Microemulsions As Transdermal Drug Delivery Vehicles

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Advances in Colloid and Interface Science 123–126 (2006) 369 – 385

www.elsevier.com/locate/cis

Microemulsions as transdermal drug delivery vehicles


Anna Kogan, Nissim Garti ⁎
Casali Institute of Applied Chemistry, The Institute of Chemistry, Givat Ram Campus, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
Available online 14 July 2006

Abstract

Microemulsions are clear, stable, isotropic mixtures of oil, water, and surfactant, frequently in combination with a cosurfactant. Microemulsions
have been intensively studied during the last decades by many scientists and technologists because of their great potential in many food and
pharmaceutical applications. The use of microemulsions is advantageous not only due to the facile and low cost preparation, but also because of the
improved bioavailability. The increased absorption of drugs in topical applications is attributed to enhancement of penetration through the skin by the
carrier. Saturated and unsaturated fatty acids serving as an oil phase are frequently used as penetration enhancers. The most popular enhancer is oleic
acid. Other permeation enhancers commonly used in transdermal formulations are isopropyl myristate, isopropyl palmitate, triacetin, isostearylic
isostearate, R(+)-limonene and medium chain triglycerides. The most popular among the enhancing permeability surfactants are phospholipids that
have been shown to enhance drug permeation in a different mode. L-α-phosphatidylcholine from egg yolk, L-α-phosphatidylcholine 60%, from
soybean and dioleylphosphatidyl ethanolamine which are in a fluid state may diffuse into the stratum corneum and enhance dermal and transdermal
drug penetration, while distearoylphosphatidyl choline which is in a gel-state has no such capability. Other very commonly used surfactants are
Tween 20®, Tween 80®, Span 20®, Azone®, Plurol Isostearique® and Plurol Oleique®. As cosurfactants commonly serve short-chain alkanols such
as ethanol and propylene glycol. Long-chain alcohols, especially 1-butanol, are known for their enhancing activity as well. Decanol was found to be
an optimum enhancer among other saturated fatty alcohols that were examined (from octanol to myristyl alcohol). Many enhancers are concentration-
dependent; therefore, optimal concentration for effective promotion should be determined. The delivery rate is dependent on the type of the drug, the
structure and ingredients of the carrier, and on the character of the membrane in use. Each formulation should be examined very carefully, because
every membrane alters the mechanism of penetration and can turn an enhancer to a retarder.
Various potential mechanisms to enhance drug penetration through the skin include directly affecting the skin and modifying the formulation
so the partition, diffusion, or solubility is altered. The combination of several enhancement techniques such as the use of iontophoresis with fatty
acids leads to synergetic drug penetration and to decrease in skin toxicity.
Selected studies of various microemulsions containing certain drugs including retinoic acid, 5-fluorouracil, triptolide, ascorbic acid, diclofenac,
lidocaine, and prilocaine hydrochloride in transdermal formulations are presented in this review.
In conclusion, microemulsions were found as an effective vehicle of the solubilization of certain drugs and as protecting medium for the
entrapped of drugs from degradation, hydrolysis, and oxidation. It can also provide prolonged release of the drug and prevent irritation despite the
toxicity of the drug. Yet, in spite of all the advantages the present formulations lack several key important characteristics such as cosmetic-
permitted surfactants, free dilution in water capabilities, stability in the digestive tracts and sufficient solubilization capacity.
© 2006 Elsevier B.V. All rights reserved.

Keywords: Microemulsions; Drug delivery system; Transdermal

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 370
2. Microemulsions as transdermal drug delivery vehicles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 371
2.1. Mechanisms of skin permeation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 371
2.2. Components of the transdermal formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 372
2.2.1. Oil phase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 372
2.2.2. Surfactants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 373

⁎ Corresponding author. Tel.: +972 2 658 6574/5; fax: +972 2 652 0262.
E-mail address: garti@vms.huji.ac.il (N. Garti).

0001-8686/$ - see front matter © 2006 Elsevier B.V. All rights reserved.
doi:10.1016/j.cis.2006.05.014
370 A. Kogan, N. Garti / Advances in Colloid and Interface Science 123–126 (2006) 369–385

2.2.3. Cosurfactants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 374


2.2.4. Aqueous phase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 374
2.3. Selected studies on microemulsions as transdermal drug vehicles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 374
2.3.1. Ascorbic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 374
2.3.2. Diclofenac. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 376
2.3.3. Lidocaine and prilocaine hydrochloride . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 379
2.3.4. Triptolide . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 380
2.3.5. 5-Fluorouracil . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 381
2.3.6. Retinoic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 381
3. Summary and conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 382
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 383
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 383

1. Introduction the physical or chemical properties of the drug, administrative


matters such as approval for use, excipients, and engineering
Pharmaceutical formulators aim to deliver the active mole- issues [2].
cule to the target organ at therapeutically relevant levels, with Some of the major challenges of drug delivery are [2] poor
negligible discomfort and side effects to the patient. This deli- solubility, short in vitro (shelf-life) and in vivo (half-life)
very is significantly influenced by the physical and chemical stability, low bioavailability, strong side effects (therefore,
properties of the drug. targeted delivery is needed) and regulatory issues. In order to
The biopharmaceutical classification system of drugs was increase the probability of a drug delivery formulation entering
defined by Amidon et al. in 1995 [1]. Class I and Class II drugs the pharmaceutical market, it should fulfill (as much as
demonstrate high gastrointestinal permeability. In contrast to possible) the main requirements [2]: ease of production, appli-
Class II, Class I covers drugs with high water solubility. Class I cability to as many drugs as possible, physical stability, ex-
drugs are well absorbed, but their bioavailability can still be low cipients that are well tolerated and accepted by regulatory
because of the first pass metabolism [1]. The rate-limiting step authorities and availability of large scale production allowable
to drug absorption is the drug dissolution. In cases of very rapid by regulatory authorities.
dissolution, the rate-controlling step will be gastric emptying. By many estimates, up to 40% of the new chemical entities
Compounds of Class II with solubility below 10 mg/ml present discovered by the pharmaceutical industry today are poorly
difficulties related to solubilization during formulation. In these soluble or lipophilic compounds. The solubility issue, compli-
cases, the rate controlling step to drug absorption is drug cating the delivery of these new drugs, also affects the delivery
dissolution. The drug dissolution profile can be affected by of many existing drugs. Researchers are making great efforts to
many formulation and in vivo variables; therefore, drugs in this discover new methods for delivery of poorly soluble drugs that
class can be expected to have variable absorption. The absorp- will be efficient and economically acceptable for drug
tion of these drugs is usually slower than for Class I. Class III manufacturers.
and IV drugs are compounds featuring high solubility and low The most common approach to improving the solubility of
permeability, and low solubility and low permeability, respec- drugs possessing a net negative electrical charge is to form salts
tively. Class III drugs feature variability in both the rate and (e.g., hydrochlorides, sulfates, nitrates, maleates, citrates, tar-
extent of absorption. However, if the dissolution is rather fast tarates) of the basic drugs. Yet, other drugs do not form such
(i.e., 85% dissolved in less than 15 min) the variability will be salts and this method of improving solubility is not possible.
due to the variations in luminal content, gastrointestinal transfer, Another route is to reduce particle size of powdered drug by
and membrane permeability which is the rate-determining step, new milling technologies or by applying new crystallization
and not due to the dosage form factors. Due to their character- processes to improve dissolution kinetics. Researchers are con-
istics, Class IV drugs exhibit many problems in their successful stantly searching for new and more efficient vehicles to carry
delivery [1]. In the conventional pharmaceutical industry and in active molecules into the blood stream. One approach to over-
modern medicine, it is well known that many promising drugs come these problems is to package the drugs into a particulate
that are discovered never make it to the market because of carrier system. Microcapsules, micro- and nanospheres, nano-
difficulties in delivery. This means that such drugs need to be powders, nanocrystals, and nanodispersions are only some of
formulated with smart drug delivery systems and/or deli- the options. Other options include delivery of the nutraceuti-
very technology to make them acceptable for the treatment of cals or drugs via liquid vehicles such as liposomes, emulsions,
patients. Some of these drugs are insoluble while others are double emulsions, microemulsions, micellar solutions and lyo-
eliminated by the acidity of the stomach, or are cleared from the tropic liquid crystals such as cubosomes and hexosomes. These
blood too rapidly to be effective. Intravenous delivery may last two vehicles are still in an experimental stage. The incor-
serve as an alternative in such cases, but may require either very poration of the drug into a carrier-system can be envisioned to
frequent administration or large a volume of drug injected each protect it against degradation in vitro as well as in vivo; the
time. The problems that occur with many drugs can be related to release can be controlled, and targeting can also be achieved.
A. Kogan, N. Garti / Advances in Colloid and Interface Science 123–126 (2006) 369–385 371

