Next Article in Journal
A Hyper-Attentive Multimodal Transformer for Real-Time and Robust Facial Expression Recognition
Previous Article in Journal
Influence of Degree of Saturation on Soil–Pile Interactions for Piles in Expansive Soils
Previous Article in Special Issue
Nanoformulations Loaded with Phytochemicals for Combating Wound Infections and Promoting Wound Healing: Current Applications and Innovations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeted Delivery Strategies for Hydrophilic Phytochemicals

by
Marta Sharafan
1,†,
Anna Dziki
1,†,
Magdalena Anna Malinowska
2,
Elżbieta Sikora
2 and
Agnieszka Szopa
3,*
1
Department of Organic Chemistry and Technology, Faculty of Chemical Engineering and Technology, CUT Doctoral School, Cracow University of Technology, 24 Warszawska St., 31-155 Cracow, Poland
2
Department of Organic Chemistry and Technology, Faculty of Chemical Engineering and Technology, Cracow University of Technology, 24 Warszawska St., 31-155 Cracow, Poland
3
Department of Medicinal Plant and Mushroom Biotechnology, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Cracow, Poland
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Appl. Sci. 2025, 15(13), 7101; https://doi.org/10.3390/app15137101
Submission received: 15 May 2025 / Revised: 14 June 2025 / Accepted: 18 June 2025 / Published: 24 June 2025
(This article belongs to the Special Issue Applications of Nanocarriers for Phytochemical Delivery)

Abstract

Featured Application

This review provides a knowledge base for designing efficient delivery systems for hydrophilic plant-derived compounds, offering practical guidance for pharmaceutical and cosmetic industries seeking to improve skin penetration, bioavailability, and formulation stability.

Abstract

Hydrophilic phytochemicals, such as flavonoids and phenolic acids, possess important biological activities, including antioxidant, anti-inflammatory, and anticancer effects. However, their application is hindered by low membrane permeability, poor chemical stability, and limited skin penetration. This review provides a comprehensive analysis of advanced delivery strategies aimed at enhancing the solubility, bioavailability, and therapeutic efficacy of selected hydrophilic compounds. Specifically, it focuses on the encapsulation of flavonoids such as quercetin, luteolin, and apigenin, as well as phenolic acids including ferulic acid, caffeic acid, and chlorogenic acid. The review discusses various nanocarrier systems: liposomes, niosomes, exosomes, and polymeric nanoparticles (e.g., nanocapsules, nanospheres) and compares their structural characteristics, preparation methods, and functional benefits. These delivery systems improve the physicochemical stability of active compounds, enable controlled and targeted release, and enhance skin and cellular absorption. Despite certain challenges related to large-scale production and regulatory constraints, such approaches offer promising solutions for the pharmaceutical and cosmetic application of hydrophilic plant-derived compounds.

1. Introduction

1.1. Background on Phytochemicals: Overview of the Importance and Therapeutic Potential of Plant-Derived Active Substances

Since ancient times, plants have played a vital role in human life. Traditionally, medicinal plants have been key ingredients in various cosmetic products and infusions, used for cosmeceutical purposes and even for addressing dermatological conditions. Nowadays, the list of plants, which appears in cosmetic formulations, continues to grow has still increasing. Plant extracts possess lots of properties including moisturizing, regenerating, anti-inflammatory as well as antioxidant and anti-aging activity, which are in great demand in cosmetics. Those functions are determined by the presence of valuable secondary metabolites, which are termed as phytochemicals [1,2,3].
Phytochemicals are plant-derived active substances, which mainly include alkaloids, terpenoids or polyphenolic compounds [4]. This review will focus on polyphenols, particularly flavonoids and phenolic acids along with the active molecules associated with them. Phytocompounds derived from the above-mentioned groups are popular ingredients in cosmetic formulations and act as important bioactive agents [5,6,7,8].
However, despite the benefits offered by those phytochemicals, issues arising from their hydrophilicity, stability and bioavailability remains challenging for designing cosmetic formulations [9,10].

1.2. The Overview on the Phytochemicals Penetartions Rates: Discuss Factors Affecting Skin Penetration

Skin is a large organ whose main role, aside from thermoregulation, is to protect the body from mechanical, biological, and chemical factors. Stratum corneum (SC), the outermost layer of the epidermis, acts as a barrier, which protects the skin from external factors including penetration of drugs or cosmetics. Active substances can penetrate the stratum corneum through several pathways. The most common is the transepidermal route, which includes two mechanisms: the intercellular route—used by the majority of compounds, and the transcellular route. Penetration can also occur through appendageal structures such as hair follicles and sweat glands. In the transepidermal pathway, compounds either pass directly through the corneocytes (transcellular) or move between them by crossing the intercellular lipid matrix—this intercellular route is the primary mode of penetration for most substances [11,12,13,14,15,16,17,18,19] (Figure 1).
The penetration rate of active substances is determined by their hydrophobic nature. The octanol/water partition coefficient P (logP) is an important factor, which has an impact on membrane permeability. Positive values for logP indicates more hydrophobic nature of the compound. While hydrophilic compounds are characterized by negative values for logP, which determine its higher aqueous phase affinity following its low penetration [20,21].
The molecules successful penetration is determined by the suitable balance between lipophilicity and hydrophilicity and also appropriate molecular mass of the compounds, which should be less than 500 Da. In addition to physicochemical properties, the formulation characteristics is equally important for effective skin penetration. For instance, formulations with greater viscosity delay the absorption of active compounds, because of the reduced diffusion coefficient. The pH of the cosmetic base is also crucial because it determines whether the active molecule is dissociated. Undissociated molecules penetrate more effectively, because of its better lipid dissolution compared to dissociated molecules, which easily separate into ions. Moreover, the presence of skin penetration enhancers (e.g., alcohols, amides, glycols) in the cosmetic base also have an influence by reducing the skins barrier resistance [11,13,14,15,16,17,18]. Furthermore, skin permeation is also affected by physiological factors (e.g., age, skin condition and metabolism), as well as sufficient physicochemical characteristics of drug carrier (e.g., size, shape or surface charge), which must be satisfied to guarantee the effective penetration deep into the skin [19,20,22,23].
All the above-mentioned factors are extremely significant and relate to many active molecules. However, the primary focus reviewed here is on hydrophilic compounds and the additional factors, which affect its delivery efficiency.

1.3. Challenges of Hydrophilic Phytochemicals: Discuss Solubility, Stability, and Bioavailability Issues Associated with Hydrophilic Active Substances

Considering the polar nature of hydrophilic phytochemicals, their effective penetration through the skin faces fundamental challenges. Since the stratum corneum extracellular lipid matrix predominantly contains lipids, e.g., cholesterol and ceramides, as well as free fatty acids, so it results on low solubility in nonpolar environments [19,24].
Hydrophilic phytochemicals are often chemically unstable under environmental stressors such as temperature, pH, oxygen, and light. These weaknesses result in poor skin absorption and limited bioavailability. For instance, polyphenols, being the most abundant plant-derived compounds, are unstable, because of the presence of hydroxyl groups (–OH) attached to benzene ring, which are highly reactive. Therefore, with an increase of hydroxyl groups the stability of polyphenols is lowered. Polyphenols also have a tendency to autooxidation, which provide peroxide and hydroperoxide formations. PH impacts polyphenols stability as well, high pH values decreases stability due to the chemical form changing [25]. For instance, anthocyanidins, which are sensitive to pH, are more stable in acidic conditions what manifest with its red color [26,27,28,29]. Furthermore, at high temperatures with alkaline conditions polyphenols undergo epimerization, leading to its instability [25].
The next challenge of hydrophilic phytochemicals resulting from its limited solubility and instability is low bioavailability. To improve their therapeutic potential, modern strategies increasingly utilize carriers. In recent years, different forms of topical administration have become available. For instance, nanostructured lipid carrier gel or nanogel are used to deliver quercetin and increase its bioavailability [30]. Lipid-derived carriers such as liposomes form the occlusive layer, which increase skin humidity and enhance the active molecules penetration [31,32].
This work aims to present a comprehensive review of selected lipid-based delivery systems as an effective tool to overcome the limitations of hydrophilic compounds.

2. Delivery Systems for Hydrophilic Phytochemicals

2.1. General Considerations

In recent years, especially organic vesicles e.g., liposomes, niosomes, exosomes or polymeric nanoparticles have been found to enable not only skin penetration through stratum corneum, but can also improve the biocompatibility, stability as well as prolongated action of the contained active ingredient [33,34].
The fundamental biological barriers related to hydrophilic compound delivery have been addressed in Section 1.2. Here we will focus specifically on the functional roles, advantages, and possible limitations of carrier technologies. As detailed in Section 1.2, hydrophilic phytochemicals face significant barriers to skin absorption due to their polar nature and low membrane permeability. Delivery systems based on nanotechnology, such as liposomes, niosomes, and polymeric nanoparticles (Figure 2), offer a viable solution by enhancing solubility, stabilizing active compounds, and facilitating their transport through the stratum corneum [35].

2.2. Liposomes

Liposomes (Figure 2) are the well-explored representatives of the carrier ensemble. They are defined as sphere-shaped vesicles, consisting of one or more phospholipid bilayers, which are self-formed in aqueous solution. Liposome’s structure is biphasic composed of hydrophilic core and hydrophobic bilayer, enabling both hydrophilic and hydrophobic compounds to be delivered. Liposomes were first discovered in 1960s by the British scientist Alec Bangham. Since that time, they have started gaining popularity as a prospective delivery system. The Dior “Capture” was the first liposomal cosmetic product introduced to the market in 1986. Since that time, the list of liposomal products are increasing [36].
Based on the physical characteristics (e.g., structure, particle size, lamellarity), liposomes can be divided into different categories: small unilamellar vesicles (SUVs), large unilamellar vesicles (LUVs), giant unilamellar vesicles (GUVs), oligolamellar vesicles, (OLVs), multilamellarvesicles (MLVs), and multivesicular vesicles (MVVs) [37] (Table 1). There is a wide list of liposome preparation methods, which includes, for instance, film hydration also known as “Bangham method”, reverse phase evaporation or solvent injection (ethanol, diethyl ether). The application of the specific method is determined by the desired liposome characteristics [38,39]. All of techniques are presented in Table 1.

2.3. Niosomes

Niosomes (Figure 2) are self-assembled vesicles, which are formed of non-ionic surfactants (e.g., alkyl ethers, sorbitan fatty acid esters) and lipids such as cholesterol. Due to the amphiphilic character, niosomes could deliver both hydrophilic and hydrophobic drugs. The first niosome-based formulation was patented by L’Oreal (France) in 1970s. Then, in 1986, Lâncome (France) introduced to the market niosome formulation under the trade name “Niosôme” [40,41].
Compared to liposomes, niosomes are more chemically stable. The active substance entrapment efficiency of niosomes is also better compared to liposomes due to the fact that the dominant compounds of liposomes such as phospholipids are unstable and tend to degrade. The presence of cholesterol in niosomes results in enhanced vesicle stability as well as entrapment efficiency. As non-ionic surfactants, from which niosomes are composed, are not carrying a charge, they also offer better stability of system. Furthermore, to increase the stability of niosomes, the charged molecules such as dicetyl phosphate or stearyl amine could be provided, which by prevent the vesicles aggregation [41,42,43,44].

2.4. Exosomes

Exosomes (Figure 2) are defined as subgroup of nanosized spherical extracellular vesicles secreted by most cells. Exosomes are made of single lipid bilayer containing nucleic acids, proteins, lipids and lipid-related enzymes and an aqueous core, which enables delivery of hydrophilic drugs [45,46,47,48,49]. Exosomes are divided to natural (animal or plant-derived) and engineered, which are recognized as modified version of natural exosomes. Engineered exosomes are modified to enable advantageous drug delivery. They increase stability and bioavailability as well as reduce toxicity. In addition to the advantages of exosomes such as biocompatibility, biodegradability, or low toxicity, they also have some limitations (Table 1). For instance, exosomes cannot be stored long, for this reason protection techniques are required e.g., freezing or spray-drying [50,51].

2.5. Polymeric Nanoparticles

Polymeric nanoparticles (Figure 2) are defined as colloidal systems composed of polymers. They are divided into nanocapsules and nanospheres. Nanospheres consist of a solid core and polymeric matrix within which active molecules are encapsulated. In the case of nanocapsules, the active compound is entrapped in an inner liquid or solid core secured by polymeric membrane, which is composed of non-ionic surfactants, phospholipids and macromolecules. Active ingredients could be encapsulated or adsorbed on the polymers surface [35,52,53,54,55,56].
Polymeric nanoparticles hold scientific interest, because, similarly to liposomes or niosomes, they could deliver hydrophilic substances. Nanocapsules could have inner water or oil core, within which active ingredients could be solubilized this gives higher drug encapsulation compared to nanospheres. In nanospheres, the active molecule is entrapped in matrix of polymer (Figure 2) [54,57,58].
Table 1 provides short comparison of the selected carriers.
Table 1. Brief characteristics of the selected carriers for hydrophilic pthytochemicals.
Table 1. Brief characteristics of the selected carriers for hydrophilic pthytochemicals.
GroupStructureSizePreparation/Isolation MethodsAdvantagesLimitationsReferences
LiposomesPhospholipid bilayer (SUV, LUV, GUV) or several lipid bilayers (MLV, MVV), hydrophilic core- SUV (20–200 nm)
- LUV (200 nm–1 µm)
- GUV (>1 µm)
- OLV (100 nm–1 µm)
- MLV (> 500 nm)
- MVV (>1 µm)
- film hydratation
- reverse phase evaporation
- solvent injection
- heating method
- microfluidic channel
- supercritical fluidic
- freeze-thawing
- freeze-drying
- detergent removal
- membrane extrusion
- sonication
- micro-emulsification
- dual asymmetric centrifuging
- biocompatible
- biodegradable
- high bioavailability
- low toxicity
- low immunogenicity
- both hydrophilic and hydrophobic compounds delivery
- controlled release of the of active compounds
- targeting delivery
- extension of drug half-life
- high cost of production
- low solubility
- possible instability (e.g., during storage)
- possible degradation via hydrolysis or oxidation
- possible encapsulated drug leakage
- special storage conditions are required
[32,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76]
NiosomesNon-ionic surfactant bilayer in conjunction with cholesterol- SUV (10–100 nm)
- LUV (100–3000 nm)
- MLV (>10 µm)
- thin film hydratation
- reverse phase evaporation
- ether injection (solvent)
- emulsion method
- lipid injection
- bubble method
- microfluidisation method
- supercritical reverse phase evaporation
- micelle solution and enzyme
- trans membrane pH gradient drug uptake process
- formation from proniosomes
- sonication
- lower cost of production
- biocompatible
- biodegradable
- low toxicity
- low immunogenicity
- both hydrophilic and hydrophobic compounds delivery
- improved chemical stability
- structurally flexible
- controlled release of active compounds
- targeting delivery
- special storage conditions are not required
- low physical stability
- possible degradation via hydrolysis
- possible aggregation
- possible encapsulated drug leakage
- possible aggregation
- limited shelf-life
[41,43,76,77,78,79]
ExosomesSingle lipid bilayer containing RNAs, proteins and lipids−30–200 nm- ultracentrifugation
- ultrafiltration
- size-exclusion chromatography
- polymer precipitation
- magnetic separation
- acoustic fluid separation
- immunological separation
- dielectrophoretic separation
- biocompatible
- biodegradable
- low toxicity
- low immunogenicity
- innate stability
- targeting delivery
- short-half life in circulation
- special storage conditions are required
[47,48,50,51,80,81,82,83,84]
Polymeric nanoparticlesSolid core with polymeric matrix

