1 s2.0 S1773224719300863 Main
1 s2.0 S1773224719300863 Main
1 s2.0 S1773224719300863 Main
A R T I C LE I N FO A B S T R A C T
Keywords: The herbal medicines have been popularized throughout the globe since time immemorial. Several plant based
Nanoemulsions therapeutic leads have been produced through integrated approaches. Many herbal bioactives have several
Herbal bioactives health benefits but limited therapeutic potential due to short half-life and low bioavailability profile. Plant
Nano drug delivery system derived molecules are either hydrophilic or lipophilic in nature. Highly hydrophilic bioactives have poor ab-
Nanocarriers
sorption via lipid membrane which slows down their biological efficacy and pharmacokinetics. The large mo-
Bioavailability
lecular size of the plant bioactive compounds also limited their therapeutic uses due to low membrane per-
meability. Nanocarriers have the ability to potentiate the efficacy of herbal bioactives by improving their
solubility, absorption profile, minimizing dose and side effects. The nanoemulsions can channel the herbal
bioactives to the particular target site and maintain blood-plasma concentration for longer periods. However, the
conventional drug delivery system failed to achieve these specific requirements. Nanoemulsions are promising to
improve solubility, stability, permeability and bioavailability of the herbal bioactives through encapsulation.
The present review describes the importance of nanoemulsions for delivery of herbal bioactives and highlights
the various aspects of its formulation consideration, challenges and future perspective for the development of
herbal bioactives.
1. Introduction drug inside the human body. NDDS have the potential to target the
drug/active molecule to particular tissues or sites, also have the ability
Herbal medicines have been used for the treatment of several dis- to get the desired drug concentration over effective treatment period
eases since ancient era. During the last decades, it has become a thrust under therapeutic index [1,2]. Nanosized particles or droplets lead to
area of research for many researchers and innovators throughout the improved solubility and bioavailability of the drug. In this context,
world due to their health benefits. The increasing demand of herbal nanoencapsulation based nanoemulsions are pioneering for en-
medicines as alternative therapies in the Western world needs more capsulating the herbal bioactives and thereby, enhanced solubility and
research in alliance with pharmaceutical nanotechnology for safe and bioavailability. Nanoemulsions are homogeneous, thermodynamically
efficacious medicines. The pharmaceutical application of herbal stable and isotropic system of aqueous, surfactants/co-surfactants (Smix)
bioactives and extracts or phytopharmaceuticals is limited with low and oil phase [3,4]. The nanoemulsion droplet size (o/w and w/o) is
solubility, permeability as well as bioavailability. It has become the reported to be 20–200 nm that is variable based on the composition of
biggest hurdle for the effective use of herbal bioactive compounds the system and the homogenization method used. Nanoemulsion
against various health complications. These drawbacks could be solved structure has been represented in Fig. 1. Upon oral administration,
by the nanoencapsulation process utilizing the nanocarriers which can nanoemulsions dispersed into tiny droplets by self-nanoemulsification
entrap the active moieties inside the lipidic or polymeric core of ex- process in the gastric environment with gastro-intestinal motility [5].
cipients. Therefore, the efficacy of phytopharmaceuticals is based on These nanocarriers can also improve the stability of the en-
effective drug delivery systems [1]. capsulated extract and bioactive of the herbs from gastric degradation
In the recent past, nano drug delivery systems (NDDS) have and results into an extended release of the formulation. They have
emerged as most powerful strategy to deliver bioactive constituent or capability to enhance the membrane diffusion which leads to the
∗
Corresponding author.
E-mail addresses: [email protected], [email protected] (R.K. Harwansh), [email protected] (R. Deshmukh),
[email protected] (M.A. Rahman).
https://doi.org/10.1016/j.jddst.2019.03.006
Received 16 January 2019; Received in revised form 25 February 2019; Accepted 6 March 2019
Available online 11 March 2019
1773-2247/ © 2019 Elsevier B.V. All rights reserved.
R.K. Harwansh, et al. Journal of Drug Delivery Science and Technology 51 (2019) 224–233
Fig. 1. Structure of nanoemulsion [A] oil in water type emulsion (o/w), [B] water in oil type of emulsion (w/o).
sustained effect thereof. For instance, several bioactive and extract of Nanosized droplet offers increased drug absorption and permeation
plants have been reported with nanoemulsion such as neem oil, eu- through the gastrointestinal tract (GIT) due to large interfacial area
calyptus oil, zedoary oil, curcumin, Phaleria macrocarpa, citronella oil, [20].
capsicum oleoresin, berberine, triptolide, etc. [6–14].
The enhanced oral bioavailability and sustained release property of 2.1. Advantage of nanoemulsions as novel nanocarrier
paclitaxel were achieved with nanoemulsions (o/w) as reported by
Choudhury et al. (2014) [15]. In another study, a capsicum oleoresin The advantages offered by nanoemulsions [21] are as follows:
(capsaicin) encapsulated nanoemulsion has been investigated for en-
hancement of its bioavailability. It was obtained due to nanosized • It improves the solubility, dissolution, absorption, bio-membrane
droplets of the formulation [16]. permeation and bioavailability of poorly soluble drugs. This may be
Nanoemulsions have been exploited well for delivery of diverse due to nanosize range and large interfacial area of the droplets.
medicines including pharmaceuticals, phytopharmaceuticals, cosme- • The herbal bioactives can be encapsulated into the nanoemulsion
ceuticals, dietary and nutraceutical for the betterment of human health. matrix so it improves their physical stability.
It can improve the drug's profile like solubility, stability, permeability • The oil or lipids are biocompatible, biodegradable and non-muta-
and bioavailability by encapsulating them into core oil and water of the genic so nanoemulsions are safe for human health.
droplets. It offers a greater advantage for oral administration of the • It offers the site-specific and prolonged release delivery of the
poorly soluble drugs in comparison to conventional delivery systems phytomolecules.
[17]. The specific feature of nanoemulsion as novel carrier system has • Reductions in the dose thereby minimize associated toxicities and
been represented in Fig. 2. provide better therapeutic effects.
