Vous êtes sur la page 1sur 57

ARTICLE IN PRESS

Steric Stabilizers for Cubic Phase


Lyotropic Liquid Crystal
Nanodispersions (Cubosomes)
Josephine Y.T. Chong*,,{, Xavier Mulet, Ben J. Boyd*,1,
Calum J. Drummond,{,1
*Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Science, Monash University
(Parkville Campus), Parkville, Victoria, Australia

CSIRO Materials Science and Engineering, Clayton, Victoria, Australia


{
School of Applied Sciences, College of Science, Engineering and Health, RMIT University, Melbourne,
Victoria, Australia
1
Corresponding authors: e-mail address: ben.boyd@monash.edu; calum.drummond@rmit.edu.au

Contents
1. An Introduction to Cubosomes: Self-Assembly of Lipids and Surfactants 2
2. Applications of Cubosomes in Nanotechnology 5
3. Preparation and Characterization of Cubosomes 14
3.1 Preparation of cubosomes 14
3.2 Characterization of cubosomes 15
4. Agents for the Stabilization of Cubosomes 15
4.1 Classes of steric stabilizers for cubosome dispersions 18
5. Future Developments in the Stabilization of Cubosomes 43
6. Conclusion 46
Acknowledgments 46
References 47

Abstract
Lyotropic liquid crystalline nanostructured particles, such as cubosomes, have grown in
popularity as drug delivery systems in the last few years. These systems require steric
stabilizers to maintain colloidal stability in an aqueous medium, with PluronicF127,
a block copolymer, being the most commonly employed stabilizer. However, in recent
years, alternative, more effective stabilizers, as well as rationally designed systems with
opportunities for further biofunctionalization have been reported. The purpose of this
chapter is to collate and collectively interpret studies in the field of steric stabilization of
this important emerging class of nanoparticles for drug and medical imaging agent
delivery.

Advances in Planar Lipid Bilayers and Liposomes # 2015 Elsevier Inc. 1


ISSN 1554-4516 All rights reserved.
http://dx.doi.org/10.1016/bs.adplan.2014.11.001
ARTICLE IN PRESS

2 Josephine Y.T. Chong et al.

1. AN INTRODUCTION TO CUBOSOMES: SELF-ASSEMBLY


OF LIPIDS AND SURFACTANTS
Cubosomes are lipid-based particles, approximately 200 nm in diam-
eter, which can be formed in excess water by dispersing cubic phases that
possess an infinite lipid bilayer draped on a minimal surface. The cubic phase
is often denoted with a Q or V annotation in the literature. The first
observation of a cubic phase in lipidwater systems was reported by Luzzati
et al. in 1960 [1]. The cubic phase possessed the crystallographic space group
with symmetry Ia3d (Q230) and its internal structure was confirmed in 1967
[2,3]. Since, six other cubic phases have been discovered with crystallo-
graphic space group symmetries Pn3m (Q224) [4,5], Im3m (Q229) [68],
Fm3m (Q225) [9], Pm3n (Q223) [4,10], Fd3m (Q227) [7,1114], and P4332
(Q212) [7]. These cubic phases may be classified as bicontinuous cubic
phases, with the exception of Q223 and Q227, which are discontinuous or
discrete micellar cubic phases. Recently, a new lyotropic liquid crystalline
phase was reported based on a three-dimensional (3D) hexagonal close-
packed arrangement of inverse micelles of space group symmetry
P63Immc [15].
Bicontinuous cubic phases have an internal structure based on the peri-
odic minimal surfaces [1618]. In the bicontinuous cubic phase, the lipid
bilayer is arranged in a periodic 3D structure. By mapping the bilayers onto
the surface of infinite periodic minimal surfaces, the mean curvature at any
point on the surface is zero. The mean curvature H is defined as (K1 + K2),
in which K1 and K2 are two principal curvatures. A minimal surface is a sur-
face with H 0 at all points, so that every point on the surface is a balanced
saddle point. There are three types of inverse cubic phases identified in lipid
systems (Fig. 1) [1618]. The first is the Q230 cubic phase, which has an Ia3d
crystallographic space group symmetry and is based on the Schoen gyroid
(G) minimal surface. The second is the Q224 cubic phase, which has a
Pn3m crystallographic space group symmetry and is based on the Schwartz
diamond (D) minimal surface. The third is the Q229 cubic phase, which has
an Im3m crystallographic space group symmetry and is based on the
Schwartz primitive (P) minimal surface. The size of the two distinct inter-
penetrating aqueous water channels in these space group symmetries can be
calculated as they are a function of the surfactant or amphiphilic lipid com-
position and the space group and can range from 4 to 20 nm in diameter,
which is sufficiently large to accommodate certain water-soluble
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 3

Figure 1 Minimal surfaces of the Schoen gyroid (G), Schwartz diamond (D), and
Schwartz primitive (P). Images were generated using Mathematica V.9, based on the
equations obtained from [19].

compounds [17,2022]. The amount of water accommodated in the cubic


phases as well as the size of the water channels increases from G to D to
P minimal surface phases [17].
Cubic phases are typically prepared using lipids. Lipids are often amphi-
philic, which means that they are partly hydrophilic and partly hydrophobic.
Usually the hydrophilic portion is depicted as a head, with the hydropho-
bic portion as the tail in illustrations. Some surfactants or amphiphilic
lipidwater systems are known to form a variety of different lyotropic liquid
crystalline phases. Lyotropic liquid crystals are often termed mesophases,
representing intermediate states of matter between an isotropic liquid and a
solid crystal. The different lipid geometries and their resultant self-assembled
structures that form in the presence of a solvent can be understood using the
critical packing parameter (CPP) concept [23]. CPP is often defined using
Eq. (1)
v
CPP (1)
al
where v is the volume of the hydrophobic tail(s), a is the polar headgroup
area, and l is the length of the hydrophobic chain of the surfactant. The
lamellar phase has a structure with no interfacial curvature (i.e., CPP 1)
because under the CPP concept the surfactants or amphiphilic lipids occupy
an apparently cylindrical space (Fig. 2).
The mesophases can be classified under two categories subdivided into
two topologically distinct regions: type 1/I (normal or oil-in-water
(O/W)) or type 2/II (reverse or water-in-oil (W/O)) [24]. Type 1
mesophases, which include discontinuous micellar cubic, hexagonal, and bi-
continuous cubic phases, are composed of surfactants or amphiphilic lipids that
have an overall geometry that occupy an apparently cone shape space (Fig. 2),
ARTICLE IN PRESS

4 Josephine Y.T. Chong et al.

Figure 2 An illustration of the main types of lyotropic liquid crystal phases depending
on the interface curvature (molecular shape or concentration in water). The mesophases
are denoted as I1, I2 (discrete micellar cubic phase), H1, H2 (hexagonal phase), V1, V2
(bicontinuous cubic phase), and L (lamellar phase).
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 5

whereby CPP < 1. This conical geometry consequently results in the forma-
tion of spheres and cylindrical rods.
In contrast, type 2 mesophases, which include the reverse discontinuous
micellar cubic, reverse hexagonal, and reverse bicontinuous cubic phases,
are composed of surfactants or amphiphilic lipids with an inverse cone shape
space (Fig. 2), whereby CPP > 1. This geometry results in the formation of
inverse spheres and cylindrical rods (Fig. 2). Factors effecting v (volume of
the hydrophobic tail(s)), a (polar headgroup area), and l (length of the hydro-
phobic chain of the surfactant) are summarized by Malmsten [25].
In lyotropic liquid crystal phases, the solvent concentration is a variable
that will partly dictate the self-assembly behavior [26]. In contrast, thermo-
tropic liquid crystal phase transitions are only dependent on temperature and
pressure [26]. The order/sequence of self-assembly phases that are typically
observed as the surfactant/lipid concentration increases in an amphiphilic
lipid/water system is micelles, micellar cubic, hexagonal, bicontinuous
cubic, lamellar, and their respective reverse phases, as illustrated in Fig. 2
[2729].
Materials which have been reported to form cubic phase systems have
been listed by Fontell in 1990 [30]. These materials include anionic and cat-
ionic soaps, zwitterionic and nonionic surfactants, and amphiphilic lipids
of biological origin, such as monoglycerides, sphingolipids, and phospho-
lipids, and also galactolipids, glycolipids, and tetra ether lipids [30]. Since
then reviews have reported that inverse bicontinuous cubic phases have
been observed for many different types of lipids, including mono-
acylglycerides, glycolipids, urea, and urea-like amphiphiles and mono-
ethanolamides [3133]. The amphiphilic lipids most commonly used in
lipid lyotropic liquid crystal research have been glyceryl monooleate
(GMO), a food emulsifier, and phytantriol, a cosmetic ingredient (Fig. 3)
due to their low cost, ease of availability, and potential biocompatibility
based on their history of use in other fields.

2. APPLICATIONS OF CUBOSOMES IN
NANOTECHNOLOGY
The aforementioned lyotropic liquid crystal systems are thermody-
namically stable and for the case of the reversed phases or lamellar phase
can be dispersed into smaller particles that retain the complex internal nano-
structure in the presence of a stabilizer. Dispersions of these bulk parent
ARTICLE IN PRESS

6 Josephine Y.T. Chong et al.

Figure 3 The chemical structures of amphiphilic lipids: (A) phytantriol and (B) glycerol
monoleate.

phases have been given the suffix -osome. For example, dispersions from
the lamellar, hexagonal, and cubic phases are known as liposomes,
hexosomes, and cubosomes, respectively. Liposomes have been exten-
sively used as drug delivery vehicles with 12 clinically approved liposomal
drug formulations currently on the market and 22 liposomal drugs undergo-
ing clinical trials [34,35]. Particles based on other lyotropic liquid crystalline
structures such as cubosomes (inverse bicontinuous cubic phase, Fig. 4) and
hexosomes (inverse hexagonal phase) are also being developed as potential
drug delivery systems [37,38].
The key advantages of these nanostructured particles compared to lipo-
somes include their ordered 3D mesoporous internal structure with poten-
tial for controllable release and their increased lipid volume fraction per
particle, which provides a large lipophilic area for containing poorly
water-soluble lipophilic therapeutics [39,40]. The lyotropic liquid crystal-
line structure and dimensions of the phase, specifically the water channels,
determine the release rate of drugs from within the lyotropic liquid crystal
phase [41,42]. The phases can accommodate molecules of varying proper-
ties [43,44]. A recent study by Zabara and Mezzenga reported the controlled
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 7

Figure 4 An illustration of the particle morphology and applications of the cubosome.


The cryo-FESEM images display the 3D particle morphology of cubosomes and are adapted
from Rizwan et al. [36].

release of encapsulated protein within Q224 cubic nanostructured particles


by doping the mesophase with a hydration-modulating agent that causes
an increase in the diameter of the water channels [45]. In a similar manner,
the swelling of the aqueous domain can also be manipulated with polysac-
charides, as illustrated by Mezzenga et al. [46]. The possibility of controlled
drug release from nonlamellar lyotropic liquid crystalline systems is one
of the main attractive features for using these systems for drug delivery.
Examples of therapeutics which have been incorporated into cubosomes
for investigating their potential as drug delivery systems are listed in
Table 1. Cubosomes have been studied for administration via ocular, der-
mal, intradermal, mucosal, intranasal, oral, percutaneous, intraperitoneal,
intratympanic, and intravenous routes, as presented in Table 1. In addition,
studies have also been performed on the interaction of nanostructured par-
ticles with model and cell membranes [102] and blood components [103],
for their biocompatibility within the body as effective drug delivery systems.
Table 1 A table listing examples of the therapeutics and lipid composition of drug-loaded cubosomes as drug nanocarriers
Administration
Bioactive molecule (peptide, drug) Matrix constituent Stabilizer route (if applicable) References
Silver sulfadiazine Glyceryl monooleate PluronicF127 Topical [47]
Hinokitiol Glyceryl monooleate PluronicF127 Dermal [48]

Soluble extracts of Korean barberry Glyceryl monooleate Pluronic F127 Dermal [49]

5(6)-Tetramethylcarboxy rhodamine-labeled Phytantriol Pluronic F127 Transcutaneous [50]
OVA257264

ARTICLE IN PRESS
Herbal extracts (obtained from Poria cocos, Glyceryl monooleate PluronicF127 Skin (for hair [51]
Thuja orientalis, Espinosilla, Lycium chinense Mill, regrowth)
Coix lacryma-jobi, and Polygonum multiflorum
Thunberg)
Alpha lipoic acid Myverol 18-99 PluronicF127 Skin [52]

Saponin adjuvant Quil A and Phytantriol Pluronic F127 In vitro (skin) [53]
monophosphoryl lipid A
Triclosan Glyceryl monooleate PluronicF127 In vitro (skin) [54]

KIOM-MA-128 (water-soluble extract) Glyceryl monooleate Pluronic F127 In vitro (skin) [55]

Houttuynia cordata (water-soluble extract) Glyceryl monooleate Pluronic F127 In vitro (skin) [56]

Tacrolimus Glyceryl monooleate Pluronic F127 Intradermal [57]
Clotrimazole Glyceryl monooleate PluronicF127 Mucosal [58]
Dexamethasone Glyceryl monooleate PluronicF127 Ocular [59]

Flurbiprofen Glyceryl monooleate Pluronic F127 Ocular [60]

Cyclosporine A Glyceryl monooleate Pluronic F127 Ocular [61]

5-FC oleyl carbamate (pro-drug) 5-FC oleyl carbamate Pluronic F127 Oral [62]

Curcumin/piperine Phytantriol Pluronic F127 Oral [63]

ARTICLE IN PRESS
Amphotericin B Phytantriol Pluronic F127 Oral [64,65]

Cinnarizine Phytantriol or glyceryl Pluronic F127 Oral [66]
monooleate
Cyclosporine A Glyceryl monooleate PluronicF127 Oral [67]

Ibuprofen Phytantriol Pluronic F127 Oral [68]

Insulin Glyceryl monooleate Pluronic F127 Oral [69]

20(S)-protopanaxadiol/piperine Glyceryl monooleate Pluronic F127 Oral [70,71]
Simvastatin Glyceryl monooleate PluronicF127 Oral [72]

Omapatrilat Glyceryl monooleate Pluronic F127 Oral [73]

Omapatrilat Glyceryl monooleate Pluronic F68 Oral [73]
Coenzyme Q10 Glyceryl monooleate PluronicF127 Oral [74]

Coenzyme Q10 Glyceryl monooleate Pluronic F108 Oral [74]

Coenzyme Q10 Glyceryl monooleate Pluronic F68 Oral [74]
Continued
Table 1 A table listing examples of the therapeutics and lipid composition of drug-loaded cubosomes as drug nanocarrierscont'd
Administration
Bioactive molecule (peptide, drug) Matrix constituent Stabilizer route (if applicable) References
Coenzyme Q10 Phytantriol PluronicF127 Oral [74]
Coenzyme Q10 Phytantriol PluronicF108 Oral [74]

Coenzyme Q10 Phytantriol Pluronic F68 Oral [74]
Paclitaxel Soy Polysorbate 80 Oral [75]
phosphatidylcholine/