In 1959, Schulman et al. visualized the existence of small The aim of this chapter is to review the current literature with
emulsion-like structures by electron microscopy and subse- respect to the use of microemulsions for cutaneous drug deli-
quently coined the term “microemulsions” [3]. The term has very and to discuss the influence of microemulsion composi-
been defined and redefined by many authors. In this review, tion, components, and structure on the drug delivery potential as
however, we will use the most general definition provided by well as the tolerability of these vehicles both in vitro as well as
Danielsson and Lindman in 1981 [4]: a microemulsion is a in vivo.
single, optically isotropic structured solution of surfactant, oil, Many other potential pharmaceutical applications of micro-
and water. emulsions have been studied such as pulmonary [60–68], intra-
Microemulsions seem to be ideal liquid vehicles for drug vaginal or intrarectal administration delivery vehicles for
delivery since they provide all the possible requirements of lipophilic drugs such as microcides, steroids, and hormones
a liquid system including thermodynamic stability (long shelf- [69–77], and intramuscular formulations of peptide or cell-
life), easy formation (zero interfacial tension and almost targeting systems [78], other drugs have been also evaluated. The
spontaneous formation), low viscosity with Newtonian review recently written by Garti and Aserin includes among
behavior, high surface area (high solubilization capacity), others the recent progress in microemulsions for oral and intra-
and very small droplet size. The small droplets have better venous delivery [79]. The space limitations that are imposed
chance to adhere to membranes and to transport bioactive on this review prevent us from further elaborating on these
molecules in a more controlled fashion. Using the microemul- applications.
sion vehicles, water-insoluble and oil-soluble components
from different plant extracts can be co-solubilized in order to 2. Microemulsions as transdermal drug delivery vehicles
attain synergistic effect for a specific therapeutic goal.
Microemulsions can be introduced into the body orally, Transdermal drug delivery has many advantages over the
topically on the skin, or nasally, as an aerosol for direct entry oral route of administration: it avoids hepatic metabolism, the
into the lungs. administration is easier and more convenient for the patient, and
Microemulsions have been subjected to numerous studies there is the possibility of immediate withdrawal of the treatment
during the last decades because of their great potential in many if necessary. Despite the great potential of transdermal delivery
applications. Due to their rather complicated phase behavior and of drugs, only a few drug formulations are available com-
the fascinating microstructures encountered in microemulsion mercially. The main reason is the barrier function of human skin
forming systems, many researchers have made significant that is considered to be the most impermeable epithelium to
efforts to obtain a better understanding of these microstructures. exogenous substances [80].
Comprehensive reviews discussing the microstructures encoun-
tered in microemulsion phases were written [5–8]. 2.1. Mechanisms of skin permeation
In order to investigate the potential of microemulsions as
delivery vehicles, it is necessary to characterize their micro- The skin itself has two main layers: the epidermis, which is
structures as well as the locus of the drug in the loaded micro- the outermost layer of the skin, covering the dermis that is the
emulsion. In some cases, due to molecular interactions between active part of the skin, holding the hair muscles, blood supply,
the loaded drug and the microemulsion, the microstructure of sebaceous glands, and nerve receptors. There is a fat layer
the system may be altered. Due to the complexity of the mic- underneath the dermis. The skin is a very heterogeneous mem-
roemulsions and the variety of the structures and components brane and has a variety of cell types, but the layer that controls
involved in construction of the microstructure, as well as the the penetration of drugs is called the stratum corneum and,
limitation associated with each technique, the characterization despite its thickness of only 15–20 μm, it provides a very
of such structures is a rather difficult task. Some of the major effective barrier to penetration. The permeation of the drug
methods relevant to the characterization of the microemulsions through the skin has several routes: transcellular, intercellular,
include viscosity [9–14] and conductivity [9,10,15–25] mea- and appendageal (through eccrine (sweat) glands or hair
surements as well as more advanced methods such as cryo-TEM follicles) (Fig. 1). Since the appendages occupy a very low
[26–34], pulsed gradient spin echo (self-diffusion) NMR surface area, this means of permeation is less significant under
[32,35–42], dynamic light scattering (DLS) [43,44], small
angle X-ray scattering (SAXS) [45–55] and small angle neutron
scattering (SANS) [56–59].
However, microemulsions suffer from high surfactant
concentrations and in most cases from high alcohol, solvent,
and co-solvent contents. High levels of non-active compounds
are always a hazard. Patent search shows that microemulsions
have been heavily patented by researchers, as well as by com-
panies, as delivery systems, but a close examination of formu-
lations that are available in the market place reveals that the
technology is far from being exhausted and applied. The ques- Fig. 1. Schematic representation of the different possible routes of penetration
tion is always why? through the skin. Reprinted with permission from Hadgraft [81].
372 A. Kogan, N. Garti / Advances in Colloid and Interface Science 123–126 (2006) 369–385

normal conditions [81]. Nevertheless, in iontophoretic delivery 2.2. Components of the transdermal formulations
this route is more significant [82].
The intercellular spaces consist of a mixture of lipids–cera- A large variety of chemicals were defined as penetration
mides, free fatty acids and their esters, and cholesterol and its enhancers, but their use in transdermal formulations is limited
sulphates that are structured in bilayers. Recent developments in because of toxicity issues or unclear mechanisms of action.
spectroscopic techniques give interesting insights at the Here, we will briefly discuss the most common penetration
molecular level that may explain the impermeability of the enhancers that are used as the components of topically applied
skin by repeated partition and diffusion across structured bi- microemulsions. In the very comprehensive review of Williams
layers [81]. Transdermal drug permeability is influenced mainly and Barry [84] the reader can find more widely used penetration
by three factors: the mobility of the drug in the vehicle, the enhancers and their possible mechanisms of action.
release of the drug from the vehicle, and drug permeation
through skin [83]. Therefore, the researchers are challenged to 2.2.1. Oil phase
come up with formulations that increase the permeability of the Saturated and unsaturated fatty acids can be used as effective
drug without irreversibly changing the skin barrier function. penetration enhancer for a variety of drugs (e.g., naloxone,
Various potential mechanisms to enhance drug penetration hydrocortisone, estradiol, and peptides like LHRH, and CCK-8)
through the skin include directly affecting the skin and modi- [81,85–89]. Aungst et al. [90] studied various fatty acids,
fying the formulation so the partition, diffusion, or solubility are alcohols, sulphoxides, surfactants, and amides as enhancers for
altered [81,84]. Here we will present briefly these potential naloxone. It was suggested that enhancers with a saturated alkyl
mechanisms that are interconnected with each other. chain of C10–C12 and a polar head, and enhancers with an
unsaturated C18 alkyl chain such as oleic acid, appeared to be
1. Direct effect on the skin [84] the optimal ones. The unsaturated cis configuration perturbs the
a. Denaturation of intracellular keratin or modification of its lipid packing more than does the trans configuration [90]. Since
conformation causes swelling and increased hydration the tail of the stratum corneum bilayer is hydrophobic, the fatty
b. Affection of desmosomes (known as macula adherens- acids can enter the bilayer, perturb it by creating separate
cell structures specialized for cell to cell adhesion) that domains, and in this way may induce highly permeable path-
maintain cohesion between corneocytes (dead cells of the ways in the stratum corneum [91].
stratum corneum) The most popular enhancer is oleic acid. It increased the flux
c. Modification of lipid bilayers reduces resistance to of salicylic acid 28-fold and of 5-flourouracil 56-fold through
penetration human skin membrane in vitro [92]. Nevertheless, in examina-
d. Altering the solvent properties of the stratum corneum to tion of hexyl nicotinate on human skin in vivo, the addition of
modify drug partitioning oleic acid to propylene glycol (PG) did not improve the
e. Use of solvent that can extract the lipids in the stratum permeation flux achieved with PG alone [93]. It is important to
corneum and decrease its resistance to penetration note that formulators should use oleic acid in their formulations
2. Modification of the formulation [84] with great care, since its application may cause changes in the
a. Supersaturation state produced by volatile solvent that morphology of the Langerhans cells that are located in the
leaves the active substance in a more thermodynamically superbasal layer of the epidermis and play a key role in the
active state initiation and coordination of the T-cell mediated immune
b. Choosing the enhancer molecules in the vehicle that are response. Their depletion from the epidermis can cause skin
good solvents for the active ingredient and which enhance immunosuppression [94].
permeation through the skin; this way the partition of the The combination of fatty acids and iontophoretic delivery
drug into the stratum corneum will be improved resulted in enhanced permeation of many drugs. In iontophoretic
c. The diffusion of the active ingredient through the skin delivery, a small electric current applied between two electrodes
may be facilitated by using enhancers that create liquid placed on the skin enhances the penetration of ionized, neutral,
pools within the bilayers like oleic acid, or disturb the or polar molecules in a controlled manner [95,96]. Appendageal
bilayers uniformly as do the Azone® molecules (Azone® sites are suggested as important sites through which electrotran-
(1-dodecylazacycloheptan-2-one or lauro capram) is the sport occurs; ion and water transport have also been reported to
first molecule specifically designed as a skin permeation be associated, at least in part, with stratum corneum lipid lamel-
enhancer. Azone® serves as surfactant and enhances the lae. Jadoul et al. [82] suggested that permeation enhancement by
skin transport of a wide variety of drugs including ste- iontophoresis was related to lipid layer stacking disorganization.
roids, antibiotics and antiviral agents [84]). Since fatty acids enhance permeation by selective perturba-
tion in the intercellular lipids of the stratum corneum, and ions
Sometimes the synergy between several enhancement effects are known to take the path with less resistance: so combining
causes a greater enhancement in permeability of the desired the use of iontophoresis and fatty acids is an alternative that
substance. More detailed explanations and various examples of researchers have used to increase the permeation of not only
the enhancement mechanisms can be found in very comprehen- low-molecular-weight drugs, but also high-molecular-weight
sive reviews recently written by Handgraft [81] and Williams and proteins such as insulin, and peptides like LHRH, CCK-8 and
Barry [84]. AVP [85,86,97–99]. Furthermore, the combination of these two
A. Kogan, N. Garti / Advances in Colloid and Interface Science 123–126 (2006) 369–385 373

