.I.; Abdel-Wahab, A.; Gamal, A.; Abdel-Razik, A.-R.H.; El-Banna, H.A.; Elsamannoudy, S.I.; Ibrahim, M.A.; Abdelghany, A.K. Therapeutic efficacy of amygdaline and amygdaline-loaded niosomes in a rat model of Alzheimer’s disease via oxidative stress, brain neurotransmitters, and apoptotic pathway. Beni-Suef Univ. J. Basic Appl. Sci. 2024, 13, 117. [Google Scholar] [CrossRef]
  145. Palozza, P.; Muzzalupo, R.; Trombino, S.; Valdannini, A.; Picci, N. Solubilization and stabilization of beta-carotene in niosomes: Delivery to cultured cells. Chem. Phys. Lipids 2006, 139, 32–42. [Google Scholar] [CrossRef] [PubMed]
  146. Ravaghi, M.; Razavi, S.H.; Mousavi, S.M.; Sinico, C.; Fadda, A.M. Stabilization of natural canthaxanthin produced by Dietzia natronolimnaea HS-1 by encapsulation in niosomes. LWT 2016, 73, 498–504. [Google Scholar] [CrossRef]
  147. Gadapa, S.; Battula, S.N.; Mor, S.; Pushpadass, H.A.; Naik, L.N.; Emerald, M.E. Green tea catechin loaded niosomes: Formulation and their characterization for food fortification. J. Food Sci. Technol. 2022, 59, 3669–3682. [Google Scholar] [CrossRef]
  148. Esmaeili Rad, M.; Egil, A.C.; Ozaydin Ince, G.; Yuce, M.; Zarrabi, A. Optimization of curcumin loaded niosomes for drug delivery applications. Colloids Surf. A Physicochem. Eng. Asp. 2022, 654, 129921. [Google Scholar] [CrossRef]
  149. Hajizadeh, M.R.; Parvaz, N.; Barani, M.; Khoshdel, A.; Fahmidehkar, M.A.; Mahmoodi, M.; Torkzadeh-Mahani, M. Diosgenin-loaded niosome as an effective phytochemical nanocarrier: Physicochemical characterization, loading efficiency, and cytotoxicity assay. DARU J. Pharm. Sci. 2019, 27, 329–339. [Google Scholar] [CrossRef]
  150. Karaman Evren, D.; Kozgus Guldu, O.; Tut, E.; Medine, E.I. Nanoencapsulation of green tea catechin (−)-Epigallocatechin-3-Gallate (EGCG) in niosomes and assessment of its anticancer activity against lung cancer. J. Drug Deliv. Sci. Technol. 2024, 93, 105412. [Google Scholar] [CrossRef]
  151. Kanpipit, N.; Mattariganont, S.; Janphuang, P.; Rongsak, J.; Daduang, S.; Chulikhit, Y.; Thapphasaraphong, S. Comparative Study of Lycopene-Loaded Niosomes Prepared by Microfluidic and Thin-Film Hydration Techniques for UVB Protection and Anti-Hyperpigmentation Activity. Int. J. Mol. Sci. 2024, 25, 1717. [Google Scholar] [CrossRef]
  152. He, R.X.; Ye, X.; Li, R.; Chen, W.; Ge, T.; Huang, T.Q.; Nie, X.J.; Chen, H.J.; Peng, D.Y.; Chen, W.D. PEGylated niosomes-mediated drug delivery systems for Paeonol: Preparation, pharmacokinetics studies and synergistic anti-tumor effects with 5-FU. J. Liposome Res. 2017, 27, 161–170. [Google Scholar] [CrossRef]
  153. Khederzadeh, A.; Ebrahimnejad, P.; Seyedabadi, M.; Babaei, A.; Amiri, F.T.; Aslani, N.; Mojarad-Jabali, S.; Mohammadi, H. Synergistic effect of curcumin and Piperine loaded Niosomal nanoparticles on acute pulmonary toxicity induced by Paraquat in mice. Toxicol. Res. 2024, 13, tfae181. [Google Scholar] [CrossRef]
  154. Tyagi, R.; Waheed, A.; Kumar, N.; Ahad, A.; Bin Jardan, Y.A.; Mujeeb, M.; Kumar, A.; Naved, T.; Madan, S. Formulation and Evaluation of Plumbagin-Loaded Niosomes for an Antidiabetic Study: Optimization and In Vitro Evaluation. Pharmaceuticals 2023, 16, 1169. [Google Scholar] [CrossRef] [PubMed]
  155. Lu, B.; Huang, Y.; Chen, Z.; Ye, J.; Xu, H.; Chen, W.; Long, X. Niosomal Nanocarriers for Enhanced Skin Delivery of Quercetin with Functions of Anti-Tyrosinase and Antioxidant. Molecules 2019, 24, 2322. [Google Scholar] [CrossRef]
  156. Gilani, S.J.; Imam, S.S.; Ahmed, A.; Chauhan, S.; Mirza, M.A.; Taleuzzaman, M. Formulation and evaluation of thymoquinone niosomes: Application of developed and validated RP-HPLC method in delivery system. Drug Dev. Ind. Pharm. 2019, 45, 1799–1806. [Google Scholar] [CrossRef]
  157. Barani, M.; Mirzaei, M.; Torkzadeh-Mahani, M.; Adeli-Sardou, M. Evaluation of Carum-loaded Niosomes on Breast Cancer Cells:Physicochemical Properties, In Vitro Cytotoxicity, Flow Cytometric, DNA Fragmentation and Cell Migration Assay. Sci. Rep. 2019, 9, 7139. [Google Scholar] [CrossRef]
  158. Amiri, B.; Ahmadvand, H.; Farhadi, A.; Najmafshar, A.; Chiani, M.; Norouzian, D. Delivery of vinblastine-containing niosomes results in potent in vitro/in vivo cytotoxicity on tumor cells. Drug Dev. Ind. Pharm. 2018, 44, 1371–1376. [Google Scholar] [CrossRef]
  159. Chauhan, M.K.; Sahoo, P.K.; Rawat, A.S.; Singh, A.; Bamrara, A.; Sharma, D. Bilosomes: A Novel Approach to Meet the Challenges in Oral Immunization. Recent Pat. Drug Deliv. Formul. 2015, 9, 201–212. [Google Scholar] [CrossRef]
  160. Can, A.; Tyler, A.I.; Mackie, A.R. Potential use of bile salts in lipid self-assembled systems for the delivery of phytochemicals. Curr. Opin. Colloid Interface Sci. 2021, 56, 101502. [Google Scholar]
  161. Mitrović, D.; Zaklan, D.; Đanić, M.; Stanimirov, B.; Stankov, K.; Al-Salami, H.; Pavlović, N. The Pharmaceutical and Pharmacological Potential Applications of Bilosomes as Nanocarriers for Drug Delivery. Molecules 2025, 30, 1181. [Google Scholar] [CrossRef]
  162. Ahmad, J.; Singhal, M.; Amin, S.; Rizwanullah, M.; Akhter, S.; Kamal, M.A.; Haider, N.; Midoux, P.; Pichon, C. Bile Salt Stabilized Vesicles (Bilosomes): A Novel Nano-Pharmaceutical Design for Oral Delivery of Proteins and Peptides. Curr. Pharm. Des. 2017, 23, 1575–1588. [Google Scholar] [CrossRef]
  163. Matloub, A.A.; Salama, A.H.; Aglan, H.A.; AbouSamra, M.M.; ElSouda, S.S.M.; Ahmed, H.H. Exploiting bilosomes for delivering bioactive polysaccharide isolated from Enteromorpha intestinalis for hacking hepatocellular carcinoma. Drug Dev. Ind. Pharm. 2018, 44, 523–534. [Google Scholar] [CrossRef]
  164. Yang, H.; Liu, Z.; Song, Y.; Hu, C. Hyaluronic acid-functionalized bilosomes for targeted delivery of tripterine to inflamed area with enhancive therapy on arthritis. Drug Deliv. 2019, 26, 820–830. [Google Scholar] [CrossRef] [PubMed]
  165. Waglewska, E.; Pucek-Kaczmarek, A.; Bazylińska, U. Novel Surface-Modified Bilosomes as Functional and Biocompatible Nanocarriers of Hybrid Compounds. Nanomaterials 2020, 10, 2472. [Google Scholar] [CrossRef] [PubMed]
  166. Kaurav, H.; Tripathi, M.; Kaur, S.D.; Bansal, A.; Kapoor, D.N.; Sheth, S. Emerging Trends in Bilosomes as Therapeutic Drug Delivery Systems. Pharmaceutics 2024, 16, 697. [Google Scholar] [CrossRef]
  167. Eid, H.M.; Ali, A.A.; Ali, A.M.A.; Eissa, E.M.; Hassan, R.M.; Abo El-Ela, F.I.; Hassan, A.H. Potential Use of Tailored Citicoline Chitosan-Coated Liposomes for Effective Wound Healing in Diabetic Rat Model. Int. J. Nanomed. 2022, 17, 555–575. [Google Scholar] [CrossRef]
  168. Akrawi, S.H.; Gorain, B.; Nair, A.B.; Choudhury, H.; Pandey, M.; Shah, J.N.; Venugopala, K.N. Development and optimization of naringenin-loaded chitosan-coated nanoemulsion for topical therapy in wound healing. Pharmaceutics 2020, 12, 893. [Google Scholar] [CrossRef]
  169. Elkomy, M.H.; Alruwaili, N.K.; Elmowafy, M.; Shalaby, K.; Zafar, A.; Ahmad, N.; Alsalahat, I.; Ghoneim, M.M.; Eissa, E.M.; Eid, H.M. Surface-Modified Bilosomes Nanogel Bearing a Natural Plant Alkaloid for Safe Management of Rheumatoid Arthritis Inflammation. Pharmaceutics 2022, 14, 563. [Google Scholar] [CrossRef]
  170. Imam, S.S.; Alshehri, S.; Altamimi, M.A.; Almalki, R.K.H.; Hussain, A.; Bukhari, S.I.; Mahdi, W.A.; Qamar, W. Formulation of Chitosan-Coated Apigenin Bilosomes: In Vitro Characterization, Antimicrobial and Cytotoxicity Assessment. Polymers 2022, 14, 921. [Google Scholar] [CrossRef]
  171. Shahbazi, R.; Jafari-Gharabaghlou, D.; Mirjafary, Z.; Saeidian, H.; Zarghami, N. Design and optimization various formulations of PEGylated niosomal nanoparticles loaded with phytochemical agents: Potential anti-cancer effects against human lung cancer cells. Pharmacol. Rep. 2023, 75, 442–455. [Google Scholar] [CrossRef]
  172. Deng, F.; Bae, Y.H. Bile acid transporter-mediated oral drug delivery. J. Control. Release 2020, 327, 100–116. [Google Scholar] [CrossRef]
  173. Zafar, A.; Yasir, M.; Khalid, M.; Amir, M.; Singh, L. Pegylated bilosomes for improvement of oral delivery of Biochanin A: Development to preclinical evaluation. S. Afr. J. Bot. 2023, 162, 633–643. [Google Scholar] [CrossRef]
  174. Wang, L.; Wang, L.; Zhang, Y.; Zhao, Z.; Liu, C.; Li, M.; Liu, J.; Wang, S.; Yang, D.; Luo, F.; et al. LS-HB-Mediated Photodynamic Therapy Inhibits Proliferation and Induces Cell Apoptosis in Melanoma. Mol. Pharm. 2022, 19, 2607–2619. [Google Scholar] [CrossRef] [PubMed]
  175. Waglewska, E.; Kulbacka, J.; Bazylinska, U. Superior Drug Delivery Performance of Multifunctional Bilosomes: Innovative Strategy to Kill Skin Cancer Cells for Nanomedicine Application. Int. J. Nanomed. 2024, 19, 4701–4717. [Google Scholar] [CrossRef]
  176. Alamoudi, A.J.; Badr-Eldin, S.M.; Ahmed, O.A.A.; Fahmy, U.A.; Elbehairi, S.E.I.; Alfaifi, M.Y.; Asfour, H.Z.; Mohamed, G.A.; Ibrahim, S.R.M.; Abdel-Naim, A.B.; et al. Optimized bilosome-based nanoparticles enhance cytotoxic and pro-apoptotic activity of costunolide in LS174T colon cancer cells. Biomed. Pharmacother. 2023, 168, 115757. [Google Scholar] [CrossRef]
  177. Hashem, F.M.; Elkhateeb, D.; Ali, M.M.; Abdel-Rashid, R.S. In-vivo and in-vitro assessment of curcumin loaded bile salt stabilized nanovesicles for oral delivery. DARU J. Pharm. Sci. 2025, 33, 9. [Google Scholar] [CrossRef]
  178. Wang, L.; Huang, X.; Jing, H.; Ma, C.; Wang, H. Bilosomes as effective delivery systems to improve the gastrointestinal stability and bioavailability of epigallocatechin gallate (EGCG). Food Res. Int. 2021, 149, 110631. [Google Scholar] [CrossRef]
  179. Zafar, A.; Alruwaili, N.K.; Imam, S.S.; Alsaidan, O.A.; Yasir, M.; Ghoneim, M.M.; Alshehri, S.; Anwer, M.K.; Almurshedi, A.S.; Alanazi, A.S. Development and evaluation of luteolin loaded pegylated bilosome: Optimization, in vitro characterization, and cytotoxicity study. Drug Deliv. 2021, 28, 2562–2573. [Google Scholar] [CrossRef]
  180. Binsuwaidan, R.; Sultan, A.A.; Negm, W.A.; Attallah, N.G.M.; Alqahtani, M.J.; Hussein, I.A.; Shaldam, M.A.; El-Sherbeni, S.A.; Elekhnawy, E. Bilosomes as Nanoplatform for Oral Delivery and Modulated In Vivo Antimicrobial Activity of Lycopene. Pharmaceuticals 2022, 15, 1043. [Google Scholar] [CrossRef]
  181. Youness, R.A.; Al-Mahallawi, A.M.; Mahmoud, F.H.; Atta, H.; Braoudaki, M.; Fahmy, S.A. Oral Delivery of Psoralidin by Mucoadhesive Surface-Modified Bilosomes Showed Boosted Apoptotic and Necrotic Effects against Breast and Lung Cancer Cells. Polymers 2023, 15, 1464. [Google Scholar] [CrossRef]
  182. Waglewska, E.; Misiaszek, T.; Bazylińska, U. Nanoencapsulation of poorly soluble sea-buckthorn pulp oil in bile salt-origin vesicles: Physicochemical characterization and colloidal stability. Colloids Surf. A Physicochem. Eng. Asp. 2022, 647, 129113. [Google Scholar] [CrossRef]
