Skip to main content

Liposome drug delivery in combating the widespread topical antibiotic resistance: a narrative review

Abstract

Background

The increasing trend of antibiotic resistance has posed challenges for scientists, especially in developing better drug formulations. The discovery of new antibiotics could take years. Therefore, the management of an ideal drug delivery system has become a primary focus nowadays.

Main body of abstract

Almost all skin diseases could be treated with the administration of topical drugs, especially infectious skin diseases. The increasing cases of antimicrobial resistance require innovative strategies and actions. In dermatokinetics, achieving optimal drug concentrations in the deepest layers of skin tissue is a significant challenge. Human skin has remarkably complex characteristics, presenting a major obstacle in efficiently maintaining drug efficacy. Nanocarriers are an important part of nanomedicine which provide excellent drug penetration through various drug delivery systems. Lipid-based nanovesicles, such as liposome, are the oldest and most potential nanovesicles for such a purpose. Several studies have shown the efficacy of liposome-contained antibiotics and offered the lowest microbial inhibition concentration (MIC). It is suggested that liposome also delivers greater drug accumulation compared to blank drugs.

Short conclusion

Liposome is a flexible lipid-based drug delivery that enhances drug permeation through skin tissue by mimicking the lipid bilayer system of the organ. It is non-toxic, less immunogenic, and easily degraded by enzyme. The incorporation of liposome into antibiotics may reduce the inefficient drug dosage since the encapsulation will protect the active compounds prior to being released from the vehicle. Thus, the lowest MIC and less clinical side effects will be obtained.

Graphical abstract

1 Background

Most dermatological diseases are commonly treated with topical applications as they can easily reach the target area within the skin tissue compared to other administration routes. However, topical therapies have several disadvantages especially due to improper and irrational use which may lead to detrimental effects, such as emerging cases of antibacterial resistance and incurable complications. There are several well-known topical antibiotics, such as gels, creams, ointments, sprays, and liquids. Most of the topical antibiotics could be purchased over the counter (OTC) without prescriptions since the medical issues are usually mild or do not require medical consultation. A concerted effort is required to prevent more resistance cases in the future. Reports on topical antibiotic resistance are limited compared to those on the systemic route. However, one systematic review has suggested that the cases of antibiotic resistance to Pseudomonas aeruginosa (P. aeruginosa) have increased drastically. P. aeruginosa is known as the main pathogen of acne. The same study reported that antibiotic resistance to P. aeruginosa has increased from 20% in 1978 to 62% in 1996. Therefore, some countries, for example, the UK through National Institute for Health and Care Excellence, have released guidelines on antimicrobial stewardship for consultation which help practitioners prescribe and monitor patients’ antibiotics [1, 2].

Skin infections are usually caused by the skin flora normal infiltration through open wounds or scars. Most infections are caused by Gram-positive microbes such as Staphylococcus aureus (S. aureus), Enterococcus faecalis (E. faecalis), and Streptococcus viridans (S. viridans). However, Gram-negative bacteria could also cause the infections, for example, Escherichia coli (E. coli), Haemophilus influenza (H. influenza), Clostridium species, Mycobacterium species, Pseudomonas species, and other anaerobes [2]. Each type of bacteria could be distinguished by the thickness of its cell wall membrane. Gram-positive bacteria possess thicker peptidoglycan compared to Gram-negative bacteria with fewer layers. Nevertheless, Gram-negative bacteria are likely to have greater intrinsic resistance against antibiotics, which potentially leads to antimicrobial resistance [3]. While minor infections can be effectively treated with conventional topical antibiotics, recent attention has been directed toward preserving drug efficacy and optimizing bioavailability. Novel drug delivery for topical applications has been an issue not only to limit the widespread of antibiotic resistance, but also to navigate efficient drug molecules to reach targeted tissues. From a dermatokinetics perspective, the fundamental problem of low-level bactericidal activity is usually related to the bioavailability of drug molecules within the tissue. Skin layers have complex structures as they serve as the first anatomical barrier against many pathogens from the environment. Having many variations, the topical route facilitates permeation across the lipid bilayer system of the skin. Conventional topical dermal drug delivery systems (TDDDS) such as ointments, creams, gels, lotions, liniments, and oils are the most popular formulas for treating many skin diseases. However, there is a need for innovative approaches which allow increased drug concentration as the primary limitation of this pathway is the constrained permeability of drug dosages [4, 5]. The pore size and physicochemical properties within an individual’s skin type can influence drug accumulation levels. A hydrophilic formulation in a topical drug delivery system provides enhanced capacity for drug molecules to reach specific layers of the skin tissue [6]. Liposome is one of the strategies to improve antibiotics ability to obtain better clinical results in treating skin infections. As reported by Sang et al. [7], liposome can be coupled with ligands or small peptides that can bind bacteria, improving the antibiotic delivery efficiency and avoiding the side effect.

Lipid nanoparticles are a nanotechnology-based advancement in pharmaceutical engineering. Many studies have been conducted to examine their ability to enhance drug delivery to reach specific target within tissues. They can be developed to be sustainable and capable of interacting with proteins in the body. Moreover, when they meet lipid-sensitive enzyme, they can trigger a controlled release mechanism [8]. Nanosized liposome is one of the options to provide the ability but studies focus on the delivery system to encapsulate topical antibiotics are still limited. Thus, this review aims to explain the complexity of skin tissue and dermatokinetics as well as liposome adaptability when it encapsulates various antibiotics to treat skin infections. It also highlights how liposome could enhance topical antibiotics as well as its inhibition ability toward bacteria compared to non-liposome coated. The review also aims to provide insights into pharmaceutical breakthroughs with liposome as a nanocarrier for topical antibiotics. Scoping assessment was conducted to collect data from recorded and ongoing studies within the last ten years (2014–2024).

2 Main text

2.1 The anatomy and physiology of skin

The skin is the widest organ that covers the body. It serves crucial functions as a physical barrier, sensory organ, and immunology interface. It accommodates homeostasis by limiting the loss of water, electrolytes, and heat. It contains Merkel cells as mechanoreceptors and Langerhans cells or dendritic cells serving immune functions [9, 10]. Skin tissue comprises three different layers: the epidermis, dermis, and subcutaneous tissue (Fig. 1). The epidermal layer is composed of squamous epithelium and is stratified based on the level of keratinization exhibited by the cells. The layers include the stratum basale (comprising basal cells) at the base, the stratum spinosum (comprising spinous or prickle cells), the stratum granulosum (comprising granular cells), and the stratum corneum (comprising corneocytes). The stratum corneum (SC) is held together by lipid bilayers organized in a brick-and-mortar configuration [9, 11]. Stratum corneum forms a hydrophobic layer with 10–30 µm thickness and functions as a prominent barrier for skin permeation. In addition to corneocytes, the skin barrier consists of a highly lipidic component comprising keratin filaments and filaggrin. Corneocytes are embedded within multilamellar lipids composed of sterols, phospholipids, and glycosphingolipids [10]. Corneocytes have a rigid structure, lack functionality (are non-viable), do not contain a nucleus, and are rich in keratin [12]. In the stratum corneum, corneocytes show a flat hexagonal shape aligned parallel to the skin surface, characterized by an irregular morphology. They have a diameter of approximately 40 µm, a width reaching up to around 1 µm, and an intercellular lipid area thickness of up to 0.1 µm. The corneocytes and lipids in the stratum corneum have a compact brick-and-mortar-like configuration, serving as a formidable barrier to the permeation and diffusion of substances applied topically or transdermally. This structure functions as a barrier impeding the skin absorption of topical products [13, 14]. In the stratum basale, cells undergo active proliferation and keratinization. They move toward the outer layers, reaching the stratum corneum. The route of skin permeation entails drug passage through the epidermis and associated skin appendages, predominant hair follicles, and sweat glands, which represent an alternative path to the intact epidermis. These skin appendages collectively cover approximately 0.1% of the human skin surface [5, 12].

Fig. 1
figure 1

Skin histology

Dermis is the deeper layer below the epidermis. The thickness of dermis is 3–5 mm, and it consists of fibrous proteins (such as collagen and elastin) and an interfibrillar gel of glycosaminoglycans, salts, and water. It also contains appendages, including sweat gland, pilosebaceous units, blood vessel, and nerves. The dermis supports the binding between epidermis and subcutis, keeping the tensile strength, elasticity, and resilience [10]. It also supports nutrient and oxygen delivery as well as waste removal from epidermis by diffusion across dermo-epidermal junction [15]. Dermis also contains a range of immune cells, including macrophages and dermal dendritic cells. The dermis serves as the pathway for drug permeation through the transcellular route (diffusion across corneocytes), intercellular route (diffusion through the lipid matrix), and the shunt pathway or appendageal route (diffusion into sweat glands, hair follicles, and sebaceous glands) [10].

However, the outer physical barrier contains non-living components (called “dead”) [14]. The subcutaneous layers, the epidermis and dermis, contain living components. The subcutaneous layer has three zones: the basal zone (with thick, dense keratin consisting of 4–10 cells), the intermediate zone (with denser keratin loosely composed of 8–12 cells), and the superficial zone (with less keratin, 2–3 cells). The subcutaneous layer is a dynamic barrier; it can be modulated by topical drug delivery systems, which can make a drug able to penetrate or fail to penetrate the layer. The establishment of the subcutaneous barrier is influenced by dead corneocytes within a flat, water-repellent sheath in an extracellular lipid matrix by the proteolytic breakdown of corneodesmosomes. These cohesive molecular bonds between corneocytes encompass approximately 30% of the overall surface volume of the compact stratum corneum layer [14]. The hypodermis layer comprises loose connective tissue and adipose tissue. The components of dermis and hypodermis are blood vessel, lymphatics, nerve cells, and skin appendages. The surface covered by the appendages is only about 0.1% of the skin continuum [10]. Although the absorption of compound is small, this route enables permeation of charged molecules and large polar compounds, such as peptide-based drugs [12].

2.2 Permeability of the skin

Understanding the skin permeation system is important before formulating a suitable topical drug delivery. Skin is the first barrier which provides strong immunity protection toward various threats from the environment such as biological, mechanical, chemical, and physical attacks [16]. Overall, the width of skin tissue could extend to approximately 2 m2 and comprises several layers. Each layer has specific functions based on its histological feature. From outward to inward, skin tissue consists of living epidermis, dermis, and hypodermis. Epidermis structure comprises five different layers: stratum basal, stratum spinosum, stratum granulosum, stratum lucidum, and stratum corneum. Among these layers, the stratum corneum should be considered for topical administration [16, 17]. Prior the permeation process, an important variable that must be considered is the partition phenomenon. In terms of drug pharmacokinetic, partition relates to the distribution of the drug. The partition occurs between the drug vehicles and the surface of the skin tissue to finally achieve the balance during the process; diffusion and partition will repeatedly take place until the whole drug molecules reach the capillary system. Defining the partition potency requires an absolute value which explains the ratio between the two involved variables. Partition coefficient (K) helps to determine the strategy to increase drug absorption through skin barrier especially if the drug is designed as a transdermal route [18, 19]. Based on the definition, K is a value used to estimate the drug concentration of a certain skin drug route toward its delivery system [20]. To achieve the coefficient, studies to retrieve the information have been conducted. In a study using human viable epidermis and stratum corneum layers, the partition coefficient was obtained by measuring the concentration of drug compound in the tissue or lipid (per gram of tissue or lipid weight) and the concentration of drug compound in the buffer at equilibrium (24 or 2 h). In another study, the partition coefficient was defined as the mass of chemical per unit mass of dry stratum corneum relative to the mass of chemical in buffer per volume of buffer. The study revealed that K value was higher in lipidized sample than that in the de-lipidized stratum corneum (fully hydrated). The size of compound, less than 500 Da, is easily penetrates through passive diffusion across lipid bilayer of cell membrane and further passes the absorption step [21]. Aside of partition coefficient, another important parameter needed to observe and analyze skin penetration ability is diffusion coefficient (D). Diffusion coefficient is obtained by measuring the steady flux values and its lag time. However, only a limited number of studies have documented the diffusion coefficient of the skin due to limitations of models and methodologies. Both parameters could be employed to calculate the skin’s absorption rate [22].

The stratum corneum of the skin serves as the main location for percutaneous drug absorption since it consists of corneocytes and is enriched with intercellular lipids. Thus, it has an hydrophobic ability. The corneocytes are connected through desmosome to ensure the strength of tissue connectivity and permeability for most hydrophilic molecules greater than 200–350 Da [6]. The intercellular lipids form a matrix which contains stratified hydrophilic and hydrophobic layers arranged into thick layers as multilamellar sheets [16, 18, 23]. Due to the rich staggered corneocytes available in stratum corneum, the opportunity for lipoidal diffusion to occur is high. It is approximately 1000 times more hydrophobic [16]. The stratum corneum plays a crucial role in moisturizing the outermost skin layer. Excessive loss of overall body water may result in dry skin, affecting the drug’s ability to permeate effectively. Various known routes for drug molecule to access systematic circulation are intracellular, intercellular, and transfollicular routes [6]. However, the basic mechanism of permeation system usually depends on the physiochemical property of the drug molecules. Highly hydrophobic drugs are difficult to permeate through the hydrophilic layer and vice versa [6, 24]. The difference between the available routes is mostly related to the size and lipid solubility of penetrated molecules. Hydrophilic compounds most likely penetrate through the crevices found inside the lipid layer surrounding the corneocytes. Intercellular pathway allows for hydrophilic molecules to pass the lipid matrix. Yet, it requires exact sequential diffusion and partitioning amid the alkyl chain length and the polar head groups of intercellular lipids [6, 16, 25]. The intracellular pathway initiates diffusion that allows the molecules to run deeply into the keratin [16]. Lower pKa value on the skin surface also allows for better permeation if the drug compound is bound under acidic vehicle due to the higher unionized level on the skin surface. If the partition coefficient is known, then further calculation to measure flow rate of skin permeation of certain drug molecules could be obtained through Fick’s second law [18].