methods can decrease skin toxicity and irritation that are The phase properties of systems contained isopropyl myristate
associated with use of high levels of either of them. Rastogi and (IPM) as an oil, lecithin from egg or soy as a low toxicity
Singh [99] investigated the influence of R(+)-limonene and fatty surfactant, butanol [128] or ethanol as cosurfactant, and water.
acids such palmitic (C16:0), palmitoleic (C16:1), stearic (C18:0), The influence of lecithin and butanol or ethanol on the area of
oleic (C18:1), linoleic (C18:2), and linolenic (C18:3) on the the microemulsion was examined. In the system containing
penetration enhancement of insulin. It was shown that iontophor- butanol oil-in-water microemulsion regions were detected at a
esis in combination with chemical enhancers synergistically in- surfactant/cosurfactant weight ratio between 1:0.6 and 1:0.33.
creased (p < 0.05) the in vitro permeability of insulin. Linolenic In the systems consisting of ethanol, microemulsion area was
acid enhanced the permeability of insulin through the epidermis obtained in only 60–80% ethanol in water w/w. It was shown
more than other fatty acids did during both passive that in order to obtain a microemulsion, the amount of lecithin
(44.45 × 10− 4 cm/h) and iontophoretic (78.03 × 10− 4 cm/h) tran- required increased as the amount of the ethanol decreased. In
sport. The authors claim that by using an iontophoretic patch size comparison to the systems containing butanol it was found that
of 10 cm2, they would be able to deliver 50 IU of insulin within at the same amount of IPM more ethanol was required. Larger
3 h [99]. amounts of ethanol are required to reduce the rigidity of the
Nair and Panchagnula [98] assessed in vitro the effect of lecithin condensed film, creating the curvature that is needed for
oleic, linoleic, and lauric fatty acids alone and in combination droplet formation. This is explained by the low interface/water
with iontophoresis on the transdermal penetration of arginine partition coefficient of ethanol because of its hydrophilicity
vasopressin (AVP), a cyclic peptide that is used in treatment of [128]. It is known that with an increase in the aqueous phase
diabetis insipidus, alzheimer, and other diseases. It was found fraction, the droplet size increases. Atwood et al. [128] found
that all fatty acids increased the AVP flux compared to rat skin that at constant aqueous phase content, droplet size increased as
that was not pretreated with enhancer, but their effectiveness in the amount of lecithin decreased. Other authors have investi-
permeability enhancement was similar. This study provided gated formulations based on lecithins [106,116,125]. Different
direct evidence that oleic acid in mixture of ethanol and water phospholipids enhance drug permeation differently. Kirjavainen
causes disruption of the stratum cornea lipid lamellae. Never- et al.'s [126] study showed that the reason for this difference
theless, this study showed that iontophoresis did not further was that different phospholipids had different effects on the
increase the permeation of AVP through linoleic pretreated skin partitioning of drugs into the lipid bilayers. The authors used
[98]. Other researchers detected similar effects with fatty acids stratum corneum lipid liposomes as a model for the lipid matrix
that increased the permeation of midodrine hydrochloride and of stratum corneum. Estradiol, progesterone, and propranolol
leuprolide acetate [100,101]. Iontophoresis was also effective were studied. They concluded that EPC (L-α-phosphatidylcho-
for enhancing transdermal penetration from microemulsions as line from egg yolk), SPC (L-α-phosphatidylcholine 60%, from
it was shown for methotrexate [102]. soybean), and DOPE (dioleylphosphatidyl ethanolamine)
Another compound that is often used as an oil phase and as a which are in a fluid state may diffuse into the stratum corneum
permeation enhancer in transdermal formulations is isopropyl and enhance dermal and transdermal drug penetration, while
myristate (IPM), but the mechanism of its action is poorly DSPC (distearoylphosphatidyl choline) which is in a gel-state is
understood [13,83,102–112]. In some cases IPM does not show a not able to do this [126].
significant effect [113]. Other oils commonly used are isopropyl There is wide use of nonionic surfactants in topical formula-
palmitate [114–116], triacetin [117], isostearylic isostearate tions as solubilizing agents. Among them, the polysorbates (etho-
[36,118], R(+)-limonene [118] and Mygliol 812® (medium xylated sorbitan esters) are very common. Tween 80® was
chain length triglyceride) [119–122]. reported to accelerate hydrocortisone and lidocaine permeation,
and Tween 20® improved the permeation of 5-flourouracil across
2.2.2. Surfactants hairless mouse skin [129–131].
More than 15 years ago, Kato et al. [123] proposed the use of Some recent results indicate that a nonionic surfactant may
low toxicity substances, e.g., phospholipids, as penetration en- also affect the skin barrier function [132,133]. In the experiments
hancers. The penetration of theophylline was enhanced by 1% conducted in vitro on rat skin it was found that Span 20®, Tween
phosphatidylcholine in propylene glycol through hairless mouse 20®, and Azone® have different mechanisms of enhancement.
skin. Numerous studies have been conducted using various leci- Span 20® and Azone® affect the intercellular lipids of the stratum
thin-based microemulsions as topical formulations for enhancing corneum by making them more fluid. This way the diffusion of
the penetration of methotrexate [124], ascorbic acid [115], lipophilic compounds through the stratum corneum (lipophilic
diclofenac [116,125], indomethacin [116], ketoprofen [117,119], pathway) is enhanced. Tween 20® enhances penetration by al-
hematoporphyrin [106], etc. Absorption of phospholipids on skin lowing the polar molecule to partition across the barrier more
can increase tissue hydration, consequently increasing drug per- easily. It is also possible that since the experiments were con-
meation. When phospholipids are applied to skin as vehicles due to ducted in aqueous medium, large micelles were formed. These
their physico-chemical properties and structures they can fuse with micelles have the potential to extract lipids from the skin. This
stratum corneum lipids, perturb its structure and facilitate drug modifies the composition of the membrane and favors permeation
delivery [84,126]. of the hydrophilic compounds [132,133]. Recently, new low-irri-
As early as 1990, Friberg and later Atwood et al. published tant surfactants based on caprylocaproyl macrogolglycerides for
works on drug delivery systems for skin applications [127,128]. microemulsions as drug delivery vehicles for topical application
374 A. Kogan, N. Garti / Advances in Colloid and Interface Science 123–126 (2006) 369–385

were studied. In order to facilitate caprylocaproyl macrogolgly- whereas oleic acid increased it. In this work ethanol, propylene
ceride based microemulsion formation, the surfactants based on glycol, and phospholipids that are known as enhancers turned
polyglycerol fatty acid esters were used as cosurfactants [13,134– out to be retarders [148].
136]. Other surfactants such as dioctyl sodium sulphosuccinate
[137,138], Plurol Isostearique® [36,102,135,139], and Plurol 2.2.4. Aqueous phase
Oleique® [13,118,120,121,140] are used very often. In most of the studies water served as the aqueous phase
[13,36,102–105,107,108,111,114–117,119–122,125,
2.2.3. Cosurfactants 137,138,140,149,150]. In some of the cases phosphate buffer of
Short-chain alkanols are widely used as permeation pH 7.4 was used [102,109,112,118,135].
enhancers. Ethanol is very common among transdermal
formulations and its addition is known to enhance the flux of 2.3. Selected studies on microemulsions as transdermal drug
several drugs. Levonorgestrel [141], estradiol [142,143], and an vehicles
ionized form of diclofenac were shown to be better transported
through rat skin when ethanol was added [144]. Sometimes The application of microemulsion vehicles for cutaneous
when using ethanol-water-based vehicles, the effect of ethanol drug delivery is becoming increasingly popular due to their high
is concentration-dependent, and, therefore, under certain solubilization potential for both lipophilic and hydrophilic
conditions it can even decrease the permeation [84,145–147]. drugs. It was demonstrated that permeation rates from
Various mechanisms have been suggested for the enhancing microemulsions were significantly higher than from conven-
activity of ethanol. It can increase the drug solubility in the tional emulsions. In emulsions (creams, lotions, etc.), the strong
vehicle [84,143] or it can alter the structure of the membrane interactions between the surfactants that occur in the interfacial
and increase the permeability of the drug [84,146]. Another membrane film limit the mobility of the drug between the
mechanism is based on the fact that ethanol is volatilized from internal and external phases within the formulation. In addition,
the applied formulation and, consequently, increases the drug emulsions are not stable formulations and strong fluctuations in
concentration to a supersaturated state with a greater driving bioavailability were detected. In microemulsions, the co-
force for permeation. In addition, ethanol may extract some of surfactant assists in lowering the interfacial tension of the
the lipid fraction from the stratum corneum and, thus, can surfactant film, resulting in spontaneous formation of a
improve the drug flux through it [84]. In some cases ethanol by microemulsion and promoting its thermodynamic stability
itself does not serve as an enhancer, but needs a co-enhancer as [36,137,151]. The thermodynamic process of drug diffusion
demonstrated in the case of penetration of morphine hydro- across the flexible interfacial surfactant film between the phases
chloride [113]. In this study, a combination of 1-menthol with of the microemulsion can increase partitioning and diffusion
ethanol showed greater penetration enhancement than if ethanol into the stratum corneum [114]. The drug may also be retained
alone was used as solvent. It was suggested that the high in the droplets of the microemulsion formulation resulting in
penetration enhancement was derived from the influence of 1- increased concentration of surfactant in dispersed systems that
menthol on the stratum corneum barrier and the effect of ethanol will decrease its permeation into the skin [107].
against the viable skin layer beneath it [84,113]. Long-chain An excellent and comprehensive review on the role of
alcohols are known for their enhancing activity as well. 1- microemulsions in percutaneous penetration of drugs was recently
Butanol was reported to be an enhancer for levonorgesterol published by Kreilgaard [80] in which the author summarized the
[141]. Decanol was found to be an optimum enhancer for main studies published before 2002 in several tables. We have
melatonin among other saturated fatty alcohols that were included the data gathered by this author as well as additional
examined (from octanol to myristyl alcohol) [141]. Propylene studies that have been conducted since then in Table 1
glycol (PG) has been found to act as an enhancer by a [13,36,83,102–112,114–122,124,125,135,137,138,140,149,
mechanism similar to that of ethanol [84], enhancing the 150,152–154]. Many studies have been conducted incorporating
permeability of hexyl nicotinate. The addition of oleic acid to many drugs in microemulsions, we decided to describe only those
PG did not show an improvement in permeation [93]. studies conducted on certain drugs.
It is important to stress that each formulation should be
examined very carefully, because every membrane alters the 2.3.1. Ascorbic acid
mechanism of penetration and can turn an enhancer to a re- Ascorbic acid (vitamin C) is an essential nutrient protecting
tarder. In vitro studies on the transdermal absorption of flur- living tissues and cells from oxidation processes by free radicals
biprofen (a chiral non-steroidal anti-inflammatory drug that is and reactive oxygen derived species. Gallarate et al. [115]
used for treatment of gout, osteoarthritis, rheumatoid arthritis, studied the stability of ascorbic acid in several o/w microemul-
and sunburn) from cellulose hydrogels were conducted by Fang sions, o/w and w/o emulsions, and a w/o/w multiple emulsion.
et al. [148] using different kinds of additives. The permeation Isopropyl palmitate or cetearyl octanoate were used as oils,
was studied on different types of skin, such as skin pre-treated dodecylglucoside and cocoamide propylbetaine were used as
by various additives, stratum corneum stripped skin, and surfactants, and 2-ethyl-1,3-hexanediol was chosen as cosur-
delipidized skin. Propylene glycol and ethanol did not increase factant. These emulsifiers are non-ionic, non-ethoxylated, and
skin absorption of the drug, and phospholipids even markedly skin compatible. The addition of only 1% w/w of phosphati-
reduced it. Ethanol reduced the skin reservoir of the drug, dylcholine to formulations allowed a two-fold reduction in the
A. Kogan, N. Garti / Advances in Colloid and Interface Science 123–126 (2006) 369–385 375