  183. Zewail, M.; Gaafar, P.M.E.; Youssef, N.; Ali, M.E.; Ragab, M.F.; Kamal, M.F.; Noureldin, M.H.; Abbas, H. Novel Siprulina platensis Bilosomes for Combating UVB Induced Skin Damage. Pharmaceuticals 2022, 16, 36. [Google Scholar] [CrossRef]
  184. Gupta, R.; Kumar, A. Transfersomes: The Ultra-Deformable Carrier System for Non-Invasive Delivery of Drug. Curr. Drug Deliv. 2021, 18, 408–420. [Google Scholar] [CrossRef] [PubMed]
  185. Matharoo, N.; Mohd, H.; Michniak-Kohn, B. Transferosomes as a transdermal drug delivery system: Dermal kinetics and recent developments. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2024, 16, e1918. [Google Scholar] [CrossRef]
  186. Chen, R.P.; Chavda, V.P.; Patel, A.B.; Chen, Z.S. Phytochemical Delivery Through Transferosome (Phytosome): An Advanced Transdermal Drug Delivery for Complementary Medicines. Front. Pharmacol. 2022, 13, 850862. [Google Scholar] [CrossRef]
  187. Opatha, S.A.T.; Titapiwatanakun, V.; Chutoprapat, R. Transfersomes: A Promising Nanoencapsulation Technique for Transdermal Drug Delivery. Pharmaceutics 2020, 12, 855. [Google Scholar] [CrossRef]
  188. Ontong, J.C.; Singh, S.; Siriyong, T.; Voravuthikunchai, S.P. Transferosomes stabilized hydrogel incorporated rhodomyrtone-rich extract from Rhodomyrtus tomentosa leaf fortified with phosphatidylcholine for the management of skin and soft-tissue infections. Biotechnol. Lett. 2024, 46, 127–142. [Google Scholar] [CrossRef]
  189. Wang, W.H.; Tyan, Y.C.; Chen, Z.S.; Lin, C.G.; Yang, M.H.; Yuan, S.S.; Tsai, W.C. Evaluation of the antioxidant activity and antiproliferative effect of the jaboticaba (Myrciaria cauliflora) seed extracts in oral carcinoma cells. BioMed Res. Int. 2014, 2014, 185946. [Google Scholar] [CrossRef]
  190. Castangia, I.; Manca, M.L.; Allaw, M.; Hellström, J.; Granato, D.; Manconi, M. Jabuticaba (Myrciaria jaboticaba) Peel as a Sustainable Source of Anthocyanins and Ellagitannins Delivered by Phospholipid Vesicles for Alleviating Oxidative Stress in Human Keratinocytes. Molecules 2021, 26, 6697. [Google Scholar] [CrossRef]
  191. Opatha, S.A.T.; Chutoprapat, R.; Khankaew, P.; Titapiwatanakun, V.; Ruksiriwanich, W.; Boonpisuttinant, K. Asiatic acid-entrapped transfersomes for the treatment of hypertrophic scars: In vitro appraisal, bioactivity evaluation, and clinical study. Int. J. Pharm. 2024, 651, 123738. [Google Scholar] [CrossRef]
  192. Retnaningtyas, E.; Susatia, B.; Khotimah, H.; Rudijanto, A.; Ali Ahmed Abousouh, A.; Setiawan, A. Centella asiatica transfersomes and Bergamot essential oil nanoemulsion combined in gel exhibited anti-photoaging effects on UVB-radiated BALB/c mice. J. King Saud Univ. Sci. 2024, 36, 103207. [Google Scholar] [CrossRef]
  193. Tuntiyasawasdikul, S.; Sripanidkulchai, B. Curcuma comosa loaded transfersomal gel for transdermal application: Formulation, in vitro and in vivo evaluation. Drug Dev. Ind. Pharm. 2021, 47, 1824–1834. [Google Scholar] [CrossRef]
  194. Nangare, S.; Bhatane, D.; Mali, R.; Shitole, M. Development of a Novel Freeze-dried Mulberry Leaf Extract-based Transfersome Gel. Turk. J. Pharm. Sci. 2021, 18, 44–55. [Google Scholar] [CrossRef] [PubMed]
  195. Sklenarova, R.; Allaw, M.; Perra, M.; Castangia, I.; Frankova, J.; Luis Pedraz, J.; Letizia Manca, M.; Manconi, M. Co-delivering of oleuropein and lentisk oil in phospholipid vesicles as an effective approach to modulate oxidative stress, cytokine secretion and promote skin regeneration. Eur. J. Pharm. Biopharm. 2023, 185, 126–136. [Google Scholar] [CrossRef] [PubMed]
  196. Chamsai, B.; Rangsimawong, W.; Suriyaamporn, P.; Opanasopit, P.; Samprasit, W. Development of radish extract-loaded transfersomes blended sunscreen formulation for tyrosinase melanin and photoprotective sunscreening effect. J. Drug Deliv. Sci. Technol. 2024, 101, 106230. [Google Scholar] [CrossRef]
  197. Sahu, N.; Alam, P.; Ali, A.; Kumar, N.; Tyagi, R.; Madan, S.; Walia, R.; Saxena, S. Optimization, In Vitro and Ex Vivo Assessment of Nanotransferosome Gels Infused with a Methanolic Extract of Solanum xanthocarpum for the Topical Treatment of Psoriasis. Gels 2024, 10, 119. [Google Scholar] [CrossRef]
  198. Abd-Allah, H.; Ragaie, M.H.; Elmowafy, E. Unraveling the pharmaceutical and clinical relevance of the influence of syringic acid loaded linoleic acid transferosomes on acne. Int. J. Pharm. 2023, 639, 122940. [Google Scholar] [CrossRef]
  199. Andleeb, M.; Shoaib Khan, H.M.; Daniyal, M. Development, Characterization and Stability Evaluation of Topical Gel Loaded With Ethosomes Containing Achillea millefolium L. Extract. Front. Pharmacol. 2021, 12, 603227. [Google Scholar] [CrossRef]
  200. Javed, N.; Ijaz, S.; Akhtar, N.; Khan, H.M.S. Nanostructured Ethosomal Gel Loaded with Arctostaphylosuva-Ursi Extract; In-Vitro/In-Vivo Evaluation as a Cosmeceutical Product for Skin Rejuvenation. Curr. Drug Deliv. 2022, 19, 706–720. [Google Scholar] [CrossRef]
  201. Lin, H.; Lin, L.; Choi, Y.; Michniak-Kohn, B. Development and in-vitro evaluation of co-loaded berberine chloride and evodiamine ethosomes for treatment of melanoma. Int. J. Pharm. 2020, 581, 119278. [Google Scholar] [CrossRef]
  202. Khan, P.; Akhtar, N. Phytochemical investigations and development of ethosomal gel with Brassica oleraceae L. (Brassicaceae) extract: An innovative nano approach towards cosmetic and pharmaceutical industry. Ind. Crop. Prod. 2022, 183, 114905. [Google Scholar] [CrossRef]
  203. Hallan, S.S.; Sguizzato, M.; Mariani, P.; Cortesi, R.; Huang, N.; Simelière, F.; Marchetti, N.; Drechsler, M.; Ruzgas, T.; Esposito, E. Design and Characterization of Ethosomes for Transdermal Delivery of Caffeic Acid. Pharmaceutics 2020, 12, 740. [Google Scholar] [CrossRef]
  204. Sasindran, S.; Easwaran, M.; Shyamala, G.; Karuppaiah, A.; Siram, K.; Veintramuthu, S. Phytochemical screening and cytotoxicity evaluation of crude extracts: Toxicity comparison of crude extracts and its ethosomal formulations. J. Cosmet. Dermatol. 2020, 19, 1794–1803. [Google Scholar] [CrossRef] [PubMed]
  205. Mota, A.H.; Prazeres, I.; Mestre, H.; Bento-Silva, A.; Rodrigues, M.J.; Duarte, N.; Serra, A.T.; Bronze, M.R.; Rijo, P.; Gaspar, M.M.; et al. A Newfangled Collagenase Inhibitor Topical Formulation Based on Ethosomes with Sambucus nigra L. Extract. Pharmaceuticals 2021, 14, 467. [Google Scholar] [CrossRef] [PubMed]
  206. Qin, D.; Cui, Y.; Zheng, M.; Yang, Z.; Wang, X. Preparation of Ethosome Gel with Total Flavonoids from Vernonia anthelmintica (L.) Willd. for the Treatment of Vitiligo. Gels 2025, 11, 73. [Google Scholar] [CrossRef]
  207. Adnan, M.; Afzal, O.; Altamimi, A.S.A.; Alamri, M.A.; Haider, T.; Faheem Haider, M. Development and optimization of transethosomal gel of apigenin for topical delivery: In-vitro, ex-vivo and cell line assessment. Int. J. Pharm. 2023, 631, 122506. [Google Scholar] [CrossRef]
  208. Vera Pérez, J.; Martínez Cortés, D.M.; Gómez y Gómez, Y. Potential use of transethosomes as a transdermal delivery system for metabolites from Chenopodium murale. Mater. Today Commun. 2022, 30, 103165. [Google Scholar] [CrossRef]
  209. Abdulbaqi, I.M.; Darwis, Y.; Assi, R.A.; Khan, N.A.K. Transethosomal gels as carriers for the transdermal delivery of colchicine: Statistical optimization, characterization, and ex vivo evaluation. Drug Des. Dev. Ther. 2018, 12, 795–813. [Google Scholar] [CrossRef]
  210. Moolakkadath, T.; Aqil, M.; Ahad, A.; Imam, S.S.; Iqbal, B.; Sultana, Y.; Mujeeb, M.; Iqbal, Z. Development of transethosomes formulation for dermal fisetin delivery: Box-Behnken design, optimization, in vitro skin penetration, vesicles-skin interaction and dermatokinetic studies. Artif. Cells Nanomed. Biotechnol. 2018, 46, 755–765. [Google Scholar] [CrossRef]
  211. Hassan, A.S.; Hofni, A.; Abourehab, M.A.S.; Abdel-Rahman, I.A.M. Ginger Extract-Loaded Transethosomes for Effective Transdermal Permeation and Anti-Inflammation in Rat Model. Int. J. Nanomed. 2023, 18, 1259–1280. [Google Scholar] [CrossRef]
  212. Yasmeen; Khan, M.A.; Iqbal, Z.; Aqil, M. Carbopol 934-based transethosomal gel of Glycyrrhizic acid for the management of skin cancer. J. Drug Deliv. Sci. Technol. 2024, 97, 105825. [Google Scholar] [CrossRef]
  213. Aodah, A.H.; Hashmi, S.; Akhtar, N.; Ullah, Z.; Zafar, A.; Zaki, R.M.; Khan, S.; Ansari, M.J.; Jawaid, T.; Alam, A.; et al. Formulation Development, Optimization by Box-Behnken Design, and In Vitro and Ex Vivo Characterization of Hexatriacontane-Loaded Transethosomal Gel for Antimicrobial Treatment for Skin Infections. Gels 2023, 9, 322. [Google Scholar] [CrossRef]
  214. Bin Jardan, Y.A.; Ahad, A.; Raish, M.; Al-Jenoobi, F.I. Preparation and Characterization of Transethosome Formulation for the Enhanced Delivery of Sinapic Acid. Pharmaceutics 2023, 15, 2391. [Google Scholar] [CrossRef] [PubMed]
  215. Paiva-Santos, A.C.; Silva, A.L.; Guerra, C.; Peixoto, D.; Pereira-Silva, M.; Zeinali, M.; Mascarenhas-Melo, F.; Castro, R.; Veiga, F. Ethosomes as Nanocarriers for the Development of Skin Delivery Formulations. Pharm. Res. 2021, 38, 947–970. [Google Scholar] [CrossRef]
  216. Alfehaid, F.S.; Nair, A.B.; Shah, H.; Aldhubiab, B.; Shah, J.; Mewada, V.; Jacob, S.; Attimarad, M. Enhanced transdermal delivery of apremilast loaded ethosomes: Optimization, characterization and in vivo evaluation. J. Drug Deliv. Sci. Technol. 2024, 91, 105211. [Google Scholar]
  217. Bodratti, A.M.; Alexandridis, P. Formulation of Poloxamers for Drug Delivery. J. Funct. Biomater. 2018, 9, 11. [Google Scholar] [CrossRef]
  218. Sguizzato, M.; Ferrara, F.; Mariani, P.; Pepe, A.; Cortesi, R.; Huang, N.; Simelière, F.; Boldrini, P.; Baldisserotto, A.; Valacchi, G.; et al. “Plurethosome” as Vesicular System for Cutaneous Administration of Mangiferin: Formulative Study and 3D Skin Tissue Evaluation. Pharmaceutics 2021, 13, 1124. [Google Scholar] [CrossRef]
  219. Touitou, E.; Natsheh, H. The Evolution of Emerging Nanovesicle Technologies for Enhanced Delivery of Molecules into and across the Skin. Pharmaceutics 2024, 16, 267. [Google Scholar] [CrossRef]
  220. Bajaj, K.J.; Parab, B.S.; Shidhaye, S.S. Nano-transethosomes: A novel tool for drug delivery through skin. Indian J. Pharm. Educ. Res. 2021, 55, S1–S10. [Google Scholar]
  221. Munir, M.; Zaman, M.; Waqar, M.A.; Hameed, H.; Riaz, T. A comprehensive review on transethosomes as a novel vesicular approach for drug delivery through transdermal route. J. Liposome Res. 2024, 34, 203–218. [Google Scholar] [CrossRef]
  222. Song, C.K.; Balakrishnan, P.; Shim, C.K.; Chung, S.J.; Chong, S.; Kim, D.D. A novel vesicular carrier, transethosome, for enhanced skin delivery of voriconazole: Characterization and in vitro/in vivo evaluation. Colloids Surf. B Biointerfaces 2012, 92, 299–304. [Google Scholar] [CrossRef]
  223. Malang, S.D.; Shambhavi; Sahu, A.N. Transethosomal gel for enhancing transdermal delivery of natural therapeutics. Nanomedicine 2024, 19, 1801–1819. [Google Scholar] [CrossRef]
