Next Article in Journal
Bovine Lactoferrin Suppresses Tumor Angiogenesis through NF-κB Pathway Inhibition by Binding to TRAF6
Next Article in Special Issue
Formulation Development of Fast Dissolving Microneedles Loaded with Cubosomes of Febuxostat: In Vitro and In Vivo Evaluation
Previous Article in Journal
mRNA in the Context of Protein Replacement Therapy
Previous Article in Special Issue
Effect of Ciprofloxacin-Loaded Niosomes on Escherichia coli and Staphylococcus aureus Biofilm Formation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nanoemulgel: A Novel Nano Carrier as a Tool for Topical Drug Delivery

by
Mahipal Reddy Donthi
1,†,
Siva Ram Munnangi
1,2,†,‡,
Kowthavarapu Venkata Krishna
1,3,§,
Ranendra Narayan Saha
1,
Gautam Singhvi
1 and
Sunil Kumar Dubey
1,4,*,‖
1
Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Pilani 333031, India
2
Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA
3
Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA
4
R&D Healthcare Division Emami Ltd., 13, BT Road, Kolkata 700056, India
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Current Affiliation: Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA.
§
Current Affiliation: Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA.
Current Affiliation: R&D Healthcare Division Emami Ltd., 13, BT Road, Kolkata 700056, India.
Pharmaceutics 2023, 15(1), 164; https://doi.org/10.3390/pharmaceutics15010164
Submission received: 20 November 2022 / Revised: 24 December 2022 / Accepted: 27 December 2022 / Published: 3 January 2023
(This article belongs to the Special Issue Local Drug Delivery System)

Abstract

:
Nano-emulgel is an emerging drug delivery system intended to enhance the therapeutic profile of lipophilic drugs. Lipophilic formulations have a variety of limitations, which includes poor solubility, unpredictable absorption, and low oral bioavailability. Nano-emulgel, an amalgamated preparation of different systems aims to deal with these limitations. The novel system prepared by the incorporation of nano-emulsion into gel improves stability and enables drug delivery for both immediate and controlled release. The focus on nano-emulgel has also increased due to its ability to achieve targeted delivery, ease of application, absence of gastrointestinal degradation or the first pass metabolism, and safety profile. This review focuses on the formulation components of nano-emulgel for topical drug delivery, pharmacokinetics and safety profiles.

1. Introduction

The recent progress in drug synthesis and high throughput screening have steered drug discovery and development toward lipophilic drug moieties. Currently, 90% of drugs in the discovery pipeline and more than 40% of the drugs present in the market are of lipophilic nature [1]. The lipophilic nature of the drugs leads to problems like poor solubility, unpredictable absorption, and inter and intra-subject variability concerning pharmacokinetics. Various techniques have been employed to increase the solubility of active moieties. These techniques include physical and chemical modification of API along with formulation strategies, which include particle size reduction, complexation, amorphization, and nano-carrier drug delivery systems as represented in Figure 1 [2,3,4].
Despite of employing various technologies for enhancing the solubility, delivering the drugs via the oral route is not always feasible owing to their low bioavailability associated with poor absorption, first-pass metabolism, chemical and enzymatic degradation [5,6]. In addition, clinical complications and low concentrations of the drug at the site of action hinder drug delivery through the oral route. For example, the oral administration of Disease-modifying anti-rheumatic drugs (DMARDs) used in the treatment of arthritis are associated with various side effects like carcinogenicity, hepatotoxicity, and hematologic toxicity [7,8]. These clinical complications can be mitigated by delivering the drug through the topical route [9].
In topical delivery, skin being a fundamental defense layer, considers the API’s as external components and restricts their entry into the body. The outer most layer of epidermis called stratum corneum is the first and firm layer to overcome for drug penetration into the skin [10]. Various mechanisms have been explored to enhance the drug permeation. One such mechanism involves disruption of skin layer structure, which can be achieved using techniques such as chemical penetration enhancers, ultrasound, iontophoresis, sonophoresis, electroporation and microneedles [11]. In contrary, the use of nanocarriers was observed to be an effective strategy for circumventing the SC barrier without exacerbating skin damage and achieving efficient drug penetration. They facilitate the drug delivery through the skin utilizing intra and inter cellular transport mechanisms, interacting with skin components to mediate transport or to create depots of the drug for sustained or stimuli-induced release. These novel carrier for topical administration includes but not limited to emulsions (nano/micro), micelles, dendrimers, liposomes, solid lipid nanoparticles and nano-structured lipid carriers [12,13,14]. Among these, nano-emulsions are found to be a potential drug delivery system because of their high drug-loading capacities, solubilizing capacities, ease of manufacturability, stability and controlled release patterns. These nano-emulsions owing to their lipophilic core allow the movement of more lipophilic molecules across the topical membranes compared to the liposomes [15]. In addition, liposomes stability has always been an issue, as they disintegrate during the penetration process. Likewise, the low drug loading capacity and uncontrolled release hinders the application of solid-lipid nanoparticles in dermal drug delivery. Similarly, micelles exhibit poor stability and encapsulation efficiency. In the same way, the toxicity and poor controlled release behavior of dendrimers limits its topical application [16].
Nano-emulsions are heterogeneous colloidal mixtures of oil and water, with one component as a dispersed phase and the other as a continuous phase. A surfactant known as an emulsifier is adsorbed at the interface between the dispersed and continuous phases, lowering the surface tension and thus stabilizing the system. These systems possess high thermodynamic stability leading to longer shelf life compared to simple emulsions, micelles or suspensions, etc. Despite having various advantages, nano-emulsions are limited by their low viscosity leading to low retention time and spreadability [17]. These problems can be resolved by modifying the nano-emulsion into a nano-emulgel by using a suitable gelling agent [18].
The nano-emulgel acts as a colloidal system consisting of a mixture of emulsion and gel. The emulsion part protects the drug from enzymatic degradation, and hydrolysis and improves the permeation like other nano-carriers. Besides enhancing the penetration of the drug through the skin, it is equally important to retain the therapeutic concentrations of the drug for a sufficient period of time. The gel part improves the viscosity and spreadability resulting in improved retention time, and also reduces the surface and interfacial tension, thus improving the thermodynamic stability. Nano-emulgel possesses various advantages having high drug loading capacity, better penetration, diffusion, and low skin irritation compared to other nano-carriers [19,20].
This article aims to provide insight into the selection of formulation ingredients of a nano-emulgel, characteristics and formulation aspects, advantages, pharmacokinetics and pharmacodynamics, and safety of the same. The objective here is to give an overview of the future and rationale behind the nano-emulgel drug delivery system.

2. Drug Delivery through a Topical Route

The characteristics of any ideal formulation are patient compliance, self-administration, non-invasiveness, fewer side effects, and better pharmacological action. The topical route administering formulations possess most of the aforementioned characteristics [21]. The benefits of the topical route of administration comprise of avoiding the hepatic first-pass effect, decreased side effects due to the local site of action, enhancement in percutaneous absorption and topical usage may even increase bioavailability with a sustained deposition [22]. Further, the reduced drug loss due to metabolism or decomposition, and the ability to specifically target the drug at the desired site are also some of the advantages. Minimization of drug breakdown coupled with constant delivery of drug for a prolonged period results in prominent movement of the drug across the barrier of stratum corneum, leading to improved bioavailability [23,24].
An increase in the bioavailability of drugs via the topical route has been proved in various research works. For example, Flurbiprofen nano-emulsion showed 4.4 times increment in bioavailability upon topical administration compared to oral delivery [25]. Zhou et al. prepared the nano-emulsion of nile red dye, which displayed 10-fold increase in the penetration of dye across the skin compared to an emulsion formulation [26]. Gannu et al. reported a 3.5-fold increase in Lacidipine bioavailability via the transdermal route of administration using microemulsions. The group reported that this improvement could be due to the avoidance of the first-pass effect on the drug upon topical application [24]. Further, an enhancement in the therapeutic and pharmacological effect of therapeutically active agents has been demonstrated with topical formulations.
Conventional topical formulations that are used are solutions, ointments, lotions, creams, patches, gels, etc. [27]. But these topical formulations have to traverse the remarkably effective and competent stratum corneum barrier along with viable epidermis of the skin as shown in Figure 2. The SC is a 10–20 µm thick lipid-interspersed matrix of terminally formed keratinocytes, which causes a huge challenge for the delivery of therapeutically active agents via the topical or transdermal route of administration [28,29,30]. Thus, reducing the amount of drug reaching the target site. This is reflected in the marketed topical preparations possessing low permeation leading to poor therapeutic effect [31,32,33]. Therefore, research in this area mainly focuses on the development of topical formulations with appropriate permeability and ensuring delivery by numerous mechanisms. The direction of research work in recent years has shifted towards novel carrier systems with the intent to alter the permeability of hydrophobic drugs through the skin. New formulation development techniques and strategies are emerging in recent years but the main drawback of the recent strategies is the usage of chemicals and non-green solvents for enhancing the permeation. The usage of these preparations for a long period would lead to various skin complications [34,35]. Besides, various limitations posed by the skin, there are certain characteristics that an active moiety should possess in order to be suitable for the topical route of administration as represented in Table 1 [36,37].

3. Nano Emulsions in Topical Delivery

An upgrade and innovation of topical and transdermal drug delivery systems led to the development of lipid-based nano-formulations. Though there are various formulations, research has deepened pertaining to nano-emulsions due to the aforementioned advantages and their ability to deliver hydrophobic drugs non-invasively and without the need for a penetration enhancer [38]. Nano-emulsions are an isotropic biphasic mixture consisting of two portions: water and oil, where one phase is dispersed in the other as nanosized droplets. The system is stabilized by the utilization of an interfacial layer of surfactants [39]. The difference between nano-emulsions and traditional emulsions is that the former has decreased propensity to undergo phase separation [40,41]. A prominent number of in-vivo studies have been carried out demonstrating the applications and feasibility of topical micro and nano-emulsions. In-vitro works have also supported the use of these topical lipidic formulations [42,43]. These nano-emulsion systems possess a translucent or transparent appearance. The thermodynamic stability of nano-emulsions is greater than other lipid carriers. Nano-emulsions exhibit an increased solubilization capacity as compared to solutions of simple micelles [29,44]. These formulations can solubilize and incorporate large amounts of active drug substances due to the increased surface area because of the nano-size of oil droplets [45]. The phenomena of creaming or sedimentation are the general issues faced in an emulsion. The improved stability in a nano-emulsion is due to Brownian motion and less gravitational force acting on the particles because of their nano-size, thus preventing the stability issues like sedimentation and creaming [46]. Numerous studies have demonstrated the enhanced permeation of drugs upon administered as nano-emulsion systems in comparison to other formulations like emulsions, creams, and ointment gels [47,48,49]. The enhanced permeation is because of the ability of nano-emulsion to overcome the firmly bonded lipid bi-layers, thus able to penetrate deep into the skin and deliver the drug to systemic circulation because of smaller sized dispersed droplets, which facilitate transcellular in addition to paracellular transport [18].

4. Nano-Emulgel Drug Delivery System

Despite possessing many advantages, nano-emulsions lack spreadability because of their low viscosity resulting in poor retention of formulation over the skin [50]. This limitation hampers the clinical applications of nano-emulsions [51]. This issue has been resolved by incorporating a gelling agent into the nano-emulsion, thus forming a nano-emulgel [52]. Huge quantities of aqueous or hydroalcoholic bases are employed in a colloidal particulate system to prepare gels [53]. Nano-emulgel is formed by incorporating the nano-emulsion into a hydrogel matrix, which reduces the thermodynamic instability of the emulsion. The improved thermodynamic stability is due to the reduction in the portability of the non-aqueous phase because of the increased consistency of the external medium. The increased retention time and thermodynamic stability enable the formulation to release the drug over a period, making nano-emulgel a controlled release dosage form for topical administration benefiting the drugs with a short half-life [19,54].
The incorporation of nano-emulsion into a gelling system helps to annihilate the disadvantages of both individual systems. The combined nano-emulgel enjoys the properties of a gel with the refined characteristics of a nano-emulsion. The Table 2 discloses the advantages of nano-emulgel over conventional emulgel owing to its particle size and thermodynamic stability. The variety of benefits offered by nano-emulgels is enhanced skin permeation, greater loading of an active moiety, less irritation, and greater spreadability. This is apparent in comparison with different nano-carriers such as solid lipid nanoparticles and liposomes. The nano-emulsion is made suitable for topical use due to the increased viscosity of the gel. To achieve the same, various gelling agents compatible with skin like xanthan gum, carbomer 980, Pluronic’s, carrageenan, and carbomer 934 are used for topical application [55]. Acceptable localization and drug dispersion through adequate percutaneous absorption across the skin is achieved in nano-emulsions. This helps to increase the efficacy locally and also systematically via the skin. This system can also be used to deliver drugs to the central nervous system (CNS) due to its ability to cross the blood-brain barrier when applied through the nasal route [56,57]. Non-irritant and non-greasy nature of nano-emulgel facilitates better patient compliance [53] In addition, pharmacokinetic properties like enhanced bioavailability and decreased side effects are added advantages for these systems [58]. The hydrogel matrix, consistency, and homogeneity have added to the growing focus on nano-emulgels. Furthermore, various studies have shown that nanoemulgel has increased stability due to less Oswalt ripening caused by decreased mobility of oil globules in gel matrix [59]. For instance, Kaur et al. developed a topical nanoemulgel loaded with TPGS containing mefenamic acid. In the pharmacodynamic investigation, the optimized nanoemulgel inhibited inflammation and enhanced percent reaction time with improved analgesic efficacy. The formulated nanoemulgel outperformed other traditional topical formulations in terms of long-term stability and drug penetration [60].
Besides these, nano-emulgel is devoid of other formulation stability limitations like the problem of destabilization faced with conventional emulgels, the problem of moisture entrapment faced with powders, the problem of cake formation faced with suspensions, the problem of coalescence of oil globules, formation of agglomerates in case of suspensions, along with the problem of poor adherence and excessive spreadability that is faced with nano-emulsions [53]. Due to these factors, nano-emulgel is often thought of as an improved and different topical drug delivery approach over the standard marketed dosage forms. This novel formulation is welcome for research targeting various skin diseases and disorders. Nano-emulgel will soon be capturing the market in the topical delivery segment as a favorable substitute over conventional forms and some are currently being marketed as in Table 3. Many preclinical (Table 4) and clinical studies (Table 5) are being conducted to evaluate the efficacy of nanoemulgel.