Table 1
Overview of transdermal drug delivery systems containing microemulsions
Drug Microemulsion Membrane/ References
skin
Oil phase Surfactants/cosurfactants Aqueous phase
[3H]H2O octanol dioctyl sodium sulphosuccinate water human [137,138]
5-Fluorouracil isopropyl myristate AOT water mouse [149]
8-Methoxsalen isopropyl myristate Tween 80®, Span 80®, 1,2-octanediol water pig [103]
Apomorphine isopropyl myristate, Epikuron 200®, 1, water, mouse [104]
hydrochloride decanol 2-propanediol benzyl alcohol Aerosil 200®
Ascorbic acid isopropyl palmitate, dodecylglucoside cocoamide propylbetaine, water – [115]
cetearyl octanoate phosphatidylcholine/2-ethyl-1,3-hexanediol
Ascorbyl Mygliol 812® Labrasol®/Plurol Oleique® water – [121]
palmitate
Diclofenac isopropyl palmitate Lecithin water human [116]
Diclofenac Diclofenac diethylamine Lecithin water human [125]
Diclofenac isopropyl myristate Labrasol®/Plurol Oleique® water – [13]
Diphenhydramine isopropyl myristate Tween 80®, Span 20® water human [108]
hydrochloride
Estradiol Epicuron 200®, oleic acid, Tween 20®, Tween 80®, water human [83]
isopropyl myristate Span 80®/ethanol, isopropanol)
Felodipine isopropyl myristate, Tween 20®, tauro-deoxycholate water, mouse [105]
benzyl alcohol Transcutol®, Carbopol®
Glucose octanol dioctyl sodium sulphosuccinate water human [137]
Hematoporphyrin isopropyl myristate, decanol, lecithin, sodium monohexyl-phosphate, water, PG mouse [106]
hexadecanol, oleic acid, monoolein benzyl alcohol
Indomethacin isopropyl palmitate Lecithin water human [116]
Ketoprofen Miglyol 812 N® lecithin/n-butanol water human [119]
Ketoprofen triacetin, oleic acid Labrasol®, Cremophor RH® water rat [117]
Lidocaine isopropyl palmitate glyceryloleate, polyoxyl 40 fatty water mouse [114]
acid derivatives /tetraglycol
Lidocaine iostearylic isostearate Labrasol®, Plurol Isostearique® water rat [36]
Methotrexate decanol lecithin, benzyl alcohol water, PG mouse [124]
Methotrexate ethyl oleate Labrasol®/Plurol Isostearique® Aq. 154 mM NaCl (pH 7.4) pig [102]
Methotrexate isopropyl myristate Tween 80®, Span 80®, 1,2-octanediol water pig [2]
Nifedipine benzyl alcohol Tween 20®, tauro-deoxycholate water, mouse [152]
Transcutol®, PG
Niflumic acid octyl octanoate sucrose monolaurate sucrose dilaurate/ water human [150]
medium chain alcohol
Piroxicam isopropyl myristate hexadecyltrimethylammonium bromide Aqueous buffer (pH 5.5) rat [109]
Prilocaine isostearylic isostearate Labrasol®, Plurol Isostearique® water rat [36]
hydrochloride
Propranolol isopropyl myristate Tween 80® water artificial [107]
Prostaglandin E1 oleic acid Labrasol®, Plurol Oleique® water a mouse [140]
Prostaglandin E1 Gelucire® Labrafac®, Lauroglycol Transcutol®, water mouse [153]
Retinoic acid isopropyl myristate Epikuron 200®, Oramix® phosphate buffer artificial, pig [112]
NS10/ethanol, 1,2 hexanediol
Sodium ascorbyl Mygliol 812® Labrasol®/Plurol Oleique® water artificial [120]
phosphate
Sodium isostearylic isostearate, oleic acid, Labrasol®/Plurol Oleique® water, buffer pH 7.4 rat, rabbit [118]
diclofenac R(+)-limonene tributyrine
Sodium Mygliol 812®, soybean oil Brij 97® water pig [122]
fluorescein
Sodium salicylate isopropyl myristate Tween 21/81/85®, bis-2-(ethylhexyl) water, gelatin pig [110]
sulphosuccinate
Sucrose ethyl oleate Labrasol®, Plurol Isostearique® water, 154 mM mouse [135]
NaCl
Triptolide isopropyl myristate Tween 80®/1,2-propylene glycol water rat [111]
Triptolide oleic acid Tween 80®/propylene glycol H2O/menthol mouse [154]
Reprinted with permission from Kreilgaard et al. [80].
a
A variety of seven different co-solvents was tested with the basic vehicle.

amount of surfactant. Ascorbic acid stability against oxidation oxidation for ascorbic acid, and therefore its degradation rate
was examined in the prepared systems and compared to the was slower than in aqueous solutions. The degradation rate
aqueous solutions at different pH values. It was found that all increased with increasing pH. The highest protection of the
examined emulsified systems provided protection against ascorbic acid was achieved by solubilizing it in a w/o/w
376 A. Kogan, N. Garti / Advances in Colloid and Interface Science 123–126 (2006) 369–385

multiple emulsion. From the kinetic studies performed, a sensitive to oxidation is located in the internal aqueous phase
pseudo first-order mechanism for ascorbic acid degradation was and, therefore, it is protected by the interface that serves as a
suggested in the experimental conditions where abundant barrier for oxygen diffusion. In o/w microemulsions, the cyclic
dissolved oxygen was present (Fig. 2). Despite the fact that ring is located in the external aqueous phase, where it is less
ascorbic acid has many favorable effects, its use in cosmetic and protected, and therefore the degradation process is favored. In
pharmaceutical products is limited due to its low stability. In addition, it was found that light also accelerates the oxidative
order to solve this problem, derivatives of vitamin C, with degradation of ascorbyl palmitate. In order to formulate an
action similar to that of ascorbic acid, but with improved optimal carrier system for this ingredient, other factors influen-
chemical stability, have been synthesized. Two derivatives that cing the stability have to be considered. The studies on sodium
are widely used in cosmetic products are lipophilic ascorbyl ascorbyl phosphate revealed that, compared to ascorbyl pal-
palmitate and hydrophilic sodium ascorbyl phosphate that differ mitate, it was equally stable towards oxidation in both types of
in stability (Fig. 3) [121]. microemulsions. Consequently, sodium ascorbyl phosphate has
W/o microemulsions consisting of caprylic/capric triglycer- been found to be convenient for use as an active ingredient in
ide 24.75%, PEG-8 caprylic/capric glycerides 47.53%, poly- topical formulations [121]. In subsequent studies different types
glycerol-6 dioleate 11.88% and purified water 15.84%, were of thickening agents, that increase viscosity, were added to the
prepared by Špiclin et al. [121]. In parallel, o/w microemulsions microemulsions described above, to optimize these topical
containing caprylic/capric triglyceride 7.43%, PEG-8 caprylic/ formulations. The influence of thickening agents on the stability
capric glycerides 38.02%, polyglycerol-6 dioleate 9.50% and of the microemulsions and the release kinetics of the solubilized
purified water 45.05% were also prepared. Ascorbic acid sodium ascorbyl phosphate were studied. Magnesium stearate,
derivatives were incorporated into the two types of microemul- colloidal silica, or cetostearyl alcohol was used as thickening
sions at 0.25–2.00 wt.% concentrations. The influence of agent for w/o type microemulsions while for o/w microemul-
several parameters such as initial concentration, location in sions sodium alginate, methyl cellulose, HPMC K4M, HPMC
microemulsion, dissolved oxygen, and storage conditions on E4M, and xanthan gum were incorporated. As a suitable thick-
the stability of the less stable derivative ascorbyl palmitate was ening agent, colloidal silica was chosen for w/o microemulsions
examined. It was found that incorporation of ascorbyl palmitate and xanthan gum for the o/w type. The presence of thickening
into microemulsions at the highest concentration (2.00 w/w%) agent and the location of sodium ascorbyl phosphate in the
reduced its degradation. In presence of oxygen, ascorbyl microemulsion influenced the in vitro drug release profiles.
palmitate is more stable in w/o microemulsions; on the contrary When incorporated in w/o microemulsions, sustained release
if microemulsions are degassed, the compound is more stable in profiles were observed from both thickened and non-thickened
the o/w type. Therefore, the location of the active ingredient in systems. This is due to the external oil phase which can act as a
the microemulsion may have a great influence on its stability barrier for the diffusion of a hydrophilic compound. In the case
against oxidation. In a w/o microemulsion the cyclic ring that is of o/w systems, larger amounts were released due to the incor-
poration of the sodium ascorbyl phosphate in the external phase
that increased the diffusion rate (Fig. 4).
The type of thickening agent added to the examined systems
influenced the rate of drug diffusion. The addition of xanthan gum
(o/w system) increased the viscosity of the system and, therefore,
decreased the amount of released compound. On the contrary, the
amount of released sodium ascorbyl phosphate was higher in the
case of thickened w/o microemulsion with colloidal silica. The
authors assumed that colloidal silica modified the physicochem-
ical characteristics of the external phase; therefore, the diffusion
of sodium ascorbyl phosphate was faster. By evaluating the
release profiles by fitting the experimental data to different orders
kinetic equations, it was concluded that the rate-determining step
for sodium ascorbyl phosphate release from microemulsions was
its transport within the carrier system [120]. One can conclude
that microemulsions are an effective solution to preserving
ascorbic acid stability against oxidation and are effective vehicles
for controlling the release kinetics of the active compound.

2.3.2. Diclofenac
Several groups have selected diclofenac (a nonsteroidal anti-
inflammatory drug) as a model compound that was solubilized
Fig. 2. Pseudo first-order plot of ascorbic acid degradation at 45 ± 0.1 °C in in a microemulsion medium [13,116,118,125]. Kriwet and
emulsified systems at pH 5.0 (ascorbic acid = 1.00 wt.%) Reprinted with Müller-Goymann [125] investigated vehicles containing phos-
permission from Gallarate et al. [115]. pholipids, diclofenac diethylamine, and water. Various colloidal
A. Kogan, N. Garti / Advances in Colloid and Interface Science 123–126 (2006) 369–385 377

Fig. 3. Chemical structures of ascorbyl palmitate (a) and sodium ascorbyl phosphate (b). Reprinted with permission from Špiclin et al. [121].

structures such as liposomal dispersions, microemulsions and addition of diclofenac diethylamine, the number of vesicles with
lamellar liquid crystals can be formed by varying the ratios of multilamellar layers decreases until the system transforms into a
the components forming the structures and method of pre- microemulsion. Since the release of the entrapped drug in the
paration. In vitro studies, using a dialysis membrane impreg- vesicle is hindered, the fewer are the vesicles, the easier is the
nated with a silicone polymer, showed that the effective drug release. Permeation through human stratum corneum was
diffusion coefficient of diclofenac diethylamine changed ra- also examined. A linear relationship between the drug release
pidly with a phase transformation of the vehicle. By examina- out of the vehicle through the artificial membrane and the rate of
tion of the systems with constant concentration of diclofenac drug permeability through the stratum corneum was observed.
diethylamine (5% w/w) and variable amounts of phospholipids, The rate-limiting step for vehicles with a high effective diffusion
it was found that the diffusion coefficient of the drug decreased coefficient like microemulsions was the diffusional resistance in
with an increase of the phospholipids content [125] (Fig. 5). The the stratum corneum. On the contrary, for vehicles with a low
systems with 6% phospholipids or less are microemulsions with diffusion coefficient the rate-limiting step was the release from
low viscosity; therefore, the release is fast. Further addition of the vehicle. Only microemulsions showed enhanced perme-
the phospholipids leads to isotropic and afterwards anisotropic ability of diclofenac diethylamine through the stratum corneum,
gel formation, decreasing the release rate. The decrease in since the phospholipids were able to interact with the structures
diffusion coefficient can be explained by the increase of the of the stratum corneum only if they were applied as micro-
viscosity of the systems with the addition of phospholipids. The emulsion [125]. From these studies it can be concluded that
authors also investigated the influence of drug concentration in phospholipid content can alter the structure of the vehicle, hence
the vehicle on the effective diffusion coefficient [125] (Fig. 6). influences the drug release. Phospholipids can be used as a
It was found that drug molecules participate in the micro- thickening agents since they increase the viscosity of the system.
structure of the resulting systems. At concentrations up to 4.5% In addition, the drug itself may participate in the organization of
diclofenac diethylamine, liposomal dispersion, dispersion of the vehicle and consequently control its own release [125].
multilamellar vesicles in a microemulsion gel, or an isotropic Other authors showed that diclofenac diethylamine partici-
gel occurred. The viscosity of these systems is high, therefore pates in the vehicle structure due to its amphiphilic properties.
the drug release is slow. Further addition of diclofenac die- Djordjevic et al. [13] investigated microemulsion systems com-
thylamine leads to microemulsion formation with a low vis- posed of water, isopropyl myristate, PEG-8 caprylic/capric gly-
cosity, and the rate of the drug release is increased. With the cerides (Labrasol®), and polyglyceryl-6 dioleate (Plurol
Oleique®), as potential diclofenac diethylamine delivery vehicles.
It was found that in the experimental conditions, conductivity and
rheological properties were influenced significantly by the
incorporation of the drug. Conductivity values for drug loaded
microemulsions were increased by a factor of 2–3 in comparison
to microemulsions without the drug. On the contrary, the drug did
not significantly influence either the pH value of the vehicles, or
the stability and the optical texture of the examined systems [13].
Escribano et al. [118] investigated several ternary solvent
systems and microemulsions in vitro using human skin in order to
find the formulation that enhanced the percutaneous permeability
of sodium diclofenac. In order to compare the effect of the vehicle
on the percutaneous absorption, a fixed concentration of the drug
was used in all examined systems. A 1 wt.% solution of sodium
diclofenac and a commercially available semisolid preparation
served as reference. Commonly known enhancers such as oleic
and lauric acids, R(+)-limonene, and Transcutol® (diethylene
Fig. 4. Release profiles of sodium ascorbyl phosphate from o/w and w/o glycol monoethyl ether) were used as the components of the
microemulsions. Reprinted with permission from Špiclin et al. [120]. vehicles. The highest values of permeability coefficient and drug
378 A. Kogan, N. Garti / Advances in Colloid and Interface Science 123–126 (2006) 369–385