ARTICLE IN PRESS
glycerol dioleate
Baicalin/KiOM-C Glyceryl monooleate PluronicF127 In vitro (small [76]
intestine
adsorption)
S-164 (water-soluble extract) Glyceryl monooleate PluronicF127 In vitro (small [77]
intestine
adsorption)
Odorranalectin/streptavidin Glyceryl monooleate PluronicF127 Intranasal [78]

Indomethacin Glyceryl monooleate Pluronic F127 Percutaneous [79]

Bromocriptine Glyceryl monooleate Pluronic F127 Intraperitoneal [80]
Adjuvants imiquimod and monophosphoryl Phytantriol PluronicF127 Intravenous [81]
lipid A
Fluorescein isothiocyanate-ovalbumin/Quil Phytantriol PluronicF127 Intravenous [82]
A
Paclitaxel Glyceryl monooleate PluronicF127/ Intravenous [83]
mPEG2kDSPE
Propofol Soy Polysorbate 80 Intravenous [84]
phosphatidylcholine/
glycerol dioleate
Somatostatin Soy Polysorbate 80 Intravenous [85]
phosphatidylcholine/

ARTICLE IN PRESS
glycerol dioleate
Earthworm fibrinolytic enzyme (protein) Glyceryl monooleate/ PluronicF127 Intratympanic [86]
propylene glycol
Ovalbumin Phytantriol or glyceryl PluronicF127 [87]
monooleate
-Chymotrypsinogen A (protein) Glyceryl monooleate MO-PEG2000 [88]
[poly(ethylene
glycol) monooleate]
Carrier-free human recombinant brain- Glyceryl monooleate/ D--Tocopherol [89]
derived neurotrophic factor eicosapentaenoic acid poly(ethylene
glycol) 1000
succinate (V1000)
Annexin V (protein) Phytantriol PluronicF127 [90]

Curcumin Glyceryl monooleate Pluronic F127 [91]
Continued
Table 1 A table listing examples of the therapeutics and lipid composition of drug-loaded cubosomes as drug nanocarrierscont'd
Administration
Bioactive molecule (peptide, drug) Matrix constituent Stabilizer route (if applicable) References
Quercetin Glyceryl monooleate PluronicF108 [92]
Camptothecin Glyceryl monooleate PluronicF108 [93]
and folic acid
Dacarbazine Glyceryl monooleate PluronicF127 [9496]

Carbamazepine (CBZ), coenzyme Q10 Glyceryl monooleate Pluronic F127 [97]

ARTICLE IN PRESS
(CoQ10), cholesterol (Chl, sterol),
phytosterols (PSs, plant sterols)
Diazepam, griseofulvin, propofol, rifampicin Myverol 18-99 PluronicF127 [98]
50-Deoxy-5-fluoro-N4- 50- PluronicF127 [99]
(phytanyloxycarbonyl) cytidine (phytanyl pro- Deoxy-5-fluoro-N4-
drug analogue of capecitabine) (phytanyloxycarbonyl)
cytidine
Hydrocortisone Phytantriol PluronicF127 [39,100]

Atropine Phytantriol Pluronic F127 [39,100]

Transretinol Phytantriol Pluronic F127 [39,100]
Diazepam Phytantriol PluronicF127 [39,100]

Prednisolone Phytantriol Pluronic F127 [39,100]

Dexamethasone Phytantriol Pluronic F127 [39,100]
Progesterone Phytantriol PluronicF127 [39,100]

Haloperidol Phytantriol Pluronic F127 [39,100]

Levofloxacin Phytantriol Pluronic F127 [39,100]
Indometacin Phytantriol PluronicF127 [39,100]

Hydrocortisone Myverol 18-99K Pluronic F127 [39,100]

Atropine Myverol 18-99K Pluronic F127 [39,100]
PluronicF127

ARTICLE IN PRESS
Transretinol Myverol 18-99K [39,100]

Diazepam Myverol 18-99K Pluronic F127 [39,100]

Prednisolone Myverol 18-99K Pluronic F127 [39,100]

Dexamethasone Myverol 18-99K Pluronic F127 [39,100]

Progesterone Myverol 18-99K Pluronic F127 [39,100]
Haloperidol Myverol 18-99K PluronicF127 [39,100]

Levofloxacin Myverol 18-99K Pluronic F127 [39,100]

Indometacin Myverol 18-99K Pluronic F127 [39,100]
DOPURu (amphiphilic ruthenium-based 1,2-Dioleoyl-sn- [101]
molecule) glycero-3-
phosphocholine
(DOPC) and 1,2-
dioleoyl-sn-glycero-3-
phosphoethanolamine
(DOPE)
ARTICLE IN PRESS

14 Josephine Y.T. Chong et al.

Recent reviews by Rizwan et al. [104] and Conn and Drummond [33]
have collected examples where lyotropic liquid crystalline nanostructured
particles accommodate biologically active molecules such as vitamins,
enzymes, and other proteins, as well as crystallizing membrane proteins,
which have important application for membrane protein crystallization, bio-
sensors, biofuel applications, as well as in drug delivery. Owing to the high-
surface area of the internal mesophase structure (up to 400 m2/g) [105], the
cubic phase can be used to incorporate these biologically active molecules
(e.g., globular proteins), which have similar dimensions to the water chan-
nels in the bicontinuous cubic phases [89].
In other recent developments, cubosomes are also being investigated for
the containment of contrast agents for medical imaging applications
[106108], and capabilities as a cell-free bio-sensing platform [109]. Apart
from drug delivery and biomedical applications, the use and application
of lyotropic liquid crystalline nanostructured particles is also relevant within
the food industry (e.g., solubilization of food bioactives within lyotropic liq-
uid crystalline mesophases) [110,111] and agriculture industry (e.g., delivery
of plant agrochemicals) [112]. Therefore, any research into the colloidal
stability and retention of internal structure of nanostructured particles is
relevant to several research fields.

3. PREPARATION AND CHARACTERIZATION OF


CUBOSOMES
3.1. Preparation of cubosomes
For the preparation of any cubosome dispersion, there are three main com-
ponents, required; these are (i) lipid, (ii) steric stabilizer, and (iii) an aqueous
solution, which is typically water or a buffer system. Boyd et al. and Guo et al.
have extensively reviewed and listed established preparation methodologies
used for producing cubosome dispersions [113,114]. In summary, there are
two main approaches typically used to produce cubosome dispersions:
(i) top-down and (ii) bottom-up approaches. The top-down approach
requires the dispersion of an extremely viscous lipid or bulk cubic phase into
the aqueous solution usually using sonication. The high energy created by
sonication can also be produced by high-pressure homogenization and
shearing. Since the report of this approach by Ljusberg-Wahren in 1996
[115], high-pressure homogenization and sonication are still the most fre-
quently used techniques in the preparation of cubosomes [113,114]. This
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 15

is probably because it is a rapid method for forming uniform dispersions with


a particle size below 200 nm and low polydispersity.
The bottom-up approach is one in which a single phase solution is
diluted into a two phase regime of cubosomes coexisting with an excess
aqueous phase. An advantage of this method compared to the top-down
approach is that it requires less energy input to generate dispersions. The
key factor in the bottom-up approach is the presence of a hydrotrope
(e.g., chloroform, ethanol), which is miscible with water-insoluble lipids
to create single phase liquid precursors and prevents the formation of
lyotropic liquid crystals at high concentration.

3.2. Characterization of cubosomes


In order to verify that the dispersed particles prepared using the desired prep-
aration technique are indeed cubosomes, characterization techniques,
such as visual assessment, dynamic light scattering, cross-polarized light
microscopy, small angle X-ray scattering (SAXS), and cryo-transmission
electron microscopy (cryoTEM), are employed. Although these may not
be the only characterization techniques used for cubosome analysis, these
are the major techniques used in the literature to date. These techniques
have been well established and used with great success in distinguishing dif-
ferent aspects of the lyotropic liquid crystalline nanostructured particle, such
as particle size and lyotropic liquid crystal phase/nanostructure type.
Quantifying the stability of dispersions (i.e., cubosome and hexosome
dispersions) was initially performed using a stability analyzer, the
LUMiFuge, which is a specialist instrument designed to quantify stability
principally for emulsion or other colloidal systems [116,117]. This method
only allowed one sample to be measured at any given time. However,
recently an accelerated stability assay has been developed enabling high-
throughput qualitative analysis for multiple samples [118].

4. AGENTS FOR THE STABILIZATION OF CUBOSOMES


Although the internal mesophase of lyotropic liquid crystalline parti-
cles is thermodynamically stable, cubosomes are often less colloidally stable
than regular emulsions in an aqueous solution. Therefore, a steric stabilizer is
required to retain colloidal stability [119]. The van der Waals forces driving
flocculation, and consequent coalescence and creaming of typical O/W
ARTICLE IN PRESS

16 Josephine Y.T. Chong et al.

emulsion systems, are destabilizing factors of the colloidal nature of cub-


osome dispersions.
An ideal stabilizer for cubosomes prevents unfavorable interaction
between the hydrophobic domains on encounter between particles, without
causing disruption to the inner cubic structure. Typically, this requires the
formation of a steric and/or electrostatic-repulsive barrier between
approaching particles. Stabilizers are therefore considered an essential com-
ponent in liquid crystalline nanostructured particle preparation. A further ele-
ment of consideration for cubosomes is their high internal interfacial area,
which may lead to stabilizer sequestration within the liquid crystalline nano-
structure, which will reduce its contribution to colloidal stability [120].
Although a charged stabilizer can be applied to provide an electrostatic barrier
to the flocculation of cubosomes, it is more common to utilize a steric stabi-
lizer, as charged surfactant molecules have a high propensity to disrupt the
internal phase structure of cubosomes [121]. Charged nanostructured parti-
cles, such as negatively charged liposomes, have also been reported to have a
shorter half-life in the blood than neutral liposomes [122,123] and positively
charged liposomes were found to be toxic and quickly removed from systemic
circulation [124]. It was reported that the surface charge (i.e., positive or neg-
ative) is a key determinant in complement-system activation by liposomes for
both human and guinea-pig serum [125,126].
Stealth and steric hindrance are provided by polymers that have been
reported to confer repellency to surfaces. They typically share similar prop-
erties, such as high hydrophilicity, the presence of hydrogen bond acceptors
but absence of hydrogen bond donors, and electrical neutrality [127,128].
Polyethylene glycol (PEG) also known as polyethylene oxide (PEO) fits this
profile, being an uncharged, hydrophilic polymer that is soluble in water.
Due to its low toxicity and immunogenicity, PEG is considered to be the
chemical moiety that yields the most effective steric repulsion barrier while
improving the pharmacokinetics and pharmacodynamics of nanoscale drug
delivery systems (e.g., stealth liposomes) [129131]. PEG has been shown to
be able to form a stealth corona around liposomes, significantly reducing the
rapid uptake of intravenously injected particulate drug carriers by cells of the
mononuclear phagocyte system (MPS) [132,133]. It has been demonstrated
theoretically [134137] and experimentally [138143] that protein
repellence of PEG coatings depends on both chain length and chain density,
which jointly determine the thickness of the adlayer [127].
Steric stabilization of nanostructured particles is also highly dependent on
the stabilizer concentration. Low stabilizer surface coverage often results in a
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 17

mushroom surface conformation of the stabilizer on the surface of the par-


ticle (Fig. 5). Increasing the density of PEG chains on the surface of the particle
often results in a brush conformation of the stabilizing polymer, which is
more effective in stabilization and protein repellence [144]. However, little
is currently known on the optimal concentration of stabilizer for the prepara-
tion of cubosome dispersions, although 10% w/w is the standard concentra-
tion often used in their preparation, as it produces an aggregate-free dispersion
[113]. Besides PEG length and concentration, it was also established by
Thies in 1976 that stabilizer composition/structure and establishing a favorable
balance between the anchoring unit (i.e., hydrophobic head) and extending
unit (i.e., hydrophilic tail) were as equally important in achieving optimum
stability performance when using copolymers as steric stabilizers [145].
This highlights the importance of assessing various copolymer structures as
stabilizers for lyotropic liquid crystalline nanostructured particles, as it is

Figure 5 An illustration of some of the factors affecting steric hindrance between nano-
structured particles: (A) the concentration of steric stabilizer used and (B) PEG length in
steric stabilizer.
ARTICLE IN PRESS

18 Josephine Y.T. Chong et al.

possible that better copolymer structure configurations which will enable


more effective stabilization have not yet been explored.
Accordingly, steric hindrance and provision of stealth onto a nanostruc-
tured particle is also dependent on the concentration of steric stabilizer
applied and the PEG length in the steric stabilizer (Fig. 5). In 1954, Heller
and Pugh found that increasing the PEG length and concentration on their
gold sols increased their stability [146]. This was later confirmed by Lee et al.
in 1989, using a range of Poloxamers (PluronicL63, P65, P105, F68,
F88, F108) and Poloxamine (Tetronic908), with increasing hydrophilic
PEG chain lengths on polystyrene beads. Whereby it was also found that
increasing the PEG length also increased the stability of the beads, with
PluronicF108 and Tetronic908 being the best stabilizers from the series
[147]. PluronicF108 was also used on polystyrene beads in 1998 achieving
similar stability results [148]. Short PEG lengths on steric stabilizers may be
unfavorable because there is insufficient distance created between neighbor-
ing particles. For this reason, typically the longer the PEG chain (e.g.,
PluronicF108) the better its effectiveness at providing stabilization to a
hydrophobic particle. However, whilst PEG is regarded as an ideal hydro-
philic domain for steric stabilizers for lyotropic liquid crystalline nanostruc-
tured particles, little is known about the ideal PEG chain length for
establishing maximum steric stabilization effectiveness onto cubosomes.

4.1. Classes of steric stabilizers for cubosome dispersions


The steric stabilizers which have been reported in the literature for preparing
cubosome dispersions have been categorized into four groups: (i) amphiphilic
block copolymers (i.e., Poloxamer and Poloxamine), (ii) PEGylated
lipids (e.g., GMO-PEG, vitamin E TPGS, Tween, 1,2-dimyristoyl-
sn-glycero-3-phosphoethanolamine (DMPE)-PEG, DOPE-PEG, 1,2-
distearoylphosphatidylethanolamine (DSPE)-PEG), (iii) designer/customized
lipidcopolymer series (e.g., poly(octadecyl acrylate)-block-poly(PEG methyl
ether acrylate) (P(ODA)-b-P(PEGA-OMe)) series), and (iv) alternative steric
stabilizers (e.g., bile salts, proteins, polysaccharide polymers, vitamins, and
nanoparticles). These are summarized in Tables 24 and are described in more
detail below.