2.3 Dermatokinetics of applied topical drugs

After a topical preparation is applied to the skin, the drug must be able to penetrate deeper layers by crossing the various layers [13]. Therefore, only a small drug concentration can penetrate the skin layers within a certain period [26]. The dermatokinetics concept emphasizes three mechanisms: (1) The stratum corneum layer limits the rate of drug absorption; (2) the drug diffusion process affects the drug concentration penetrating the dermis layer; the concentration is equal to the drug concentration in the stratum corneum; and (3) the drug concentration in the skin layers (stratum corneum, epidermis, dermis) describes the dermatological effectiveness of the drug [13]. The delivery of drugs to the skin involves several processes: (a) drug release from the dosage form; (b) drug penetration through the stratum corneum; (c) drug diffusion across the stratum corneum, primarily via intercellular lipids; (d) drug separation from the stratum corneum into the viable epidermal layer; (e) drug diffusion through the epidermal layer to the underlying dermis; and (f) drug absorption by capillaries, leading to systemic circulation [12].

Water component is essential in most topical drug delivery systems because it quickly penetrates the surface layer. Water can cause the subcutaneous layer to soften and thicken (toward the surface and not laterally), reduce the depth of the groove, and reduce subcutaneous corneocyte undulations. Topical formulations alter subcutaneous hydration by modulating water content in the stratum disjunctum, with slight changes in water content in the stratum compactum. The insoluble and protective corneocyte sheath is also affected by stratum corneum hydration, which influences protease activity. The sheath is irregular, deformed, and brittle in its underlying layers, but 80% of superficial subcutaneous corneocytes are polygonal and very strong. This decrease in subcutaneous corneocyte maturation can be influenced by environmental exposure or skin dryness, which can reduce the resistance of the sheath. The impact of heterogeneous skin morphology determines how quickly the drug can penetrate layers layer of the skin and be effective for topical treatment. Drugs will pass through the subcutaneous layer more easily if they contain additional formulations in the form of one or more chemicals to increase their penetration ability, have a small molecular weight (≤ 500 Da), have high solubility, and are sufficiently lipophilic (logarithm of octanol partition candidate—water coefficient log P in the range 0–5). However, those with a larger molecular weight are more lipophilic and more difficult to pass through epidermis due to their poor solubility in water [14, 27].

Transportation of drugs in and out of the subcutaneous lipid layer by permeation along the corneocytes mainly occurs via the lipid pathway only. The thickness of the subcutaneous extracellular lipid layer varies and depends on the number of lipid bilayer layers at any given cross section. Each bilayer illustrated is approximately ~ 11 nm wide and consists of narrow segments (~ 4.5 nm) and broad segments (~ 6.5 nm) arranged in alternating parallels [12, 28]. The thick lipid region extends 41 mm between subcutaneous corneocytes, while the thinnest interface is ~ 250 nm long [14, 29]. Other structures that can be a barrier to drug entry into the subcutaneous extracellular lipid space are the densely packed junctions and corneodesmosomes that appear in the stratum corneum’s lowest cell layers and the stratum granulosum [27,28,29].

Biopsy results showed > 100 lipid bilayers in one location. Corneodesmosomes are distributed in various body locations and can be influenced by previous treatments or disease. Corneodesmosomes are commonly found on corneocytes’ inner and outer subcutaneous surfaces in winter-induced xerotic skin, outer palmoplantar, xerotic, ichthyotic, and soap-induced hyperkeratotic skin [26, 29]. Hyperkeratosis can also be caused by genetic mutations that affect corneodesmosome proteolysis, such as in psoriasis patients. Thus, if corneodesmosome degradation continues, it will increase subcutaneous thickness. This does not mean that thicker subcutaneous tissue is less permeable. However, water and other solutes can have a tenfold higher permeability through the 400- to 600-µm-thick plantar subcutaneous compared to the thinner abdominal subcutaneous (20–30 µm), with a subcutaneous diffusion coefficient difference of up to 150 times. Because it has a thicker layer, the time lag for solutes passing through the plantar skin maybe ten times longer than the abdominal skin. Therefore, drug transport through subcutaneous lipids varies and is influenced by the location, be it the depth or location of the subcutaneous (at the edge of the corneocyte or on the surface of the corneocyte) and environmental influences, as well as skin diseases associated with corneodesmosomes [5, 26, 29].

When initially applied, the drugs will pass through hair follicles [5]. Drug absorption into the skin is influenced by several factors, such as molecular size, lipophilicity, formulation pH, penetrant concentration, chemical additives, skin hydration, and skin enzymes. The absorption ability of a drug is inversely proportional to its molecular weight and mainly affects the diffusion coefficient. Molecules larger than 500 Da will have more difficulties in penetrating the subcutaneous tissue. The permeability of drug molecules will be optimal at a lipid/water partition coefficient of 1 or greater. Drug formulations with excessively high or low pH levels can potentially damage or irritate the skin. Therefore, a neutral pH value (typically above the skin’s isoelectric point, pH 4) may be more appropriate for topical application. The degree of drug ionization at a certain pH will affect its ability to diffuse through the lipophilic intercellular areas in the subcutaneous layer and form pairs with ions in the skin to produce neutral compounds to cross the skin barrier. Drug delivery through the skin can avoid the first-pass effect in the liver, the physiological environment, and chemical or metabolic degradation in the gastrointestinal tract, such as changes in pH and luminal microflora. In addition, the slow plasma concentration of topical medications can reduce the side effects. Absorption of the drug into the skin occurs more slowly through passive diffusion. The speed of drug delivery across the stratum corneum depends on the solubility of the drug in water, the oil/water partition coefficient, the drug concentration in the formulation vehicle, the size and shape of the molecule, the surface area of the skin to be exposed, and the thickness of the stratum corneum [26].

Complete penetration ability can be achieved if all parameters are stable. Key parameters crucial for determining the efficacy of topical medication consist of Cmax (maximum drug concentration in the skin layers), Tmax (time required to reach Cmax), and AUC (area under the curve) in all skin layers. However, these parameters could only be observed through in vitro or in vivo experiments. Various methods have been employed in previous studies to assess topical products and their dermatokinetic profile such as tape stripping, microdialysis and open-flow microperfusion, vasoconstrictor assay, and microscopic and spectroscopic methods [13]. Tape stripping is simpler compared to other methods as it relies solely on the adhesive quality of applicators. However, various factors may introduce bias, such as skin conditions like scar tissue, pH, TEWL (transepidermal water loss), hydration, sebum content, and adhesion with the stratum corneum cells. This method is more effective when the study specifically aims to observe barrier restoration following adhesion [30]. To develop clinically effective topical products, it is essential that they demonstrate substantial skin bioequivalence (BE). Some studies have shown that pharmacokinetic endpoints should typically fall within specified BE limits (usually 0.80–1.25) on a log transformed basis (95% CI). Dermal open-flow microperfusion (dOFM) enables the assessment of intradermal biochemistry and drug concentrations by sampling dermal interstitial fluid for up to 48 h. This method of dermatokinetics assessment enhances monitoring, particularly for lipophilic drugs and larger proteins, using an inserted probe into the intradermal region. The area surrounding the probes is then scanned with longitudinal ultrasound to evaluate drug accumulation [31].

2.4 Various drug delivery strategies for topical route

Topical drugs are the safest option with minimal side effects and are primarily used to treat skin conditions. Their ease of application and high drug concentration make them a popular choice for long-term treatment [32]. The anatomical and physiological barriers in the skin pose challenges for effective penetration by foreign substances which affect the development of topical drugs. Topical drug delivery forms include solid (powder, plaster, ointments), semi-solid (gels, creams, ointments), liquid (lotions, solutions, emulsions, suspensions), and miscellaneous (patches, topical aerosols, rubbing alcohols, liquid cleansers) [33].

Topical delivery systems can target skin melanocytes for whitening, act as anesthetics for pain relief, serve as anti-infective agents, and activate Langerhans cells. The appropriate topical agent or delivery system is crucial, with consideration given to the specific skin cell type being targeted [34, 35]. The earliest method for improving skin absorption is by choosing the appropriate drug. Essential techniques such as selecting raw materials, utilizing eutectic systems, employing ion pairing, and understanding chemical potential are necessary for producing high-quality products. Topical drug absorption is primarily influenced by two factors: physiological aspects (such as skin thickness, pH, and type) and physicochemical properties including molecular weight and ionization [35]. Gels and creams are topical medications commonly available. It is essential to optimize the drug vehicle, particularly for topical applications. However, not all traditional topical forms can effectively incorporate active drug ingredients, such as antibiotics. Creating a conjugation or network system by combining materials is often challenging and may take years to develop the ideal formula. One strategy commonly used by many researchers is utilizing existing drug delivery components with more efficient release systems, such as lipid-based drug delivery. In addition to lipids, non-organic materials like gold and silver have also been found to be easily adaptable for use as drug delivery systems [36]. A study has shown that blank cationic liposomes combined with carboxyl-modified gold nanoparticles (AuC) efficiently penetrated S. aureus, with fluorescence microscopy indicating higher activity at lower pH levels (pH 4.5). This suggests the formulation could enhance antimicrobial efficacy within bacteria upon liposome loading [37].

Nanotechnology is a vital breakthrough in various fields, particularly pharmaceuticals. However, many traditional topical drug delivery systems still rely on larger materials. Skin permeability becomes the first obstacle to overcome before achieving clinical efficacy. The utilization of large materials in drug delivery presents significant challenges, such as in vivo instability, limited bioavailability, poor solubility, inadequate absorption, target-specific delivery issues, diminished effectiveness, and potential adverse drug effects [38]. The use of topical antibiotics is the main and important component in the treatment of skin infections. Several skin infections such as dermatitis, acne, and impetigo require antimicrobial agents that are easy to use without side effects. Generally, topical antibiotics work by suppressing bacterial growth and changing the condition of the skin so that they can support the repair of skin cells. This will help heal from infections. The antibiotics mupirocin, neomycin, and aminoglycosides have a broad spectrum so they are often recommended for the use of topical antibiotics. Irrational use of topical antibiotic may cause more harm than benefits. Despite the growing number of challenges, especially antibiotic resistance, various nanoplatforms have been introduced, studied, and modified alongside the current drugs. The primary aim is to achieve optimal formulas for specific drug delivery to effectively target and retain drug molecules within the intended tissues. Over the years, nanotechnology has led to significant advancements in medicine, particularly in the field of nanomedicine. The first generation of nanoparticle-based therapies included lipid systems like liposomes and micelles, which are now (Food and Drug Administration) FDA-approved for medical use [39].

Nanotechnology-based formulations, especially nanoparticle formulations, have many types and forms. For decades, researchers have tried to create adjustable drug delivery methods for skin diseases. However, the strong barrier in skin tissue limits the range of available strategies. Solutions have been explored, ranging from active methods like microneedles, electrophoresis, and sonophoresis to passive approaches such as carrier-mediated delivery involving chemical permeation enhancers and nanovesicular carriers [40,41,42]. Nanovesicles are typically classified based on their lipid solubility into phospholipid and non-phospholipid types. Various types of nanovesicles include ethosomes, liposomes, glycerosomes, transethosomes, bilosomes, polymersomes, exosomes, transfersomes, phytosomes, and niosomes. Numerous studies indicate the use of nanovesicles to enhance skin permeability, particularly in hydrogel form. When designing specific nanovesicles, it is crucial to evaluate and adjust key parameters such as size, composition, drug diffusion into skin layers, surface charge, and deformability. It is important to determine the size of nanovesicles. Nanovesicles over 600 nm may not penetrate deeper layers of the skin, particularly the subcutaneous layer. Smaller sizes (< 300 nm) are preferred as they can potentially penetrate even deeper into the dermal region. Nanovesicles loaded with various drugs are widely reported to effectively treat skin diseases, as summarized in Table 1.

Table 1 Summary of studies related to nanovesicles application for various skin infectious diseases

The natural characteristics of liposome enable encapsulation of both hydrophilic and lipophilic substances/drugs. Some liposome-based nanovesicles products have been approved by FDA and commercialized for medicinal purpose since they are non-toxic and effective. Liposomal-based transdermal drug has commonly been used, foe examples diclofenac, ketoprofen, and azithromycin [6]. Another phospholipid-based nanovesicle is the ethosome, composed mainly of phospholipids mixed with alcohol (20–40%, such as ethanol). Ethanol enhances the penetration flexibility of the ethosome. This nanocarrier boasts enhanced drug loading capacity and deformability, making it superior to liposomes. Modified versions like transethosomes include phospholipids, higher ethanol content, and surfactants. The increased ethanol concentration in transethosomes enhances permeation, which makes them a viable alternative for transdermal drug delivery for treating various skin conditions. Ethosomes, a lipid delivery system, have shown improved efficiency in percutaneous drug delivery due to their rounded shape that accumulates greater deposits in deeper skin layers [34]. Transdermal drugs using liposomal have been used to load a range of oral medications, including anti-hypertensive agents like valsartan, along with antifungal treatments like econazole nitrate and mitoxantrone. Transfersomes consist of a lipid bilayer incorporating phospholipids, ethanol, and an aqueous component. Typically, surfactants are added to optimize consistency. The distinct structure of transfersomes enables deep skin penetration [33]. Niosomes are non-ionic surfactants which are thermodynamically stable. They are commonly used for controlling and targeting drug delivery due to their sustained release. Typically ranging from 10 to 1000 nm in size, they offer enhanced skin layer penetration compared to liposomes [34, 73].