Inner liquid or solid core secured by polymeric shelf
- nanospheres (10–200 nm)


- nanocapsules (50–300 nm)
- dialysis
- emulsification diffusion
- interfacial polymerization
- nanoprecipitation
- phase inversion temperature
- salting out
- super critical fluid technology
- solvent evaporation
- biodegradable
- low toxicity
- high stability
- both hydrophilic and hydrophobic compounds delivery
- long shelf life
- both hydrophilic and hydrophobic compounds delivery
- controlled release of active compounds
- high loading capacity
- targeting delivery
- possible degradation
- possible monomer aggregation
[53,85,86,87,88,89]
While the aforementioned delivery platforms offer a wide range of strategies for enhancing the dermal and systemic availability of hydrophilic compounds, their effectiveness ultimately depends on the molecular characteristics of the compounds themselves. Therefore, the following section presents a detailed analysis of selected polyphenols, focusing on their chemical structure, solubility, biological activity, and formulation-specific delivery challenges. This compound-level perspective provides essential context for understanding how delivery systems are matched to molecular needs.

3. Hydrophilic Phytochemicals

Hydrophilic phytochemicals are characterized by a high content of hydroxyl (–OH) and carboxyl (–COOH) groups, which increases their water solubility but simultaneously hinders their permeability across cell membranes and limits bioavailability [90,91,92,93]. Their molecular weight (500–4000 Da), polycyclic structures, and abundance of hydrogen bond donors further reduce passive diffusion and contribute to rapid excretion and metabolic degradation. Jacob et al. [94] highlighted that additional limitations in bioavailability result from their instability under environmental conditions—exposure to light, oxygen, pH changes, and elevated temperature can lead to photodegradation, hydrolysis, and oxidation [94]. Recent studies have shown that some phenolic substances (e.g., epicatechin, chlorogenic acid, isoquercetin) are only stable at low temperatures and degrade rapidly at 40 °C [92,94,95]. The excess of hydrogen bond donor groups (–OH and –NH2) further impairs passive membrane diffusion, and their complex molecular structures (e.g., polycyclic features) promote swift elimination and metabolic breakdown [94]. They are also subjected to enzymatic degradation and interactions with microbiota and pH, necessitating the development of protective delivery systems [94,96]. In response, various strategies like advanced skin delivery systems are commonly applied [97].
Hydrophilic particles can undergo destabilization already at the stage of their extraction from plants raw materials. For instance, Khoddami et al. [98] emphasized the importance of polyphenol solubility, which depends on the number of hydroxyl groups, substituents, and degree of conjugation. The choice of solvents (ethanol, methanol, water) is crucial—high-polarity solvents improve extraction efficiency but may also cause degradation of thermolabile constituents [98,99]. As alternatives, hydrotropic solvents (e.g., sodium cumenesulfonate (NaCS), sodium n-butyl benzene sulfonate (NaNBBS)) offer high extraction efficiency, are eco-friendly, non-toxic, and reusable [100].
The application of auxiliary extraction techniques such as MAE (microwave-assisted extraction), UAE (ultrasound-assisted extraction), or EAE (enzyme-assisted extraction) increases extraction efficiency while limiting degradation of active compounds [98,99,101]. High hydrostatic pressure (HHPE) and pulsed electric field (PEF) improve cell membrane permeability, allowing cytoplasmic release without damaging compound structures [98].
Due to low membrane permeability (low value of logP) [102] and high polarity, efficient delivery of hydrophilic compounds requires the use of transport systems. The appropriate selection of surfactants, particle size, and viscosity enhances solubility in lipid environments and boosts bioavailability [103]. Hansen Solubility Parameters (HSP) are used to predict the compatibility of compounds (e.g., phenolic acids and flavonoids) with ionic liquids, emphasizing the need for supportive systems for their isolation and formulation [104].
A great advantage of hydrophilic phytochemicals is their strong biological activity. Their antioxidant and anti-inflammatory properties determine their ability to neutralize free radicals, chelate heavy metals, and modulate pro-inflammatory enzyme expression [91,103,105,106,107]. A correlation between phenolic content and antioxidant potential of the extracts has been confirmed [108,109]. Interestingly, thermal processing may even enhance the bioactivity of certain compounds [107]. Jacob et al. [94] stated that, the abundance of hydrogen bond donor groups, typical for hydrophilic compounds, limits membrane permeability but supports receptor binding and neutralization of reactive oxygen species (ROS) [94].
Hydrophilic compounds represent a large and diverse group of plant metabolites. These substances are widely distributed in natural raw materials and play important roles in various biological processes. Below, we present examples of the most common classes of hydrophilic compounds found in botanical sources, highlighting their presence in natural extracts and their potential applications in cosmetic formulations.

3.1. Flavonoids

Flavonoids constitute a vast class of plant-derived polyphenolic compounds characterized by a C6–C3–C6 skeleton, consisting of two aromatic rings linked by a three-carbon bridge forming a heterocyclic ring [110,111]. Their structure determines physicochemical properties such as solubility, polarity, biological activity, and bioavailability [7]. The hydrophilicity of flavonoids largely depends on the number and positioning of hydroxyl groups (–OH) as well as the presence of sugar moieties (glycosylation). The incorporation of glucose, rhamnose, or other sugars increases their water solubility and improves their pharmacokinetic properties, positively influencing absorption and distribution in the body [112,113,114,115,116]. Hydroxyl groups play a key role in forming hydrogen bonds with water molecules, and their location affects not only solubility but also the capacity to interact with biological receptors [110,111,117].
As mentioned above, flavonoids can occur as free aglycones or in glycosylated forms. Glycosides display higher solubility in aqueous environments, which enhances their biological availability [112,114,115]. In contrast, aglycones are less polar, which may favor their penetration through lipid membranes but simultaneously reduce their solubility in plasma. According to Journal of Immunology Research, highly hydrophilic flavonoids tend to localize near the surface of cellular membranes, where they form a hydrophilic protective layer. This localization aids in protecting membranes against oxidative stress and free radical-induced damage [116].

3.1.1. Impact of Structural Modifications on Flavonoid Hydrophilicity and Bioavailability

Flavonoid hydrophilicity—and thus their bioavailability—depends not only on the native molecular structure but also on applied chemical and biotechnological modifications. Hydroxyl groups (–OH), glycosylation, and structural transformations that increase water solubility play key roles.

3.1.2. Hydroxyl Groups, Glycosylation, and Polarity

Water solubility of flavonoids strongly depends on the number and placement of hydroxyl groups, the presence of glycosides, and structural modifications such as sulfonation, acetylation, or glycosylation [115,117,118,119,120]. As demonstrated by Heim et al. (2002), a higher number of hydroxyl groups significantly increases molecular polarity and hydrogen bonding potential, directly improving water solubility [119]. As described earlier in the context of quercetin, luteolin, and apigenin, glycosylation with sugar moieties like glucose or rhamnose enhances hydrophilicity, leading to greater water solubility and bioavailability [119]. For instance, quercetin-3-O-glucoside exhibits much higher solubility compared to its aglycone form, which directly influences its absorption and distribution in biological systems [119]. Similarly, luteolin-7-O-glucoside and glycosylated derivatives of apigenin demonstrate improved solubility and stability in aqueous environments, which enhances their biological effectiveness [119]. Despite its high number of hydroxyl groups, quercetin in its aglycone form has low water solubility (approximately 0.92 g/L at 25 °C) [115]. However, glycosylation or encapsulation in β-cyclodextrins increases its solubility and stability, forming hydrophilic–lipophilic inclusion complexes that improve bioavailability and protect against degradation [115].

3.1.3. Acetamide and Sulfate Modifications

Further chemical modifications, such as acetamide and sulfate substitutions, have been shown to enhance flavonoid hydrophilicity. Research by Isika (2022) demonstrated that replacing hydroxyl groups with acetamide moieties in flavonoids like quercetin and luteolin significantly improves their solubility and pharmacokinetic profile while retaining antioxidant properties [111]. This structural adjustment not only increases polarity but also enhances molecular stability.
Highly sulfated flavonoid derivatives, such as quercetin sulfate or (+)-catechin sulfate, exhibit extreme hydrophilicity due to their strong negative charge and elevated polarity [121]. These modifications, analyzed using capillary electrophoresis, provide enhanced solubility and biological stability in aqueous environments, offering potential for novel therapeutic applications [121].

3.1.4. Cyclodextrin Complexation

The use of cyclodextrins, particularly β-cyclodextrin, facilitates the formation of inclusion complexes, where the hydrophobic segments of flavonoid molecules are encapsulated within the hydrophilic cavity of the cyclodextrin [113]. This strategy has been successfully applied to flavonoids like baicalin, kaempferol, and quercetin, substantially improving their water solubility and stability [113]. Encapsulation not only enhances solubility but also protects these compounds from environmental degradation, thereby improving their bioavailability.

3.1.5. Membrane Localization and Hydrophilic Properties

The increased hydrophilicity of flavonoids influences their localization in biological membranes. According to Oteiza et al., more polar flavonoids tend to accumulate near the surface of cellular membranes, forming a hydrophilic layer that limits oxidative penetration and protects cellular structures from oxidative stress [116]. This is consistent with findings described for quercetin, which localizes within the lipid bilayer, enhancing its antioxidant and anti-inflammatory effects. The strategic positioning of hydrophilic flavonoids in membrane regions rich in polar phospholipids strengthens their interaction with membrane-bound receptors, amplifying their biological efficacy [113].

3.2. Quercetin

3.2.1. Structure and Solubility

Quercetin (Figure 3a) is a flavonol of a 3,3′,4′,5,7-pentahydroxyflavone structure, containing five hydroxyl groups, making it one of the most hydrophilic aglycones among flavonoids [111,114,119]. These groups facilitate numerous hydrogen bonds, influencing interactions with biological membranes and oxidative enzymes [7].

3.2.2. Biological Activity

Despite its high polarity, the aglycone form of quercetin has low water solubility—approximately 0.92 g/L at 25 °C [115]. However, glycosylated derivatives such as quercetin-3-O-glucoside show significantly higher solubility and bioavailability [115]. Encapsulation of quercetin in β-cyclodextrins enhances its solubility and stability by forming hydrophilic–lipophilic inclusion complexes, thereby improving bioavailability and protecting the compound from environmental and physiological degradation [113,122]. Quercetin is a potent antioxidant; its activity derives from the number and position of hydroxyl groups that enable neutralization of hydroxyl and peroxyl radicals [115,119,122,123]. In addition to its antioxidant properties, it inhibits inflammation-related enzymes such as COX and LOX (lipoxygenase), making it a promising agent for the prevention of metabolic and chronic inflammatory disorders [7,118,123]. Moreover, in vitro studies have shown that quercetin can suppress epidermal growth factor receptor (EGFR) activity in gastric cancer cells, indicating its potential as a targeted anticancer agent [124].
Quercetin acts as a potent antioxidant and anti-inflammatory agent, inhibits MMP activity, protects collagen, and improves skin elasticity and hydration. In vivo application of 1% quercetin after UV exposure increased glutathione levels and skin hydration, while reducing oxidative stress markers. It also inhibits tyrosinase activity, contributing to skin lightening and anti-pigmentation effects [125].

3.2.3. Delivery Barriers and Carrier-Based Strategies

Applied in w/o microemulsions, it efficiently penetrates the stratum corneum and epidermis without causing irritation, protecting against UVB-induced photodamage [126]. For instance, quercetin-loaded phospholipid vesicles modified with edge activators, such as sodium cholate, increase dermal absorption by fluidizing the lipid bilayers of the stratum corneum.

3.3. Luteolin

3.3.1. Structural Properties and Biological Functions

Luteolin is a flavone with a 3′,4′,5,7-tetrahydroxyflavone structure (Figure 3b), containing four hydroxyl groups [119,123]. Although slightly less hydrophilic than quercetin, its polarity remains substantial, affecting its solubility and biological interactions. The highest solubility of luteolin is observed in polar protic solvents—especially mixtures of water with methanol or ethanol [99,127]. Luteolin exhibits broad biological activity, including antioxidant, anti-inflammatory, and anticancer effects [128,129]. As an aglycone, however, it suffers from limited bioavailability due to its moderate solubility in water and vulnerability to degradation. Glycosides like luteolin-7-O-glucoside exhibit significantly improved solubility and chemical stability, making them more effective formulations [115,117]. Despite the advantages of glycosylation, biotransformation of luteolin into its glycoside form remains inefficient, with a yield of approximately 8%, lower than that of other flavonoids [115]. According of Dias 2021, luteolin may inhibiting protein kinase activity and reducing expression of pro-inflammatory enzymes such as cyclooxygenase-2 (COX-2) [7,130]. Luteolin reduces the expression of pro-inflammatory cytokines (IL-6, IL-8, TNF-α) and the activity of COX-2 and iNOS, alleviating psoriatic skin [30]. In nanosuspensions and bilosomes, it accumulates more effectively in the skin, showing stronger anti-inflammatory and photoprotective activity [90]. Formulas containing Pluronic F127 and alginate improve its physicochemical stability and skin permeation [131]. Recent studies have demonstrated that luteolin exhibits anti-photoaging properties by inhibiting the MAPK pathway in UVB-exposed skin cells, thereby mitigating oxidative stress and inflammation, which supports its dermatological application [132].