4.1.1 Amphiphilic block copolymers


Two classes of amphiphilic block copolymers have been reported as
steric stabilizers for cubosomes to date. These are the Poloxamer and
Poloxamine.
Table 2 Amphiphilic block copolymers used as steric stabilizers for cubosomes reported in the general literature
Space group of inner
Stabilizer Lipid matrix constituent structure References
Poloxamer
PluronicF127 Phytantriol Pn3m (Q224) [36,39,53,6466,68,
81,82,87,90,108,
109,118,120,
149160]

ARTICLE IN PRESS
PluronicF127 Phytantriol/dipalmitoyl phosphatidylserine (DPPS) Pn3m (Q224) or [154]
Im3m (Q229)
PluronicF127 1-O-(5,9,13,17-tetramethyloctadecanoyl)erythritol Pn3m (Q224) and [161]
(EROCO C22) Im3m (Q229)
PluronicF127 1-O-(5,9,13,17-tetramethyloctadecyl)--D- Pn3m (Q224) and [161163]
xylopyranoside (-XP) Im3m (Q229)
PluronicF127 glycolipid 1-O-phytanyl--D-xyloside (-XP) Pn3m (Q224) and [163]
Im3m (Q229)
PluronicF127 5-FC oleyl carbamate Pn3m (Q224) and [62]
Ia3d (Q230)
PluronicF127 50-Deoxy-5-fluoro-N4-(phytanyloxycarbonyl) Pn3m (Q224) and [99]
cytidine Ia3d (Q230)
PluronicF127 Monolinolein (MLO) Pn3m (Q224) at [164]
25 C
Continued
Table 2 Amphiphilic block copolymers used as steric stabilizers for cubosomes reported in the general literaturecont'd
Space group of inner
Stabilizer Lipid matrix constituent structure References
PluronicF127 Monolinolein (MLO)/oil Pn3m (Q224) [165168]
Fd3m (Q227) [165168]
224
Pluronic F127 Monolinolein (MLO)/diglycerol monooleate Pn3m (Q ) or [169]
(DGMO) or soybean PC/oil Im3m (Q229)
PluronicF127 Monoelaidin Im3m (Q229) [170]

ARTICLE IN PRESS
PluronicF127 Myverol 18-99K Pn3m (Q224) or [39,98,108,152,171]
Im3m (Q229)
PluronicF127 RYLO MG 19 Pn3m (Q224) and [96,171]
Im3m (Q229)
PluronicF127 Glyceryl monooleate Pn3m (Q224) or [48,49,51,54,55,
Im3m (Q229) 5761,66,67,
6973,7680,87,
91,9497,102,111,
119,152,155,159,
160,170,172188]
PluronicF127 Glyceryl monooleate/propylene glycol Pn3m (Q224) and [86]
Im3m (Q229)
PluronicF127 Glyceryl monooleate/soya phospholipids Im3m (Q229) [189]
229
Pluronic F127 Glyceryl monooleate/oil Im3m (Q ) [190]
Fd3m (Q227) [190]
PluronicF127 Glyceryl monooleate/1-glycerol monooleyl ether Pn3m (Q224) and [191]
(GME) Im3m (Q229)
PluronicF127 Dimodan U/J (96% monoglycerides: 62% linoleate Pn3m (Q224) and [192]
and 25% oleate)/tetradecane (oil) Im3m (Q229)
PluronicF127 Dimodan U/J (96% monoglycerides: 62% linoleate Pn3m (Q224) and [193195]
and 25% oleate) Im3m (Q229)
PluronicF127/mPEG2kDSPE Im3m (Q229)

ARTICLE IN PRESS
Glyceryl monooleate [83]
PluronicF127/mPEG350DSPE Phytantriol Pn3m (Q224), Im3m [196]
(Q229)
PluronicF127/mPEG750DSPE Phytantriol Pn3m (Q224) and [196]
Im3m (Q229)
PluronicF127/mPEG2kDSPE Phytantriol Pn3m (Q224) and [196]
Im3m (Q229)
PluronicF127/-casein Phytantriol Pn3m (Q224) [152]
mixture
PluronicF108 Phytantriol Pn3m (Q224) [118,160]
224
Pluronic F108 Glyceryl monooleate Pn3m (Q ) [92,93,160]
224
Pluronic F87 Phytantriol Pn3m (Q ) [160]
PluronicF87 Glyceryl monooleate Im3m (Q229) [160]
224
Pluronic F68 Phytantriol Pn3m (Q ) [160]
Continued
Table 2 Amphiphilic block copolymers used as steric stabilizers for cubosomes reported in the general literaturecont'd
Space group of inner
Stabilizer Lipid matrix constituent structure References
PluronicF68 Glyceryl monooleate Cubosome [73,160]
PluronicF68 Myverol 18-99K Cubosome [113]
229
Pluronic P123 Phytantriol Im3m (Q ) [160]
224
Pluronic P105 Phytantriol Pn3m (Q ) [160]
PluronicP105 Glyceryl monooleate Im3m (Q229) [160]

ARTICLE IN PRESS
229
Pluronic P104 Phytantriol Im3m (Q ) [160]
229
Pluronic P104 Glyceryl monooleate Im3m (Q ) [160]
224
Pluronic P84 Phytantriol Pn3m (Q ) and [160]
Im3m (Q229)
Polaxamine
Poloxamine 908 Myverol 18-99K Cubosome [113]
Poloxamine 908/ Myverol 18-99K Cubosome [113]
PluronicF127 combinations
Table 3 PEGylated lipid copolymers used as steric stabilizers for cubosomes reported in the general literature
PEG PEG Space group of inner
Stabilizer MW units Lipid matrix constituent structure References
PEGylated lipid
1,2-Dimyristoyl-sn- 550 12 Dielaidoylphosphatidylethanola Im3m (Q229) [197,198]
glycero-3-phosphoethanolamine-N- mine (DEPE)
PEG (DMPE-PEG550)
PEGylated monoolein (MO-PEG660) 660 15 1,2- Ia3d (Q230) and Pn3m [199]

ARTICLE IN PRESS
Dioleoylphosphatidylethanolami (Q224)
ne (DOPE)
1,2-Distearorylphosphatidylethanol 750 17 1,2- Cubosome [200]
amine-PEG (DSPE-PEG750) Dioleoylphosphatidylethanolami
ne (DOPE)
Polyoxyethylene (20) sorbitan 900 20 Myverol 18-99K Cubosome [113]
monopalmitate (Tween40)
Polyoxyethylene (20) sorbitan 900 20 Glyceryl monooleate Cubosome [102]
monooleate (Tween80)
Polyoxyethylene (20) sorbitan 900 20 Soy phosphatidylcholine/ Cubosome [75,84,85]
monooleate (Tween80) glycerol dioleate
Polyoxyethylene (20) sorbitan 900 20 Soy PE (L-- Cubosome [38]
monooleate (Tween80) phosphatidylethanolamine)
D-alpha-Tocopheryl PEO1000 succinate 1000 22 Phytantriol Im3m (Q229) [38]
(vitamin E TPGS)
Continued
Table 3 PEGylated lipid copolymers used as steric stabilizers for cubosomes reported in the general literaturecont'd
PEG PEG Space group of inner
Stabilizer MW units Lipid matrix constituent structure References
229
PEG1K20PHYT30 1000 22 Phytantriol Im3m (Q ) [201]
PEG1K25PHYT25 1000 22 Phytantriol Im3m (Q229) [201]
224
PEG1K30PHYT20 1000 22 Phytantriol Pn3m (Q ) and Im3m [201]
(Q229)
PEG1K40PHYT10 1000 22 Phytantriol Pn3m (Q224) and Im3m [201]
(Q229)

ARTICLE IN PRESS
1,3-Didodecyloxy-propane-2-ol-PEG 1300 30 GMO (RYLO MG 90) Ia3d (Q230) [202]
(DDP(EO)30)
PEG-40-stearate 1800 40 Phytantriol Im3m (Q229) [203]
1,2-Dioleoylphosphatidylethanolami 2000 45 Glyceryl monooleate and Cubosome [204]
ne-PEG (DOPE-PEG) cis-5,8,11,14,17-
eicosapentaenoic
acid (20:5, EPA)
PEGylated monoolein 2000 45 Glyceryl monooleate Pn3m (Q224) or Im3m [88]
(MO-PEG2000) (Q229)
1,2-Distearorylphosphatidylethanol 2000 45 1,2- Cubosome [200,205]
amine-PEG (DSPE-PEG2000) Dioleoylphosphatidylethanolami
ne (DOPE)
1,2-distearoylphosphatidylethanol 2000 45 Soy phosphatidyl choline (SPC) Cubosome [205]
amine-PEG (DSPE-PEG2000) and glycerol dioleate (GDO)
PEG-45-stearate 2000 45 Phytantriol Im3m (Q229) [203]
229
PEG2K10PHYT40 2000 45 Phytantriol Im3m (Q ) [201]
PEG2K20PHYT30 2000 45 Phytantriol Pn3m (Q224) and Im3m [201]
(Q229)

ARTICLE IN PRESS
PEG2K25PHYT25 2000 45 Phytantriol Im3m (Q229) [201]
229
PEG2K30PHYT20 2000 45 Phytantriol Im3m (Q ) [201]
PEG2K40PHYT10 2000 45 Phytantriol Im3m (Q229) [201]
229
PEG-50-stearate 2200 50 Phytantriol Im3m (Q ) [203]
229
1,3-Didodecyloxy-propane-2-ol-PEG 2300 52 GMO (RYLO MG 90) Im3m (Q ) and Ia3d [202,206]
(DDP(EO)52) (Q230) (coexisting with L3
phase)
PEG-55-stearate 2400 55 Phytantriol Im3m (Q229) [203]
PEG3K10PHYT40 3000 68 Phytantriol Pn3m (Q224) and Im3m [201]
(Q229)
PEG3K20PHYT30 3000 68 Phytantriol Pn3m (Q224) [201]
224
PEG3K25PHYT25 3000 68 Phytantriol Pn3m (Q ) [201]
PEG3K30PHYT20 3000 68 Phytantriol Pn3m (Q224) [201]
Continued
Table 3 PEGylated lipid copolymers used as steric stabilizers for cubosomes reported in the general literaturecont'd
PEG PEG Space group of inner
Stabilizer MW units Lipid matrix constituent structure References
224
PEG3K40PHYT10 3000 68 Phytantriol Pn3m (Q ) [201]
PEG4K10PHYT40 4000 90 Phytantriol Pn3m (Q224) [201]
224
PEG4K20PHYT30 4000 90 Phytantriol Pn3m (Q ) [201]
224
PEG4K25PHYT25 4000 90 Phytantriol Pn3m (Q ) [201]
PEG4K30PHYT20 4000 90 Phytantriol Pn3m (Q224) [201]

ARTICLE IN PRESS
224
PEG4K40PHYT10 4000 90 Phytantriol Pn3m (Q ) [201]
229
1,3-Didodecyloxy-propane-2-ol-PEG 4100 92 GMO (RYLO MG 90) Im3m (Q ) and Ia3d [202,206]
(DDP(EO)92) (Q230) (coexisting
with L3 phase)
PEG-100-stearate (Myrj59) 4400 100 Phytantriol Pn3m (Q224) [118,203]
229
1,3-Didodecyloxy-2-glycidyl-glycerol- 5000 114 GMO (RYLO MG 90) Im3m (Q ) and Ia3d [202,206]
PEG (Q230) (coexisting
(DDGG4-(EO)114) with L3 phase)
PEG6K10PHYT40 6000 136 Phytantriol Pn3m (Q224) [201]
224
PEG6K20PHYT30 6000 136 Phytantriol Pn3m (Q ) [201]
PEG6K25PHYT25 6000 136 Phytantriol Pn3m (Q224) [201]
224
PEG6K30PHYT20 6000 136 Phytantriol Pn3m (Q ) [201]
224
PEG6K40PHYT10 6000 136 Phytantriol Pn3m (Q ) [201]
1,3-Didodecyloxy-2-glycidyl-glycerol- 6000 136 GMO (RYLO MG 90) Im3m (Q229) and Ia3d [202,206]
PEG-1,3-didodecyloxy-2- (Q230) (coexisting
glycidyl-glycerol (DDGG2-(EO)136- with L3 phase)
DDGG2)
PEG-150-stearate 6600 150 Phytantriol Pn3m (Q224) [118]
224
PEG8K10PHYT40 8000 181 Phytantriol Pn3m (Q ) [201]
224

ARTICLE IN PRESS
PEG8K20PHYT30 8000 181 Phytantriol Pn3m (Q ) [201]
PEG8K25PHYT25 8000 181 Phytantriol Pn3m (Q224) [201]
224
PEG8K30PHYT20 8000 181 Phytantriol Pn3m (Q ) [201]
224
PEG8K40PHYT10 8000 181 Phytantriol Pn3m (Q ) [201]
224
PEG10K10PHYT40 10,000 227 Phytantriol Pn3m (Q ) [201]
224
PEG10K20PHYT30 10,000 227 Phytantriol Pn3m (Q ) [201]
PEG10K25PHYT25 10,000 227 Phytantriol Pn3m (Q224) [201]
224
PEG10K30PHYT20 10,000 227 Phytantriol Pn3m (Q ) [201]
224
PEG10K40PHYT10 10,000 227 Phytantriol Pn3m (Q ) [201]
PEG14K10PHYT40 14,000 317 Phytantriol Pn3m (Q224) [201]
224
PEG14K20PHYT30 14,000 317 Phytantriol Pn3m (Q ) [201]
Continued
Table 3 PEGylated lipid copolymers used as steric stabilizers for cubosomes reported in the general literaturecont'd
PEG PEG Space group of inner
Stabilizer MW units Lipid matrix constituent structure References
224
PEG14K25PHYT25 14,000 317 Phytantriol Pn3m (Q ) [201]
PEG14K30PHYT20 14,000 317 Phytantriol Pn3m (Q224) [201]
224
PEG14K40PHYT10 14,000 317 Phytantriol Pn3m (Q ) [201]
224
P(ODA)6-b-P(PEGA-OMe)27 Phytantriol Pn3m (Q ) [207]
Glyceryl monooleate Im3m (Q229) [207]

ARTICLE IN PRESS
224
P(ODA)6-b-P(PEGA-OMe)35 Phytantriol Pn3m (Q ) [207]
229
Glyceryl monooleate Im3m (Q ) [207]
224
P(ODA)6-b-P(PEGA-OMe)39 Phytantriol Pn3m (Q ) [207]
229
Glyceryl monooleate Im3m (Q ) [207]
P(ODA)10-b-P(PEGA-OMe)23 Phytantriol Pn3m (Q224) [207]
229
Glyceryl monooleate Im3m (Q ) [207]
224
P(ODA)10-b-P(PEGA-OMe)31 Phytantriol Pn3m (Q ) [207]
Glyceryl monooleate Im3m (Q229) [207]
224
P(ODA)10-b-P(PEGA-OMe)34 Phytantriol Pn3m (Q ) [207]
229
Glyceryl monooleate Im3m (Q ) [207]
PEG-based copolymers bearing lipid-mimetic anchors Glyceryl monooleate/sodium Cubosome (coexisting with [208]
cholate L3 phase)
Table 4 Additional miscellaneous steric stabilizers for cubosomes reported in the general literature
Space group of
Stabilizer Lipid matrix constituent inner structure References
Casein
-Casein Glyceryl monooleate Pn3m (Q224) [152]
Casein Myverol 18-99K Cubosome [113]
Albumin