There are several methods for preparing nanovesicles before loading them with active compounds. The common methods for liposomes include the thin-film method and injection methods (similar to the ethanol and ether methods). Among these, the thin-film method is the most popular. In this technique, a thin lipid film forms on the inner wall of a rotary evaporator flask, which is then hydrated with a water or buffer solution. Subsequent vigorous shaking and sonication in an ultrasonic bath help the film detach from the flask and form liposomes. The objective of injection method is to ensure that the lipid suspension (for either hydrophobic or hydrophilic organic solvents) is injected into the water. For example, in ethanol injection, the first step is preparing liposomes ranging between 30 and 170 nm. When preparing liposomes using this method, the lipids dissolved in an organic solvent (in this case ethanol) are injected into the water for stirring, and then, the solvent is removed, followed by hydrating the solution during stirring for another 15 min. Ethanol can be removed from the liposome suspension through either rotary evaporation or centrifugation with a silica gel column. The ether injection method, resembling ethanol injection, enhances lipid solubility and ensures robust liposome stability since the solution does not dissolve in water. Unlike the injection method, emulsification forms the organic phase. This technique offers potentially higher encapsulation efficiencies compared to injection methods [74].

The cold method is the most commonly used approach for ethosome preparation. Phospholipids, drugs, and other lipid materials are dissolved in ethanol within a covered vessel at room temperature under vigorous stirring. The mixture is heated to 30 °C in a water bath. Water in a separate vessel is also heated to 30 °C and then added to the mixture, then stirred for five minutes in a covered vessel. Sonication or extrusion can be utilized to adjust the vesicle size of the ethosomes formulation. Finally, the formulation must be properly stored in a refrigerator. In the hot method, drugs are initially dissolved in a mixture of ethanol and propylene glycol. This mixture is then added to the phospholipid dispersion in water at 40 °C. After a five-minute mix, the preparation undergoes sonication at 4 °C for three cycles of five minutes each, with a five-minute break between each cycle, using the Probe Sonicator. The formulation is homogenized at 15,000 psi pressure in three cycles using a high-pressure homogenizer to obtain nanosized ethosomes. In the Classic Mechanical Dispersion Method, organic ingredients like Soya phosphatidylcholine are combined and dissolved in a mixture of chloroform to methanol (3:1 ratio) in a round-bottom flask. Removing the organic solvents using a rotary vacuum evaporator above the lipid transition temperature forms a thin lipid film on the flask’s wall. Remaining solvent traces are eliminated by vacuum exposure overnight. Hydration involves different concentrations of a hydroethanolic mixture containing the drug, with the flask rotated at a suitable temperature. The classic method dissolves phospholipids and drugs in ethanol, heating to 30 °C in a water bath, followed by slow addition of double distilled water while stirring at a constant 700 rpm in a closed vessel. Further homogenization involves passing the mixture through a polycarbonate membrane using a hand extruder in three cycles [75].

2.5 Liposome as strategy to increase skin permeability for topical antibiotics

2.5.1 Definition of liposome

In 1961, Alec Bangham first created liposomes and its invention has revolutionized the pharmaceutical field. The application of liposome is now established in various areas such as drug, biomolecules, and gene delivery. Liposomes are spherical vesicles consisting of a lipid bilayer encapsulating an aqueous core. They typically contain phospholipids or synthetic amphiphiles along with sterols like cholesterol to influence membrane permeability. With hydrophilic heads and hydrophobic tails, liposomes can vary in size, composition, charge, and lamellarity. They have emerged as a favored nanocarrier for numerous biomedical uses. The advancements in liposome technology have led to various liposome-based drug formulations for human use, with many products undergoing clinical trials. In 1974, Allison and Gregoriadis noted that liposomes were the first system known to provide adjuvant action due to their identified immunological role and properties. Specifically, negatively charged liposomes containing dicetyl phosphate were found to enhance immune responses against diphtheria toxoid. Liposomes are used as adjuvants in two vaccine systems: Inflexal and Epaxal. The first successful achievement in liposome-based products was the introduction of Doxil® to the US market in 1995 for the treatment of patients with ovarian cancer and AIDS-related Kaposi’s sarcoma after the failure of prior systemic chemotherapy or intolerance to such therapy. Gabizon and Barenholz initiated the development of Doxil® in Israel and the USA. It was the first nanosized liposomal product to obtain regulatory approval [76,77,78,79]. In transdermal drug delivery, liposomes have a limitation: they often adhere to the inner walls of skin cells, leading to the breakdown of phospholipid bonds and premature leakage of the encapsulated drug before reaching deep skin layers [78]. There are four main types of liposomal delivery systems: conventional liposomes, sterically-stabilized liposomes, ligand-targeted liposomes, and combinations of these. Conventional liposomes are made up of a lipid bilayer containing cationic, anionic, or neutral (phospho)lipids and cholesterol, enclosing an aqueous volume. Its formulations reduced the toxicity of compounds in vivo, through modifying pharmacokinetics and biodistribution to enhance drug delivery to diseased tissue in comparison to free drug. However, the delivery system was prone to rapid elimination from the bloodstream, therefore limiting its therapeutic efficacy. Sterically-stabilized liposomes were made to improve liposome stability and enhance their circulation times in the blood. The hydrophilic polymer, polyethylene glycol (PEG), has been shown to be the optimal choice for obtaining sterically-stabilized liposomes. This not only reduces the elimination of drugs by prolonging blood circulation and providing accumulation at pathological sites, but also attenuates side effects. Steric stabilization strongly influences the pharmacokinetics of liposomes, with reported half-lives varying from 2 to 24 h in rodents (mice and rats) and as high as 45 h in humans, depending on the particle size and the characteristics of the coating polymer. Ligand-targeted liposomes offer a vast potential for site specific delivery of drugs to designated cell types or organs in vivo, which selectively express or over-express specific ligands (e.g., receptors or cell adhesion molecules) at the site of disease. Many types of ligands are available, such as antibodies, peptides/proteins, and carbohydrates. The coupling of antibodies, particularly monoclonal antibodies, to create immunoliposomes represents one of the more versatile ligands that can be affixed to liposome surfaces [80, 81]. Improved traditional liposome compositions achieve the deeper permeation of active ingredients to different skin strata. Liposomes of ultraflexible vesicles are common vectors in transdermal drug delivery systems that are relatively liquid and deformed. It demonstrates improving skin penetration and many clinical trials were registered and some of them has been authorized as topically applied medicinal products such as Pevaryl® Lipogel, Maxilene® cream, Lipoxysan®, Supra-vir® cream [82, 83].

2.5.2 Physiochemical characters of liposome

As lipid-based delivery drug, the basic component contained in liposome is amphipathic phospholipid molecules arranged as bilayers with two distinct features on the surface and considered to share similar characteristics with biological membranes. Lipids and fatty acids are joined together to form the primary physical body of liposome (Fig. 2A). The hydrophobic part will be directed into the inner side of the molecule, while the hydrophilic part facing to the outside with both parts is attached spontaneously [84,85,86]. Surfactant is often added to multiply the liposome’s stability and elasticity. Under hydrated environment, each component of liposome is self-assembly and manifests a physical shell of hydrophobic acyl chain. Therefore, it is thermodynamically stable and could be enhanced through various interactions, such as hydrogen bonds, van der Waals forces, or other electrostatic interactions [87, 88]. The hydrophilic surface is highly polar by nature thus it easily attracts water molecules. The water-resistant surface is owned by the hydrophobic surface, and it is characterized as non-polar sites. Due to its physiochemical nature, liposome could trap both hydrophobic and hydrophilic compounds effectively [85, 89]. In general, liposome could be designed from either natural or synthetic lipids [87].

Fig. 2
figure 2

Liposome is lipid bilayer with an internal aqueous cavity. The lipid bilayer system (A) is similar to human tissue membrane that has hydrophilic heads with a hydrophilic area in the middle. The nucleus part of the liposome (B) consists of aqueous area that is important for most hydrophilic drug molecules to be kept inside the core

The utility of liposome as drug carriers has been well known for recent years. Enormous active compounds are trialed to examine liposome’s ability in encapsulating them. Various reported studies priorly have claimed the enhancement capacity of drugs molecule to reach targeted cells after bursting out from the liposome complex molecules. The difference polarities owned by liposome allow interaction with several substances, mostly drug molecules, such as antibiotics, antifungal, anti-virus, and genes. Better bioavailability is considered to improve such wanted therapeutic effects. However, enzymatic destruction often challenges the drug molecule’s quality and quantity inside the tissue or cell. The administration route will also affect since many proteins could degrade the liposome early. Even in systematic delivery, reticuloendothelial system (RES) responds to the molecules and metabolizes them instantly. The poor design and characterization are also responsible to the problem. Then, strategies are required to against the issues [86, 90, 91].

To produce high-quality liposome, it is necessary to rely on two basic matters, its size and membrane lamellarity [86]. Liposome is determined based on the synthesis process, the unilamellar vesicle with one bilayer membrane, oligolamellar vesicle with 2–5 bilayer membranes, and multilamellar vesicles with five or more bilayer membranes. The distinguished features among the three types of liposome preparations are seen through the entrapping procedure. The unilamellar vesicle needs to be trapped within a single internal aqueous compartment, while the multilamellar liposome requires some lipid combinations and organic solvent such as cholesterol glycerol, or egg lecithin and mixed with chloroform and methanol as the solvents [34, 92].

2.5.3 Nanoencapsulation method of liposome as nanocarrier

Liposomes are well-investigated nanocarriers for targeted drug delivery. They have improved therapies by stabilizing therapeutic compounds, overcoming obstacles to cellular and tissue uptake, and improving biodistribution of compounds to target sites in vivo. The large aqueous center and biocompatible lipid exterior permit the delivery of a variety of macromolecules, such as DNA, proteins, and imaging agents [80, 93].

Liposomes function as a drug delivery system, penetrating the SC as whole vesicles. This allows them to localize drugs within the SC and the viable epidermis, aiming to reduce systemic absorption and thereby minimize side effects. Besides, the advantages are suitable biocompatibility, low toxicity, and similarity to skin component. Enhancement of the liposomes ability to penetrate the epidermis and dermis layers is conducted by developing new liposomal formulations [94, 95]. Size of liposome is the important factors in vivo applications. The size of liposomes affects drug loading, biodistribution, the rate of drug clearance from the body, targeting efficacy to specific organs, and therapeutic [96].

Nanoencapsulation is the inclusion of bioactive substances or entrapment of natural compounds in carriers of nanoscale dimensions. This drug delivery method aims to enhance the solubility of various active pharmaceutical ingredients (APIs) and improve stability by shielding them from harsh environments [97]. This modification of phospholipids enhances self-sealing properties in liposomes and aqueous materials. It expands their applications across diverse fields including agriculture, food processing, cosmetics, tissue engineering, and pharmaceuticals. Phospholipid modification serves as an encapsulation structure for modifying, protecting, and targeting the delivery of essential bioactive compounds in cancer therapy [98].

Nanocarriers are extensively utilized in formulation and development for their various advantages in drug delivery. They serve as a potent method to enhance bioavailability and solubility, prolong drug action duration, and enhance drug stability. Nanoliposomes can be achieved through passive loading encapsulation, where compounds are trapped during vesicle formation: hydrophilic compounds in the aqueous phase, hydrophobic compounds in the lipid bilayer, and amphiphilic molecules in the lipid-soluble region between bilayers. On the other hand, active or remote loading entails trapping bioactive compounds within intact vesicles. This process leverages ammonium sulfate and calcium acetate to enhance encapsulation efficiency, allowing higher drug-to-lipid ratios through pumping or forcing mechanisms while ensuring controlled release by locking bioactive compounds inside the liposomes (Fig. 2B) [41, 98, 99].

2.5.4 Liposome as drug delivery for skin diseases

Topical administration of antibiotics is a common method to treat bacterial skin infections. It is convenient and less invasive compared to other treatments, efficiently accessing various skin layers. Treating a local insect bite infection with a topical antibiotic ointment is often faster and has fewer side effects than oral treatments, as it minimizes the number of drug molecules reaching the target tissue. Additionally, the topical route is cost-effective [100, 101]. The increasing issue of antimicrobial resistance poses a significant threat to antibiotics across all classes, particularly affecting Gram-negative bacteria. These bacteria can undergo complex processes to enhance their defense against antibiotic penetration, particularly within the cell membrane system. In contrast to Gram-positive bacteria, Gram-negative bacteria feature two distinct membrane layers: an outer membrane rich in a thick lipid component called lipopolysaccharide (LPS) and an inner cytoplasmic membrane abundant in peptidoglycan. These robust components effectively hinder polar molecules while exhibiting greater lipophilicity (Fig. 3A and B) [102,103,104]. Therefore, a strategy is needed to enhance bacterial eradication and decrease resistance cases effectively. Different vehicles like ointments and gels can transport antibiotics for topical administration. However, developing a durable and effective drug formulation can be a time-consuming process [89]. Many scientists combine natural-derived compounds with antibiotics to generate innovative ideas. Lipid-based drug carriers like liposomes are favored for their ability to penetrate deep into bacterial cytoplasm [105]. Hydrophilic drug molecules typically carry a positive charge that hinders bonding with the bacterial outer membrane. Bacterial cell membrane sizes, primarily composed of lipid structures, vary based on their chemical composition. Gram-positive bacteria possess a thicker cell wall (20–80 nm) compared to Gram-negative bacteria (< 10 nm) [106, 107]. The outer membrane of Gram-negative bacteria, rich in lipoprotein components (~ 70%), allows small molecules like amino acids and sugars to permeate via specific channels called porins. Similar to bacterial characteristics, liposomes also consist of two distinct lipid layers, enhancing their capacity to fuse together [49].