The encapsulation of herbal bioactives in novel systems offers dif- • In case of topical application, it avoids first-pass metabolisms hence
ferent advantages in comparison with traditional delivery systems improves plasma drug concentration.
which include stabilization of drug in the aqueous environment of • Offers value added drug delivery system for nutraceutical and
stomach containing food components, protection from gastric de- dietary supplements.
gradation, control the release of active constituents and bioavailability • Facilitating patient compliance.
improvement. Nanosized droplets offer an improved pharmacokinetic
profile of bioactive from herbs. Many authors have shown that reducing 2.2. Formulation aspect of nanoemulsions
the size of the globules leads to an increased surface area which offers
higher drug solubility and absorption profile of the drug. This property Excipients screening are the major steps for the formulation of na-
helps to improve the bio-accessibility and cellular uptake of the active noemulsions. The major excipients for formulation of nanoemulsions
molecules by passive transport through gastrointestinal walls [18]. are oil, Smix and water. Drug solubility study helps to select the best oils
Nanoemulsions are promising for delivery of herbal bioactives in an and surfactants for development of the nanoemulsion formulations. A
effective and efficacious manner. The present review highlights the pseudoternary phase diagram is mainly used for determining the actual
aspects of current status, challenges and future perspective of herbal ratio of water:oil:Smix in the nanoemulsions. The maximum drug solu-
bioactives with implication of nanoemulsions as herbal drug delivery bility, encapsulation efficiency and drug loading in the formulation
systems. depend upon the ratio of ternary system of the formulation [22,23]. The
specific properties of the three components (oil, water and Smix) and
2. Formulation considerations and techniques for nanoemulsion their formulations have been represented in Fig. 3.
225
R.K. Harwansh, et al. Journal of Drug Delivery Science and Technology 51 (2019) 224–233
concentration for longer periods. Hydrolyzed vegetable oils, medium- 2.2.2. Surfactants
chain and modified long triglycerides are broadly used as excipients in Surfactants are interfacial tension lowering agents in oil and water
nanoemulsions [24,25]. interface of an emulsion. It is generally used in lower concentration to
reduce the oil-water surface tension during fabrication of nanoemul-
sion. Surfactants are nonionic, zwitterionic, cationic and anionic. It
Fig. 3. A hypothetical pseudo-ternary phase diagram of oil, water and Smix with emphasis on emulsification process and various formulations.
226
R.K. Harwansh, et al. Journal of Drug Delivery Science and Technology 51 (2019) 224–233
Table 1
Example of oils, surfactants and co-surfactants.
Excipients Description/main fatty acid (FA) composition HLB
Lipids/Oils
Akoline MCM Caprylic/capric glycerides [MAG and DAG of C8 and C10 with small quantities of TAG] 5–6
Beeswax –
Capmul MCM Caprylic/capric glycerides 5–6
Caprylic/capric TAG TAG of C8 and C10 –
Captex 355 Glycerol caprylate caprate [TAG of C8 and C10] –
Corn oil – –
Ethyl oleate Oleic acid ethyl ester 11
Isopropyl myristate FA ester [Isopropyl ester of C14 FA-myristic acid] 11.5
Isostearyl isostearate – 7
Labrafac CCa Caprylic/capric triglyceride [TAG of C8-12 FA] 1
Labrafac™ lipophile WL1349a Medium chain triglycerides 2
Miglyol 812 Medium-chain TAG –
Oleic acid – –
Olive oil - 7
Peanut oil – –
Sesame oil – –
Soyabean oil Soya oil 7
Vegetable oil Long-chain TAG [TAG of C18, C16 and C14 FA] –
Viscoleo Fractionated coconut oil [TAG of C8-12 FA] –
Surfactants
Acconon CC-6 Ethoxylated glycerides 12.5
Acconon MC-8 Caprylocaproyl macrogolglycerides (polyoxyglycerides) and C10:0 and some free PEG 400 14–15
Brij 35 Polyoxyethylene lauryl ether 9.5
Caproyl 90™a Propylene glycol monocaprylate [C8 FA mono-ester of propylene glycol] 6
Caproyl™ PGMCa Propylene glycol caprylate 5
Captex 350a Coconut oil transesterified with C8 and C10 fatty acids –
Cremophor ELa Polyoxy-35-castor oil [Glycerol-PEG ricinoleate, FA ester of PEG] 12–14
Cremophor RH40a Polyoxy-40- hydrogenated castor oil [FA ester of glycerol-PEG, FA ester of PEG, free PEG and ethoxylate glycerol] 14–16
Labrafac™ PGa Propylene glycol dicaprylocaprate 2
Labrafil® M 2125CSa Polyoxyethylated glycerides [Linoleoyl macrogolglycerides] 4
Labrafil® M1944CSa Polyoxyethlated oleic glycerides [Oleoyl macrogolglycerides]; Mainly C18:1 mono- and diester of PEG 300 and MAG, DAG and TAG 4
Labrasola Caprylocaproyl macrogol-8-glycerides [FA C8:0/C10:0 mono and diesters of PEG 400 and MAG, DAG and TAG with mainly C8:0 and 14
C10:0 and some free PEG 400]
Lauroglycol™ 90a Propylene glycol monolaurate [C12 FA mono-ester of propylene glycol] 5
Lauroglycol™ FCCa Propylene glycol laurate 4
Maisine™ 35-1a Glyceryl monolinoleate [MAG and DAG of C18 and C16 FA with small quantities of TAG] 4
Plurol isosteariquea Polyglyceryl-6-Isostearate 10.8
Span 80 Sorbitan monooleate [Sorbitan (Z)-mono-9-octadecenoate] 4.3
TPGS D-alpha Tocopheryl polyethylene glycol 1000 succinate –
Transcutol CGa Diethyleneglycol monoethyl ether 4.2
Transcutol Pa Diethyleneglycol monoethyl ether 4.2
Tween 20 Polysorbate 20 [Polyoxyethylene 20-sorbitan monolaurate]; PEG-sorbitan esterified with C12 FA 16.5
Tween 80 Polysorbate 80 [Polyoxyethylene 20-sorbitan monooleate]; PEG-sorbitan esterified with 80 C18:1 FA 15
Tween 85 Polyxyethylene 20-sorbitan trioleate 11
Co-surfactantsa
Ethanol Ethyl alcohol 7.9
Glycerin – –
Polypylene glycol – –
PEG-400 Polyethylene glycol-400 15.5
Carbitol Diglycol monoethyl ether –
a
Solubilizer, penetration and bioavailability enhancer.
consists of both hydrophilic head and a hydrophobic tail region which ethanol, isopropyl alcohol, ethylene glycol, transcutol P, carbitol,
can act as an emulsifier to produce nanoemulsion. Hydrophilic-lipo- transcutol CG etc. [32].
philic balance (HLB, 3–8 for w/o and 8–18 for o/w) value of the sur-
factants plays an important role in nanoemulsion formulations. 2.2.4. Surfactant and co-surfactant (Smix)
Surfactants (30–60% v/v) are used to produce a stable emulsion. A vide Smix is a mixer of surfactant and co-surfactant which is a de-
category of surfactants has been shown in Table 1 [26,27]. termining factor for producing nanoemulsion. It can work better in a
ratio of oil for selecting the proper region of nanoemulsion in the phase
2.2.3. Co-surfactants diagram. It is generally considered that all of the excipients used in
An alone surfactant cannot lower the surface tension of both oil and nanoemulsion formulation should have generally regarded as safe
water interface efficiently to make the nanoemulsions. Co-surfactants (GRAS) listed components for safety aspect [33–35]. The list of phar-
can work better in combination with a surfactant which is generally maceutically acceptable excipients for the formulation of nanoemulsion
used in development of the nanoemulsions. It can be used in low con- is represented in Table 1.
centration due to severe side effects at higher concentration [28–30].