ARTICLE IN PRESS
Albumin Myverol 18-99K Cubosome [113]
Lecithin
Partially hydrolyzed emulsifier lecithin Dimodan U/J (96% monoglycerides: 62% linoleate Im3m (Q229) [195]
(EmultopEP) and 25% oleate)
Modified cellulose
Hydroxypropyl methyl cellulose Glyceryl monooleate Pn3m (Q224) [161]
acetate succinate (HPMCAS)
Hydroxypropyl methyl cellulose 1-O-(5,9,13,17-tetramethyloctadecanoyl)erythritol Pn3m (Q224) [161]
acetate succinate (HPMCAS) (EROCO C22)
Hydroxypropyl methyl cellulose 1-O-(5,9,13,17-tetramethyloctadecyl)--D-xylopyranoside Pn3m (Q224) [161]
acetate succinate (HPMCAS) (-XP)
Modified starch
HI-CAP100 (hydrophobically Glyceryl monooleate Cubosome [209]
modified with octenyl succinate
groups)
Continued
Table 4 Additional miscellaneous steric stabilizers for cubosomes reported in the general literaturecont'd
Space group of
Stabilizer Lipid matrix constituent inner structure References
CAPSUL-E (hydrophobically Glyceryl monooleate Cubosome [209]
modified with octenyl succinate
groups)
Dextran Glyceryl monooleate Cubosome [209]
Laponite
Laponite XLG Phytantriol Pn3m (Q224) [151]

ARTICLE IN PRESS
224
Laponite XLG Dimodan U/J (96% monoglycerides: 62% linoleate Pn3m (Q ) [193]
and 25% oleate)
Laponite XLG Dimodan U/J (96% monoglycerides: 62% linoleate Pn3m (Q224) [192]
and 25% oleate)/tetradecane (oil)
Silica nanoparticles

Silica nanoparticles Phytantriol/tetradecane (oil) Pn3m (Q224) [210]


No stabilizer
1,2-Dioleoyl-sn-glycero-3-phosphocholine (DOPC) and Im3m (Q229) [101]
1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE)
Glyceryl monooleate/cis-5,8,11,14,17-eicosapentaenoic Cubosome [204]
acid/1,2-dioleyl-sn-glycero-3-phosphoethanolamine-N-
[methoxy(poly(ethylene glycol))-2000] (DOPE-PEG2000)
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 31

4.1.1.1 Poloxamer
4.1.1.1.1 Poloxamer 407/PluronicF127 By far the most widely
and frequently used steric stabilizer for cubosomes is Poloxamer 407 (also
known as PluronicF127), a nonionic triblock copolymer composed of
PEG and polypropylene oxide (PPO): PEG100PPO65PEG100, with a molec-
ular weight of approximately 12,600 Da (Figs. 6 and 7). PluronicF127 is a
nonionic macromolecule that is used widely in pharmaceutical formulations
and personal care products. In lyotropic liquid crystalline dispersions,
PluronicF127 acts as a steric stabilizer through the incorporation or adsorp-
tion of its hydrophobic PPO block onto the surface of the nanostructured
particle. Whilst the PPO domain/block acts as an anchor to the particle,
the hydrophilic PEG chains extend to cover the surface, providing steric
shielding and stabilizing the colloidal particles in aqueous solutions [211].
PluronicF127 has been employed to stabilize cubosome dispersions in
various lipid systems, including GMO, glycerol monolinoleate, and
phytantriol. The GMO system has been the most extensively studied. At
low stabilizer concentrations (<4%, w/w, vs. GMO), PluronicF127 stabi-
lized GMO dispersions form Q224 cubosomes with Pn3m space group sym-
metry, whilst at higher stabilizer concentrations (i.e., 7.4 or 10%, w/w, vs.
GMO), Q229 cubosomes with Im3m space group symmetry are formed
[173]. Although using low stabilizer concentrations of PluronicF127 can

A B
4000 L121 P123 F127 4000 L121 P123 F127

L101 P103 P104 P105 F108 L101 P103 P104 P105 F108
Hydrophobe (MW of PPO)

Hydrophobe (MW of PPO)

L92 L92

L81 P84 P85 F87 L81 P84 P85 F87

L61 L62 L64 F68 L61 L62 L64 F68

L43 L43

950 L35 F38 950 L35 F38


0 10 20 30 40 50 60 70 80 0 10 20 30 40 50 60 70 80

Hydrophile (% of PEG) Hydrophile (% of PEG)


Key: Not assessed Im3m Pn3m

Figure 6 Lyotropic liquid crystalline phases obtained from (A) phytantriol and
(B) monoolein dispersions using the Poloxamers/Pluronic surfactants. Image
reproduced from [160] with permission from The Royal Society of Chemistry.
ARTICLE IN PRESS

32 Josephine Y.T. Chong et al.

Figure 7 The chemical structures of stabilizers Pluronic and Tetronic. The blue (gray
in the print version) shading indicates the hydrophilic domain, whilst the yellow (light
gray in the print version) shading indicates the hydrophobic domain. A graphic illustra-
tion of the stabilizer structure is shown on the left hand side, with the dashed line rep-
resentative of the surface of a nanostructured particle.

produce Q224 cubosomes, the overall quality of the dispersed sample is


typically poor, with visual aggregates present. Therefore, higher stabilizer
concentrations (i.e., 10% w/w vs. lipid) are typically employed in cubosome
preparation, as they establish dispersions that are aggregate-free.
It is important to preserve the Pn3m space group symmetry within GMO
cubosomes because not only does the change to an Im3m space group sym-
metry indicate a disruption and destabilization of the lyotropic liquid crystal
system, but also the release rate of encapsulated drugs from a cubic phase
system with an Im3m space group symmetry is much faster than it is for
one with a Pn3m space group symmetry [41].
In contrast to the case of GMO, the use of high PluronicF127
concentrations with either glycerol monolinoleate [164] or phytantriol
[149], as the main lipid, results in retention of the Pn3m diamond (D)
bicontinuous cubic phase within their dispersions. The PluronicF127 con-
tent was as high as 33% relative to lipid in some of the phytantriol dispersions
in water [149]. Mixed phase (Q229 and Q224) cubosome dispersions can also
be obtained from the cubic phase-forming lipid, 1-O-(5,9,13,17-
tetramethyloctadecyl)--D-xylopyranoside (-XP), system when dispersed
with 5.1% (w/w) PluronicF127 [162]. Mixed phases were also observed
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 33

from a ternary system composed of pine needle oil monoglycerides (89.5 wt


% GMO), PluronicF127, and water, whereby an extended inverted cubic
phase was observed in which four different cubic structures (Q230
(G periodic minimal surface), Q224 (D periodic minimal surface), Q229
(P periodic minimal surface), and Q229 (Neoviuss periodic minimal surface)
were detected by X-ray diffraction [187].
4.1.1.1.2 Poloxamer 338/PluronicF108 PluronicF108, with the
structure PEG132PPO50PEG132, is another Poloxamer class amphiphile
which has recently been successfully used to sterically stabilize cubosomes.
It has longer hydrophilic arms than PluronicF127 with a molecular weight
of 14,600 Da. Chong et al. reported its ability to conserve the native Pn3m
space group symmetry of the bulk phase in both GMO and phytantriol cub-
osome dispersions (Fig. 6) in 2011 [118,160]. Since then, Swarnakar et al.,
Murgia et al., and Caltagirone et al. have used PluronicF108 to stabilize
drug-loaded cubic nanostructured particles from aqueous dispersions based
on GMO (GMOPluronicF108water system) [74,92,93]. Swarnakar
et al. also reported cubic nanostructured particles from aqueous dispersions
based on phytantriol (phytantriolPluronicF108water system) [74].
4.1.1.1.3 Other Poloxamers/Pluronics Other Poloxamers/
Pluronics which have been used for the stabilization of cubosome disper-
sions of GMO and/or phytantriol are summarized in Table 2. Chong et al.
reported the use of Pluronics with lower molecular weights than
PluronicF127 (i.e., PluronicF87, F68, P123, P105, P104, P84) as steric
stabilizers (Fig. 6) [160]. Pluronics with PEG chains greater or equal to
37 PEG units were reported to stabilize phytantriol cubosome dispersions,
which had Pn3m space group symmetry. In contrast, all the Pluronics that
were able to produce cubosome dispersions based on GMO were reported
to have Im3m space group symmetry. Tamayo-Esquivel et al., Swarnakar
et al., and Boyd et al. also reported cubic nanostructured particles from aque-
ous dispersions based on GMO (GMOPluronicF68water system)
[73,74,113] and phytantriol (phytantriolPluronicF68water system) [74],
employing PluronicF68 as a steric stabilizing agent.

4.1.1.2 Poloxamine
4.1.1.2.1 Poloxamine 908/Tetronic908 Poloxamine 908, also
known as Tetronic908, is a tetrafunctional PEGPPO ethylenediamine
block copolymer (Fig. 7). Boyd et al. reported stabilizing aqueous dispersions
of Myverol 18-99K in Poloxamine 908 solution [113]. Aqueous
ARTICLE IN PRESS

34 Josephine Y.T. Chong et al.

dispersions of Myverol 18-99K in water were also achieved using a com-


bination of Poloxamer 407 and Poloxamine 908, as stabilizing
agents [113].

4.1.2 PEGylated lipids


In addition to commercially available block copolymer stabilizers,
PEGylated lipids (i.e., PEG-lipids) have also been reported to sterically sta-
bilize inverse bicontinuous cubic phase particles. To date only a few
PEGylated lipids have been published as effective stabilizers for cubosomes
(Fig. 8) [38,88,197200,202,204206,208]. The lipid anchors of these
PEGylated lipids are explored in greater detail below.

4.1.2.1 Glyceryl monooleate


Johnsson et al. reported cubic nanostructured particles prepared from the
dispersion of DOPE with PEGylated monoolein (MO-PEG660) as a steric
stabilizing agent [199]. These cubosomes were identified with the mixed
cubic phases Ia3d and Pn3m space groups (Q230 and Q224).
Angelov et al. reported cubic nanostructured particles from aqueous dis-
persions of GMO in water using a PEGylated monoolein with a longer PEG
chain (MO-PEG2000) as the steric stabilizing agent [88]. These cubosomes
had mixed Im3m and Pn3m space group symmetry.

4.1.2.2 Sorbitan monooleate and sorbitan monopalmitate


Polyoxyethylene (20) sorbitan monooleate, otherwise known as polysorbate
80 or Tween80, is a commercially available PEGylated lipid which has
been reported to stabilize cubosomes (Fig. 8). Polysorbate 80 is a solubilizing
agent ubiquitously used in nutritives, creams, ointments, lotions, and mul-
tiple medical preparations (e.g., vitamin oils, vaccines, and anticancer agents)
and as an additive in tablets. Barauskas et al. reported cubic nanostructured
particles from aqueous dispersions of GMO (GMOTween80water sys-
tem) with Tween80, as a steric stabilizing agent [102]. Cubosomes have
also been prepared using soy phosphatidyl choline (SPC) and glycerol dio-
leate (GDO) in water, sterically stabilized with Tween80 [75,84,85].
Barauskas et al. have also stabilized cubic nanostructured particles from
aqueous dispersions of soy PE (L--phosphatidylethanolamine) (Soy PE
Tween80water system) using Tween80 [38].
It is interesting to note that Boyd et al. observed immediate phase sep-
aration when using polysorbate 80 to stabilize dispersions of Myverol
18-99K, using 10% w/w solution in lipid [113]. Although polysorbate
ARTICLE IN PRESS

Figure 8 The chemical structures of some of the PEGylated lipid stabilizers used in the
literature for the preparation of cubosome dispersions. The blue (gray in the print ver-
sion) shading indicates the hydrophilic domain and the yellow (light gray in the print
version) shading indicates the hydrophobic domain. A graphic illustration of the stabi-
lizer structure is shown on the left hand side, with the dashed line representative of the
nanostructured particle's surface.
ARTICLE IN PRESS

36 Josephine Y.T. Chong et al.

80 was successfully used to stabilize GMO cubosomes, Myverol is a mix-


ture of monoglycerides, and it is possible the impurities influence the effect
of the polysorbate stabilizer. Boyd et al. have also assessed other polysorbates
for their effectiveness at stabilizing Myverol dispersions. These were poly-
sorbate 20 (polyoxyethylene (20) sorbitan monolaurate), polysorbate
40 (polyoxyethylene (20) sorbitan monopalmitate), and polysorbate
60 (polyoxyethylene (20) sorbitan monostearate), which are commercially
known as Tween20, 40, and 60, respectively [113].
Similar to polysorbate 80, it was found that polysorbates 20 and 60 stabi-
lized Myverol systems in water displayed immediate phase separation.
Only polysorbate 40 (Fig. 8) was found to produce a coarse dispersion of
Myverol in water. This indicates the importance of the lipophilic domain
length of a polysorbate stabilizer and that an optimal hydrocarbon chain
length is apparently required to sufficiently anchor the stabilizer to the nano-
structured particles and effectively stabilize these systems.

4.1.2.3 D-alpha-tocopheryl (vitamin E)


Barauskas et al. reported cubic nanostructured particles from aqueous disper-
sions of phytantriol in water, with D-alpha-tocopheryl PEO1000 succinate
(vitamin E TPGS) as the steric stabilizer [38]. Cubosomes were identified
with cubic phase Q229, with Im3m space group symmetry.

4.1.2.4 Phospholipids (DOPE, DSPE, and DMPE)


The use of PEGylated phospholipids as a steric stabilizer for cubic nanostruc-
tured particles was reported by Angelov et al. [204], Johnsson and Edwards
[200], Zeng et al. [205], and Koynova et al. [197,198]. Angelov et al. reported
cubic nanostructured particles from aqueous dispersions of GMO and
cis-5,8,11,14,17-eicosapentaenoic acid (20:5, EPA) in water, with 1,2-
dioleoylphosphatidylethanolamine-PEG (DOPE-PEG2000) as the steric
stabilizer [204].
Johnsson and Edwards reported cubic nanostructured particles from
aqueous dispersions of DOPE-PEG-derivatized phospholipids (PEG lipid
+ water systems) with PEG lipids (DSPE-PEG (DSPE-PEG750 or DSPE-
PEG2000)) as a steric stabilizing agent [200]. Zeng et al. also used DSPE-
PEG2000 as a stabilizing agent for forming cubic nanostructured particles
from aqueous dispersions of SPC and GDO [205].
Koynova et al. reported cubic nanostructured particles from aqueous dis-
persions of dielaidoylphosphatidylethanolamine (DEPE) with PEGylated
lipid, PEGylated DMPE (DMPE-PEG550) as a steric stabilizing agent
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 37

[197,198]. Cubosomes were identified with cubic phase Q229, with Im3m
space group symmetry.

4.1.2.5 1,3-Didodecyloxy-propane-2-ol and


1,3-didodecyloxy-2-glycidyl-glycerol
Rangelov and Almgren reported cubosomes with Ia3d space group from
aqueous dispersions of GMO (RYLO MG 90) in water, using 1,3-
didodecyloxy-propane-2-ol (DDP)-PEG (DDP(EO)30) as the steric stabi-
lizer [202]. These findings were further supported by Almgren and Rangelov
who reported cubic nanostructured particles from aqueous dispersions of
GMO (RYLO MG 90) in water, using DDP-PEG (DDP(EO)52 or
DDP(EO)92) as the steric stabilizer [206]. However, cubosomes were also
identified with an additional cubic phase Q229, with Im3m space group.
Almgren and Rangelov [202,206] also reported cubic nanostructured
particles from aqueous dispersions of GMO (RYLO MG 90) in water, with
1,3-didodecyloxy-2-glycidyl-glycerol (DDGG)-PEG (DDGG4-(EO)114) as
the steric stabilizer. They also tested a PEGylated lipid with a triblock copol-
ymer structure, with the lipids located on both terminal ends of the PEG
domain (DDGG2-(EO)136-DDGG2) [202,206]. Cubosomes formed were
identified with cubic phase Q230, with Ia3d space group and cubic phase
Q229, with Im3m space group symmetry.

