Nanoemulgel A Novel Nano Carrier As A Tool For Top
Nanoemulgel A Novel Nano Carrier As A Tool For Top
1 Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus,
Rajasthan 333031, India
2 Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi,
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 amalga-
mated preparation of different systems aims to deal with these limitations. The novel system pre-
pared 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.
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 epi-
dermis 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 permea-
tion. One such mechanism involves disruption of skin layer structure, which can be
achieved using techniques such as chemical penetration enhancers, ultrasound, iontopho-
resis, sonophoresis, electroporation and microneedles [11]. In contrary, the use of nanocar-
riers 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 in-
cludes but not limited to emulsions (nano/micro), micelles, dendrimers, liposomes, solid
lipid nanoparticles and nano-structured lipid carriers [12–14]. Among these, nano-emul-
sions are found to be a potential drug delivery system because of their high drug-loading
capacities, solubilizing capacities, ease of manufacturability, stability and controlled re-
lease 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
Pharmaceutics 2023, 15, 164 3 of 28
thermodynamic stability leading to longer shelf life compared to simple emulsions, mi-
celles 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 spread-
ability 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.
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].
Properties Conditions
t1/2 ≤10 h
≤500 Daltons
Molecular mass
The limit can be exceeded by altering the permeability of skin
Molecular size Small
Polarity Non-polar is desirable
Log P 0.8–5
pKa Higher
Irritation on skin Non-irritating
Skin Permeability coefficient ≥0.5 × 10–3 cm/h
number of in-vivo studies have been carried out demonstrating the applications and fea-
sibility 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 trans-
lucent or transparent appearance. The thermodynamic stability of nano-emulsions is
greater than other lipid carriers. Nano-emulsions exhibit an increased solubilization ca-
pacity as compared to solutions of simple micelles [29,44]. These formulations can solu-
bilize and incorporate large amounts of active drug substances due to the increased sur-
face area because of the nano-size of oil droplets [45]. The phenomena of creaming or sed-
imentation 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 [47–49]. The enhanced permeation is because of the
ability of nano-emulsion to overcome the firmly bonded lipid bi-layers, thus able to pen-
etrate 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].
topical nanoemulgel loaded with TPGS containing mefenamic acid. In the pharmacody-
namic investigation, the optimized nanoemulgel inhibited inflammation and enhanced
percent reaction time with improved analgesic efficacy. The formulated nanoemulgel out-
performed other traditional topical formulations in terms of long-term stability and drug
penetration [60].
Active Route of
Composition In Vivo Model Therapeutic Outcome Reference
Ingredient Administration
Oil: Labrofac PG + Psoriatic mice treated with the
Curcumin BALB/c mice Topical [69]
transcutol HP curcumin nano-emulgel showed
Pharmaceutics 2023, 15, 164 7 of 28
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 poly-
mers that can swell on the absorption of a liquid. The various components in the nano-
Pharmaceutics 2023, 15, 164 9 of 28
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.
Active Composition
Disease/
S.No Pharmaceutica References
Disorder Oil Surfactant Co-Surfactant Gelling Agent
l Ingredient
Cremophor Labrafil
1 Anti-inflammatory Curcumin Emu oil Carbopol [85]
RH40 M2125CS
Diclofenac Isopropyl Labrafil
2 Anti-inflammatory Tween 20 Carbopol 980 [86]
sodium myristate M2125CS
Almond and
3 Anti-inflammatory Meloxicam peppermint oil Tween 80 Ethanol Carbopol 940 [87]
(1:2)
Antimicrobial and
4 Anti- Quercetin Cinnamon oil Tween 80 Carbitol Poloxamer [88]
Inflammatory
TranscutolP,
5 Antifungal Itraconazole Eugenol Labrasol Carbolpol [89]
Lecithin
6 Antifungal Fluconazole Capmul MCM Tween 80 Transcutol P Carbopol 934 [90]
Diethylene
Labrafac: glycol
7 Anti-hyperglycemic Glibenclamide Tween 80 Carbopol 934 [91]
Triacetin (1:1) monoethyl
ether
Oleic acid: IPM
8 Antihypertensive Carvedilol Tween 20 Carbitol Carbopol-934 [92]
(3:1)
Immunosuppressiv
9 Cyclosporine Oleic acid Tween 80 Transcutol P Guar gum. [93]
e agent
10 Anti-cancer Chrysin Capryol 90 Tween 80 Transcutol HP Pluronic F127 [94]
Sodium
Atorvastatin Propylene
11 Wound Healing Liquid Paraffin Tween 80 carboxymethyl [95]
Calcium glycol
cellulose
Sodium
12 Anti-inflammatory Curcumin Myrrh Oil Tween 80 Ethanol carboxymethyl [96]
cellulose
Propylene
13 Wound Healing Curcumin Labrofac PG Tween 80 Carbopol 940 [69]
glycol 400
Terbinafine
14 Anti-fungal Peceol oil Tween 80 Propanol Carbopol 940 [97]
HCl
Soluphus
Dimethylaceta (10% w/v) &
15 Anti-fungal Ebselen Captex Kolliphor ELP [98]
mide HPMC K4M
(2.5% w/v)
The consistency of the lipids may vary from liquid to high molecular solids. The hydro-
phobicity of an oil plays a crucial role in forming a stable emulsion, wherein poor hydro-
phobicity of the oil is shown to increase the emulsification, concurrently affecting the sol-
ubility 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 ac-
tion 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 contrib-
ute 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 keto-
profen [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 en-
hanced 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 mois-
turizes the skin and has high amounts of unsaturated fatty acids like oleic acid, thus im-
proving 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-inflamma-
tory 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 de-
velopment 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 hy-
drolyzed to form an appropriate oil, which upon combining with a suitable surfactant
enhances the solubility of hydrophobic compounds for nano-emulgel formulation [104].