  224. Nayak, B.S.; Mohanty, B.; Mishra, B.; Roy, H.; Nandi, S. Transethosomes: Cutting edge approach for drug permeation enhancement in transdermal drug delivery system. Chem. Biol. Drug Des. 2023, 102, 653–667. [Google Scholar] [CrossRef] [PubMed]
  225. Mita, S.R.; Abdassah, M.; Supratman, U.; Shiono, Y.; Rahayu, D.; Sopyan, I.; Wilar, G. Nanoparticulate System for the Transdermal Delivery of Catechin as an Antihypercholesterol: In Vitro and In Vivo Evaluations. Pharmaceuticals 2022, 15, 1142. [Google Scholar] [CrossRef] [PubMed]
  226. Kim, J.-E.; Oh, G.-H.; Jang, G.-H.; Kim, Y.-M.; Park, Y.-J. Transformer-ethosomes with palmitoyl pentapeptide for improved transdermal delivery. J. Drug Deliv. Sci. Technol. 2019, 52, 460–467. [Google Scholar]
  227. Sguizzato, M.; Ferrara, F.; Hallan, S.S.; Baldisserotto, A.; Drechsler, M.; Malatesta, M.; Costanzo, M.; Cortesi, R.; Puglia, C.; Valacchi, G.; et al. Ethosomes and Transethosomes for Mangiferin Transdermal Delivery. Antioxidants 2021, 10, 768. [Google Scholar] [CrossRef]
  228. Jahan, S.; Ali, A.; Sultana, N.; Qizilbash, F.F.; Ali, H.; Aqil, M.; Mujeeb, M.; Ali, A. An overview of phospholipid enriched-edge activator-based vesicle nanocarriers: New paradigms to treat skin cancer. J. Drug Target. 2025, 33, 17–41. [Google Scholar] [CrossRef]
  229. Abourehab, M.A.S.; Ansari, M.J.; Singh, A.; Hassan, A.; Abdelgawad, M.A.; Shrivastav, P.; Abualsoud, B.M.; Amaral, L.S.; Pramanik, S. Cubosomes as an emerging platform for drug delivery: A review of the state of the art. J. Mater. Chem. B 2022, 10, 2781–2819. [Google Scholar] [CrossRef]
  230. Sivadasan, D.; Sultan, M.H.; Alqahtani, S.S.; Javed, S. Cubosomes in Drug Delivery—A Comprehensive Review on Its Structural Components, Preparation Techniques and Therapeutic Applications. Biomedicines 2023, 11, 1114. [Google Scholar] [CrossRef]
  231. Al-Mahallawi, A.M.; Abdelbary, A.A.; El-Zahaby, S.A. Norfloxacin loaded nano-cubosomes for enhanced management of otitis externa: In vitro and in vivo evaluation. Int. J. Pharm. 2021, 600, 120490. [Google Scholar] [CrossRef]
  232. Gorantla, S.; Saha, R.N.; Singhvi, G. Exploring the affluent potential of glyceryl mono oleate–myristol liquid crystal nanoparticles mediated localized topical delivery of Tofacitinib: Study of systematic QbD, skin deposition and dermal pharmacokinetics assessment. J. Mol. Liq. 2022, 346, 117053. [Google Scholar] [CrossRef]
  233. Nath, A.G.; Dubey, P.; Kumar, A.; Vaiphei, K.K.; Rosenholm, J.M.; Bansal, K.K.; Gulbake, A. Recent Advances in the Use of Cubosomes as Drug Carriers with Special Emphasis on Topical Applications. J. Lipids 2024, 2024, 2683466. [Google Scholar] [CrossRef]
  234. Barriga, H.M.G.; Holme, M.N.; Stevens, M.M. Cubosomes: The Next Generation of Smart Lipid Nanoparticles? Angew. Chem. Int. Ed. 2019, 58, 2958–2978. [Google Scholar] [CrossRef]
  235. Faisal, M.M.; Gomaa, E.; Ibrahim, A.E.; El Deeb, S.; Al-Harrasi, A.; Ibrahim, T.M. Verapamil-Loaded Cubosomes for Enhancing Intranasal Drug Delivery: Development, Characterization, Ex Vivo Permeation, and Brain Biodistribution Studies. AAPS PharmSciTech 2024, 25, 95. [Google Scholar] [CrossRef] [PubMed]
  236. Peng, X.; Zhou, Y.; Han, K.; Qin, L.; Dian, L.; Li, G.; Pan, X.; Wu, C. Characterization of cubosomes as a targeted and sustained transdermal delivery system for capsaicin. Drug Des. Dev. Ther. 2015, 9, 4209–4218. [Google Scholar] [CrossRef]
  237. Khattab, A.; Ismail, S.; Abd-Elrazek, A.M. Mucoadhesive Chitosan Composite Sponge as a Carrier for β-Sitosterol Cubosomes for Thermal Burn Treatment. AAPS PharmSciTech 2024, 25, 148. [Google Scholar] [CrossRef]
  238. Alyami, M.H.; Hamdan, D.I.; Khalil, H.M.A.; Orabi, M.A.A.; Aborehab, N.M.; Osama, N.; Abdelhafez, M.M.; Al-Mahallawi, A.M.; Alyami, H.S. Preparation and in vivo evaluation of nano sized cubosomal dispersion loaded with Ruta graveolens extracts as a novel approach to reduce asthma-mediated lung inflammation. Saudi Pharm. J. 2024, 32, 101968. [Google Scholar] [CrossRef]
  239. Kumari, S.; Goyal, A.; Garg, M. Box-Behnken Design (BBD) Based Optimization of Beta-Carotene Loaded Cubosomes for Anti-Oxidant Activity Using DPPH Assay. BioNanoScience 2023, 13, 466–480. [Google Scholar] [CrossRef]
  240. Li, Y.; Angelova, A.; Hu, F.; Garamus, V.M.; Peng, C.; Li, N.; Liu, J.; Liu, D.; Zou, A. pH Responsiveness of Hexosomes and Cubosomes for Combined Delivery of Brucea javanica Oil and Doxorubicin. Langmuir ACS J. Surf. Colloids 2019, 35, 14532–14542. [Google Scholar] [CrossRef]
  241. Rakotoarisoa, M.; Angelov, B.; Garamus, V.M.; Angelova, A. Curcumin- and Fish Oil-Loaded Spongosome and Cubosome Nanoparticles with Neuroprotective Potential against H2O2-Induced Oxidative Stress in Differentiated Human SH-SY5Y Cells. ACS Omega 2019, 4, 3061–3073. [Google Scholar] [CrossRef]
  242. Singh, S.; Singh, G.; Attri, S.; Kaur, P.; Rashid, F.; Bedi, N.; Haque, S.; Janahi, E.M.; Arora, S. Development and optimization of nanoparticles loaded with erucin, a dietary isothiocyanate isolated from Eruca sativa: Antioxidant and antiproliferative activities in ehrlich-ascites carcinoma cell line. Front. Pharmacol. 2022, 13, 1080977. [Google Scholar] [CrossRef]
  243. Zhu, C.; Duan, W.; Jing, H.; Long, J.; Huang, Y.; Huang, D.; Wu, C. Improving the stability and transdermal permeability of phycocyanin loaded cubosomes. Front. Nanotechnol. 2024, 6, 1359219. [Google Scholar]
  244. Raafat, K.M.; Abdelwahab, I.A.; El-Zahaby, S.A. Nano-cubosomes of the phyto-active principle in Withania somnifera: LC-MS-NMR, anti-microbial, and insights of the anti-neuropathic and anti-inflammatory mechanism. Fitoterapia 2024, 178, 106196. [Google Scholar] [CrossRef] [PubMed]
  245. Salaheldin Abdelhamid Ibrahim, S.; Bassam, S.M.; El-Hawary, S.; Sheta, E.; Masoud, I.M.; El-Zahaby, S.A.; Al-Mahallawi, A.M.; Hammad, G.O. The gastroprotective effect of Yucca filamentosa standardized crude leaves extract versus its nano-cubosomal formulation in ethanol-induced gastric injury. Int. Immunopharmacol. 2024, 137, 112440. [Google Scholar] [CrossRef]
  246. Has, C.; Sunthar, P. A comprehensive review on recent preparation techniques of liposomes. J. Liposome Res. 2020, 30, 336–365. [Google Scholar] [CrossRef]
  247. Patil, Y.P.; Jadhav, S. Novel methods for liposome preparation. Chem. Phys. Lipids 2014, 177, 8–18. [Google Scholar] [CrossRef]
  248. Dragićević, N. Invasomes as Drug Nanocarriers for Innovative Pharmaceutical Dosage Forms; CRC Press: Boca Raton, FL, USA, 2021. [Google Scholar]
  249. Roostaee, M.; Derakhshani, A.; Mirhosseini, H.; Banaee Mofakham, E.; Fathi-Karkan, S.; Mirinejad, S.; Sargazi, S.; Barani, M. Composition, preparation methods, and applications of nanoniosomes as codelivery systems: A review of emerging therapies with emphasis on cancer. Nanoscale 2024, 16, 2713–2746. [Google Scholar] [CrossRef]
  250. Nayak, D.; Rathnanand, M.; Tippavajhala, V.K. Unlocking the Potential of Bilosomes and Modified Bilosomes: A Comprehensive Journey into Advanced Drug Delivery Trends. AAPS PharmSciTech 2023, 24, 238. [Google Scholar] [CrossRef]
  251. Fernández-García, R.; Lalatsa, A.; Statts, L.; Bolás-Fernández, F.; Ballesteros, M.P.; Serrano, D.R. Transferosomes as nanocarriers for drugs across the skin: Quality by design from lab to industrial scale. Int. J. Pharm. 2020, 573, 118817. [Google Scholar] [CrossRef]
  252. Hameed, H.; Faheem, S.; Khan, M.A.; Hameed, A.; Ereej, N.; Ihsan, H. Ethosomes: A potential nanovesicular carrier to enhancing the drug delivery against skin barriers. J. Microencapsul. 2024, 41, 204–225. [Google Scholar] [CrossRef]
  253. Ascenso, A.; Raposo, S.; Batista, C.; Cardoso, P.; Mendes, T.; Praça, F.G.; Bentley, M.V.; Simões, S. Development, characterization, and skin delivery studies of related ultradeformable vesicles: Transfersomes, ethosomes, and transethosomes. Int. J. Nanomed. 2015, 10, 5837–5851. [Google Scholar] [CrossRef]
  254. Yang, X.; Jiang, Q.; Du, P.; Zhao, J.; Zhang, T. Preparation and characterization of solidified oleanolic acid–phospholipid complex aiming to improve the dissolution of oleanolic acid. Asian J. Pharm. Sci. 2016, 11, 241–247. [Google Scholar] [CrossRef]
  255. Yu, F.; Li, Y.; Chen, Q.; He, Y.; Wang, H.; Yang, L.; Guo, S.; Meng, Z.; Cui, J.; Xue, M.; et al. Monodisperse microparticles loaded with the self-assembled berberine-phospholipid complex-based phytosomes for improving oral bioavailability and enhancing hypoglycemic efficiency. Eur. J. Pharm. Biopharm. 2016, 103, 136–148. [Google Scholar] [CrossRef]
  256. Andishmand, H.; Yousefi, M.; Jafari, N.; Azadmard-Damirchi, S.; Homayouni-Rad, A.; Torbati, M.; Hamishehkar, H. Designing and fabrication of colloidal nano-phytosomes with gamma-oryzanol and phosphatidylcholine for encapsulation and delivery of polyphenol-rich extract from pomegranate peel. Int. J. Biol. Macromol. 2024, 256, 128501. [Google Scholar] [CrossRef]
  257. Peanparkdee, M.; Yooying, R. Enhancement of solubility, thermal stability and bioaccessibility of vitexin using phosphatidylcholine-based phytosome. NFS J. 2023, 31, 28–38. [Google Scholar] [CrossRef]
  258. Demir, B.; Barlas, F.; Guler, E.; Gumus, P.; Can, M.; Yavuz, M.; Coskunol, H.; Timur, S. Gold nanoparticle loaded phytosomal systems: Synthesis, characterization and in vitro investigations. RSC Adv. 2014, 4, 34687–34695. [Google Scholar]
  259. Chi, C.; Zhang, C.; Liu, Y.; Nie, H.; Zhou, J.; Ding, Y. Phytosome-nanosuspensions for silybin-phospholipid complex with increased bioavailability and hepatoprotection efficacy. Eur. J. Pharm. Sci. 2020, 144, 105212. [Google Scholar] [CrossRef]
  260. Rani, A.; Kumar, S.; Khar, R.K. Murraya koenigii extract loaded phytosomes prepared using antisolvent precipitation technique for improved antidiabetic and hypolidemic activity. Indian J. Pharm. Educ. Res. 2022, 56, s326–s338. [Google Scholar] [CrossRef]
  261. Damle, M.; Mallya, R. Development and Evaluation of a Novel Delivery System Containing Phytophospholipid Complex for Skin Aging. AAPS PharmSciTech 2016, 17, 607–617. [Google Scholar] [CrossRef]
  262. Karole, S.; Gupta, G. Preparation and evaluation of phytosomes containing ethanolic extract of leaves of Bombax ceiba for hepatoprotective activity. Pharma Innov. J. 2019, 8, 22–26. [Google Scholar]
  263. Habbu, P.; Madagundi, S.; Shastry, R.; Vanakudri, R.; Kulkarni, V. Preparation and evaluation of antidiabetic activity of Allium cepa-phospholipid complex (phytosome) in streptozotocin induced diabetic rats. RGUHS J. Pharm. Sci. 2015, 5, 132–141. [Google Scholar]
  264. Dewi, M.K.; Muhaimin, M.; Joni, I.M.; Hermanto, F.; Chaerunisaa, A.Y. Fabrication of Phytosome with Enhanced Activity of Sonneratia alba: Formulation Modeling and in vivo Antimalarial Study. Int. J. Nanomed. 2024, 19, 9411–9435. [Google Scholar] [CrossRef]