5. Formulation Components

Nano-emulgels are made up of two individual systems; the gelling agent and the nano-emulsion i.e., emulsion consisting of nano droplets which are of o/w or w/o type. Both emulsion types possess an aqueous and an oily phase. The gel base consists of polymers that can swell on the absorption of a liquid. The various components in the nano-emulgel formulation are provided in Table 6 [53,84]. The overview of the selection criteria of the essential components in a nano-emulgel have been discussed below.

5.1. Oil Phase

The selection of oil and its quantity depends on the application and utility of the nano-emulgel. The permeability, stability, and viscosity of the prepared nano-emulsion depends on the type and quantity of chosen lipid component, i.e., oil phase. Primarily in case of pharmaceutical and cosmetic applications, the oil phase is made up of either naturally or synthetically originated lipids, unless the oil phase itself is an active ingredient. The consistency of the lipids may vary from liquid to high molecular solids. The hydrophobicity of an oil plays a crucial role in forming a stable emulsion, wherein poor hydrophobicity of the oil is shown to increase the emulsification, concurrently affecting the solubility of lipophilic moieties [99]. Thus, choosing an oil is an essential prerequisite for nano-emulgel development as a novel drug delivery system [100].
Natural oils exhibit an additional medicinal significance leading to an increase in the researcher’s interest to use these additive properties supporting the pharmacological action of the active moiety. For example, oleic acid is frequently used oil in nano-emulgel formulations and is obtained from vegetable and animal sources. It is a biodegradable and biocompatible omega-nine fatty acid and has elevated solubilization characteristics along with improving percutaneous absorption [101]. Antioxidants present in oleic acid contribute to cellular membrane integrity. It also repairs cell damage and showcases formulation stabilization [55,102]. Arora et al. confirmed that an increase in oleic acid content in the preparation increases the rate of permeation. In their study, using 6% oleic acid instead of 3% in the preparation nanoemulgel of drastically improved the permeability of ketoprofen [55].
Another natural oil called Emu oil is being appreciated for its analgesic, antipruritic, and antioxidant characteristics. Jeengar and group prepared nano-emulgel of curcumin with emu oil to treat the disease of joint synovium, the formulation demonstrated enhanced permeability and better pharmacological activity compared to pure curcumin [85,103]. The use of emu oil has been encouraged in the cosmetic field as well [85]. It moisturizes the skin and has high amounts of unsaturated fatty acids like oleic acid, thus improving the penetration of the drug [104].
The therapeutically active agent may also be used as the oil component in nano-emulgel preparation. Active moieties from Swietenia macrophylla have anti-inflammatory action and are self-employed as an oily phase in nano-emulgel. The therapeutic effect was found to be better in this nano-carrier preparation as opposed to the parent form [44]. Further, the edible oils considered to be the preferred lipid excipient of choice for the development of emulsions, are not frequently chosen due to their poor ability to dissolve large amounts of lipophilic drugs. Therefore, these oils are chemical modification or hydrolyzed to form an appropriate oil, which upon combining with a suitable surfactant enhances the solubility of hydrophobic compounds for nano-emulgel formulation [104].

5.2. Surfactant System

Surfactants are an essential ingredient in nano-emulsion, which are utilized in the stabilization of the unstable mix of two immiscible phases. This is achieved by a decreasing the interfacial tension amongst the two phases and alteration of dispersion entropy. The surfactant should show quick adsorption along the interface of the liquids. The final result is a decrease of interfacial tension and inhibition of coalescence of the individual nano-sized droplets [105].
The HLB value of the surfactant is an important variable for selecting the proper surfactant. The surfactants are either w/o type (HLB of 3–8) or o/w type (HLB of 8–16). In w/o emulsions, low HLB value surfactants i.e., less than 8 are utilized. Alternately Spans and Tweens are used for o/w emulsion as their HLB value is more than 8. A mixture of Span and Tween provides better stability to an emulsion system compared to pure Span or Tween containing preparations. Thus, using a proper mixture of surface-active agents is essential to formulate an ideal nano-emulsion. Based on the charge, the surfactants are of four main categories i.e., cationic, non-ionic, anionic, and zwitterionic nature. Examples of cationic surfactants are hexadecyl trimethyl ammonium bromide, cetyl trimethyl ammonium bromide, quaternary ammonium compounds, and dodecyl dimethyl ammonium bromide [106,107]. Poloxamer 124 and 188, Tween 20 and Caproyl 90 are some of the non-ionic surfactants [108,109]. Anionic surfactants are sodium dodecyl sulphate and sodium bis-2-ethylhexylsulfosuccinate [110]. Phospholipids such as phosphatidylcholine are part of zwitterion surfactants [111]. Toxicity should be considered while selecting the surfactant as it may lead to irritation of the gastrointestinal tract or skin based on the route of administration. Ionic surfactants are usually not preferred due to their toxicity and non-biocompatibility. The safety, biocompatibility and being unaffected by pH or ionic strength alteration make non-ionic surfactants an appropriate choice [112].
The surfactants derived from natural sources such as bacteria, fungi, and animals are being considered as a potential option, due to their safety, biodegradability, and biocompatibility. Bio-surfactants show a similar mechanism in decreasing surface tension along the interface due to amphiphilic properties. This is mainly due to the presence of non-polar short fatty acids and polar functionalities as the tail and head respectively [113]. They are more bio-compatible and safer than synthetic surfactants.

5.3. Co-Surfactant System

Co-surfactants support surfactants during the emulsification of oil in the water phase. Co-surfactants are required for decreasing the interfacial tension and improving the emulsification [114]. Flexibility is added to the interfacial film along with attaining transient negative interfacial tension due to co-surfactants. The association between the surfactant and co-surfactant along with the partitioning of the drug in immiscible phases decides the drug release from the nano-emulgel. Hence co-surfactant selection is equally important as surfactant. The commonly used co-surfactants are PEG- 400, transcutol® HP, absolute ethyl alcohol, and carbitol [115]. Alcohol based co-surfactants are most preferred because of their ability to partition between the oil and water phase thereby improving their miscibility.
The concentration of co-surfactant being used has to be chosen cautiously, since it may affect the emulsification by surfactant. Also, a combination of surfactant and co-surfactant with closer HLB values does not produce a stable emulsion as produced by non-ionic surfactants with different HLB values. The reason may be due to the solubilization of higher HLB value surfactants in the aqueous phase. Whereas, lower HLB value surfactants solubilize in the non-aqueous phase, enabling more intense association with the mixture of surfactant and co-surfactant [116]. Therefore, the choice of various formulation components and the rationale behind them is a very demanding and stimulating exercise.

5.4. Gelling Agents

Gelling agents upon addition to the appropriate media as a colloidal mixture forms a weakly cohesive three-dimensional structural network with a high degree of cross-linking either physically or chemically providing consistency to nano-emulgel [117,118,119]. In topical applications, these agents are used to stabilize the formulation, to attain optimum delivery of the drug across the skin. They play an important role in determining various parameters of the formulation like consistency, rheological properties, bio-adhesive properties, pharmacokinetics, spreadability, and extrudability. Based on the origin, these gelling agents are divided into natural, synthetic, and semi-synthetic. The Table 7 gives information on the concentration and pharmaceutical adaptability of various gelling agents used to prepare nano-emulgel. Natural gelling agents are bio-polysaccharides or their derivatives and proteins. The pectin, carrageenan, alginic acid, locust bean gum, and gelatine, etc., are bio-polysaccharides, while xanthan gum, starch, dextran, and acacia gum, etc., are derivatives of bio-polysaccharides. Though they provide excellent biocompatibility and biodegradability, the major limitation of natural gelling agents is microbial degradation [119,120]. Like natural gelling agents, semisynthetic gelling agents also offer good biocompatibility and biodegradability [121]. These agents are usually the derivatives of cellulose like hydroxypropyl cellulose, ethyl cellulose, sodium alginate, etc. The semisynthetic agents are comparatively more stable than natural gelling agents and are more responsive to chemical, biological and environmental changes like pH and temperature [122]. Synthetic gelling agents are prepared by chemical synthesis, some of them are FDA-approved e.g., carbomers and poloxamers [123,124]. Carbomers are polymerized acrylic acids, while poloxamers are triblock non-iconic copolymers comprising two hydrophilic units of polyoxyethylene attached to a central hydrophobic chain of polypropylene [124,125]. The FDA-approved synthetic agents are non-toxic and offer a wide range of rheological properties based on the molecular weight of the polymer, thus suitable for a wide range of applications.

6. Preparation of Nano-Emulgel

Nano-emulgel is a non-equilibrium formulation of structured liquids requiring energy, surfactant, or both for its preparation. They are spontaneously formulated by mixing the components. This is undertaken by introducing energy in the biphasic system or decreasing the interfacial tension between the interfaces of the two immiscible phases [135].
There are various nano-emulgel preparation methods reported based on the order of mixing of oil and aqueous phase [136]. Lupi et al. (2014) as illustrated in Figure 3A solubilized the drug in the oil phase and gelling agent in the water phase separately. The oil phase is added to the aqueous gel phase under stirring followed by homogenization to form an emulsion. The sol form of gelling agent in the emulsion is converted to gel by various mechanisms like adding a complexing agent or adjusting to the required pH [137]. Dong et al. (2015) as illustrated in Figure 3B divided the total quantity of water required for the preparation into two parts. One part of the divided quantity is used to prepare pre-emulsion and the other part is used for the preparation of gel. Later, these two components are mixed together under stirring [138]. Jeengar et al. (2016) prepared the emulsion and gel separately, followed by mixing them together in a 1:1 w/w ratio [85].
Nano-emulgel formulation preparation can be further divided into two types based on the implementation of high-energy and low-energy emulsification techniques. High energy method involves the use of mechanical devices to produce a highly disruptive force in which both phases undergo size reduction. Hence this method may lead to the heating up of components in the formulation causing thermodynamic instability of the formulation and making it not suitable for thermo-labile drugs. Microfluidizers, high-pressure homogenizers, and ultrasonicated are high-energy methods employed to obtain a nanosized emulsion. This method is used for preparing nano-formulation of sizes of about 1 nm.
Phase inversion, self-emulsification, temperature, and phase transition are techniques of low energy approach. These methods provide the required thermodynamic stability to the nano-emulsion. The spontaneous method involves mixing oil, surfactant, and water in the best ratio possible and is most applicable for thermolabile compounds. The emulsification process is based on the surfactant and co-surfactant characteristics and their order of addition. Temperature-based alterations in HLB are utilized for non-ionic surfactants like Tween 20 Tween 60, Tween 80, Labrasol [139]. This method is mostly utilized for phase transition during phase inversion. Application of cooling with constant stirring will lead to a reversal of emulsion prepared at inversion temperature. Reduction in phase inversion temperature facilitates the inclusion of thermolabile components using this technique [140]. The second step incorporates gelling agent to change the liquid state to gel in the nano-emulsion. The thixotropic nature of the gelling agent facilitates the gel-solution conversion when shear stress is applied to the preparation keeping the volume constant. This leads to thickening in o/w nano-emulsion because of the creation of a gelled structure.

7. Permeability of Nano-Emulgel

In the preparation of emulsion-based gels, it is necessary to examine the important process parameters that have a significant effect on the size and formulation stability. In order to accomplish this, we must select the proper preparation process at the early stages. Emulsions are developed using various techniques, such as mechanical (or rotor-stator), high-pressure, microfluidization, and ultrasonic methods. The mechanical system comprises a colloid mill, that has a complex geometry, and the droplets of an emulsion generated by this system are several microns in size, making it the least desirable approach for manufacturing nanoemulsions [141]. Achieving an optimum droplet size is highly challenging. However, a droplet size of less than a micron can be achieved using high-pressure homogenization and sonication techniques, which in turn helps extend the shelf life of emulsions by lowering the creaming rate. For this reason, homogenization and sonication are considered to be efficient methods for the development of nanoemulsion [142,143]. In addition, increasing the homogenization speed or duration by itself is not enough to decrease the size of the globules, however, the use of the optimum concentration of an emulsifier is necessary to maintain control over the re-coalescence of the emulsion. For instance, Sabna Kotta et al. made a nanoemulsion utilizing the phase inversion and homogenization methods. In this formulation, gelucire 44/14 was used as a surfactant and transcutol-HP as a co-surfactant. They employed both the proposed techniques to produce nano-sized emulsion globules. In the case of homogenization, the large globule size was observed, despite increased pressure, and increased cycles at lower concentrations of an emulsifier. This demonstrates that the globule size of the formulation could not be decreased by homogenization alone. When the optimum concentration of an emulsifier is combined with increasing homogenization pressure and cycles, the size of the globules decreases. Because homogenization alone can break down globule size to nano, but with a lower concentration of surfactant, the newly formed globule surface would be improperly covered with a surfactant, resulting in re-coalescence. With the optimum concentration of an emulsifier and increased homogenization pressure and cycles, a smaller globule size with a good polydispersity index could be achieved. As a result, the author came to the conclusion that, throughout the preparation process, the desired particle size was obtained with a lower PDI by the combination of the surfactant, homogenization pressure, and cycle duration [142].
Mohammed S. et al. used ultrasonication to develop a thymoquinone-loaded topical nanoemulgel for wound healing. They used black seed oil (oil vehicle), Kolliphor El (surfactant), and Transcutol HP (co-surfactant). Nanoemulgel was prepared using different time intervals (3, 5, and 10 min) of ultrasonication at a 40% amplitude. When the concentration of surfactant decreased with 10 min of ultrasonication, the globular size increased. Meanwhile, increasing the concentration of surfactant with 10 min of sonication time resulted in a smaller globular size. The authors concluded that sonication is more effective when the appropriate concentration is used [70]. Monitoring the process control parameters and taking into account the composition of the excipients is both necessary steps in the process of optimizing the formulation.