Fig. 5. Effective diffusion coefficient of diclofenac diethylamine from vehicle with 5% diclofenac diethylamine vs. content of phospholipids. Reprinted with
permission from Kriwet and Müller-Goymann [125].

flux, and the amount of drug that permeated and was retained in that the microemulsion would alter the skin lipids and show
the skin at 24 h were observed for 1 wt.% drug formulations enhanced permeation, it behaved like a solution. This may be
containing Transcutol® 59.2 wt.%, oleic acid 14.9 wt.%, and R attributed to the lower percentage of the enhancing agents (19%)
(+)-limonene 5 wt.% as enhancers, and water 19.9 wt.%. This as compared to 73% in other formulations. Therefore, the com-
formulation had greater anti-inflammatory activity, both local and ponents of the vehicle are no less important than the structure of
systemic, than the semisolid preparation. It was found that the the vehicle [118].
synergistic enhancement effect with Transcutol®/oleic acid/D- In other studies, diclofenac and indomethacin in a microemul-
limonene was greater than that of Transcutol®/oleic acid or sion gel were used as model drugs. The enhancement of drug
Transcutol®/lauric acid. In the comparison of permeation penetration was studied. The microemulsion gel consisted of
enhancement between oleic acid and lauric acid, it was shown soybean phosphatidylcholine (lecithin), isopropyl palmitate, and
that oleic acid provides more penetration capacity due to its cis a small amount of water. Studies using Fourier transform infrared
double bond at C9, which causes a kink in the alkyl chain and (FTIR) spectroscopy, differential scanning calorimetry (DSC), as
disrupts the skin lipids more efficiently. Despite the expectation well as low-temperature scanning electron microscopy, indicated

Fig. 6. Effective diffusion coefficient of diclofenac diethylamine in systems with 6% phospholipids vs. content of diclofenac diethylamine. Reprinted with permission
from Kriwet and Müller-Goymann [125].
A. Kogan, N. Garti / Advances in Colloid and Interface Science 123–126 (2006) 369–385 379

Table 2 transdermal flux of lidocaine up to four times, compared to a


Microemulsion compositions conventional oil-in-water emulsion, and increased the transder-
System Water Isostearylic isostearate Labrasol® Plurol Isostearique® mal flux of prilocaine hydrochloride almost 10 times compared
(%) (%) (%) (%) to hydrogels (Table 3). The flux seems to be dependent on drug
A 20 10 35 a 35 a solubility in the microemulsion as well as on drug mobility in
B 20 10 47 b 23 b the individual vehicle. It was found that the solubilities of the
C 20 10 53 c 17 c
drugs in the microemulsions were greater in comparison to the
D 7 70 11.5 a 11.5 a
E 11 26 42 b 21 b solubilities obtained in the components of the microemulsion.
F 55 8 25 b 12 b Due to high solubilization property of microemulsions which
G 65 3 24 c 8c leads to high concentration gradients, the transdermal flux in-
Reprinted with permission from Kreilgaard et al. [36]. creased. Lidocaine showed a 28–62% increase in solubility and
a
Labrasol®/Plurol Isostearique® 1:1 (w/w). prilocaine hydrochloride showed an increase of 24–40%. The
b
Labrasol®/Plurol Isostearique® 2:1 (w/w). large increase in solubility is due to surfactant film interface
c
Labrasol®/Plurol Isostearique® 3:1 (w/w).
between the oil and water phases that leads to additional
solubilization sites for drugs, compared to the molecular or-
that isopropyl palmitate affected the stratum corneum lipid orga- ganization of bulk surfactants. A linear relationship between
nization, but in vivo human skin irritation tests showed no sig- self-diffusion of the drugs in the vehicles and transdermal flux
nificant irritancy [116]. was found. It suggests that with given components, the per-
cutaneous delivery potential of microemulsion vehicles may be
2.3.3. Lidocaine and prilocaine hydrochloride decelerated by hindrance to diffusion due to the internal struc-
Lidocaine is a local anesthetic and cardiac depressant com- ture of the microemulsion [36].
monly used to restore a regular heartbeat in patients with ar- Sintov and Shapiro [114] also chose lidocaine as a lipophilic
rhythmia. Prilocaine hydrochloride is also used as an anesthetic. drug model. To reduce irritation, special types of microemul-
Kreilgaard et al. [36] investigated the influence of the structure sions were suggested. Glycerol oleate and polyoxyl [40 EO]
and composition of microemulsions on their transdermal fatty acid derivatives/tetraglycol (cosurfactant)/isopropyl pal-
delivery potential for a lipophilic (lidocaine) and a hydrophilic mitate/water were used to construct a pseudo-ternary phase
model drug (prilocaine hydrochloride), and compared the drug diagram at fixed cosurfactant/surfactant ratios (Fig. 7). Lido-
delivery potential of microemulsions to conventional vehicles. caine was solubilized and penetration studies using rat skin in
They used self-diffusion coefficients, as determined by the vitro showed that the transdermal flux of lidocaine was signi-
pulsed-gradient spin-echo NMR (PGSE-NMR), to study the ficantly improved by microemulsion composed of glycerol
transdermal flux using Franz-type diffusion cells. The authors oleate-PEG 400 stearate compared with that of glycerol oleate-
prepared phase diagrams where w/o and o/w microemulsions PEG 400 hydroxylated castor oil [114]. Fig. 8 shows a
were formed, and seven formulations for further studies were comparison of in vitro transdermal penetration between micro-
chosen (Table 2). The microemulsions were formulated with emulsion, mixture of surfactants, micellar, and macroemulsion
4.8 wt.% of lidocaine and 2.4 wt.% of prilocaine hydrochloride. systems. Macroemulsion and micellar systems presented a lower
It was demonstrated clearly that microemulsions increased the flux of lidocaine relative to the microemulsion. The authors

Table 3
Steady-state flux (J; 20–28 h) and permeation coefficient (κp) of lidocaine and prilocaine hydrochloride (HCl) through rat skin from microemulsion systems A–G with
near-maximum drug load and comparison with EMLA 5%, xylocain 5% o/w-cream and xylocain 2% hydrogel
Formulation Lipophilic model drug Hydrophilic model drug
[Lidocaine] J κp [Prilocaine HCl] J κp
(%, w/w) (μg × h- 1 × cm− 2) (μg × h− 1 × cm− 2) (%, w/w) (μg × h− 1 × cm− 2) (μg × h− 1 × cm− 2)
A 23 36.2 ± 4.2 1.6 ± 0.2 5 8.1 ± 2.6 1.6 ± 0.5
B 27 41.1 ± 8.9 1.5 ± 0.3 5 6.3 ± 1.3 1.3 ± 0.3
C 24 44.8 ± 4.3 1.9 ± 0.2 5 6.4 ± 2.1 1.3 ± 0.4
D 17 56.5 ± 4.6 3.3 ± 0.3 – – –
E 25 50.8 ± 8.6 2.0 ± 0.3 2.4a 6.2 ± 0.7 2.6 ± 0.3
F 12 44.6 ± 4.3 3.7 ± 0.4 13 24.7 ± 4.3 1.9 ± 0.3
G 9.1 78.3 ± 3.9 8.7 ± 0.4 14 29.7 ± 9.9 2.1 ± 0.7
EMLAb 2.5 52.3 ± 19 20.9 ± 7.6 – – –
EMLAb, c 2.5c 57.8 ± 18.8c 23.1 ± 7.5c – – –
Xylocain 5% 5 22.2 ± 6 4.4 ± 1.2 – – –
Xylocain 2% – – – 2d 3.1 ± 1.0 1.6 ± 0.5
Reprinted with permission from Kreilgaard et al. [36].
a
Near-maximum concentration equals 2.4%.
b
n = 5.
c
Prilocaine-free base.
d
Lidocainehydrochloride.
380 A. Kogan, N. Garti / Advances in Colloid and Interface Science 123–126 (2006) 369–385

suggested that the presence of droplets of the microemulsion of needed to reach steady-state drug penetration rates (lag time). In
only a few nanometers in size had a significant contribution to the addition, there is an increase in the accumulation of drug
percutaneous penetration of lidocaine. quantities in skin layers. The decrease in lag time is an important
The use of a patch was also studied in this work. The results issue, in particular for lidocaine and for topical analgesia in
showed that patches based on a microemulsion had almost two- general, since commercial products require a relatively long ap-
fold higher fluxes than those obtained after applications of the plication time to ease pains [114].
corresponding liquid microemulsion and EMLA cream. (EMLA
is a commercial product containing 1:1 eutectic mixture of 2.5 wt. 2.3.4. Triptolide
% lidocaine and 2.5 wt.% prilocaine in a o/w macroemulsion.) It Triptolide is used as an immunosuppressive, anti-fertility, and
was concluded that patches were a very effective delivery device. anti-cancer drug. In order to decrease the adverse side effects
It was also demonstrated that applying a liquid microemulsion or caused by its toxicity, to prevent hydrolysis that may be caused by
a patch based on a microemulsion significantly shortened the time long-term storage of triptolide in an aqueous environment and be