4.1.2.6 Octadecanoic acid (stearic acid)


Chong et al. reported cubic nanostructured particles from aqueous disper-
sions of phytantriol in water, using PEG-stearates (Myrj) as steric stabilizers
[118,203]. From the series of PEG-stearates assessed (i.e., PEG-stearates
with 10, 20, 25, 40, 45, 50, 55, 100, and 150 PEG units), only those with
40 or greater PEG units were able to form aqueous dispersions of phytantriol
in water (Fig. 9) [203]. Furthermore, the PEG-stearates that were able to
retain the Pn3m space group of phytantriol, had 100 or greater PEG units
(i.e., PEG-100-stearate (Myrj59) and PEG-150-stearate) [118,203]. It is
also interesting to note that the addition of an additional stearate chain to
the PEG-150-stearate stabilizer disabled its effectiveness as a steric stabilizer,
as PEG-150-distearate was unable to provide stable dispersions of
phytantriol in water [118]. This reinforces the importance of the position
of hydrophilic moieties within an amphiphilic polymer, for its application
as a steric stabilizer. Chong et al. also reported that PEG-stearates were
unsuccessful in stabilizing monoolein dispersions.
Although several PEGylated lipid stabilizers with different PEG lengths
have been identified, the most frequently used PEG length in PEGylated lipid
ARTICLE IN PRESS

38 Josephine Y.T. Chong et al.

Figure 9 Illustration of PEG-100-stearate (Myrj59), the SAXS-scattering profile when


used to stabilize phytantriol-based cubosomes, and the visual assessment results for
dispersions using commercially available stabilizers, including the PEG-stearate copol-
ymer series. Images reproduced with permission from [203].

stabilizers is 45 PEG units (i.e., PEG2000) [88,200,204,205]. The use of


PEG2000 was initiated for liposome stabilization, whereby the use of
PEG2000 hindered aggregation of lipid nanostructured particles under physi-
ological conditions [88,212214]. The shortest and the longest chain lengths
for PEGylated lipid stabilizers that have been reported are 10 PEG units on
average [197,198,203] and 150 PEG units on average [118], respectively.
These PEGylated lipids have mainly been used to stabilize dispersions of
GMO, resulting in Q229 cubic phase dispersions, with an Im3m space group.
However, other lipid dispersions stabilized using PEGylated lipids as steric
stabilizers include DEPE, DOPE, and a mixture of SPC and GDO. The first
PEGylated lipid that had been reported to stabilize phytantriol dispersions in
water was D-alpha-tocopheryl poly(ethylene glycol) 1000 succinate (vitamin
E TPGS), which was used at around 10% w/w stabilizer concentration. This
resulted in a cubosome with the P-type cubic phase internal structure (Im3m
space group) [38]. Since, PEG-100-stearate (Myrj59) [118,203] and PEG-
150-stearate [118] were reported to successfully stabilize phytantriol disper-
sions in water, with stabilizer concentrations as low as 1% w/w. In contrast
to vitamin E TPGS stabilized dispersions, these cubosome dispersions had
Pn3m space group symmetry [118,203].

4.1.3 Designer/customized lipidcopolymer series


4.1.3.1 PEGylated-phytanyl copolymer series
Chong et al. reported cubosomes with Pn3m and Im3m space groups, from
aqueous dispersions of phytantriol, using random copolymers from a series
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 39

of PEGylated-phytanyl copolymers, as the steric stabilizing agent [201].


The molecular weight of the PEG used in the preparation of the copoly-
mers and the ratio of the PEG-to-lipid within the copolymer composition
were varied to enable structureperformance relationships to be established
to guide further work on rationally designed systems (Fig. 10). The impor-
tant relationships established were that an asymmetric amphiphilic structure
with a larger hydrophilic domain (e.g., high hydrophilic lipophilic balance
(HLB)) and adequate PEG length were essential for establishing an effective
steric stabilizer. Copolymers with PEG lengths between 68 and 136 PEG
units on average, with HLB > 17, were found to be effective steric stabi-
lizers for phytantriol cubosome dispersions. Chong et al. also reported a
maximum hydrophilic threshold, whereby the effectiveness of the copoly-
mer at providing effective long-term steric stability diminishes when the
hydrophilic domain exceeds 60% of the copolymers total amphiphilic
structure.

4.1.3.2 Polymeric PEGylated lipid copolymer series


In addition to PEGylated lipids, a recently developed customized polymeric
PEGylated lipid system, P(ODA)-b-P(PEGA-OMe) was reported by
Chong et al. to successfully sterically stabilize aqueous dispersions of both
phytantriol and monoolein cubosomes in water [207]. Cubosomes with
Pn3m space group symmetry were prepared, from aqueous dispersions of
phytantriol, using the P(ODA)-b-P(PEGA-OMe) copolymers as the steric
stabilizing agents [207]. Similar to PluronicF127, the P(ODA)-b-P(PEGA-
OMe) copolymers also sterically stabilized monoolein cubosomes, with
Im3m space group symmetry (Fig. 11) [207].
The effectiveness of the designer copolymers was equivalent to
PluronicF127, highlighting the potential of developing custom steric sta-
bilizers for cubosomes [207]. Specifically, these custom polymeric
PEGylated lipid copolymers were designed to allow functionalization at
the terminal end of the hydrophilic moiety (i.e., P(PEGA-OMe)), to enable
the addition of active targeting functionalities for targeting in vivo delivery
applications.

4.1.4 Alternative steric stabilizers


In addition to amphiphilic block copolymers, PEGylated lipids, and custom-
ized lipid copolymers, other amphiphilic stabilizing agents have been
reported for cubosomes, including bile salts, amphiphilic proteins (i.e.,
ARTICLE IN PRESS

40 Josephine Y.T. Chong et al.

Figure 10 Accelerated stability assay results for (A) F127 (control stabilizer) at 0.3, 0.5,
0.7, 1, and 1.2 wt% stabilizer concentration, (B) PEG20PHYT30 copolymer series, where
PEG MW: 214K, (C) PEG4K-PHYT copolymer (from 20, 25, 30 and 40 PEG mol% copol-
ymer series), and (D) PEG6K-PHYT copolymer (from 20, 30 and 40 PEG mol% copolymer
series). ASA results after first spin 1800 rpm are represented in black columns, whilst ASA
results after second spin 2000 rpm are represented in blue (gray in the print version)
columns. Steric stabilizer concentration for ASA results presented in (B)(D) is 1 wt%,
with control standard steric stabilizer F127 at 1 wt% presented to the right. Image
reproduced with permission from [201].
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 41

A B
600 2

500
Intensity (AU)

400

300 4 6

200 12
10 14
100

0.05 0.1 0.15 0.2 500 nm


1
q ( )

Figure 11 (A) SAXS diffraction pattern showing an Im3m space group symmetry and
(B) cryo-TEM image of monoolein dispersion stabilized using reduced P(ODA)10-b-P
(PEGA-OMe)34 copolymer at 25 C. Images reproduced from [207] with permission from
The Royal Society of Chemistry.

casein and albumin), modified polysaccharide polymers (i.e., modified cel-


lulose and starch), poly(vinyl) alcohol, and nanoparticles (i.e., silica and clay
nanoparticles).

4.1.4.1 Bile salts


The first fragmented bilayer cubic phase structure was observed in 1979
[215]. The cubic phase formed from a monoglyceridewater mixture was
dispersed in the presence of micellar solutions of bile salts [119,208,216].
The cubic phase was stabilized by the formation of a lamellar envelope com-
posed of bile salt and monoglycerides shielding the inner cubic structure.

4.1.4.2 Amphiphilic protein


4.1.4.2.1 -Casein -Casein is an amphiphilic protein and a very effec-
tive emulsifier and so was employed as a stabilizer in a disperse mono-
glyceridewater cubic phase. Here, the protein is believed to partition
into the outer layer of the lipid particle, yielding a hydrophilic coating
and therefore easy to disperse [152,217].

4.1.4.2.2 Albumin Albumin is a ubiquitous protein which is soluble in


water and can be found in egg white, milk, and blood serum. Boyd et al.
briefly reported coarse dispersions of Myverol 18-99K in water using
albumin as a steric stabilizer [113].
ARTICLE IN PRESS

42 Josephine Y.T. Chong et al.

4.1.4.3 Partially hydrolyzed lecithin (EmultopEP)


EmultopEP, a partially hydrolyzed lecithin, is a well-accepted food grade
emulsifier and contains a relatively large quantity of lysophospholipids. It
was thought to be a good emulsifying/stabilizing agent as it was able to sta-
bilize O/W emulsions. Sagalowicz et al. reported cubic nanostructured par-
ticles, with Im3m space group symmetry, from aqueous dispersions of
Dimodan U/J (a lipid containing 96% monoglycerides: 62% linoleate and
25% oleate) in water, using EmultopEP as the steric stabilizer [195].

4.1.4.4 Modified polysaccharide polymers


4.1.4.4.1 Hydrophobically modified ethyl hydroxyethyl cellulose
(modified cellulose) Almgren et al. applied hydrophobically modified
ethyl hydroxyethyl cellulose (HMEHEC) to the GMO-based cubic phase
[218]. Although cubosomes were formed, HMEHEC polymers do not
make successful cubosome stabilizers as they were observed to have inter-
acted so strongly with lipids that shortly after dispersion of the sample,
the internal nanostructure of the cubosome transformed into lamellar and
reversed hexagonal phase.

4.1.4.4.2 Hydroxypropyl methyl cellulose acetate succinate


(modified cellulose) Uyama et al. reported cubic nanostructured particles
from aqueous dispersions of three lipids: (i) GMO, (ii) 1-O-(5,9,13,17-
tetramethyloctadecanoyl)erythritol (EROCO C22), and (iii) 1-O-
(5,9,13,17-tetramethyloctadecyl)--D-xylopyranoside (-XP), using
hydroxypropyl methyl cellulose acetate succinate (HPMCAS) as a stabilizing
agent [161]. The cubosomes formed were identified to have cubic phase
Q224, with Pn3m space group symmetry. The motivation for using modified
cellulose was because cellulose products are widely used in the cosmetics,
food, and pharmaceutical industries, such as in eye drops and inhalants.
HPMCAS is a commercially available generic coating agent and widely used
in dry coating or solid dispersion systems [219221] and demonstrated by
Uyama et al., to be applicable as a stabilizer for cubosomes, which allows
for sufficient dispersion stability without any internal structure
modification [161].

4.1.4.4.3 HI-CAP100, CAPSUL-E, dextran (modified starch) Spicer


et al. presented a pseudoternary phase diagram of GMO with
hydrophobically modified starch in water and prepared cubosomes by the
rehydration of spray-dried starchGMO mixtures [209]. In that system,
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 43

starch was mixed threefold higher than the weight of GMO, and the particle
size was 600 nm on average.

4.1.4.5 Amphiphilic poly(vinyl) alcohol


Poly(vinyl) alcohol is a water-soluble synthetic polymer, which has been
used in papermaking and textiles. Tamayo-Esquivel et al. has reported using
poly(vinyl) alcohol to stabilize aqueous GMO dispersions, resulting in sub-
200 nm particles [73].

4.1.4.6 Nanoparticles
4.1.4.6.1 Laponite XLG (clay nanoparticles) Muller et al. and
Salonen et al. have reported cubic nanostructured particles from aqueous dis-
persions of two lipids: (i) phytantriol [151] and (ii) Dimodan U/J (consisting
of 96% monoglycerides, which contain 62% linoleate and 25% oleate), with
[192] and without [193] tetradecane (oil) in water, using Laponite XLG (clay
nanoparticles) as the steric stabilizer. The cubosomes formed were identified
with cubic phase Q224, with Pn3m space group symmetry.

4.1.4.6.2 Silica (silica nanoparticles) Salonen et al. have reported


cubic nanostructured particles from aqueous dispersions of phytantriol
and tetradecane (oil) in water, using silica nanoparticles as the steric
stabilizing agent [210]. The cubosomes formed were identified with cubic
phase Q224, with Pn3m space group symmetry.

5. FUTURE DEVELOPMENTS IN THE STABILIZATION


OF CUBOSOMES
Although advancements in high-throughput technology have accel-
erated the rate of screening for steric stabilizers for cubosomes, the current
list of steric stabilizers for cubosomes is still relatively small and limited to a
few classes of primarily steric stabilizers. This limits potential progression of
these materials into viable products by reducing the available formulation
space. Despite the applicability of the possible classes of steric stabilizers
for cubosomes, there is an overwhelming reliance and frequency of use with
one particular steric stabilizer, PluronicF127. PluronicF127 is still
currently the main steric stabilizer for cubosome preparation of drug-
encapsulated cubosomes for drug delivery applications. This may be in
part due to only recent attempts to establish an understanding of the
ARTICLE IN PRESS

44 Josephine Y.T. Chong et al.

structureperformance relationships dictating the colloidal stability for these


relatively complex self-assembled particles.
Although PluronicF127 is an established and effective steric stabilizer
for cubosomes, some issues have been raised regarding its use. Physicochem-
ically, PluronicF127 is thought to adsorb onto the particle surface [120];
however, this polymer has been shown to affect the internal structure of
GMO-based cubosomes, inducing a transition from the parent Pn3m to
Im3m space group [172,173,181,187]. In addition, the cubic phase is trans-
formed to a lamellar phase (vesicles) at high PluronicF127 concentrations
[187]. Thus, there is generally a need to identify more alternative steric
stabilizers, both commercially available and custom-synthesized stabilizers.
Designing effective customized steric stabilizers generally requires iden-
tifying intelligent/smart design properties, which can only be established
after screening many different stabilizers, and under different conditions.
The structureproperty relationships currently identified to be important
in optimizing colloidal stability include PEG length in PEGylated stabilizers,
and having multiple PEG chains (e.g., triblock and brush copolymers). For
example, stabilizers PluronicF127 and Myrj59 both have approximately
100 PEG units and were successfully able to sterically stabilize phytantriol
cubosome dispersions, which retained the native Pn3m space group symme-
try [203]. However, PluronicF127 out-performs Myrj59 when compar-
ing colloidal stability of their stabilized dispersions using the accelerated
stability assay, which may possibly be due to Pluronic stabilizers having
two PEG arms compared with Myrj stabilizers having just one PEG arm
[118]. As increased PEGylation enhances the colloidal stability, polymers
which are more effective steric stabilizers tend to have higher water solubility
(e.g., high HLB value).
It is also important to ensure that when designing and synthesizing a steric
stabilizer, the hydrophilic moiety is not surrounded by hydrophobic groups.
For example, PEG-150-distearate and nonreduced P(ODA)-b-P(PEGA-
OMe) copolymers were not as successful in maintaining stable dispersions
as their single hydrophobic block counterparts, namely PEG-150-stearate
and the reduced P(ODA)-b-P(PEGA-OMe) copolymers [118,207]. This
is likely caused by the aggregation of the hydrophobic ends and increased
interparticle bridging. With reverse addition-fragmentation chain transfer
(RAFT) polymerization being a popular technique for synthesizing excit-
ing, novel polymer architectures, it is also important to note that some
RAFT end-groups can be hydrophobic and can impact the performance
of the polymer as a steric stabilizer.
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 45