of zwitterion surfactants [111]. Toxicity should be considered while selecting the surfac-
tant 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 biocom-
patibility. 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.
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.
Table 7. Various gelling agents and their pharmaceutical adaptability for use in topical emulgel.
Concentration
Gelling Agent Pharmaceutical Adaptability Reference
Range (%w/w)
• Forms neutral gels
HPMC 2–6% • Can provide good stability [126,127]
• Resists microbial growth
• Forms high viscous gel
Carbomer (Carbopol)
• Forms gel at very low concentration
Grades–ETD 2020, 171, 910, 934, 0.1–1.5% [126,128]
• Provides controlled release
934P, 940, 1342 NF, 1971P
• pH dependent gelling
• It withstands autoclaving. Therefore, can be used
NaCMC 3–6% in sterile gels [129,130]
• Stable between pH 2 to 10
• Possess better solubility in cold water
Poloxamer
20–30% • Thermoreverisble gelation–gel at room tempera- [131,132]
Grades–124, 182, 188, 407
ture and liquid at refrigerated conditions
Combination of HPMC & • Combination can improve stability of emulsion
1.2% [133,134]
Carbopol compared to individual components
6. Preparation of Nano-Emulgel
Nano-emulgel is a non-equilibrium formulation of structured liquids requiring en-
ergy, 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 de-
creasing 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 solu-
bilized 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].
Pharmaceutics 2023, 15, 164 13 of 28
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.
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 tech-
niques of low energy approach. These methods provide the required thermodynamic sta-
bility 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
Pharmaceutics 2023, 15, 164 14 of 28
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 com-
prises a colloid mill, that has a complex geometry, and the droplets of an emulsion gener-
ated 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 chal-
lenging. However, a droplet size of less than a micron can be achieved using high-pres-
sure homogenization and sonication techniques, which in turn helps extend the shelf life
of emulsions by lowering the creaming rate. For this reason, homogenization and soni-
cation 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 concentra-
tion of an emulsifier is necessary to maintain control over the re-coalescence of the emul-
sion. 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 concentra-
tions 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 improp-
erly covered with a surfactant, resulting in re-coalescence. With the optimum concentra-
tion 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 ob-
tained 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 (sur-
factant), 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 concen-
tration of surfactant decreased with 10 min of ultrasonication, the globular size increased.
Meanwhile, increasing the concentration of surfactant with 10 min of sonication time re-
sulted in a smaller globular size. The authors concluded that sonication is more effective
when the appropriate concentration is used [70]. Monitoring the process control parame-
ters and taking into account the composition of the excipients is both necessary steps in
the process of optimizing the formulation.
Pharmaceutics 2023, 15, 164 15 of 28
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 constit-
uents 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 path-
ways 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].
Figure 4. Graphical representation of entry of nano-emulgel into skin [144]. Adapted from Nano-
medicine, 3 December 2010; 5(9): 1385–1399. Copyright (2010) Future Science Group.
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 up-
per slide, Length of the slide, and Time taken to detach the slides.
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 var-
iations 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 asso-
ciated 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 pro-
duce 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 re-
duction 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
Pharmaceutics 2023, 15, 164 20 of 28
Current Granted
Disease
Patent Number API Title Assignee/Invent /Publication Reference
Indication
ors Year
Transdermal non-
Aromatase aqueous nanoemulgels
US11185504B2 breast cancer Qatar University 2021 [172]
inhibitors
for systemic delivery of
aromatase inhibitor
Methods of treating
Anti-
inflammatory disorders
inflammatory
and global inflammation
nutraceuticals
with compositions Nanosphere
e.g., resveratrol, Inflammatory
CA3050535C comprising phospholipid Health Sciences 2021 [173]
cinnamaldehyde Disorders
nanoparticle Inc
, green tea
encapsulations of anti-
polyphenols,
inflammatory
lipoic acid etc.
nutraceuticals
Immune diseases
Tripterygium glycosides Second Military
e.g., clinical
Tripterygium nanoemulsion gel and Medical
CN107303263B rheumatoid 2020 [174]
glycosides preparation method University
arthritis and
thereof SMMU
psoriasis etc.