  265. Freag, M.S.; Elnaggar, Y.S.; Abdallah, O.Y. Lyophilized phytosomal nanocarriers as platforms for enhanced diosmin delivery: Optimization and ex vivo permeation. Int. J. Nanomed. 2013, 8, 2385–2397. [Google Scholar] [CrossRef]
  266. Human, C.; Aucamp, M.; de Beer, D.; van der Rijst, M.; Joubert, E. Food-grade phytosome vesicles for nanoencapsulation of labile C-glucosylated xanthones and dihydrochalcones present in a plant extract matrix—Effect of process conditions and stability assessment. Food Sci. Nutr. 2023, 11, 8093–8111. [Google Scholar] [CrossRef]
  267. Telange, D.R.; Patil, A.T.; Pethe, A.M.; Tatode, A.A.; Anand, S.; Dave, V.S. Kaempferol-phospholipid complex: Formulation, and evaluation of improved solubility, in vivo bioavailability, and antioxidant potential of kaempferol. J. Excip. Food Chem. 2016, 7, 89–116. [Google Scholar]
  268. Li, Y.; Yang, D.J.; Chen, S.L.; Chen, S.B.; Chan, A.S. Process parameters and morphology in puerarin, phospholipids and their complex microparticles generation by supercritical antisolvent precipitation. Int. J. Pharm. 2008, 359, 35–45. [Google Scholar] [CrossRef]
  269. Goel, H.; Saini, K.; Razdan, K.; Khurana, R.K.; Elkordy, A.A.; Singh, K.K. Chapter 3—In vitro physicochemical characterization of nanocarriers: A road to optimization. In Nanoparticle Therapeutics: Production Technologies, Types of Nanoparticles, and Regulatory Aspects; Kesharwani, P., Singh, K.K., Eds.; Academic Press: Cambridge, MA, USA, 2022; pp. 133–179. [Google Scholar] [CrossRef]
  270. Gaikwad, V.L.; Choudhari, P.B.; Bhatia, N.M.; Bhatia, M.S. Chapter 2—Characterization of pharmaceutical nanocarriers: In vitro and in vivo studies. In Nanomaterials for Drug Delivery and Therapy; Grumezescu, A.M., Ed.; William Andrew Publishing: Norwich, NY, USA, 2019; pp. 33–58. [Google Scholar] [CrossRef]
  271. Zuhrotun, A.; Oktaviani, D.J.; Hasanah, A.N. Biosynthesis of Gold and Silver Nanoparticles Using Phytochemical Compounds. Molecules 2023, 28, 3240. [Google Scholar] [CrossRef]
  272. AlMadalli, H.J.; Abdul Rasool, B.K.; Shehab, N.G.; Sala, F.D.; Borzacchiello, A. Pomegranate extract-loaded sphingosomes for the treatment of cancer: Phytochemical investigations, formulation, and antitumor activity evaluation. PLoS ONE 2024, 19, e0293115. [Google Scholar] [CrossRef]
  273. Maurizi, L.; Lasalvia, A.; Fabiano, M.G.; D’Intino, E.; Del Cioppo, F.; Fraschetti, C.; Filippi, A.; Ammendolia, M.G.; Conte, A.L.; Forte, J.; et al. Lentisk (Pistacia lentiscus) Oil Nanoemulsions Loaded with Levofloxacin: Phytochemical Profiles and Antibiofilm Activity against Staphylococcus spp. Pharmaceutics 2024, 16, 927. [Google Scholar] [CrossRef]
  274. Bennion, B.J.; Be, N.A.; McNerney, M.W.; Lao, V.; Carlson, E.M.; Valdez, C.A.; Malfatti, M.A.; Enright, H.A.; Nguyen, T.H.; Lightstone, F.C.; et al. Predicting a Drug’s Membrane Permeability: A Computational Model Validated with in Vitro Permeability Assay Data. J. Phys. Chem. B 2017, 121, 5228–5237. [Google Scholar] [CrossRef]
  275. Bansal, G.; Suthar, N.; Kaur, J.; Jain, A. Stability Testing of Herbal Drugs: Challenges, Regulatory Compliance and Perspectives. Phytother. Res. 2016, 30, 1046–1058. [Google Scholar] [CrossRef]
  276. Isleroglu, H.; Turker, I. Ultrasonic-assisted extraction and thermal stability of phytochemicals from fenugreek leaves. J. Appl. Res. Med. Aromat. Plants 2022, 30, 100390. [Google Scholar]
  277. İnanç Horuz, T.; Maskan, M. Effect of the phytochemicals curcumin, cinnamaldehyde, thymol and carvacrol on the oxidative stability of corn and palm oils at frying temperatures. J. Food Sci. Technol. 2015, 52, 8041–8049. [Google Scholar] [CrossRef]
  278. Baba, W.N.; Abdelrahman, R.; Maqsood, S. Production and utilization of non-covalent dairy-based proteins complexed with date palm leave polyphenols for improving curcumin stability. Food Biosci. 2023, 53, 102690. [Google Scholar] [CrossRef]
  279. Thomas Pannakal, S.; Eilstein, J.; Prasad, A.; Ekhar, P.; Shetty, S.; Peng, Z.; Bordier, E.; Boudah, S.; Paillat, L.; Marrot, L.; et al. Comprehensive characterization of naturally occurring antioxidants from the twigs of mulberry (Morus alba) using on-line high-performance liquid chromatography coupled with chemical detection and high-resolution mass spectrometry. Phytochem. Anal. 2022, 33, 105–114. [Google Scholar] [CrossRef]
  280. Tabassum, S.; Ahmad, S.; Rehman Khan, K.U.; Tabassum, F.; Khursheed, A.; Zaman, Q.U.; Bukhari, N.A.; Alfagham, A.; Hatamleh, A.A.; Chen, Y. Phytochemical Profiling, Antioxidant, Anti-Inflammatory, Thrombolytic, Hemolytic Activity In Vitro and In Silico Potential of Portulacaria afra. Molecules 2022, 27, 2377. [Google Scholar] [CrossRef]
  281. Rathaur, P.; Johar, K., Sr. Metabolism and Pharmacokinetics of Phytochemicals in the Human Body. Curr. Drug Metab. 2019, 20, 1085–1102. [Google Scholar] [CrossRef]
  282. Ghalbi Ahangari, M.; Farimani, M.M.; Erfani, M.; Goudarzi, M. Technetium-99m radiolabeling through conjugation with l,l-ethylene dicysteine chelator of a trimethoxylated flavone and its bioevaluation in rat with induced C6 glioma tumor as a new cancer diagnostic agent. Radiochim. Acta 2024, 112, 327–337. [Google Scholar] [CrossRef]
  283. Sousa Carvalho, G.F.; Marques, L.K.; Sousa, H.G.; Silva, L.R.; Leão Ferreira, D.C.; Pires de Moura do Amaral, F.; Martins Maia Filho, A.L.; Figueredo-Silva, J.; Alves, W.D.S.; Oliveira, M.; et al. Phytochemical study, molecular docking, genotoxicity and therapeutic efficacy of the aqueous extract of the stem bark of Ximenia americana L. in the treatment of experimental COPD in rats. J. Ethnopharmacol. 2020, 247, 112259. [Google Scholar] [CrossRef]
  284. Aurori, M.; Niculae, M.; Hanganu, D.; Pall, E.; Cenariu, M.; Vodnar, D.C.; Bunea, A.; Fiţ, N.; Andrei, S. Phytochemical Profile, Antioxidant, Antimicrobial and Cytoprotective Effects of Cornelian Cherry (Cornus mas L.) Fruit Extracts. Pharmaceuticals 2023, 16, 420. [Google Scholar] [CrossRef]
  285. Bacchetti, T.; Campagna, R.; Sartini, D.; Cecati, M.; Morresi, C.; Bellachioma, L.; Martinelli, E.; Rocchetti, G.; Lucini, L.; Ferretti, G.; et al. C. spinosa L. subsp. rupestris Phytochemical Profile and Effect on Oxidative Stress in Normal and Cancer Cells. Molecules 2022, 27, 6488. [Google Scholar] [CrossRef]
  286. Gul, B.; Anwar, R.; Saleem, M.; Noor, A.; Ullah, M.I. Cassia absus-mediated upregulation of IL-4, IL-10 and downregulation of IL-1β, IL-6, TNF-α, NF-κB, IFN-γ in CFA-induced arthritis model. Inflammopharmacology 2023, 31, 1241–1256. [Google Scholar] [CrossRef]
  287. Jantan, I.; Arshad, L.; Septama, A.W.; Haque, M.A.; Mohamed-Hussein, Z.A.; Govender, N.T. Antiviral effects of phytochemicals against severe acute respiratory syndrome coronavirus 2 and their mechanisms of action: A review. Phytother. Res. 2023, 37, 1036–1056. [Google Scholar] [CrossRef]
  288. Ndezo Bisso, B.; Njikang Epie Nkwelle, R.; Tchuenguem Tchuenteu, R.; Dzoyem, J.P. Phytochemical Screening, Antioxidant, and Antimicrobial Activities of Seven Underinvestigated Medicinal Plants against Microbial Pathogens. Adv. Pharmacol. Pharm. Sci. 2022, 2022, 1998808. [Google Scholar] [CrossRef]
  289. Noushida, N.; Nayak, R.P.; Sultana, R.; Salian, T.R.; Alobid, S.; Almadani, M.E.; Ahmad, F.; Gilkaramenthi, R.; Asdaq, S.M.B.; Almoteer, A.I. Cardioprotective effects of Callicarpa tomentosa leaf extract in Wistar albino rats against isoproterenol-induced myocardial necrosis: Phytochemical analysis and in vitro antioxidant study. J. King Saud Univ.-Sci. 2024, 36, 103100. [Google Scholar]
  290. da Silva, G.G.; Pimenta, L.P.S.; Melo, J.O.F.; Mendonça, H.O.P.; Augusti, R.; Takahashi, J.A. Phytochemicals of Avocado Residues as Potential Acetylcholinesterase Inhibitors, Antioxidants, and Neuroprotective Agents. Molecules 2022, 27, 1892. [Google Scholar] [CrossRef]
  291. Yolin Angel, P.; Jeyakumar, P.; Jasmin Suriya, A.R.; Sheena, A.; Karuppiah, P.; Periyasami, G.; Stalin, A.; Murugan, K. Topical antifungal keratitis therapeutic potential of Clitoria ternatea Linn. flower extract: Phytochemical profiling, in silico modelling, and in vitro biological activity assessment. Front. Microbiol. 2024, 15, 1343988. [Google Scholar] [CrossRef]
  292. Rani, A.; Uzair, M.; Ali, S.; Qamar, M.; Ahmad, N.; Abbas, M.W.; Esatbeyoglu, T. Dryopteris juxtapostia Root and Shoot: Determination of Phytochemicals; Antioxidant, Anti-Inflammatory, and Hepatoprotective Effects; and Toxicity Assessment. Antioxidants 2022, 11, 1670. [Google Scholar] [CrossRef]
  293. El Faqer, O.; Bendiar, S.; Rais, S.; Elkoraichi, I.; Dakir, M.; Elouaddari, A.; El Amrani, A.; Oudghiri, M.; Mtairag, E.M. Phytochemical characterization and immunomodulatory effects of aqueous, ethanolic extracts and essential oil of Syzygium aromaticum L. on human neutrophils. Sci. Afr. 2022, 18, e01395. [Google Scholar]
  294. Ullah, M.; Mehmood, S.; Khan, R.A.; Ali, M.; Fozia, F.; Waqas, M.; Ullah, R.; Alotaibi, A.; Sultan, M.A. Assessment of antidiabetic potential and phytochemical profiling of Viscum album, a traditional antidiabetic plant. J. Food Qual. 2022, 2022, 5691379. [Google Scholar]
  295. Zhang, J.; Jiang, M.; Zhao, H.; Han, L.; Jin, Y.; Chen, W.; Wang, J.; Zhang, Z.; Peng, C. Synthesis of Paeonol-Ozagrel Conjugate: Structure Characterization and In Vivo Anti-Ischemic Stroke potential. Front. Pharmacol. 2020, 11, 608221. [Google Scholar] [CrossRef]
  296. Chudasma, M.P.; Shah, S.A.; Qureshi, M.H.N.; Shah, N.; Shah, D.; Trivedi, R.; Shah, V.H. Brief insight on nanovesicular liposomes as drug-delivery carriers for medical applications. J. Explor. Res. Pharmacol. 2023, 8, 222–236. [Google Scholar]
  297. Su, S.; Peter, M.K. Recent Advances in Nanocarrier-Assisted Therapeutics Delivery Systems. Pharmaceutics 2020, 12, 837. [Google Scholar] [CrossRef]
  298. Wi, T.I.; Won, J.E.; Lee, C.M.; Lee, J.W.; Kang, T.H.; Shin, B.C.; Han, H.D.; Park, Y.M. Efficacy of Combination Therapy with Linalool and Doxorubicin Encapsulated by Liposomes as a Two-in-One Hybrid Carrier System for Epithelial Ovarian Carcinoma. Int. J. Nanomed. 2020, 15, 8427–8436. [Google Scholar] [CrossRef]
  299. Castro, N.R.; Pinto, C.S.C.; Santos, E.P.; Mansur, C.R.E. Hybrid Vesicular Nanosystems Based on Lipids and Polymers Applied in Therapy, Theranostics, and Cosmetics. Crit. Rev. Ther. Drug Carr. Syst. 2020, 37, 271–303. [Google Scholar] [CrossRef]
Figure 1. Different types of nanovesicular carriers utilized for the delivery of phytochemicals.
Figure 1. Different types of nanovesicular carriers utilized for the delivery of phytochemicals.
Pharmaceutics 17 00464 g001
Table 1. Chemical structure, sources, activities, and formulation challenges of major phytochemicals.
Table 1. Chemical structure, sources, activities, and formulation challenges of major phytochemicals.