The future developments for the stabilization of cubosomes involve


adding functionalization (i.e., targeting and MRI imaging agent) to the sta-
bilizer, for active targeting capabilities [92,93,106,108,188]. An extensive
review of cubosome targeting, drug delivery, and medical imaging was
recently written by Mulet et al. [106]. A recent study by Caltagirone et al.
reported the functionalization of PluronicF108 with folate, for targeting
cubosomes to cancer cells. The targeted monoolein cubosomes were loaded
with camptothecin, an anticancer therapeutic, and provided active drug
targeting [93]. Although this is the first report of active targeting activity
in cubosomes, there are a vast array of opportunities to be explored. With
possible exploration of functionalizing and development of more custom-
ized steric stabilizers (e.g., P(ODA)-b-P(PEGA-OMe) copolymers),
improvements in stealth behavior and colloidal stability, in addition to a
targeting capability, are likely to result. Regardless, exceptional colloidal sta-
bility is essential for these systems to be eventually developed into clinical
products/therapeutics.
In addition to steric stabilization, there are other stabilizers (e.g., charged
stabilizers), which have not been covered in detail in this chapter but have
the potential to be explored further for the advancement of the stabilization
of lyotropic liquid crystalline nanostructured particles. Other directions of
research in the stabilization of lyotropic liquid crystalline nanostructured
particles may include using charged polymers to stabilize dispersions
[158,159,222,223]. In particular, Angelov et al. stabilized a dispersion of
fusogenic monoolein, using cationic lipid dioctadecyldimethylammonium
bromide and PEGylated lipid 1,2-dioleoyl-sn-glycero-3-phospho-
ethanolamine-N-(methoxy-(polyethyleneglycol)-2000) ammonium salt
(DOPE-PEG2000) [223]. These nanostructured particles were able to encap-
sulate neurotropic plasmid DNA, for the study of using lyotropic liquid crys-
talline nanostructured particles as nanocarriers for DNA complexes for the
development of prospective nanoparticle-based gene therapies.
Lastly, it may be that the future developments in lyotropic liquid crys-
talline nanostructured particles may not require addition of a separate stabi-
lizer. Studies investigating stimuli responsiveness of lyotropic liquid
crystalline nanostructured particles, such as inducing a stable vesicle to unsta-
ble cubosome transformation in situ as reported recently by Du et al., have
highlighted the possibility of maintaining stable lyotropic liquid crystalline
nanostructured particle dispersions in the absence of a stabilizer, by
maintaining a specific pH (e.g., pH 8 in Dus study) [224]. Therefore,
one of the future directions for maintaining stable lyotropic liquid crystalline
ARTICLE IN PRESS

46 Josephine Y.T. Chong et al.

nanostructured particle dispersions may depend on investigating different


solvent conditions, such as temperature and pH.

6. CONCLUSION
We have discussed the stabilization of cubosomes. Due to their small
size and ordered 3D mesoporous internal structure, with a high surface area
for substance loading, cubosomes are being widely investigated as drug
delivery systems, to encapsulate hydrophilic, lipophilic, or amphiphilic ther-
apeutics, and/or imaging agents. However, these bicontinuous cubic
lyotropic liquid crystalline nanostructured particles require the presence
of a stabilizer for their colloidal stability. This review covers stabilizers which
come from amphiphilic block copolymers, PEGylated lipids, designer/cus-
tomized copolymer series, and alternative stabilizers. Although these few
groups of stabilizers have been identified and reported as suitable stabilizers
for these systems, researchers still rely quite heavily on commercially avail-
able stabilizers, such as PluronicF127.
However, advances in polymer synthesis techniques (e.g., RAFT poly-
merization) allow for new polymer structures (e.g., PEGylated brush copol-
ymers) to be investigated for their use as customized steric stabilizers for
cubosome systems. An important molecular design feature for creating an
effective customized stabilizer for cubosomes is having an asymmetric
amphiphilic polymer structure with a larger hydrophilic domain/moiety
(e.g., high HLB value). This can be achieved through the use of longer
PEG chains (e.g., high PEG MW) and/or multiple PEG chains (e.g., brush
or comb-like design). Designing and synthesizing new stabilizers for
lyotropic liquid crystalline nanostructured particles enable these stabilizers
to eventually be functionalized with a targeting moiety. This will enable
active targeting of these systems, which may be important for certain clinical
applications for different therapeutics. Future studies will focus on investi-
gating and screening for more effective stabilizers (e.g., both commercial
and customized) and/or solvent conditions for maintaining cubosome
dispersions.

ACKNOWLEDGMENTS
This work was supported by CSIRO and RMIT funding.
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 47

REFERENCES
[1] V. Luzzati, et al., La structure des collodes dassociation. I. Les phases liquide
cristallines des syste`mes amphiphileeau, Acta Crystallogr. 13 (1960) 660667.
[2] V. Luzzati, P.A. Spegt, Polymorphism of lipids, Nature 215 (1967) 701704.
[3] V. Luzzati, et al., Structure of cubic phases of lipidwater systems, Nature 220 (1968)
485488.
[4] A. Tardieu, V. Luzzati, Polymorphism of lipids: a novel cubic phasea cage-like net-
work of rods with enclosed spherical micelles, Biochim. Biophys. Acta 219 (1970)
1117.
[5] W. Longley, T.J. McIntosh, A bicontinuous tetrahedral structure in a liquid-crystalline
lipid, Nature 303 (1983) 612614.
[6] M. Caffrey, Kinetics and mechanism of transitions involving the lamellar, cubic,
inverted hexagonal and fluid isotropic phases of hydrated monoacylglycerides moni-
tored by time-resolved X-ray diffraction, Biochemistry 26 (1987) 63496363.
[7] P. Mariani, V. Luzzati, H. Delacroix, Cubic phases of lipid-containing systems: struc-
ture analysis and biological implications, J. Mol. Biol. 204 (1988) 165189.
[8] P.J. Maddaford, C. Toprakcioglu, Structure of cubic phases in the ternary system
didodecyldimethylammonium bromide/water/hydrocarbon, Langmuir 9 (1993)
28682878.
[9] O. Diat, G. Porte, J.-F. Berret, Orientation and twins separation in a micellar cubic
crystal under oscillating shear, Phys. Rev. B 54 (1996) 1486914872.
[10] R.R. Balmbra, J.S. Clunie, J.F. Goodman, Cubic mesomorphic phases, Nature
222 (1969) 11591160.
[11] V. Luzzati, et al., Lipid polymorphism: a correction. The structure of the cubic phase of
extinction symbol Fdconsists of two types of disjointed reverse micelles embedded
in a three-dimensional hydrocarbon matrix, Biochemistry 31 (1992) 279285.
[12] P. Mariani, et al., Polymorphism of a lipid extract from Pseudomonas fluorescens:
structure analysis of a hexagonal phase and of a novel cubic phase of extinction symbol
Fd, Biochemistry 29 (1990) 67996810.
[13] J. Charvolin, J.F. Sadoc, Periodic-systems of frustrated fluid films and micellar cubic
structures in liquid crystals, J. Phys. 49 (1988) 521526.
[14] H. Delacroix, T. Gulik-Krzywicki, J.M. Seddon, Freeze fracture electron microscopy
of lyotropic lipid systems: quantitative analysis of the inverse micellar cubic phase of
space group Fd3m (Q227), J. Mol. Biol. 258 (1996) 88103.
[15] G.C. Shearman, et al., A 3-D hexagonal inverse micellar lyotropic phase, J. Am.
Chem. Soc. 131 (2009) 16781679.
[16] H.A. Schwarz, Gesammelte matematische Abhandlung, vol. 1, Springer-Verlag,
Berlin, 1890.
[17] A.H. Schoen, Infinite periodic minimal surfaces without self-intersections: NASA
Technical Note, TD-5541, Springfield, 1970.
[18] L.E. Scriven, Equilibrium bicontinuous structure, Nature 263 (1976) 123125.
[19] J.D. Enlow, Mathematical Modelling of Surfactant Liquid Crystal X-ray Diffraction,
Department of Mathematics and Statistics, University of Otago, New Zealand, 2001,
222.
[20] D. Yang, B. Armitage, S.R. Marder, Cubic liquid-crystalline nanoparticles, Angew.
Chem. Int. Ed. Engl. 43 (2004) 44024409.
[21] B. Angelov, Modulation of nanochannels hydration in lipid cubic phases studied by
SANS and SAXS, in: NSTI-Nanotech, 2007, pp. 58.
[22] B. Angelov, et al., Diamond-type lipid cubic phase with large water channels, J. Am.
Chem. Soc. 125 (2003) 71887189.
ARTICLE IN PRESS

48 Josephine Y.T. Chong et al.

[23] J.N. Israelachvili, D.J. Mitchell, B.W. Ninham, Theory of self-assembly of lipid bila-
yers and vesicles, Biochim. Biophys. Acta 470 (1977) 185201.
[24] V. Luzzati, A. Tardieu, Lipid phasesstructure and structural transitions, Annu. Rev.
Phys. Chem. 25 (1974) 7994.
[25] M. Malmsten, in: Surfactants and Polymers in Drug Delivery. Drugs and the Pharma-
ceutical Sciences, vol. 122, Marcel Dekker, New York, NY, 2002.
[26] A.M. Figueiredo Neto, S.R.A. Salinas, The Physics of Lyotropic Liquid Crystals:
Phase Transitions and Structural Properties, Monographs on the Physics & Chemistry
of Materials, Oxford University Press, New York, NY, 2005.
[27] P.A. Winsor, Binary and multicomponent solutions of amphiphilic compounds. Sol-
ubilization and the formation, structure, and theoretical significance of liquid crystal-
line solutions, Chem. Rev. 68 (1968) 140.
[28] G.J.T. Tiddy, Surfactant-water liquid crystal phases, Phys. Rep. 57 (1980) 146.
[29] J. Charvolin, Crystals of interfacesthe cubic phases of amphiphile water-systems,
J. Geophys. Res. 46 (1985) 173190.
[30] K. Fontell, Cubic phases in surfactant and surfactant-like lipid systems, Colloid Polym.
Sci. 268 (1990) 264285.
[31] C. Fong, T. Le, C.J. Drummond, Lyotropic liquid crystal engineering-ordered nano-
structured small molecule amphiphile self-assembly materials by design, Chem. Soc.
Rev. 41 (2012) 12971322.
[32] T. Kaasgaard, C.J. Drummond, Ordered 2-D and 3-D nanostructured amphiphile self-
assembly materials stable in excess solvent, Phys. Chem. Chem. Phys. 8 (2006)
49574975.
[33] C.E. Conn, C.J. Drummond, Nanostructured bicontinuous cubic lipid self-
assembly materials as matrices for protein encapsulation, Soft Matter 9 (2013)
34493464.
[34] H.-I. Chang, M.-K. Yeh, Clinical development of liposome-based drugs: formulation,
characterization, and therapeutic efficacy, Int. J. Nanomed. 7 (2012) 4960.
[35] J.-X. Zhang, et al., Application of liposomes in drug developmentfocus on gastro-
enterological targets, Int. J. Nanomed. 8 (2013) 13251334.
[36] S.B. Rizwan, et al., Characterisation of bicontinuous cubic liquid crystalline systems of
phytantriol and water using cryo field emission scanning electron microscopy (cryo
FESEM), Micron 38 (2007) 478485.
[37] A. Yaghmur, O. Glatter, Characterization and potential applications of nanostructured
aqueous dispersions, Adv. Colloid Interface Sci. 147148 (2009) 333342.
[38] J. Barauskas, M. Johnsson, F. Tiberg, Self-assembled lipid superstructures: beyond ves-
icles and liposomes, Nano Lett. 5 (2005) 16151619.
[39] X. Mulet, et al., High throughput preparation and characterisation of amphiphilic
nanostructured nanoparticulate drug delivery vehicles, Int. J. Pharm. 395 (2010)
290297.
[40] W.-K. Fong, et al., Controlling the nanostructure of gold nanorod-lyotropic liquid-
crystalline hybrid materials using near-infrared laser irradiation, Langmuir 28 (2012)
1445014460.
[41] K.W.Y. Lee, et al., Nanostructure of liquid crystalline matrix determines in vitro
sustained release and in vivo oral absorption kinetics for hydrophilic model drugs,
Int. J. Pharm. 365 (2009) 190199.
[42] R. Negrini, R. Mezzenga, Diffusion, molecular separation, and drug delivery from
lipid mesophases with tunable water channels, Langmuir 28 (2012) 1645516462.
[43] J. Clogston, et al., Controlling release from the lipidic cubic phase by selective alkyl-
ation, J. Control. Release 102 (2005) 441461.
[44] J. Clogston, M. Caffrey, Controlling release from the lipidic cubic phase. Amino acids,
peptides, proteins and nucleic acids, J. Control. Release 107 (2005) 97111.
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 49