Besifloxacin for the
Vyome
EP3099301B1 Besifloxacin treatment of resistant Acne vulgaris 2019 [175]
Therapeutics Ltd.
acne
Pharmaceutics 2023, 15, 164 21 of 28
Hemant
In-situ gelling
WO2020240451 Hanumant
Brinzolamide nanoemulsion of glaucoma 2020 [176]
A1 BHALERAO,
brinzolamide
Sajeev Chandran
Sudha suresh Dr.
WO2020121329 Minoxidil and Minoxidil and castor oil androgenic
Rathodsoniya 2020 [177]
A1 castor oil nanoemulgel for alopecia alopecia
ramesh devasani
Nanoemulgel based on
ucúuba fat (Virola
BR102019014044 surinamensis) for Rayanne Rocha
Ketoconazole Onychomycosis 2021 [178]
A2 transungual Pereira et al.
administration of
antimicotics
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 de-
livering lipophilic moieties mainly due to improved permeation, and better pharmacoki-
netics, which subsequently improves the pharmacological effect. Patient compliance is
also elevated due to its non-greasy and improved spreading properties on topical admin-
istration. 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.; Writ-
ing, 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 interests 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, doi:10.1016/J.APSB.2015.07.003.
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.
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 Can-
cer Cells. J. Pharm. Investig. 2021, 51, 701–714.
4. Alekya, T.; Narendar, D.; Mahipal, D.; Arjun, N.; Nagaraj, B. Design and Evaluation of Chronomodulated Drug Delivery of
Tramadol Hydrochloride. Drug Res. (Stuttg). 2018, 68, 174–180.
5. Homayun, B.; Lin, X.; Choi, H.J. Challenges and Recent Progress in Oral Drug Delivery Systems for Biopharmaceuticals. Phar-
maceutics 2019, 11, 129, doi:10.3390/PHARMACEUTICS11030129.
6. Donthi, M.R.; Dudhipala, N.R.; Komalla, D.R.; Suram, D.; Banala, N. Preparation and Evaluation of Fixed Combination of Ke-
toprofen Enteric Coated and Famotidine Floating Mini Tablets by Single Unit Encapsulation System. J. Bioequiv. Availab. 2015,
07, 1–5, doi:10.4172/jbb.1000254.
Pharmaceutics 2023, 15, 164 22 of 28
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, doi:10.1016/J.EJMECH.2018.09.027.
8. Rajitha, R.; Narendar, D.; Arjun, N.; Nagaraj, B. Colon Delivery of Naproxen: Preparation, Characterization and Clinical Eval-
uation in Healthy Volunteers. Int. J. Pharm. Sci. Nanotechnol. 2016, 9, 3383–3389.
9. Garg, N.K.; Singh, B.; Tyagi, R.K.; Sharma, G.; Katare, O.P. Effective Transdermal Delivery of Methotrexate through Nanostruc-
tured Lipid Carriers in an Experimentally Induced Arthritis Model. Colloids Surf. B. Biointerfaces 2016, 147, 17–24,
doi:10.1016/J.COLSURFB.2016.07.046.
10. Szumała, P.; Macierzanka, A. Topical Delivery of Pharmaceutical and Cosmetic Macromolecules Using Microemulsion Systems.
Int. J. Pharm. 2022, 615, 121488.
11. Gupta, R.; Dwadasi, B.S.; Rai, B.; Mitragotri, S. Effect of Chemical Permeation Enhancers on Skin Permeability: In Silico Screen-
ing Using Molecular Dynamics Simulations. Sci. Rep. 2019, 9, 1–11.
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.
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.
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, doi:10.1021/ACSCHEMNEURO.1C00022/ASSET/IMAGES/MEDIUM/CN1C00022_M001.GIF.
15. Nafisi, S.; Maibach, H.I. Nanotechnology in Cosmetics. Cosmet. Sci. Technol. Theor. Princ. Appl. 2017, 337–369, doi:10.1016/B978-
0-12-802005-0.00022-7.
16. Tiwari, N.; Osorio-Blanco, E.R.; Sonzogni, A.; Esporr\’\in-Ubieto, D.; Wang, H.; Calderon, M. Nanocarriers for Skin Applica-
tions: Where Do We Stand? Angew. Chemie Int. Ed. 2022, 61, e202107960.
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, doi:10.1016/J.BIOPHA.2018.10.021.
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 De-
livery. Pharmaceutics 2017, 9, 37, doi:10.3390/PHARMACEUTICS9040037.
19. Aithal, G.C.; Narayan, R.; Nayak, U.Y. Nanoemulgel: A Promising Phase in Drug Delivery. Curr. Pharm. Des. 2020, 26, 279–291,
doi:10.2174/1381612826666191226100241.
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, doi:10.2174/1574891X14666190717111531.
21. Murthy, S.N. Approaches for Delivery of Drugs Topically. AAPS PharmSciTech 2019 211 2019, 21, 1–2, doi:10.1208/S12249-019-
1582-X.
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, doi:10.3109/1061186X.2012.716845.