8. Permeability of Nano-Emulgel

Skin shows an inherent property of acting as a protective barrier against external agents. Therefore, penetration through the skin is a major complication associated with topical delivery systems. The outermost layer of skin is the stratum corneum, which is followed by stratum granulosum and stratum lucidum. The stratum corneum is loosely composed of keratinized cells, waxy lipids, fatty acids, and cholesterol. All these constituents of stratum corneum help in retaining moisture and provide a hydrophobic barrier over the skin [18]. After the stratum corneum, there is the epidermis which is followed by dermis and subcutaneous layer. After crossing the subcutaneous layer, the active moiety will finally reach the systemic circulation. The primary hurdle for the drug moiety after reaching out from gel matrix is crossing the stratum corneum, from here the nano sized droplet due to the virtue of small diameter traverses basically through two different pathways as shown in Figure 4. One is cell to cell transfer involving concentration gradient-based movement called transcellular transport or intracellar transport, while the other is a passage through intercellular spaces or paracellular transport [118]. Whereas there is a third pathways called transappendageal transport, its influence on drug penetration is limited because hair follicles and glandular ducts make up negligible portion of the total surface area of the skin [16].
Generally, ex-vivo permeation studies involve the examination of nano-emulgel formulation on isolated tissue in a simulated biological medium. Ex-vivo studies give a comparative analysis of penetration with different types of topical dosage forms and an idea about the flux rate of the drug inside the skin. Jeengar et al. made a nano-emulsion with emu oil as the oil phase. Optimized nanoemulsion was amalgamated with Carbopol gel to form nano emulgel and used for topical delivery of curcumin as an anti-inflammatory agent in rheumatoid arthritis. Ex-vivo permeation studies showed that permeation through the skin was higher for nano-emulgel as the retention of the formulation was higher compared to the nano-emulsion [145,146]. Elmateeshy and group formulated nano-emulgel by incorporating terbinafine HCl (TB) nano-emulsion formed from peceol as oil phase and (TWEEN 80/propanolol) as surfactant mixture and Carbopol as a gelling agent. The enhanced permeation of peceol oil-based nano-emulgel was observed in ex-vivo studies compared to available marketed products [97]. Similarly, Mulia et al. developed nano-emulgel for mangosteen extract composed of o/w nano-emulsion with virgin coconut oil as the oil phase and Tween 80/SPAN 80 as surfactant mixtures. The gel base was made with xanthan gum and phenoxyethanol was supplemented as a preservative. In vitro permeation studies demonstrated elevated penetration compared to nano-emulsion [29,147,148]. In addition, Bhattacharya et al., formulated celecoxib nano-emulgel with carbopol-940 hydrogel base, while Tween 80 and Acconon MC8-2EP as surfactants. Both in-vitro drug release and ex-vivo studies showed positive results. After the twelfth hour of diffusion, the optimized formulation displayed 95.5% cumulative release of the drug, whereas the commercially available formulation showed only 56.90% release. A higher penetration coefficient is displayed by nano-emulgel compared to commercial formulation [149]. In the same way, Chin et al. also developed a nano-emulgel of telmisartan for intranasal delivery using different molecular weight chitosan polymers. The ex-vivo penetration studies showed an improved permeation profile. The group also demonstrated the improvement in permeation is attributed to the molecular weight of the polymer, where the medium molecular weight chitosan provides higher permeation [150].
A nano-emulgel preparation for delivery through the transdermal route of tacrolimus was formulated by Begur et al. using almond gum as gel and oleic acid as the lipophilic phase. Cremophor was used as a surfactant to improve penetration. Examinations on rat abdominal skin showed a substantial increase in penetration [34]. Similarly, butenafine an antifungal agent available as a cream in the market, Syamala et al. prepared a nano-emulgel formulation for the same drug and found considerable results. Ex-vivo penetration studies showed a substantial increase in permeation over marketed creams [97]. In the same way, an increment in permeation of ketoconazole by about 53% was observed by delivering the drug in nano-emulgel formulation compared to normal marketed cream. The quality of life of the patient could be improved by implementing these types of dosage forms [151]. These studies showcase the ability of nano-emulgel in enhancing the permeation of the active moiety compared to nano-emulsion and conventional topical dosage forms. The permeation of the nano-emulgel is affected by various factors like gelling agents, surfactants, and permeation enhancers, etc. The gelling agents improve the permeation by improving the adherence of formulation upon the skin. While the surfactant alone or in combination with a co-surfactant will improve the permeation by disrupting the lipid bilayer. All these components can improve the permeation of active moiety.

9. Characterization Studies of Nano-Emulgel

The pharmaceutical product must be evaluated to ensure quality and consistency between different batches. These tests help in understanding the product’s behavior and stability. According to USP, there are few universal tests for any given dosage form e.g., description, identification, assay, and impurities. A topical dosage form should undergo a few specific tests set by USP on a case-by-case basis: uniformity of dosage units, water content, microbial limits, antimicrobial and antioxidant content, pH, particle size, sterility, and API’s polymorphic nature. Apart from the tests required for topical dosage form, nanoemulgel consists of nanosized globules, which need to be evaluated for zeta potential, droplet size and polydispersity index (PDI). Along with these physiochemical tests, a dosage form needs to be evaluated for its in-vitro release, spreadability, bio-adhesive tests, skin-irritation, ex-vivo permeability and in-vivo bioavailability can be performed to understand the behavior of nanoemulgel. Methods and techniques for analyzing significant properties of a nanoemulgel are briefly described below:

9.1. Zeta Potential

The particles in a solution usually possess a layer of ions on their surface, referred to as the stern layer. Adjacent to the stern layer, there exists a diffuse layer of loosely bounded ions, which along with the stern layer collectively called an electrical double layer. There is a boundary between the ions in the diffuse layer that move with the particle and the ions that remain with the bulk dispersant. The zeta potential is the electrostatic potential at this “slipping plane” boundary [152]. Zeta potential measurement provides an indirect measure of the net charge and is a tool to compare batch-to-batch consistency. The higher the zeta potential, the greater the repulsion resulting in increased stability of the formulation. For example, the high zeta potential of emulsion globules prevents them from coalescing. A surface charge modifier may also be used to adjust the surface charge. For instance, if a negatively charged surface modifier is used, the zeta-potential value becomes negative, and vice-versa [153,154]. Surface active ingredients (such as anionic or cationic surfactants) thus play an important role in emulsion stability, and the zeta potential can be measured using various instruments such as the ZC-2000 (Zeecom-2000, Microtec Co. Ltd., Chiba, Japan), Malvern Nanosizer/Zetasizer® nano-ZS ZEN 3600 (Malvern Instruments, Westborough, MA, USA), and others.

9.2. Droplet Size Measurement and Polydispersity Index (PDI)

The size of globule in nanoemulgel is referred as its hydrodynamic diameter, which is a diameter of equivalent hard sphere that diffuses at the same rate as the active moiety [155]. The PDI determines the distribution of droplet size and is defined as the standard deviation of droplet size divided by mean droplet size. The droplet size and the polydispersity index are closely connected to the stability and drug release, as well as the ex-vivo and in-vivo performance of the dosage form. In addition, it is important to measure consistency between different batches. The globule size and PDI of the formulation can be measured using a zeta sizer or master sizer. The globule size of the emulsion can be determined using the principle of dynamic light scattering, in which the transitional diffusion coefficient is measured by monitoring the interaction between the laser beam and dispersion, as well as the Polydispersity index [156,157].

9.3. Rheological Characterizations

Rheology is the study of the deformation and flow of materials. The rheological characterization of materials reveals the influence of excipient concentrations like oils, surfactants, and gelling agents on the formulation’s viscoelastic flow behavior. If a formulation’s viscosity and flow characteristics vary, this may influence its stability, drug release, and other in-vivo parameters. In this instance, the formulation’s shear thinning tendency generates a thin layer on the skin surface, improving permeability, whereas a thicker formulation decreases permeation. Therefore, the rheological behavior is an extremely important factor in the formulation of nanoemulgel and several unique types of viscometers can be used to determine the rheological behavior [20]. FDA recommends the evaluation of complete flow curves whenever possible, plotted as both heat stress versus shear rate and viscosity versus shear rate across multiple shear rates until low or high plateaus are observed. If a formulation exhibits plastic flow, yield stress values should be evaluated.

9.4. Spreadability Testing

The spreadability property of the topical dosage form ensures the evenly spreading of the dosage form, thus delivering a stranded dose subsequently affecting the efficacy. The viscosity of the nanoemulgel greatly affects the spreadability property [158]. To date, no standard method has been established for measuring the spreadability of the dosage form. A few tests, that are commonly used for a good approximation of spreadability are a parallel-plate method and human subject assessment etc. The parallel-plate method (slip and drag method) is a widely employed technique because of its simplicity and relatively economic [158]. The instrumental setup consists of two glass slides of the same length, one of which is stationarily attached to the wooden block, and the other glass slide is mobile attached to a pulley at one end to measure spreadability. Spreadability is determined by the emulgel’s ‘Slip’ and ‘Drag’ qualities. The nanoemulgel dosage form will be placed on a stationary glass slide, which is then squeezed in between stationary and mobile glass slides. The formulation is squeezed firmly for uniformly spreading formulation between two slides and to remove any air bubbles. The known weights are added to the pulley until the upper slide slips off from the lower slide. The time required for slipping off is recorded, which is used to calculate spreadability using the following equation [159].
S = M L / T
where, S, M, L and T respectively represent the spreadability, weight bounded to the upper slide, Length of the slide, and Time taken to detach the slides.

9.5. In-Vitro Release Test (IVRT)

The efficacy and safety of the API are associated with drug release from the dosage form. The IVRT serves as a tool for assessing the quality of the drug product [160]. According to FDA, the IVRT studies for semi-solid dosage forms are conducted using either the vertical diffusion cell or an immersion cell. The vertical diffusion cell consists of receptor and donor chambers, separated by a receptor membrane. The donor chamber holds the sample of dosage form, while the receptor chamber holds the receptor media. The receptor media can be a buffer or hydro-alcoholic solution, selected based on the solubility, sink condition, and stability of the API. The skin-like receptor membrane is selected based on the effective pore size, high permeability and expected inertness towards the API. If necessary, the receptor membrane should be saturated with release media. The temperature of the media should be maintained around 32 °C ± 1 °C for topical administering products, for products intended for mucosal membrane the temperature should be 37 °C ± 1 °C. A Teflon-coated magnetic stirrer is used for stirring the receptor media. While the immersion cell model has a cell body, which acts as a reservoir [161]. The cell body is covered with a membrane and closed using a leakproof seal (retaining ring cap) that ensures no leakage of the dosage form. The retaining ring cap possesses an opening on the top, and it should be adjusted in such a way that the membrane is in contact with the dosage form on the bottom and release media on the top. The whole setup is used along with the USP-2 apparatus, wherein the immersion cell is placed in flat bottomed dissolution vessel with a usual volume of 150–200 mL. A mini spin-paddle is used for stirring or agitating the media [162].

9.6. Bio-Adhesive Property

Bio-adhesive strength is used to determine the force required to detach the drug carrier system from a biological surface. This property is important for a topical dosage form if prolonged contact is required [163]. This test is usually performed using rat or pig skin, the latter is preferred because of its resemblance to human skin. There are various techniques to measure this property but none of them is approved by FDA. The texture analyzer is one such technique, where the upper mobile probe and stationary lower base plate will be covered with skin. The dosage form is placed on the skin of the base plate. The upper probe is lowered to contact the lower base plate and the contact is maintained for at least a minute. The upper probe is lifted slowly until the separation of skin sheets. The force required to separate the two skin sheets will be measured by the instrument and represented as the area under the force-distance curve [164].

10. Safety Issues

One of the crucial concerns while developing a skin-related formulation is toxicity and skin irritation [165]. Impairment of enzyme activity, disturbance in normal physiological functions, and sometimes carcinogenic effects (For e.g., being caused by Sodium do-decyl benzene sulfonate) are some common toxicity issues related to surfactants [166]. Smith and the group analyzed the effect of two surface active agents’ sodium dodecyl sulfate and dodecyl trimethyl ammonium bromide on penetration and skin perturbation. They concluded that disruption in the layer of skin is primarily caused by elevated concentrations of micelle agglomerate and monomers [167].
Irritation caused by topical nano-emulgel can be examined by applying it on the shaven skin of a rat, then observation of redness and other signs of inflammation on the skin were made and then graded based on the number of eurythmic spots to assess their clinical implication as give in Table 8 [167]. In general, a grade scale up to 2 is safe. Azeem et al. prepared ropinirole nano-emulgel using caproyl 90, tween 20 and carbital. The skin irritation studies showed a grade 2 erythema index, which is safe [168]. Gannu et al. also performed skin irritation studies of nano-emulgel prepared using non-ionic surfactant the Tween 80 and co-surfactant labrasol. They observed no signs of skin irritation as the used surfactants are generally considered safe [25]. Usually, major toxic effects are observed with cationic surfactants so they are avoided in preparations associated with topical delivery. While nonionic surfactants are mostly preferred as they cause minimum perturbation of the skin layer [151,166].

11. Challenges

The impartment of large drug entities with molecular weight exceeding 400 Dalton is hindered in this dosage form, as they show difficulty during size reduction and are found to leach out of the gel mesh network. A limited number of safe surfactants and co-surfactants are available for emulgel preparation. Not much maneuvering can be done with the selection of surfactant as it can have hazardous consequences. The abundance of surfactant in emulgel can lead to skin problems like contact dermatitis, erythema, redness of skin, skin layer perturbation [169]. High susceptibility of the gelling agent toward variations in pH and temperature can lead to the breaking of gel structure and the leaching of chemicals [170].
Capriciousness in nano-emulsion is caused due to Ostwald ripening, which is associated with nano size of oil droplets, preferably nano-emulsion is prepared shortly before its application. Optimizing the speed of the stirrer in the homogenizer (as required to produce an inflexible and non-cracking gel), mixing appropriate quantities of surface-active agents, and selecting a reliable packing material are very pivotal tasks associated with the stability of nano-emulgel [171]. Highly specialized instruments are required for size reduction to nanoscale, which requires handling by skilled labour. Expensive sustenance of high energy homogenizers and production cost is one of the critical limitations associated with scale up of nano-emulgel formulation. Besides these disadvantages, the comforting prospect of nano-emulgel is increased adherent property and elevated embranglement of the drug in the gel mesh [53]. Also, prevalent drawbacks associated with conventional topical dosage forms i.e., emulsion, ointment, lotions etc. such as creaming, phase disruption, oxidation induced degradation of ointments are overcome by forming emulgel [44].

12. Current and Future Prospects of Nanoemulgel

Delivering hydrophobic drugs to the biological systems has been a major challenge in formulation development owing to their low solubility, leading to poor bioavailability. Some of the topical formulations include creams, ointments, and lotions. They possess good emollient characteristics, however, has slow drug release kinetics due to the presence of hydrophobic oleaginous bases such as petrolatum, beeswax, and vegetable oils, which inhibit the incorporation of water or aqueous phase. On contrary, topical aqueous-based formulations like gels enhance the drug release from the medication since it provides an aqueous environment for medicament. Therefore, hydrophobic APIs are blended with oily bases to form an emulgel, which further undergoes nanonization to form a nanoemulgel with enhanced properties. The superior properties of a nanoemulgel like thermodynamic stability, permeation enhancement, and sustained release make it an excellent dosage form. There are several marketed emulgels and patents being filed (Table 9) for the same, demonstrating its tremendous progress in this field. By making advancements in the ongoing research, nanoemulgel, as a delivery system would outshine, in formulating the drugs that are being eliminated from the development pipeline owing to their poor bioavailability, therapeutic non-efficacy, etc. Despite these advantages, the manufacturing of nano-emulsion limits its commercialization. However, with the progressing technology, commercially feasible and profitable manufacturing techniques could be possible in the future. With the advantages of nano-emulgel over other formulations, a tremendous increase in the production of nano-emulgel can be foreseen.