Mostly applicable surfactants are C3-C8 chain alcohols which increase 2.3. Preparation method of nanoemulsions
the fluidity by reducing the interfacial tension between oil and water
phase [31]. Examples of commonly applicable co-surfactants (Table 1) Nanoemulsions are non-equilibrated formulations, therefore large
are polyethylene glycol 400 (PEG 400), propylene glycol, glycerin, amount of surfactants and energies are required for fabricating suitable
227
R.K. Harwansh, et al. Journal of Drug Delivery Science and Technology 51 (2019) 224–233
nanosize emulsions. Generally, spontaneous nano-emulsfication, high solubilization of water and oil molecules by using non-ionic surfactants
or low-energy techniques are used for producing nanodroplets of the like polyethoxylated type. It is a basis of nanoemulsion formulation.
formulation [36,37]. The schematic of nanoemulsion preparation has Generally, nanoemulsions (o/w) are fabricated by mixing the water,
been shown in Fig. 4. The detailed methods of preparation of nanoe- nonionic surfactants and oil in specific concentration at room
mulsion are described below. temperature. Upon heating o/w type nanodroplets changed to w/o
type nanodroplets due to hydrophobic nature of surfactants. At this
2.3.1. Low-energy methods stage, surfactant monolayer has negative curvature [41]. But the
This technique employs low energy for manufacturing of nanoe- curvature becomes zero at low interfacial tension in the range of
mulsions. It is also known as the condensation process of nanoemulsi- 10−2-10−5 mNm−1 which may be due to intermediate HLB value with
fication which occurred due to phase transitions. It includes phase in- respect to temperature [47,48]. At this stage, the ternary system
version temperature (PIT), phase transition and spontaneous- involves formation of lamellar liquid crystalline and bicontinuous
emulsification process [38]. These processes are primarily reliant on the type emulsions (Fig. 3). This system can produce the nanodroplets
inflection of interfacial properties of components like oil, surfactant and upon rapid cooling at above PIT or normal condition. Dilution of
Smix to give suitable formulation. The intrinsic physical and chemical microemulsion with oil/water results in nanoemulsions (w/o or o/w
characteristics and interfacial behavior of the ternary phase played type) and this phenomenon is known as catastrophic phase inversion
crucial role in nanoemulsion production [39,40]. process [38,42].
PIT involves a quick alteration in the curvature of non-ionic sur-
factants at constant the composition and temperature while emulsion 2.3.1.2. Solvent displacement technique. It is spontaneous
inversion point technique employed the changes in composition of the nanoemulsification technique for producing polymeric nanoparticles
formulations at a constant temperature which is broadly used in in- through nano-precipitation concept. Briefly, firstly oily components are
dustry [41–44]. In this method, the internal capacity (energy) of the dispersed into organic solvents like alcohol, diethyl ether, acetone etc.
formulation is applied for producing the fine droplets, so that low-en- Then, it is incorporated into Smix containing water system with
ergy method is very effective for preparing nanoemulsion. For the na- agitation and mixed properly which results in nanodroplets
noemulsification, HLB value, temperature and ingredients ratio of the formation. In last stage, excess amount of solvent is removed by using
formulation are prime factors [45–48]. The low-energy emulsification rotatory vacuum evaporator [44–46,49].
methods are now gaining attention due to their wide application and
benefits as a nanoemulsion formulation and its stability aspects. The 2.3.1.3. Phase inversion composition (PIC) technique. This technique is
detailed discussions about these techniques have been illustrated un- also called as a self-nanoemulsification process. It can produce nano-
derneath. assemblies without utilization of heat and solvent at an environment of
room temperature. Nanoemulsions are the kinetically stable system at
2.3.1.1. PIT technique. PIT is a temperature dependent technique for nanosize (around 50 nm) range. A specific method of preparation
228
R.K. Harwansh, et al. Journal of Drug Delivery Science and Technology 51 (2019) 224–233
229
R.K. Harwansh, et al. Journal of Drug Delivery Science and Technology 51 (2019) 224–233
Fig. 6. Schematic of skin and major routes of drug penetration, (1) via the sweat ducts (intercellular pathway), (2) directly across the stratum corneum (intracellular
pathway) and (3) through the hair follicles and sebaceous gland (folicullar pathway).
4. Challenges and future perspective of herbal bioactives infection and disease are inappropriate to fulfill the need of the com-
munity so much so minimizing undesired action at the target sites are
Herbal bioactives have been popularized worldwide for their several evenly essential. Drug targeting to a particular site like a tumor can be
health effects but there are still challenges like low half-life, low solu- achieved with NEs which can modulate the absorption, distribution,
bility, low permeability as well as poor bioavailability hence, low metabolism and excretion profile of herbal drugs. Commencement and
biological effect. Most of the active principle compounds (40%) pro- development of suitable herbal bioactive formulations are utmost im-
duced through high-throughput techniques having low solubility. These portant for promotion of human health by using nano drug delivery
are major challenges for development of appropriate herbal products approach [88,89].
for management of the disease. It requires continuous research in the Novel drug delivery systems like NEs can overcome the drawbacks
novel carriers for delivery of herbal bioactives to achieve the effective of conventional drug delivery systems. It can potentially increase the
level of blood concentration in the body [1,54]. therapeutic effect of herbal bioactives. Although, clinical implementa-
The fabrication of nanocarrier aims to increase the absorption and tion of herbal drugs still remain challenges as alternative drug therapies
pharmacokinetics of poorly bioavailable herbal bioactives through size for the treatment of diseases. Interaction of drug loaded nanocarriers
reduction, encapsulation and stability. An effective level of drug de- with biological receptors and tissues results in interfering with the
livery is an important aspect to attain the improved therapeutic efficacy immune response is challengeable for clinical transformation to drug
at target site. In case of herbal bioactives, large molecular size and lipid therapies. Another challenge in regard to formulation and development
solubilization are prime rate limiting parameters for bio-membrane of herbal bioactive based NDDS at industrial scale are the feasibility and
permeation. To cross the membrane with enhanced pharmacokinetic availability of the innovative techniques. Nanomedicine drug targeting
profile are biggest challenges in development of herbal formulations. and probing suffered from biocompatibility and toxicological issues
Oil or lipid based nanocarriers (NEs) may be the solution to these which remain challenges and need to be resolved for the fulfilment of
challenges. They can improve the solubility, permeability, GIT ab- international standards of drug therapies [90].