Fig. 3
figure 3

According to membrane structures, bacteria could be identified as Gram-positive and Gram-negative bacteria. The apparent thick peptidoglycan is shown in Gram-positive bacteria (A) and could be seen through Gram staining. Meanwhile, Gram-negative bacteria (B) only possesses thinner peptidoglycan wall but has two layers of cell membrane

For effective permeation through the skin barrier, it is essential to consider the size of the drug molecule and its carrier. Drug carriers larger than 300 nm in diameter will likely face challenges passing through skin pores [108]. Liposomes have various sizes. Small unilamellar vesicles (SUVs) are 25–50 nm in diameter, large unilamellar vesicles (LUVs) range from 50–500 nm, and large multilamellar vesicles (LMVs) are 500–10,000 nm in diameter. Smaller liposomes are generally more efficient, making LUVs the preferred size. LUVs, containing phosphatidylethanolamine, enhance liposome penetration by easily fusing with skin lipids (Fig. 4) [109]. Cholesterol is usually provided to ensure liposome stability. With its unique physiochemical characteristic, it is important to assess whether each loaded drug molecule is well capsulated. Various specific drug-trapping methods have been reported. However, each drug may generate different results. The main principal to obtain successful preparation is to elicit drug’s solubility and to find a perfect homogenous compound. Theoretically, lipophilic drug molecules can be retained within the lipid bilayer compartment of the liposome, whereas hydrophilic ones blend easily in the aqueous phase [110]. In addition to liposome flexibility in size, a key objective in topical drug delivery is to minimize allergic reactions to the compounds. Skin irritation often arises from chemical substances like surfactants. Lipid-based materials generally induce fewer side effects on the local immune system. In a study by Wu et al. [111], licorice flavonoid licochalcone A loaded into keratin liposomes (LAL) demonstrated a range of benefits for guinea pig skin. Skin irritation tests revealed no visible signs of local inflammation after application to hairless skin during three observation periods. Further analysis indicated a significant distribution of loaded substances in the skin tissue, with LAL showing higher fluorescence intensity compared to the non-liposome group [111].

Fig. 4
figure 4

Three different type of liposome according to its molecule size

Understanding the basic pharmacokinetics of specific antibiotics, particularly in topical administration, is as important as selecting appropriate delivery materials. Fluoroquinolones and aminoglycosides are concentration-dependent antibiotics that require peak concentrations to effectively eliminate targeted bacteria. Dosing issues often correlate linearly with toxic effects; higher doses lead to greater side effects. The goal is to maximize pharmacological benefits while minimizing dosage. Incorporating antibiotics into liposomes, especially in nanosized forms, is feasible. β-lactams, on the other hand, are often time-dependent antibiotics. To ensure their full bactericidal potential, frequent systemic release is necessary after administration. Timing plays a vital role in maintaining the required drug concentrations. However, maintaining a stable plasma concentration of antibiotics requires a consistent environment, but the human body’s complexity can lead to changes based on specific conditions. Addressing these changes is crucial. Compact carriers like liposomes can enhance stability and phagocytic activity for improved efficacy. In topical applications, the focus is on optimizing the permeation system to prevent inadequate antibiotic concentrations in plasma, which can elevate resistance risks. Novel antibiotic discoveries are time-intensive, while mortality rates continue to rise annually. Unlike conventional delivery materials, liposomes enable nanoparticle formulations and robust combinations with various compounds, including polymers and non-organic materials like silver and gold. Sang et al. [7] incorporated polymyxin B (PMB) into a modified liposomal system (P-lipo) and observed that the larger liposome size increased the penetration of PMB concentration into P. aeruginosa by interacting with membrane-anchoring lipid A on the outer membrane. In vivo tests supported these findings, showing a significant survival rate of up to 60% in mice injected with D-galactosamine hydrochloride-induced shock septicemia when administered P-lipo compared to the non-liposome group. Overall, P-lipo demonstrated enhanced effectiveness, particularly in terms of its ability to bind with bacterial lipopolysaccharides (LPS) [7].

2.5.5 Liposome’s ability in enhancing antibiotics permeation across skin barrier

Designing drug molecules for gaining the benefits is a challenge. To address the increasing antibiotic resistance, more research is focused on this inevitable event. Skin infections caused by bacterial pathogens such as skin normal flora, S. aureus contribute to specific antibiotic resistance called MRSA. The number of MRSA cases continue to increase every year throughout countries, as reported by Centers for Disease Control and Prevention (CDC). More than 70,000 severe infections and nearly over 9000 deaths per year have been recorded [113, 114]. The basic characteristic of MRSA is its ability to resist most beta-lactam antibiotics such as flucloxacillin, methicillin, oxacillin, and cefoxitin. The bacteria is often present in the pus of diabetic foot ulcers or chronic impetigo. However, vancomycin, the preferred treatment, is limited and inaccessible in many low- and middle-income countries [115, 116]. Unfortunately, more vancomycin-resistant S. aureus infections have been found in the last two decades with at least 11 van gene clusters that contribute to the resistance process (VanA, VanB, VanD, VanF, VanI, VanM, VanC, VanE, VanG, VanL, and VanN phenotypes). To eliminate further infections, CDC recommends systemic antibiotic for vancomycin-resistant S. aureus (VRSA) [117]. A new breakthrough in drug delivery systems allows the combination of multiple antibiotics in one carrier, including drugs from different antibiotic classes and natural substances. Given the increasing adaptability and resilience of pathogens against antibiotics, additional strategies are essential to mitigate potential harm, such as designing improved vehicles for delivering antibiotics [113, 118, 119].

The increasing research interest in liposomes as futuristic drug carriers not only enhances medicine significantly but also meets high demand for improvements. In dermatology, liposomes are extensively utilized in various drug formulations like ointments and gels. The primary concern is maintaining stability for effective penetration through skin layers. Nanopharmaceuticals aid in addressing this challenge by chemical manipulation of liposomal profiles [120, 121]. Liposomes, rich in lipid content, can penetrate deep into skin tissues due to their lipid bilayer structure. Coating antibiotics with liposomes enhances solubility, facilitating efficient transport through the skin with its negatively charged state. Introducing a more positive charge in the design can significantly amplify the visible effects (Fig. 5) [47, 122].

Fig. 5
figure 5

Entrapment ability of liposome to coat various drug molecules increases the bioavailable of drug and further enables higher plasma concentration with as smallest dosage given as possible

Liposomes can enhance the antibiotic’s ability to penetrate infection sites compared to antibiotics alone [38, 112]. Chloramphenicol-loaded DA (deoxycholic acid) liposomes double the chloramphenicol concentration deposition in the follicular level of nude mouse skin compared to an aqueous control group, with 5.8 μg/cm2 in the stratum corneum versus 4.6 μg/cm2 for chloramphenicol alone [123]. Nanosized liposomes demonstrated superior results. A study assessing nanosized rifampicin against S. aureus displayed enhanced bacterial colony reduction relative to rifampicin alone by the fifth day. Adding cationic lipids like monoolein (MO) and N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium methyl-sulfate (DOTAP) (NanoRIF) increased rifampicin’s cytotoxicity toward S. aureus. Mice treated with 25 mg/kg NanoRIF showed significantly fewer or undetectable bacteria (p < 0.05) compared to the untreated group, with the 12.5 mg/kg group exhibiting even better average reductions [105]. An additional strategy to improve sustained drug release from liposomal carriers involves combining them with water-based gel formulations. Liposome-in-hydrogel formulations demonstrate enhanced membrane-active antimicrobial activity compared to Chlorhexidine alone [124]. Another study indicates that loading vancomycin into liposomes enhances the phagocytic ability of cultured leukocytes against S. aureus. Overall, topical nanoliposomes are preferred due to their potential for recognition by bacterial cell walls through endosomal mechanisms and reduced interaction with natural biofluid [125].

2.6 Current research: in vitro and in vivo

2.6.1 Fusidic acid

Fusidic acid (FA) is a broad-spectrum antibiotic primarily used to treat various pyoderma types caused by Gram-positive bacteria, including impetigo, erythrasma, bullous impetigo, psoriasis, folliculitis, furuncles, carbuncles, and infected wounds and burns. This antibiotic, known for its low water solubility and steroidal effect, shows decreased drug concentration over time if given systemically (orally or via injection), partly due to associated adverse reactions. The topical route is often preferred due to higher patient compliance [126, 127]. Mutations in different nucleotides of the bacterial protein synthesis gene, fusA, pose an increased risk that can arise from irrational usage [128, 129]. Using nanotechnology to enhance fusidic acid (FA) bioavailability is evident in its encapsulation within nanoemulsions. Nanoemulsions (NE) are a type of nanolipid formulation, akin to liposomes, niosomes, and ethosomes. Encapsulating FA with NE leads to a significantly higher release rate (p < 0.05) compared to NE-hydrogel formulations, likely due to the presence of FA in the outer aqueous phase of the NE formulation, facilitating easier and greater drug release. In optimized NE formulations, approximately 90.66 ± 3.43% of FA is released within 6 h. The antibacterial activity against S. aureus was assessed using the disk diffusion test, showing a larger killing zone with FA-NE compared to blank FA and NE formulations alone [130]. Encapsulating fusidic acid in niosomes has shown promising results, as reported by Waqas et al. [130]. The size of FA when formulated with niosomes ranges between 377.2 and 725.4 nm. The smaller size of niosome-loaded FA may be attributed to the high cholesterol content, which increases surface energy and reduces particle size. Skin permeation studies reveal a significantly higher flux rate in an ex vivo model of mice skin with niosome-loaded FA (80.02 μg/cm2/h) compared to blank gel alone (15.98 μg/cm2/h) [127]. Another study compared the permeation profiles of FA in liposomal gel to plain FA hydrogel and creams. Over a 24 h period, approximately 75 ± 1.2% of FA was absorbed into the mice skin. Furthermore, retention tests showed higher values (1.620 ± 0.8 mg/cm2) compared to commercial cream (0.476 ± 1.4 mg/cm2) and plain hydrogel (0.659 ± 1.6 mg/cm2) [126].

2.6.2 Daptomycin

While S. aureus is part of the human skin microbiome, many skin infections are primarily caused by this bacterium. The clinical severity of such infections varies based on immunity levels. Topical antibiotics offer significant benefits against these bacteria, but excessive and improper use can result in prolonged healing times. Daptomycin is a novel cyclic lipopeptide antibiotic derived from the fungus Streptomyces roseosporus, capable of effectively targeting various Gram-positive pathogens, including MRSA, VRSA, vancomycin-resistant Enterococci (VRE), glycopeptide-resistant S. aureus (GRSA), Coagulase-negative staphylococci (CoNS), penicillin-resistant Streptococcus pneumoniae (PRSP), Clostridium difficile, Clostridium perfringens, Finegoldia magna, Bacillus megaterium, and P. acnes. [131, 132].

A study demonstrated the efficacy of daptomycin-loaded flexible liposomes using a diffusion cell method against S. aureus until visible biofilm formation occurred. The cell viability ratio of the control group to the experimental group was nearly 1:108. An in vivo experiment comparing control and daptomycin-loaded flexible liposome-treated mice showed that after four days of daptomycin application to the mice’s hairless skin, scattered and less visible S. aureus biofilm was observed through scanning electron microscopy. However, a similar outcome was noted in the positive control group that received intravenous antibiotic injections [48].

2.6.3 Tetracycline

Acne vulgaris (AV) is a major skin disease that mostly occurs during puberty. Alongside various infection skin diseases such as pyoderma, AV could develop tremendous manifestation starting from blackhead to excessive pustules. The well-known pathogen involved in AV is P. acnes. The wide range of clinical appearance in AV requires different medications. Topical antibiotic is one of the most applied medicines to treat such disease. Tetracycline family is considered to generate great magnitude of bactericidal activity and acts by inhibiting bacteria’ protein synthesis process [133, 134]. Specifically, tetracycline could disable the chemotaxis mechanism of neutrophils induced by P. acnes and reduce Interleukin-8 (IL-8) expression and phospholipase A2 to reactive oxygen species production [135].

Eroğlu et al. [54] reported that using tetracycline HCl and tretinoin-loaded hydrogel liposome on S. aureus and S. epidermidis significantly reduced bacteria life growth. The MIC was slightly different between non-hydrogel and hydrogel formulations, with the non-hydrogel showing a lower MIC compared to the hydrogel formulation.

2.6.4 Rifampicin

The skin plays a vital role as a protective barrier for the body. Any disruption in skin continuity, whether traumatic or not, can compromise immunity against external threats like pathogen infiltration. Rifampicin is known for its broad-spectrum antibacterial effects, targeting bacterial RNA synthesis. Specifically, rifampicin binds to the active site of RNA polymerase, inhibiting RNA elongation [136]. It is commonly utilized in treating chronic wounds, particularly those associated with progressive infectious skin conditions like diabetic ulcers. However, achieving a stable topical form of this antibiotic requires further research, especially in finding suitable drug delivery methods. Wallenwein et al. [137] conducted a study on encapsulating Rifampicin in nanoliposomes under an oxidative stress model. Wounds that are damaged or heal slowly often involve oxidative processes. When compared to ascorbic acid, rifampicin-loaded liposomes demonstrated similar efficacy in reducing oxidation levels and protecting rifampicin from lysosomal degradation. Additionally, higher levels of oxidation products resulted in more rifampicin molecules being released from the liposome vesicles [137]. Another study showed that rifampicin-loaded liposomes exhibited greater efficacy in killing Mycobacterium abscessus compared to a group without the antibiotic encapsulated in liposomes [138].