13. Conclusions

The selection of ingredients and their appropriate ratios play a vital role in deciding the properties of a nano-emulgel. Deviation from this could affect the conversion of a nano-emulsion to a nano-emulgel and its thermodynamic stability. The nano-emulgel is more stable compared to that of a nano-emulsion mainly due to its less mobile dispersed phase and the decreased interfacial tension. Thus, the former is a better alternative in delivering lipophilic moieties mainly due to improved permeation, and better pharmacokinetics, which subsequently improves the pharmacological effect. Patient compliance is also elevated due to its non-greasy and improved spreading properties on topical administration. Despite of its advantages, nano-emulgel is still at its infancy in the prospect of the pharmaceutical industry. However, various emulgels are being marketed e.g., Voltron emulgel, which holds out hope for the commercialization of nano-emulgel in near future. Hence it has the potential to become a center of attention due to its safety, efficacy, and user-friendly nature for topical drug delivery. Despite some disadvantages, nano-emulgel is a tool for the future which may be an alternative to traditional formulations.

Author Contributions

Writing—original draft, M.R.D.; Writing—review & editing, S.R.M.; Writing, review & editing, K.V.K.; Resources, Supervision, R.N.S.; editing, Resources, Supervision, G.S.; Conceptualization, Review & editing, Resources, Supervision S.K.D. All authors have read and agreed to the published version of the manuscript.

Funding

This review received no external funding.

Conflicts of Interest

The authors declare that they have no known competing financial interest or personal relationships that could have appeared to influence the work reported in this paper.