sorption and minimize the food-stuffs effect on rate of absorption of Nanoemulsions are very effective and versatile nanocarrier system
highly lipophilic molecules, thus enhanced bioavailability [87]. used not only for delivery of allopathic drug but also for plant derived
The biological half-life is another major aspect for determining the medicines that have been proved through various platforms for phar-
pharmacological effect of a drug. Shorter half-life compound can maceutical application. It has greater encapsulation efficiency rather
eliminate rapidly as compared to greater half-life bioactives. Liver than the other delivery systems which could be useful for solubilization
metabolism leads to detoxification and fragmentation of the active of poorly soluble compounds, especially hydrophobic moieties. Future
compounds results in low bioavailability. Higher renal filtration and perspective of nanoemulsion based herbal drug delivery delineates di-
elimination rate also lower the bioavailability of herbal drugs. In this verse kind of application including oral, parenteral, ocular, pulmonary,
context, nanoencapsulation with nanocarriers are promising for fabri- topical, transdermal, vaccine, protein and peptides drug targeting. It
cating and overcoming the limitation of herbal bioactives. Several kinds could be promising for delivery of nutraceuticals for several health
of research on herbal bioactive based nanocarriers are being carried out benefits. Nanoemulsions can produce the prolonged and sustained re-
to get the desired therapeutic efficacy at the same dose or reduced dose lease action to maintain the therapeutic level of the drug.
with minimum adverse effects. So nanocarriers like nanoemulsions are Nanoemulsions based novel herbal formulations will lead to maximum
exploited successfully in the herbal drug delivery to gain the maximum economic growth for cosmetic, dietary and nutraceutical and pharma-
therapeutic response. Thus, NEs based many herbal formulations are ceutical companies [55].
being used with molecular mechanism understanding of disease for
therapeutic purposes. Unfavorable drug effects against the cause of
230
Table 2
R.K. Harwansh, et al.
Encapsulation and delivery of bioactive compounds from herbs through the nanoemulsion techniques.
Herbal bioactives Applications Biological activities Special features Route of administrations References
Oleanolic acid (OA) and Dermal controlled release Anti-inflammatory and anti-carcinogenic Mean droplet size 140 ± 47 nm (OA) and Transdermal and in vivo [74]
ursolic acid (UA) activity 201 ± 21 nm (UA)
Ferulic acid Enhanced skin permeability, sustained release action UVA protection efficacy against oxidative Droplet size 102.3 ± 1.14 nm, ZP Transdermal, in vivo [71]
stress, antioxidant -35.2 ± 0.41 mV, 98.88 ± 0.12 %EE
Catechin Enhanced skin permeability and bioavailability, sustained release Photoprotection against UVA induced %EE 99.02 ± 0.13, droplet size Transdermal and oral [75]
oxidative stress 98.6 ± 1.05 nm, ZP -27.3 ± 0.20 mV
Betulinic acid Enhanced gastro-intestinal permeability, bioavailability and Hepatoprotection and antioxidant Droplet size 50.3 ± 0.56 nm, ZP Oral (in vivo) [56]
sustained release action -10.2 ± 0.11 mV; 99.95 ± 0.12 %EE
β-Elemene Improved solubility and enhanced antitumor activity against Anti-tumor Average particle size 52.68 nm and ZP In vitro and in vivo [76]
Hep3B cancer cells -26.43 mV
Emodin Enhanced transcellular permeation, improved oral bioavailability – Droplet size Oral [77]
116 ± 6.5 nm
Zedoary turmeric oil SNEDDS for enhanced bioavailability and therapeutic efficacy Antibacterial, antioxidant, Droplets size 68.3 ± 1.6 nm, ZP Oral [60]
hepatoprotective and antitumor -41.2 ± 1.3 mV
Colchicine Improved intestinal membrane permeability and bioavailability Eugenol as penetration enhancer, Particle size 41.2 ± 7.2 nm Oral [61]
anticancer
Glycyrrhizin Increased percutaneous permeation, sustained release action Useful in the treatment of dermatitis, Droplet size 22 nm Transdermal [4]
eczema, psoriasis, antibacterial and anti-
inflammatory
Berberine Sustained drug release, improved oral bioavailability Anti-inflammatory, anticancer and anti- Droplet size 19.53 ± 1.58 nm, ZP -9.14 mV Oral [13]
231
angiogenic activity
Capsaicin Enhanced oral bioavailability and stability Analgesic, pain reliever anticancer, Particle size 53.4 ± 0.83 nm, ZP Oral [16]
cardiovascular -42.1 ± 0.52 mV
Curcumin Increased solubility, improved oral bioavailability Anticancer, antioxidant, anti-tumor, Droplet median size 11.2 nm In vitro, in vivo oral [78]
antimicrobial, anti-inflammatory
Epigallocatechin Increased absorption Nutraceutical, systemic antioxidant, 50–100 mg dose Oral [79]
anticancer, hypolipidemics
Paclitaxel Sustained release profile, increased bioavailability Anticancer, targeted for breast cancer Absolute oral bioavailability (55.9%) In vitro, [15]
In vivo
Quercetin Enhanced oral bioavailability, reduced dose, enhanced penetration Antiobesity, Antioxidant, anticancer, anti- Particle size 19.3 ± 0.17 nm, ZP Oral and in vivo, intranasal, [57,58,80–82]
in blood brain barrier, increased antioxidant activity and 74 times inflammatory, anti-ageing, anti-wrinkle 0.34 ± 0.13 mV, %EE > 99%, droplet size in vitro, oral, topical
higher drug release, exerted better therapeutic efficacy, enhanced 10–100 nm
skin
Permeability
Resveratrol Improved bioavailability controlled release, pharmacokinetics Antioxidant, anti-inflammatory, anti- Dose 4 mg/kg, particle size 5 μm, %EE 92.5% Oral, in vivo [18,83]
profile tumor
Silymarin Improved bioavailability Hepatoprotective Droplet size 68.22 ± 0.00314 nm, PDI 0.216, Oral, buccal [59]
%EE 69.22 ± 0.6%
β-Carotene Enhanced bioaccessibility and bioavailability, increased stability, Antioxidant, anticancer, cardiovascular Particle size 140–170 nm, 0.5% dose, 66% In vitro [84,85]
sustained release delivery, effective delivery system for GIT disease bioaccessibility
Pterostilbene Improved solubility and Stability. Sustained drug release Anticancer, antidiabetic, and Mean droplet size 55.8 nm In vitro, Oral delivery [86]
cardiovascular diseases
Where, % EE = Entrapment efficiency, ZP = Zeta potential, PDI = Polydispersity index, GIT = Gastrointestinal tract.