Fig. 7. Pseudo-ternary phase diagrams of (a) microemulsion system made of isopropyl palmitate/water/tetraglycol/glyceryl oleate and PEG-40 hydrogenated castor oil
at three different cosurfactant/surfactant ratios and (b) microemulsion system made of isopropyl palmitate/water/tetraglycol/glyceryl oleate and PEG-40 stearate, at
three different cosurfactant/surfactant ratios. Reprinted with permission from Sintov and Shapiro [114].
A. Kogan, N. Garti / Advances in Colloid and Interface Science 123–126 (2006) 369–385 381

that the microemulsions containing lower amounts of Tween 80®


and propylene glycol provided higher permeation flux, which
may be attributed to the increased thermodynamic activity of the
drug in these microemulsions. A microemulsion containing
0.025% triptolide, 6% oleic acid, 20% Tween 80®, 10% pro-
pylene glycol, 62.975% water and 1% menthol was found to be
the most suitable for triptolide delivery. The irritation studies on
the skin of rabbits did not show any visible irritation for 7 days. In
comparison, the aqueous solution induced significant erythema
and edema. It may be concluded that a microemulsion is an
effective method for triptolide delivery since it protects it from
degradation, provides prolonged release, and despite the toxicity
of the drug it does not cause irritation [154]. This formulation is
better than the one previously suggested by Mei et al. [111]
consisting of 40% isopropyl myristate, 50% Tween 80®/pro-
Fig. 8. Percutaneous penetration of lidocaine from a microemulsion liquid pylene glycol (5:1), and water, which was a w/o type of
vehicle of system made of isopropyl palmitate/water/tetraglycol/glyceryl oleate microemulsion. In the latest formulation, triptolide is solubilized
and PEG-40 stearate through rat skin in vitro (n = 5) during 3 h. The transport in the outer oily phase and therefore might directly contact the
kinetics (squares) is compared to the penetration profiles of lidocaine from a surface of the skin. From the safety point of view this is not
surfactant mixture in tetraglycol (diamonds), oil-free micellar system (circles),
suitable for long-term use.
and a macroemulsion (triangles). Reprinted with permission from Sintov and
Shapiro [114].
2.3.5. 5-Fluorouracil
5-Fluorouracil is an antineoplastic drug which, on intravenous
promoted by pH values less than 3 or more than 8, a micro- administration, produces severe systemic gastrointestinal, hema-
emulsion system with controlled and prolonged percutaneous tological, neural, cardiac and dermatological effects. Therefore,
delivery of triptolide needs to be developed. The purpose of the transdermal delivery remains the choice route for administration.
studies by Mei et al. [111]and Chen et al. 154] was to investigate Very recently, Gupta et al. [149] investigated in vitro transdermal
microemulsions for transdermal delivery of triptolide. Pseudo- permeation through hairless mouse skin of this hydrophilic drug,
ternary phase diagrams were developed and various microemul- which was encapsulated in AOT/water/isopropylmyristate water-
sion formulations were prepared using oleic acid as an oil, Tween in-oil microemulsions. Incorporation into microemulsions
80® as a surfactant, and propylene glycol as a cosurfactant. The increased the flux of the drug 2–6 fold in comparison to the
droplet size of the microemulsions was characterized by photo- aqueous solution of the drug. The results revealed that
correlation spectroscopy and it varied from 12 to 83 nm. The microemulsions increased the average acyl lipid disorder as
microemulsion formulations that were stored at 37 °C did not well as hydration of the stratum corneum, thus increasing the drug
show any changes in size or phase separation for 6 months. No flux. The skin flux was dependent on the concentration of water
degradation of triptolide was detected either, since the drug was and AOT. Skin toxicity studies indicate that the AOT/water/
incorporated in the oily phase, preventing its contact with the isopropylmyristate ME is safe for the transdermal permeation of
external phase and pH values of the constructed microemulsions this drug [149].
that were in the range of 4–6 inhibited hydrolysis. Permeation
studies from microemulsion vehicles and aqueous solutions 2.3.6. Retinoic acid
containing 20% propylene glycol using mouse skin were also Trotta et al. [112] examined the significance of ion pairing on
performed. A steady increase of triptolide during 24 h was ob- the topical permeation of retinoic acid (RA) using microemul-
served. On the contrary, the permeation flux from the aqueous sions as delivery vehicles. Retinoic acid is used in treating acne
solution decreased significantly after 10 h, which revealed the vulgaris. Both oil-in-water (o/w) and water-in-oil (w/o) micro-
inability of this formulation to provide prolonged delivery of emulsion formulations were prepared using water, isopropyl
triptolide. In a microemulsion the drug can be released from the myristate, lecithin, caprylyl-capryl glucoside and ethanol or 1,2
internal to the external phase and then from the external phase to hexanediol. The mean apparent diameters of these vehicles were
the skin. The release from the internal phase supplements the 20–30 nm. Phenylalanine methyl ester, phenylalanine ethyl
depletion of the drug in the external phase, supplying sustained ester, histidine methyl ester, tryptophan methyl ester, and valine
and controlled release of the triptolide into the skin. Zero-order methyl ester were used as counter ions. The study revealed that
release kinetics was obtained. Despite the great permeation the permeability of the RA from an ethanol–pH 6.4 buffer
enhancing properties of oleic acid, the formulation with higher mixture through polydimethylsiloxane (PDMS) membrane was
oleic acid concentration did not show a significant enhancing significantly increased in presence of the counter ions. These
effect. It was concluded that penetration of oleic acid was results were confirmed by examination of the permeability of
perturbed by the encapsulation of the surfactant and the the RA from the ethanol–pH 6.4 buffer mixture through pig
cosurfactant. Menthol was found to be a suitable permeation skin. In order to clarify whether the enhancement was due to the
enhancer for triptolide loaded microemulsions. It was also found ion pair formation or partly due to direct damage of the skin by
382 A. Kogan, N. Garti / Advances in Colloid and Interface Science 123–126 (2006) 369–385

the amino esters, the same permeability experiments were done components that are well known as enhancers such as ethanol
with a Cortisone, a lipophilic drug that is unable to form an ion [141–144], propylene glycol [93] and phospholipids [106,115,
pair. Since its flux was not significantly increased in presence of 116,119,124,125], turned out to be retarders [145,148]. Thicken-
phenylalanine ethyl ester hydrochloride, the authors concluded ing agents can serve as promoters or as retarders too [132]. In
that RA flux increase might be attributed to the ion pair order to enhance the penetration ability of certain drugs one must
formation. Nevertheless, ion pairing in ethanolic solution could modify its solubility, as well as diffusion and partition coefficients
not help in optimizing RA targeting since drug accumulated/ in each membrane.
drug delivered ratios of 0.6–0.9 and 0.4 μg h cm− 1 10− 3 were It is understood that in many cases the presence of a co-
obtained for ion pair and RA, respectively. In the presence of enhancer to an enhancer optimizes the permeability, such as in
counter ions, the permeabilities of RA from microemulsions the case of 1-menthol addition to ethanol that showed greater
using PDMS membranes decreased. These results were also penetration enhancement of morphine hydrochloride [113]. It is
confirmed using pig skin. w/o microemulsions exhibited the important to note that the combination of several enhancing
lowest accumulation of the drug in the skin. In comparison, components leads to a synergistic effect and a greater enhance-
o/w microemulsions containing counter ions (amino esters) ment effect than is observed for each component alone. In
showed drug accumulation four to five times higher than the addition, the synergy between the iontophoresis and enhancers,
corresponding counter ion-free microemulsions. These kinds such as fatty acids, has resulted in increase of both low and high
of o/w microemulsions are very appealing vehicles for treat- molecular weight drug permeation as well as in decrease of skin
ment of skin diseases and can be used to optimize drug toxicity [85,86,97–99].
targeting without a concomitant increase in systemic absorp- Many enhancers are concentration-dependent; therefore
tion. Despite the same components of these formulations, optimal concentration for effective promotion should be
relative percentages of water, oil, and surfactant lead to different determined. Sometimes when using an ethanol–water vehicle
microstructures that provide different drug permeation and thus the effect of ethanol is concentration dependent, and therefore,
different skin accumulation [112]. can even decrease the permeation [145].
Ion pair creation provides prolonged activity due to the
3. Summary and conclusions formation of a reservoir in topical formulations. This delay ef-
fect leads to increased bioavailability which promotes a
Microemulsions can be formed by numerous oil, surfactant, decrease in daily intake of the drug and side effects.
co-surfactant, and aqueous constituents. The main advantages Many in vitro and in vivo studies have examined the
of microemulsions as vehicles for drug delivery are their high relevance of the microemulsions as a transdermal route for
solubilization capacities for both hydrophilic and hydrophobic administration. Nevertheless, more in vivo studies should be
drugs, their thermodynamic stability, the ease of formation, and conducted to support the in vitro findings. In topical formula-
the relatively low cost of formulation preparation. In some tions, in many cases the irritation issue is not considered. In
cases, the enhanced accumulation of the drug can significantly addition, chronic application consequences should be studied.
help optimize the targeting of the drug without increasing the For instance, for very common enhancers such as oleic acid, the
systemic side effects. benefit of penetration enhancement must be considered very
Microemulsions topically applied were shown to signifi- carefully since immunosuppression of the skin might be affected
cantly increase the cutaneous absorption of the drugs. The [94]. Local and systemic toxicity should be further studied to
vehicles frequently act as penetration enhancers, depending on determine therapeutic benefit/risk ratio and to evaluate the
the oil/surfactant constituents, which involves the risk of in- possible influence on the human body. Since some enhancers
ducing local irritation. The process of skin permeation is very can harm the skin, researchers should examine not only the
complex, but recent studies have improved the knowledge immediate influence of the formulations both in vitro and in
relative to the permeability process and its controllers. The un- vivo, but also examine the skin after chronic application.
derstanding at the molecular level will enable the design of Despite all the complexity, it is possible to predict the
topical formulations with better rationale that will increase the appropriate enhancer based on similarity in physicochemical
bioavailability and decrease the side effects. properties such as molecular weight, hydrophilic/lipophilic
One of the main problems is that after applying a topical classification, and solubility.
formulation on the skin, the formulation composition may The microemulsion structure facilitates drug solubilization in
change. Accordingly, volatile compounds are promptly evapo- greater amounts than in its individual components. A much
rated, small molecules can get absorbed by themselves, and others better understanding of the relationship between the structure of
may interact with the skin in an irreversible manner causing the microemulsion droplets and the nature of the solubilized
irritation and possible permanent damage. Therefore, many drugs drug has been gained from the recent studies, but yet many of
that penetrate the skin sufficiently to produce a therapeutic effect the results are strictly empirical and difficult to explain and even
fail due to the incompatibility of the formulation in which they are more difficult to predict or to forecast. Attention is being paid to
entrapped. In addition, the choice of the formulation components the quantification of the maximum solubilization of a substance
is very complicated. Depending on the structure of the micro- in the concentrate and on its behavior upon dilution. Attempts
emulsion, the character of the drug, and the membrane properties, have been made to determine the locus of the quest molecule in
the same substances can act as enhancers or as retarders. The the core of the microemulsion or at the interface. Understanding
A. Kogan, N. Garti / Advances in Colloid and Interface Science 123–126 (2006) 369–385 383