References

  1. Kalepu, S.; Nekkanti, V. Insoluble Drug Delivery Strategies: Review of Recent Advances and Business Prospects. Acta Pharm. Sin. B 2015, 5, 442–453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Donthi, M.R.; Munnangi, S.R.; Krishna, K.V.; Marathe, S.A.; Saha, R.N.; Singhvi, G.; Dubey, S.K. Formulating Ternary Inclusion Complex of Sorafenib Tosylate Using β-Cyclodextrin and Hydrophilic Polymers: Physicochemical Characterization and In Vitro Assessment. AAPS PharmSciTech 2022, 23, 1–15. [Google Scholar] [CrossRef] [PubMed]
  3. Singh, G.; Singh, D.; Choudhari, M.; Kaur, S.D.; Dubey, S.K.; Arora, S.; Bedi, N. Exemestane Encapsulated Copolymers L121/F127/GL44 Based Mixed Micelles: Solubility Enhancement and in Vitro Cytotoxicity Evaluation Using MCF-7 Breast Cancer Cells. J. Pharm. Investig. 2021, 51, 701–714. [Google Scholar] [CrossRef]
  4. Alekya, T.; Narendar, D.; Mahipal, D.; Arjun, N.; Nagaraj, B. Design and Evaluation of Chronomodulated Drug Delivery of Tramadol Hydrochloride. Drug Res. 2018, 68, 174–180. [Google Scholar] [CrossRef] [PubMed]
  5. Homayun, B.; Lin, X.; Choi, H.J. Challenges and Recent Progress in Oral Drug Delivery Systems for Biopharmaceuticals. Pharmaceutics 2019, 11, 129. [Google Scholar] [CrossRef] [Green Version]
  6. Donthi, M.R.; Dudhipala, N.R.; Komalla, D.R.; Suram, D.; Banala, N. Preparation and Evaluation of Fixed Combination of Ketoprofen Enteric Coated and Famotidine Floating Mini Tablets by Single Unit Encapsulation System. J. Bioequiv. Availab. 2015, 7, 1–5. [Google Scholar] [CrossRef]
  7. Wang, W.; Zhou, H.; Liu, L. Side Effects of Methotrexate Therapy for Rheumatoid Arthritis: A Systematic Review. Eur. J. Med. Chem. 2018, 158, 502–516. [Google Scholar] [CrossRef]
  8. Rajitha, R.; Narendar, D.; Arjun, N.; Nagaraj, B. Colon Delivery of Naproxen: Preparation, Characterization and Clinical Evaluation in Healthy Volunteers. Int. J. Pharm. Sci. Nanotechnol. 2016, 9, 3383–3389. [Google Scholar] [CrossRef]
  9. Garg, N.K.; Singh, B.; Tyagi, R.K.; Sharma, G.; Katare, O.P. Effective Transdermal Delivery of Methotrexate through Nanostructured Lipid Carriers in an Experimentally Induced Arthritis Model. Colloids Surf. B Biointerfaces 2016, 147, 17–24. [Google Scholar] [CrossRef]
  10. Szumała, P.; Macierzanka, A. Topical Delivery of Pharmaceutical and Cosmetic Macromolecules Using Microemulsion Systems. Int. J. Pharm. 2022, 615, 121488. [Google Scholar] [CrossRef]
  11. Gupta, R.; Dwadasi, B.S.; Rai, B.; Mitragotri, S. Effect of Chemical Permeation Enhancers on Skin Permeability: In Silico Screening Using Molecular Dynamics Simulations. Sci. Rep. 2019, 9, 1456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Saka, R.; Jain, H.; Kommineni, N.; Chella, N.; Khan, W. Enhanced Penetration and Improved Therapeutic Efficacy of Bexarotene via Topical Liposomal Gel in Imiquimod Induced Psoriatic Plaque Model in BALB/c Mice. J. Drug Deliv. Sci. Technol. 2020, 58, 101691. [Google Scholar] [CrossRef]
  13. Pandi, P.; Jain, A.; Kommineni, N.; Ionov, M.; Bryszewska, M.; Khan, W. Dendrimer as a New Potential Carrier for Topical Delivery of SiRNA: A Comparative Study of Dendriplex vs. Lipoplex for Delivery of TNF-$α$ SiRNA. Int. J. Pharm. 2018, 550, 240–250. [Google Scholar] [CrossRef] [PubMed]
  14. Sarathlal, K.C.S.; Kakoty, V.; Krishna, K.V.; Dubey, S.K.; Chitkara, D.; Taliyan, R. Neuroprotective Efficacy of Co-Encapsulated Rosiglitazone and Vorinostat Nanoparticle on Streptozotocin Induced Mice Model of Alzheimer Disease. ACS Chem. Neurosci. 2021, 12, 1528–1541. [Google Scholar] [CrossRef]
  15. Nafisi, S.; Maibach, H.I. Nanotechnology in Cosmetics. In Cosmetic Science and Technology: Theoretical Principles and Applications; Elsevier: Amsterdam, The Netherlands, 2017; pp. 337–369. [Google Scholar] [CrossRef]
  16. Tiwari, N.; Osorio-Blanco, E.R.; Sonzogni, A.; Esporr\’\in-Ubieto, D.; Wang, H.; Calderon, M. Nanocarriers for Skin Applications: Where Do We Stand? Angew. Chem. Int. Ed. 2022, 61, e202107960. [Google Scholar] [CrossRef]
  17. Shukla, T.; Upmanyu, N.; Agrawal, M.; Saraf, S.; Saraf, S.; Alexander, A. Biomedical Applications of Microemulsion through Dermal and Transdermal Route. Biomed. Pharmacother. 2018, 108, 1477–1494. [Google Scholar] [CrossRef]
  18. Nastiti, C.M.R.R.; Ponto, T.; Abd, E.; Grice, J.E.; Benson, H.A.E.; Roberts, M.S. Topical Nano and Microemulsions for Skin Delivery. Pharmaceutics 2017, 9, 37. [Google Scholar] [CrossRef]
  19. Aithal, G.C.; Narayan, R.; Nayak, U.Y. Nanoemulgel: A Promising Phase in Drug Delivery. Curr. Pharm. Des. 2020, 26, 279–291. [Google Scholar] [CrossRef]
  20. Anand, K.; Ray, S.; Rahman, M.; Shaharyar, A.; Bhowmik, R.; Bera, R.; Karmakar, S. Nano-Emulgel: Emerging as a Smarter Topical Lipidic Emulsion-Based Nanocarrier for Skin Healthcare Applications. Recent Pat. Antiinfect. Drug Discov. 2019, 14, 16–35. [Google Scholar] [CrossRef]
  21. Murthy, S.N. Approaches for Delivery of Drugs Topically. AAPS PharmSciTech 2019, 21, 1–2. [Google Scholar] [CrossRef]
  22. Iqbal, M.A.; Md, S.; Sahni, J.K.; Baboota, S.; Dang, S.; Ali, J. Nanostructured Lipid Carriers System: Recent Advances in Drug Delivery. J. Drug Target. 2012, 20, 813–830. [Google Scholar] [CrossRef] [PubMed]
  23. Bhowmik, D.; Gopinath, H.; Kumar, B.P.; Duraivel, S.; Kumar, K.P.S. Recent Advances In Novel Topical Drug Delivery System. Pharma Innov. J. 2012, 1, 12–31. [Google Scholar]
  24. Gannu, R.; Palem, C.R.; Yamsani, V.V.; Yamsani, S.K.; Yamsani, M.R. Enhanced Bioavailability of Lacidipine via Microemulsion Based Transdermal Gels: Formulation Optimization, Ex Vivo and in Vivo Characterization. Int. J. Pharm. 2010, 388, 231–241. [Google Scholar] [CrossRef] [PubMed]
  25. Bhaskar, K.; Anbu, J.; Ravichandiran, V.; Venkateswarlu, V.; Rao, Y.M. Lipid Nanoparticles for Transdermal Delivery of Flurbiprofen: Formulation, in Vitro, Ex Vivo and in Vivo Studies. Lipids Health Dis. 2009, 8, 1–15. [Google Scholar] [CrossRef] [Green Version]
  26. Zhou, J.; Zhou, M.; Yang, F.F.; Liu, C.Y.; Pan, R.L.; Chang, Q.; Liu, X.M.; Liao, Y.H. Involvement of the Inhibition of Intestinal Glucuronidation in Enhancing the Oral Bioavailability of Resveratrol by Labrasol Containing Nanoemulsions. Mol. Pharm. 2015, 12, 1084–1095. [Google Scholar] [CrossRef]
  27. Chang, R.K.; Raw, A.; Lionberger, R.; Yu, L. Generic Development of Topical Dermatologic Products: Formulation Development, Process Development, and Testing of Topical Dermatologic Products. AAPS J. 2013, 15, 41–52. [Google Scholar] [CrossRef] [Green Version]
  28. Cevc, G. Lipid Vesicles and Other Colloids as Drug Carriers on the Skin. Adv. Drug Deliv. Rev. 2004, 56, 675–711. [Google Scholar] [CrossRef]
  29. Chellapa, P.; Mohamed, A.T.; Keleb, E.I.; Elmahgoubi, A.; Eid, A.M.; Issa, Y.S.; Elmarzugi, N.A. Nanoemulsion and Nanoemulgel as a Topical Formulation. IOSR J. Pharm. 2015, 5, 43–47. [Google Scholar]
  30. Marto, J.; Baltazar, D.; Duarte, A.; Fernandes, A.; Gouveia, L.; Militão, M.; Salgado, A.; Simões, S.; Oliveira, E.; Ribeiro, H.M. Topical Gels of Etofenamate: In Vitro and in Vivo Evaluation. Pharm. Dev. Technol. 2015, 20, 710–715. [Google Scholar] [CrossRef]
  31. Lau, W.; White, A.; Gallagher, S.; Donaldson, M.; McNaughton, G.; Heard, C. Scope and Limitations of the Co-Drug Approach to Topical Drug Delivery. Curr. Pharm. Des. 2008, 14, 794–802. [Google Scholar] [CrossRef]
  32. Raza, K.; Kumar, M.; Kumar, P.; Malik, R.; Sharma, G.; Kaur, M.; Katare, O.P. Topical Delivery of Aceclofenac: Challenges and Promises of Novel Drug Delivery Systems. Biomed Res. Int. 2014, 2014, 406731. [Google Scholar] [CrossRef] [PubMed]
  33. Somagoni, J.; Boakye, C.H.A.; Godugu, C.; Patel, A.R.; Faria, H.A.M.; Zucolotto, V.; Singh, M. Nanomiemgel--a Novel Drug Delivery System for Topical Application--in Vitro and in Vivo Evaluation. PLoS ONE 2014, 9, e115952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Begur, M.; Vasantakumar Pai, K.; Gowda, D.V.; Srivastava, A.; Raghundan, H.V. Development and Characterization of Nanoemulgel Based Transdermal Delivery System for Enhancing Permeability of Tacrolimus. Adv. Sci. Eng. Med. 2016, 8, 324–332. [Google Scholar] [CrossRef]
  35. Ngawhirunpat, T.; Worachun, N.; Opanasopit, P.; Rojanarata, T.; Panomsuk, S. Cremophor RH40-PEG 400 Microemulsions as Transdermal Drug Delivery Carrier for Ketoprofen. Pharm. Dev. Technol. 2013, 18, 798–803. [Google Scholar] [CrossRef]
  36. Bos, J.D.; Meinardi, M.M.H.M. The 500 Dalton Rule for the Skin Penetration of Chemical Compounds and Drugs. Exp. Dermatol. 2000, 9, 165–169. [Google Scholar] [CrossRef]
  37. Phad, A.R.; Dilip, N.T.; Sundara Ganapathy, R. Emulgel: A Comprehensive Review for Topical Delivery of Hydrophobic Drugs. Asian J. Pharm. 2018, 12, 382. [Google Scholar]
  38. Azeem, A.; Rizwan, M.; Ahmad, F.J.; Iqbal, Z.; Khar, R.K.; Aqil, M.; Talegaonkar, S. Nanoemulsion Components Screening and Selection: A Technical Note. AAPS PharmSciTech 2009, 10, 69. [Google Scholar] [CrossRef]
  39. Gao, F.; Zhang, Z.; Bu, H.; Huang, Y.; Gao, Z.; Shen, J.; Zhao, C.; Li, Y. Nanoemulsion Improves the Oral Absorption of Candesartan Cilexetil in Rats: Performance and Mechanism. J. Control. Release 2011, 149, 168–174. [Google Scholar] [CrossRef]
  40. Kim, B.S.; Won, M.; Lee, K.M.; Kim, C.S. In Vitro Permeation Studies of Nanoemulsions Containing Ketoprofen as a Model Drug. Drug Deliv. 2008, 15, 465–469. [Google Scholar] [CrossRef] [Green Version]
  41. Akhter, S.; Jain, G.; Ahmad, F.; Khar, R.; Jain, N.; Khan, Z.; Talegaonkar, S. Investigation of Nanoemulsion System for Transdermal Delivery of Domperidone: Ex-Vivo and in Vivo Studies. Curr. Nanosci. 2008, 4, 381–390. [Google Scholar] [CrossRef]
  42. El Maghraby, G.M. Transdermal Delivery of Hydrocortisone from Eucalyptus Oil Microemulsion: Effects of Cosurfactants. Int. J. Pharm. 2008, 355, 285–292. [Google Scholar] [CrossRef] [PubMed]
  43. Huang, Y.B.; Lin, Y.H.; Lu, T.M.; Wang, R.J.; Tsai, Y.H.; Wu, P.C. Transdermal Delivery of Capsaicin Derivative-Sodium Nonivamide Acetate Using Microemulsions as Vehicles. Int. J. Pharm. 2008, 349, 206–211. [Google Scholar] [CrossRef] [PubMed]
  44. Eid, A.M.; El-Enshasy, H.A.; Aziz, R.; Elmarzugi, N.A. Preparation, Characterization and Anti-Inflammatory Activity of Swietenia Macrophylla Nanoemulgel. J. Nanomed. Nanotechnol. 2014, 5, 1–10. [Google Scholar] [CrossRef]
  45. Algahtani, M.S.; Ahmad, M.Z.; Ahmad, J. Nanoemulgel for Improved Topical Delivery of Retinyl Palmitate: Formulation Design and Stability Evaluation. Nanomaterials 2020, 10, 848. [Google Scholar] [CrossRef]
  46. Bernardi, D.S.; Pereira, T.A.; Maciel, N.R.; Bortoloto, J.; Viera, G.S.; Oliveira, G.C.; Rocha-Filho, P.A. Formation and Stability of Oil-in-Water Nanoemulsions Containing Rice Bran Oil: In Vitro and in Vivo Assessments. J. Nanobiotechnol. 2011, 9, 44. [Google Scholar] [CrossRef]
  47. Bolzinger, M.A.; Briançon, S.; Pelletier, J.; Fessi, H.; Chevalier, Y. Percutaneous Release of Caffeine from Microemulsion, Emulsion and Gel Dosage Forms. Eur. J. Pharm. Biopharm. 2008, 68, 446–451. [Google Scholar] [CrossRef]
  48. Fini, A.; Bergamante, V.; Ceschel, G.C.; Ronchi, C.; Moraes, C.A.F. Control of Transdermal Permeation of Hydrocortisone Acetate from Hydrophilic and Lipophilic Formulations. AAPS PharmSciTech 2008, 9, 762. [Google Scholar] [CrossRef] [Green Version]
  49. Teichmann, A.; Heuschkel, S.; Jacobi, U.; Presse, G.; Neubert, R.H.H.; Sterry, W.; Lademann, J. Comparison of Stratum Corneum Penetration and Localization of a Lipophilic Model Drug Applied in an o/w Microemulsion and an Amphiphilic Cream. Eur. J. Pharm. Biopharm. 2007, 67, 699–706. [Google Scholar] [CrossRef]
  50. Khurana, S.; Jain, N.K.; Bedi, P.M.S. Nanoemulsion Based Gel for Transdermal Delivery of Meloxicam: Physico-Chemical, Mechanistic Investigation. Life Sci. 2013, 92, 383–392. [Google Scholar] [CrossRef]
  51. Mou, D.; Chen, H.; Du, D.; Mao, C.; Wan, J.; Xu, H.; Yang, X. Hydrogel-Thickened Nanoemulsion System for Topical Delivery of Lipophilic Drugs. Int. J. Pharm. 2008, 353, 270–276. [Google Scholar] [CrossRef]
  52. Pund, S.; Pawar, S.; Gangurde, S.; Divate, D. Transcutaneous Delivery of Leflunomide Nanoemulgel: Mechanistic Investigation into Physicomechanical Characteristics, in Vitro Anti-Psoriatic and Anti-Melanoma Activity. Int. J. Pharm. 2015, 487, 148–156. [Google Scholar] [CrossRef] [PubMed]
  53. Dev, A.; Chodankar, R.; Shelke, O. Emulgels: A Novel Topical Drug Delivery System. Pharm. Biol. Eval. 2015, 2, 64–75. [Google Scholar]
  54. Sengupta, P.; Chatterjee, B. Potential and Future Scope of Nanoemulgel Formulation for Topical Delivery of Lipophilic Drugs. Int. J. Pharm. 2017, 526, 353–365. [Google Scholar] [CrossRef] [PubMed]
  55. Arora, R.; Aggarwal, G.; Harikumar, S.L.; Kaur, K. Nanoemulsion Based Hydrogel for Enhanced Transdermal Delivery of Ketoprofen. Adv. Pharm. 2014, 2014, 468456. [Google Scholar] [CrossRef]
  56. Gorain, B.; Choudhury, H.; Tekade, R.K.; Karan, S.; Jaisankar, P.; Pal, T.K. Comparative Biodistribution and Safety Profiling of Olmesartan Medoxomil Oil-in-Water Oral Nanoemulsion. Regul. Toxicol. Pharmacol. 2016, 82, 20–31. [Google Scholar] [CrossRef]
  57. Dubey, S.K.; Ram, M.S.; Krishna, K.V.; Saha, R.N.; Singhvi, G.; Agrawal, M.; Ajazuddin; Saraf, S.; Saraf, S.; Alexander, A. Recent Expansions on Cellular Models to Uncover the Scientific Barriers Towards Drug Development for Alzheimer’s Disease. Cell. Mol. Neurobiol. 2019, 39, 181–209. [Google Scholar] [CrossRef]
  58. Formariz, T.P.; Sarmento, V.H.V.; Silva-Junior, A.A.; Scarpa, M.V.; Santilli, C.V.; Oliveira, A.G. Doxorubicin Biocompatible O/W Microemulsion Stabilized by Mixed Surfactant Containing Soya Phosphatidylcholine. Colloids Surf. B Biointerfaces 2006, 51, 54–61. [Google Scholar] [CrossRef]