3.3.2. Carrier-Based Delivery Strategies

Lipid-based delivery systems have shown promise in enhancing the solubility and bioavailability of luteolin. These carriers contribute to improved physicochemical stability, prolong the compound’s residence time in the skin, and enable a more controlled release.
These advanced delivery systems represent a promising strategy for optimizing the clinical effectiveness of luteolin in dermatological and anti-inflammatory applications.

3.4. Apigenin

3.4.1. Structure and Solubility

Apigenin (4′,5,7-trihydroxyflavone) contains three hydroxyl groups (Figure 3c), making it less hydrophilic than quercetin or luteolin, yet still soluble in water–alcohol systems [112,119,123]. The hydrophilicity of apigenin can be enhanced by glycosylation and pH modulation. In acidic environments, it shows increased affinity for lipid membranes, facilitating its cellular penetration [116]. Glycosylation substantially improves the solubility of apigenin, with enzymatic methods using Staphylococcus saprophyticus strains proving effective [112]. This not only enhances the physicochemical properties of the compound but also its stability and bioavailability.

3.4.2. Biological Activity

Apigenin demonstrates cytotoxic effects against cancer cells, primarily through the induction of apoptosis and inhibition of angiogenesis, as confirmed in studies on skin melanoma [111,114,118]. Its hydroxyl groups allow it to act as both an antioxidant and a metal ion chelator, protecting cells against oxidative stress [133]. Thanks to moderate hydrophilicity and a small number of functional groups, apigenin easily penetrates biological membranes, enabling intracellular action and interaction with key molecular pathways [116,129]. Apigenin improves skin barrier function by increasing filaggrin expression, lipid synthesis, and lamellar body formation, contributing to improved skin hydration and integrity [134]. It also exhibits significant anticancer effects by inhibiting mTOR, MAPK, PI3K/Akt, and Wnt/β-catenin pathways and inducing apoptosis [135]. Formulations with carriers such as PLGA and transfersomes significantly enhance its bioavailability and skin penetration [136].
In summary, although quercetin, luteolin, and apigenin differ structurally and vary in polarity and molecular weight, a consistent trend emerges in their delivery outcomes: encapsulation in lipid-based nanocarriers improves dermal targeting by enhancing compound stability, modulating release kinetics, and facilitating efficient skin penetration. Notably, each flavonoid responds differently depending on the carrier type, suggesting the importance of customized formulation strategies to optimize therapeutic performance.

3.5. Phenolic Acids

According to Materska (2010), phenolic acids represent the most hydrophilic fraction of analyzed herbal extracts, surpassing other bioactive compound classes in solubility [137]. Their hydrophilicity is influenced not only by their intrinsic chemical structure but also by the application of carriers that enhance their solubility and bioavailability. The degree of hydrophilicity also depends on the chemical form—glycosides and esters can alter solubility and bioavailability [98]. Phenolic acids are among the most widely distributed natural phenolic compounds in plants. Their structure includes an aromatic ring—responsible for molecular stability and free radical neutralization—along with a hydroxyl group (–OH), which contributes to antioxidant activity, and a carboxyl group (–COOH), which imparts high polarity and acidic properties to the molecule [102,108,138]. Based on side chain length, phenolic acids are divided into two major subclasses: hydroxybenzoic acids derivatives (C6–C1), such as gallic, protocatechuic, vanillic, and syringic acids, and hydroxycinnamic acids derivatives (C6–C3), including caffeic, ferulic, p-coumaric, and sinapic acids [108,137,139].
The hydrophilicity of phenolic acids results from the presence of multiple hydroxyl and carboxyl groups that enable hydrogen bonding with water. As a result, these acids are readily soluble in water and polar organic solvents such as methanol, ethanol, and acetone [108,137,140]. Gallic acid, for instance, demonstrates high solubility (up to 11 g/L), while ferulic acid exhibits moderate yet significant solubility [108,137,140]. Due to their structural features, phenolic acids are primarily localized in aqueous compartments of the body (e.g., cytoplasm), where they effectively scavenge reactive oxygen species (ROS) [108,138,141].
According to Materska (2010) phenolic acids were found to represent the most hydrophilic fraction of analyzed herbal extracts, surpassing other bioactive compound classes in solubility [137]. The degree of hydrophilicity also depends on the chemical form—glycosides and esters can alter solubility and bioavailability [98].
In nature, phenolic acids exist in both free forms—easily extractable from cell sap—and bound forms such as glycosides (e.g., ferulic acid bound to sugars), esters (e.g., chlorogenic acid as an ester of caffeic and quinic acids), amides (with amino acids or polyamines), and as components of lignin and cell wall polysaccharides like arabinoxylans in cereals [98,108,137,139]. The chemical form plays a crucial role in solubility, absorption, and transport—compounds bound within the plant matrix require enzymatic or microbial hydrolysis for release [98,108]. Phenolic acids exhibit antioxidant activity—via hydrogen or electron donation to neutralize ROS—chelating activity (metal ion binding and Fenton reaction inhibition), anti-inflammatory effects (inhibition of COX and LOX enzymes), antimicrobial properties (antibacterial and antifungal), and anticancer effects (impacting cell cycle and apoptosis) [102,108,140].

3.5.1. Ferulic Acid

Ferulic acid (4-hydroxy-3-methoxycinnamic acid, C10H10O4) is a hydroxycinnamic phenolic acid containing hydroxyl (–OH), methoxy (–OCH3), and carboxyl (–COOH) groups (Figure 4a). These groups confer good hydrophilicity and hydrogen bonding capability [108,142]. It naturally occurs mainly in cereal products, often in a bound form with cell wall polysaccharides such as arabinoxylans, which significantly limits its bioavailability [108,142,143,144]. Ferulic acid is well soluble in aqueous–alcoholic solutions especially in ethanol and methanol mixed with water (typically 40%, 70%, or 80% concentrations), but only moderately soluble in pure water [98,137]. Since it commonly exists in esterified form, its extraction often requires enzymatic or alkaline hydrolysis. Although other alcohols such as propanol and butanol can be used for extraction, they are less effective for phenolic compounds due to their lower polarity compared to ethanol and methanol. Propanol shows limited efficiency in solubilizing polar phenolic compounds, while butanol is mainly applied in later stages of serial extraction and not as a primary solvent. Mixtures of ethanol or methanol with water have been found to enhance the yield of phenolic extraction significantly, optimizing both solubility and extraction efficiency [98,137].

3.5.2. Biological Activity

Its bioavailability largely depends on chemical form and food matrix. In a study by Adama et al. (2002), ferulic acid bound to dietary fiber (e.g., in whole wheat flour) showed significantly lower bioavailability (<10%) compared to its free form, which can be absorbed at over 50% [142,143,144]. It circulates and is excreted primarily as conjugated metabolites, which may exhibit different biological activity from the parent compound [108,145]. Ferulic acid shows strong antioxidant, anti-inflammatory, antimicrobial, and neuroprotective properties. It scavenges free radicals, inhibits lipid peroxidation, modulates signaling pathways (e.g., Nrf2 (nuclear factor erythroid 2-related factor 2), NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells)), and protects DNA from oxidative damage. It improves skin elasticity, protects collagen and elastin, and stabilizes vitamin C in cosmetic formulations, enhancing its effectiveness [146]. Chemical modifications, such as amino acid esters, increase its solubility and bioavailability, improving its transdermal penetration [147]. It also acts anti-inflammatory, protects DNA from damage, and inhibits angiogenesis in skin cancer cells [135]. Its potential is applied in food, cosmetic, and pharmaceutical industries [108,145].

3.5.3. Carrier-Based Delivery Strategies

To further optimize its bioavailability and skin penetration, ferulic acid has been effectively encapsulated in several carrier systems, including solid lipid nanoparticles (SLN), nanostructured lipid carriers (NLC), liposomes, and nanoemulsions [135]. Liposomal formulations enhance its solubility and facilitate deeper penetration through the stratum corneum by mimicking the phospholipid layers of cell membranes, allowing ferulic acid to integrate seamlessly into the lipid matrix of the skin [148,149,150]. Nanoemulsions, characterized by their ultra-small droplet size, further improve the diffusion of ferulic acid through the skin barrier, enhancing its absorption into deeper layers [149,150]. Lipid-based carriers like SLN and NLC not only stabilize ferulic acid but also protect it from oxidative degradation and UV-induced damage, extending its therapeutic effects. These nanoparticles reorganize the lipid structure of the stratum corneum, reducing its barrier properties and facilitating improved transdermal delivery [148,149,150]. The biphasic release profile of SLN and NLC ensures both immediate and controlled release, enhancing the compound’s retention in skin tissues and minimizing irritation [149]. Moreover, hydrogels have been explored as carriers for ferulic acid, providing sustained release and prolonged skin retention, which enhances its antioxidant and protective effects [149]. Although detailed studies on ferulic acid’s individual transdermal mechanism are limited [148], its encapsulation in carriers clearly optimizes its permeation and stability. The incorporation into solid lipid nanoparticles, liposomes, nanoemulsions, and hydrogels not only enhances its solubility but also improves its bioavailability and protective effects against oxidative stress, making it a valuable component in advanced dermatological formulations [135,148,149,150].

3.6. Caffeic Acid

3.6.1. Structure and Solubility

Caffeic acid (3,4-dihydroxycinnamic acid, C9H8O4) contains two hydroxyl groups and one carboxyl group (Figure 4b), providing it with high polarity and good solubility in water and aqueous–alcoholic solutions [102,108,137]. It is found in carrots, basil, oregano, cereals, and coffee [151,152]. After ingestion, caffeic acid is efficiently absorbed in the small intestine and subsequently metabolized in the liver (via glucuronidation, sulfation, and methylation), with bioavailability ranging from 5% to 20% depending on food source and chemical form [151,152].

3.6.2. Biological Activity

Caffeic acid exhibits antioxidant, anti-inflammatory, and anticancer activities. It also shows synergy with other polyphenols, such as chlorogenic or ferulic acids, which may enhance its biological effects [108,109,145]. Caffeic acid has chemopreventive properties against skin cancer by inhibiting ERK1/2 kinases and reducing MAPK pathway signaling and pro-inflammatory factor expression [153]. It protects skin cell DNA from UVB-induced damage by activating DNA repair proteins of the NER pathway [136]. It also shows stability and high affinity for lipid emulsions used in topical formulations [154].

3.6.3. Carrier-Based Delivery Strategies

Recent advancements in nanotechnology have significantly improved the bioavailability and transdermal delivery of caffeic acid through its encapsulation in various carrier systems, including ethosomes, hydrogels, nanoemulsions, microparticles, and liposomes [148,149,150,155,156]. Ethosomes—lipid-based vesicles enriched with ethanol—facilitate deep penetration of caffeic acid through the stratum corneum (SC) due to their flexible structure and enhanced lipid solubility. Ethanol increases the fluidity of the SC lipids, allowing ethosomes to effectively transport caffeic acid to deeper skin layers, maintaining its antioxidant properties for extended periods [155]. Another promising delivery system for caffeic acid involves hydrogels and nanoemulsions, which significantly improve its skin retention and penetration. Hydrogels enable controlled, sustained release of caffeic acid, ensuring long-lasting antioxidant protection [135]. Nanoemulsions, characterized by their ultrafine droplet size, enhance the diffusion of caffeic acid across the SC, increasing its solubility and enabling effective delivery to deeper epidermal layers [148]. Both carriers modify the lipid structure of the SC, reducing resistance to permeation and enhancing local bioavailability and stability [148,150]. Furthermore, microparticles (MPs) formulated in oil-in-water (O/W) emulsions have been investigated for their potential to target hair follicles and extend the residence time of caffeic acid in the skin [156]. MPs composed of chitosan encapsulate caffeic acid, facilitating its accumulation within hair follicles and enhancing its anti-inflammatory and antioxidant effects. This targeted delivery not only improves local bioavailability but also minimizes systemic exposure, reducing potential side effects [150]. Studies have demonstrated that MPs increase the fluidity of the SC lipids, creating microchannels that promote deeper penetration of caffeic acid and prolong its therapeutic action [156]. Additionally, liposomes have been employed to boost caffeic acid’s transdermal absorption. Their phospholipid bilayer structure mimics cell membranes, enhancing the diffusion of caffeic acid across the skin barrier and providing localized antioxidant effects. Encapsulation in liposomes also protects caffeic acid from oxidative degradation, stabilizing the compound and allowing for controlled, sustained release in topical formulations [150]. Although structural analogies with ferulic acid suggest that caffeic acid may benefit similarly from lipid-based carriers [149], direct studies are still limited. Current evidence clearly indicates that carrier-based delivery systems significantly enhance its bioavailability, stability, and therapeutic efficacy, making caffeic acid a promising candidate for advanced dermal and systemic applications.

3.7. Chlorogenic Acid

3.7.1. Structure and Solubility

Chlorogenic acid (CGA) is an ester of caffeic and quinic acids. It contains multiple hydroxyl and carboxyl groups (Figure 4c), which contribute to its high hydrophilicity. It is well soluble in water and 70% ethanol and remains stable under low pH conditions. It is found in coffee, carrots, olive oil, and whole grain products [98,137,151]. CGA is hydrolyzed in the intestine into caffeic and quinic acids, which are then absorbed and metabolized into conjugated forms. Its bioavailability is variable and influenced by fiber and lignin content in the plant matrix, as well as microbial enzyme activity [142,144]. Like its precursor caffeic acid, CGA exhibits potent antioxidant, anti-inflammatory, anticancerproperties (e.g., inhibition of digestive enzymes like α-glucosidase). Chlorogenic acid exhibits anti-inflammatory effects, increases skin hydration, and is highly soluble in water and ethanol, making it ideal for cosmetic applications.

3.7.2. Biological Activity

Its strong antioxidant effect is due to multiple hydroxyl and carboxyl groups capable of neutralizing reactive oxygen species [146].