23. Bhowmik, D.; Gopinath, H.; Kumar, B.P.; S.Duraivel; Kumar, K.P.S. Recent Advances In Novel Topical Drug Delivery System.
Pharma Innov. J. 2012, 1, 12–31.
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,
doi:10.1016/J.IJPHARM.2009.12.050.
25. Bhaskar, K.; Anbu, J.; Ravichandiran, V.; Venkateswarlu, V.; Rao, Y.M. Lipid Nanoparticles for Transdermal Delivery of Flurbi-
profen: Formulation, in Vitro, Ex Vivo and in Vivo Studies. Lipids Health Dis. 2009, 8, 1–15, doi:10.1186/1476-511X-8-6.
26. Zhou, J.; Zhou, M.; Yang, F.F.; Liu, C.Y.; Pan, R. Le; 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, doi:10.1021/MP5005838.
27. Chang, R.K.; Raw, A.; Lionberger, R.; Yu, L. Generic Development of Topical Dermatologic Products: Formulation Develop-
ment, Process Development, and Testing of Topical Dermatologic Products. AAPS J. 2013, 15, 41–52, doi:10.1208/S12248-012-
9411-0.
28. Cevc, G. Lipid Vesicles and Other Colloids as Drug Carriers on the Skin. Adv. Drug Deliv. Rev. 2004, 56, 675–711,
doi:10.1016/J.ADDR.2003.10.028.
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.
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,
doi:10.3109/10837450.2014.915571.
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, doi:10.2174/138161208784007653.
Pharmaceutics 2023, 15, 164 23 of 28
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, doi:10.1155/2014/406731.
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, 12, e115952, doi:10.1371/JOUR-
NAL.PONE.0115952.
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, doi:10.1166/ASEM.2016.1859.
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, doi:10.3109/10837450.2011.627871.
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, doi:10.1034/J.1600-0625.2000.009003165.X.
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.
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, doi:10.1208/S12249-008-9178-X.
39. Gao, F.; Zhang, Z.; Bu, H.; Huang, Y.; Gao, Z.; Shen, J.; Zhao, C.; Li, Y. Nanoemulsion Improves the Oral Absorption of Can-
desartan Cilexetil in Rats: Performance and Mechanism. J. Control. Release 2011, 149, 168–174, doi:10.1016/J.JCON-
REL.2010.10.013.
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, doi:10.1080/10717540802328599.
41. Akhter, S.; Jain, G.; Ahmad, F.; Khar, R.; Jain, N.; Khan, Z.; Talegaonkar, S. Investigation of Nanoemulsion System for Trans-
dermal Delivery of Domperidone: Ex-Vivo and in Vivo Studies. Curr. Nanosci. 2008, 4, 381–390, doi:10.2174/157341308786306071.
42. El Maghraby, G.M. Transdermal Delivery of Hydrocortisone from Eucalyptus Oil Microemulsion: Effects of Cosurfactants. Int.
J. Pharm. 2008, 355, 285–292, doi:10.1016/J.IJPHARM.2007.12.022.
43. Huang, Y. Bin; 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, doi:10.1016/J.IJPHARM.2007.07.022.
44. Eid, A.M.; El-Enshasy, H.A.; Aziz, R.; Elmarzugi, N.A. Preparation, Characterization and Anti-Inflammatory Activity of
Swietenia Macrophylla Nanoemulgel. 2014, 5, 1–10, doi:10.4172/2157-7439.1000190.
45. Algahtani, M.S.; Ahmad, M.Z.; Ahmad, J. Nanoemulgel for Improved Topical Delivery of Retinyl Palmitate: Formulation De-
sign and Stability Evaluation. Nanomater. (Basel, Switzerland) 2020, 10, doi:10.3390/NANO10050848.
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. Nanobiotechnology 2011, 9, 44,
doi:10.1186/1477-3155-9-44.
47. Bolzinger, M.A.; Briançon, S.; Pelletier, J.; Fessi, H.; Chevalier, Y. Percutaneous Release of Caffeine from Microemulsion, Emul-
sion and Gel Dosage Forms. Eur. J. Pharm. Biopharm. 2008, 68, 446–451, doi:10.1016/J.EJPB.2007.10.018.
48. Fini, A.; Bergamante, V.; Ceschel, G.C.; Ronchi, C.; Moraes, C.A.F. Control of Transdermal Permeation of Hydrocortisone Ace-
tate from Hydrophilic and Lipophilic Formulations. AAPS PharmSciTech 2008, 9, 762, doi:10.1208/S12249-008-9107-Z.
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, doi:10.1016/J.EJPB.2007.04.006.
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, doi:10.1016/J.LFS.2013.01.005.
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, doi:10.1016/J.IJPHARM.2007.11.051.
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,
doi:10.1016/J.IJPHARM.2015.04.015.
53. Dev, A.; Chodankar, R.; Shelke, O. Emulgels: A Novel Topical Drug Delivery System. undefined 2015.
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, doi:10.1016/J.IJPHARM.2017.04.068.