2.6.5 Amphotericin B

Cutaneous leishmaniasis, affecting nearly 700,000 to 1 million people annually, is primarily caused by the protozoan parasite Leishmania from the Trypanosomatidae family, leading to skin ulcers. Amphotericin B, a second-line antibiotic for leishmaniasis, acts by inducing membrane cell leakage in protozoa. Amphotericin B, a second-line antibiotic for leishmaniasis, acts by inducing membrane cell leakage in protozoa [139]. To get a greater effect against Leishmania protozoa, the topical route is the most convenient and least toxic method of administration. Smaller particle sizes are preferable for topical skin drugs. In a study by Jaafari et al. [140], the efficacy of amphotericin B-loaded liposomes (Lip-AmB) in killing Leishmania major protozoa infecting cultured macrophages was investigated. The experimental group demonstrated non-inferiority in suppressing protozoa viability compared to Fungizone© as the positive control. Notably, significant results were observed in an in vivo study. After 8 to 12 weeks of exposure to Lip-AmB 0.4%, the parasitic count in the mice spleen was significantly lower than in the placebo group (p < 0.001) [140].

2.6.6 Azithromycin

S. aureus is a prevalent skin microbiome, contributing to approximately 76% of various skin infection diseases, particularly pyoderma. In cases of diabetic ulcers, bacterial infiltration into deeper skin layers can exacerbate the condition. This bacterium typically colonizes the superficial skin layer and adheres to corneocytes. Besides pyogenic skin lesions, S. aureus is recognized as a contributing factor in atopic dermatitis (AD) [141, 142]. Azithromycin is a macrolide antibiotic which disrupts bacterial protein synthesis by binding near the peptidyl transferase center on 23S rRNA [143]. Pathogenic bacteria are the main targets of this antibiotic. However, the rise of MRSA cases globally raises concerns for clinicians due to significant cost implications. MRSA contributes to community-acquired nosocomial infections and increased morbidity and mortality from multidrug resistance (MDR). Several studies have highlighted that azithromycin’s poor water solubility makes it challenging to find effective carriers. Microemulsions represent one strategy to enhance azithromycin penetration into skin tissue [144, 145]. Another strategy is loading the antibiotic into liposomes. Rukavina et al. [47] investigated the efficacy of azithromycin-loaded liposomes in treating MRSA infections and analyzed their permeation through porcine ear skin. The study revealed that azithromycin-loaded liposomes inhibited about 90% of bacterial growth at concentrations ranging from 0.5 to 8 μg/ml, compared to free azithromycin requiring higher doses (8 to 32 μg/ml) to achieve a similar effect. The primary objective of topical drugs is to attain maximum concentrations at the target site while preventing excessive penetration beyond the superficial tissue layer. The experiment demonstrated that entrapping the antibiotic within cationic liposomes (CATL) optimized drug concentration in the stratum corneum by approximately 35%, whereas free azithromycin was more concentrated in deeper skin layers [47].

3 Conclusion

Bacteria primarily consist of essential macromolecules like lipids, proteins, and glycans. Antimicrobial agents are designed to target crucial components of bacteria’s machinery system, disrupting them by interacting with specific ligands or receptors both inside and outside the bacterial cell. Nevertheless, the increasing survival rates of bacteria and the indiscriminate prescription of antimicrobials have contributed to a higher prevalence of antimicrobial resistance. To eradicate pathogenic bacterial colonies in the human body, multiple factors are essential. Irregular dosage regimens, delayed medication intake, and abnormal health conditions can alter the normal pharmacokinetics of antimicrobial agents. For instance, insufficient plasma concentrations compared to the bacteria’s MIC can promote further resistance. The challenge of antimicrobial resistance is widely acknowledged by scientists as unavoidable. Consequently, innovative breakthroughs are imperative for future solutions [146,147,148]. Recently, futuristic drug delivery system has become more popular. Liposomes enables higher intracellular drug loading due to their nanosized design and adaptability. Liposomes are rich in phospholipids, thus they easily penetrate tissues with lipid bilayer structures. Additionally, liposomes can incorporate a variety of organic and non-organic molecules like silver, gold, and magnets. The primary goal is to achieve efficient and optimal internalized drug concentrations to effectively combat pathogenic bacteria. While many liposomal products have been approved by FDA for clinical use, their application is predominantly limited to systemic and transdermal routes. The utilization of topical liposome drug delivery, especially for antibiotics, is not yet widespread, likely due to ongoing studies. However, topical application is known for being noninvasive, convenient, and effective in treating skin infections. Misuse and overuse of this delivery system may contribute to the rise of antibiotic resistance. Some studies suggest that there is no significant difference between applying postsurgical topical antibiotics alone and non-antibiotic agents like paraffin or petrolatum for infection prevention. Although achieving efficient drug absorption poses complexities in pharmacokinetic studies, the key principle is to administer the smallest effective dosage possible or opt for the minimum therapeutic dose [13, 35, 149, 150].

In conclusion, the use of liposomes to improve the permeation of antimicrobial drugs for topical applications holds promise in addressing antibiotic resistance. More studies are required to strengthen the findings, continued with clinical setting research.

Abbreviations

MIC:

Microbial inhibition concentration

OTC:

Over the counter

P. aeruginosa :

Pseudomonas aeruginosa

S. aureus :

Staphylococcus aureus

E. faecalis :

Enterococcus faecalis

S. viridans :

Streptococcus viridans

E. coli :

Escherichia coli

H. influenza :

Haemophilus influenza

TDDDS:

Topical dermal drug delivery systems

SC:

Stratum corneum

K:

Partition coefficient

D:

Diffusion coefficient

C max :

Maximum drug concentration in the skin layers

T max :

Time required to reach Cmax

AUC:

Area under the curve

AuC:

Gold nanoparticles

FDA:

Food and drug administration

TH:

Liposome: terbinafine

CFU:

Colony-forming units

CDC:

Centers for disease control and prevention

TEWL:

Transepidermal water loss

dOFM:

Dermal open-flow microperfusion

P. acnes :

Propionibacterium acnes

MRSA:

Methicillin-resistant S. aureus

S. epidermidis :

Staphylococcus epidermidis

PK:

Proteinase K

RA:

Retinoic acid

SME:

Soyethyl morpholinium ethosulfate

SD:

Standard deviation

CPT:

Cryptotanshinone

RES:

Reticuloendothelial system

NISV:

Non-ionic surfactant vesicle

VISA:

Vancomycin-intermediate S. aureus

API:

Active pharmaceutical ingredients

LPS:

Lipopolysaccharide

SUVs:

Small unilamellar vesicles

LUVs:

Large unilamellar vesicles

LMVs:

Large multilamellar vesicles

LAL:

Licochalcone A-liposome

P-lipo:

Polymyxin B (PMB)-liposome

VRSA:

Vancomycin-resistant S. aureus

DA:

Deoxycholic acid

MO:

Monoolein

DOTAP:

N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium methyl-sulfate

NanoRIF:

Monoolein (MO) and N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium methyl-sulfate (DOTAP)

NE:

Nanoemulsions

FA:

Fusidic acid

S. roseosporus :

Streptomyces roseosporus

VRE:

Vancomycin-resistant enterococci

GRSA:

Glycopeptide-resistant S. aureus

CoNS:

Coagulase-negative Staphylococcus

PRSP:

Penicillin-resistant Streptococcus pneumoniae

AV:

Acne vulgaris

M. abscessus :

Mycobacterium abscessus

IL-8:

Interleukin-8

CL:

Cutaneous leishmaniasis

Lip-AmB:

Amphotericin B-loaded liposome

AD:

Atopic dermatitis

MDR:

Multi-drug resistant

CATL:

Cationic liposome

References

  1. Walsh TR, Efthimiou J, Dréno B (2016) Systematic review of antibiotic resistance in acne: an increasing topical and oral threat. Lancet Infect Dis 16:23–33. https://doi.org/10.1016/S1473-3099(15)00527-7

    Article  CAS  Google Scholar 

  2. Amirthalingam S, Yi KS, Ching LT, Mun NY (2015) Topical antibacterials and global challenges on resistance development. Trop J Pharm Res 14:919–924. https://doi.org/10.4314/tjpr.v14i5.24

    Article  Google Scholar 

  3. Sun J, Rutherford ST, Silhavy TJ, Huang KC (2022) Physical properties of the bacterial outer membrane. Nat Rev Microbiol 20:236–248. https://doi.org/10.1038/s41579-021-00638-0

    Article  CAS  PubMed  Google Scholar 

  4. Iyer A, Jyothi VGSS, Agrawal A, Khatri DK, Srivastava S, Singh SB, Madan J (2021) Does skin permeation kinetics influence efficacy of topical dermal drug delivery system?: Assessment, prediction, utilization, and integration of chitosan biomacromolecule for augmenting topical dermal drug delivery in skin. J Adv Pharm Technol Res 12:345–355

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Nair A, Jacob S, Al-Dhubiab B, Attimarad M, Harsha S (2013) Basic considerations in the dermatokinetics of topical formulations. Artic Braz J Pharm Sci 49:423–434. https://doi.org/10.1590/S1984-82502013000300004

    Article  CAS  Google Scholar 

  6. Hmingthansanga V, Singh N, Banerjee S, Manickam S, Velayutham R, Natesan S (2022) Improved topical drug delivery: role of permeation enhancers and advanced approaches. Pharmaceutics 14:1–31. https://doi.org/10.3390/pharmaceutics14122818

    Article  CAS  Google Scholar 

  7. Sang N, Jiang L, Wang Z, Zhu Y, Lin G, Li R, Zhang J (2022) Bacteria-targeting liposomes for enhanced delivery of cinnamaldehyde and infection management. Int J Pharm 612:1–15. https://doi.org/10.1016/j.ijpharm.2021.121356

    Article  CAS  Google Scholar 

  8. Ullah N, Khan D, Ahmad N, Zafar A, Shah KU, Rehman A, ur, (2023) Lipase-sensitive fusidic acid polymeric nanoparticles based hydrogel for on-demand delivery against MRSA-infected burn wounds. J Drug Deliv Sci Technol. https://doi.org/10.1016/j.jddst.2022.104110

    Article  Google Scholar 

  9. Abd E, Yousef SA, Pastore MN, Telaprolu K, Mohammed YH, Namjoshi S, Grice JE, Roberts MS (2016) Skin models for the testing of transdermal drugs. Clin Pharmacol 8:163–176. https://doi.org/10.2147/CPAA.S64788

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Neupane R, Boddu SHS, Renukuntla J, Babu RJ, Tiwari AK (2020) Alternatives to biological skin in permeation studies: current trends and possibilities. Pharmaceutics 12:1–25. https://doi.org/10.3390/pharmaceutics12020152

    Article  CAS  Google Scholar 

  11. Shpichka A, Butnaru D, Bezrukov EA, Sukhanov RB, Atala A, Burdukovskii V, Zhang Y, Timashev P (2019) Skin tissue regeneration for burn injury. Stem Cell Res Ther 10:1–6

    Article  Google Scholar 

  12. Ruela ALM, Perissinato AG, de Lino MES, Mudrik PS, Pereira GR (2016) Evaluation of skin absorption of drugs from topical and transdermal formulations. Braz J Pharm Sci 52:527–544. https://doi.org/10.1590/s1984-82502016000300018

    Article  CAS  Google Scholar 

  13. Rapalli VK, Singhvi G (2021) Dermato-pharmacokinetic: assessment tools for topically applied dosage forms. Expert Opin Drug Deliv 18:423–426. https://doi.org/10.1080/17425247.2021.1856071

    Article  PubMed  Google Scholar 

  14. Roberts MS, Cheruvu HS, Mangion SE, Alinaghi A, Benson HAE, Mohammed Y, Holmes A, van der Hoek J, Pastore M, Grice JE (2021) Topical drug delivery: History, percutaneous absorption, and product development. Adv Drug Deliv Rev 177:113929

    Article  CAS  PubMed  Google Scholar 

  15. Ng KW, Lau WM (2015) Skin deep: the basics of human skin structure and drug penetration. In: Dragicevic N, Maibach HI (eds) Percutaneous penetration enhancers chemical methods in penetration enhancement. Springer, Berlin, Heidelberg, pp 3–11

    Chapter  Google Scholar 

  16. Yu YQ, Yang X, Wu XF, Bin FY (2021) Enhancing permeation of drug molecules across the skin via delivery in nanocarriers: novel strategies for effective transdermal applications. Front Bioeng Biotechnol 9:646554

    Article  PubMed  PubMed Central  Google Scholar 

  17. Ghasemiyeh P, Mohammadi-Samani S (2020) Potential of nanoparticles as permeation enhancers and targeted delivery options for skin: advantages and disadvantages. Drug Des Dev Ther 14:3271–3289. https://doi.org/10.2147/DDDT.S264648

    Article  CAS  Google Scholar 

  18. Obata Y, Takayama K (2017) Related topic: determination of partition coefficient from vehicle to skin. Skin permeation and disposition of therapeutic and cosmeceutical compounds. Springer, Taikoo, pp 385–389

    Chapter  Google Scholar 

  19. Sugibayashi K (2017) Skin permeation of chemicals. Skin permeation and disposition of therapeutic and cosmeceutical compounds. Springer, Taikoo, pp 13–53

    Chapter  Google Scholar 

  20. The National Institute for Occupational Safety and Health (NIOSH) (2022) Skin permeation calculator. https://www.cdc.gov/niosh/topics/skin/skinpermcalc.html. Accessed 6 Mar 2024

  21. Ellison CA, Tankersley KO, Obringer CM, Carr GJ, Manwaring J, Rothe H, Duplan H, Géniès C, Grégoire S, Hewitt NJ, Jamin CJ, Klaric M, Lange D, Rolaki A, Schepky A (2020) Partition coefficient and diffusion coefficient determinations of 50 compounds in human intact skin, isolated skin layers and isolated stratum corneum lipids. Toxicol in Vitro. https://doi.org/10.1016/j.tiv.2020.104990

  22. Rothe H, Obringer C, Manwaring J, Avci C, Wargniez W, Eilstein J, Hewitt N, Cubberley R, Duplan H, Lange D, Jacques-Jamin C, Klaric M, Schepky A, Grégoire S (2017) Comparison of protocols measuring diffusion and partition coefficients in the stratum corneum. J Appl Toxicol 37:806–816. https://doi.org/10.1002/jat.3427