Journal of Drug Delivery Science and Technology 51 (2019) 224–233
R.K. Harwansh, et al. Journal of Drug Delivery Science and Technology 51 (2019) 224–233
5. Conclusion [2] A. Alexander, Ajazuddin, R.J. Patel, S. Saraf, S. Saraf, Recent expansion of phar-
maceutical nanotechnologies and targeting strategies in the field of phytopharma-
ceuticals for the delivery of herbal extracts and bioactives, J. Control. Release 241
Herbal medicines have been popularized as a traditional system of (2016) 110–124.
therapy in healthcare throughout the world. However, the traditional [3] R.K. Harwansh, K.C. Patra, S.K. Pareta, Nanoemulsion as potential vehicles for
herbal drug delivery systems are old and outdated. Herbal bioactives transdermal delivery of pure phytopharmaceuticals and poorly soluble drug, Int. J.
Drug Deliv. 3 (2011) 209–218.
have become a thrust area of research for exploiting their several health [4] R.K. Harwansh, K.C. Patra, S.K. Pareta, J. Singh, M.A. Rahman, Nanoemulsions as
benefits through nanotechnology. Nanoemulsions have been recognized vehicles for transdermal delivery of glycyrrhizin, Braz. J. Pharm. Sci. 47 (2011)
as novel drug delivery systems for administering and targeting the 769–778.
[5] C.J.H. Porter, C.W. Pouton, J.F. Cuine, W.N. Charman, Enhancing intestinal drug
herbal bioactives and extracts. Although, a lot of research is conducting solubilization using lipid-based delivery systems, Adv. Drug Deliv. Rev. 60 (2008)
in this area and it is still in the exploratory phase. Several phyto- 673–691.
pharmaceuticals exhibited improved pharmacological effect at reduced [6] J. Jerobin, P. Makwana, R.S. Suresh Kumar, R. Sundaramoorthy, A. Mukherjee,
N. Chandrasekaran, Antibacterial activity of neem nanoemulsion and its toxicity
or similar dose after encapsulating into nanoemulsions in comparison
assessment on human lymphocytes in vitro, Int. J. Nanomed. 10 (2015) 77–86.
with traditional herb extracts or crude herbs. Thus, NEs have great [7] C.H. Anjali, Y. Sharma, A. Mukherjee, N. Chandrasekaran, Neem oil (Azadirachta
potential for development of a novel formulation of herbal drugs which indica) nanoemulsion-a potent larvicidal agent against Culex quinquefasciatus, Pest
results in an economical and effective delivery of a drug. The devel- Manag. Sci. 68 (2012) 158–163.
[8] S. Sugumar, S.K. Clarke, M.J. Nirmala, B.K. Tyagi, A. Mukherjee,
opment of suitable nanocarriers like nanoemulsions for herbal drugs is N. Chandrasekaran, Nanoemulsion of eucalyptus oil and its larvicidal activity
pioneered for enhancing or controlling the required therapeutic level against Culex quinquefasciatus, Bull. Entomol. Res. 104 (2014) 393–402.
and bioavailability. Nanoemulsions have many notable merits for in- [9] M. Pant, S. Dubey, P.K. Patanjali, S.N. Naik, S. Sharma, Insecticidal activity of
eucalyptus oil nanoemulsion with karanja and jatropha aqueous filtrates, Int.
creasing the solubility and in vivo permeability of low bioavailable Biodeterior. Biodegrad. 91 (2014) 119–127.
phytopharmaceuticals. In the current scenario, NEs are attaining in- [10] H. Rachmawati, M.A. Novel, R.M. Nisa, G. Berlian, O.M. Tandrasasmita, A. Rahma,
creased attention as novel nanocarriers for delivery of herbal bioac- C. Riani, R.R. Tjandrawinata, Co-delivery of curcumin-loaded nanoemulsion and
Phaleria macrocarpa extract to NIH 3T3 cell for antifibrosis, J. Drug Deliv. Sci.
tives. The simplicity of nanoemulsion fabrication also attracted interest Technol. 39 (2017) 123–130.
towards increased loading, improved therapeutic efficacy and stability [11] E. Osman Mohamed Ali, N.A. Shakil, V.S. Rana, D.J. Sarkar, S. Majumder,
of herbal drugs as compared to conventional delivery systems. It has P. Kaushik, B.B. Singh, J. Kumar, Antifungal activity of nano emulsions of neem and
citronella oils against phytopathogenic fungi, Rhizoctonia solani and Sclerotium
been widely exploited for delivery and targeting of actives from herbs rolfsii, Ind. Crops Prod. 108 (2017) 379–387.
through a different route of administration. Hence, NEs are promising [12] S. Surassmo, S.G. Min, P. Bejrapha, M.J. Choi, Effects of surfactants on the physical
for effective delivery of herbal therapeutics and others. properties of capsicum oleoresin-loaded nanocapsules formulated through the
emulsion-diffusion method, Food Res. Int. 43 (2010) 8–17.
[13] S. Pund, G. Borade, G. Rasve, Improvement of anti-inflammatory and anti-angio-
Conflicts of interest genic activity of berberine by novel rapid dissolving nanoemulsifying technique,
Phytomedicine 21 (2014) 307–314.
The authors confirmed no conflicts of interest. [14] M. Yang, Y. Gu, D. Yang, X. Tang, J. Liu, Development of triptolide-nanoemulsion
gels for percutaneous administration: physicochemical, transport, pharmacokinetic
and pharmacodynamic characteristics, J. Nanobiotechnol. 15 (2017) 88.
Acknowledgment [15] H. Choudhury, B. Gorain, S. Karmakar, E. Biswas, G. Dey, R. Barik, M. Mandal,
T.K. Pal, Improvement of cellular uptake, in vitro antitumor activity and sustained
release profile with increased bioavailability from a nanoemulsion platform, Int. J.
Authors (RKH and RD) are deeply gratitude to the GLA University, Pharm. 460 (2014) 131–143.