  59. Ahmad, M.Z.; Ahmad, J.; Alasmary, M.Y.; Akhter, S.; Aslam, M.; Pathak, K.; Jamil, P.; Abdullah, M.M. Nanoemulgel as an Approach to Improve the Biopharmaceutical Performance of Lipophilic Drugs: Contemporary Research and Application. J. Drug Deliv. Sci. Technol. 2022, 72, 103420. [Google Scholar] [CrossRef]
  60. Kaur, G. TPGS Loaded Topical Nanoemulgel of Mefenamic Acid for the Treatment of Rheumatoid Arthritis. Int. J. Pharm. Pharm. Res. 2019, 15, 64–107. [Google Scholar]
  61. Tesch, S.; Gerhards, C.; Schubert, H. Stabilization of Emulsions by OSA Starches. J. Food Eng. 2002, 54, 167–174. [Google Scholar] [CrossRef]
  62. Ingle, A.P.; Shende, S.; Gupta, I.; Rai, M. Recent Trends in the Development of Nano-Bioactive Compounds and Delivery Systems. In Biotechnological Production of Bioactive Compounds; Elsevier: Amsterdam, The Netherlands, 2020; pp. 409–431. [Google Scholar]
  63. Ashara, K.C. Microemulgel: An overwhelming approach to improve therapeutic action of drug moiety. Saudi Pharm. J. 2014, 24, 452–457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Sultana, N.; Akhtar, J.; Khan, M.I.; Ahmad, U.; Arif, M.; Ahmad, M.; Upadhyay, T. Nanoemulgel: For Promising Topical and Systemic Delivery. In Drug Development Life Cycle; IntechOpen: London, UK, 2022. [Google Scholar]
  65. Zhou, H.; Yue, Y.; Liu, G.; Li, Y.; Zhang, J.; Gong, Q.; Yan, Z.; Duan, M. Preparation and Characterization of a Lecithin Nanoemulsion as a Topical Delivery System. Nanoscale Res. Lett. 2010, 5, 224–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Li, P.; Nielsen, H.M.; Müllertz, A. Oral Delivery of Peptides and Proteins Using Lipid-Based Drug Delivery Systems. Expert Opin. Drug Deliv. 2012, 9, 1289–1304. [Google Scholar] [CrossRef] [PubMed]
  67. Bahadur, S.; Pardhi, D.M.; Rautio, J.; Rosenholm, J.M.; Pathak, K. Intranasal Nanoemulsions for Direct Nose-to-Brain Delivery of Actives for Cns Disorders. Pharmaceutics 2020, 12, 1230. [Google Scholar] [CrossRef] [PubMed]
  68. Chatterjee, B.; Gorain, B.; Mohananaidu, K.; Sengupta, P.; Mandal, U.K.; Choudhury, H. Targeted Drug Delivery to the Brain via Intranasal Nanoemulsion: Available Proof of Concept and Existing Challenges. Int. J. Pharm. 2019, 565, 258–268. [Google Scholar] [CrossRef] [PubMed]
  69. Algahtani, M.S.; Ahmad, M.Z.; Nourein, I.H.; Albarqi, H.A.; Alyami, H.S.; Alyami, M.H.; Alqahtani, A.A.; Alasiri, A.; Algahtani, T.S.; Mohammed, A.A.; et al. Preparation and Characterization of Curcumin Nanoemulgel Utilizing Ultrasonication Technique for Wound Healing: In Vitro, Ex Vivo, and in Vivo Evaluation. Gels 2021, 7, 213. [Google Scholar] [CrossRef]
  70. Algahtani, M.S.; Ahmad, M.Z.; Shaikh, I.A.; Abdel-Wahab, B.A.; Nourein, I.H.; Ahmad, J. Thymoquinone Loaded Topical Nanoemulgel for Wound Healing: Formulation Design and In-Vivo Evaluation. Molecules 2021, 26, 3863. [Google Scholar] [CrossRef] [PubMed]
  71. Alyoussef, A.; El-Gogary, R.I.; Ahmed, R.F.; Ahmed Farid, O.A.; Bakeer, R.M.; Nasr, M. The Beneficial Activity of Curcumin and Resveratrol Loaded in Nanoemulgel for Healing of Burn-Induced Wounds. J. Drug Deliv. Sci. Technol. 2021, 62, 102360. [Google Scholar] [CrossRef]
  72. Abdallah, M.; Lila, A.; Unissa, R.; Elsewedy, H.S.; Elghamry, H.A.; Soliman, M.S. Preparation, Characterization and Evaluation of Anti-Inflammatory and Anti-Nociceptive Effects of Brucine-Loaded Nanoemulgel. Colloids Surf. B Biointerfaces 2021, 205, 111868. [Google Scholar] [CrossRef]
  73. Zakir, F.; Ahmad, A.; Mirza, M.A.; Kohli, K.; Ahmad, F.J. Exploration of a Transdermal Nanoemulgel as an Alternative Therapy for Postmenopausal Osteoporosis. J. Drug Deliv. Sci. Technol. 2021, 65, 102745. [Google Scholar] [CrossRef]
  74. Mao, Y.; Chen, X.; Xu, B.; Shen, Y.; Ye, Z.; Chaurasiya, B.; Liu, L.; Li, Y.; Xing, X.; Chen, D. Eprinomectin Nanoemulgel for Transdermal Delivery against Endoparasites and Ectoparasites: Preparation, in Vitro and in Vivo Evaluation. Taylor Fr. 2019, 26, 1104–1114. [Google Scholar] [CrossRef] [Green Version]
  75. Gadhave, D.; Tupe, S.; Tagalpallewar, A.; Gorain, B.; Choudhury, H.; Kokare, C. Nose-to-Brain Delivery of Amisulpride-Loaded Lipid-Based Poloxamer-Gellan Gum Nanoemulgel: In Vitro and in Vivo Pharmacological Studies. Int. J. Pharm. 2021, 607, 121050. [Google Scholar] [CrossRef]
  76. Qu, Y.; Li, A.; Ma, L.; Iqbal, S.; Sun, X.; Ma, W.; Li, C. Nose-to-Brain Delivery of Disulfiram Nanoemulsion in Situ Gel Formulation for Glioblastoma Targeting Therapy. Int. J. Pharm. 2021, 597, 120250. [Google Scholar] [CrossRef]
  77. Topical Metformin Emulgel VS Salicylic Acid Peeling in Treatment of Acne Vulgaris—Full Text View—ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/NCT05536193 (accessed on 1 December 2022).
  78. Comparison of the Bioavailability of Diclofenac in a Combination Product (Diclofenac 2% + Capsaicin 0.075% Topical Gel) with Two Diclofenac Only Products, Diclofenac Mono Gel 2% and Voltarol® 12 Hour Emulgel 2.32% Gel, in Healthy Volunteers—Full Text View—ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03074162 (accessed on 1 December 2022).
  79. Effects of Visnadin, Ethyl Ximeninate, Coleus Barbatus and Millet in Emulgel on Sexual Function in Postmenopausal Women—Full Text View—ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/NCT04579991 (accessed on 1 December 2022).
  80. Clinical Assessment of Voriconazole Self Nano Emulsifying Drug Delivery System Intermediate Gel—Full Text View—ClinicalTrials.Gov. Available online: https://www.clinicaltrials.gov/ct2/show/NCT04110860 (accessed on 1 December 2022).
  81. Clinical Assessment of Itraconazole Self Nano Emulsifying Drug Delivery System Intermediate Gel—Full Text View—ClinicalTrials.Gov. Available online: https://www.clinicaltrials.gov/ct2/show/NCT04110834 (accessed on 1 December 2022).
  82. Study of Efficacy and Tolerability of SYSTANE Complete in Patients with Dry Eye Disease—Full Text View—ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03492541 (accessed on 1 December 2022).
  83. Santhosh, A.; Kumar, A.; Pramanik, R.; Gogia, A.; Prasad, C.P.; Gupta, I.; Gupta, N.; Cheung, W.Y.; Pandey, R.M.; Sharma, A.; et al. Randomized Double-Blind, Placebo-Controlled Study of Topical Diclofenac in the Prevention of Hand-Foot Syndrome in Patients Receiving Capecitabine (the D-TORCH Study). Trials 2022, 23, 420. [Google Scholar] [CrossRef]
  84. Eswaraiah, S.; Swetha, K. Emulgel: Review on Novel Approach to Topical Drug Delivery. Available online: https://asianjpr.com/AbstractView.aspx?PID=2014-4-1-2 (accessed on 1 December 2022).
  85. Jeengar, M.K.; Rompicharla, S.V.K.; Shrivastava, S.; Chella, N.; Shastri, N.R.; Naidu, V.G.M.; Sistla, R. Emu Oil Based Nano-Emulgel for Topical Delivery of Curcumin. Int. J. Pharm. 2016, 506, 222–236. [Google Scholar] [CrossRef]
  86. Md, S.; Alhakamy, N.A.; Aldawsari, H.M.; Kotta, S.; Ahmad, J.; Akhter, S.; Alam, M.S.; Khan, M.A.; Awan, Z.; Sivakumar, P.M. Improved Analgesic and Anti-Inflammatory Effect of Diclofenac Sodium by Topical Nanoemulgel: Formulation Development—In Vitro and in Vivo Studies. J. Chem. 2020, 2020, 4071818. [Google Scholar] [CrossRef] [Green Version]
  87. Drais, H.K.; Hussein, A.A. Formulation Characterization and Evaluation of Meloxicam Nanoemulgel to Be Used Topically. Iraqi J. Pharm. Sci. 2017, 26, 9–16. [Google Scholar]
  88. Aithal, G.C.; Nayak, U.Y.; Mehta, C.; Narayan, R.; Gopalkrishna, P.; Pandiyan, S.; Garg, S. Localized In Situ Nanoemulgel Drug Delivery System of Quercetin for Periodontitis: Development and Computational Simulations. Molecules 2018, 23, 1363. [Google Scholar] [CrossRef] [Green Version]
  89. Wankar, J.; Ajimera, T. Design, Development and Evaluation of Nanoemulsion and Nanogel of Itraconazole for Transdermal Delivery. J. Sci. Res. Pharm. 2014, 3, 6–11. [Google Scholar]
  90. Pathak, M.K.; Chhabra, G.; Pathak, K. Design and Development of a Novel PH Triggered Nanoemulsified In-Situ Ophthalmic Gel of Fluconazole: Ex-Vivo Transcorneal Permeation, Corneal Toxicity and Irritation Testing. Drug Dev. Ind. Pharm. 2013, 39, 780–790. [Google Scholar] [CrossRef]
  91. Wais, M.; Samad, A.; Nazish, I.; Khale, A.; Aqil, M.; Khan, M. Formulation development ex-vivo and in-vivo evaluation of nanoemulsion for transdermal delivery of glibenclamide. Int. J. Pharm. Pharm. Sci. 2013, 5, 747–754. [Google Scholar]
  92. Pratap, S.B.; Brajesh, K.; Jain, S.K.; Kausar, S. Development and Characterization of A Nanoemulsion Gel for Transdermal Delivery of Carvedilol. Int. J. Drug Dev. Res. 2012, 4, 151–161. [Google Scholar]
  93. Begur, M.; Pai, V.; Gowda, D.V.; AtulSrivastava; Raghundan, H.V.; Shinde, C.G.; Manusri, N. Enhanced Permeability of Cyclosporine from a Transdermally Applied Nanoemulgel. Der Pharm. Sin. 2015. Available online: https://hal.archives-ouvertes.fr/hal-03627803 (accessed on 1 December 2022).
  94. Nagaraja, S.; Basavarajappa, G.M.; Attimarad, M.; Pund, S. Topical Nanoemulgel for the Treatment of Skin Cancer: Proof-of-Technology. Pharmaceutics 2021, 13, 902. [Google Scholar] [CrossRef]
  95. Morsy, M.A.; Abdel-Latif, R.G.; Nair, A.B.; Venugopala, K.N.; Ahmed, A.F.; Elsewedy, H.S.; Shehata, T.M. Preparation and Evaluation of Atorvastatin-Loaded Nanoemulgel on Wound-Healing Efficacy. Pharmaceutics 2019, 11, 609. [Google Scholar] [CrossRef] [Green Version]
  96. Soliman, W.E.; Shehata, T.M.; Mohamed, M.E.; Younis, N.S.; Elsewedy, H.S. Enhancement of Curcumin Anti-Inflammatory Effect via Formulation into Myrrh Oil-Based Nanoemulgel. Polymers 2021, 13, 577. [Google Scholar] [CrossRef]
  97. Elmataeeshy, M.E.; Sokar, M.S.; Bahey-El-Din, M.; Shaker, D.S. Enhanced Transdermal Permeability of Terbinafine through Novel Nanoemulgel Formulation; Development, in Vitro and in Vivo Characterization. Future J. Pharm. Sci. 2018, 4, 18–28. [Google Scholar] [CrossRef]
  98. Vartak, R.; Menon, S.; Patki, M.; Billack, B.; Patel, K. Ebselen Nanoemulgel for the Treatment of Topical Fungal Infection. Eur. J. Pharm. Sci. 2020, 148, 105323. [Google Scholar] [CrossRef]
  99. Vandamme, T.F. Microemulsions as Ocular Drug Delivery Systems: Recent Developments and Future Challenges. Prog. Retin. Eye Res. 2002, 21, 15–34. [Google Scholar] [CrossRef]
  100. Bashir, M.; Ahmad, J.; Asif, M.; Khan SU, D.; Irfan, M.; Ibrahim, A.Y.; Asghar, S.; Khan, I.U.; Iqbal, M.S.; Haseeb, A.; et al. Undefined Nanoemulgel, an Innovative Carrier for Diflunisal Topical Delivery with Profound Anti-Inflammatory Effect: In Vitro and in Vivo Evaluation. Int. J. Nanomed. 2021, 16, 1457. [Google Scholar] [CrossRef]
  101. Williams, A.C.; Barry, B.W. Penetration Enhancers. Adv. Drug Deliv. Rev. 2012, 64, 128–137. [Google Scholar] [CrossRef]
  102. Aggarwal, G.; Dhawan, B.; Harikumar, S. Enhanced Transdermal Permeability of Piroxicam through Novel Nanoemulgel Formulation. Int. J. Pharm. Investig. 2014, 4, 65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Jeengar, M.K.; Sravan Kumar, P.; Thummuri, D.; Shrivastava, S.; Guntuku, L.; Sistla, R.; Naidu, V.G.M. Review on Emu Products for Use as Complementary and Alternative Medicine. Nutrition 2015, 31, 21–27. [Google Scholar] [CrossRef]
  104. Tayel, S.A.; El-Nabarawi, M.A.; Tadros, M.I.; Abd-Elsalam, W.H. Positively Charged Polymeric Nanoparticle Reservoirs of Terbinafine Hydrochloride: Preclinical Implications for Controlled Drug Delivery in the Aqueous Humor of Rabbits. AAPS PharmSciTech 2013, 14, 782–793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Silva, H.D.; Cerqueira, M.A.; Vicente, A.A. Influence of Surfactant and Processing Conditions in the Stability of Oil-in-Water Nanoemulsions. J. Food Eng. 2015, 167, 89–98. [Google Scholar] [CrossRef] [Green Version]
  106. Rajpoot, K.; Tekade, R.K. Microemulsion as Drug and Gene Delivery Vehicle: An inside Story. In Drug Delivery Systems; Academic Press: Cambridge, MA, USA, 2019; pp. 455–520. [Google Scholar] [CrossRef]
  107. Rousseau, D.; Rafanan, R.R.; Yada, R. Microemulsions as Nanoscale Delivery Systems. Compr. Biotechnol. Second Ed. 2011, 4, 675–682. [Google Scholar] [CrossRef]
  108. Khachane, P.V.; Jain, A.S.; Dhawan, V.V.; Joshi, G.V.; Date, A.A.; Mulherkar, R.; Nagarsenker, M.S. Cationic Nanoemulsions as Potential Carriers for Intracellular Delivery. Saudi Pharm. J. 2015, 23, 188–194. [Google Scholar] [CrossRef] [Green Version]
  109. Shakeel, F.; Haq, N.; Alanazi, F.K.; Alsarra, I.A. Impact of Various Nonionic Surfactants on Self-Nanoemulsification Efficiency of Two Grades of Capryol (Capryol-90 and Capryol-PGMC). J. Mol. Liq. 2013, 182, 57–63. [Google Scholar] [CrossRef]
  110. Mantzaridis, C.; Mountrichas, G.; Pispas, S. Complexes between High Charge Density Cationic Polyelectrolytes and Anionic Single- and Double-Tail Surfactants. J. Phys. Chem. B 2009, 113, 7064–7070. [Google Scholar] [CrossRef]
  111. Zakharova, L.Y.; Pashirova, T.N.; Fernandes, A.R.; Doktorovova, S.; Martins-Gomes, C.; Silva, A.M.; Souto, E.B. Self-Assembled Quaternary Ammonium Surfactants for Pharmaceuticals and Biotechnology. In Organic Materials as Smart Nanocarriers for Drug Delivery; William Andrew Publishing: Norwich, NY, USA, 2018; pp. 601–618. [Google Scholar] [CrossRef]
  112. Bali, V.; Ali, M.; Ali, J. Study of Surfactant Combinations and Development of a Novel Nanoemulsion for Minimising Variations in Bioavailability of Ezetimibe. Colloids Surf. B. Biointerfaces 2010, 76, 410–420. [Google Scholar] [CrossRef]
  113. Hu, J.; Chen, D.; Jiang, R.; Tan, Q.; Zhu, B.; Zhang, J. Improved Absorption and in Vivo Kinetic Characteristics of Nanoemulsions Containing Evodiamine–Phospholipid Nanocomplex. Int. J. Nanomed. 2014, 9, 4411–4420. [Google Scholar] [CrossRef] [Green Version]
  114. Poré, J. Emulsions, Micro-Émulsions, Émulsions Multiples; Editions Techniques des Industries des Corps Gras: Neuilly sur Seine, France, 1992; ISBN 9782950724106. [Google Scholar]