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sano T (2017) Related topic: Skin permeation of topical formulations. Skin permeation and disposition of therapeutic and cosmeceutical compounds. Springer, Taikoo, pp 93–99

    Chapter  Google Scholar 

  24. Ooi K (2017) Related topic: chemical permeation through impaired skin. Skin permeation and disposition of therapeutic and cosmeceutical compounds. Springer, Taikoo, pp 87–91

    Chapter  Google Scholar 

  25. Szunerits S, Boukherroub R (2018) Heat: a highly efficient skin enhancer for transdermal drug delivery. Front Bioeng Biotechnol 6:15

    Article  PubMed  PubMed Central  Google Scholar 

  26. Chandrashekar NS, Rani HS (2008) Physicochemical and pharmacokinetic parameters in drug selection and loading for transdermal drug delivery. Indian J Pharm Sci 70:94–96. https://doi.org/10.4103/0250-474X.40340

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Haftek M (2014) “Memory” of the stratum corneum: exploration of the epidermis’ past. Br J Dermatol 171:6–9

    Article  CAS  PubMed  Google Scholar 

  28. Desoqi MH, El-Sawy HS, Khafagy E, Ghorab M, Gad S (2021) Drug delivery systems for topical treatment of inflammatory skin diseases. Rec Pharm Biomed Sci 5:59–64. https://doi.org/10.21608/RPBS.2021.62807.1094

    Article  Google Scholar 

  29. Ishida-Yamamoto A, Igawa S, Kishibe M (2018) Molecular basis of the skin barrier structures revealed by electron microscopy. Exp Dermatol 27:841–846

    Article  CAS  PubMed  Google Scholar 

  30. Escobar-Chávez JJ, Merino-Sanjuán V, López-Cervantes M, Urban-Morlan Z, Piñón-Segundo E, Quintanar-Guerrero D, Ganem-Quintanar A (2008) The tape-stripping technique as a method for drug quantification in skin. J Pharm Pharm Sci 11:104–130. https://doi.org/10.18433/J3201Z

    Article  PubMed  Google Scholar 

  31. Bodenlenz M, Tiffner KI, Raml R, Augustin T, Dragatin C, Birngruber T, Schimek D, Schwagerle G, Pieber TR, Raney SG, Kanfer I, Sinner F (2017) Open flow microperfusion as a dermal pharmacokinetic approach to evaluate topical bioequivalence. Clin Pharmacokinet 56:91–98. https://doi.org/10.1007/s40262-016-0442-z

    Article  CAS  PubMed  Google Scholar 

  32. de Oliveira RS, Fantaus SS, Guillot AJ, Melero A, Beck RCR (2021) 3d-printed products for topical skin applications: from personalized dressings to drug delivery. Pharmaceutics 13:1946

    Article  PubMed  PubMed Central  Google Scholar 

  33. Bani KS, Bhardwaj K (2021) Topical drug delivery therapeutics, drug absorption and penetration enhancement techniques. J Drug Deliv Ther 11:105–110. https://doi.org/10.22270/jddt.v11i4.4864

    Article  CAS  Google Scholar 

  34. Raina N, Rani R, Thakur VK, Gupta M (2023) New insights in topical drug delivery for skin disorders: from a nanotechnological perspective. ACS Omega 8:19145–19167. https://doi.org/10.1021/acsomega.2c08016

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Benson HAE, Grice JE, Mohammed Y, Namjoshi S, Roberts MS (2019) Topical and transdermal drug delivery: from simple potions to smart technologies. Curr Drug Deliv 16:444–460. https://doi.org/10.2174/1567201816666190201143457

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Singh Malik D, Mital N, Kaur G (2016) Topical drug delivery systems: a patent review. Expert Opin Ther Pat 26:213–228

    Article  CAS  PubMed  Google Scholar 

  37. Gao W, Vecchio D, Li J, Zhu J, Zhang Q, Fu V, Li J, Thamphiwatana S, Lu D, Zhang L (2014) Hydrogel containing nanoparticle-stabilized liposomes for topical antimicrobial delivery. ACS Nano 8:2900–2907. https://doi.org/10.1021/nn500110a

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ferreira M, Ogren M, Dias JNR, Silva M, Gil S, Tavares L, Aires-Da-silva F, Gaspar MM, Aguiar SI (2021) Liposomes as antibiotic delivery systems: a promising nanotechnological strategy against antimicrobial resistance. Molecules 26:1–25. https://doi.org/10.3390/molecules26072047

    Article  CAS  Google Scholar 

  39. Patra JK, Das G, Fraceto LF, Campos EVR, Rodriguez-Torres MDP, Acosta-Torres LS, Diaz-Torres LA, Grillo R, Swamy MK, Sharma S, Habtemariam S, Shin HS (2018) Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnol 16:1–33. https://doi.org/10.1186/s12951-018-0392-8

    Article  CAS  Google Scholar 

  40. Touitou E, Natsheh H (2024) The evolution of emerging nanovesicle technologies for enhanced delivery of molecules into and across the skin. Pharmaceutics 16:1–23. https://doi.org/10.3390/pharmaceutics16020267

    Article  CAS  Google Scholar 

  41. Aguilar-Pérez KM, Avilés-Castrillo JI, Medina DI, Parra-Saldivar R, Iqbal HMN (2020) Insight into nanoliposomes as smart nanocarriers for greening the twenty-first century biomedical settings. Front Bioeng Biotechnol 8:579536

    Article  PubMed  PubMed Central  Google Scholar 

  42. Salatin S, Lotfipour F, Jelvehgari M (2019) A brief overview on nano-sized materials used in the topical treatment of skin and soft tissue bacterial infections. Expert Opin Drug Deliv 16:1313–1331. https://doi.org/10.1080/17425247.2020.1693998

    Article  CAS  PubMed  Google Scholar 

  43. Shah N, Patel K (2023) Formulation, development and characterization of liposome-based gel of eberconazole nitrate for topical delivery. Int J Pharm Investig 13:485–495. https://doi.org/10.5530/ijpi.13.3.060

    Article  CAS  Google Scholar 

  44. Bicho J, Marto J, Salgado A, Raposo S, Simões S, Ribeiro HM (2016) Lipid nanocarriers for ketoconazole topical delivery. Gavin J Dermatol Res Therapy 2016:7–13

    Google Scholar 

  45. Bavarsad N, Kouchak M, Mohamadipour P, Sadeghi-Nejad B (2016) Preparation and physicochemical characterization of topical chitosan-based film containing griseofulvin-loaded liposomes. J Adv Pharm Technol Res 7:91–98. https://doi.org/10.4103/2231-4040.184591

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kinski S, Antonieta AM, Nicolas C, Alfredo I (2019) Evaluation of the permeation and penetration of two formulations of terbinafine chlorhydrate incorporated in liposomes (Cream 1%) vs. a conventional formulation (Cream 1%), in an <i>in Vitro-ex Vivo</i> Model. J Biosci Med (Irvine) 07:119–133. https://doi.org/10.4236/jbm.2019.78010

    Article  CAS  Google Scholar 

  47. Rukavina Z, Šegvić Klarić M, Filipović-Grčić J, Lovrić J, Vanić Ž (2018) Azithromycin-loaded liposomes for enhanced topical treatment of methicillin-resistant Staphyloccocus aureus (MRSA) infections. Int J Pharm 553:109–119. https://doi.org/10.1016/j.ijpharm.2018.10.024

    Article  CAS  PubMed  Google Scholar 

  48. Li C, Zhang X, Huang X, Wang X, Liao G, Chen Z (2013) Preparation and characterization of flexible nanoliposomes loaded with daptomycin, a novel antibiotic, for topical skin therapy. Int J Nanomed 8:1285–1292. https://doi.org/10.2147/IJN.S41695

    Article  CAS  Google Scholar 

  49. Hajiahmadi F, Alikhani MY, Shariatifar H, Arabestani MR, Ahmadvand D (2019) The bactericidal effect of liposomal vancomycin as a topical combating system against Methicillin-resistant Staphylococcus aureus skin wound infection in mice. Med J Islam Repub Iran 33:1–7. https://doi.org/10.34171/mjiri.33.153

    Article  Google Scholar 

  50. Hurler J, Sørensen KK, Fallarero A, Vuorela P, Škalko-Basnet N (2013) Liposomes-in-hydrogel delivery system with mupirocin: in vitro antibiofilm studies and in vivo evaluation in mice burn model. Biomed Res Int 2013:1–8. https://doi.org/10.1155/2013/498485

    Article  CAS  Google Scholar 

  51. Li S, Wang Y, Wang S, Xie J, Fu T, Li S (2023) In situ gelling hydrogel loaded with berberine liposome for the treatment of biofilm-infected wounds. Front Bioeng Biotechnol 11:1–13. https://doi.org/10.3389/fbioe.2023.1189010

    Article  Google Scholar 

  52. Anjani QK, Pandya AK, Demartis S, Domínguez-Robles J, Moreno-Castellanos N, Li H, Gavini E, Patravale VB, Donnelly RF (2023) Liposome-loaded polymeric microneedles for enhanced skin deposition of rifampicin. Int J Pharm 646:1–20. https://doi.org/10.1016/j.ijpharm.2023.123446

    Article  CAS  Google Scholar 

  53. Peralta MF, Guzmán ML, Pérez AP, Apezteguia GA, Fórmica ML, Romero EL, Olivera ME, Carrer DC (2018) Liposomes can both enhance or reduce drugs penetration through the skin. Sci Rep 8:1–11. https://doi.org/10.1038/s41598-018-31693-y

    Article  CAS  Google Scholar 

  54. Eroğlu İ, Aslan M, Yaman Ü, Gultekinoglu M, Çalamak S, Kart D, Ulubayram K (2020) Liposome-based combination therapy for acne treatment. J Liposome Res 30:263–273. https://doi.org/10.1080/08982104.2019.1630646

    Article  CAS  PubMed  Google Scholar 

  55. Tsai MJ, Lin CY, Trousil J, Sung CT, Lee MH, Fang JY, Yang SC (2023) Proteinase K/retinoic acid-loaded cationic liposomes as multifunctional anti-acne therapy to disorganize biofilm and regulate keratinocyte proliferation. Int J Nanomedicine 18:3879–3896. https://doi.org/10.2147/IJN.S416966

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Arooj A, Rehman AU, Iqbal M, Naz I, Alhodaib A, Ahmed N (2023) Development of adapalene loaded liposome based gel for acne. Gels 9:1–16. https://doi.org/10.3390/gels9020135

    Article  CAS  Google Scholar 

  57. Patel A, Dayaramani R (2022) Novel formulation for facial acne using liposomal gel containing lipid soluble naphthoic acid derivative. Indian J Pharm Educ Res 56:245–252. https://doi.org/10.5530/ijper.56.2s.95

    Article  CAS  Google Scholar 

  58. Chorachoo J, Amnuaikit T, Voravuthikunchai SP (2013) Liposomal encapsulated rhodomyrtone: a novel antiacne drug. Evid-based Complement Altern Med. https://doi.org/10.1155/2013/157635

    Article  Google Scholar 

  59. Rahman SA, Abdelmalak NS, Badawi A, Elbayoumy T, Sabry N, El Ramly A (2016) Tretinoin-loaded liposomal formulations: from lab to comparative clinical study in acne patients. Drug Deliv 23:1184–1193. https://doi.org/10.3109/10717544.2015.1041578

    Article  CAS  PubMed  Google Scholar 

  60. Pasca PM, Miere F, Antonescu A, Fritea L, Banica F, Vicas SI, Laslo V, Zaha DC, Cavalu S (2022) Novel liposomal formulation with azelaic acid: preparation, characterization, and evaluation of biological properties. Appl Sci (Switzerland). https://doi.org/10.3390/app122413039

    Article  Google Scholar 

  61. Madan S, Nehate C, Barman TK, Rathore AS, Koul V (2019) Design, preparation, and evaluation of liposomal gel formulations for treatment of acne: in vitro and in vivo studies. Drug Dev Ind Pharm 45:395–404. https://doi.org/10.1080/03639045.2018.1546310

    Article  CAS  PubMed  Google Scholar 

  62. Huanbutta K, Rattanachitthawat N, Luangpraditkun K, Sriamornsak P, Puri V, Singh I, Sangnim T (2022) Development and evaluation of ethosomes loaded with zingiber zerumbet linn rhizome extract for antifungal skin infection in deep layer skin. Pharmaceutics 14:1–12. https://doi.org/10.3390/pharmaceutics14122765

    Article  CAS  Google Scholar 

  63. Aljohani AA, Alanazi MA, Munahhi LA, Hamroon JD, Mortagi Y, Qushawy M, Soliman GM (2023) Binary ethosomes for the enhanced topical delivery and antifungal efficacy of ketoconazole. OpenNano 11:1–15. https://doi.org/10.1016/j.onano.2023.100145

    Article  Google Scholar 

  64. Shen T, Tian B, Liu W, Yang X, Sheng Q, Li M, Wang H, Wang X, Zhou H, Han Y, Ding C, Sai S (2024) Transdermal administration of farnesol-ethosomes enhances the treatment of cutaneous candidiasis induced by Candida albicans in mice. Microbiol Spectr. https://doi.org/10.1128/spectrum.04247-23

    Article  PubMed  PubMed Central  Google Scholar 

  65. Afifudin Al-Anshori A, Nafisah Putri A, Noviana Ismi A, Kharis Suhud M, Plumeriastuti H, Maslachah L (2023) Efficacy of transdermal delivery nano ethosomal gel from ashitaba leaves on in-vivo burn wound healing in albino rats. J Med Vet 6:145–154. https://doi.org/10.20473/jmv.vol6.iss2.2023