3.7.3. Carrier-Based Delivery Strategies

Recent advancements in nanotechnology have significantly improved the bioavailability and transdermal delivery of chlorogenic acid through encapsulation in various carrier systems, including ethosomes, liposomes, nanophytovesicles (NPVs), and chlorogenic acid–hydrogenated soy phosphatidylcholine complex (CA–HSPC) [148,150,157] Ethosomes, which are lipid-based vesicles enriched with high concentrations of ethanol, effectively enhance the permeability of chlorogenic acid through the stratum corneum (SC). Ethanol increases the fluidity of the lipid bilayer, disrupting the barrier properties of the SC and allowing CGA to penetrate deeper into the epidermis. In vitro studies have demonstrated that ethosomal formulations not only improve the deposition of chlorogenic acid in the skin but also boost its antioxidant and therapeutic efficacy, especially in cosmetic and dermatological applications [148]. In addition to ethosomes, chlorogenic acid is also efficiently delivered through liposomes, which facilitate its solubility and transport across the lipid layers of the skin. Liposomal encapsulation enhances CGA’s penetration through the stratum corneum by mimicking the phospholipid arrangement of cellular membranes, leading to deeper tissue absorption [148,150]. A particularly innovative system for CGA delivery is nanophytovesicles (NPVs), which are specialized phospholipid-based carriers designed to enhance lipid solubility and skin permeability. NPVs containing Phospholipon® 90H and LIPOID® S100 create microscopic channels within the SC, significantly increasing CGA’s permeation through the lipid matrix. Phospholipon® 90H is specifically effective in enhancing stratum corneum penetration, while LIPOID® S100 facilitates deeper interactions with cellular membranes, allowing for improved bioavailability [146]. Additionally, NPVs boost lipid solubility more than 25-fold compared to the pure form of CGA, enhancing its stability and therapeutic impact. This carrier system also bypasses the hepatic first-pass effect, ensuring greater systemic availability of chlorogenic acid after transdermal application [157]. CGA encapsulated in Phospholipid Complex (CA–HSPC) has demonstrated increased stability, protection against oxidative degradation, and enhanced penetration through the stratum corneum. The HSPC (hydrogenated soy phosphatidylcholine) layer acts as a protective barrier, prolonging the release of chlorogenic acid while simultaneously improving its therapeutic effects. This controlled-release mechanism not only reduces the frequency of application but also provides sustained antioxidant protection, particularly against UVA-induced photoaging [148]. The use of these carrier systems—ethosomes, liposomes, nanophytovesicles, and phospholipid complexes—substantially improves the skin penetration, stability, and bioavailability of chlorogenic acid. By optimizing its delivery, these technologies enhance CGA’s potential for antioxidant, anti-inflammatory, and anti-aging effects in dermatological and cosmetic formulations.
Ferulic, caffeic, and chlorogenic acids, once ingested and absorbed, primarily circulate as conjugated metabolites—glucuronides and sulfates. These forms not only facilitate excretion but may also exert regulatory and protective functions depending on their biological activity and site of action [108,109,145].
The practical implications of these physicochemical characteristics and delivery mechanisms are further explored in the following section, which provides a comparative overview of their dermal and therapeutic applications.

4. Applications of Delivery Systems for Hydrophilic Phytochemicals

Building upon the structural properties and encapsulation strategies discussed in the previous section, this part of the review highlights practical applications of nanocarrier-based delivery systems for selected hydrophilic plant-derived compounds.
The table below (Table 2) presents a concise comparative overview of selected scientific publications focused on delivery systems for hydrophilic phytochemicals. The compounds summarized here, including apigenin, quercetin, luteolin, ferulic acid, chlorogenic acid, caffeic acid, curcumin, genistein, rutin, and others, are known for their antioxidant, anti-inflammatory or therapeutic properties. These phytochemicals have been encapsulated using diverse carrier systems to improve solubility, stability, skin penetration, and targeted bioavailability.
The analysis is organized according to key formulation and therapeutic aspects, including type of carrier, method of encapsulation, pharmacokinetic modifications, and targeted dermal effects. The table also expands the scope by including representative examples of compounds not discussed in detail earlier in the manuscript, thereby providing a broader view of the design and application of nanocarrier-based systems for plant-derived hydrophilic substances.
Overall, the summarized data underscores the pivotal role of nanocarriers in optimizing the dermal delivery of phytochemicals. The incorporation of lipid-based systems, polymeric matrices, and hybrid formulations enables enhanced skin permeation, greater compound stability, and effective site-specific action—underscoring their promise in transdermal therapeutic applications.

5. Summary

Advanced drug delivery systems such as liposomes, niosomes, exosomes, and polymeric nanoparticles offer a promising approach for improving the overall performance of hydrophilic compounds. They enhance its stability, bioavailability and considering its amphiphilic character, they also constitute an effective strategy for the phytochemicals transport through the stratum corneum allowing the increasing of hydrophilic molecules absorption.
While the majority of available research is descriptive and in vitro-based, a critical comparative evaluation reveals distinct advantages and limitations among the nanocarrier types. Liposomes and ethosomes are supported by the most consistent in vivo evidence for skin applications, demonstrating improved drug retention in the epidermis and superior permeation profiles compared to conventional topical formulations [175,176,177]. Ethosomes, for instance, have been shown to deliver active substances more efficiently through the stratum corneum due to their high ethanol content, which fluidizes skin lipids [178]. In contrast, solid lipid nanoparticles (SLNs) offer greater stability and control over release profiles but may face limitations in drug loading capacity and skin permeation depth [179].
Despite these advances, clinical translation remains limited. Main barriers to adoption include: (1) high production and scaling costs, particularly for complex nanocarriers like polymeric micelles or lipid-polymer hybrids; (2) regulatory uncertainty concerning safety and long-term stability; and (3) lack of standardization in preparation methods, which affects reproducibility [180,181].
Further comparative studies, particularly clinical trials, are essential to substantiate the therapeutic superiority of individual delivery platforms and to determine the most viable candidates for clinical translation in dermatological care.