the locus of solubilization can be useful in protecting the drug mathematical models to predict the behavior of real formula-
and for its sustained or immediate release. The potential in tions. Currently, the use of in vitro models and in vivo animal
controlling the delivery rate has been demonstrated only to models is the only way to predict in vivo human behavior.
some extent and was demonstrated to be dependent on the However, the design of such experiments should be done with
structure and ingredients of the microemulsion. care because human membranes either in vivo or extracted (in
It should be noted that most of the studies conducted with vitro) can behave differently from membranes taken from other
topical formulations are done in vitro/in vivo on animal's skin mammals.
showing greater permeability than that obtained with the same During the last decade much progress has been made in
formulation in humans. It is obvious that both the type of exploring new types of microemulsion vehicles for drug deli-
examined membrane and the kind of vehicle play significant very. More advanced analytical methods are becoming avail-
roles. The same components with varying concentration can able; therefore, it becomes easier to better characterize the
form different vehicles affecting differently the rate of drug correlation between the nature of the microdroplets and the
permeability, and the amount of drug accumulated into the skin. bioavailability of the embedded drug. Despite the enormous
In most cases, the lower the viscosity of the vehicle the faster is amount of information gathered in this field, there are still many
the release [125,112]. questions left unanswered and good deal of work to be done.
There are some cases where the observations do not coincide
with the expectations. Despite expectation of drug permeation Acknowledgement
enhancement due to the formation of a microemulsion vehicle,
in some cases no enhancement occurs due to the lower per- We are very grateful to Dr A. Aserin and Ms. E. Oxman for
centage of the enhancing components in the vehicle in com- their helpful remarks.
parison to other formulations [118]. Therefore, the components,
their concentrations and the form of the vehicle are of great
interest to the pharmaceutical scientist. References
Phase transitions are known to occur in microemulsion
[1] Amidon GL, Lennernas H, Shah VP, Crison JR. Pharm Res 1995;12:413.
microstructures as a result of changes in pH, temperature, dilu- [2] Muller RH, Keck CM. J Biotechnol 2004;113:151.
tion, etc., as well as due to the presence of the guest molecules, [3] Schulman JH, Stoeckenius W, Prince LM. J Phys Chem 1959;63:1677.
the solubilized drugs. Phase transitions were studied only by [4] Danielsson I, Lindman B. Colloids Surf 1981;3:391.
few scientists in past reports, and their effect on drug behavior [5] Hellweg T. Curr Opin Colloid Interface Sci 2002;7:50.
[6] Langevin D. Annu Rev Phys Chem 1992;43:341.
in the microemulsion was not fully clarified. Phase equilibria
[7] Strey R. Colloid Polym Sci 1994;272:1005.
and structural phase transformations within microemulsions in [8] Strey R. Curr Opin Colloid Interface Sci 1996;1:402.
the presence of drugs are of high importance in drug delivery. [9] Bennett KE, Hatfield JC, Davis HT, Macosko CW, Scriven LE. In: Robb
Drugs tend to precipitate and crystallize as large crystals upon ID, editor. Microemulsions. New York: Plenum Press; 1982. p. 65–84.
storage or dilution. These aspects were not considered in the past, [10] Yu ZJ, Neuman RD. Langmuir 1995;11:1081.
[11] Angelico R, Palazzo G, Colafemmina G, Cirkel PA, Giustini M, Ceglie A.
but are relevant issues in relation to sustained or slow delivery.
J Phys Chem B 1998;102:2883.
Therefore, stability examinations of drug-loaded microemulsions [12] Ray S, Moulik SP. Langmuir 1994;10:2511.
should be performed. In view of the fact that the entrapped drug [13] Djordjevic L, Primorac M, Stupar M, Krajisnik D. Int J Pharm
may participate in the formation of the vehicle and, consequently, 2004;271:11.
influence its release, it is very important to investigate the drug [14] Paul BK, Moulik SP. Proc Ind Natl Sci Acad 2000;66A:499.
[15] Feldman Y, Kozlovich N, Nir I, Garti N. Phys Rev E 1995;51:478.
impact on the physicochemical properties of the vehicle.
[16] D'Angelo M, Fioretto D, Onori G, Palmieri L. Phys Rev E 1996;54:993.
The dilution effects on the stability of the microemulsions [17] Moulik SP, Digout LG, Aylward WM, Palepu R. Langmuir 2000;16:
were in most studies neglected and not considered. However, it 3101.
is self-obvious that once the formulation is incorporated into the [18] Feldman Y, Kozlovich N, Nir I, Garti N, Archipov V, Idiyatullin Z, et al.
digestive tracts dilution occurs and the microemulsion in most J Phys Chem 1996;100:3745.
[19] Mukhopadhyay L, Bhattacharya PK, Moulik SP. Colloids Surf 1990;50:
cases decomposes and the drug is released in the uncontrolled
295.
patterns and has the tendency to precipitate. [20] Mukhopadhyay L, Bhattacharya PK, Moulik SP. Ind J Chem 1993;32A:
It was shown by very limited number of studies that micro- 485.
emulsions were efficient drugs protectors from environmental [21] Ray S, Paul S, Moulik SP. J Colloid Interface Sci 1996;183:6.
damages such as oxidative degradation [121], hydrolysis, in- [22] Ray S, Bisal SR, Moulik SP. J Chem Soc Faraday Trans 1993;89:3277.
[23] Moulik SP, De GC, Bhowmik BB, Panda AK. J Phys Chem B
appropriate pH environment [154]. Many more such studies are
1999;103:7122.
required. [24] Hait SK, Moulik SP, Rodgors MP, Burke SE, Palepu R. J Phys Chem B
Modeling the microemulsion phase transition behavior and 2001;105:7145.
the permeation process through the different membranes can be [25] Hait SK, Sanyal A, Moulik SP. J Phys Chem B 2002;106:12642.
a very useful tool to study the change of physicochemical pro- [26] Talmon Y. Colloids Surf 1986;19:237.
[27] Danino D, Kaplun A, Talmon Y, Zana R. ACS Symp Ser 1994;578:105.
perties in case of drug upload. This way, hundreds of ex-
[28] Danino D, Gupta R, Satyavolu J, Talmon Y. J Colloid Interface Sci
periments will be eliminated automatically and will save 2002;249:180.
researchers and the pharmaceutical industry a lot of money [29] Konikoff FM, Danino D, Weihs D, Rubin M, Talmon Y. Hepatology
and precious time. In this area, there is a huge lack of reliable 2000;31:261.
384 A. Kogan, N. Garti / Advances in Colloid and Interface Science 123–126 (2006) 369–385