    Article  Google Scholar 

  66. Yu Z, Lv H, Han G, Ma K (2016) Ethosomes loaded with cryptotanshinone for acne treatment through topical gel formulation. PLoS ONE 11:1–11. https://doi.org/10.1371/journal.pone.0159967

    Article  CAS  Google Scholar 

  67. Rani S, David N, Hui MS, Pin CF, Ci FY, Rajabalaya R (2013) Formulation and in vitro evaluation of ethosomes as vesicular carrier for enhanced topical delivery of isotretinoin. Int J Drug Deliv 5:28–34

    Google Scholar 

  68. Niroumand U, Motazedian MH, Ahmadi F, Asgari Q, Bahreini MS, Ghasemiyeh P, Mohammadi-Samani S (2024) Preparation and characterization of artemether-loaded niosomes in Leishmania major-induced cutaneous leishmaniasis. Sci Rep 14:1–15. https://doi.org/10.1038/s41598-024-60883-0

    Article  CAS  Google Scholar 

  69. Sangboonruang S, Semakul N, Obeid MA, Ruano M, Kitidee K, Anukool U, Pringproa K, Chantawannakul P, Ferro VA, Tragoolpua Y, Tragoolpua K (2021) Potentiality of melittin-loaded niosomal vesicles against vancomycin-intermediate staphylococcus aureus and staphylococcal skin infection. Int J Nanomed 16:7639–7661. https://doi.org/10.2147/IJN.S325901

    Article  CAS  Google Scholar 

  70. Naveen KR, Sayani B (2021) A niosomal gel of cefoperazone sodium for topical application. Acta Pharm Sci 59:483–499. https://doi.org/10.23893/1307-2080.APS.05929

    Article  CAS  Google Scholar 

  71. Goyal MK, Qureshi J (2019) Formulation and evaluation of itraconazole niosomal gel for topical application. J Drug Deliv Ther. https://doi.org/10.22270/jddt.v9i4-s.3680

    Article  Google Scholar 

  72. Shah A, Boldhane S, Pawar A, Bothiraja C (2020) Advanced development of a non-ionic surfactant and cholesterol material based niosomal gel formulation for the topical delivery of anti-acne drugs. Mater Adv 1:1763–1774. https://doi.org/10.1039/d0ma00298d

    Article  CAS  Google Scholar 

  73. Muzzalupo R, Tavano L (2015) Niosomal drug delivery for transdermal targeting: recent advances. Res Rep Transdermal Drug Deliv 4:23–33. https://doi.org/10.2147/rrtd.s64773

    Article  CAS  Google Scholar 

  74. Šturm L, Ulrih NP (2021) Basic methods for preparation of liposomes and studying their interactions with different compounds, with the emphasis on polyphenols. Int J Mol Sci 22:6547

    Article  PubMed  PubMed Central  Google Scholar 

  75. Verma P, Pathak K (2010) Therapeutic and cosmeceutical potential of ethosomes: an overview. J Adv Pharm Technol Res 1:274–282

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Bulbake U, Doppalapudi S, Kommineni N, Khan W (2017) Liposomal formulations in clinical use: an updated review. Pharmaceutics 9:1–33. https://doi.org/10.3390/pharmaceutics9020012

    Article  CAS  Google Scholar 

  77. Perrie Y, Crofts F, Devitt A, Griffiths HR, Kastner E, Nadella V (2016) Designing liposomal adjuvants for the next generation of vaccines. Adv Drug Deliv Rev 99:85–96

    Article  CAS  PubMed  Google Scholar 

  78. Jijie R, Barras A, Boukherroub R, Szunerits S (2017) Nanomaterials for transdermal drug delivery: beyond the state of the art of liposomal structures. J Mater Chem B 5:8653–8675. https://doi.org/10.1039/c7tb02529g

    Article  CAS  PubMed  Google Scholar 

  79. Ramadon D, McCrudden MTC, Courtenay AJ, Donnelly RF (2022) Enhancement strategies for transdermal drug delivery systems: current trends and applications. Drug Deliv Transl Res 12:758–791. https://doi.org/10.1007/s13346-021-00909-6

    Article  PubMed  Google Scholar 

  80. Sercombe L, Veerati T, Moheimani F, Wu SY, Sood AK, Hua S (2015) Advances and challenges of liposome assisted drug delivery. Front Pharmacol 6:1–13. https://doi.org/10.3389/fphar.2015.00286

    Article  CAS  Google Scholar 

  81. Yokota M, Tokudome Y (2017) Related topic: liposomes-recent advances and future perspective. In: Sugibayashi K (ed) Skin permeation and disposition of therapeutic and cosmeceutical compounds, 1st edn. Springer, Taikoo, pp 137–142

    Chapter  Google Scholar 

  82. Sudhakar K, Fuloria S, Subramaniyan V, Sathasivam KV, Azad AK, Swain SS, Sekar M, Karupiah S, Porwal O, Sahoo A, Meenakshi DU, Sharma VK, Jain S, Charyulu RN, Fuloria NK (2021) Ultraflexible liposome nanocargo as a dermal and transdermal drug delivery system. Nanomaterials 11:2557

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Schlich M, Musazzi UM, Campani V, Biondi M, Franzé S, Lai F, De Rosa G, Sinico C, Cilurzo F (2022) Design and development of topical liposomal formulations in a regulatory perspective. Drug Deliv Transl Res 12:1811–1828. https://doi.org/10.1007/s13346-021-01089-z

    Article  PubMed  Google Scholar 

  84. Pierre MBR, Dos Santos Miranda Costa I (2011) Liposomal systems as drug delivery vehicles for dermal and transdermal applications. Arch Dermatol Res 303:607–621. https://doi.org/10.1007/s00403-011-1166-4

    Article  CAS  PubMed  Google Scholar 

  85. Carita AC, Eloy JO, Chorilli M, Lee RJ, Leonardi GR (2017) Recent advances and perspectives in liposomes for cutaneous drug delivery. Curr Med Chem 25:606–635. https://doi.org/10.2174/0929867324666171009120154

    Article  CAS  Google Scholar 

  86. Large DE, Abdelmessih RG, Fink EA, Auguste DT (2021) Liposome composition in drug delivery design, synthesis, characterization, and clinical application. Adv Drug Deliv Rev 176:1–14. https://doi.org/10.1016/j.addr.2021.113851

    Article  CAS  Google Scholar 

  87. Kanásová M, Nesměrák K (2017) Systematic review of liposomes’ characterization methods. Monatsh Chem 148:1581–1593. https://doi.org/10.1007/s00706-017-1994-9

    Article  CAS  Google Scholar 

  88. El Maghraby GMM, Williams AC, Barry BW (2010) Can drug-bearing liposomes penetrate intact skin? J Pharm Pharmacol 58:415–429. https://doi.org/10.1211/jpp.58.4.0001

    Article  CAS  Google Scholar 

  89. Ray P, Singh S, Gupta S (2019) Topical antimicrobial therapy: current status and challenges. Indian J Med Microbiol 37:299–308. https://doi.org/10.4103/ijmm.ijmm_19_443

    Article  PubMed  Google Scholar 

  90. Ostrózka-Cieślik A, Sarecka-Hujar B (2017) The use of nanotechnology in modern pharmacotherapy. In: Grumezescu AM (ed) Multifunctional systems for combined delivery, biosensing and diagnostics, 1st edn. Elsevier, pp 139–158

    Chapter  Google Scholar 

  91. Sriwidodo UAK, Wathoni N, Zothantluanga JH, Das S, Luckanagul JA (2022) Liposome-polymer complex for drug delivery system and vaccine stabilization. Heliyon 8:1–16. https://doi.org/10.1016/j.heliyon.2022.e08934

    Article  CAS  Google Scholar 

  92. Alavi M, Karimi N, Safaei M (2017) Application of various types of liposomes in drug delivery systems. Adv Pharm Bull 7:3–9. https://doi.org/10.15171/apb.2017.002

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Dan N (2018) Structure and kinetics of synthetic, lipid-based nucleic acid carriers: lipoplexes. In: Grumezescu AM (ed) Lipid nanocarriers for drug targeting, 1st edn. Elsevier, Amsterdam, pp 529–562

    Chapter  Google Scholar 

  94. Cui M, Wiraja C, Wan S, Chew T, Xu C (2020) Nano-delivery systems for topical management of skin disorders. Mol Pharm 18:491–505

    Article  PubMed  Google Scholar 

  95. Waghule T, Gorantla S, Rapalli VK, Shah P, Dubey SK, Saha RN, Singhvi G (2020) Emerging trends in topical delivery of curcumin through lipid nanocarriers: effectiveness in skin disorders. AAPS PharmSciTech 21:1–12

    Article  Google Scholar 

  96. Has C, Sunthar P (2020) A comprehensive review on recent preparation techniques of liposomes. J Liposome Res 30:336–365. https://doi.org/10.1080/08982104.2019.1668010

    Article  CAS  PubMed  Google Scholar 

  97. Witika BA, Makoni PA, Matafwali SK, Chabalenge B, Mwila C, Kalungia AC, Nkanga CI, Bapolisi AM, Walker RB (2020) Biocompatibility of biomaterials for nanoencapsulation: current approaches. Nanomaterials 10:1–40. https://doi.org/10.3390/nano10091649

    Article  CAS  Google Scholar 

  98. Subramani T, Ganapathyswamy H (2020) An overview of liposomal nano-encapsulation techniques and its applications in food and nutraceutical. J Food Sci Technol 57:3545–3555. https://doi.org/10.1007/s13197-020-04360-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Xia H, Tang Y, Huang R, Liang J, Ma S, Chen D, Feng Y, Lei Y, Zhang Q, Yang Y, Huang Y (2022) Nanoliposome use to improve the stability of phenylethyl resorcinol and serve as a skin penetration enhancer for skin whitening. Coatings. https://doi.org/10.3390/coatings12030362

    Article  Google Scholar 

  100. Williamson DA, Carter GP, Howden BP (2017) Current and emerging topical antibacterials and antiseptics: agents, action, and resistance patterns. Clin Microbiol Rev 30:827–860. https://doi.org/10.1128/CMR

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Dallo M, Patel K, Hebert AA (2023) Topical antibiotic treatment in dermatology. Antibiotics 12:1–13. https://doi.org/10.3390/antibiotics12020188

    Article  CAS  Google Scholar 

  102. Scorciapino MA, Acosta-Gutierrez S, Benkerrou D, D’Agostino T, Malloci G, Samanta S, Bodrenko I, Ceccarelli M (2017) Rationalizing the permeation of polar antibiotics into Gram-negative bacteria. J Phys Condens Matter 29:1–16. https://doi.org/10.1088/1361-648X/aa543b

    Article  Google Scholar 

  103. Epand RM, Walker C, Epand RF, Magarvey NA (2016) Molecular mechanisms of membrane targeting antibiotics. Biochim Biophys Acta Biomembr 1858:980–987. https://doi.org/10.1016/j.bbamem.2015.10.018

    Article  CAS  Google Scholar 

  104. Malanovic N, Lohner K (2016) Gram-positive bacterial cell envelopes: the impact on the activity of antimicrobial peptides. Biochim Biophys Acta Biomembr 1858:936–946. https://doi.org/10.1016/j.bbamem.2015.11.004

    Article  CAS  Google Scholar 

  105. Walduck A, Sangwan P, Vo QA, Ratcliffe J, White J, Muir BW, Tran N (2020) Treatment of Staphylococcus aureus skin infection in vivo using rifampicin loaded lipid nanoparticles. RSC Adv 10:33608–33619. https://doi.org/10.1039/d0ra06120d

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Strahl H, Errington J (2017) Bacterial membranes: structure, domains, and function. Annu Rev Microbiol 71:519–538. https://doi.org/10.1146/annurev-micro-102215

    Article  CAS  PubMed  Google Scholar 

  107. Mai-Prochnow A, Clauson M, Hong J, Murphy AB (2016) Gram positive and Gram negative bacteria differ in their sensitivity to cold plasma. Sci Rep 6:1–11. https://doi.org/10.1038/srep38610

    Article  CAS  Google Scholar 

  108. Hua S (2015) Lipid-based nano-delivery systems for skin delivery of drugs and bioactives. Front Pharmacol 6:1–5. https://doi.org/10.3389/fphar.2015.00219

    Article  CAS  Google Scholar 

  109. Kamra M, Diwan A (2017) Liposomes in dermatological disease. J Appl Pharm Res 5:1–8

    CAS  Google Scholar 

  110. Zoabi A, Touitou E, Margulis K (2021) Recent advances in nanomaterials for dermal and transdermal applications. Colloids Interfaces 5:1–69. https://doi.org/10.3390/colloids5010018

    Article  CAS  Google Scholar 

  111. Wu W, Wang Z, Wu Y, Wu H, Chen T, Xue Y, Wang Y, Jiang C, Shen C, Liu L, Zhu H, Liu Q (2022) Mechanisms of penetration enhancement and transport utilizing skin keratine liposomes for the topical delivery of licochalcone A. Molecules 27:1–12. https://doi.org/10.3390/molecules27082504

    Article  CAS  Google Scholar 

  112. Ferreira M, Pinto SN, Aires-da-silva F, Bettencourt A, Aguiar SI, Gaspar MM (2021) Liposomes as a nanoplatform to improve the delivery of antibiotics into staphylococcus aureus biofilms. Pharmaceutics 13:1–25. https://doi.org/10.3390/pharmaceutics13030321

    Article  CAS  Google Scholar 

  113. Nandhini P, Kumar P, Mickymaray S, Alothaim AS, Somasundaram J, Rajan M (2022) Recent developments in methicillin-resistant Staphylococcus aureus (MRSA) treatment: a review. Antibiotics 11:1–21. https://doi.org/10.3390/antibiotics11050606