  115. Wang, Z.; Mu, H.-J.; Zhang, X.-M.; Ma, P.-K.; Lian, S.-N.; Zhang, F.-P.; Chu, S.-Y.; Zhang, W.-W.; Wang, A.-P.; Wang, W.-Y.; et al. Lower Irritation Microemulsion-Based Rotigotine Gel: Formulation Optimization and in Vitro and in Vivo Studies. Int. J. Nanomed. 2015, 10, 633–644. [Google Scholar]
  116. Syed, H.K.; Peh, K.K. Identification of Phases of Various Oil, Surfactant/ Co-Surfactants and Water System By Ternary Phase Diagram. Acta Pol. Pharm. 2014, 71, 301–309. [Google Scholar] [PubMed]
  117. Shah, H.; Jain, A.; Laghate, G.; Prabhudesai, D. Pharmaceutical Excipients. In Remington; Academic Press: Cambridge, MA, USA, 2021; pp. 633–643. [Google Scholar] [CrossRef]
  118. Ojha, B.; Jain, V.K.; Gupta, S.; Talegaonkar, S.; Jain, K. Nanoemulgel: A Promising Novel Formulation for Treatment of Skin Ailments. Polym. Bull. 2021, 79, 1–25. [Google Scholar] [CrossRef]
  119. Dubey, S.K.; Alexander, A.; Sivaram, M.; Agrawal, M.; Singhvi, G.; Sharma, S.; Dayaramani, R. Uncovering the Diversification of Tissue Engineering on the Emergent Areas of Stem Cells, Nanotechnology and Biomaterials. Curr. Stem Cell Res. Ther. 2020, 15, 187–201. [Google Scholar] [CrossRef]
  120. Ajazuddin; Alexander, A.; Khichariya, A.; Gupta, S.; Patel, R.J.; Giri, T.K.; Tripathi, D.K. Recent Expansions in an Emergent Novel Drug Delivery Technology: Emulgel. J. Control. Release 2013, 171, 122–132. [Google Scholar] [CrossRef]
  121. Deshmukh, K.; Basheer Ahamed, M.; Deshmukh, R.R.; Khadheer Pasha, S.K.; Bhagat, P.R.; Chidambaram, K. Biopolymer Composites with High Dielectric Performance: Interface Engineering. In Biopolymer Composites in Electronics; Elsevier: Amsterdam, The Netherlands, 2017; pp. 27–128. [Google Scholar] [CrossRef]
  122. Vlaia, L.; Coneac, G.; Olariu, I.; Vlaia, V.; Lupuleasa, D. Cellulose-Derivatives-Based Hydrogels as Vehicles for Dermal and Transdermal Drug Delivery. In Emerging Concepts in Analysis and Applications of Hydrogels; IntechOpen: London, UK, 2016. [Google Scholar] [CrossRef] [Green Version]
  123. Hashemnejad, S.M.; Badruddoza, A.Z.M.; Zarket, B.; Ricardo Castaneda, C.; Doyle, P.S. Thermoresponsive Nanoemulsion-Based Gel Synthesized through a Low-Energy Process. Nat. Commun. 2019, 10, 2749. [Google Scholar] [CrossRef] [Green Version]
  124. Perale, G.; Veglianese, P.; Rossi, F.; Peviani, M.; Santoro, M.; Llupi, D.; Micotti, E.; Forloni, G.; Masi, M. In Situ Agar–Carbomer Hydrogel Polycondensation: A Chemical Approach to Regenerative Medicine. Mater. Lett. 2011, 65, 1688–1692. [Google Scholar] [CrossRef]
  125. Braun, S. Encapsulation of Cells (Cellular Delivery) Using Sol–Gel Systems. Compr. Biomater. 2011, 4, 529–543. [Google Scholar] [CrossRef]
  126. Daood, N.M.; Jassim, Z.E.; Ghareeb, M.M.; Zeki, H. Studying the Effect of Different Gelling Agent on The Preparation and Characterization of Metronidazole as Topical Emulgel. Asian J. Pharm. Clin. Res. 2019, 12, 571–577. [Google Scholar] [CrossRef]
  127. Kathe, K.; Kathpalia, H. Film Forming Systems for Topical and Transdermal Drug Delivery. Asian J. Pharm. Sci. 2017, 12, 487–497. [Google Scholar] [CrossRef] [PubMed]
  128. Rapalli, V.K.; Mahmood, A.; Waghule, T.; Gorantla, S.; Kumar Dubey, S.; Alexander, A.; Singhvi, G. Revisiting techniques to evaluate drug permeation through skin. Expert Opin. Drug Deliv. 2021, 18, 1829–1842. [Google Scholar] [CrossRef] [PubMed]
  129. Ibrahim, M.M.; Shehata, T.M. The Enhancement of Transdermal Permeability of Water Soluble Drug by Niosome-Emulgel Combination. J. Drug Deliv. Sci. Technol. 2012, 22, 353–359. [Google Scholar] [CrossRef]
  130. Dixit, A.S.; Charyulu, N.; Nayari, H. Design and Evaluation of Novel Emulgel Containing Acyclovir for Herpes Simplex Keratitis. Lat. Am. J. Pharm. 2011, 30, 844–852. [Google Scholar]
  131. Salem, H.F.; Kharshoum, R.M.; Abou-Taleb, H.A.; Naguib, D.M. Nanosized Nasal Emulgel of Resveratrol: Preparation, Optimization, in Vitro Evaluation and in Vivo Pharmacokinetic Study. Drug Dev. Ind. Pharm. 2019, 45, 1624–1634. [Google Scholar] [CrossRef] [PubMed]
  132. De Souza Ferreira, S.B.; Bruschi, M.L. Investigation of the Physicochemical Stability of Emulgels Composed of Poloxamer 407 and Different Oil Phases Using the Quality by Design Approach. J. Mol. Liq. 2021, 332, 115856. [Google Scholar] [CrossRef]
  133. Shahin, M.; Abdel Hady, S.; Hammad, M.; Mortada, N. Novel Jojoba Oil-Based Emulsion Gel Formulations for Clotrimazole Delivery. AAPS PharmSciTech 2011, 12, 239–247. [Google Scholar] [CrossRef] [PubMed]
  134. El-Setouhy, D.A.; Ahmed El-Ashmony, S.M. Ketorolac Trometamol Topical Formulations: Release Behaviour, Physical Characterization, Skin Permeation, Efficacy and Gastric Safety. J. Pharm. Pharmacol. 2010, 62, 25–34. [Google Scholar] [CrossRef]
  135. Anton, N.; Vandamme, T.F. The Universality of Low-Energy Nano-Emulsification. Int. J. Pharm. 2009, 377, 142–147. [Google Scholar] [CrossRef]
  136. Sharma, V.; Nayak, S.K.; Paul, S.R.; Choudhary, B.; Ray, S.S.; Pal, K. Emulgels. In Polymeric Gels; Woodhead Publishing: Sawston, UK, 2018; pp. 251–264. [Google Scholar] [CrossRef]
  137. Lupi, F.R.; Gabriele, D.; Seta, L.; Baldino, N.; de Cindio, B.; Marino, R. Rheological Investigation of Pectin-Based Emulsion Gels for Pharmaceutical and Cosmetic Uses. Rheol. Acta 2015, 54, 41–52. [Google Scholar] [CrossRef]
  138. Dong, L.; Liu, C.; Cun, D.; Fang, L. The Effect of Rheological Behavior and Microstructure of the Emulgels on the Release and Permeation Profiles of Terpinen-4-Ol. Eur. J. Pharm. Sci. 2015, 78, 140–150. [Google Scholar] [CrossRef]
  139. Solè, I.; Pey, C.M.; Maestro, A.; González, C.; Porras, M.; Solans, C.; Gutiérrez, J.M. Nano-Emulsions Prepared by the Phase Inversion Composition Method: Preparation Variables and Scale Up. J. Colloid Interface Sci. 2010, 344, 417–423. [Google Scholar] [CrossRef] [PubMed]
  140. Lovelyn, C.; Attama, A.A.; Lovelyn, C.; Attama, A.A. Current State of Nanoemulsions in Drug Delivery. J. Biomater. Nanobiotechnol. 2011, 2, 626–639. [Google Scholar] [CrossRef] [Green Version]
  141. Van der Schaaf, U.S.; Nanoemulsions, H.K. Fabrication of Nanoemulsions by Rotor-Stator Emulsification. In Nanoemulsions; Academic Press: Cambridge, MA, USA, 2018; pp. 141–174. [Google Scholar] [CrossRef]
  142. Kotta, S.; Khan, A.W.; Ansari, S.H.; Sharma, R.K.; Ali, J. Formulation of Nanoemulsion: A Comparison between Phase Inversion Composition Method and High-Pressure Homogenization Method. Drug Deliv. 2015, 22, 455–466. [Google Scholar] [CrossRef] [PubMed]
  143. Juttulapa, M.; Piriyaprasarth, S.; Takeuchi, H.; Sriamornsak, P. Effect of High-Pressure Homogenization on Stability of Emulsions Containing Zein and Pectin. Asian J. Pharm. Sci. 2017, 12, 21–27. [Google Scholar] [CrossRef]
  144. Bei, D.; Meng, J.; Youan, B.B.C. Engineering Nanomedicines for Improved Melanoma Therapy: Progress and Promises. Nanomedicine 2010, 5, 1385–1399. [Google Scholar] [CrossRef] [Green Version]
  145. Gorantla, S.; Singhvi, G.; Rapalli, V.K.; Waghule, T.; Dubey, S.K.; Saha, R.N. Targeted Drug-Delivery Systems in the Treatment of Rheumatoid Arthritis: Recent Advancement and Clinical Status. Ther. Deliv. 2020, 11, 269–284. [Google Scholar] [CrossRef]
  146. Prathyusha, E.; Prabakaran, A.; Ahmed, H.; Dethe, M.R.; Agrawal, M.; Gangipangi, V.; Sudhagar, S.; Krishna, K.V.; Dubey, S.K.; Pemmaraju, D.B.; et al. Investigation of ROS Generating Capacity of Curcumin-Loaded Liposomes and Its in Vitro Cytotoxicity on MCF-7 Cell Lines Using Photodynamic Therapy. Photodiagnosis Photodyn. Ther. 2022, 40, 103091. [Google Scholar] [CrossRef]
  147. Mulia, K.; Ramadhan, R.M.A.; Krisanti, E.A. Formulation and Characterization of Nanoemulgel Mangosteen Extract in Virgin Coconut Oil for Topical Formulation. MATEC Web Conf. 2018, 156, 01013. [Google Scholar] [CrossRef] [Green Version]
  148. Chellapa, P.; Eid, A.M.; Elmarzugi, N.A. Preparation and Characterization of Virgin Coconut Oil Nanoemulgel. J. Chem. Pharm. Res. 2015, 7, 787–793. Available online: https://www.jocpr.com (accessed on 1 December 2022).
  149. Bhattacharya, S.; Prajapati, B.G. Formulation and Optimization of Celecoxib Nanoemulgel. Asian J. Pharm. Clin. Res. 2017, 10, 353–365. [Google Scholar] [CrossRef] [Green Version]
  150. Chin, L.Y.; Tan, J.Y.P.; Choudhury, H.; Pandey, M.; Sisinthy, S.P.; Gorain, B. Development and Optimization of Chitosan Coated Nanoemulgel of Telmisartan for Intranasal Delivery: A Comparative Study. J. Drug Deliv. Sci. Technol. 2021, 62, 102341. [Google Scholar] [CrossRef]
  151. Khullar, R.; Kumar, D.; Seth, N.; Saini, S. Formulation and Evaluation of Mefenamic Acid Emulgel for Topical Delivery. Saudi Pharm. J. 2012, 20, 63–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Clogston, J.D.; Patri, A.K. Zeta Potential Measurement. Methods Mol. Biol. 2011, 697, 63–70. [Google Scholar] [CrossRef] [PubMed]
  153. Krishna, K.V.; Saha, R.N.; Dubey, S.K. Biophysical, Biochemical, and Behavioral Implications of ApoE3 Conjugated Donepezil Nanomedicine in a Aβ1-42Induced Alzheimer’s Disease Rat Model. ACS Chem. Neurosci. 2020, 11, 4139–4151. [Google Scholar] [CrossRef]
  154. Khosa, A.; Krishna, K.V.; Saha, R.N.; Dubey, S.K.; Reddi, S. A Simplified and Sensitive Validated RP-HPLC Method for Determination of Temozolomide in Rat Plasma and Its Application to a Pharmacokinetic Study. J. Liq. Chromatogr. Relat. Technol. 2018, 41, 692–697. [Google Scholar] [CrossRef]
  155. Manus Maguire, C.; Rösslein, M.; Wick, P.; Prina-Mello, A. Characterisation of Particles in Solution–a Perspective on Light Scattering and Comparative Technologies. Taylor Fr. 2018, 19, 732–745. [Google Scholar] [CrossRef] [Green Version]
  156. Sneha, K.; Kumar, A. Nanoemulsions: Techniques for the Preparation and the Recent Advances in Their Food Applications. Innov. Food Sci. Emerg. Technol. 2022, 76, 102914. [Google Scholar] [CrossRef]
  157. Khosa, A.; Krishna, K.V.; Dubey, S.K.; Saha, R.N. Lipid Nanocarriers for Enhanced Delivery of Temozolomide to the Brain. Methods Mol. Biol. 2020, 2059, 285–298. [Google Scholar] [CrossRef]
  158. Garg, A.; Aggarwal, D.; Garg, S.; America, A.S.-T.N. Spreading of Semisolid Formulations: An Update. Pharm. Technol. N. Am. 2002, 26, 84. [Google Scholar]
  159. Nikumbh, K.V.; Sevankar, S.G.; Patil, M.P. Formulation Development, in Vitro and in Vivo Evaluation of Microemulsion-Based Gel Loaded with Ketoprofen. Drug Deliv. 2015, 22, 509–515. [Google Scholar] [CrossRef] [PubMed]
  160. Shah, V.P.; Simona Miron, D.; Ștefan Rădulescu, F.; Cardot, J.M.; Maibach, H.I. In Vitro Release Test (IVRT): Principles and Applications. Int. J. Pharm. 2022, 626, 122159. [Google Scholar] [CrossRef]
  161. Sheshala, R.; Anuar, N.K.; Abu Samah, N.H.; Wong, T.W. In Vitro Drug Dissolution/Permeation Testing of Nanocarriers for Skin Application: A Comprehensive Review. AAPS PharmSciTech 2019, 20, 164. [Google Scholar] [CrossRef] [PubMed]
  162. Kanfer, I.; Rath, S.; Purazi, P.; Mudyahoto, N.A. In Vitro Release Testing of Semi-Solid Dosage Forms. Dissolut. Technol. 2017, 24, 52–60. [Google Scholar] [CrossRef]
  163. Shaikh, R.; Raj Singh, T.; Garland, M.; Woolfson, A.; Donnelly, R. Mucoadhesive Drug Delivery Systems. J. Pharm. Bioallied Sci. 2011, 3, 89–100. [Google Scholar] [CrossRef] [PubMed]
  164. Amorós-Galicia, L.; Nardi-Ricart, A.; Verdugo-González, C.; Arroyo-García, C.M.; García-Montoya, E.; Pérez-Lozano, P.; Suñé-Negre, J.M.; Suñé-Pou, M. Development of a Standardized Method for Measuring Bioadhesion and Mucoadhesion That Is Applicable to Various Pharmaceutical Dosage Forms. Pharmaceutics 2022, 14, 1995. [Google Scholar] [CrossRef]
  165. Yuan, C.; Xu, Z.Z.; Fan, M.; Liu, H.; Xie, Y.; Zhu, T. Study on Characteristics and Harm of Surfactants. J. Chem. Pharm. Res. 2014, 6, 2233–2237. [Google Scholar]
  166. Lewis, M.A. Chronic Toxicities of Surfactants and Detergent Builders to Algae: A Review and Risk Assessment. Ecotoxicol. Environ. Saf. 1990, 20, 123–140. [Google Scholar] [CrossRef]
  167. James-Smith, M.A.; Hellner, B.; Annunziato, N.; Mitragotri, S. Effect of Surfactant Mixtures on Skin Structure and Barrier Properties. Ann. Biomed. Eng. 2011, 39, 1215–1223. [Google Scholar] [CrossRef] [Green Version]
  168. Azeem, A.; Ahmad, F.J.; Khar, R.K.; Talegaonkar, S. Nanocarrier for the Transdermal Delivery of an Antiparkinsonian Drug. AAPS PharmSciTech 2009, 10, 1093–1103. [Google Scholar] [CrossRef]
  169. Patel, B.B.; Patel, J.K.; Chakraborty, S.; Shukla, D. Revealing Facts behind Spray Dried Solid Dispersion Technology Used for Solubility Enhancement. Saudi Pharm. J. 2015, 23, 352–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Wang, W.; Hui, P.C.L.; Kan, C.W. Functionalized Textile Based Therapy for the Treatment of Atopic Dermatitis. Coatings 2017, 7, 82. [Google Scholar] [CrossRef] [Green Version]
  171. Gutiérrez, J.M.; González, C.; Maestro, A.; Solè, I.; Pey, C.M.; Nolla, J. Nano-Emulsions: New Applications and Optimization of Their Preparation. Curr. Opin. Colloid Interface Sci. 2008, 13, 245–251. [Google Scholar] [CrossRef]
  172. Sallam, A.A.N.; Younes, H.M. Transdermal Non-Aqueous Nanoemulgels for Systemic Delivery of Aromatase Inhibitor. EP3727363A1, 28 October 2020. [Google Scholar]
  173. Kaufman, R.C. Methods of Treating Inflammatory Disorders and Global Inflammation with Compositions Comprising Phospholipid Nanoparticle Encapsulations of NSAIDS 2018. CA2970917C, 17 September 2019. [Google Scholar]
  174. Xiaoling, F.; Xiao, W.; Xianyi, S. Tripterygium Glycoside-Containing Micro-Emulsified Gel Transdermal Preparation and Preparation Method Thereof. CN107303263B, 31 July 2020. [Google Scholar]
  175. Sengupta, S.; Chawrai, S.R.; Ghosh, S.; Ghosh, S.; Jain, N.; Sadhasivam, S.; Buchta, R.; Bhattacharyya, A. Besifloxacin for the Treatment of Resistant Acne. EP3099301B1, 18 December 2019. [Google Scholar]