NameStructureBotanical SourcesTypeBiological RoleFormulation Challenges
AllicinPharmaceutics 17 00464 i001Allium sativumThiosulfinateAntimicrobial, antioxidant, anti-inflammatory, cardioprotective, anticancer Highly unstable, short shelf life, strong odor, low oral bioavailability
ArtemisininPharmaceutics 17 00464 i002Artemisia annuaSesquiterpene lactoneAntimalarial, anticancer, anti-inflammatory, antiviral, antiparasiticLow solubility, short-half life, chemical instability, limited shelf life, drug resistance
β-carotenePharmaceutics 17 00464 i003Daucus carota, Ipomoea batatas, Cucurbita maxima, Spinacia oleraceaCarotenoidProvitamin A, antioxidantInsoluble in water, prone to oxidation
β-LapachonePharmaceutics 17 00464 i004Tabebuia avellanedaeQuinoneAntioxidant, anti-inflammatory, cardioprotective, anticancer, antimicrobial Poor solubility, low bioavailability, toxicity, short half-life
Betulinic acidPharmaceutics 17 00464 i005Betula pendula, Syzygium formosanum, Ziziphus mauritiana, Diospyros melanoxylonPentacyclic triterpenoidAnti-cancer, anti-HIV, anti-inflammatoryLow water solubility, limited bioavailability
CanthaxanthinPharmaceutics 17 00464 i006Cantharellus cinnabarinus, Capsicum annuum, Daucus carotaKeto-carotenoidAntioxidantInsoluble in water, prone to oxidation
CatechinsPharmaceutics 17 00464 i007Camellia sinensis, Theobroma cacao, Vitis viniferaPolyphenolic flavonoidAntioxidant, anti-inflammatory, cardiovascular, anticancer, metabolic health, neuroprotective, antimicrobial Poor solubility, low stability, rapid metabolism, low oral bioavailability,
CinnamaldehydePharmaceutics 17 00464 i008Cinnamomum cassia, Cinnamomum verumPhenylpropanoidAntimicrobial, antiparasitic, antioxidant, antidiabetic, anticancer, cardiovascular, neuroprotectiveVolatility, low stability, poor solubility, mucosal irritation, rapid metabolism
CurcuminPharmaceutics 17 00464 i009Curcuma longaDiarylheptanoidAnti-inflammatory, antioxidant, anti-cancerPoor bioavailability, rapid metabolism
EchinacosidePharmaceutics 17 00464 i010Echinacea purpurea, Cistanche deserticola, Leonurus japonicusPhenylethanoid glycoside Antioxidant, anti-inflammatory, neuroprotective, immunomodulatory, antiaging, wound healing, hepatoprotectiveLow stability, poor solubility, low bioavailability, short half-life
FisetinPharmaceutics 17 00464 i011Nelumbo nuciferaFlavonoidAntioxidant, anti-inflammatory, senolytic, neuroprotective, anti-cancer, cardioprotectivePoor solubility, sensitive to heat, oxygen and light, first-pass metabolism
Ginsenoside Rg3Pharmaceutics 17 00464 i012Panax ginseng, Panax notoginsengSaponinAnticancer, neuroprotective, anti-inflammatory, cardioprotective, antifatigue, immunomodulatory, antidiabetic Poor solubility, low bioavailability, unstable, low permeability, first-pass metabolism
HonokiolPharmaceutics 17 00464 i013Magnolia officinalis, Magnolia grandifloraBiphenolAnticancer, neuroprotective, anti-inflammatory, antimicrobial, antiviral, cardioprotective, anxiolytic, antidepressantPoor solubility, rapid metabolism, low stability, low intestinal permeability
LuteinPharmaceutics 17 00464 i014Spinacia oleracea, Brassica oleraceaCarotenoidAntioxidantInstability to light, poor solubility
LycopenePharmaceutics 17 00464 i015Solanum lycopersicum L., Citrullus lanatus, Psidium guajavaAcyclic carotenoidCardiovascular, reduce oxidative stressPoor solubility
PaeonolPharmaceutics 17 00464 i016Paeonia suffruticosa, Paeonia lactifloraPhenolic compoundAnti-inflammatory, analgesic, antioxidant, antimicrobial, immunomodulatory, cardioprotective, anticancerPoor solubility, volatility, low stability, low bioavailability
PiperinePharmaceutics 17 00464 i017Piper nigrum, Piper longumAlkaloidAntioxidant, anti-inflammatory, antimicrobial, anti-carcinogenic, neuroprotectivePoor solubility, low bioavailability, light/heat sensitive
PlumbaginPharmaceutics 17 00464 i018Plumbago zeylanica, Diospyros lotus, Drosera rotundifoliaNaphthoquinoneAnticancer, anti-inflammatory, antioxidant, cardioprotective, neuroprotective, antidiabetic, antimicrobialLow stability, poor solubility, low oral bioavailability, short half-life, toxicity
QuercetinPharmaceutics 17 00464 i019Capparis spinosa, Malus domestica, Vaccinium corymbosumFlavonoidAntioxidant, anti-inflammatory, anticancer, antiviral, antimicrobial, cardioprotective, neuroprotectivePoor solubility, low bioavailability, stability, low permeability
ResveratrolPharmaceutics 17 00464 i020Vitis vinifera, Arachis, Punica granatum, Polygonum cuspidatumStilbeneAntioxidant, anti-inflammatory, anti-carcinogenic Slightly soluble in water
Salvianolic acid BPharmaceutics 17 00464 i021Salvia miltiorrhizaPolyphenolAntioxidant, anti-inflammatory, cardioprotective, neuroprotective, anti-fibrotic, anticancer, antidiabeticLow oral bioavailability, instability, first-pass metabolism, short half-life, poor permeability
ThymoquinonePharmaceutics 17 00464 i022Nigella sativaBenzoquinoneHepato-protective, neuroprotective, nephroprotective, cardioprotective, anti-inflammatory, antimicrobial, anti-carcinogenic, antidiabetic, immunomodulatoryPoor solubility, instability, low bioavailability, bitter taste
Ursolic acidPharmaceutics 17 00464 i023Rosmarinus officinalis, Prunella vulgaris, Ocimum sanctumTriperpenoidAntioxidant, anti-inflammatory, anti-cancer, antimicrobial, cardioprotectivePoor solubility, low bioavailability, unstable
VerbascosidePharmaceutics 17 00464 i024Verbascum thapsusPhenylethanoid glycoside Antioxidant, anti-inflammatory, neuroprotective, hepatoprotective, antimicrobial, nephroprotective, wound healing, anticancerLow stability, poor solubility, low bioavailability, short half-life
WogoninPharmaceutics 17 00464 i025Scutellaria baicalensisFlavonoidAnticancer activity, neuroprotective, anti-inflammatory, antiviralPoor solubility, low stability, first-pass metabolism
Table 2. Comparison of advantages, limitations, and key attributes of nanovesicular carriers.
Table 2. Comparison of advantages, limitations, and key attributes of nanovesicular carriers.
CarrierAdvantagesLimitationsSkin PermeationEntrapment EfficiencyDrug LoadingScalabilityStabilityCost
PhytosomesEnhanced bioavailability, natural origin, suitable for phytoactivesLimited to amphiphilic/lipophilic phytochemicals, cost of phospholipidModerateHighModerate to highGood, relatively simple formulation methodsModerate, sensitive to oxidationHigh due to phospholipids
LiposomesBiocompatible, versatile, enhanced drug deliveryLow stability, high cost, potential leakageModerate, depending on formulationModerate to highModerateChallenging due to cost and stability issuesLow to moderate, prone to oxidationHigh due to phospholipids
InvasomesEnhanced active delivery through stratum corneumVolatility of terpenes, moderate stabilityHighModerate to highModerateModerate, due to terpene handlingModerate, sensitive to terpene lossModerate, terpene inclusion increases cost slightly
NiosomesStable, cost-effective, customizableLess biocompatible compared to liposomes, may require specific surfactantsModerate to high, dependent on surfactant typeHighModerate to highGood, cost-effective methods availableGood, more stable than liposomesLow to moderate, cost-effective materials
BilosomesEnhanced oral bioavailability, protects against gastric degradation, stable under physiological conditionsLimited skin delivery application, bile salts may interact with other excipientsLow to moderateHighModerate to highGood, suitable for large-scale productionHigh, stable in GI tract and physiological pHLow to moderate, bile salts are cost-effective
TransferosomesHigh deformability, deep skin penetrationRequires special handling, potential instabilityHigh due to deformabilityModerate to highModerateModerate, requires specific techniquesModerate, edge activators may affect stabilityModerate, depends on edge activators
EthosomesImproved skin permeation due to ethanol, effective for transdermal deliveryEthanol may irritate, stability issuesHigh due to ethanol’s effect on skin lipidsHighModerate to highModerate to high, depends on ethanol handlingModerate, ethanol can evaporate over timeModerate, ethanol adds to cost
TransethosomesCombines ethosome and transferosome benefits, superior penetrationComplex composition requires precise formulationVery high, superior to ethosomes and transferosomesVery highHighModerate, due to complexityModerate, requires optimized storageModerate to high, complex formulation increases cost
CubosomesHigh stability, controlled and sustained drug release, large surface area, high drug-loading capacityComplex preparation process, potential viscosity issues, limited scalabilityModerate to high, depends on formulationHigh, due to large internal surface area and cubic structureHigh, suitable for both hydrophilic and lipophilic drugsModerate, requires specialized techniques like high pressure homogenizationHigh, resistant to degradation, but sensitive to temperature and pH fluctuationsModerate, influenced by surfactant and stabilizer costs
Table 3. Phytochemical name, preparation technique, composition, and highlights of liposomal delivery of phytochemicals.
Table 3. Phytochemical name, preparation technique, composition, and highlights of liposomal delivery of phytochemicals.
PhytochemicalPreparation TechniqueCompositionDisease/Therapeutic ApplicationHighlightsRef.
β-carotene and luteinThin film hydration with ultrasonicationDipalmitoyl-phosphatidylcholine, L-α-phosphatidyl-ethanolamine, L-α-cephalin (3-snphosphatidyl-ethanolamine), stearylamineAntioxidants, cancer, cardiovascular, skin diseases, eye health, anti-inflammatory effectsβ-carotene adheres to the liposomal boundary, causing external morphological changes. Encapsulation efficiencies of lutein and β-carotene are over 98.8% and 87%, respectively. Lutein in cationic liposomes demonstrates better in vitro release stability (30%) compared to β-carotene (45%) between 3 and 6 h, with a lower leakage rate ensuring higher lutein retention. [102]
β-lapachoneThin film hydrationSoybean phosphatidylcholine, 1,2-distearoyl-sn-glycero-3-phosphoethanol amine-[N-(Carbonyl-methoxypolyethylene glycol-2000), cholesterolBreast cancerUncoated and concanavalin A (ConA) coated β-lapachone liposomes exhibit nano size (100–150 nm). Hemagglutination assays verified ConA avidity, showing that Lipo-ConA and Lipo-PEG-ConA could hemagglutinate red blood cells. Site-specific liposomes demonstrated enhanced toxicity. ConA-coated liposomes achieved greater internalization in MCF7 cells compared to uncoated liposomes. [103]
AllicinReverse phase evaporation Lecithin, cholesterol Diarrhea, flatulence, edema, arthritis, worm infestation and pulmonary complaintsOptimized allicin liposomes showed higher EE, low vesicle size and good stability. The formulation demonstrated sustained release behavior, highlighting their potential for controlled release for wider applications. [104]
ArtemisininThin film hydrationSoy lecithin, cholesterol,1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-(polyethylene glycol)-2000AntimalarialArtemisinin-loaded liposomes, modified with a reversibly activatable cell penetrating peptide ((HE)10-G5-R6 or HE-R6), demonstrated sustained release, enhanced internalization, and increased cytotoxicity compared to non-modified liposomes. Additionally, modified liposomes exhibited prolonged tumor retention and improved tumor suppression. [105]
Betulin and betulinic acidLipid hydration with extrusion Dry egg-phosphatidylcholine/dipalmitoyl-phosphatidylcholineCytotoxic propertiesBetulin and betulinic acid influence mitochondrial bioenergetics and membrane behavior by inhibiting oxidative phosphorylation, affecting H2O2 production, and inducing mitochondrial and liposome aggregation. Laurdan fluorescence studies reveal that these compounds cause phase heterogeneity in lipid systems, linked to their interaction with lipid bilayers. [106]
CanthaxanthinHydration with sonicationSoy lecithinPhotoprotectors, antioxidants, anti-inflammatory, immunity enhancer and reproductive health.Canthaxanthin-loaded vesicles (63–87 nm) were stable, biocompatible, and effective in protecting skin cells. Vesicles preserved fibroblast viability, reduced oxidative and inflammatory responses, and promoted cell migration, showcasing the potential for skin protection and regeneration through their antioxidant, anti-inflammatory, and antiaging effects. [107]
Catechins and curcuminMicrofluidic Dipalmitoylphosphatidylcholine, cholesterolTumorDual-loaded liposomes with curcumin and catechin exhibited nanoscale size (<200 nm) with encapsulation efficiencies of 100% for curcumin and 16.77% for catechin. While curcumin, catechin, and liposomes individually enhanced antiproliferation activity in colon cancer cells, the dual-loaded liposomes showed significantly greater inhibitory effects (p < 0.05). The results highlight the potential of dual encapsulation.[108]
CinnamaldehydeEthanol injection Egg yolk lecithin, tween 80AntibacterialHigher cinnamaldehyde loading reduced liposome membrane fluidity. Antibacterial studies revealed that liposome-encapsulated cinnamaldehyde retained its ability to inhibit Staphylococcus aureus by disrupting cell membrane integrity, demonstrating greater persistence than pure cinnamaldehyde. [109]
CurcuminFilm hydration with hand extrusion1,2-dioleoyl-sn-glycero-3-phosphocholine, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000], cholesterolAnticancerLiposomes loaded with curcumin demonstrated low particle size and excellent physical stability. Dilution showed rapid partitioning to the lipid bilayer. The in vivo efficacy of curcumin is unlikely to improve with liposomes, as cytotoxicity and uptake studies revealed reduced effectiveness in the 3D cell model. [85]
EchinacosideThin film hydration Soybean phosphatidylcholine, cholesterol, peptide angiopep-2, DSPE-PEG2K-MAL, DSPE-PEG 2KParkinson’s diseasePeptide angiopep-2 modified PEGylated echinacoside-loaded liposomes showed significantly higher brain uptake than others. It also showed superior efficacy in reducing MPTP-induced behavioral impairments, oxidative stress, dopamine depletion, and dopaminergic neuron death. This formulation significantly enhanced the neuroprotective effects of drug in the Parkinson’s disease model. [110]