Author Contributions

Data collection: A.D. and M.S., design of the study: A.D., M.S., M.A.M., E.S. and A.S.; analysis and interpretation of the data: A.D., M.S., M.A.M., E.S. and A.S.; drafting the manuscript: A.D., M.S., M.A.M., E.S. and A.S.; critical revision of the manuscript: M.A.M., E.S. and A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Ministry of Higher Education of Poland, grand no. N42/DBS/000427.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The research was created thanks to the PHC Polonium 2022 joint research project between France and Poland, entitled ‘Cosmetic potential of viticulture byproducts as novel functional ingredients for skin barrier recovery’. The authors would also like to express their gratitude to the ‘Le Studium Loire Valley Institute for Advanced Studies’ organization for its support.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Faccio, G. Plant Complexity and Cosmetic Innovation. iScience 2020, 23, 101358. [Google Scholar] [CrossRef] [PubMed]
  2. Michalak, M. Plant-Derived Antioxidants: Significance in Skin Health and the Ageing Process. Int. J. Mol. Sci. 2022, 23, 585. [Google Scholar] [CrossRef]
  3. Xie, M.; Jiang, Z.; Lin, X.; Wei, X. Application of plant extracts cosmetics in the field of anti-aging. J. Dermat. Sci. Cosmet. Technol. 2024, 1, 100014. [Google Scholar] [CrossRef]
  4. Sławińska, N.; Olas, B. Selected Seeds as Sources of Bioactive Compounds with Diverse Biological Activities. Nutrients 2022, 15, 187. [Google Scholar] [CrossRef]
  5. Kumar, A.; Nirmal, P.; Kumar, M.; Jose, A.; Tomer, V.; Oz, E.; Proestos, C.; Zeng, M.; Elobeid, T.; Sneha, K.; et al. Major Phytochemicals: Recent Advances in Health Benefits and Extraction Method. Molecules 2023, 28, 887. [Google Scholar] [CrossRef]
  6. Gonfa, Y.H.; Tessema, F.B.; Bachheti, A.; Rai, N.; Tadesse, M.G.; Singab, A.N.; Chaubey, K.K.; Bachheti, R.K. Anti-inflammatory activity of phytochemicals from medicinal plants and their nanoparticles: A review. Curr. Res. Biotechnol. 2023, 6, 100152. [Google Scholar] [CrossRef]
  7. Dias, M.C.; Pinto, D.C.G.A.; Silva, A.M.S. Plant flavonoids: Chemical characteristics and biological activity. Molecules 2021, 26, 5377. [Google Scholar] [CrossRef]
  8. Kiokias, S.; Proestos, C.; Oreopoulou, V. Phenolic acids of plant origin-a review on their antioxidant activity in vitro (O/W emulsion systems) along with their in vivo health biochemical properties. Foods 2020, 9, 534. [Google Scholar] [CrossRef] [PubMed]
  9. Kim, B.; Cho, H.E.; Moon, S.H.; Ahn, H.J.; Bae, S.; Cho, H.D.; An, S. Transdermal delivery systems in cosmetics. Biomed. Dermatol. 2020, 4, 10. [Google Scholar] [CrossRef]
  10. Cao, H.; Saroglu, O.; Karadag, A.; Diaconeasa, Z.; Zoccatelli, G.; Conte-Junior, C.A.; Gonzalez-Aguilar, G.A.; Ou, J.; Bai, W.; Zamarioli, C.M.; et al. Available technologies on improving the stability of polyphenols in food processing. Food Front. 2021, 2, 109–139. [Google Scholar] [CrossRef]
  11. Neubert, R.H.H. Mechanisms of penetration and diffusion of drugs and cosmetic actives across the human Stratum corneum. Eur. J. Pharm. Biopharm. 2024, 202, 114394. [Google Scholar] [CrossRef]
  12. Trommer, H.; Neubert, R.H.H. Overcoming the stratum corneum: The modulation of skin penetration. A review. Ski. Pharmacol. Physiol. 2006, 19, 106–121. [Google Scholar] [CrossRef]
  13. Boncheva, M. The physical chemistry of the stratum corneum lipids. Int. J. Cosmet. Sci. 2014, 36, 505–515. [Google Scholar] [CrossRef]
  14. Dayan, N. Pathways for Skin Penetration. Cosmet. Toilet. 2005, 120, 67–76. [Google Scholar]
  15. Śliwowska, A. Penetration of active subtsnces through the skin. Factors affecting this phenomenon and methods for its modification. Aesthetic Cosmetol. Med. 2020, 9, 399–405. [Google Scholar]
  16. Souto, E.B.; Fangueiro, J.F.; Fernandes, A.R.; Cano, A.; Sanchez-Lopez, E.; Garcia, M.L.; Severino, P.; Paganelli, M.O.; Chaud, M.V.; Silva, A.M. Physicochemical and biopharmaceutical aspects influencing skin permeation and role of SLN and NLC for skin drug delivery. Heliyon 2022, 8, e08938. [Google Scholar] [CrossRef] [PubMed]
  17. Lane, M.E. Skin penetration enhancers. Int. J. Pharm. 2013, 447, 12–21. [Google Scholar] [CrossRef]
  18. Barry, B.W. Mode of action of penetration enhancers in human skin. J. Control Release 1987, 6, 85–97. [Google Scholar] [CrossRef]
  19. Sikora, E.; Miastkowska, M.; Lasoń, E. Selected Skin Delivery Systems; Wydawnictwo PK: Cracov, Poland, 2020; pp. 1–118. [Google Scholar]
  20. Vitorino, C.; Sousa, J.; Pais, A. Overcoming the Skin Permeation Barrier: Challenges and Opportunities. Curr. Pharm. Des. 2015, 21, 2698–2712. [Google Scholar] [CrossRef]
  21. Adachi, K.; Shimizu, M.; Shono, F.; Funatsu, K.; Yamazaki, H. Octanol/water partition coefficients estimated using retention times in reverse-phase liquid chromatography and calculated in silico as one of the determinant factors for pharmacokinetic parameter estimations of general chemical substances. J. Toxicol. Sci. 2024, 49, 127–137. [Google Scholar] [CrossRef]
  22. Tiwari, N.; Osorio-Blanco, E.R.; Sonzogni, A.; Esporrín-Ubieto, D.; Wang, H.; Calderón, M. Nanocarriers for Skin Applications: Where Do We Stand? Angew Chem. Int. Ed. Engl. 2022, 61, e202107960. [Google Scholar] [CrossRef] [PubMed]
  23. Bruschi, M.L. Strategies to Modify the Drug Release from Pharmaceutical Systems, 1st ed.; Woodhead Publishing: Cambridge, UK, 2015; pp. 87–194. [Google Scholar]
  24. Mojumdar, E.H.; Gooris, G.S.; Groen, D.; Barlow, D.J.; Lawrence, M.J.; Demé, B.; Bouwstra, J.A. Stratum corneum lipid matrix: Location of acyl ceramide and cholesterol in the unit cell of the long periodicity phase. Biochim. Biophys. Acta Biomembr. 2016, 1858, 1926–1934. [Google Scholar] [CrossRef] [PubMed]
  25. Deng, J.; Yang, H.; Capanoglu, E.; Cao, H.; Xiao, J. Technological aspects and stability of polyphenols. In Polyphenols: Properties, Recovery, and Applications, 1st ed.; Galanakis, C.M., Ed.; Woodhead Publishing: Cambridge, UK, 2018; pp. 295–323. [Google Scholar]
  26. Parisi, O.I.; Puoci, F.; Restuccia, D.; Farina, G.; Iemma, F.; Picci, N. Polyphenols and Their Formulations: Different Strategies to Overcome the Drawbacks Associated with Their Poor Stability and Bioavailability. In Polyphenols in Human Health and Disease; Watson, R.R., Preedy, V.R., Zibadi, S., Eds.; Academic Press: Cambridge, MA, USA, 2013; Volume 1, pp. 29–45. [Google Scholar]
  27. Wahyuningsih, S.; Wulandari, L.; Wartono, M.W.; Munawaroh, H.; Ramelan, A.H. The Effect of pH and Color Stability of Anthocyanin on Food Colorant. IOP Conf. Ser. Mater. Sci. Eng. 2017, 193, 012047. [Google Scholar] [CrossRef]
  28. Chua, L.S.; Thong, H.Y.; Soo, J. Effect of pH on the extraction and stability of anthocyanins from jaboticaba berries. Food Chem. Adv. 2024, 5, 100835. [Google Scholar] [CrossRef]
  29. Pina, F.; Oliveira, J.; De Freitas, V. Anthocyanins and derivatives are more than flavylium cations. Tetrahedron 2015, 71, 3107–3114. [Google Scholar] [CrossRef]
  30. Farhan, M. The Promising Role of Polyphenols in Skin Disorders. Molecules 2024, 29, 865. [Google Scholar] [CrossRef]
  31. Soni, V.; Chandel, S.; Jain, P.; Asati, S. Role of liposomal drug-delivery system in cosmetics. In Nanobiomaterials in Galenic Formulations and Cosmetics; Grumezescu, A.M., Ed.; Elsevier: Amsterdam, The Netherlands, 2016; pp. 93–120. [Google Scholar]
  32. Rahimpour, Y.; Hamishehkar, H. Liposomes in cosmeceutics. Expert Opin. Drug Deliv. 2012, 9, 443–455. [Google Scholar] [CrossRef]
  33. Gupta, V.; Mohapatra, S.; Mishra, H.; Farooq, U.; Kumar, K.; Iqbal, Z. Nanotechnology in Cosmetics and Cosmeceuticals—A Review. Gels 2022, 8, 173. [Google Scholar] [CrossRef]
  34. Salvioni, L.; Morelli, L.; Ochoa, E.; Labra, M.; Fiandra, L.; Palugan, L.; Prosperi, D.; Colombo, M. The emerging role of nanotechnology in skincare. Adv. Colloid Interface Sci. 2021, 293, 102437. [Google Scholar] [CrossRef]
  35. Trucillo, P. Drug Carriers: Classification, Administration, Release Profiles, and Industrial Approach. Processes 2021, 9, 470. [Google Scholar] [CrossRef]
  36. Ahmadi Ashtiani, H.R.; Bishe, P.; Lashgari, N.A.; Nilforoushzadeh, M.A.; Zare, S. Liposomes in Cosmetics. J. Ski. Stem Cell 2016, 3, e65815. [Google Scholar] [CrossRef]
  37. Liu, P.; Chen, G.; Zhang, J. A Review of Liposomes as a Drug Delivery System: Current Status of Approved Products, Regulatory Environments, and Future Perspectives. Molecules 2022, 27, 1372. [Google Scholar] [CrossRef] [PubMed]
  38. 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] [PubMed]
  39. Šturm, L.; Ulrih, N.P. Basic methods for preparation of liposomes and studying their interactions with different compounds, with the emphasis on polyphenols. Int. J. Mol. Sci. 2021, 22, 6547. [Google Scholar] [CrossRef]
  40. Mawazi, S.M.; Ann, T.J.; Widodo, R.T. Application of Niosomes in Cosmetics: A Systematic Review. Cosmetics 2022, 9, 127. [Google Scholar] [CrossRef]
  41. Liga, S.; Paul, C.; Moacă, E.-A.; Péter, F. Niosomes: Composition, Formulation Techniques, and Recent Progress as Delivery Systems in Cancer Therapy various. Pharmaceutics 2024, 16, 223. [Google Scholar] [CrossRef]
  42. Lens, M. Niosomes as Vesicular Nanocarriers in Cosmetics: Characterisation, Development and Efficacy. Pharmaceutics 2025, 17, 287. [Google Scholar] [CrossRef] [PubMed]
  43. Karim, K.; Mandal, A.; Biswas, N.; Guha, A.; Chatterjee, S.; Behera, M.; Kuotsu, K. Niosome: A future of targeted drug delivery systems. J. Adv. Pharm. Technol. Res. 2010, 1, 374–380. [Google Scholar]
  44. Hazira, R.M.N.; Reddy, M.S. Niosomes: A nanocarrier drug delivery system. GSC Biol. Pharm. Sci. 2023, 22, 120–127. [Google Scholar] [CrossRef]
  45. Chen, Y.F.; Luh, F.; Ho, Y.S.; Yen, Y. Exosomes: A review of biologic function, diagnostic and targeted therapy applications, and clinical trials. J. Biomed. Sci. 2024, 31, 67. [Google Scholar] [CrossRef]
  46. Frydrychowicz, M.; Kolecka-Bednarczyk, A.; Madejczyk, M.; Yasar, S.; Dworacki, G. Exosomes-structure, biogenesis and biological role in non-small-cell lung cancer. Scand. J. Immunol. 2015, 81, 2–10. [Google Scholar] [CrossRef] [PubMed]
  47. Rajput, A.; Varshney, A.; Bajaj, R.; Pokharkar, V. Exosomes as New Generation Vehicles for Drug Delivery: Biomedical Applications and Future Perspectives. Molecules 2022, 27, 7289. [Google Scholar] [CrossRef]
  48. Isola, A.L.; Chen, S. Exosomes: The Messengers of Health and Disease. Curr. Neuropharmacol. 2017, 15, 157–165. [Google Scholar] [CrossRef]
  49. Fan, T.; Sun, N.; He, J. Exosome-Derived LncRNAs in Lung Cancer. Front. Oncol. 2020, 10, 1728. [Google Scholar] [CrossRef] [PubMed]
  50. Zhang, Y.; Bi, J.; Huang, J.; Tang, Y.; Du, S.; Li, P. Exosome: A review of its classification, isolation techniques, storage, diagnostic and targeted therapy applications. Int. J. Nanomed. 2020, 15, 6917–6934. [Google Scholar] [CrossRef]
  51. Chen, Z.; Xiong, M.; Tian, J.; Song, D.; Duan, S.; Zhang, L. Encapsulation and assessment of therapeutic cargo in engineered exosomes: A systematic review. J. Nanobiotechnol. 2024, 22, 18. [Google Scholar] [CrossRef]
  52. Elmowafy, M.; Shalaby, K.; Elkomy, M.H.; Alsaidan, O.A.; Gomaa, H.A.M.; Abdelgawad, M.A.; Mostafa, E.M. Polymeric Nanoparticles for Delivery of Natural Bioactive Agents: Recent Advances and Challenges. Polymers 2023, 15, 1123. [Google Scholar] [CrossRef] [PubMed]
  53. Zielińska, A.; Carreiró, F.; Oliveira, A.M.; Neves, A.; Pires, B.; Nagasamy Venkatesh, D.; Durazzo, A.; Lucarini, M.; Eder, P.; Silva, A.M.; et al. Polymeric Nanoparticles: Production, Characterization, Toxicology and Ecotoxicology. Molecules 2020, 25, 3731. [Google Scholar] [CrossRef]
  54. Deng, S.; Gigliobianco, M.R.; Censi, R.; Di Martino, P. Polymeric nanocapsules as nanotechnological alternative for drug delivery system: Current status, challenges and opportunities. Nanomaterials 2020, 10, 847. [Google Scholar] [CrossRef]
  55. Thakur, R.; Sharma, A.; Arora, V. Nanoparticles Methods for Hydrophobic Drugs—A Novel Approach: Graphical Abstract. Mater. Open 2023, 1, 2350002. [Google Scholar] [CrossRef]
  56. Kothamasu, P.; Kanumur, H.; Ravur, N.; Maddu, C.; Parasuramrajam, R.; Thangavel, S. Nanocapsules: The weapons for novel drug delivery systems. BioImpacts 2012, 2, 71–81. [Google Scholar]
  57. Arpicco, S.; Battaglia, L.; Brusa, P.; Cavalli, R.; Chirio, D.; Dosio, F. Recent studies on the delivery of hydrophilic drugs in nanoparticulate systems. J. Drug Deliv. Sci. Technol. 2016, 32, 298–312. [Google Scholar] [CrossRef]
  58. Neophytou, C.M.; Constantinou, A.I. Drug Delivery Innovations for Enhancing the Anticancer Potential of Vitamin E Isoforms and Their Derivatives. BioMed Res. Int. 2015, 2015, 584862. [Google Scholar] [CrossRef] [PubMed]
  59. Dymek, M.; Sikora, E. Liposomes as biocompatible and smart delivery systems–the current state. Adv. Colloid Interface Sci. 2022, 309, 102757. [Google Scholar] [CrossRef] [PubMed]
  60. Sforzi, J.; Palagi, L.; Aime, S. Liposome-based bioassays. Biology 2020, 9, 202. [Google Scholar] [CrossRef] [PubMed]
  61. Chaves, M.A.; Ferreira, L.S.; Baldino, L.; Pinho, S.C.; Reverchon, E. Current Applications of Liposomes for the Delivery of Bioactives: A Review on the Encapsulation of Vitamins. Nanomaterials 2023, 13, 1557. [Google Scholar] [CrossRef]
  62. Luiz, H.; Oliveira Pinho, J.; Gaspar, M.M. Advancing Medicine with Lipid-Based Nanosystems—The Successful Case of Liposomes. Biomedicines 2023, 11, 435. [Google Scholar] [CrossRef]
  63. Barba, A.A.; Bochicchio, S.; Dalmoro, A.; Lamberti, G. Lipid delivery systems for nucleic-acid-based-drugs: From production to clinical applications. Pharmaceutics 2019, 11, 360. [Google Scholar] [CrossRef]