Mathura, India for providing necessary facilities to accomplish this [16] A.Y. Choi, C.T. Kim, H.Y. Park, H.O. Kim, N.R. Lee, K.E. Lee, H.S. Gwak,
work. Authors are also grateful to Prof. Anoop Kumar Gupta, Dean, Pharmacokinetic characteristics of capsaicin-loaded nanoemulsions fabricated with
alginate and chitosan, J. Agric. Food Chem. 61 (2013) 2096–2102.
Academic Affairs, GLA University, Mathura, India for his motivation [17] Y. Li, J. Zheng, H. Xiao, D.J. McClements, Nanoemulsion-based delivery systems for
and support toward the research. poorly water-soluble bioactive compounds: influence of formulation parameters on
polymethoxyflavone crystallization, Food Hydrocolloids 27 (2012) 517–528.
[18] M. Sessa, M.L. Balestrieri, G. Ferrari, L. Servillo, D. Castaldo, N. D'Onofrio, F. Donsì,
Appendix A. Supplementary data
R. Tsao, Bioavailability of encapsulated resveratrol into nanoemulsion-based de-
livery systems, Food Chem. 147 (2014) 42–50.
Supplementary data to this article can be found online at https:// [19] F. Shakeel, N. Haq, F.K. Alanazi, I.A. Alsarra, Impact of various nonionic surfactants
on self-nanoemulsification efficiency of two grades of Capryol (Capryol-90 and
doi.org/10.1016/j.jddst.2019.03.006.
Capryol-PGMC), J. Mol. Liq. 182 (2013) 57–63.
[20] M.A. Rahman, R. Harwansh, M.A. Mirza, S. Hussain, A. Hussain, Oral lipid based
List of abbreviation drug delivery system (LBDDS): formulation, characterization and application: a
review, Curr. Drug Deliv. 8 (2011) 1–16.
[21] M. Jaiswal, R. Dudhe, P.K. Sharma, Nanoemulsion: an advanced mode of drug
GIT Gastrointestinal tract delivery system, 3 Biotech 5 (2015) 123–127.
GRAS Generally regarded as safe [22] S.A. Charman, W.N. Charman, M.C. Rogge, T.D. Wilson, F.J. Dukto, C.W. Pouton,
HLB Hydrophilic-lipophilic balance Self-emulsifying drug delivery systems: formulation and biopharmaceutic evalua-
tion of an investigational lipophilic compound, Pharm. Res. (N. Y.) 9 (1992) 87–93.
NDDS Nano drug delivery systems [23] P.M. Bummer, Physical chemical considerations of lipid-based oral drug delivery-
NEs Nanoemulsions solid lipid nanoparticles, Crit. Rev. Ther. Drug Carrier Syst. 21 (2004) 1–19.
o/w Oil in water nanoemulsion [24] P.P. Constantinides, Lipid microemulsions for improving drug dissolution and oral
absorption: physical and biopharmaceutical aspects, Pharm. Res. (N. Y.) 12 (1995)
PIC Phase inversion composition 1561–1572.
PIT Phase inversion temperature [25] M.J. Lawrence, G.D. Rees, Microemulsion based media as a novel drug delivery
Smix Surfactants and co-surfactants mixture system, Adv. drug delivery, Rev. 45 (2002) 89–121.
[26] M.G. Wakerly, C.W. Pouton, B.J. Meakin, F.S. Morton, Self-emulsification of ve-
SNEDDSs Self-nanoemulsifying drug delivery systems
getable oil nonionic surfactant mixture: a proposed mechanism of action, ACS
TDDS Topical and transdermal drug delivery systems Symp. Ser. 311 (1986) 242–255.
w/o Water in oil nanoemulsion [27] D.Q.M. Craig, S.A. Barker, D. Banning, S.W. Booth, An investigation into the me-
chanisms of self-emulsification using particle size analysis and low frequency di-
electric spectroscopy, Int. J. Pharm. 114 (1995) 103–110.
References [28] D. Attwood, Microemulsions, in: J. Kreuter (Ed.), Colloidal Drug Delivery Systems,
Marcel Dekker, New York, 1994, pp. 31–71.
[1] P.K. Mukherjee, R.K. Harwansh, S. Bhattacharyya, Bioavailability of herbal pro- [29] M.J. Lawrence, Surfactant systems: microemulsions and vesicles as vehicles for drug
ducts: approach toward improved pharmacokinetics, in: P.K. Mukherjee (Ed.), delivery, Eur. J. Drug Metab. Pharmacokinet. 3 (1994) 257–269.
Evidence-Based Validation of Herbal Medicine, Elsevier, Amsterdam, 2015, pp. [30] M. Kreilgaard, E.J. Pedersen, J.W. Jaroszewski, NMR characterization and trans-
217–245. dermal drug delivery potential of microemulsion systems, J. Control. Release 69
(2000) 421–433.
232
R.K. Harwansh, et al. Journal of Drug Delivery Science and Technology 51 (2019) 224–233
[31] S. Tenjarla, Microemulsions: an overview and pharmaceutical applications, Crit. applications, Iran, J. Pharm. Sci. 7 (2011) 139–150.
Rev. Ther. Drug Carrier Syst. 16 (1999) 461–521. [63] H.A.E. Benson, Transdermal drug delivery: penetration enhancement techniques,
[32] M.J. Lawrence, Microemulsions as drug delivery vehicles, Curr. Opin. Colloid Curr. Drug Deliv. 2 (2005) 23–33.
Interface Sci. 1 (1996) 826–832. [64] K.S. Paudel, M. Milewski, C.L. Swadley, N.K. Brogden, P. Ghosh, A.L. Stinchcomb,
[33] D. Attwood, G. Ktistis, Y. McCormick, M.J. Story, Solubilization of indomethacin by Challenges and opportunities in dermal/transdermal delivery, Ther. Deliv. 1 (2010)
polysorbate 80 in mixed water-sorbitol solvents, J. Pharm. Pharmacol. 41 (1989) 109–131.
83–86. [65] M.R. Prausnitz, S. Mitragotri, R. Langer, Current status and future potential of
[34] D. Attwood, C. Mallon, C.J. Taylor, Phase studies of oil-in-water phospholipid mi- transdermal drug delivery, Nat. Rev. Drug Discov. 3 (2004) 115–124.
croemulsions, Int. J. Pharm. 84 (1992) R5–R8. [66] A. Alexander, S. Dwivedi, Ajazuddin, T.K. Giri, S. Saraf, S. Saraf, D.K. Tripathi,
[35] G. Kitistis, I. Niopas, A study on the in vitro percutaneous absorption of propranolol Approaches for breaking the barriers of drug permeation through transdermal drug
from disperse system, J. Pharm. Pharmacol. 50 (1998) 413–419. delivery, J. Control. Release 164 (2012) 26–40.