[45] A. Zabara, R. Mezzenga, Modulating the crystal size and morphology of in meso-
crystallized lysozyme by precisely controlling the water channel size of the hosting
mesophase, Soft Matter 9 (2013) 10101014.
[46] R. Mezzenga, et al., Polysaccharide-induced order-to-order transitions in lyotropic
liquid crystals, Langmuir 21 (2005) 61656169.
[47] N.M. Morsi, G.A. Abdelbary, M.A. Ahmed, Silver sulfadiazine based cubosome
hydrogels for topical treatment of burns: development and in vitro/in vivo character-
ization, Eur. J. Pharm. Biopharm. 86 (2014) 178189.
[48] T.K. Kwon, J.-C. Kim, Preparation and in vitro skin permeation of cubosomes con-
taining Hinokitiol, J. Dispers. Sci. Technol. 31 (2010) 10041009.
[49] T. Kwon, et al., In vitro skin permeation of cubosomes containing water soluble
extracts of Korean barberry, Colloid J. 72 (2010) 205210.
[50] T. Rattanapak, et al., Transcutaneous immunization using microneedles and cub-
osomes: mechanistic investigations using Optical Coherence Tomography and
Two-Photon Microscopy, J. Control. Release 172 (2013) 894903.
[51] S.R. Seo, et al., In vivo hair growth-promoting efficacies of herbal extracts and their
cubosomal suspensions, J. Ind. Eng. Chem. 19 (2013) 13311339.
[52] S. Sherif, E.R. Bendas, S. Badawy, The clinical efficacy of cosmeceutical application of
liquid crystalline nanostructured dispersions of alpha lipoic acid as anti-wrinkle, Eur. J.
Pharm. Biopharm. 86 (2014) 251259.
[53] T. Rattanapak, et al., Comparative study of liposomes, transfersomes, ethosomes and
cubosomes for transcutaneous immunisation: characterisation and in vitro skin pene-
tration, J. Pharm. Pharmacol. 64 (2012) 15601569.
[54] T.K. Kwon, S.K. Hong, J.C. Kim, In vitro skin permeation of cubosomes containing
triclosan, J. Ind. Eng. Chem. 18 (2012) 563567.
[55] S.K. Hong, J.Y. Ma, J.-C. Kim, In vitro skin permeation enhancement of KIOM-
MA-128 by monoolein cubosomes, J. Dispers. Sci. Technol. 33 (2012) 15031508.
[56] T.K. Kwon, J.-C. Kim, In vitro skin permeation and anti-atopic efficacy of lipid
nanocarriers containing water soluble extracts of Houttuynia cordata, Drug Dev.
Ind. Pharm. 40 (2014) 13501357.
[57] R.K. Thapa, et al., Preparation, characterization, and release study of Tacrolimus-
loaded liquid crystalline nanoparticles, J. Dispers. Sci. Technol. 34 (2013) 7277.
[58] E. Esposito, et al., Clotrimazole nanoparticle gel for mucosal administration, Mater.
Sci. Eng. C 33 (2013) 411418.
[59] L. Gan, et al., Self-assembled liquid crystalline nanoparticles as a novel ophthalmic
delivery system for dexamethasone: improving preocular retention and ocular bio-
availability, Int. J. Pharm. 396 (2010) 179187.
[60] S. Han, et al., Novel vehicle based on cubosomes for ophthalmic delivery of
flurbiprofen with low irritancy and high bioavailability, Acta Pharmacol. Sin.
31 (2010) 990998.
[61] Y. Chen, et al., Ocular delivery of cyclosporine A based on glyceryl monooleate/
poloxamer 407 liquid crystalline nanoparticles: preparation, characterization,
in vitro corneal penetration and ocular irritation, J. Drug Target. 20 (2012) 856863.
[62] X. Gong, et al., Nanostructured self-assembly materials from neat and aqueous solu-
tions of C18 lipid pro-drug analogues of capecitabinea chemotherapy agent. Focus
on nanoparticulate cubosomes (TM) of the oleyl analogue, Soft Matter 7 (2011)
57645776.
[63] Y.S. Tu, et al., Preparation, characterisation and evaluation of curcumin with piperine-
loaded cubosome nanoparticles, J. Microencapsul. 31 (2014) 551559.
[64] Z. Yang, et al., Optimization of the preparation process for an oral phytantriol-based
amphotericin B cubosomes. J. Nanomater. 2011 (2011), http://dx.doi.org/
10.1155/2011/308016.
ARTICLE IN PRESS

50 Josephine Y.T. Chong et al.

[65] Z. Yang, et al., Development of amphotericin B-loaded cubosomes through the


SolEmuls technology for enhancing the oral bioavailability, AAPS PharmSciTech
13 (2012) 14831491.
[66] T.-H. Nguyen, et al., Nanostructured liquid crystalline particles provide long duration
sustained-release effect for a poorly water soluble drug after oral administration,
J. Control. Release 153 (2011) 180186.
[67] J. Lai, et al., Pharmacokinetics and enhanced oral bioavailability in beagle dogs of
cyclosporine A encapsulated in glyceryl monooleate/poloxamer 407 cubic
nanoparticles, Int. J. Nanomed. 5 (2010) 1323.
[68] L. Dian, et al., Cubic phase nanoparticles for sustained release of ibuprofen: formula-
tion, characterization, and enhanced bioavailability study, Int. J. Nanomed. 8 (2013)
845854.
[69] H. Chung, et al., Self-assembled nanocubicle as a carrier for peroral insulin delivery,
Diabetologia 45 (2002) 448451.
[70] X. Jin, et al., A nanostructured liquid crystalline formulation of 20(S)-protopanaxadiol
with improved oral absorption, Fitoterapia 84 (2013) 6471.
[71] X. Jin, et al., Enhanced oral absorption of 20(S)-protopanaxadiol by self-assembled liq-
uid crystalline nanoparticles containing piperine: in vitro and in vivo studies, Int. J.
Nanomed. 8 (2013) 641652.
[72] J. Lai, et al., Glyceryl monooleate/poloxamer 407 cubic nanoparticles as oral drug
delivery systems: I. In vitro evaluation and enhanced oral bioavailability of the poorly
water-soluble drug Simvastatin, AAPS PharmSciTech 10 (2009) 960966.
[73] D. Tamayo-Esquivel, et al., Evaluation of the enhanced oral effect of omapatrilat
monolein nanoparticles prepared by the emulsification-diffusion method,
J. Nanosci. Nanotechnol. 6 (2006) 31343138.
[74] N.K. Swarnakar, K. Thanki, S. Jain, Lyotropic liquid crystalline nanoparticles of
CoQ10: implication of lipase digestibility on oral bioavailability, in vivo antioxidant
activity, and in vitroin vivo relationships, Mol. Pharm. 11 (2014) 14351449.
[75] N. Zeng, et al., Lipid-based liquid crystalline nanoparticles as oral drug delivery vehi-
cles for poorly water-soluble drugs: cellular interaction and in vivo absorption, Int. J.
Nanomed. 7 (2012) 37033718.
[76] S.K. Hong, J.Y. Ma, J.-C. Kim, Preparation and characterization of cubosomal
KIOM-C suspension and investigation on in vitro small intestinal absorption of
Baicalin, J. Dispers. Sci. Technol. 34 (2013) 252258.
[77] S.K. Hong, J.Y. Ma, J.-C. Kim, In vitro small intestinal absorption enhancement of
S-164 by monoolein cubic phase nanoparticles, J. Dispers. Sci. Technol. 34 (2013)
511515.
[78] H. Wu, et al., A novel small odorranalectin-bearing cubosomes: preparation, brain
delivery and pharmacodynamic study on amyloid-beta(2535)-treated rats following
intranasal administration, Eur. J. Pharm. Biopharm. 80 (2012) 368378.
[79] E. Esposito, et al., Cubosome dispersions as delivery systems for percutaneous admin-
istration of Indomethacin, Pharm. Res. 22 (2005) 21632173.
[80] E. Esposito, et al., Nanoparticulate lipid dispersions for bromocriptine delivery:
characterization and in vivo study, Eur. J. Pharm. Biopharm. 80 (2012) 306314.
[81] S.B. Rizwan, et al., Cubosomes containing the adjuvants imiquimod and mono-
phosphoryl lipid A stimulate robust cellular and humoral immune responses,
J. Control. Release 165 (2013) 1621.
[82] S. Gordon, et al., Chitosan hydrogels containing liposomes and cubosomes as partic-
ulate sustained release vaccine delivery systems, J. Liposome Res. 22 (2012) 193204.
[83] V. Jain, et al., Paclitaxel loaded PEGylated gleceryl monooleate based nanoparticulate
carriers in chemotherapy, Biomaterials 33 (2012) 72067220.
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 51

[84] M. Johnsson, et al., Physicochemical and drug delivery aspects of lipid-based liquid
crystalline nanoparticles: a case study of intravenously administered propofol,
J. Nanosci. Nanotechnol. 6 (2006) 30173024.
[85] C. Cervin, et al., A combined in vitro and in vivo study on the interactions between
somatostatin and lipid-based liquid crystalline drug carriers and bilayers, Eur. J. Pharm.
Sci. 36 (2009) 377385.
[86] H. Liu, et al., Protein-bearing cubosomes prepared by liquid precursor dilution: inner
ear delivery and pharmacokinetic study following intratympanic administration,
J. Biomed. Nanotechnol. 9 (2013) 17841793.
[87] S.B. Rizwan, et al., Preparation of phytantriol cubosomes by solvent precursor dilution
for the delivery of protein vaccines, Eur. J. Pharm. Biopharm. 79 (2011) 1522.
[88] B. Angelov, et al., Protein-containing PEGylated cubosomic particles: freeze-fracture
electron microscopy and synchrotron radiation circular dichroism study, J. Phys.
Chem. B 116 (2012) 76767686.
[89] B. Angelov, et al., Multicompartment lipid cubic nanoparticles with high protein
upload: millisecond dynamics of formation, ACS Nano 8 (2014) 52165226.
[90] H.-H. Shen, et al., Targeted detection of phosphatidylserine in biomimetic membranes
and in vitro cell systems using annexin V-containing cubosomes, Biomaterials
34 (2013) 83618369.
[91] E. Esposito, et al., Curcumin containing monoolein aqueous dispersions: a
preformulative study, Mater. Sci. Eng. C 33 (2013) 49234934.
[92] S. Murgia, et al., Drug-loaded fluorescent cubosomes: versatile nanoparticles for
potential theranostic applications, Langmuir 29 (2013) 66736679.
[93] C. Caltagirone, et al., Cancer-cell-targeted theranostic cubosomes, Langmuir
30 (2014) 62286236.
[94] D. Bei, J. Marszalek, B.B.C. Youan, Formulation of dacarbazine-loaded cubosomes
Part I: influence of formulation variables, AAPS PharmSciTech 10 (2009) 10321039.
[95] D. Bei, J. Marszalek, B.B.C. Youan, Formulation of dacarbazine-loaded cubosomes
Part II: influence of process parameters, AAPS PharmSciTech 10 (2009) 10401047.
[96] D. Bei, et al., Formulation of dacarbazine-loaded cubosomes. Part III. Physicochem-
ical characterization, AAPS PharmSciTech 11 (2010) 12431249.
[97] R. Efrat, et al., Solubilization of hydrophobic guest molecules in the monoolein dis-
continuous Q(L) cubic mesophase and its soft nanoparticles, Langmuir 25 (2009)
13161326.
[98] B.J. Boyd, Characterisation of drug release from cubosomes using the pressure ultra-
filtration method, Int. J. Pharm. 260 (2003) 239247.
[99] X. Gong, et al., Lyotropic liquid crystalline self-assembly material behavior and
nanoparticulate dispersions of a phytanyl pro-drug analogue of capecitabinea che-
motherapy agent, ACS Appl. Mater. Interfaces 3 (2011) 15521561.
[100] T.C. Le, et al., Predicting the complex phase behavior of self-assembling drug delivery
nanoparticles, Mol. Pharm. 10 (2013) 13681377.
[101] G. Mangiapia, et al., Cubosomes for ruthenium complex delivery: formulation and
characterization, Soft Matter 7 (2011) 1057710580.
[102] J. Barauskas, et al., Interactions of lipid-based liquid crystalline nanoparticles with
model and cell membranes, Int. J. Pharm. 391 (2010) 284291.
[103] J.C. Bode, et al., Interaction of dispersed cubic phases with blood components, Int. J.
Pharm. 448 (2013) 8795.
[104] S.B. Rizwan, et al., Bicontinuous cubic liquid crystals as sustained delivery systems for
peptides and proteins, Expert Opin. Drug Deliv. 7 (2010) 11331144.
[105] M.J. Lawrence, Surfactant systems: their use in drug delivery, Chem. Soc. Rev.
23 (1994) 417424.
ARTICLE IN PRESS

52 Josephine Y.T. Chong et al.

[106] X. Mulet, B.J. Boyd, C.J. Drummond, Advances in drug delivery and medical imaging
using colloidal lyotropic liquid crystalline dispersions, J. Colloid Interface Sci.
393 (2013) 120.
[107] G.Z. Liu, et al., Colloidal amphiphile self-assembly particles composed of gadolinium
oleate and myverol: evaluation as contrast agents for magnetic resonance imaging,
Langmuir 26 (2010) 23832391.
[108] B.W. Muir, et al., Metal-free and MRI visible theranostic lyotropic liquid crystal
nitroxide-based nanoparticles, Biomaterials 33 (2012) 27232733.
[109] S.J. Fraser, et al., Development of cubosomes as a cell-free biosensing platform, Aust. J.
Chem. 64 (2011) 4653.
[110] I. Amar-Yuli, et al., Solubilization of food bioactives within lyotropic liquid crystalline
mesophases, Curr. Opin. Colloid Interface Sci. 14 (2009) 2132.
[111] K. Larsson, Lyotropic liquid crystals and their dispersions relevant in foods, Curr.
Opin. Colloid Interface Sci. 14 (2009) 1620.
[112] P.P. Nadiminti, et al., Nanostructured liquid crystalline particles as an alternative deliv-
ery vehicle for plant agrochemicals, ACS Appl. Mater. Interfaces 5 (2013) 18181826.
[113] B.J. Boyd, Y.D. Dong, T. Rades, Nonlamellar liquid crystalline nanostructured par-
ticles: advances in materials and structure determination, J. Liposome Res. 19 (2009)
1228.
[114] C. Guo, et al., Lyotropic liquid crystal systems in drug delivery, Drug Discov. Today
15 (2010) 10321040.
[115] H. Ljusberg-Wahern, L. Nyberg, K. Larsson, Dispersion of the cubic liquid crystalline
phasestructure preparation and functionality aspects, Chim. Oggi 14 (1996) 4043.
[116] D. Libster, et al., Soft matter dispersions with ordered inner structures, stabilized by
ethoxylated phytosterols, Colloids Surf. B 74 (2009) 202215.
[117] I. Amar-Yuli, et al., Hexosome and hexagonal phases mediated by hydration and poly-
meric stabilizer, Langmuir 23 (2007) 36373645.
[118] J.Y.T. Chong, et al., Accelerated stability assay (ASA) for colloidal systems, ACS
Comb. Sci. 16 (2014) 205210.
[119] K. Larsson, Cubic lipidwater phasesstructures and biomembrane aspects, J. Phys.
Chem. 93 (1989) 73047314.
[120] A.J. Tilley, C.J. Drummond, B.J. Boyd, Disposition and association of the steric sta-
bilizer Pluronic (R) F127 in lyotropic liquid crystalline nanostructured particle disper-
sions, J. Colloid Interface Sci. 392 (2013) 288296.
[121] K. Lindell, et al., Influence of a charged phospholipid on the release pattern of timolol
maleate from cubic liquid crystalline phases, in: B. Lindman, B.W. Ninham (Eds.), The
Colloid Science of Lipids, Steinkopff, 1998, pp. 111118.
[122] K. Nishikawa, H. Arai, K. Inoue, Scavenger receptor-mediated uptake and metabo-
lism of lipid vesicles containing acidic phospholipids by mouse peritoneal macro-
phages, J. Biol. Chem. 265 (1990) 52265231.
[123] K. Funato, R. Yoda, H. Kiwada, Contribution of complement system on destabiliza-
tion of liposomes composed of hydrogenated egg phosphatidylcholine in rat fresh
plasma, Biochim. Biophys. Acta 1103 (1992) 198204.
[124] J. Senior, Fate and behavior of liposomes in vivo: a review of controlling factors, Crit.
Rev. Ther. Drug Carrier Syst. 3 (1987) 123193.
[125] A. Chonn, P. Cullis, D. Devine, The role of surface charge in the activation of the
classical and alternative pathways of complement by liposomes, J. Immunol.
146 (1991) 42344241.
[126] M.L. Immordino, F. Dosio, L. Cattel, Stealth liposomes: review of the basic science,
rationale, and clinical applications, existing and potential, Int. J. Nanomed. 1 (2006)
297315.
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 53