  64. Riccardi, D.; Baldino, L.; Reverchon, E. Liposomes, transfersomes and niosomes: Production methods and their applications in the vaccinal field. J. Transl. Med. 2024, 22, 339. [Google Scholar] [CrossRef]
  65. Effiong, D.E.; Uwah, T.O.-O.; Udofa, E. Nanotechnology in Cosmetics: Basics, Current Trends and Safety Concerns—A Review. Adv. Nanopart. 2019, 9, 1–22. [Google Scholar]
  66. Azadi, Y.; Ahmadpour, E.; Ahmadi, A. Targeting Strategies in Therapeutic Applications of Toxoplasmosis: Recent Advances in Liposomal Vaccine Delivery Systems. Curr. Drug Targets 2019, 21, 541–558. [Google Scholar] [CrossRef]
  67. Falciani, C.; Accardo, A.; Brunetti, J.; Tesauro, D.; Lelli, B.; Pini, A.; Bracci, L.; Morelli, G. Target-Selective Drug Delivery through Liposomes Labeled with Oligobranched Neurotensin Peptides. ChemMedChem 2011, 6, 678–685. [Google Scholar] [CrossRef]
  68. Giofrè, S.; Renda, A.; Sesana, S.; Formicola, B.; Vergani, B.; Leone, B.E.; Denti, V.; Paglia, G.; Gropusso, S.; Romeo, V.; et al. Dual Functionalized Liposomes for Selective Delivery of Poorly Soluble Drugs to Inflamed Brain Regions. Pharmaceutics 2022, 14, 2402. [Google Scholar] [CrossRef] [PubMed]
  69. Martin, G.P.; Lloyd, A.W. Basic Principles of Liposomes for Drug Use. In Liposome Dermatics; Braun-Falco, I., Korting, H.C., Maibach, H.I., Eds.; Griesbach Conference; Springer: Berlin/Heidelberg, Germany, 1992; pp. 20–26. [Google Scholar]
  70. Honda, M.; Asai, T.; Oku, N.; Araki, Y.; Tanaka, M.; Ebihara, N. Liposomes and nanotechnology in drug development: Focus on ocular targets. Int. J. Nanomed. 2013, 8, 495–504. [Google Scholar] [CrossRef] [PubMed]
  71. Maja, L.; Željko, K.; Mateja, P. Sustainable technologies for liposome preparation. J. Supercrit. Fluids 2020, 165, 104984. [Google Scholar] [CrossRef]
  72. Gatto, M.S.; Johnson, M.P.; Najahi-Missaoui, W. Targeted Liposomal Drug Delivery: Overview of the Current Applications and Challenges. Life 2024, 14, 672. [Google Scholar] [CrossRef]
  73. Enaru, B.; Fărcaş, A.; Stănilă, A.; Socaci, S.; Diaconeasa, Z. Novel Methods for Liposome Formulation: Advancements and Innovations. Bull. Univ. Agric. Sci. Vet. Med. Cluj-Napoca Food Sci. Technol. 2023, 80, 1–13. [Google Scholar] [CrossRef]
  74. Andra, V.V.S.N.L.; Pammi, S.V.N.; Bhatraju, L.V.K.P.; Ruddaraju, L.K. A Comprehensive Review on Novel Liposomal Methodologies, Commercial Formulations, Clinical Trials and Patents. BioNanoScience 2022, 12, 274–291. [Google Scholar] [CrossRef]
  75. Gunda, R.K.; Kumar, J.N.S.; Bhargavi, G.; Bhavani, S.P.; Sandhya, B.; Padmaja, K.; Praveen, S. A Review on Formulation and Evaluation of Liposomal Drugs. Br. J. Multidiscip. Adv. Stud. 2023, 4, 31–44. [Google Scholar] [CrossRef]
  76. Kaul, S.; Gulati, N.; Verma, D.; Mukherjee, S.; Nagaich, U. Role of Nanotechnology in Cosmeceuticals: A Review of Recent Advances. J. Pharm. 2018, 2018, 3420204. [Google Scholar] [CrossRef]
  77. Moammeri, A.; Chegeni, M.M.; Sahrayi, H.; Ghafelehbashi, R.; Memarzadeh, F.; Mansouri, A.; Akbarzadeh, I.; Abtahi, M.S.; Hejabi, F.; Ren, Q. Current advances in niosomes applications for drug delivery and cancer treatment. Mater. Today Bio 2023, 23, 100837. [Google Scholar] [CrossRef] [PubMed]
  78. Kauslya, A.; Borawake, P.D.; Shinde, J.V.; Chavan, R.S. Niosomes: A Novel Carrier Drug Delivery System. J. Drug Deliv. Ther. 2021, 11, 162–170. [Google Scholar] [CrossRef]
  79. Sharma, S.; Sharma, D.; Thakur, P.; Devi, P.; Sharma, M.D. A Comprehensive Review on Niosomes; Targeted Drug Delivery System. YMER 2024, 22, 1969–1987. [Google Scholar]
  80. Bai, G.; Truong, T.M.; Pathak, G.N.; Benoit, L.; Rao, B. Clinical applications of exosomes in cosmetic dermatology. Skin Health Dis. 2024, 4, e348. [Google Scholar] [CrossRef]
  81. Thakur, A.; Shah, D.; Rai, D.; Parra, D.C.; Pathikonda, S.; Kurilova, S.; Cili, A. Therapeutic Values of Exosomes in Cosmetics, Skin Care, Tissue Regeneration, and Dermatological Diseases. Cosmetics 2023, 10, 65. [Google Scholar] [CrossRef]
  82. Gurung, S.; Perocheau, D.; Touramanidou, L.; Baruteau, J. The exosome journey: From biogenesis to uptake and intracellular signalling. Cell Commun. Signal. 2021, 19, 47. [Google Scholar] [CrossRef]
  83. Patil, S.M.; Sawant, S.S.; Kunda, N.K. Exosomes as drug delivery systems: A brief overview and progress update. Eur. J. Pharm. Biopharm. 2020, 154, 259–269. [Google Scholar] [CrossRef] [PubMed]
  84. Palakurthi, S.S.; Shah, B.; Kapre, S.; Charbe, N.; Immanuel, S.; Pasham, S.; Thalla, M.; Jain, A.; Palakurthi, S. A comprehensive review of challenges and advances in exosome-based drug delivery systems. Nanoscale Adv. 2024, 6, 5803–5826. [Google Scholar] [CrossRef]
  85. Verma, G.; Rajagopalan, M.D.; Valluru, R.; Sridhar, K.A. Chapter 7—Nanoparticles: A Novel Approach to Target Tumors. In Nano- and Microscale Drug Delivery Systems; Grumezescu, A.M., Ed.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 113–129. [Google Scholar]
  86. Yadav, H.K.S.; Almokdad, A.A.; Shaluf, S.I.M.; Debe, M.S. Polymer-Based Nanomaterials for Drug-Delivery Carriers. In Nanocarriers for Drug Delivery: Nanoscience and Nanotechnology in Drug Delivery; Mohapatra, S.S., Ranjan, S., Dasgupta, N., Mishra, R.K., Thomas, S., Eds.; Elsevier: Amsterdam, The Netherlands, 2018; pp. 531–556. [Google Scholar]
  87. Maqbool, I.; Noreen, S. A Review of Novel Techniques for Nanoparticles Preparation. Glob. Drug Des. Dev. Rev. 2019, 4, 41–50. [Google Scholar] [CrossRef]
  88. Pereira, A.S.; Daniel-da-Silva, A.L.; Trindade, T. From nanoparticles to nanocomposites: A brief overview. In Nanocomposite Particles for Bio-Applications: Materials and Bio-Interfaces; Trindade, T., Daniel da Silva, A.L., Eds.; Jenny Stanford Publishing: Singapore, 2011; pp. 1–20. [Google Scholar]
  89. Pisal, M.; Barbade, P.; Dudhal, S. Nanocapsule. Int. J. Pharm. Sci. Rev. Res. 2020, 60, 53–62. [Google Scholar]
  90. Ozkan, G.; Ceyhan, T.; Çatalkaya, G.; Rajan, L.; Ullah, H.; Daglia, M.; Capanoglu, E. Encapsulated phenolic compounds: Clinical efficacy of a novel delivery method. Phytochem. Rev. 2024, 23, 781–819. [Google Scholar] [CrossRef]
  91. Gutiérrez-Grijalva, E.P.; Picos-Salas, M.A.; Leyva-López, N.; Criollo-Mendoza, M.S.; Vazquez-Olivo, G.; Heredia, J.B. Flavonoids and phenolic acids from Oregano: Occurrence, biological activity and health benefits. Plants 2018, 7, 2. [Google Scholar] [CrossRef]
  92. 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]
  93. Gugleva, V.; Ivanova, N.; Sotirova, Y.; Andonova, V. Dermal drug delivery of phytochemicals with phenolic structure via lipid-based nanotechnologies. Pharmaceuticals 2021, 14, 837. [Google Scholar] [CrossRef] [PubMed]
  94. 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. [Google Scholar] [CrossRef] [PubMed]
  95. McClements, D.J.; Decker, E.A.; Weiss, J. Emulsion-based delivery systems for lipophilic bioactive components. J. Food Sci. 2007, 72, 109–124. [Google Scholar] [CrossRef]
  96. Mouhid, L.; Corzo-Martínez, M.; Torres, C.; Vázquez, L.; Reglero, G.; Fornari, T.; Ramírez de Molina, A. Improving in vivo efficacy of bioactive molecules: An overview of potentially antitumor phytochemicals and currently available lipid-based delivery systems. J. Oncol. 2017, 2017, 7351976. [Google Scholar] [CrossRef]
  97. McClements, D.J. Encapsulation, protection, and delivery of bioactive proteins and peptides using nanoparticle and microparticle systems: A review. Adv. Colloid Interface Sci. 2018, 253, 1–22. [Google Scholar] [CrossRef]
  98. Khoddami, A.; Wilkes, M.A.; Roberts, T.H. Techniques for analysis of plant phenolic compounds. Molecules 2013, 18, 2328–2375. [Google Scholar] [CrossRef]
  99. Alara, O.R.; Abdurahman, N.H.; Ukaegbu, C.I. Extraction of phenolic compounds: A review. Curr. Res. Food Sci. 2021, 4, 200–214. [Google Scholar] [CrossRef]
  100. Nagarajan, J.; Heng, W.W.; Galanakis, C.M.; Nagasundara Ramanan, R.; Raghunandan, M.E.; Sun, J.; Ismail, A.; Beng-Ti, T.; Prasad, K.N. Extraction of phytochemicals using hydrotropic solvents. Sep. Sci. Technol. 2016, 51, 1151–1165. [Google Scholar] [CrossRef]
  101. Zhang, L.; McClements, D.J.; Wei, Z.; Wang, G.; Liu, X.; Liu, F. Delivery of synergistic polyphenol combinations using biopolymer-based systems: Advances in physicochemical properties, stability and bioavailability. Crit. Rev. Food Sci. Nutr. 2020, 60, 2083–2097. [Google Scholar] [CrossRef]
  102. Li, Z.; Jiang, H.; Xu, C.; Gu, L. A review: Using nanoparticles to enhance absorption and bioavailability of phenolic phytochemicals. Food Hydrocoll. 2015, 43, 153–164. [Google Scholar] [CrossRef]
  103. Vieira da Silva, B.; Barreira, J.C.M.; Oliveira, M.B.P.P. Natural phytochemicals and probiotics as bioactive ingredients for functional foods: Extraction, biochemistry and protected-delivery technologies. Trends Food Sci. Technol. 2016, 50, 144–158. [Google Scholar] [CrossRef]
  104. Rahman, N.R.A.; Yunus, N.A.; Mustaffa, A.A. Selection of optimum ionic liquid solvents for flavonoid and phenolic acids extraction. IOP Conf. Ser. Mater. Sci. Eng. 2017, 206, 012061. [Google Scholar] [CrossRef]
  105. Tungmunnithum, D.; Thongboonyou, A.; Pholboon, A.; Yangsabai, A. Flavonoids and Other Phenolic Compounds from Medicinal Plants for Pharmaceutical and Medical Aspects: An Overview. Medicines 2018, 5, 93. [Google Scholar] [CrossRef]
  106. Frezza, C.; Venditti, A.; Serafini, M.; Bianco, A. Chapter 4—Phytochemistry, Chemotaxonomy, Ethnopharmacology, and Nutraceutics of Lamiaceae. In Studies in Natural Products Chemistry, 1st ed.; Atta-ur-Rahman, Ed.; Elsevier: Amsterdam, The Netherlands, 2019; Volume 62, pp. 125–178. [Google Scholar]
  107. Roy, M.K.; Juneja, L.R.; Isobe, S.; Tsushida, T. Steam processed broccoli (Brassica oleracea) has higher antioxidant activity in chemical and cellular assay systems. Food Chem. 2009, 114, 263–269. [Google Scholar] [CrossRef]
  108. Kumar, N.; Goel, N. Phenolic acids: Natural versatile molecules with promising therapeutic applications. Biotechnol. Rep. 2019, 24, e00370. [Google Scholar] [CrossRef]
  109. Tang, Y.; Tsao, R. Phytochemicals in quinoa and amaranth grains and their antioxidant, anti-inflammatory, and potential health beneficial effects: A review. Mol. Nutr. Food Res. 2017, 61, 1600767. [Google Scholar] [CrossRef]
  110. Tripoli, E.; La Guardia, M.; Giammanco, S.; Di Majo, D.; Giammanco, M. Citrus flavonoids: Molecular structure, biological activity and nutritional properties: A review. Food Chem. 2007, 104, 466–479. [Google Scholar] [CrossRef]
  111. Isika, D.K.; Sadik, O.A. Selective Structural Derivatization of Flavonoid Acetamides Significantly Impacts Their Bioavailability and Antioxidant Properties. Molecules 2022, 27, 8133. [Google Scholar] [CrossRef] [PubMed]
  112. Chu, J.; Wu, X.; Li, B.; He, B. Efficient glucosylation of flavonoids by organic solvent-tolerant Staphylococcus saprophyticus CQ16 in aqueous hydrophilic media. J. Mol. Catal. B Enzym. 2014, 99, 8–13. [Google Scholar] [CrossRef]
  113. Park, S. Cyclic glucans enhance solubility of bioavailable flavonoids. Molecules 2016, 21, 1556. [Google Scholar] [CrossRef]
  114. Ullah, A.; Munir, S.; Badshah, S.L.; Khan, N.; Ghani, L.; Poulson, B.G.; Emwas, A.-H.; Jaremko, M. Important flavonoids and their role as a therapeutic agent. Molecules 2020, 25, 5243. [Google Scholar] [CrossRef]
  115. Plaza, M.; Pozzo, T.; Liu, J.; Gulshan Ara, K.Z.; Turner, C.; Nordberg Karlsson, E. Substituent effects on in vitro antioxidizing properties, stability, and solubility in flavonoids. J. Agric. Food Chem. 2014, 62, 3321–3333. [Google Scholar] [CrossRef]
  116. Oteiza, P.I.; Erlejman, A.G.; Verstraeten, S.V.; Keen, C.L.; Fraga, C.G. Flavonoid-membrane interactions: A protective role of flavonoids at the membrane surface? Clin. Dev. Immunol. 2005, 12, 19–27. [Google Scholar] [CrossRef]
  117. Xie, L.; Deng, Z.; Zhang, J.; Dong, H.; Wang, W.; Xing, B.; Liu, X. Comparison of Flavonoid O-Glycoside, C-Glycoside and Their Aglycones on Antioxidant Capacity and Metabolism during In Vitro Digestion and In Vivo. Foods 2022, 11, 882. [Google Scholar] [CrossRef]
  118. Chen, S.; Wang, X.; Cheng, Y.; Gao, H.; Chen, X. A Review of Classification, Biosynthesis, Biological Activities and Potential Applications of Flavonoids. Molecules 2023, 28, 4982. [Google Scholar] [CrossRef] [PubMed]
  119. Heim, K.E.; Tagliaferro, A.R.; Bobilya, D.J. Flavonoid antioxidants: Chemistry, metabolism and structure-activity relationships. J. Nutr. Biochem. 2002, 13, 572–584. [Google Scholar] [CrossRef]
  120. Han, F.; Xiao, Y.; Lee, I.S. Microbial transformation of prenylquercetins by Mucor hiemalis. Molecules 2020, 25, 528. [Google Scholar] [CrossRef]
  121. Dantuluri, M.; Gunnarsson, G.T.; Riaz, M.; Nguyen, H.; Desai, U.R. Capillary electrophoresis of highly sulfated flavanoids and flavonoids. Anal. Biochem. 2005, 336, 316–322. [Google Scholar] [CrossRef]
  122. Tripoli, E.; Giammanco, M.; Tabacchi, G.; Di Majo, D.; Giammanco, S.; La Guardia, M. The phenolic compounds of olive oil: Structure, biological activity and beneficial effects on human health. Nutr. Res. Rev. 2005, 18, 98–113. [Google Scholar] [CrossRef] [PubMed]
  123. Kumar, S.; Pandey, A.K. Chemistry and biological activities of flavonoids: An overview. Sci. World J. 2013, 2013, 162750. [Google Scholar] [CrossRef] [PubMed]
  124. Liu, Y.; Li, Y.; Jiang, Y.; Zheng, X.; Wang, T.; Li, J.; Zhang, B.; Zhu, J.; Wei, X.; Huang, R.; et al. Quercetin Promotes Apoptosis of Gastric Cancer Cells through the EGFR-ERK Signaling Pathway. J. Food Biochem. 2024, 2024, 9945178. [Google Scholar] [CrossRef]
  125. Hatahet, T.; Morille, M.; Hommoss, A.; Devoisselle, J.M.; Müller, R.H.; Bégu, S. Quercetin topical application, from conventional dosage forms to nanodosage forms. Eur. J. Pharm. Biopharm. 2016, 108, 41–53. [Google Scholar] [CrossRef]