[36] T.P.U. Ravi, T. Padma, Nanoemulsions for drug delivery through different routes, [67] R.J. Batchelder, R.J. Calder, C.P. Tomas, C.M. Heard, In vitro transdermal delivery
Res. Biotechnol. 2 (2011) 1–13. of the major catechins and caffeine from extract of Camellia sinensis, Int. J. Pharm.
[37] T.G. Mason, S.M. Graves, J.N. Wilking, M.Y. Lin, Extreme emulsification: formation 283 (2004) 45–51.
and structure of nanoemulsions, J. Phys. Condens. Matter 9 (2006) 193–199. [68] J.Y. Fang, H.T.L. Wang, Y.L. Huang, C.L. Fang, Enhancement of the transdermal
[38] L. Wang, X. Li, G. Zhang, J. Dong, J. Eastoe, Oil-in-water nanoemulsions for pes- delivery of catechins by liposomes incorporating anionic surfactants and ethanol,
ticide formulations, J. Colloid Interface Sci. 314 (2007) 230–235. Int. J. Pharm. 310 (2006) 131–138.
[39] S. Lamaallam, H. Bataller, C. Dicharry, J. Lachaise, Formation and stability of mini- [69] J. Suppasrivasuseth, R.A. Bellantone, F.M. Plakogiannis, G. Stagni, Permeability
emulsions produced by dispersion of water/oil/surfactants concentrates in a large and retention studies of (-)epicatechin gel formulations in human cadaver skin,
amount of water, Colloid. Surf. Physicochem. Eng. Asp. 270 (2005) 44–51. Drug Dev. Ind. Pharm. 32 (2006) 1007–1017.
[40] C. Solans, J. Esquena, A.M. Forgiarini, N. Uson, D. Morales, P. Izquierdo, [70] Y.P. Lu, Y.R. Lou, J.G. Xie, Q.Y. Peng, J. Liao, C.S. Yang, M.T. Huang, Topical ap-
Nanoemulsions: formation and properties, in: K.L. Mittal, D.O. Shah (Eds.), plications of caffeine or (-)-epicatechin gallate (EGCG) inhibit carcinogenesis and
Surfactants in Solution: Fundamentals and Applications, Marcel Dekker, New York, selectively increase apoptosis in UVB-induced skin tumours in mice, Proc. Natl.
2002, p. 525. Acad. Sci. Unit. States Am. 99 (2002) 12455–12460.
[41] P. Izquierdo, J. Feng, J. Esquena, T.F. Tadros, J.C. Dederen, M.J. Garcia, The in- [71] R.K. Harwansh, P.K. Mukherjee, S. Bahadur, R. Biswas, Enhanced permeability of
fluence of surfactant mixing ratio on nano-emulsion formation by the pit method, J. ferulic acid loaded nanoemulsion based gel through skin against UVA mediated
Colloid Interface Sci. 285 (2005) 388–394. oxidative stress, Life Sci. 141 (2015) 202–211.
[42] K. Shinoda, H. Saito, The effect of temperature on the phase equilibria and the types [72] H. Rachmawati, D.K. Budiputra, R. Mauludin, Curcumin nanoemulsion for trans-
of dispersions of the ternary system composed of water, cyclohexane, and non-ionic dermal application: formulation and evaluation, Drug Dev. Ind. Pharm. 41 (2015)
surfactant, J. Colloid Interface Sci. 26 (1968) 70–74. 560–566.
[43] A. Forgiarini, J. Esquena, C. Gonzalez, C. Solans, Formation of nano-emulsions by [73] H. Chaudhary, K. Kohli, V. Kumar, A novel nano-carrier transdermal gel against
low-energy emulsification methods at constant temperature, Langmuir 17 (2001) inflammation, Int. J. Pharm. 465 (2014) 175–186.
2076–2083. [74] H.L. Alvarado, G. Abrego, E.B. Souto, M.L. Garduño-Ramirez, B. Clares, M.L. García,
[44] C.M. Pey, A. Maestro, I. Solé, C. González, C. Solans, J.M. Gutiérrez, Optimization of A.C. Calpena, Nanoemulsions for dermal controlled release of oleanolic and ursolic
nano-emulsions prepared by low-energy emulsification methods at constant tem- acids: in vitro, ex vivo and in vivo characterization, Colloids Surf., B 130 (2015)
perature using a factorial design study, Colloid. Surf. Physicochem. Eng. Asp. 288 40–47.
(2006) 144–150. [75] R.K. Harwansh, P.K. Mukherjee, A. Kar, S. Bahadur, N.A. Al-Dhabi,
[45] O. Sonneville-Aubrun, J.T. Simonnet, F. L'Alloret, Nanoemulsions: a new vehicle for V. Duraipandiyan, Enhancement of photo-protection potential of catechin through
skincare products, Adv. Colloid Interface Sci. 108 (2004) 145–149. nanoemulsion gel against UVA induced skin damage, J. Photochem. Photobiol., B
[46] C. Solans, P. Izquierdo, J. Nolla, N. Azemar, M.J. Garcia-Celma, Nanoemulsions, 160 (2016) 318–329.
Curr. Opin. Colloid Interface Sci. 10 (2005) 102–110. [76] T. Gao, Z. Wei, Y. Zhao, In vitro and in vivo anti-tumor characterizations of β-
[47] I. Sole, A. Maestro, C.M. Pey, C. Gonzalez, C. Solans, J.M. Gutierrez, Nanoemulsions elemene-loaded nanoemulsion, Nanomed. Nanotechnol. Biol. Med. 12 (2016)
preparation by low energy methods in an ionic surfactant system, Colloids Surf., A: 449–575.
Physiochem. Eng. Aspects 288 (2006) 138–143. [77] T. Zhang, D. Dong, D. Lu, S. Wang, B. Wu, Cremophor EL-based nanoemulsion
[48] I. Sole, C.M. Pey, A. Maestro, C. Gonzalez, M. Porras, C. Solans, J.M. Gutierrez, enhances transcellular permeation of emodin through glucuronidation reduction in
Nanoemulsions prepared by phase inversion composition method: preparation UGT1A1-overexpressing MDCKII cells, Int. J. Pharm. 501 (2016) 190–198.
variables and scale up, J. Colloid Interface Sci. 344 (2010) 417–423. [78] R. Nazari-Vanani, L. Moezi, H. Heli, In vivo evaluation of a self-nanoemulsifying
[49] M. Porras, C. Solans, C. González, J.M. Gutiérrez, Properties of water-in-oil (W/O) drug delivery system for curcumin, Biomed. Pharmacother. 88 (2017) 715–720.
nano-emulsions prepared by a low-energy emulsification method, Colloid. Surf. [79] Y.J. Kim, S. Houng, J.H. Kim, Y. Kim, H.G. Ji, S. Lee, Nanoemulsified green tea
Physicochem. Eng. Asp. 324 (2008) 181–188. extract shows improved hypocholesterolemic effects in C57BL/6 mice, J. Nutr.