    Article  CAS  Google Scholar 

  114. Centers for Disease Control and Prevention (2020) Preventing infections of methicillin-resistant Staphylococcus aureus (MRSA). https://www.cdc.gov/mrsa/healthcare/inpatient.html#:~:text=CDC%20estimates%20that%20MRSA%20is,implement%20prevention%20actions%2C%20and. Accessed 6 Mar 2024

  115. Turbawaty DK, Logito V, Tjandrawati A (2021) Methicillin-resistant Staphylococcus aureus (MRSA) patterns and antibiotic susceptibility in surgical and non-surgical patients in a tertiary hospital in Indonesia. Majalah Kedokteran Bandung 53:148–154. https://doi.org/10.15395/mkb.v53n3.2396

    Article  Google Scholar 

  116. Iskandar K, Molinier L, Hallit S, Sartelli M, Hardcastle TC, Haque M, Lugova H, Dhingra S, Sharma P, Islam S, Mohammed I, Naina Mohamed I, Hanna PA, El HS, Jamaluddin NAH, Salameh P, Roques C (2021) Surveillance of antimicrobial resistance in low- and middle-income countries: a scattered picture. Antimicrob Resist Infect Control 10:1–19. https://doi.org/10.1186/s13756-021-00931-w

    Article  Google Scholar 

  117. Cong Y, Yang S, Rao X (2020) Vancomycin resistant Staphylococcus aureus infections: a review of case updating and clinical features. J Adv Res 21:169–176. https://doi.org/10.1016/j.jare.2019.10.005

    Article  PubMed  Google Scholar 

  118. Kuok CF, Hoi SO, Hoi CF, Chan CH, Fong IH, Ngok CK, Meng LR, Fong P (2017) Synergistic antibacterial effects of herbal extracts and antibiotics on methicillin-resistant Staphylococcus aureus: a computational and experimental study. Exp Biol Med 242:731–743. https://doi.org/10.1177/1535370216689828

    Article  CAS  Google Scholar 

  119. Ghosh R, De M (2023) Liposome-based antibacterial delivery: an emergent approach to combat bacterial infections. ACS Omega 8:35442–35451. https://doi.org/10.1021/acsomega.3c04893

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Raszewska-Famielec M, Flieger J (2022) Nanoparticles for topical application in the treatment of skin dysfunctions—an overview of dermo-cosmetic and dermatological products. Int J Mol Sci 23:1–54. https://doi.org/10.3390/ijms232415980

    Article  CAS  Google Scholar 

  121. Hallaj-Nezhadi S, Hassan M (2015) Nanoliposome-based antibacterial drug delivery. Drug Deliv 22:581–589. https://doi.org/10.3109/10717544.2013.863409

    Article  CAS  PubMed  Google Scholar 

  122. Dong D, Thomas N, Thierry B, Vreugde S, Prestidge CA, Wormald PJ (2015) Distribution and Inhibition of liposomes on Staphylococcus aureus and Pseudomonas aeruginosa biofilm. PLoS ONE 10:1–16. https://doi.org/10.1371/journal.pone.0131806

    Article  CAS  Google Scholar 

  123. Hsu CY, Yang SC, Sung CT, Weng YH, Fang JY (2017) Anti-MRSA malleable liposomes carrying chloramphenicol for ameliorating hair follicle targeting. Int J Nanomedicine 12:8227–8238. https://doi.org/10.2147/IJN.S147226

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Pettersen AK (2020) Liposomal formulations for membrane active antimicrobials-Assuring safety through an optimised drug delivery system

  125. Vishwasrao K, Surti A, Srinivasan R (2018) Susceptibility of methicillin resistant Staphylococcus aureus to vancomycin using liposomal drug delivery system. Quant Biol. https://doi.org/10.48550/arXiv.1801.04824

    Article  Google Scholar 

  126. Wadhwa S, Singh B, Sharma G, Raza K, Katare OP (2016) Liposomal fusidic acid as a potential delivery system: a new paradigm in the treatment of chronic plaque psoriasis. Drug Deliv 23:1204–1213. https://doi.org/10.3109/10717544.2015.1110845

    Article  CAS  PubMed  Google Scholar 

  127. Waqas MK, Sadia H, Khan MI, Omer MO, Siddique MI, Qamar S, Zaman M, Butt MH, Mustafa MW, Rasool N (2022) Development and characterization of niosomal gel of fusidic acid: in-vitro and ex-vivo approaches. Des Monomers Polym 25:165–174. https://doi.org/10.1080/15685551.2022.2086411

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Boloki HA, Al-Musaileem WF, Alfouzan W, Verghese T, Udo EE (2021) Fusidic acid resistance determinants in methicillin-resistant Staphylococcus aureus isolated in kuwait hospitals. Med Princ Pract 30:542–549. https://doi.org/10.1159/000518408

    Article  PubMed  PubMed Central  Google Scholar 

  129. Zhao H, Wang X, Wang B, Xu Y, Rao L, Wan B, Guo Y, Wu X, Yu J, Chen L, Li M, Yu F (2021) The prevalence and determinants of fusidic acid resistance among methicillin-resistant Staphylococcus aureus clinical isolates in China. Front Med (Lausanne). https://doi.org/10.3389/fmed.2021.761894

    Article  PubMed  PubMed Central  Google Scholar 

  130. Elsewedy HS, Shehata TM, Genedy SM, Siddiq KM, Asiri BY, Alshammari RA, Bukhari SI, Kola-Mustapha AT, Ramadan HA, Soliman WE (2024) Enhancing the topical antibacterial activity of fusidic acid via embedding into cinnamon oil nano-lipid carrier. Gels 10:268. https://doi.org/10.3390/gels10040268

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Heidary M, Khosravi AD, Khoshnood S, Nasiri MJ, Soleimani S, Goudarzi M (2018) Daptomycin. J Antimicrob Chemother 73:1–11. https://doi.org/10.1093/jac/dkx349

    Article  CAS  PubMed  Google Scholar 

  132. Karas JA, Carter GP, Howden BP, Turner AM, Paulin OKA, Swarbrick JD, Baker MA, Li J, Velkov T (2020) Structure-activity relationships of daptomycin lipopeptides. J Med Chem 63:13266–13290. https://doi.org/10.1021/acs.jmedchem.0c00780

    Article  CAS  PubMed  Google Scholar 

  133. Baldwin H (2020) Oral antibiotic treatment options for acne vulgaris. J Clin Aesthet Dermatol 13:26–32

    PubMed  PubMed Central  Google Scholar 

  134. Francis NA, Ridd MJ, Thomas-Jones E, Shepherd V, Butler CC, Hood K, Huang C, Addison K, Longo M, Marwick C, Wootton M, Howe R, Roberts A, Haq MIU, Madhok V, Sullivan F (2016) A randomised placebo-controlled trial of oral and topical antibiotics for children with clinically infected eczema in the community: the ChildRen with eczema, antibiotic management (CREAM) study. Health Technol Assess (Rockv) 20:1–84. https://doi.org/10.3310/hta20190

    Article  Google Scholar 

  135. Orylska-Ratynska M, Placek W, Owczarczyk-Saczonek A (2022) Tetracyclines—an important therapeutic tool for dermatologists. Int J Environ Res Public Health 19:1–13. https://doi.org/10.3390/ijerph19127246

    Article  CAS  Google Scholar 

  136. Hardie KR, Fenn SJ (2022) JMM profile: rifampicin: a broad-spectrum antibiotic. J Med Microbiol 71:1–5. https://doi.org/10.1099/jmm.0.001566

    Article  CAS  Google Scholar 

  137. Wallenwein CM, Ashtikar M, Hofhaus G, Haferland I, Thurn M, König A, Pinter A, Dressman J, Wacker MG (2023) How wound environments trigger the release from Rifampicin-loaded liposomes. Int J Pharm 633:1–11. https://doi.org/10.1016/j.ijpharm.2023.122606

    Article  CAS  Google Scholar 

  138. Rinaldi F, Hanieh PN, Sennato S, De Santis F, Forte J, Fraziano M, Casciardi S, Marianecci C, Bordi F, Carafa M (2021) Rifampicin-liposomes for Mycobacterium abscessus infection treatment: intracellular uptake and antibacterial activity evaluation. Pharmaceutics. https://doi.org/10.3390/pharmaceutics

    Article  PubMed  PubMed Central  Google Scholar 

  139. Mann S, Frasca K, Scherrer S, Henao-Martínez AF, Newman S, Ramanan P, Suarez JA (2021) A review of leishmaniasis: current knowledge and future directions. Curr. Trop. Med. Rep. 8:121–132. https://doi.org/10.1007/s40475-021-00232-7/Published

    Article  PubMed  PubMed Central  Google Scholar 

  140. Jaafari MR, Hatamipour M, Alavizadeh SH, Abbasi A, Saberi Z, Rafati S, Taslimi Y, Mohammadi AM, Khamesipour A (2019) Development of a topical liposomal formulation of Amphotericin B for the treatment of cutaneous leishmaniasis. Int J Parasitol Drugs Drug Resist 11:156–165. https://doi.org/10.1016/j.ijpddr.2019.09.004

    Article  PubMed  PubMed Central  Google Scholar 

  141. Schulz A, Jiang L, de Vor L, Ehrström M, Wermeling F, Eidsmo L, Melican K (2019) Neutrophil recruitment to noninvasive MRSA at the stratum corneum of human skin mediates transient colonization. Cell Rep 29:1074–1081. https://doi.org/10.1016/j.celrep.2019.09.055

    Article  CAS  PubMed  Google Scholar 

  142. Towell AM, Feuillie C, Vitry P, Da Costa TM, Mathelié-Guinlet M, Kezic S, Fleury OM, Mcaleer MA, Dufrêne YF, Irvine AD, Geoghegan JA (2021) Staphylococcus aureus binds to the N-terminal region of corneodesmosin to adhere to the stratum corneum in atopic dermatitis. PNAS 118:1–9. https://doi.org/10.1073/pnas.2014444118/-/DCSupplemental

    Article  Google Scholar 

  143. Heidary M, Ebrahimi Samangani A, Kargari A, Kiani Nejad A, Yashmi I, Motahar M, Taki E, Khoshnood S (2022) Mechanism of action, resistance, synergism, and clinical implications of azithromycin. J Clin Lab Anal 36:1–16. https://doi.org/10.1002/jcla.24427

    Article  CAS  Google Scholar 

  144. Abruzzo A, Parolin C, Rossi M, Vitali B, Cappadone C, Bigucci F (2022) Development and characterization of azithromycin-loaded microemulsions: a promising tool for the treatment of bacterial skin infections. Antibiotics 11:1–19. https://doi.org/10.3390/antibiotics11081040

    Article  CAS  Google Scholar 

  145. Zaky MA, Zaky AG, Elsawy MF, Shehata KF, Elaziz MSA (2023) Efficacy of topical azithromycin versus systemic doxycycline in treatment of meibomian gland dysfunction. J Ophthalmol 2023:1–7. https://doi.org/10.1155/2023/4182787

    Article  CAS  Google Scholar 

  146. Hossain MJ, Jabin N, Ahmmed F, Sultana A, Abdur Rahman SM, Islam MR (2023) Irrational use of antibiotics and factors associated with antibiotic resistance: findings from a cross-sectional study in Bangladesh. Health Sci Rep 6:1–11. https://doi.org/10.1002/hsr2.1465

    Article  Google Scholar 

  147. Palmer ME, Andrews LJ, Abbey TC, Dahlquist AE, Wenzler E (2022) The importance of pharmacokinetics and pharmacodynamics in antimicrobial drug development and their influence on the success of agents developed to combat resistant gram negative pathogens: a review. Front Pharmacol 13:1–10. https://doi.org/10.3389/fphar.2022.888079

    Article  CAS  Google Scholar 

  148. MacDermott-Opeskin HI, Gupta V, O’Mara ML (2022) Lipid-mediated antimicrobial resistance: A phantom menace or a new hope? Biophys Rev 14:145–162. https://doi.org/10.1007/s12551-021-00912-8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Blackburn J, Ousey K, Patton D, Moore Z, Avsar P (2023) What is the evidence that there is antimicrobial resistance associated with the use of topical antimicrobial preparations? Wound Pract Res 31:40–48. https://doi.org/10.33235/wpr.31.1.40-48

    Article  Google Scholar 

  150. Lin YK, Yang SC, Hsu CY, Sung JT, Fang JY (2021) The antibiofilm nanosystems for improved infection inhibition of microbes in skin. Molecules 26:1–29. https://doi.org/10.3390/molecules26216392

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Author information

Authors and Affiliations

Authors

Contributions

Leonny Dwi Rizkita contributed to conceptualization. Leonny Dwi Rizkita and Pratiwi Wikaningtyas were involved in data curation. Leonny Dwi Rizkita, Rachma Greta Perdana Putri, and Muflihah Rizkawati contributed to investigation. Muhammad Farid was involved in project administration. Pratiwi Wikaningtyas contributed to supervision. Leonny Dwi Rizkita, Rachma Greta Perdana Putri, Muflihah Rizkawati, and Pratiwi Wikaningtyas were involved in validation. Leonny Dwi Rizkita and Rachma Greta Perdana Putri contributed to visualization. Leonny Dwi Rizkita, Rachma Greta Perdana Putri, Muflihah Rizkawati, and Muhammad Farid were involved in writing—original draft. Leonny Dwi Rizkita and Pratiwi Wikaningtyas contributed to writing—review and editing.

Corresponding author

Correspondence to Leonny Dwi Rizkita.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Availability of data and material

Not applicable.

Competing interests

The authors declare no competing interest either in financial or interest during the overall making of the paper.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rizkita, L.D., Putri, R.G.P., Farid, M. et al. Liposome drug delivery in combating the widespread topical antibiotic resistance: a narrative review. Beni-Suef Univ J Basic Appl Sci 13, 90 (2024). https://doi.org/10.1186/s43088-024-00545-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43088-024-00545-2

Keywords