  126. Vicentini, F.T.M.C.; Simi, T.R.M.; Del Ciampo, J.O.; Wolga, N.O.; Pitol, D.L.; Iyomasa, M.M.; Bentley, M.V.L.B.; Fonseca, M.J.V. Quercetin in w/o microemulsion: In vitro and in vivo skin penetration and efficacy against UVB-induced skin damages evaluated in vivo. Eur. J. Pharm. Biopharm. 2008, 69, 948–957. [Google Scholar] [CrossRef]
  127. Rajhard, S.; Hladnik, L.; Vicente, F.A.; Srčič, S.; Grilc, M.; Likozar, B. Solubility of luteolin and other polyphenolic compounds in water, nonpolar, polar aprotic and protic solvents by applying FTIR/HPLC. Processes 2021, 9, 1952. [Google Scholar] [CrossRef]
  128. Liga, S.; Paul, C.; Péter, F. Flavonoids: Overview of Biosynthesis, Biological Activity, and Current Extraction Techniques. Plants 2023, 12, 2732. [Google Scholar] [CrossRef]
  129. Halevas, E.G.; Avgoulas, D.I.; Katsipis, G.; Pantazaki, A.A. Flavonoid-liposomes formulations: Physico-chemical characteristics, biological activities and therapeutic applications. Eur. J. Med. Chem. Rep. 2022, 5, 100059. [Google Scholar] [CrossRef]
  130. Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef]
  131. Elmowafy, M.; Shalaby, K.; Al-Sanea, M.M.; Hendawy, O.M.; Salama, A.; Ibrahim, M.F.; Ghoneim, M.M. Influence of stabilizer on the development of luteolin nanosuspension for cutaneous delivery: An in vitro and in vivo evaluation. Pharmaceutics 2021, 13, 1812. [Google Scholar] [CrossRef] [PubMed]
  132. Mu, J.; Ma, H.; Chen, H.; Zhang, X.; Ye, M. Luteolin Prevents UVB-Induced Skin Photoaging Damage by Modulating SIRT3/ROS/MAPK Signaling: An in vitro and in vivo Studies. Front. Pharmacol. 2021, 12, 728261. [Google Scholar] [CrossRef] [PubMed]
  133. Karak, P. Biological Activities of Flavonoids: An Overview. Int. J. Pharm. Sci. Res. 2019, 10, 1567–1574. [Google Scholar]
  134. Hou, M.; Sun, R.; Hupe, M.; Kim, P.; Park, K.; Crumrine, D.; Lin, T.-K.; Santiago, J.; Mauro, T.; Elias, P.; et al. Topical Apigenin Improves Epidermal Permeability Barrier Homeostasis in Normal Murine Skin by Divergent Mechanisms. Exp. Dermatol. 2014, 22, 210–215. [Google Scholar] [CrossRef]
  135. Singh, H.; Mishra, A.K.; Mohanto, S.; Kumar, A.; Mishra, A.; Amin, R.; Darwin, C.R.; Emran, T.B. A recent update on the connection between dietary phytochemicals and skin cancer: Emerging understanding of the molecular mechanism. Ann. Med. Surg. 2024, 85, 104790. [Google Scholar] [CrossRef]
  136. Abd-El-Aziz, N.M.; Hifnawy, M.S.; Lotfy, R.A.; Younis, I.Y. LC/MS/MS and GC/MS/MS metabolic profiling of Leontodon hispidulus, in vitro and in silico anticancer activity evaluation targeting hexokinase 2 enzyme. Sci. Rep. 2024, 14, 6872. [Google Scholar] [CrossRef]
  137. Materska, M. Evaluation of the Lipophilicity and Stability. Acta Sci. Pol. Technol. Aliment 2010, 9, 61–69. [Google Scholar]
  138. Petruczynik, A.; Waksmundzka-Hajnos, M. Application of hydrophilic interaction chromatography in phytochemical analysis. Acta Chromatogr. 2013, 25, 1–25. [Google Scholar] [CrossRef]
  139. Avinash, G.; Sharma, N.; Prasad, K.R.; Kaur, R.; Singh, G.; Pagidipala, N.; Thulasinathan, T. Unveiling the distribution of free and bound phenolic acids, flavonoids, anthocyanins, and proanthocyanidins in pigmented and non-pigmented rice genotypes. Front. Plant Sci. 2024, 15, 1324825. [Google Scholar] [CrossRef]
  140. Casadey, R.; Broglia, M.; Barbero, C.; Criado, S.; Rivarola, C. Controlled release systems of natural phenolic antioxidants encapsulated inside biocompatible hydrogels. React. Funct. Polym. 2020, 156, 104729. [Google Scholar] [CrossRef]
  141. Konczak, I.; Zabaras, D.; Dunstan, M.; Aguas, P. Antioxidant capacity and hydrophilic phytochemicals in commercially grown native Australian fruits. Food Chem. 2010, 123, 1048–1054. [Google Scholar] [CrossRef]
  142. Adam, A.; Crespy, V.; Levrat-Verny, M.A.; Leenhardt, F.; Leuillet, M.; Demigné, C.; Rémésy, C. The bioavailability of ferulic acid is governed primarily by the food matrix rather than its metabolism in intestine and liver in rats. J. Nutr. 2002, 132, 1962–1968. [Google Scholar] [CrossRef] [PubMed]
  143. Amara, A.; Frainay, C.; Jourdan, F.; Naake, T.; Neumann, S.; Novoa-del-Toro, E.M.; Salek, R.M.; Salzer, L.; Scharfenberg, S.; Witting, M. Networks and Graphs Discovery in Metabolomics Data Analysis and Interpretation. Front. Mol. Biosci. 2022, 9, 841373. [Google Scholar] [CrossRef] [PubMed]
  144. Zhao, Z.; Moghadasian, M.H. Chemistry, natural sources, dietary intake and pharmacokinetic properties of ferulic acid: A review. Food Chem. 2008, 109, 691–702. [Google Scholar] [CrossRef] [PubMed]
  145. Chaudhary, A.; Jaswal, V.S.; Choudhary, S.; Sonika; Sharma, A.; Beniwal, V.; Tuli, H.S.; Sharma, S. Ferulic Acid: A Promising Therapeutic Phytochemical and Recent Patents Advances. Recent Pat. Inflamm. Allergy Drug Discov. 2019, 13, 115–123. [Google Scholar] [CrossRef]
  146. Zduńska, K.; Dana, A.; Kolodziejczak, A.; Rotsztejn, H. Antioxidant properties of ferulic acid and its possible application. Ski. Pharmacol. Physiol. 2018, 31, 332–336. [Google Scholar] [CrossRef]
  147. Janus, E.; Pinheiro, L.R.; Nowak, A.; Kucharska, E.; Świątek, E.; Podolak, N.; Perużyńska, M.; Piotrowska, K.; Duchnik, W.; Kucharski, Ł.; et al. New Ferulic Acid and Amino Acid Derivatives with Increased Cosmeceutical and Pharmaceutical Potential. Pharmaceutics 2023, 15, 117. [Google Scholar] [CrossRef]
  148. Keshavarz, F.; Dorfaki, M.; Bardania, H.; Khosravani, F.; Nazari, P.; Ghalamfarsa, G. Quercetin-loaded Liposomes Effectively Induced Apoptosis and Decreased the Epidermal Growth Factor Receptor Expression in Colorectal Cancer Cells: An In Vitro Study. Iran. J. Med. Sci. 2023, 48, 321–328. [Google Scholar]
  149. Shukla, D.; Nandi, N.; Singh, B.; Singh, A.; Kumar, B.; Narang, R.; Singh, C. Ferulic acid-loaded drug delivery systems for biomedical applications. J. Drug Deliv. Sci. Technol. 2022, 75, 103621. [Google Scholar] [CrossRef]
  150. Bhattacharyya, S.; Majhi, S.; Saha, B.P.; Mukherjee, P.K. Chlorogenic acid-phospholipid complex improve protection against UVA induced oxidative stress. J. Photochem. Photobiol. B 2014, 130, 293–298. [Google Scholar] [CrossRef]
  151. Servili, M.; Selvaggini, R.; Esposto, S.; Taticchi, A.; Montedoro, G.F.; Morozzi, G. Health and sensory properties of virgin olive oil hydrophilic phenols: Agronomic and technological aspects of production that affect their occurrence in the oil. J. Chromatogr. A 2004, 1054, 113–127. [Google Scholar] [CrossRef]
  152. Sun, T.; Simon, P.W.; Tanumihardjo, S.A. Antioxidant phytochemicals and antioxidant capacity of biofortified carrots (Daucus carota L.) of various colors. J. Agric. Food Chem. 2009, 57, 4142–4147. [Google Scholar] [CrossRef] [PubMed]
  153. Yang, G.; Fu, Y.; Malakhova, M.; Kurinov, I.; Zhu, F.; Yao, K.; Li, H.; Chen, H.; Li, W.; Lim, D.Y.; et al. Caffeic acid directly targets ERK1/2 to attenuate solar UV-induced skin carcinogenesis. Cancer Prev. Res. 2014, 7, 1056–1066. [Google Scholar] [CrossRef] [PubMed]
  154. Scalia, S.; Franceschinis, E.; Bertelli, D.; Iannuccelli, V. Comparative evaluation of the effect of permeation enhancers, lipid nanoparticles and colloidal silica on in vivo human skin penetration of quercetin. Ski. Pharmacol. Physiol. 2013, 26, 57–67. [Google Scholar] [CrossRef] [PubMed]
  155. Nagula, R.L.; Wairkar, S. Recent advances in topical delivery of flavonoids: A review. J. Control Release 2019, 296, 190–201. [Google Scholar] [CrossRef]
  156. dos Santos, C.O.L.; Spagnol, C.M.; Guillot, A.J.; Melero, A.; Corrêa, M.A. Caffeic acid skin absorption: Delivery of microparticles to hair follicles. Saudi Pharm. J. 2019, 27, 791–797. [Google Scholar] [CrossRef]
  157. Trivedi, H.R.; Puranik, P.K. Chlorogenic acid-optimized nanophytovesicles: A novel approach for enhanced permeability and oral bioavailability. Future J. Pharm. Sci. 2023, 9, 116. [Google Scholar] [CrossRef]
  158. Kyriakoudi, A.; Spanidi, E.; Mourtzinos, I.; Gardikis, K. Innovative delivery systems loaded with plant bioactive ingredients: Formulation approaches and applications. Plants 2021, 10, 1238. [Google Scholar] [CrossRef]
  159. Costa, R.; Costa Lima, S.A.; Gameiro, P.; Reis, S. On the development of a cutaneous flavonoid delivery system: Advances and limitations. Antioxidants 2021, 10, 1376. [Google Scholar] [CrossRef]
  160. Alharbi, W.S.; Almughem, F.A.; Almehmady, A.M.; Jarallah, S.J.; Alsharif, W.K.; Alzahrani, N.M.; Alshehri, A.A. Phytosomes as an emerging nanotechnology platform for the topical delivery of bioactive phytochemicals. Pharmaceutics 2021, 13, 1475. [Google Scholar] [CrossRef]
  161. Dwivedi, K.; Mandal, A.K.; Afzal, O.; Altamimi, A.S.A.; Sahoo, A.; Alossaimi, M.A.; Almalki, W.H.; Alzahrani, A.; Barkat, M.A.; Almeleebia, T.M.; et al. Emergence of Nano-Based Formulations for Effective Delivery of Flavonoids against Topical Infectious Disorders. Gels 2023, 9, 671. [Google Scholar] [CrossRef]
  162. Kazi, M.; Alhajri, A.; Alshehri, S.M.; Elzayat, E.M.; Al Meanazel, O.T.; Shakeel, F.; Noman, O.; Altamimi, M.A.; Alanazi, F.K. Enhancing oral bioavailability of apigenin using a bioactive self-nanoemulsifying drug delivery system (Bio-SNEDDS): In vitro, in vivo and stability evaluations. Pharmaceutics 2020, 12, 749. [Google Scholar] [CrossRef]
  163. Chavda, V.P.; Patel, A.B.; Mistry, K.J.; Suthar, S.F.; Wu, Z.X.; Chen, Z.S.; Hou, K. Nano-Drug Delivery Systems Entrapping Natural Bioactive Compounds for Cancer: Recent Progress and Future Challenges. Front. Oncol. 2022, 12, 867655. [Google Scholar] [CrossRef] [PubMed]
  164. Singh, M.; OS, B. Plant Latex: A Rich Source of Haemostatic Proteases, Herbal Medicine in India, 1st ed.; Springer: Singapore, 2020; pp. 143–153. [Google Scholar]
  165. Panda, S.; Vijayalakshmi, S.; Pattnaik, S.; Swain, R.P. Nanosponges: A novel carrier for targeted drug delivery. Int. J. PharmTech Res. 2015, 8, 213–224. [Google Scholar]
  166. Aizik, G.; Ostertag-Hill, C.A.; Chakraborty, P.; Choi, W.; Pan, M.; Mankus, D.V.; Lytton-Jean, A.K.R.; Kohane, D.S. Injectable hydrogel based on liposome self-assembly for controlled release of small hydrophilic molecules. Acta Biomater. 2024, 183, 101–110. [Google Scholar] [CrossRef] [PubMed]
  167. Abd, E.; Namjoshi, S.; Mohammed, Y.H.; Roberts, M.S.; Grice, J.E. Synergistic skin penetration enhancer and nanoemulsion formulations promote the human epidermal permeation of caffeine and naproxen. J. Pharm. Sci. 2016, 105, 212–220. [Google Scholar] [CrossRef]
  168. Rathee, J.; Malhotra, S.; Pandey, M.; Jain, N.; Kaul, S.; Gupta, G.; Nagaich, U. Recent Update on Nanoemulsion Impregnated Hydrogel: A Gleam into the Revolutionary Strategy for Diffusion-Controlled Delivery of Therapeutics. AAPS PharmSciTech 2023, 24, 151. [Google Scholar] [CrossRef]
  169. Tokudome, Y.; Nakamura, K.; Itaya, Y.; Hashimoto, F. Enhancement of skin penetration of hydrophilic and lipophilic compounds by pH-sensitive liposomes. J. Pharm. Pharm. Sci. 2015, 18, 249–257. [Google Scholar] [CrossRef]
  170. Lee, S.; Woo, H.; Lee, Y.; Kwon, Y.N.; Choi, Y.W. Skin Penetration and Localization Characteristics of Lipogel Containing Ascorbyl Palmitate. J. Pharm. Investig. 2001, 31, 225–232. [Google Scholar]
  171. Rangsimawong, W.; Obata, Y.; Opanasopit, P.; Takayama, K.; Ngawhirunpat, T. Investigation of skin penetration enhancing mechanism of limonene-containing liposome entrapped hydrophilic compound. Isan J. Pharm. Sci. 2017, 13, 10–17. [Google Scholar]
  172. Verma, D.D.; Verma, S.; Blume, G.; Fahr, A. Liposomes increase skin penetration of entrapped and non-entrapped hydrophilic substances into human skin: A skin penetration and confocal laser scanning microscopy study. Eur. J. Pharm. Biopharm. 2003, 55, 271–277. [Google Scholar] [CrossRef]
  173. Ban, J.; Mo, Z.; Cui, X.; Xu, Y.; Lyu, Z. Comparative study of liposomes and liposomes-in-polymer hydrogel as transdermal carriers for improving the topical delivery of imperatorin. J. Holist. Integr. Pharm. 2021, 2, 32–41. [Google Scholar] [CrossRef]
  174. Mengesha, Y. Nanotechnology-Enhanced Controlled-Release Systems in Topical Therapeutics. Precis. Nanomed. 2024, 7, 1365–1385. [Google Scholar]
  175. Touitou, E.; Dayan, N.; Bergelson, L.; Godin, B.; Eliaz, M. Ethosomes-novel vesicular carriers for enhanced delivery: Characterization and skin penetration properties. J. Control. Release 2000, 65, 403–418. [Google Scholar] [CrossRef] [PubMed]
  176. Honeywell-Nguyen, P.L.; Bouwstra, J.A. Vesicles as a tool for transdermal and dermal delivery. Drug Discov. Today Technol. 2005, 2, 67–74. [Google Scholar] [CrossRef]
  177. Elsayed, M.M.A.; Abdallah, O.Y.; Naggar, V.F.; Khalafallah, N.M. Lipid vesicles for skin delivery of drugs: Reviewing three decades of research. Int. J. Pharm. 2007, 332, 1–16. [Google Scholar] [CrossRef]
  178. Müller, R.H.; Radtke, M.; Wissing, S.A. Solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) in cosmetic and dermatological preparations. Adv. Drug Deliv. Rev. 2002, 54, 131–155. [Google Scholar] [CrossRef]
  179. Danaei, M.; Dehghankhold, M.; Ataei, S.; Hasanzadeh Davarani, F.; Javanmard, R.; Dokhani, A.; Khorasani, S.; Mozafari, M.R. Impact of particle size and polydispersity index on the clinical applications of lipidic nanocarrier systems. Pharmaceutics 2018, 10, 57. [Google Scholar] [CrossRef]
  180. Hua, S.; de Matos, M.B.C.; Metselaar, J.M.; Storm, G. Current trends and Challenges in the Clinical Translation of Nanoparticulate Nanomedicines Pathways for Translational Development and Commercialization. Front. Pharmacol. 2018, 9, 790. [Google Scholar] [CrossRef]