55. Arora, R.; Aggarwal, G.; Harikumar, S.L.; Kaur, K. Nanoemulsion Based Hydrogel for Enhanced Transdermal Delivery of Ke-
toprofen. Adv. Pharm. 2014, 2014, 1–12, doi:10.1155/2014/468456.
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,
doi:10.1016/J.YRTPH.2016.10.020.
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, doi:10.1007/S10571-019-00653-Z.
Pharmaceutics 2023, 15, 164 24 of 28
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, doi:10.1016/J.COLSURFB.2006.05.005.
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.
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.
61. Tesch, S.; Gerhards, C.; Schubert, H. Stabilization of Emulsions by OSA Starches. J. Food Eng. 2002, 54, 167–174.
62. Ingle, A.P.; Shende, S.; Gupta, I.; Rai, M. Recent Trends in the Development of Nano-Bioactive Compounds and Delivery Sys-
tems. In Biotechnological production of bioactive compounds; Elsevier, 2020; pp. 409–431.
63. Ashara, K.C. Microemulgel An Overwhelming Approach To Improve Therapeutic Action Of Drug Moiety. 2014, 24, 452-457.
64. Sultana, N.; Akhtar, J.; Khan, M.I.; Ahmad, U.; Arif, M.; Ahmad, M.; Upadhyay, T.; Nanoemulgel: For Promising Topical and
Systemic Delivery. 2022.
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.
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.
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.
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.
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, doi:10.3390/GELS7040213/S1.
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.
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,
doi:10.1016/J.JDDST.2021.102360.
72. Abdallah, M.; Lila, A.; Unissa, R.; H.E.-C. and S.B., Preparation, Characterization and Evaluation of Anti-Inflammatory and
Anti-Nociceptive Effects of Brucine-Loaded Nanoemulgel. Colloids and Surfaces B: Biointerfaces, 2021, 205, 111868.
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, doi:10.1016/J.JDDST.2021.102745.
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, doi:10.1080/10717544.2019.1682720.
75. Gadhave, D.; Tupe, S.; Tagalpallewar, A.; B.G.-I.J. Nose-to-Brain Delivery of Amisulpride-Loaded Lipid-Based Poloxamer-Gel-
lan Gum Nanoemulgel: In Vitro and in Vivo Pharmacological Studies. Int. J. of Pharmaceutics, 2021, 607, 121050
76. Qu, Y.; Li, A.; Ma, L.; Iqbal, S.; Sun, X.; Ma, W.; C.L.-I.J. Nose-to-Brain Delivery of Disulfiram Nanoemulsion in Situ Gel Formu-
lation for Glioblastoma Targeting Therapy. Int. J. of Pharmaceutics, 2021, 597, 120250.
77. Topical Metformin Emulgel VS Salicylic Acid Peeling in Treatment of Acne Vulgaris - Full Text View - ClinicalTrials.Gov Avail-
able online: https://clinicaltrials.gov/ct2/show/NCT05536193.
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.
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.
80. Clinical Assessment of Voriconazole Self Nano Emulsifying Drug Delivery System Intermediate Gel - Full Text View - Clinical-
Trials.Gov Available online: https://www.clinicaltrials.gov/ct2/show/NCT04110860.
81. Clinical Assessment of Itraconazole Self Nano Emulsifying Drug Delivery System Intermediate Gel - Full Text View - Clinical-
Trials.Gov Available online: https://www.clinicaltrials.gov/ct2/show/NCT04110834.
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.
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, doi:10.1186/S13063-022-06353-2.
84. Eswaraiah S., S.K. Emulgel: Review on Novel Approach to Topical Drug Delivery Available online: https://asianjpr.com/Ab-
stractView.aspx?PID=2014-4-1-2.
Pharmaceutics 2023, 15, 164 25 of 28
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, doi:10.1016/J.IJPHARM.2016.04.052.
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 Develop-
ment—in Vitro and in Vivo Studies. J. Chem. 2020, 2020, doi:10.1155/2020/4071818.
87. Drais, H.K.; Hussein, A.A. Formulation Characterization and Evaluation of Meloxicam Nanoemulgel to Be Used Topically. Iraqi
J. Pharm. Sci. 2017, 26.
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,
doi:10.3390/MOLECULES23061363.
89. Wankar, J.; Ajimera, T. Design, Development and Evaluation of Nanoemulsion and Nanogel of Itraconazole for Transdermal
Delivery. 2014.
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, doi:10.3109/03639045.2012.707203.
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. undefined 2013.
92. Pratap, S.B.; Brajesh, K.; S.K, J.; Kausar, S. Development and Characterization of A Nanoemulsion Gel for Transdermal Delivery
of Carvedilol. Int. J. Drug Dev. Res. 4, 151–161.
93. Begur, M.; Pai, V.; Gowda, D. V.; AtulSrivastava; Raghundan, H. V.; Shinde, C.G.; Manusri, N. Enhanced Permeability of Cy-
closporine from a Transdermally Applied Nanoemulgel. undefined 2015.