Epigallocatechin gallate and quercetinThin film hydration Lecithin, cholesterol, tween 80AntioxidantLiposomes showed nanosized with encapsulation efficiencies of 64.05% for epigallocatechin gallate and 61.73% for quercetin. After 30 days, size increased slightly by 4.05%, with no significant changes in stability. The DPPH assay confirmed a synergistic antioxidant effect of the encapsulated compounds. [111]
FisetinFilm hydration Dioleyl-phosphatidylcholine, cholesterol, 2-dioctadecylcarbamoyl-methoxyacetylamino) acetic Acid-(Ω-methoxy)-polyethylene glycol 2000 esterGlioblastomaThe formulation exhibited a particle size of 173 ± 8 nm, with drug loadings of 1.7 ± 0.3% for fisetin and 0.8 ± 0.1% for cisplatin, and demonstrated stability over time. Encapsulation preserved the antiangiogenic activity of fisetin, and the formulation displayed an additive therapeutic effect of fisetin and cisplatin on GBM cells. [112]
Genistein and plumbaginLipid film L-α-phosphatidylcholine, 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] ammonium saltProstate tumorNanoliposomes (~100 nm) encapsulating plumbagin and genistein demonstrated ~80% inhibition of prostate tumor growth in xenograft models without significant toxicity. The formulation inhibited the PI3K/AKT3 signaling pathway and reduced Glut-1 transporter levels, slowing tumor growth. The observed antitumor effects were linked to reduced cell proliferation and decreased blood vessel formation. [113]
HonokiolThin film hydration with sonicationSoybean phospholipids, cholesterol, DSPE-mpeg2000OsteosarcomaHyaluronic acid phospholipid conjugates were used to combine honokiol-loaded liposomes which have low particle size and high encapsulation efficiency. The developed formulation inhibited cell proliferation, induced apoptosis, blocked the cell cycle and disrupted mitochondrial activity.[114]
LycopeneThin film hydrationSoybean phosphatidylcholine, cholesterolAntitumorLycopene-loaded liposomes possess low particle size. In vitro, the combination of lycopene and doxorubicin significantly increased cytotoxicity, while in vivo it reduced tumor size in B16 melanoma bearing mice and alleviated doxorubicin induced cardiotoxicity. [115]
PaeonolFilm dispersion Lecithin, cholesterol, polyethylene glycol-p-nitrophenyl ester, dioleoyl-phosphatidyl-ethanolamine Hypertrophic scarsThe particle size of prepared formulation was 235.7 nm. They significantly lowered scar proliferation, improved collagen structure, and reduced VEGF, TGF-β1, and TNF-α levels, demonstrating strong therapeutic potential for scar treatment. [116]
PiperineThin layer dispersion Cholesterol, phosphatidylcholine, sodium cholate, chitosanBreast cancerThe drug included liposomes and chitosan-coated drug-loaded liposomes had nanometric sizes (165.7 to 243.4 nm). A biphasic release, improved permeation, and better in vitro antioxidant activity were noticed with liposomes when compared to piperine. The anticancer study with selected liposomes showed significant reduction of IC50 (p < 0.001) compared to pure drug. [117]
PodophyllotoxinThin layer dispersion Lecithin, cholesterolProstate cancerIn vitro drug release showed a gradual, time-dependent release pattern, with a cumulative release of 70.3% after 24 h. Cell viability tests on PC3 cells indicated that PPT-Lips had more effective anticancer activity than free PPT. [118]
QuercetinThin film hydrationSoybean lecithin, cholesterol, polyethylene glycol 4000 Diabetic nephropathyThe antidiabetic effects of liposomes were demonstrated in rats with diabetic nephropathy. Liposomes and free drug improved biochemical and pathological markers of nephropathy. Liposomes showed higher drug plasma than pure drug. [119]
Resveratrol and curcuminThin film evaporation with ultrasonicationEgg yolk phosphatidylcholine, tween 80AntioxidantThe liposome formulation co-loading curcumin and resveratrol in a 5:1 ratio exhibited small particle size. It demonstrated superior antioxidant properties and better stability compared to liposomes loaded with individual polyphenols. [120]
Salvianolic acid B Soybean phospholipid, hydrogenated soybean phosphatidylcholine, 1,2-dipalmitoyl-sn-glycero-3-phosphocholine, 1,2-dipalmitoyl-sn-glycero-3-phospho-(1-rac-glycerol)Idiopathic pulmonary fibrosisLiposomes made of saturated neutral and anionic phospholipids have higher stability and permeability compared to unsaturated or cationic phospholipids. Better efficacy was observed in bleomycin induced idiopathic pulmonary fibrosis mice model when inhaled salvianolic acid B-loaded liposomes, attributed to inflammation inhibition and regulation of the coagulation fibrinolytic system. [121]
ThymoquinoneThin layer evaporation with ultrasonicationEgg phosphatidylcholine, phospholipon 90 G, plurol oleique, sodium hyaluronate Dry eye diseaseDrug-loaded liposomes reduced proinflammatory markers (Interleukin-1β, Interleukin-6, and tumor necrosis factor) and mitochondrial reactive oxygen species. These findings demonstrated the developed liposomes can enhance the efficacy of thymoquinone in treating dry eye disease. [122]
Ursolic acid and ginsenoside Rg3Reverse evaporation and pH gradient Lecithin, cholesterol and (1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[poly(ethylene glycol)]-2000) with glycyrrhetinic acidHepatocellular carcinomaLiposomes containing ursolic acid and ginsenoside Rg3 showed good EE, drug loading, and sustained drug release. Cell experiments confirmed that codelivery of these actives significantly reduced cell viability, increased apoptosis, and elevated the proportion of cells.[123]
VerbascosideFilm hydration Soy phosphatidylcholine, cholesterolAntineuropathic activityPrepared liposomes have low particle size and sustained drug release (~82.28% in 24 h). In chronic constriction injury rat models, the liposomal formulation (100 mg/kg, i.p.) provided an extended antihyperalgesic effect compared to drug solution, with effects appearing within 15 min and lasting up to 60 min. [124]
Vincristine Ethanol injection with extrusionSphingomyelin, cholesterol, 1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy (polyethylene glycol)-2000]Hematological malignancies and solid tumorsLiposomes with drug lipid ratio (1:5) showed reduced degradation (2.9% in 12 months at 2–8 °C) and extended in vivo half-life (22.7 h). This formulation demonstrated lower toxicity and better antitumor efficacy in a human melanoma model. [125]
WogoninReverse evaporation Soybean phospholipids, cholesterol, 3-succinyl 30-stearyl glycyrrhetinic acidLiver cancerThree types of formulations were tested for antitumor efficacy: drug solution, liposomes, and glycyrrhetinic acid-modified liposomes. The modified liposomes exhibit greater entrapment, cellular uptake, liver accumulation, prolonged retention, and anticancer effects compared to others. [126]
Table 4. Phytochemical name, preparation technique, composition, and highlights of niosomal delivery of phytochemicals.
Table 4. Phytochemical name, preparation technique, composition, and highlights of niosomal delivery of phytochemicals.
PhytochemicalPreparation TechniqueCompositionDisease/
Therapeutic
Application
HighlightsRef.
AmygdalinThin film hydration Dihexadecyl phosphate, tween 60, cholesterolAlzheimer’s diseaseNanocarrier enhanced working memory and recognition, reduced oxidative stress, and restored brain monoamine and neurotransmitter levels. Gene expression analysis revealed significant downregulation of BAX, with upregulation of BCL2, acetylcholinesterase, and monoamine oxidase. [144]
Artemisinin and metforminThin film hydration Cholesterol, span 60Lung cancerPEGylated magnetic niosomes developed facilitated rapid tumor-targeted drug delivery. In vitro studies on A549 lung cancer cells showed significant toxicity under a magnetic field, increased pro-apoptotic Bax expression, decreased antiapoptotic Bcl2 expression, and effective cellular uptake via endocytosis. [143]
β-caroteneThin film hydration Spans 40, 60, 80, tween 20, 40, 60, cholesterol Cancer, cardiovascular diseases, arteriosclerosis, and macular degenerationThe developed formulation has good stability against sunlight, temperatures, oxidative stress, and was stable in culture medium. It was effectively taken up by immortalized and transformed cells at concentrations of 0.1–2 μM, suggesting the formulation as an efficient method for β carotene delivery. [145]
CanthaxanthinThin film hydration with sonication Span 60, cholesterolAntioxidant and free radical scavenging Niosomes prepared with span 60 and cholesterol at a 1:1 molar ratio showed smaller, uniform sizes with higher EE. Stability tests indicated higher degradation under light and higher temperatures. The addition of cholesterol significantly improved drug stability. [146]
CatechinThin film hydrationTween 60, lauryl alcohol, cetyl alcohol, cholesterolAntioxidantDeveloped carrier has nanosize, narrow size distribution, good EE (85.82%), and sustained catechin release. Milk fortified with niosomes maintained its sensory and physicochemical properties while exhibiting enhanced antioxidant activity. [147]
CurcuminThin film hydration with ultrasonicationSpan 60, cholesterolAnticancerDeveloped niosomes showed high entrapment (~99.8%), loading (68.33%), and sustained release (~25.49 in 336 h). The formulation was biocompatible and exhibited dose-dependent toxicity against cancer cells (IC50 at 200 µg/mL, containing 66.75 µg drug). [148]
DiosgeninThin film hydration Span 40, tween 40, cholesterolAnticancerDiosgenin niosomes exhibited nanosize, higher loading efficiency (~89%), controlled and sustained drug release. The viability of HepG2 cell line treated with free diosgenin was 61.25%, which decreased to 28.32% upon niosomes, significantly enhancing its anticancer efficacy. [149]
Epigallocatechin-3-gallateThin film hydrationSpan-60, tween-60, cholesterol, DSPE-PEG (2000)-carboxylic acidLung cancerPrepared niosomes showed ideal pharmaceutical characteristics. In vitro studies of cetuximab conjugated to niosomes on A549 lung carcinoma and BEAS-2B normal bronchial cells assessed cytotoxicity, apoptosis, fluorescence imaging, and theranostic potential. [150]
Fumaria officinalis active rich fractionEther injection Span 60, brij 52, cholesterolAntidiabetic, anti-neuropathic, anti-inflammatoryOptimized formulation (Nio-2) with stylopine (48.3%) and sanguinarine (51.6%) demonstrated high entrapment and stability. Fumaria officinalis, active rich fraction, and Nio-2 showed significant antidiabetic and anti-inflammatory activity, hence suggested as an alternative therapy in inflammations and neuropathic pain. [140]
LycopeneMicrofluidic and thin film hydration with probe sonicationSpan 60, cholesterol UVB protection and anti-hyperpigmentation Niosomes displayed good uniformity, nano size, and low PDI when processed through a hydrodynamic flow-focusing platform. The drug release showed Korsmeyer–Peppas kinetics, and the formulation showed enhanced stability, strong UVB protection, and anti-melanogenesis effects. [151]
PaeonolModified ethanol injection Span 60, cholesterol, polyethylene glycol monostearateAnticancerOptimized PEGylated niosomes exhibited nano size, good entrapment, and sustained release. Pharmacokinetic studies in rats showed a higher elimination half-life (87.5 vs. 17.0 min) and a higher AUC (38.0 vs. 19.48 μg/mL·min) compared to the free drug. Enhanced cytotoxicity was observed against HepG2 cells, with IC50 values of 22.47 μg/mL. [152]
Piperine and curcuminThin film hydration Span 60, cholesterolAntioxidant, Protection against Paraquat-induced pulmonary toxicityOptimized niosomes demonstrated high encapsulation efficiency (>85%), nanosize (264–286 nm), a narrow PDI (0.19–0.23), and stability for 90 days. Co-treatment with curcumin and piperine-loaded nanoparticles effectively restore mitochondrial function, hence reduce acute pulmonary toxicity. [153]
PlumbaginSolvent evaporationSpan 20, cholesterolAntidiabeticThe optimized niosomes demonstrated antioxidant activity and enzyme inhibition of α-amylase (90.69%) and α-glucosidase (88.43%). The data in this study show significant potential for diabetes management by inhibiting oxygen radicals and key enzymes involved in glucose metabolism by the developed carrier. [154]
QuercetinHydration with high pressure homogenizationSpan 60, creemophor RH40Skin whitening and antioxidant The best formulation was prepared with span 60 Cremophor RH40 ratio of 9:11. Niosomes enhanced quercetin’s solubility and photostability while offering sustained release and superior transdermal penetration. Skin retention of quercetin niosomes was 2.95 times higher than that of quercetin solution. [155]
ThymoquinoneSolvent evaporation and hydration with sonicationSpan 20, 40, 60, tween 20, 40, 80, cholesterol, soya lecithinAnticancer, antioxidant, anti-inflammatory, and antimicrobialOptimized niosomes (TMQNIOS) demonstrated low vesicle size (~157.32 to 211.44 nm) and encapsulation (~60–80%). Drug release followed Higuchi’s release kinetics and greater permeation across intestinal mucosa from developed carriers. [156]
Thymoquinone and carum extractThin film hydrationSpan 60, tween 60, ergosterolBreast cancerTwo formulations, namely Nio/TQ (thymoquinone) and Nio/Carum (carum extract), were developed. The MTT assay showed these carriers have greater anticancer activity than their pure form against MCF-7 cancer cell line. Cell cycle analysis revealed G2/M arrest in the TQ, Nio/TQ, and Nio/Carum formulations. [157]
Vincristine Thin film hydration Span 60, tween 60, cholesterol-PEG 600: diacetyl phosphateLung cancerPEGylated drug-containing niosomes demonstrated significantly higher cytotoxicity against TC-1 lung cancer cells and stronger tumor inhibitory effects in lung tumor bearing C57BL/6 mice compared to free drug, suggesting its potential as a promising delivery system for vinblastine in cancer treatment. [158]
Table 5. Phytochemical name, preparation technique, composition, and highlights of bilosomal delivery of phytochemicals.
Table 5. Phytochemical name, preparation technique, composition, and highlights of bilosomal delivery of phytochemicals.
PhytochemicalPreparation TechniqueCompositionDisease/
Therapeutic
Potential
HighlightsRef.