[30] Ickenstein LM, Arfvidsson MC, Needham D, Mayer LD, Edwards K. [67] Courrier HM, Vandamme ThF, Krafft MP. Colloids Surfaces A
Biochim Biophys Acta Biomembr 2003;1614:135. Physicochem Eng Asp 2004;244:141.
[31] Danino D, Talmon Y, Zana R. Colloids Surf A Physicochem Eng Asp [68] Krafft MP, Chittofrati A, Riess JG. Curr Opin Colloid Interface Sci
2000;169:67. 2003;8:251.
[32] Regev O, Ezrahi S, Aserin A, Garti N, Wachtel E, Kaler EW, et al. [69] D'Cruz OJ, Uckun FM. Contraception 2001;64:113.
Langmuir 1996;12:668. [70] D'Cruz OJ, Yiv SH, Waurzyniak B, Uckun FM. Fertil Steril 2001;75:115.
[33] Jian X, Ganzuo L, Zhiqiang Z, Guowei Z, Kejian J. Colloids Surf A [71] D'Cruz OJ, Uckun FM. Fertil Steril 2003;79:864.
Physicochem Eng Asp 2001;191:269. [72] D'Cruz OJ, Waurzyniak B, Yiv SH, Uckun FM. Contraception
[34] Magdassi S, Ben Moshe M, Talmon Y, Danino D. Colloids Surf A 2000;61:69.
Physicochem Eng Asp 2003;212:1. [73] D'Cruz OJ, Uckun FM. Toxicol Appl Pharmacol 2001;170:104.
[35] Andersson M, Löfroth J. Int J Pharm 2003;257:305. [74] D'Cruz OJ, Uckun FM. J Appl Toxicol 2001;21:317.
[36] Kreilgaard M, Pedersen EJ, Jaroszewski JW. J Control Release [75] D'Cruz OJ, Waurzyniak B, Uckun FM. Toxicology 2002;170:31.
2000;69:421. [76] D'Cruz OJ, Erbeck D, Waurzyniak B, Uckun FM. Toxicology
[37] Caboi F, Lazzari P, Pani L, Monduzzi M. Chem Phys Lipids 2002;179:61.
2005;135:147. [77] D'Cruz OJ, Zhu Z, Yiv SH, Chen CL, Waurzyniak B, Uckun FM.
[38] Lopez F, Cinelli G, Ambrosonea L, Colafemmina G, Ceglie A, Palazzo Contraception 1999;59:319.
G. Colloids Surf A Physicochem Eng Asp 2004;237:49. [78] Ho H, Hsiao CC, Sheu MT. J Pharm Sci 1996;85:138.
[39] Koa CJ, Kob YJ, Kim DM, Park HJ. Colloids Surf A Physicochem Eng [79] Garti N, Aserin A. In: Benita S, editor. Microencapsulation: Methods and
Asp 2003;216:55. Industrial Applications, Culinary and Hospitality Industry Publications
[40] Fanun M, Wachtel E, Antalek B, Aserin A, Garti N. Colloids Surf A Services, 2nd ed, vol. 158. New-York: Taylor and Francis Group, LLC;
Physicochem Eng Asp 2001;180:173. 2005. p. 345–428. chapter 12.
[41] Spernath A, Yaghmur A, Aserin A, Hoffman RE, Garti N. J Agric Food [80] Kreilgaard M. Adv Drug Deliv Rev 2002;54:S77.
Chem 2003;51:2359. [81] Hadgraft J. Int J Pharm 2001;224:1.
[42] Myers D. Surfaces, Interfaces and Colloids. 2nd ed. New York: Wiley; [82] Jadoul A, Doucet J, Durand D, Preat V. J Control Release 1996;42:165.
1999. p. 333. [83] Peltola S, Saarinen-Savolainen P, Kiesvaara J, Suhonen TM, Urtti A. Int J
[43] Kang BK, Chon SK, Kim SH, Jeong SY, Kim MS, Cho SH, et al. Int J Pharm 2003;254:99.
Pharm 2004;286:147. [84] Williams AC, Barry BW. Adv Drug Deliv Rev 2004;56:603.
[44] Porras M, Solans C, Gonzalez C, Martínez A, Guinart A, Gutiérrez JM. [85] Bhatia KS, Gao S, Freeman TP, Singh J. J Pharm Sci 1997;86:111.
Colloids Surf A Physicochem Eng Asp 2004;249:115. [86] Bhatia KS, Singh J. J Pharm Sci 1998;87:462.
[45] Bagger-Jörgensen H, Olsson U, Mortensen K. Langmuir 1997;13:1413. [87] Barry BW. J Control Release 1987;6:85.
[46] Glatter O, Orthaber D, Stradner A, Scherf G, Fanun M, Garti N, et al. [88] Pershing LK, Parry GE, Lambert LD. Pharm Res 1993;10:1745.
J Colloid Interface Sci 2001;241:215. [89] Tanojo H, Junginger HE, Boddé HE. J Control Release 1997;47:31.
[47] Brunner-Popela J, Mittelbach R, Strey R, Schubert KV, Kaler EW, Glatter [90] Aungst BJ, Rogers NJ, Shefter E. Int J Pharm 1986;33:225.
O. J Chem Phys 1999;110:10623. [91] Tanojo H, BosvanGeest A, Bouwstra JA, Junginger HE, Bodde HE.
[48] Lopez-Montilla JC, Herrera-Morales PE, Pandey S, Shah DO. J Dispers Thermochim Acta 1997;293:77.
Sci Technol 2002;23:219. [92] Goodman M, Barry BW. Int J Pharm 1989;57:29.
[49] Dungan SR. In: Solans C, Kunieda H, editors. Microemulsions in foods: [93] Tanojo H, Boelsma E, Junginger HE, Ponec M, Harry E. J Controlled
properties and applications. Industrial Applications of Microemulsions. Release 1999;58:97.
New York: Marcel Dekker; 1997. p. 148. [94] Touitou E, Godin B, Karl Y, Bujanover S, Becker Y. J Control Release
[50] Engström S, Larsson K. In: Kumar P, Mittal KL, editors. Microemulsions 2002;80:1.
in foods, Handbook of Microemulsion Science and Technology. New [95] Rao G, Glikfeld P, Guy RH. Pharm Res 1993;10:1751.
York: Marcel Dekker; 1999. p. 789. [96] Kim A, Green PG, Rao G, Guy RH. Pharm Res 1993;10:1315.
[51] Kawakami K, Yoshikawa T, Moroto Y, Kanaoka E, Takahashi K, [97] Pillai O, Nair V, Panchagnula R. Int J Pharm 2004;269:109.
Nishihara Y, et al. J Control Release 2002;81:65. [98] Nair BV, Panchagnula R. Pharm Res 2003;47:563.
[52] Gasco MR. In: Solans C, Kunieda H, editors. Microemulsions in the [99] Rastogi SK, Singh J. Pharm Dev Technol 2005;10(1):97.
pharmaceutical field: perspectives and applications, in Industrial Applica- [100] Rastogi SK, Singh J. Pharm Dev Technol 2004;9(4):341.
tions of Microemulsions. New York: Marcel Dekker; 1997. p. 98–120. [101] Wang Y, Fan Q, Michniak B. Effects of fatty acids and iontophoresis on
[53] Lawrence MJ. Curr Opin Colloid Interface Sci 1996;1:826. the delivery of midodrine hydrochloride and the structure of human skin.
[54] Podlogar F, Gašperlin M, Tomšič M, Jamnik A, Bešter Rogač M. Int J Pharm Res 2003;20(10):1612–8.
Pharm 2004;276:115. [102] Alvarez-Figueroa MJ, Blanco-Me´ndez J. Int J Pharm 2001;215:57.
[55] Yaghmur L, De Campo O, Glatter ME, Garti N. Phys Chem Chem Phys [103] Baroli B, Lopez-Quintela MA, Delgado-Charro MB, Fadda AM, Blanco-
2004;6:1524. Mendez J. J Control Release 2000;69:209.
[56] Burnett GR, Rees GD, Steytler DC, Robinson BH. Colloids Surf A [104] Peira E, Scolari P, Gasco MR. Int J Pharm 2001;226:47.
Physicochem Eng Asp 2004;250:171. [105] Trotta M, Morel S, Gasco MR. Pharmazie 1997;52:50.
[57] Silas JA, Kaler EW. J Colloid Interface Sci 2001;243:248. [106] Trotta M, Gasco MR, Caputo O, Sancin P. STP Pharma Sci 1994;4:150.
[58] Silas JA, Kaler EW. J Colloid Interface Sci 2003;257:291. [107] Ktistis G, Niopas I. J Pharm Pharmacol 1998;50:413.
[59] Pedersen JS. Curr Opin Colloid Interface Sci 1999;4:190. [108] Schmalfuss U, Neubert R, Wohlrab W. J Control Release 1997;46:279.
[60] Corswant VC, Thoren P, Engström SJ. J Pharm Sci 1998;87:200. [109] Dalmora ME, Dalmora SL, Oliveira AG. Int J Pharm 2001;222:45.
[61] Courrier HM, Krafft MP, Butz N, Porte C, Frossard N, Remy-Kristensen [110] Kantaria S, Rees GD, Lawrence MJ. J Control Release 1999;60:355.
A, et al. Biomaterials 2003;24:689. [111] Mei Z, Chen H, Weng T, Yang Y, Yang X. Eur J Pharm Biopharm
[62] Patel N, Marlow M, Lawrence MJ. J Colloid Interface Sci 2003;258:345. 2003;56:189.
[63] Hiranita T, Nakamura S, Kawachi M, Courrier HM, Vandamme ThF, [112] Trotta M, Ugazio E, Peira E, Pulitano C. J Control Release 2003;86:315.
Krafft MP, et al. J Colloid Interface Sci 2003;265:83. [113] Morimoto Y, Sugibayashi K, Kobayashi D, Shoji H, Yamazaki J, Kimura
[64] Courrier HM, Vandamme TF, Krafft MP, Nakamura S, Shibata O. M. Int J Pharm 1993;91:9.
Colloids Surf A Physicochem Eng Asp 2003;215:33. [114] Sintov AC, Shapiro L. J Control Release 2004;95:173.
[65] Krafft MP, Goldmann M. Curr Opin Colloid Interface Sci 2003;8:243. [115] Gallarate M, Carlotti ME, Trotta M, Bovo S. Int J Pharm 1999;188:233.
[66] Courrier HM, Butz N, Vandamme ThF, Critical reviews in therapeutic [116] Dreher F, Walde P, Walther P, Wehrli E. J Control Release 1997;45:131.
drug carrier systems, 19 (2002) 425. [117] Rhee YS, Choi JG, Park ES, Chi SC. Int J Pharm 2001;228:161.
A. Kogan, N. Garti / Advances in Colloid and Interface Science 123–126 (2006) 369–385 385

[118] Escribano E, Calpena AC, Queralt J, Obach R, Doménech J. Eur J Pharm [139] Delgado-Charro MB, Iglesias-Vilas G, Blanco-Mendez J, Lopez-
Sci 2003;19:203. Quintela MA, Marty JP, Guy RH. Eur J Pharm Biopharm 1997;43:37.
[119] Paolino D, Ventura CA, Nistico S, Puglisiand G, Fresta M. Int J Pharm [140] Ho HO, Chen LC, Chiang HS, Spur BP, Wong PYK, Sheu MT.
2002;244:21. Proceedings 25th International Symposium on Controlled Release of
[120] Špiclin P, Homar M, Zupančič-Valant A, Gašperlin M. Int J Pharm Bioactive Materials, Las Vegas, 21–26 June; 1998. p. 579.
2003;256:65. [141] Friend D, Catz P, Heller J, Reid J, Baker R. J Control Release 1988;7:243.
[121] Špiclin P, Gašperlin M, Kmetec V. Int J Pharm 2001;222:271. [142] Liu P, Kurihara-Bergstrom T, Good WR. Pharm Res 1991;8:938.
[122] Valenta C, Schultz K. J Control Release 2004;95:257. [143] Pershing LK, Lambert LD, Knutson K. Pharm Res 1990;7:170.
[123] Kato A, Ishibashi Y, Miyake Y. J Pharm Pharmacol 1987;39:399. [144] Obata Y, Takayama K, Maitani Y, Machida Y, Nagai T. Int J Pharm
[124] Trotta M, Pattarino F, Gasco MR. Pharm Acta Helv 1996;71:135. 1993;89:191.
[125] Kriwet K, Müller-Goymann CC. Int J Pharm 1995;125:231. [145] Kurihara-Bergstrom T, Knutson K, De Noble LJ, Goates CY. Pharm Res
[126] Kirjavainen M, Mönkkönen J, Saukkosaari M, Valjakka-Koskela R, 1990;7:762.
Kiesvaara J, Urtti A. J Control Release 1999;58:207. [146] Megrab NA, Williams AC, Barry BW. Int J Pharm 1995;116:101.
[127] Friberg SE. J Soc Cosmet Chem 1990;41:155. [147] Thomas NS, Panchagnula R. Pharm Sci 2003;18:71.
[128] Attwood D, Mallon C, Taylor CJ. Int J Pharm 1992;84:R5. [148] Fang JY, Hwang TL, Leu YL. Int J Pharm 2003;250:313.
[129] Sarpotdar PP, Zatz JL. Drug Dev Ind Pharm 1986;12:1625. [149] Gupta RR, Jain SK, Varshney M. Colloids Surf B Biointerfaces
[130] Sarpotdar PP, Zatz JL. J Pharm Sci 1986;75:176. 2005;41:25.
[131] Rigg PC, Barry BW. J Invest Dermatol 1990;94:235. [150] Bolzinger MA, Thevenin MA, Carduner C, Poelman MC. Int J Pharm
[132] López A, Linares F, Cortell C, Herraez M. Int J Pharm 2000;202:133. 1998;176:39.
[133] Fang JY, Yu SY, Wu PC, Huang YB, Tsai YH. Int J Pharm 2001;215:91. [151] Lee PJ, Langer R, Shastri VP. Pharm Res 2003;20:264.
[134] Gašperlin M, Špiclin P. Sci Pharm 2001;69:157. [152] Boltri L, Morel S, Trotta M, Gasco MR. J Pharm Belg 1994;49:315.
[135] Delgado-Charro MB, Iglesias-Vilas G, Blanco-Mendez J, López- [153] Ho HO, Huang MC, Chen LC, Hsia A, Chen KT, Chiang HS, et al. Chin
Quintela MA, Guy RH. Eur J Pharm Biopharm 1997;43:37. Pharm J Taiwan 1998;50:257.
[136] Kreilgaard M. Pharm Res 2001;18:367. [154] Chen H, Chang X, Weng T, Zhao X, Gao Z, Yang Y, et al. J Control
[137] Osborne DW, Ward AJ, O'Neill KJ. J Pharm Pharmacol 1991;43:450. Release 2004;98:427.
[138] Osborne DW, Ward AJ, O'Neill KJ. Drug Dev Ind Pharm 1988;14:1202.

You might also like