94. Nagaraja, S.; Basavarajappa, G.M.; Attimarad, M.; Pund, S. Topical Nanoemulgel for the Treatment of Skin Cancer: Proof-of-
Technology. Pharm. 2021, Vol. 13, Page 902 2021, 13, 902, doi:10.3390/PHARMACEUTICS13060902.
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. Pharm. 2019, Vol. 11, Page 609 2019, 11, 609,
doi:10.3390/PHARMACEUTICS11110609.
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. Polym. 2021, Vol. 13, Page 577 2021, 13, 577,
doi:10.3390/POLYM13040577.
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. Futur. J. Pharm. Sci. 2018, 4, 18–28,
doi:10.1016/J.FJPS.2017.07.003.
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, doi:10.1016/J.EJPS.2020.105323.
99. Vandamme, T.F. Microemulsions as Ocular Drug Delivery Systems: Recent Developments and Future Challenges. Prog. Retin.
Eye Res. 2002, 21, 15–34, doi:10.1016/S1350-9462(01)00017-9.
100. Bashir, M.; Ahmad, J.; Asif, M.; … S.K.-I.J.; 2021, undefined Nanoemulgel, an Innovative Carrier for Diflunisal Topical Delivery
with Profound Anti-Inflammatory Effect: In Vitro and in Vivo Evaluation. ncbi.nlm.nih.gov.
101. Williams, A.C.; Barry, B.W. Penetration Enhancers. Adv. Drug Deliv. Rev. 2012, 64, 128–137, doi:10.1016/J.ADDR.2012.09.032.
102. Aggarwal, G.; Dhawan, B.; Harikumar, S. Enhanced Transdermal Permeability of Piroxicam through Novel Nanoemulgel For-
mulation. Int. J. Pharm. Investig. 2014, 4, 65, doi:10.4103/2230-973X.133053.
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, doi:10.1016/J.NUT.2014.04.004.
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, doi:10.1208/S12249-013-9964-Y.
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, doi:10.1016/J.JFOODENG.2015.07.037.
106. Rajpoot, K.; Tekade, R.K. Microemulsion as Drug and Gene Delivery Vehicle: An inside Story. Drug Deliv. Syst. 2019, 455–520,
doi:10.1016/B978-0-12-814487-9.00010-7.
107. Rousseau, D.; Rafanan, R.R.; Yada, R. Microemulsions as Nanoscale Delivery Systems. Compr. Biotechnol. Second Ed. 2011, 4, 675–
682, doi:10.1016/B978-0-08-088504-9.00304-4.
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, doi:10.1016/J.JSPS.2014.07.007.
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, doi:10.1016/J.MOLLIQ.2013.03.011.
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, doi:10.1021/JP8095874.
Pharmaceutics 2023, 15, 164 26 of 28
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. Org. Mater. as Smart Nanocarriers Drug Deliv. 2018,
601–618, doi:10.1016/B978-0-12-813663-8.00014-2.
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, doi:10.1016/J.COLSURFB.2009.11.021.
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. Nanomedicine 2014, 9, 4411–4420, doi:10.2147/IJN.S59812.
114. Poré, J. Emulsions, Micro-Émulsions, Émulsions Multiples; Editions Techniques des Industries des Corps Gras: Neuilly sur Seine,
1992; ISBN 9782950724106.
115. Zheng Wang1; Hong-Jie Mu1; Xue-Mei Zhang1; Peng-Kai Ma1; Sheng-Nan Lian1; Feng-Pu Zhang1; Sheng-Ying Chu1; Wen-
Wen Zhang1; Ai-Ping Wang1, 2; Wen-Yan Wang2; et al. Lower Irritation Microemulsion-Based Rotigotine Gel: Formulation
Optimization and in Vitro and in Vivo Studies. Int. J. Nanomedicine 2015, 10, 633–644.
116. Haroon K. Syed and Kok K. Peh. Identification of Phases of Various Oil, Surfactant/ Co-Surfactants and Water System By Ter-
nary Phase Diagram. Acta Pol. Pharm. ñDrug Res. 2014, 71, 301–309.
117. Shah, H.; Jain, A.; Laghate, G.; Prabhudesai, D. Pharmaceutical Excipients. Remington 2021, 633–643, doi:10.1016/B978-0-12-
820007-0.00032-5.
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, 1–25, doi:10.1007/S00289-021-03729-3/TABLES/5.
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, doi:10.2174/1574888X15666200103124821.
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, doi:10.1016/J.JCONREL.2013.06.030.
121. Deshmukh, K.; Basheer Ahamed, M.; Deshmukh, R.R.; Khadheer Pasha, S.K.; Bhagat, P.R.; Chidambaram, K. Biopolymer Com-
posites With High Dielectric Performance: Interface Engineering. Biopolym. Compos. Electron. 2017, 27–128, doi:10.1016/B978-0-
12-809261-3.00003-6.