[50] N. Anton, T. Vandamme, The universality of low-energy nano-emulsification, Int. J. Biochem. 23 (2012) 186–191.
Pharm. 377 (2009) 142–147. [80] G. Chen-yua, Y. Chun-fen, L. Qi-lu, T. Qi, X. Yan-wei, L. Wei-na, Z. Guang-xi,
[51] S. Graves, K. Meleson, J. Wilking, Structure of concentrated nanoemulsions, J. Development of a quercetin-loaded nanostructured lipid carrier formulation for
Chem. Phys. 122 (2005) 134703, , https://doi.org/10.1063/1.1874952. topical delivery, Int. J. Pharm. 430 (2012) 292–298.
[52] S.M. Jafari, Y. He, B. Bhandari, Optimization of nanoemulsion production by mi- [81] H. Pool, S. Mendoza, H. Xiao, D.J. McClements, Encapsulation and release of hy-
crofluidization, Eur. Food Res. Technol. 225 (2007) 733–741. drophobic bioactive components in nanoemulsion-based delivery systems: impact
[53] C. Quin, D.J. McClement, Formation of nanoemulsions stabilized by model food of physical form on quercetin bioaccessibility, Food Funct 4 (2013) 162–174.
grade emulsifiers using high pressure homogenization: factors affecting particle [82] S. Bose, Y. Du, P. Takhistov, B. Michniak-Kohn, Formulation optimization and to-
size, Food Hydrocolloids 25 (2011) 1000–1008. pical delivery of quercetin from solid lipid based nanosystems, Int. J. Pharm. 441
[54] P.K. Mukherjee, M. Venkatesh, K. Maiti, K. Mukherjee, B.P. Saha, Value added (2013) 56–66.
herbal drug delivery systems - perspectives and developments, Indian J. Pharm. [83] A. Amri, J.C. Chaumeil, S. Sfar, C. Charrueau, Administration of resveratrol: what
Educ. Res. 43 (2009) 329–337. formulatio n solutions to bioavailability limitations? J. Control. Release 158 (2012)
[55] L. Zhang, L. Zhang, M. Zhang, Y. Pang, Z. Li, A. Zhao, J. Feng, Self-emulsifying drug 182–193.
delivery system and the applications in herbal drugs, Drug Deliv. 22 (2015) [84] C. Qian, E.A. Decker, H. Xiao, D.J. McClements, Nanoemulsion delivery systems:
475–486. influence of carrier oil on β-carotene bioaccessibility, Food Chem. 135 (2012)
[56] R.K. Harwansh, P.K. Mukherjee, S. Biswas, Nanoemulsion as a novel carrier system 1440–1447.
for improvement of betulinic acid oral bioavailability and hepatoprotective activity, [85] H.D. Silva, M.A. Cerqueira, B.W.S. Souza, C. Ribeiro, M.C. Avides, M.A.C. Quintas,
J. Mol. Liq. 237 (2017) 361–371. J.S.R. Coimbra, M.G. Carneiro-da-Cunha, A.A. Vicente, Nanoemulsions of β-car-
[57] T.H. Tran, Y. Guo, D. Song, R.S. Bruno, X. Lu, Quercetin-containing self-nanoe- otene using a high-energy emulsification evaporation technique, J. Food Eng. 102
mulsifying drug delivery system for improving oral bioavailability, J. Pharm. Sci. (2011) 130–135.
103 (2014) 840–852. [86] Y. Zhang, Z. Shang, C. Gao, M. Du, S. Xu, H. Song, T. Liu, Nanoemulsion for so-
[58] R. Pangeni, S.W. Kang, M. Oak, E.Y. Park, J.W. Park, Oral delivery of quercetin in lubilization, stabilization, and in vitro release of pterostilbene for oral delivery,
oil-in-water nanoemulsion: in vitro characterization and in vivo anti-obesity effi- AAPS PharmSciTech 15 (2014) 1000–1008.
cacy in mice, J. Funct. Foods 38 (2017) 571–581. [87] M.A. Rahman, A. Hussain, Z. Iqbal, R.K. Harwansh, L.R. Singh, S. Ahmad,
[59] R. Parveen, S. Baboota, J. Ali, A. Ahuja, S.S. Vasudev, S. Ahmad, Oil based nano- Nanosuspension: a potential nanoformulation for improved delivery of poorly
carrier for improved oral delivery of silymarin: in vitro and in vivo studies, Int. J. bioavailable drug, Micro Nanosyst. 5 (2013) 273–287.
Pharm. 413 (2011) 245–253. [88] B. Mishra, B.B. Patel, S. Tiwari, Colloidal nanocarriers: a review on formulation
[60] Y. Zhao, C. Wang, A.H. Chow, K. Ren, T. Gong, Z. Zhang, Y. Zheng, Self-nanoe- technology, types and applications toward targeted drug delivery, Nanomedicine 6
mulsifying drug delivery system (SNEDDS) for oral delivery of Zedoary essential oil: (2010) 9–24.
formulation and bioavailability studies, Int. J. Pharm. 383 (2010) 170–177. [89] F. Aqil, R. Munagala, J. Jeyabalan, M.V. Vadhanam, Bioavailability of phyto-
[61] Q. Shen, Y. Wang, Y. Zhang, Improvement of colchicine oral bioavailability by chemicals and its enhancement by drug delivery Systems, Cancer Lett. 334 (2013)
incorporating eugenol in the nanoemulsion as an oil excipient and enhancer, Int. J. 133–141.
Nanomed. 6 (2011) 1237–1243. [90] B.V. Bonifácio, P.B. Silva, M.A. Ramos, K.M. Negri, T.M. Bauab, M. Chorilli,
[62] S.S. Abolmaali, A.M. Tamaddon, F.S. Farvadi, S. Daneshamuz, H. Moghimi, Nanotechnology-based drug delivery systems and herbal medicines: a review, Int. J.
Pharmaceutical nanoemulsions and their potential topical and transdermal Nanomed. 9 (2014) 1–15.
233