  181. Sharifi, S.; Mahmoud, N.N.; Voke, E.; Landry, M.P.; Mahmoudi, M. Importance of Standardizing Analytical Characterization Methodology for Improved Reliability of the Nanomedicine Literature. Nano-Micro Lett. 2022, 14, 172. [Google Scholar] [CrossRef]
Figure 1. The main skin penetration routes of active substances.
Figure 1. The main skin penetration routes of active substances.
Applsci 15 07101 g001
Figure 2. The structure of the selected carriers of hydrophilic compounds.
Figure 2. The structure of the selected carriers of hydrophilic compounds.
Applsci 15 07101 g002
Figure 3. Chemical structure of high hydrophylic flavonoides; (a) Quercetin (b) Luteolin (c) Apigenin.
Figure 3. Chemical structure of high hydrophylic flavonoides; (a) Quercetin (b) Luteolin (c) Apigenin.
Applsci 15 07101 g003
Figure 4. Chemical structure of high hydrophilic phenolic acids; (a) Ferulic acid (b) Caffeic acid (c) Chlorogenic acid.
Figure 4. Chemical structure of high hydrophilic phenolic acids; (a) Ferulic acid (b) Caffeic acid (c) Chlorogenic acid.
Applsci 15 07101 g004
Table 2. Summary of delivery strategies for hydrophilic phytochemicals based on reviewed scientific data.
Table 2. Summary of delivery strategies for hydrophilic phytochemicals based on reviewed scientific data.
Chemical Compound Encapsulated in the CarrierType of CarrierRole of CarrierMethod of EncapsulationPharmacokineticsTherapeutic EffectReferences
ApigeninTransfersomes, phytosomes, ethosomes, liposomes, hydrogelsEnhanced solubility and stability; improved skin penetration; controlled and prolonged release; protection from oxidative degradationLipid hydration, sonication, hydrogel encapsulation, phospholipid complexationProlonged release; improved retention in the stratum corneum; enhanced skin distributionAnti-inflammatory and antioxidant effects; targeted delivery to inflamed skin areas[148,158,159,160,161,162,163]
QuercetinLiposomes, niosomes, nanoliposomes, phytosomes, polymeric micelles, nanocrystals, lipid nanoparticles (SLNs, PEVs), nanoemulsions, nanofibers, hydrogelsEnhanced solubility and stability; protection from enzymatic and oxidative degradation; improved penetration through stratum corneumLipid hydration, sonication, micro-/nanoemulsion, phospholipid complexation, polymer/lipid encapsulationSlower elimination; prolonged and controlled release; improved metabolic and structural stability in physiological conditionsIncreased transdermal bioavailability; targeted anti-inflammatory, antioxidant, and anticancer activity (e.g., EGFR (Epidermal Growth Factor Receptor) inhibition, deeper skin layer delivery)[93,129,148,155,159,160,161,163]
LuteolinNiosomes, transfersomes, phytosomes, nanoemulsions, liposomes, SLNs, hydrogelsImproved physicochemical stability, enhanced skin penetration, anti-inflammatory and antioxidant supportEncapsulation in invasomes, transfersomal/nanosystemic formulations, phospholipid complexationProlonged release, deeper diffusion, better skin distribution, improved structural retentionIncreased transdermal bioavailability; targeted anti-inflammatory and antioxidant effects[148,155,159,160,161,163]
Ferulic AcidPhytosomes, SLNs, NLCs, nanoemulsions, liposomes, hydrogelsEnhanced stability, solubility, and skin penetration; protection from oxidative degradation; prolonged releaseLipid matrix encapsulation (SLN, NLC), phospholipid complexation, hydrogel and nanoemulsion integrationSlower release, improved structural integrity, protection in physiological conditionsIncreased bioavailability via deeper skin penetration and resistance to metabolic degradation; targeted antioxidant and anticancer effects[93,148,149,161]
Chlorogenic AcidPhytosomes, liposomes, nanocapsules, nanoemulsions, hydrogels, NPVs (Phospholipon® 90H, LIPOID® S100), CA–HSPC complexImproved solubility, stability, and skin penetration; protection from oxidation; enhanced SC retentionLipid hydration, phospholipid complexation (HSPC, NPVs), hydrogel embeddingProlonged release, enhanced SC retention, improved structural stability in physiological conditionsIncreased transdermal bioavailability; targeted antioxidant action and UV protection in inflammatory sites[148,150,157,160,161]
Caffeic AcidEthosomes, liposomes, phytosomes, SLNs, hydrogels, polymeric/lipid nanoparticles, chitosan microparticlesEnhanced stability, solubility, and skin penetration; targeted release in skin and follicles; antioxidant protectionLipid hydration, phospholipid complexation, hydrogel encapsulation, biodegradable nanoparticle formation, spray-drying (microparticles)Slower or prolonged release, better distribution and penetration through stratum corneum and folliclesImproved bioavailability and retention; antioxidant action; potential for folliculitis treatment[93,148,155,156,160,161]
CurcuminTransferosomes, nanosponges, liposomes, liposome-in-hydrogelEnhanced penetration through SC, protection from oxidation, prolonged release, increased bioavailabilityHigh-pressure technique, solvent diffusion, lipid hydration, chitosan hydrogel formationProlonged action, reduced degradation, improved structural stabilityHigher transdermal bioavailability; targeted delivery to deep skin layers; applications in inflammation, endodontics, and periodontics[155,164,165,166]
GenisteinNanoemulsion, Liposomes, Polymeric MicellesEnhanced skin permeability, protection from degradation, increased stability and bioavailabilityEncapsulation in lipid and polymeric nanoparticles or micellesFaster absorption, prolonged release, stability in physiological environmentImproved transdermal bioavailability; targeted antioxidant activity through the skin[160,163]
RutinNanoemulsion, Liposomes, NiosomesImproved skin permeability, stability, solubility; protection from oxidationEncapsulation in liposomes and niosomes using lipid hydrationFaster absorption, improved stability, prolonged actionEnhanced bioavailability; effective in anti-inflammatory therapy and skin delivery[160,163]
MorusinNiosomeEnhanced stability and skin penetrationLipid-based encapsulation in niosomesIncreased dermal penetration and bioavailabilityTargeted delivery through the skin[163]
CapsaicinTransferosomeEnhanced skin penetration, reduced systemic side effectsHigh-pressure encapsulation techniqueImproved dermal absorption compared to conventional formulationsTargeted delivery to pain receptors and peripheral nerves[164]
Vincristine sulfateTransferosomeSite-specific delivery with minimized systemic toxicityHigh-pressure encapsulation techniqueEnhanced skin penetrationTargeted anticancer delivery with reduced effect on healthy tissue[164]
Cannabidiol EthosomeEnhanced skin permeability and localized deliveryEthosomal formulationIncreased accumulation in stratum corneumImproved therapeutic targeting of cutaneous endocannabinoid system[164]
CaffeineNanoemulsionImproved solubility and enhanced penetration through the stratum corneumNanoemulsion with eucalyptus/oleic oil and Volpo-N10 emulsification systemBetter diffusion through SC, increased solubility, enhanced skin retentionEffective delivery to deeper skin layers[167,168]
NaproxenNanoemulsionImproved solubility and deeper skin penetrationNanoemulsion using Volpo-N10, ethanol, eucalyptus/oleic oil, and PBS (Phosphate-Buffered Saline) bufferIncreased solubility in the stratum corneum, enhanced diffusionEnhanced transdermal bioavailability and targeted delivery to deeper skin layers[167,168]
KaempferolSubmicron EmulsionEnhanced solubility and skin penetrationSubmicron emulsion using PEG-400 (Polyethylene glycol) or eucalyptus oilImproved penetration through the stratum corneumIncreased skin bioavailability with targeted action in the stratum corneum[159]
Sulbutamol SulfateEthosomeEnhanced skin penetration for systemic and localized deliveryEthosomal formulationImproved penetration through the stratum corneumTargeted delivery to respiratory and cutaneous receptors[164]
Ammonium GlycyrrhizinateEthosomeAnti-inflammatory action on the skinIn vitro percutaneous permeation through human skinImproved availability in deeper skin layersImproved therapeutic effectiveness[164]
CyclodextrinsCyclodextrinsStabilization against oxidation and improved bioavailabilityComplexation with β-cyclodextrinStabilization against oxidationHigher bioavailability with β-CD application[164]
Econazole NitratePolymeric NanospongeImproved skin penetration, therapeutic stabilizationUltrasonic techniqueIncreased stability and prolonged actionEnhanced transdermal bioavailability[165]
ResveratrolPEGylated liposomeEnhanced stability, prolonged presence in tissues PEGylation of liposomesProlonged action in tissuesEnhanced bioavailability and protection from degradation[155]
CalceinpH-sensitive LiposomeEnhanced penetration through the SC depending on pHLipid layer technique and freeze-thaw cyclesFaster penetration at pH 5.0Targeted delivery to the stratum corneum at pH 5.0[169]
NBD-PE
(N-(7-Nitrobenz-2-oxa-1,3-diazol-4-yl)phosphatidylethanolamine)
pH-sensitive LiposomeFacilitated skin penetration in acidic conditionsLipid layer technique and freeze-thaw cyclesIncreased penetration at pH 5.0Targeted delivery to the stratum corneum at pH 5.0[169]
Ascorbyl Palmitate (AsP)EthosomeAnti-inflammatory action, assessed by in vitro skin permeationEthosomal formulationImproved availability in deeper skin layersEnhanced therapeutic effectiveness, targeted dermal action[170]
Sodium Fluorescein (NaFI)Limonene-containing LiposomeEnhanced skin penetration due to limonene, modification of stratum corneumThin-layer lipid hydration technique, sonication, addition of limoneneIncreased fluidization of the lipid membrane, improved penetration Improved transdermal bioavailability[171]
Carboxyfluorescein (CF)LiposomeEnhanced skin penetration through liposomal carriersRotational evaporation technique, formation of thin lipid layer, hydrationImproved distribution to the stratum corneum and deeper skin layersImproved transdermal bioavailability[172]
TetracaineLiposomeLocal skin anesthesiaThin lipid layer hydration, sonication methodFaster penetration through the stratum corneumImproved bioavailability in the stratum corneum[172]
Betamethasone dipropionateLiposomeImproved effectiveness in eczema treatmentHydration and sonication techniqueImproved penetration and retention in deeper skin layersHigher bioavailability and therapeutic effectiveness[172]
HesperidinLipid-Polymer Hybrid Nanoparticles (LPHNPs), Microemulsion-based ointmentEnhanced skin penetration, controlled release, protection from degradationEncapsulation in lipid-polymer hybrid nanoparticles; eucalyptus oil-based water emulsionInitial burst followed by prolonged release; improved diffusion through the skinImproved bioavailability in deeper skin layers, with targeted action and enhanced stability[159,172]
HesperetinMicroemulsion, Topical FilmImproved bioavailability, enhanced skin penetrationMicroemulsion with eucalyptus oil, film matrixFaster penetration through the stratum corneumEnhanced transdermal bioavailability[159]
NaringeninUbmicron Emulsion, Gel, Elastic LiposomeEnhanced stability, improved bioavailability, better skin penetrationEncapsulation in submicron emulsion and elastic liposomesIncreased skin diffusion, controlled releaseTargeted antioxidant and anti-inflammatory action[159]
CatechinsNanotransfersomes, Grape Seed Extract Cream, Multilamellar phosphatidylcholine liposomes, Ethanol-enriched liposomesImproved skin absorption and penetration, protection from degradationNanotransfersomes with hyaluronic acid, grape seed extract cream; multilamellar and ethanol-based liposomesLonger retention in the skin, increased penetration, slower releaseHigher bioavailability through the stratum corneum; targeted antioxidant and protective action in skin layers[159]
MyricetinLipid Nanoparticles, LiposomesProtection from degradation, increased stabilityMultilamellar liposomes, lipid nanoparticlesImproved stability, prolonged actionEnhanced bioavailability in skin layers, targeted anti-inflammatory action[160]
ImperatorinLipid-Polymer Hybrid Nanoparticles (LPHNPs)Prolonged action and controlled releaseEncapsulation in lipid-polymer hybrid nanoparticlesInitial burst followed by sustained releaseEnhanced skin penetration[173]
NorfloxacillinLipid-Polymer Hybrid Nanoparticles (LPHNPs)Prolonged action and controlled releaseEncapsulation in lipid-polymer hybrid nanoparticlesInitial burst release and prolonged releaseEnhanced skin penetration[174]
IndomethacinPolymeric NanoparticlesEnhanced penetration, controlled releaseEncapsulation in polymeric nanocapsules and nanospheresDiffusion-based release (Higuchi model)Increased bioavailability via deeper skin layers[174]
Amphotericin BPolycaprolactone (PCL) NanoparticlesSkin penetration and controlled releaseEncapsulation in PCL polymeric nanoparticlespH-dependent release (faster at pH 7.4)Enhanced delivery to deeper skin layers[174]
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

Sharafan, M.; Dziki, A.; Malinowska, M.A.; Sikora, E.; Szopa, A. Targeted Delivery Strategies for Hydrophilic Phytochemicals. Appl. Sci. 2025, 15, 7101. https://doi.org/10.3390/app15137101

AMA Style

Sharafan M, Dziki A, Malinowska MA, Sikora E, Szopa A. Targeted Delivery Strategies for Hydrophilic Phytochemicals. Applied Sciences. 2025; 15(13):7101. https://doi.org/10.3390/app15137101

Chicago/Turabian Style

Sharafan, Marta, Anna Dziki, Magdalena Anna Malinowska, Elżbieta Sikora, and Agnieszka Szopa. 2025. "Targeted Delivery Strategies for Hydrophilic Phytochemicals" Applied Sciences 15, no. 13: 7101. https://doi.org/10.3390/app15137101

APA Style

Sharafan, M., Dziki, A., Malinowska, M. A., Sikora, E., & Szopa, A. (2025). Targeted Delivery Strategies for Hydrophilic Phytochemicals. Applied Sciences, 15(13), 7101. https://doi.org/10.3390/app15137101

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