  176. Bhalerao, H.; Koteshwara, K.; Chandran, S. Design, Optimisation and Evaluation of in Situ Gelling Nanoemulsion Formulations of Brinzolamide. Drug Deliv. Transl. Res. 2020, 10, 529–547. [Google Scholar] [CrossRef] [PubMed]
  177. Suresh, S.; Rathod, S.; Devasani, R. Minoxidil and Castor Oil Nanoemulgel for Alopecia. WO2020121329A1, 16 June 2020. [Google Scholar]
  178. Somvico, R.R.P.O.C.S.J.M.R.C.P. Nanoemulgel Based on Ucúuba Fat (Virola Surinamensis) for Transungual Administration of Antimicotics. BR102019014044A2, 13 October 2021. [Google Scholar]
Figure 1. Strategies to improve solubility and bioavailability of lipophilic drugs.
Figure 1. Strategies to improve solubility and bioavailability of lipophilic drugs.
Pharmaceutics 15 00164 g001
Figure 2. Skin morphology.
Figure 2. Skin morphology.
Pharmaceutics 15 00164 g002
Figure 3. Schematic representation for the preparation of nano-emulgel by (A) adding Oil (oil + drug) phase to aqueous (water + gelling agent) phase (B) adding nano-emulsion to aqueous (water + gelling agent) phase.
Figure 3. Schematic representation for the preparation of nano-emulgel by (A) adding Oil (oil + drug) phase to aqueous (water + gelling agent) phase (B) adding nano-emulsion to aqueous (water + gelling agent) phase.
Pharmaceutics 15 00164 g003
Figure 4. Graphical representation of entry of nano-emulgel into skin [144]. Adapted from Nanomedicine, 3 December 2010; 5(9): 1385–1399. Copyright (2010) Future Science Group.
Figure 4. Graphical representation of entry of nano-emulgel into skin [144]. Adapted from Nanomedicine, 3 December 2010; 5(9): 1385–1399. Copyright (2010) Future Science Group.
Pharmaceutics 15 00164 g004
Table 1. Primary requirement of active moiety for topical delivery.
Table 1. Primary requirement of active moiety for topical delivery.
PropertiesConditions
t1/2≤10 h
Molecular mass≤500 Daltons
The limit can be exceeded by altering the permeability of skin
Molecular sizeSmall
PolarityNon-polar is desirable
Log P0.8–5
pKaHigher
Irritation on skinNon-irritating
Skin Permeability coefficient≥0.5 × 10–3 cm/h
Table 2. Comparison between conventional emulgel and nano-emulgel.
Table 2. Comparison between conventional emulgel and nano-emulgel.
ParameterConventional EmulgelNano-Emulgel
Thermodynamic stabilityNot stable because of natural tendence of coalescence leading to sedimentation or creaming [61]Stable–because of their smaller particle size, Brownian motion provides enough stability against gravity, preventing sedimentation or creaming [54]
Particle sizeGreater than >500 nm [18]Less than 100 nm [62]
BioavailabilityComparatively less bioavailable than Nano-emulgel [63]Enhanced bioavailability, attributed to small size and large surface area [64]
PermeationComparatively lower permeation [65] High permeation owing to its lower particle size [54,65]
PreparationRequire high energy techniques [66]It can be prepared either by using high or low energy techniques [20]
Systemic absorptionVery minimalHigher compared to conventional emulgel due to the small particle size and large surface area [54]
Ability to cross BBBCannot cross BBB [67]Can Cross BBB because of its small particle size [68]
Table 3. Examples of marketed emulgels for topical application.
Table 3. Examples of marketed emulgels for topical application.
Marketed ProductActive Pharmaceutical IngredientManufacturing Company
Voltaren EmulgelDiclofenac diethylamineGlaxoSmithKline
Isofen EmulgelIbuprofenBeit Jala Pharmaceutical Co.
Benzolait EmulgelBenzoyl peroxide & BiguanideRoydermal
Miconaz-H EmulgelMiconazole nitrate & HydrocartisoneMedical Union Pharmaceuticals
Derma FeetUreaHerbitas
Adwiflam EmulgelDiclofenac diethylamine, Methyl Salicylate & MentholSaja Pharmaceuticals
Nucoxia EmulgelEtoricoxibZydus Cadila Healthcare LTD
Table 4. Pre-clinical Studies on the nano-emulgel dosage form.
Table 4. Pre-clinical Studies on the nano-emulgel dosage form.
Active IngredientCompositionIn Vivo ModelRoute of AdministrationTherapeutic OutcomeReference
CurcuminOil: Labrofac PG + transcutol HP
Surfactant mixture: Tween 20 + solutol HS15
Gelling agent: Carbopol 934
BALB/c miceTopicalPsoriatic mice treated with the curcumin nano-emulgel showed faster and earlier healing than those treated with curcumin plus betamethasone-17-valerate gel[69]
ThymoquinoneOil: Black seed oil
Surfactant mixture: Kolliphor EL + transcutol HP
Gelling agent: Carbopol 940
Wistar ratTopicalNano-emulgel administration of thymoquinone improves its therapeutic efficiency in wound healing studies in Wistar rats[70]
Curcumin and ResveratrolOil: Labrofac PG
Surfactant mixture: Tween 80
Gelling agent: Carbopol
Wistar ratTopicalCurcumin and resveratrol nano-emulgel technology revealed drastically increased curcumin and resveratrol deposition in skin layers. The in-vivo investigation revealed that the NEG formulation resulted in improved burn healing, with histological findings comparable to standard control skin. Thymoquinone nano-emulgel delivery method improves thymoquinone therapeutic effectiveness in wound healing studies in Wistar rats.[71]
BrucineOil: Myrrh oil
Surfactant mixture: Tween 80 + PEG 400
Gelling agent: Carboxymethylcellulose sodium
BALB/c mice and Wistar ratsTopicalBrucine-loaded nanoemulgel has shown improved anti-inflammatory and anti-nociceptive efficacy.[72]
CurcuminOil: Labrofac PG
Surfactant mixture: Tween 80 + PEG 400
Gelling agent: Carbopol 940
Albino ratsTopicalCurcumin nanoemulgel improved the wound-healing efficacy of curcumin compared to the conventional gel formulation.[69]
Raloxifene hydrochlorideOil: Peceol
Surfactant mixture: Tween 20 + transcutol HP
Gelling agent: Chitosan
Wistar ratsTopicalRaloxifene hydrochloride (RH) loaded nanoemulgel formulation for enhanced bioavailability and anti-anti-osteoporotic efficacy of RH. The bioavailability improved by 26-fold compared oral marketed product.[73]
EprinomectinOil: Castor oil
Surfactant mixture: Tween 80 + Labrasol
Gelling agent: Carbomer 940-1
Wistar ratsTopicalNaoemulgel formulation showed improved skin permeability of 1.45-fold compared to emulgel and had no skin-irritating property[74]
AmisulprideOil: Maisine CC
Surfactant mixture: Labrosol + transcutol HP
Gelling agent: Poloxamer 407, Gellan gum
Wistar ratsIntranasalImproved pharmacokinetic profile. The Cmax of API in brain after administering through in-situ nano-emulgel improved by 3.39-fold compared to intravenous administration of nano-emulsion.[75]
DisulfiramOil: Ethyl oleate
Surfactant mixture: Tween 80 + transcutol HP
Gelling agent: Deacetylated gellan gum
Sprague Dawley ratsIntranasalImproved survival rate of rats and reduced tumor progression (Glioblastoma). The survival time of in-situ nano-emulgel treated group is 1.6 times higher than control group[76]
Table 5. Clinical studies on emulgel dosage form.
Table 5. Clinical studies on emulgel dosage form.
Identifier NoActive ConstituentTitile of the StudyConditionsReferance
NCT05536193Metformin and salicylic acidTopical Metformin Emulgel VS Salicylic Acid Peeling in Treatment of Acne VulgarisAcne Vulgaris[77]
NCT03074162Diclofenac sodium & CapsaicinComparison of the Bioavailability of Diclofenac in a Combination Product (Diclofenac 2% + Capsaicin 0.075% Topical Gel) With Two Diclofenac Only Products, Diclofenac Mono Gel 2% and Voltarol® 12 Hour Emulgel 2.32% Gel, in Healthy VolunteersInflammatory[78]
NCT04579991Visnadin, ethyl ximeninate, coleus barbatusEffects of Visnadin, Ethyl Ximeninate, Coleus Barbatus and Millet in Emulgel on Sexual Function in Postmenopausal WomenFemale Sexual FunctionVulvovaginal AtrophyPostmenopausal Atrophic Vaginitis[79]
NCT04110860VoriconazoleClinical Assessment of Voriconazole Self Nano Emulsifying Drug Delivery System Intermediate GelTinea Versicolor[80]
NCT04110834ItraconazoleClinical Assessment of Itraconazole Self Nano Emulsifying Drug Delivery System Intermediate GelTinea Versicolor[81]
NCT03492541Propylene glycol-based eye dropsEvaluation of the Clinical Efficacy and Tolerability of SYSTANE Complete in Adult Patients With Dry Eye Disease Following Topical Ocular Use for 4 Weeks: A Multicenter TrialDry eye disease[82]
NCT05641246Carbamide
diclofenac
Effect of Topical Diclofenac on Clinical Outcome in Breast Cancer Patients Treated With Capecitabine: A Randomized Controlled Trial.Hand and Foot Syndrome[83]
Table 6. Details of commonly used excipients in nano-emulgel formulations.
Table 6. Details of commonly used excipients in nano-emulgel formulations.
S.NoDisease/
Disorder
Active Pharmaceutical IngredientCompositionReferences
OilSurfactantCo-SurfactantGelling Agent
1Anti-inflammatoryCurcuminEmu oilCremophor RH40Labrafil M2125CSCarbopol[85]
2Anti-inflammatoryDiclofenac sodiumIsopropyl myristateTween 20Labrafil M2125CSCarbopol 980[86]
3Anti-inflammatoryMeloxicamAlmond and peppermint oil (1:2)Tween 80EthanolCarbopol 940[87]
4Antimicrobial and Anti-
Inflammatory
QuercetinCinnamon oilTween 80CarbitolPoloxamer[88]
5AntifungalItraconazoleEugenolLabrasolTranscutolP, LecithinCarbolpol[89]
6AntifungalFluconazoleCapmul MCMTween 80Transcutol PCarbopol 934[90]
7Anti-hyperglycemicGlibenclamideLabrafac: Triacetin (1:1)Tween 80Diethylene glycol monoethyl
ether
Carbopol 934[91]
8AntihypertensiveCarvedilolOleic acid: IPM (3:1)Tween 20CarbitolCarbopol-934[92]
9Immunosuppressive agentCyclosporineOleic acidTween 80Transcutol PGuar gum.[93]
10Anti-cancerChrysinCapryol 90Tween 80Transcutol HPPluronic F127[94]
11Wound HealingAtorvastatin
Calcium
Liquid ParaffinTween 80Propylene glycolSodium carboxymethyl cellulose[95]
12Anti-inflammatoryCurcuminMyrrh OilTween 80EthanolSodium carboxymethyl cellulose[96]
13Wound HealingCurcuminLabrofac PGTween 80Propylene glycol 400Carbopol 940[69]
14Anti-fungalTerbinafine HClPeceol oilTween 80PropanolCarbopol 940[97]
15Anti-fungalEbselenCaptexKolliphor ELPDimethylacetamideSoluphus (10% w/v) & HPMC K4M (2.5% w/v)[98]
Table 7. Various gelling agents and their pharmaceutical adaptability for use in topical emulgel.
Table 7. Various gelling agents and their pharmaceutical adaptability for use in topical emulgel.
Gelling AgentConcentration Range (%w/w)Pharmaceutical AdaptabilityReference
HPMC2–6%
  • Forms neutral gels
  • Can provide good stability
  • Resists microbial growth
[126,127]
Carbomer (Carbopol)
Grades–ETD 2020, 171, 910, 934, 934P, 940, 1342 NF, 1971P
0.1–1.5%
  • Forms high viscous gel
  • Forms gel at very low concentration
  • Provides controlled releasep
  • H dependent gelling
[126,128]
NaCMC3–6%
  • It withstands autoclaving. Therefore, can be used in sterile gels
  • Stable between pH 2 to 10
[129,130]
Poloxamer
Grades–124, 182, 188, 407
20–30%
  • Possess better solubility in cold water
  • Thermoreverisble gelation–gel at room temperature and liquid at refrigerated conditions
[131,132]
Combination of HPMC & Carbopol1.2%
  • Combination can improve stability of emulsion compared to individual components
[133,134]
Table 8. Skin irritation grading scale and their clinical implications.
Table 8. Skin irritation grading scale and their clinical implications.
Clinical PortrayalGrade
No erythema0
Slight erythema that is barely perceptible1
Moderate erythema that is visible2
Erythema and papules3
Severe Edema4
Erythema, edema, and papules5
Vesicular eruption6
Strong reaction spreading beyond the application sight7
Table 9. Recent patents on nano-emulgel.
Table 9. Recent patents on nano-emulgel.
Patent NumberAPITitleDisease IndicationCurrent Assignee/InventorsGranted/Publication YearReference
US11185504B2Aromatase inhibitorsTransdermal non-aqueous nanoemulgels for systemic delivery of aromatase inhibitorbreast cancerQatar University2021[172]
CA3050535CAnti-inflammatory nutraceuticals e.g., resveratrol, cinnamaldehyde, green tea polyphenols, lipoic acid etc.Methods of treating inflammatory disorders and global inflammation with compositions comprising phospholipid nanoparticle encapsulations of anti-inflammatory nutraceuticalsInflammatory DisordersNanosphere Health Sciences Inc2021[173]
CN107303263BTripterygium glycosidesTripterygium glycosides nanoemulsion gel and preparation method thereofImmune diseases e.g., clinical rheumatoid arthritis and psoriasis etc.Second Military Medical University SMMU2020[174]
EP3099301B1BesifloxacinBesifloxacin for the treatment of resistant acneAcne vulgarisVyome Therapeutics Ltd.2019[175]
WO2020240451A1BrinzolamideIn-situ gelling nanoemulsion of brinzolamideglaucomaHemant Hanumant BHALERAO, Sajeev Chandran2020[176]
WO2020121329A1Minoxidil and castor oilMinoxidil and castor oil nanoemulgel for alopeciaandrogenic alopeciaSudha suresh Dr. Rathodsoniya ramesh devasani2020[177]
BR102019014044A2KetoconazoleNanoemulgel based on ucúuba fat (Virola surinamensis) for transungual administration of antimicoticsOnychomycosisRayanne Rocha Pereira et al.2021[178]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Donthi, M.R.; Munnangi, S.R.; Krishna, K.V.; Saha, R.N.; Singhvi, G.; Dubey, S.K. Nanoemulgel: A Novel Nano Carrier as a Tool for Topical Drug Delivery. Pharmaceutics 2023, 15, 164. https://doi.org/10.3390/pharmaceutics15010164

AMA Style

Donthi MR, Munnangi SR, Krishna KV, Saha RN, Singhvi G, Dubey SK. Nanoemulgel: A Novel Nano Carrier as a Tool for Topical Drug Delivery. Pharmaceutics. 2023; 15(1):164. https://doi.org/10.3390/pharmaceutics15010164

Chicago/Turabian Style

Donthi, Mahipal Reddy, Siva Ram Munnangi, Kowthavarapu Venkata Krishna, Ranendra Narayan Saha, Gautam Singhvi, and Sunil Kumar Dubey. 2023. "Nanoemulgel: A Novel Nano Carrier as a Tool for Topical Drug Delivery" Pharmaceutics 15, no. 1: 164. https://doi.org/10.3390/pharmaceutics15010164

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

Article Metrics

Back to TopTop