Enteromorpha intestinalis-sulfated polysaccharide-protein complexes Thin film hydrationSodium cholate, span 65Hepatocellular carcinomaOptimized formulation consisted of spherical vesicles (181.1 ± 16.80 nm) with moderate drug entrapment efficiency (71.60 ± 0.25%) and exhibited controlled release behavior. In disease-induced rats, treatment with bilosomes significantly reduced serum levels of α-fetoprotein, endoglin, lipocalin-2, and heat shock protein 70 compared to untreated rats. Liver histology revealed focal degeneration of pleomorphic hepatocytes with mild fibrosis extending from the portal area.[163]
TripterineThin film hydrationSoybean phosphatidylcholine,1, 2-stearoyl-3-trimethylammonium-propane, Sodium deoxycholate, hyaluronic acidArthritisHyaluronic acid-coated tri-loaded bilosomes had a particle size of 118.5 nm and high entrapment efficiency (99.56%). Demonstrated excellent cellular uptake and significantly enhanced intra-articular bioavailability—799.9% > Tri solution. In arthritic animal models, coated tri-loaded bilosomes showed markedly improved pharmacokinetics and superior antiarthritic efficacy compared to uncoated Tri-BLs, leading to notable inflammation reduction.[164]
BerberineThin film hydrationSoybean lecithin, sodium deoxycholate, cholesterol, chitosan, carbopol 974 NFRheumatoid arthritisOptimized chitosan-coated, berberine-loaded bilosomes had a mean particle size of 202.3 nm, entrapment efficiency of 83.8%, and a surface charge of +30.8 mV. Demonstrated a sustained in vitro release profile, enhanced ex vivo skin permeability and confirmed non-irritant nature via histological studies. Topical application of the berberine-loaded bilosomal gel significantly reduced inflammation in a rat model of carrageenan-induced paw edema.[169]
ApigeninSolvent evaporation methodCholesterol, sodium deoxy cholate, tween 80, phosphatidylcholine, chitosanAntimicrobial, anticarcinogenic Optimized chitosan-coated bilosome formulation showed an increased vesicle size (298 ± 3.56 nm), positive surface charge (+17 mV), high encapsulation efficiency (88.1 ± 1.48%), and enhanced drug release (69.37 ± 1.34%). TEM analysis revealed smooth, non-aggregated vesicles. Antimicrobial and cytotoxicity studies confirmed its superior efficacy, displaying larger inhibition zones and greater activity against two cancer cell lines.[170]
Biochanin AThin film hydration Sodium deoxy cholate, span 60, polyethylene glycol-2000SAAnti-inflammatorySelected PEGylated bilosome formulation displayed a vesicle size of 216 ± 6.62 nm, entrapment efficiency of 80.54 ± 1.02%, polydispersity index of 0.231, and zeta potential of –15.4 mV. X-ray diffraction confirmed successful drug encapsulation within the bilosomal matrix. It showed a sustained release profile (88.23 ± 3.54% over 24 h) and high ex vivo intestinal permeation (56.97 ± 2.76% in 6 h). Compared to pure Biochanin A dispersion, the optimized Pegylated formulation demonstrated 4.7-fold greater bioavailability and enhanced anti-inflammatory activity.[173]
CurcuminThin film hydration methodL-α-phosphatidylcholine, sodium cholate, pluronic® P123, and cholesterolMelanomaPEGylated bilosomes demonstrated protective effects and enhanced cellular uptake in A375, Me45, and HaCaT cell lines. They showed selective targeting of tumor cells, with significantly greater cytotoxicity against Me45 melanoma cells, reducing their viability to below 20% after 24 h of phyto-photodynamic treatment.[175]
Costunolide Thin film hydration Span 85, cholesterol, bile saltColon cancerFormulation exhibited superior cytotoxicity against LS174T colon cancer cells and demonstrated safety and selectivity in normal colonic epithelial cells, as compared to raw drug. Bilosomes were found to be more effective in up-regulating proapoptotic genes, down-regulating antiapoptotic BCL2 mRNA, enhancing cytochrome C release, increasing reactive oxygen species generation, and disrupting mitochondrial membrane integrity. [176]
CurcuminThin film hydration Span 60, sodium deoxycholate, cholesterolHepatoprotective and renal protective effectThe formulation showed ideal pharmaceutical characteristics, drug release, good stability, and reduced cytotoxicity compared to free drug. The formulation demonstrated effective hepatic and renal protection in liver cirrhosis-induced rats, preserving organ function and histological integrity. [177]
Epigallocatechin gallate Ethanol injectionTween 40, cholesterolAntioxidant, anti-cancer, regulation of fatty acid metabolism, improvement of intestinal immunityBilosomes demonstrated superior gastrointestinal stability, improved drug bioavailability by 1.98 times. These results highlight the potential of bilosomes as an effective delivery system to enhance the stability and bioavailability of epigallocatechin gallate.[178]
LuteolinThin film hydration Cholesterol, span 60, bile salt, PEG 2000Antioxidant, antimicrobial, and breast cancerThe selected PEGylated bilosomes have nanosize, higher encapsulation (~75.05), showed biphasic release, and greater permeation. Additionally, the nanocarriers showed higher antioxidant activity, greater cell viability on cancer cell lines, and superior antibacterial activity against Staphylococcus aureus and Escherichia coli compared to pure drug. [179]
LycopeneLipid hydration Span 60, cholesterol, sodium cholateAntibacterial effectThe formulation showed strong antibacterial activity against multidrug-resistant K. pneumoniae. In vivo studies in a mouse lung infection model demonstrated significant therapeutic effects, including reduced lung inflammation and congestion, restored normal lung architecture, and decreased pulmonary fibrosis. [180]
PsoralidinThin film hydration Phosphatidylcholine, cholesterol, span 60, sodium deoxycholateBreast and lung cancerDeveloped formulation has shown better mucoadhesivity. In vitro studies revealed significantly improved apoptotic and necrotic effects on breast (MCF-7) and lung (A549) cancer cell lines, highlighting the potential of bilosomes as an effective oral treatment. [181]
Sea-buckthorn pulp oilThin film hydration with ultrasonication L-α-phosphatidylcholine, cholesterol, pluronic P123Antioxidant, anti-inflammatory, wound healing, gastroprotectiveAttenuated total reflection Fourier-transform infrared spectroscopy provided insights into bilosomes oil interactions. This study presents a reliable method to develop highly stable bilosome delivery systems with nanosize suitable for application in food and cosmetics. [182]
SpirulinaThin film hydration Phosphatidylcholine, cholesterol, sodium deoxycholatePhotoaging effectIn vivo studies evaluated the photoprotective and antiaging effects of formulation through biochemical analysis of antioxidant, anti-inflammatory, and anti-wrinkling markers. Biochemical and histopathological findings confirmed that formulation offered superior antiaging benefits compared to pure drug. [183]
Table 6. Phytochemical name, preparation technique, composition, and highlights of transferosomal, ethosomal, and transethosomal delivery of phytochemicals.
Table 6. Phytochemical name, preparation technique, composition, and highlights of transferosomal, ethosomal, and transethosomal delivery of phytochemicals.
PhytochemicalPreparation
Technique
CompositionHighlightsDisease/
Therapeutic Application
Ref.
Transferosomes
Asiatic acidHigh pressure homogenization Sodium deoxycholate, tween 80 Small particle size (27.15–63.54 nm), high encapsulation (90.84%), and 71.65% anti-inflammatory activity were observed with the formulation. In clinical trials, the transfersomal gel showed no adverse effects, along with significant improvements in melanin index and skin elasticity at 2, 4, and 8 weeks, demonstrating its potential as an effective treatment for hypertrophic scars. Hypertrophic scars[191]
Centella asiaticaThin film hydration Soyaphosphatidyl choline, tween 80, propylene glycolThe Centella asiatica transfersomes and Bergamot essential oil nanoemulsions combination showed effectiveness in protecting against UVB-induced skin damage in BALB/c mice. It reduced oxidative stress by enhancing SOD activity, lowering MDA levels, and suppressing proinflammatory cytokines. It also promoted collagen production, highlighting its potential as a therapeutic agent for UVB-induced skin protection. Antiphotoaging effects[192]
Curcuma comosa extractThin film hydration Phospholipon 90G, transcutol P, cremophor RH 40Optimal transfersomes, containing diarylheptanoids dispersed in Carbopol gel followed zero order release kinetics and exhibited greater permeation. In vivo pharmacokinetics in rats showed a peak concentration of ~220 ng/mL and sustained plasma levels for over 12 h. Estrogenic activity[193]
Jabuticaba peelDispersion technique with ultrasonicationLipoid S75, tween 20, hydroxyethyl cellulose, sodium hyaluronateThe extract was encapsulated in transfersomes, with or without polymer modifications (hydroxyethyl cellulose and sodium hyaluronate). Polymer-enriched transfersomes had larger sizes but were more stable and showed superior bioactivity compared to the extract solution, effectively reducing hydrogen peroxide toxicity and accelerating wound healing in cell models.Antioxidant, wound healing[190]
Mulberry leaf extractThin film hydration Phospholipon 90G, tween 80Optimized transferosome gel showed higher antioxidant activity, drug content, EE, ex vivo drug release, spreadability, homogeneity, and stability. Antioxidant [194]
Oleuropein and lentisk oilLipid hydration Soy phospholipid, tween 80Developed transferosomes formulation effectively reduced the overexpression of inflammatory markers, particularly MMP-1 and IL-6, mitigated oxidative stress induced by hydrogen peroxide, and accelerated wound healing in fibroblast monolayers in vitro. Skin regeneration[195]
Radish sprouts extractThin film hydration with sonicationPhospholipon 90G, polysorbate 20, 40, 60, and 80The extract-loaded transferosomes reduce tyrosinase activity and melanin content while being safe for skin cells. Combined with sunscreen emulgels, the formulation enhanced cytotoxic safety, demonstrated stability, provided effective UVA and UVB protection and prolonged efficacy. UV protection and antiaging effect[196]
Solanum xanthocarpum methanolic extractThin film hydration Phospholipon 90G, cholesterol, sodium cholateThe developed formulation showed nanosize, high entrapment, loading and antioxidant activity. Ex vivo studies confirmed significantly better skin permeation (82.86%) and retention compared to the conventional gel (35.28%), highlighting its potential for effective topical delivery. Psoriasis[197]
Rhodomyrtus tomentosa leaf extractThin film hydrationL-α-phosphatidylcholine, cholesterol, tween 80, tween 20, span 80, span 20, sodium deoxycholateSpherical nanovesicles (405.3  ±  2.0 nm), high encapsulation efficiency (81.90  ±  0.31%), low polydispersity index (0.16  ±  0.08), and stable zeta potential (−61.62  ±  0.86 mV). The MIC and MBC values ranged from 8 to 256 and 64 to 1024 μg/mL, respectively. Exhibited strong antioxidant activity against DPPH and ABTS radicals and moderate tyrosinase inhibition. A significant reduction in nitric oxide production (6.78–88.25%) was observed.Soft tissue infections[188]
Syringic acidEthanol injectionPhosphatidylcholine, tween 80, d-α-tocopheryl polyethylene glycol 1000 succinate, diphenyl-1-picrylhydrazyl Optimized transferosomes demonstrated significant antioxidant activity (IC50 = 11.1 µg/mL), and excellent skin deposition (78.72%). Additionally, it reduced acne lesions by 79.5%, outperforming Adapalene® gel (18.7% reduction) without causing irritation or erythema, highlighting its potential as an effective and skin friendly acne treatment. Acne[198]
Ethosomes
Achillea millefolium L. extractCold Lipoid, propylene glycol, ethanolOptimized ethosomal carrier demonstrated 88% free radical scavenging activity, along with high phenolic and flavonoid contents. The topical gel was stable and showed release of 79.8%. Antioxidant [199]
Arctostaphylos uva-ursi extractLipid dispersionPhospholipid, ethanol, propylene glycolOptimized formulations possess all pharmaceutical characteristics. The gel formulation demonstrated significant effects in reducing skin erythema, melanin, and sebum levels while enhancing skin hydration and elasticity. Skin rejuvenation and depigmentation[200]
Berberine chlride and evodiamineModified single step injection techniqueSoybean lecithin, cholesterol, ethanol, propylene glycolThe developed formulation showed highest drug deposition in the epidermis cell viability tests confirmed that the optimized ethosomes enhanced the inhibitory effect on B16 melanoma cells. Melanoma[201]
Brassicaceae extractColdPhospholipon 90 G, cholesterol, ethanolOptimized ethosomal gel showed characteristics suitable for skin application. The release of bioactives follows the Korsmeyer–Peppas model and shows higher flux values.Photoprotectiion[202]
Caffeic acidLipid dispersionSoybean lecithin, pluronic F127Permeability of caffeic acid increased drastically when formulated as ethosomes. Skin-covered oxygen electrode experiments confirmed improved delivery and antioxidant activity of caffeic acid to porcine skin compared to a simple solution. Antioxidant[203]
Phyllanthus nruri, croton tiglium, zingiber officinale extractSolvent dispersion with probe sonicationSoya lecithin, ethanolStudies in cell line (HaCaT) revealed crude extracts-loaded ethosomal formulation inhibits testosterone and improves the cell viability similar to minoxidil. Preclinical safety was confirmed through in vitro cytotoxicity and histopathological studies. Antiandrogenic and cytoprotectant[204]
Sambucus nigra L. extractLipid hydration Soybean phosphatidylcholine, ethanolPrepared ethosomes showed slow release profile over 24 h and exhibited collagenase inhibition activity and excellent skin compatibility in human applications. These results indicate the potential of extract-loaded ethosomes as a promising cosmeceutical ingredient for skin care. Skin care[205]
Vernonia anthelmintica (L.) Willd.Injection with ultrasonicationSoybean lecithin, cholesterol, ethanolEthosome formulation exhibited good physicochemical properties. In vitro studies showed significant therapeutic efficacy against vitiligo caused by hydroquinone exposure. Histological analysis of mouse skin showed increased melanin, elevated enzyme activities, and reduced antioxidant levels. Vitiligo[206]
Transethosomes
ApigeninThin film hydrationSoya phospholipid, span 80Optimized formulation demonstrated good EE, low vesicle size, and greater drug release. Ex vivo permeation showed superior apigenin delivery while cytotoxicity studies confirmed that formulation significantly reduced cell viability more effectively than the conventional gel.Skin cancer[207]
Chenopodium murale extractInjectionSoybean phosphatidylcholine, cholesterol, tween 80Drug-loaded transethosomes exhibited good physicochemical properties. The formulation maintained the extract’s macrostructural integrity, enabling enhanced skin permeation and achieving higher drug release. Anti-inflammatory, antimicrobial, antioxidant and wound healing[208]
ColchicineCold Phospholipon 90G, tween 20, sodium taurocholate, or labrafilFormulation was optimized by 22 full factorial design and has exhibited desirable properties for transdermal drug delivery. Ex vivo studies confirmed higher flux compared to normal gel and demonstrated satisfactory stability. Gout, and familial Mediterranean fever[209]
FisetinLipid film hydration with ultrasonicationLipoid S100, sodium cholateTransethosomes vesicles showed suitable physicochemical properties for transdermal delivery. In vivo data indicated that the formulation fluidized the rigid membranes of rat skin and deeper skin penetration. Skin cancer[210]
Ginger extractCold injectionPhospholipon 90G, cholesterolTransethosomal formulation demonstrated enhanced flux and skin deposition compared to free drug hydrogel. In vivo studies confirmed significant edema reduction by the transethosomal hydrogel compared to free drug and ketoprofen gels. Treated animals exhibited marked decrease in reactive oxygen species and prostaglandin E2 levels. Anti-inflammatory effect[211]
Glycyrrhizic acidLipid film hydrationPhospholipid 90G, sodium cholateTransethosome was optimized using the Box–Behnken design, which exhibited ideal characteristics for skin application. Ex vivo data demonstrated enhanced permeation and distribution of the drug, which highlighted superior skin deposition and retention of the formulation compared to conventional gels. Skin cancer[212]
HexatriacontaneLipid film hydration Lipoid S 100, sodium cholateDeveloped formulation showed optimized characteristics and Higuchi release kinetics. Dermatokinetic studies confirmed higher drug deposits in epidermal layers. Formulation exhibited effective antibacterial activity against S. aureus and E. coli. Skin infections[213]
Sinapic acidThin film hydrationPhospholipon 90 G, sodium deoxycholateOptimized transethosome formulation demonstrated characteristics ideal for skin delivery. Higher antioxidant activity and greater penetration across the Strat-M® membrane were observed. Anti-inflammatory, antioxidant, anticancer, and antibacterial activities[214]
Table 7. Phytochemical name, preparation technique, composition, and highlights of cubosomal delivery of phytochemicals.
Table 7. Phytochemical name, preparation technique, composition, and highlights of cubosomal delivery of phytochemicals.