122. Vlaia, L.; Coneac, G.; Olariu, I.; Vlaia, V.; Lupuleasa, D. Cellulose-Derivatives-Based Hydrogels as Vehicles for Dermal and
Transdermal Drug Delivery. Emerg. Concepts Anal. Appl. Hydrogels 2016, doi:10.5772/63953.
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 101 2019, 10, 1–10, doi:10.1038/s41467-019-10749-1.
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,
doi:10.1016/J.MATLET.2011.02.036.
125. Braun, S. Encapsulation of Cells (Cellular Delivery) Using Sol–Gel Systems. Compr. Biomater. 2011, 4, 529–543, doi:10.1016/B978-
0-08-055294-1.00141-0.
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, doi:10.22159/AJ-
PCR.2019.V12I3.31504.
127. Kathe, K.; Kathpalia, H. Film Forming Systems for Topical and Transdermal Drug Delivery. Asian J. Pharm. Sci. 2017, 12, 487–
497, doi:10.1016/J.AJPS.2017.07.004.
128. Rapalli VK, Mahmood A, Waghule T, Gorantla S, Kumar Dubey S, Alexander A, Singhvi G. Revisiting techniques to evaluate
drug permeation through skin. Expert Opinion on Drug Delivery. 2021 Dec 2;18(12):1829-42.
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, doi:10.1016/S1773-2247(12)50059-6.
130. Dixit, A.S.; Charyulu, N.; Nayari, H. Design and Evaluation of Novel Emulgel Containing Acyclovir for Herpes Simplex Kera-
titis. Lat. Am. J. Pharm. 2011, 30, 844–852.
131. Salem, H.F.; Kharshoum, R.M.; Abou-Taleb, H.A.; Naguib, D.M. Nanosized Nasal Emulgel of Resveratrol: Preparation, Opti-
mization, in Vitro Evaluation and in Vivo Pharmacokinetic Study. https://doi.org/10.1080/03639045.2019.1648500 2019, 45, 1624–
1634, doi:10.1080/03639045.2019.1648500.
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, doi:10.1016/J.MOL-
LIQ.2021.115856.
133. Shahin, M.; Abdel Hady, S.; Hammad, M.; Mortada, N. Novel Jojoba Oil-Based Emulsion Gel Formulations for Clotrimazole
Delivery. AAPS PharmSciTech 2011 121 2011, 12, 239–247, doi:10.1208/S12249-011-9583-4.
134. El-Setouhy, D.A.; Ahmed El-Ashmony, S.M. Ketorolac Trometamol Topical Formulations: Release Behaviour, Physical Charac-
terization, Skin Permeation, Efficacy and Gastric Safety. J. Pharm. Pharmacol. 2010, 62, 25–34, doi:10.1211/JPP.62.01.0002.
135. Anton, N.; Vandamme, T.F. The Universality of Low-Energy Nano-Emulsification. Int. J. Pharm. 2009, 377, 142–147,
doi:10.1016/J.IJPHARM.2009.05.014.
136. Sharma, V.; Nayak, S.K.; Paul, S.R.; Choudhary, B.; Ray, S.S.; Pal, K. Emulgels. Polym. Gels 2018, 251–264, doi:10.1016/B978-0-
08-102179-8.00009-0.
Pharmaceutics 2023, 15, 164 27 of 28
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, doi:10.1007/S00397-014-0809-8/FIGURES/4.
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, doi:10.1016/J.EJPS.2015.07.003.
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,
doi:10.1016/J.JCIS.2009.11.046.
140. Lovelyn, C.; Attama, A.A.; Lovelyn, C.; Attama, A.A. Current State of Nanoemulsions in Drug Delivery. J. Biomater. Nanobi-
otechnol. 2011, 2, 626–639, doi:10.4236/JBNB.2011.225075.
141. Schaaf, U. van der; Nanoemulsions, H.K. Fabrication of Nanoemulsions by Rotor-Stator Emulsification. Nanoemulsions. Aca-
demic Press, 2018, 141-174.
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. https://doi.org/10.3109/10717544.2013.866992 2015, 22, 455–
466, doi:10.3109/10717544.2013.866992.
143. Juttulapa, M.; Piriyaprasarth, S.; Takeuchi, H.; Sriamornsak, P. Effect of High-Pressure Homogenization on Stability of Emul-
sions Containing Zein and Pectin. Asian J. Pharm. Sci. 2017, 12, 21–27, doi:10.1016/J.AJPS.2016.09.004.
144. Bei, D.; Meng, J.; Youan, B.B.C. Engineering Nanomedicines for Improved Melanoma Therapy: Progress and Promises. Nano-
medicine 2010, 5, 1385–1399, doi:10.2217/NNM.10.117.
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, doi:10.4155/TDE-2020-0029.
146. Prathyusha, E.; A, P.; Ahmed, H.; Dethe, M.R.; Agrawal, M.; Gangipangi, V.; Sudhagar, S.; Krishna, K.V.; Dubey, S.K.; Pemma-
raju, 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,
doi:10.1016/J.PDPDT.2022.103091.