PhytochemicalPreparation TechniqueCompositionDisease/Therapeutic ApplicationHighlightsRef.
β-caroteneHigh pressure homogenizationGlyceryl monooleate, poloxamer 407AntioxidantDesign of experiment-based optimized formulation has 1800 mg glyceryl monooleate and 200 mg poloxamer 407. The optimized formulation demonstrated higher drug release. Incorporating β carotene into cubosomal formulations enhanced ~2-fold in its antioxidant potential. [239]
Brucea javanica oil, doxorubicin hydrochlorideHigh pressure homogenizationGlycerol monooleate, pluronic F127AntitumorpH-responsive cubosomes exhibited phase transitions under different pH conditions and enhanced interaction in acidic tumor environments. Demonstrated antitumor activity against MCF-7 and doxorubicin-resistant MCF-7 cells, with the potential to overcome doxorubicin resistance. [240]
Curcumin, fish oilThin film hydrationMonoolein, PEG 1000NeuroprotectionOptimized cubosome showed negligible cytotoxicity at concentrations of 300 and 500 nM. In neuronally derived SH-SY5Y cells, this formulation effectively mitigated hydrogen peroxide-induced oxidative stress by reducing intracellular ROS levels and apoptosis. Higher neuroprotective potential noticed here suggests combination treatments in neurodegenerative disorders. [241]
ErucinSolvent evaporationMonoolein, pluronic 84 Antioxidant and antiproliferative activitiesThe antioxidant potential of the developed vesicle was evaluated using DNA nicking, DPPH, and phosphomolybdate assays, and demonstrated significantly improved results compared to erucin alone. Additionally, it exhibited enhanced anticancer activity with a lower IC50 value in MTT assays. [242]
PhycocyaninEmulsification with homogenizationGlyceryl monooleate, poloxamer 407AntioxidantOptimized formulation is ideal for skin therapy, better stability and drug release followed the Higuchi model. Drug formulation showed superior skin permeability compared to the phycocyanin solution and was readily taken up by keratinocytes, providing prolonged antioxidative effects. [243]
Ruta graveolens extractHot emulsification Glyceryl monooleate, pluronic F108Anti-asthmaticCubosomal formulation reduced asthma scores and improved lung function by restoring the FEV1/FVC ratio to normal levels. The nanocarrier demonstrated strong antioxidant and anti-inflammatory effects by reducing malondialdehyde, IL-4, IL-7, TGF-β, and Ig-E levels while increasing superoxide dismutase and INF-γ levels in bronchoalveolar lavage fluid. [238]
Withanolide AHot emulsificationGlycerol monooleate, poloxamer 403, pluronic F127Anti-diabetic, anti-neuropathic, anti-inflammatory, and anti-bacterial activitiesDeveloped formulation showed superior antidiabetic, antineuropathic, anti-inflammatory, and antibacterial activities than the drug alone. Effects like HbA1c normalization, insulin secretagogue potential, oxidative stress reduction, and modulation of pro-inflammatory cytokines indicate the potential of this carrier in oral therapy.[244]
Yucca filamentosaHot emulsificationGlyceryl monooleate, pluronic F108 Gastroprotective effectThe formulation showed superior gastroprotective, antioxidant, and anti-inflammatory effects in rat models of ethanol induced gastric injury and was comparable to famotidine. Developed formulations demonstrated potential in preventing peptic ulcer recurrence through modulation of the HMGB-1/RAGE/TLR4/NF-κB pathway. [245]
Table 8. Overview of preparation techniques, excipients, and stability characteristics of different vesicular nanocarriers.
Table 8. Overview of preparation techniques, excipients, and stability characteristics of different vesicular nanocarriers.
Vesicular NanocarrierPreparation TechniquesCommonly Used ExcipientsKey Features and StabilityRef.
LiposomesThin film hydration, electroformation, vesicle fusion, solvent injection, detergent dialysis, reverse phase evaporation, solvent spherule, size reduction, microfluidic, supercritical fluid, freeze-drying of double emulsions, membrane contactor method.Phospholipids (e.g., phosphatidylcholine), cholesterol, cryoprotectants (e.g., trehalose)Encapsulate hydrophilic and lipophilic drugs. Stability: Sensitive to oxidation, requires antioxidants or lyophilization for improved stability.[246]
InvasomesThin film hydration, sonication, extrusionPhospholipids, cholesterol, terpenes (e.g., cineole, limonene)Enhanced dermal and transdermal drug delivery via terpene-mediated membrane fluidization. Stability: Moderate; terpene volatility may impact long-term stability.[248]
NiosomesThin film hydration, ether injection, micro fluidization, reverse phase evaporationNon-ionic surfactants (e.g., Span 60, Tween 80), cholesterolCost-effective alternative to liposomes. Stability: More stable than liposomes; however, sensitive to temperature and pH changes.[249]
BilosomesThin film hydration, sonication, extrusionBile salts (e.g., sodium deoxycholate), phospholipids, cholesterolImproved oral bioavailability via bile salt stabilization. Stability: Enhanced resistance to gastrointestinal degradation; stable under physiological pH.[165,250]
TransferosomesThin film hydration with edge activators, ultrasonic dispersion, ethanol injectionPhospholipids, edge activators (e.g., sodium cholate, span 80), ethanolHighly deformable vesicles for transdermal delivery. Stability: Sensitive to environmental conditions like ethanol evaporation or oxidation.[251]
EthosomesHot or cold method, sonication, extrusionEthanol, phospholipids (e.g., phosphatidylcholine), propylene glycolEnhanced skin penetration. Stability: Sensitive to ethanol evaporation; proper sealing is needed to maintain integrity.[252]
TransethosomesCombination of ethosomes and transferosomes techniques, sonication, extrusionPhospholipids, ethanol, edge activators (e.g., Tween 80)Combines properties of ethosomes and transferosomes. Stability: Sensitive to ethanol loss and oxidation; requires proper storage.[221,253]
CubosomesTop-down approach, bottom-up approachLipids (e.g., glyceryl monooleate), polymers (e.g., pluronic F127)High internal surface area for drug encapsulation. Stability: More stable than vesicular systems; sensitive to dilution and surfactant imbalance.[229]
Table 9. Typical in vitro and in vivo tests of vesicular carriers utilized for phytochemical delivery applications.
Table 9. Typical in vitro and in vivo tests of vesicular carriers utilized for phytochemical delivery applications.
Test TypePrincipleEquipment UsedEvaluation ParametersRef.
Particle size analysisDetermines vesicle size distributionDynamic light scattering (DLS)Particle size, polydispersity index[271]
Zeta potentialMeasures surface charge of vesiclesZeta potential analyzerSurface charge, colloidal stability[272]
In vitro drug releaseMeasures drug diffusion through a membraneFranz diffusion cell, dialysis membraneDrug release rate, cumulative release profile[273]
PermeabilitySimulates passive drug permeationPermeability apparatus, membrane modelsPermeability coefficient, flux rate[274]
Stability testingAssesses long-term stabilityStability chambers, DLS, HPLCParticle size, drug retention, degradation products[275]
Thermal stabilityEvaluates stability under varying temperature conditionsThermal analyzer, stability chamberDegradation rate, active ingredient stability[276]
Oxidative stabilityAssesses stability in the presence of oxygen or reactive speciesOxidative chamber, spectrophotometerOxidation products, active ingredient retention[277]
Photostability testAssesses stability under ultraviolet (UV) or light exposureUV chamber, spectrophotometerDegradation products, remaining active phytochemical[278]
Antioxidant assayEvaluates antioxidant activity (e.g., DPPH, ABTS, FRAP, ORAC, FIC, CUPRAC)UV–vis spectrophotometerPercentage inhibition, antioxidant capacity[279]
Anti-inflammatory assayMeasures inhibition of inflammatory mediatorsEnzyme-linked immunosorbent assay (ELISA), cell culture modelsCytokine levels (e.g., TNF-α, IL-6)[280]
Pharmacokinetic studyMeasures drug absorption, distribution, metabolism, and excretionHPLC, mass spectrometryCmax, Tmax, AUC, half-life[281]
Biodistribution studyAnalyzes drug accumulation in tissuesFluorescent imaging, gamma countersOrgan-specific drug concentration, clearance[282]
Therapeutic efficacyEvaluate pharmacological effects (e.g., anticancer, neuroprotection)Animal models, histopathologyDisease regression, biomarker levels[283]
Cell viability Assesses cell metabolic activity post-exposure (e.g., MTT)Microplate reader, cultured cellsCell viability, IC50[284]
Oxidative stress markersAnalyzes ROS, lipid peroxidation, or protein oxidationELISA, biochemical assaysROS levels, malondialdehyde, protein oxidation[285]
Cytokine profilingMeasures inflammatory and immunomodulatory effectsELISA, multiplex assaysCytokine expression (e.g., IL-10, IFN-γ)[286]
Antiviral activity assayEvaluates inhibition of viral replicationCell culture models, PCRViral load, cytopathic effect[287]
Antimicrobial assayDetermines antimicrobial efficacyMicrodilution plates, colony counterMinimum inhibitory concentration, zone of inhibition[288]
Cardioprotective assayAssesses protection against cardiac injury or dysfunctionAnimal models, echocardiographyCardiac biomarkers, ECG, histology[289]
Neuroprotective assayEvaluate protective effects against neuronal damageNeuronal cell lines, animal modelsNeuronal viability, ROS levels, neuroinflammation markers[290]
Anticancer assayMeasures inhibition of cancer cell growthMTT assay, flow cytometryCell viability, apoptosis rate, tumor volume[291]
Hepatoprotective assayAssesses protection against liver damageAnimal models, liver function testsLiver enzymes (ALT, AST), histopathology[292]
Immunomodulatory assayEvaluates modulation of immune responseELISA, multiplex cytokine assaysCytokine profile, immune cell activation[293]
Antidiabetic assayMeasures blood glucose regulation and insulin sensitivityAnimal models, glucometerBlood glucose, insulin levels, HbA1c[294]
Table 10. A compilation of recently filed patents for pharmaceutical nanosuspensions, highlights of their innovation.
Table 10. A compilation of recently filed patents for pharmaceutical nanosuspensions, highlights of their innovation.
Application IDPublication DateTitleSummary of InventionPotential Benefits
118682515 October 2007 Liposomal curcumin for treatment of neurofibromatosisThe invention presents a method to treat neurofibromatosis types 1 and 2 using curcumin or its analogues encapsulated in colloidal drug delivery systems, such as liposomes, nanoparticles, or micelles. The system targets Merlin and related pathway proteins and is administered parenterally with a pharmaceutically acceptable carrier.Neurofibromatosis
WO/2009/06178714 May 2009Coated devices and method of making coated devices that reduce smooth muscle cell proliferation and platelet activityThe invention features drug eluting stents and coated devices that release resveratrol and quercetin to maintain blood flow, reduce smooth muscle cell proliferation, and limit restenosis.Atherosclerosis, stenosis, and clotting disorders.
201010158483.X22 March 2010Allicin liposome for resisting microbe, protozoan and tumor and preparation method thereofThe invention introduces an allicin liposome formulation to treat microbes, protozoa, tumors, and viruses. Its enhanced properties expand allicin’s applications beyond bacterial and fungal infections, offering new therapeutic possibilities.Antimicrobial, protozoal and viral infection
201210041878.023 February 2012Wogonin liposome preparation modified with glycyrrhetinic acid and preparation method thereofThe invention presents a glycyrrhetinic acid-modified wogonin liposome, prepared through the reverse evaporation method, which enhances efficacy against liver tumor cells and improves liver cancer treatment.Hepatic carcinoma
1477716015 March 2013Compositions and methods including celecoxib and plumbagin relating to treatment of cancerThe invention presents a method for treating proliferative diseases, especially skin cancer, using a combination of celecoxib and plumbagin for improved therapeutic outcomes.Skin cancer
WO/2016/00578614 January 2016Liposomal formulations comprising thymoquinone and taxane, and methods of treating cancer using sameThe invention describes liposomal compositions with taxane and thymoquinone, offering synergistic anticancer effects, improved drug encapsulation, enhanced stability, and consistent taxane release for more effective cancer treatment.Synergistic Anticancer effects
201711105675.210 November 2017Brain-targeting liposome with paeonol-ozagrel conjugate and preparation method thereofThe invention describes a brain-targeting liposome containing a paeonol-ozagrel conjugate, designed to enhance drug delivery across the BBB. It prolongs drug circulation time and enables targeted treatment of various thrombotic cardiovascular and cerebrovascular diseases.cardiac and cerebral ischemia or infarction
202311320134.712 October 2023Echinacoside liposome, LRP1 targeted echinacoside-loaded liposome, and preparation method and application of echinacoside-loaded liposome and LRP1 targeted echinacoside-loaded liposomeThe invention features an LRP1-targeted echinacoside liposome that enhances brain delivery, improves dopamine levels, reduces oxidative stress, and restores motor function in neurodegenerative disease models.Neurodegenerative diseases
202311327683.713 October 2023Aptamer-macrophage membrane composite nano delivery system for targeting breast cancer and method for analyzing anti-breast cancer action mechanismThe invention describes a nano-delivery system utilizing betulinic acid encapsulated in macrophage membranes modified with nucleic acid aptamers for targeted breast cancer treatment. Betulinic acid effectively inhibits cancer cell proliferation and induces apoptosis by blocking the PI3K/AKT and MAPK signaling pathways.Breast cancer
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

Jacob, S.; Kather, F.S.; Boddu, S.H.S.; Rao, R.; Nair, A.B. Vesicular Carriers for Phytochemical Delivery: A Comprehensive Review of Techniques and Applications. Pharmaceutics 2025, 17, 464. https://doi.org/10.3390/pharmaceutics17040464

AMA Style

Jacob S, Kather FS, Boddu SHS, Rao R, Nair AB. Vesicular Carriers for Phytochemical Delivery: A Comprehensive Review of Techniques and Applications. Pharmaceutics. 2025; 17(4):464. https://doi.org/10.3390/pharmaceutics17040464

Chicago/Turabian Style

Jacob, Shery, Fathima Sheik Kather, Sai H. S. Boddu, Rekha Rao, and Anroop B. Nair. 2025. "Vesicular Carriers for Phytochemical Delivery: A Comprehensive Review of Techniques and Applications" Pharmaceutics 17, no. 4: 464. https://doi.org/10.3390/pharmaceutics17040464

APA Style

Jacob, S., Kather, F. S., Boddu, S. H. S., Rao, R., & Nair, A. B. (2025). Vesicular Carriers for Phytochemical Delivery: A Comprehensive Review of Techniques and Applications. Pharmaceutics, 17(4), 464. https://doi.org/10.3390/pharmaceutics17040464

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