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, doi:10.1051/MATECCONF/201815601013.
148. Chellapa, P.; Eid, A.M.; Elmarzugi, N.A. Preparation and Characterization of Virgin Coconut Oil Nanoemulgel. Available online
www.jocpr.com J. Chem. Pharm. Res. 2015, 7, 787–793.
149. Bhattacharya, S.; Prajapati, B.G. Formulation and Optimization of Celecoxib Nanoemulgel. Asian J. Pharm. Clin. Res. 2017, 10,
353–365, doi:10.22159/AJPCR.2017.V10I8.19510.
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,
doi:10.1016/J.JDDST.2021.102341.
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, doi:10.1016/J.JSPS.2011.08.001.
152. Clogston, J.D.; Patri, A.K. Zeta Potential Measurement. Methods Mol. Biol. 2011, 697, 63–70, doi:10.1007/978-1-60327-198-1_6.
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,
doi:10.1021/acschemneuro.0c00430/asset/images/medium/cn0c00430_m003.gif.
154. Khosa, A.; Krishna, K. V; Saha, R.N.; Dubey, S.K.; Reddi, S. A Simplified and Sensitive Validated RP-HPLC Method for Deter-
mination of Temozolomide in Rat Plasma and Its Application to a Pharmacokinetic Study. J. Liq. Chromatogr. Relat. Technol. 2018,
41, 692–697.
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, doi:10.1080/14686996.2018.1517587.
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, doi:10.1016/J.IFSET.2021.102914.
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, doi:10.1007/978-1-4939-9798-5_15.
158. Garg, A.; Aggarwal, D.; Garg, S.; America, A.S.-T.N.; Spreading of Semisolid Formulations: An Update. Pharmaceutical Tech-
nology North America. 2002, 26, 84-84
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, doi:10.3109/10717544.2013.859186.
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, doi:10.1016/J.IJPHARM.2022.122159.
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, doi:10.1208/S12249-019-1362-7.
162. Kanfer, I.; Rath, S.; Purazi, P.; Technol, N.M.-D.; 2017, undefined In Vitro Release Testing of Semi-Solid Dosage Forms. re-
searchgate.net 2017, 24, 52–60, doi:10.14227/DT240317P52.
163. Shaikh, R.; Raj Singh, T.; Garland, M.; Woolfson, A.; Donnelly, R. Mucoadhesive Drug Delivery Systems. J. Pharm. Bioallied Sci.
2011, 3, 89–100, doi:10.4103/0975-7406.76478.
Pharmaceutics 2023, 15, 164 28 of 28
164. Amorós-Galicia, L.; Nardi-Ricart, A.; Verdugo-González, C.; Arroyo-García, C.M.; García-Montoya, E.; Pérez-Lozano, P.; Ma
Suñé-Negre, J.; Suñé-Pou, M. Development of a Standardized Method for Measuring Bioadhesion and Mucoadhesion That Is
Applicable to Various Pharmaceutical Dosage Forms. mdpi.com, doi:10.3390/pharmaceutics14101995.
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.
166. Lewis, M.A. Chronic Toxicities of Surfactants and Detergent Builders to Algae: A Review and Risk Assessment. Ecotoxicol. En-
viron. Saf. 1990, 20, 123–140, doi:10.1016/0147-6513(90)90052-7.
167. James-Smith, M.A.; Hellner, B.; Annunziato, N.; Mitragotri, S. Effect of Surfactant Mixtures on Skin Structure and Barrier Prop-
erties. Ann. Biomed. Eng. 2011, 39, 1215–1223, doi:10.1007/S10439-010-0190-4.
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, doi:10.1208/S12249-009-9306-2/TABLES/6.
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, doi:10.1016/j.jsps.2013.12.013.
170. Wang, W.; Hui, P.C.L.; Kan, C.W. Functionalized Textile Based Therapy for the Treatment of Atopic Dermatitis. Coatings 2017,
Vol. 7, Page 82 2017, 7, 82, doi:10.3390/COATINGS7060082.
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, doi:10.1016/J.COCIS.2008.01.005.
172. Sallam, A.A.N.; Younes, H.M. Transdermal Non-Aqueous Nanoemulgels for Systemic Delivery of Aromatase Inhibitor.
EP3727363A1, 28 October 2020.
173. Kaufman, R.C. Methods of Treating Inflammatory Disorders and Global Inflammation with Compositions Comprising Phos-
pholipid Nanoparticle Encapsulations of NSAIDS 2018. CA2970917C, 17 September 2019.
174. Xiaoling, F.; Xiao, W.; Xianyi, S. Tripterygium Glycoside-Containing Micro-Emulsified Gel Transdermal Preparation and Prep-
aration Method Thereof. CN107303263B, 31 July 2020.
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.
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, doi:10.1007/S13346-019-00697-0.
177. Suresh, S.; Rathod, S.; Devasani, R. Minoxidil and Castor Oil Nanoemulgel for Alopecia. WO2020121329A1, 16 June 2020
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.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual au-
thor(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.