[127] U. Wattendorf, H.P. Merkle, PEGylation as a tool for the biomedical engineering of
surface modified microparticles, J. Pharm. Sci. 97 (2008) 46554669.
[128] E. Ostuni, et al., A survey of structureproperty relationships of surfaces that resist the
adsorption of protein, Langmuir 17 (2001) 56055620.
[129] J.M. Harris, N. Martin, M. Modi, Pegylation, Clin. Pharmacokinet. 40 (2001)
539551.
[130] J.M. Harris, R.B. Chess, Effect of pegylation on pharmaceuticals, Nat. Rev. Drug
Discov. 2 (2003) 214221.
[131] V.P. Torchilin, Immunoliposomes and PEGylated immunoliposomes: possible use for
targeted delivery of imaging agents, Immunomethods 4 (1994) 244258.
[132] A.L. Klibanov, et al., Amphipathic polyethyleneglycols effectively prolong the circu-
lation time of liposomes, FEBS Lett. 268 (1990) 235237.
[133] R. Gref, et al., Stealth corona-core nanoparticles surface modified by polyethylene
glycol (PEG): influences of the corona (PEG chain length and surface density) and
of the core composition on phagocytic uptake and plasma protein adsorption, Colloids
Surf. B 18 (2000) 301313.
[134] M. Morra, On the molecular basis of fouling resistance, J. Biomater. Sci. Polym. Ed.
11 (2000) 547569.
[135] S.I. Jeon, J.D. Andrade, Proteinsurface interactions in the presence of polyethylene
oxide: II. Effect of protein size, J. Colloid Interface Sci. 142 (1991) 159166.
[136] S.I. Jeon, et al., Proteinsurface interactions in the presence of polyethylene oxide: I.
Simplified theory, J. Colloid Interface Sci. 142 (1991) 149158.
[137] A. Halperin, Polymer brushes that resist adsorption of model proteins: design param-
eters, Langmuir 15 (1999) 25252533.
[138] B. Zhu, et al., Chain-length dependence of the protein and cell resistance of
oligo(ethylene glycol)-terminated self-assembled monolayers on gold, J. Biomed.
Mater. Res. 56 (2001) 406416.
[139] K.L. Prime, G.M. Whitesides, Adsorption of proteins onto surfaces containing end-
attached oligo(ethylene oxide): a model system using self-assembled monolayers,
J. Am. Chem. Soc. 115 (1993) 1071410721.
[140] T. McPherson, et al., Prevention of protein adsorption by tethered poly(ethylene
oxide) layers: experiments and single-chain mean-field analysis, Langmuir 14 (1998)
176186.
[141] M. Malmsten, K. Emoto, J.M. Van Alstine, Effect of chain density on inhibition of
protein adsorption by poly(ethylene glycol) based coatings, J. Colloid Interface Sci.
202 (1998) 507517.
[142] P. Kingshott, H. Thissen, H.J. Griesser, Effects of cloud-point grafting, chain length,
and density of PEG layers on competitive adsorption of ocular proteins, Biomaterials
23 (2002) 20432056.
[143] S. Pasche, et al., Poly(L-lysine)-graft-poly(ethylene glycol) assembled monolayers on
niobium oxide surfaces: a quantitative study of the influence of polymer interfacial
architecture on resistance to protein adsorption by ToF-SIMS and in situ OWLS,
Langmuir 19 (2003) 92169225.
[144] J.V. Jokerst, et al., Nanoparticle PEGylation for imaging and therapy, Nanomedicine
6 (2011) 715728.
[145] C. Thies, Adsorption of styrene/butadiene copolymers and stabilization of silica dis-
persed in perchloroethylene, J. Colloid Interface Sci. 54 (1976) 1321.
[146] W. Heller, T.L. Pugh, Steric protection of hydrophobic colloidal particles by
adsorption of flexible macromolecules, J. Chem. Phys. 22 (1954) 1778.
[147] J. Lee, P.A. Martic, J.S. Tan, Protein adsorption on pluronic copolymer-coated poly-
styrene particles, J. Colloid Interface Sci. 131 (1989) 252266.
ARTICLE IN PRESS

54 Josephine Y.T. Chong et al.

[148] J.A. Neff, K.D. Caldwell, P.A. Tresco, A novel method for surface modification to
promote cell attachment to hydrophobic substrates, J. Biomed. Mater. Res.
40 (1998) 511519.
[149] Y.-D. Dong, et al., Bulk and dispersed aqueous phase behavior of phytantriol: effect of
vitamin E acetate and F127 polymer on liquid crystal nanostructure, Langmuir
22 (2006) 95129518.
[150] B.J. Boyd, et al., Self-assembled geometric liquid-crystalline nanoparticles imaged in
three dimensions: hexosomes are not necessarily flat hexagonal prisms, Langmuir
23 (2007) 1246112464.
[151] F. Muller, A. Salonen, O. Glatter, Phase behavior of phytantriol/water bicontinuous
cubic Pn3m cubosomes stabilized by laponite disc-like particles, J. Colloid Interface
Sci. 342 (2010) 392398.
[152] J. Zhai, et al., Revisiting -casein as a stabilizer for lipid liquid crystalline nanostruc-
tured particles, Langmuir 27 (2011) 1475714766.
[153] T. Kojarunchitt, et al., Development and characterisation of modified poloxamer 407
thermoresponsive depot systems containing cubosomes, Int. J. Pharm. 408 (2011)
2026.
[154] H.H. Shen, et al., The influence of dipalmitoyl phosphatidylserine on phase behaviour
of and cellular response to lyotropic liquid crystalline dispersions, Biomaterials
31 (2010) 94739481.
[155] C.E. Conn, et al., Enhanced uptake of an integral membrane protein, the dopamine
D2L receptor, by cubic nanostructured lipid nanoparticles doped with Ni(II) chelated
EDTA amphiphiles, Soft Matter 7 (2011) 567578.
[156] H.H. Shen, et al., The interaction of cubosomes with supported phospholipid bilayers
using neutron reflectometry and QCM-D, Soft Matter 7 (2011) 80418049.
[157] S.J. Fraser, et al., Surface immobilization of bio-functionalized cubosomes: sensing of
proteins by quartz crystal microbalance, Langmuir 28 (2012) 620627.
[158] T.E. Hartnett, et al., Size and phase control of cubic lyotropic liquid crystal
nanoparticles, J. Phys. Chem. B 118 (2014) 74307439.
[159] S.J. Fraser, et al., Controlling nanostructure and lattice parameter of the inverse
bicontinuous cubic phases in functionalised phytantriol dispersions, J. Colloid Interface
Sci. 408 (2013) 117124.
[160] J.Y.T. Chong, et al., Steric stabilisation of self-assembled cubic lyotropic liquid
crystalline nanoparticles: high throughput evaluation of triblock polyethylene oxide
polypropylene oxidepolyethylene oxide copolymers, Soft Matter 7 (2011) 47684777.
[161] M. Uyama, et al., Useful modified cellulose polymers as new emulsifiers of cubosomes,
Langmuir 25 (2009) 43364338.
[162] T. Abraham, M. Hato, M. Hirai, Glycolipid based cubic nanoparticles: preparation and
structural aspects, Colloids Surf. B 35 (2004) 107118.
[163] T. Abraham, M. Hato, M. Hirai, Polymer-dispersed bicontinuous cubic glycolipid
nanoparticles, Biotechnol. Prog. 21 (2005) 255262.
[164] L. de Campo, et al., Reversible phase transitions in emulsified nanostructured lipid
systems, Langmuir 20 (2004) 52545261.
[165] A. Yaghmur, et al., Oil-loaded monolinolein-based particles with confined inverse
discontinuous cubic structure (Fd3m), Langmuir 22 (2005) 517521.
[166] A. Yaghmur, et al., Emulsified microemulsions and oil-containing liquid crystalline
phases, Langmuir 21 (2004) 569577.
[167] A. Salonen, S. Guillot, O. Glatter, Determination of water content in internally self-
assembled monoglyceride-based dispersions from the bulk phase, Langmuir 23 (2007)
91519154.
[168] S. Guillot, et al., Direct and indirect thermal transitions from hexosomes to emulsified
micro-emulsions in oil-loaded monoglyceride-based particles, Colloids Surf. A
291 (2006) 7884.
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 55

[169] A. Yaghmur, et al., Control of the internal structure of MLO-based isasomes by the
addition of diglycerol monooleate and soybean phosphatidylcholine, Langmuir
22 (2006) 99199927.
[170] A. Yaghmur, et al., Self-assembly in monoelaidin aqueous dispersions: direct vesicles to
cubosomes transition, PLoS One 3 (2008) e3747.
[171] E. Esposito, et al., Lipid-based supramolecular systems for topical application: a
preformulatory study, AAPS PharmSci 5 (2003) 6276.
[172] J. Gustafsson, et al., Cubic lipidwater phase dispersed into submicron particles,
Langmuir 12 (1996) 46114613.
[173] J. Gustafsson, et al., Submicron particles of reversed lipid phases in water stabilized by a
nonionic amphiphilic polymer, Langmuir 13 (1997) 69646971.
[174] K. Larsson, Colloidal dispersions of ordered lipidwater phases, J. Dispers. Sci.
Technol. 20 (1999) 2734.
[175] K. Larsson, Aqueous dispersions of cubic lipidwater phases, Curr. Opin. Colloid
Interface Sci. 5 (2000) 6469.
[176] K. Larsson, Bicontinuous cubic lipidwater particles and cubosomal dispersions,
in: T. Osamu (Ed.), Studies in Surface Science and Catalysis, Elsevier, 2004, pp. 4151.
[177] C. Neto, et al., Imaging soft matter with the atomic force microscope: cubosomes and
hexosomes, J. Phys. Chem. B 103 (1999) 38963899.
[178] P.T. Spicer, et al., Novel process for producing cubic liquid crystalline nanoparticles
(cubosomes), Langmuir 17 (2001) 57485756.
[179] J. Barauskas, et al., Cubic phase nanoparticles (cubosome): principles for controlling
size, structure, and stability, Langmuir 21 (2005) 25692577.
[180] M. Monduzzi, H. Ljusberg-Wahren, K. Larsson, A 13C NMR study of aqueous dis-
persions of reversed lipid phases, Langmuir 16 (2000) 73557358.
[181] M. Nakano, et al., Small-angle X-ray scattering and 13C NMR investigation on the
internal structure of cubosomes Langmuir 17 (2001) 39173922.
[182] O. Svensson, K. Thuresson, T. Arnebrant, Interactions between chitosan-modified
particles and mucin-coated surfaces, J. Colloid Interface Sci. 325 (2008) 346350.
[183] O. Svensson, K. Thuresson, T. Arnebrant, Interactions between drug delivery particles
and mucin in solution and at interfaces, Langmuir 24 (2008) 25732579.
[184] P. Vandoolaeghe, et al., Adsorption of cubic liquid crystalline nanoparticles on model
membranes, Soft Matter 4 (2008) 22672277.
[185] P. Vandoolaeghe, et al., Neutron reflectivity studies of the interaction of cubic-phase
nanoparticles with phospholipid bilayers of different coverage, Langmuir 25 (2009)
40094020.
[186] P. Vandoolaeghe, et al., Adsorption of intact cubic liquid crystalline nanoparticles on
hydrophilic surfaces: lateral organization, interfacial stability, layer structure, and inter-
action mechanism, J. Phys. Chem. C 113 (2009) 44834494.
[187] T. Landh, Phase-behavior in the system pine oil monoglycerides-poloxamer-407-
water at 20-degrees-C, J. Phys. Chem. 98 (1994) 84538467.
[188] A. Gupta, et al., Nanoassemblies of Gd-DTPA-monooleyl and glycerol monooleate
amphiphiles as potential MRI contrast agents, J. Mater. Chem. B 2 (2014) 12251233.
[189] B. Siekmann, et al., Preparation and structural investigations of colloidal dispers-
ions prepared from cubic monoglyceridewater phases, Int. J. Pharm. 244 (2002)
3343.
[190] M. Nakano, et al., Dispersions of liquid crystalline phases of the monoolein/oleic acid/
Pluronic F127 system, Langmuir 18 (2002) 92839288.
[191] G. Popescu, et al., Liquid crystalline phases and their dispersions in aqueous mixtures of
glycerol monooleate and glyceryl monooleyl ether, Langmuir 23 (2006) 496503.
[192] A. Salonen, F. Muller, O. Glatter, Dispersions of internally liquid crystalline systems
stabilized by charged disklike particles as pickering emulsions: basic properties and
time-resolved behavior, Langmuir 24 (2008) 53065314.
ARTICLE IN PRESS

56 Josephine Y.T. Chong et al.

[193] F. Muller, A. Salonen, O. Glatter, Monoglyceride-based cubosomes stabilized by lap-


onite: separating the effects of stabilizer, pH and temperature, Colloids Surf. A
358 (2010) 5056.
[194] A. Salonen, et al., Material transfer in cubosomeemulsion mixtures: effect of alkane
chain length, Langmuir 26 (2010) 1067010676.
[195] L. Sagalowicz, et al., Influence of vitamin E acetate and other lipids on the phase
behavior of mesophases based on unsaturated monoglycerides, Langmuir 29 (2013)
82228232.
[196] C. Nilsson, et al., PEGylation of phytantriol-based lyotropic liquid crystalline
particlesthe effect of lipid composition, PEG chain length, and temperature on
the internal nanostructure, Langmuir 30 (2014) 63986407.
[197] R. Koynova, B. Tenchov, G. Rapp, Effect of PEG-lipid conjugates on the phase
behavior of phosphatidylethanolamine dispersions, Colloids Surf. A 149 (1999)
571575.
[198] R. Koynova, B. Tenchov, G. Rapp, Low amounts of PEG-lipid induce cubic phase in
phosphatidylethanolamine dispersions, Biochim. Biophys. Acta 1326 (1997) 167170.
[199] M. Johnsson, J. Barauskas, F. Tiberg, Cubic phases and cubic phase dispersions in a
phospholipid-based system, J. Am. Chem. Soc. 127 (2005) 10761077.
[200] M. Johnsson, K. Edwards, Phase behavior and aggregate structure in mixtures of
dioleoylphosphatidylethanolami
ne and poly(ethylene glycol)-lipids, Biophys. J. 80 (2001) 313323.
[201] J.Y.T. Chong, et al., Novel steric stabilizers for lyotropic liquid crystalline
nanoparticles: PEGylated-phytanyl copolymers. Langmuir (2014), http://dx.doi.
org/10.1021/la501471z.
[202] S. Rangelov, M. Almgren, Particulate and bulk bicontinuous cubic phases obtained
from mixtures of glyceryl monooleate and copolymers bearing blocks of lipid-mimetic
anchors in water, J. Phys. Chem. B 109 (2005) 39213929.
[203] J.Y.T. Chong, et al., High-throughput discovery of novel steric stabilizers for cubic
lyotropic liquid crystal nanoparticle dispersions, Langmuir 28 (2012) 92239232.
[204] B. Angelov, et al., Earliest stage of the tetrahedral nanochannel formation in cubosome
particles from unilamellar nanovesicles, Langmuir 28 (2012) 1664716655.
[205] N. Zeng, et al., Preparation and characterization of paclitaxel-loaded DSPE-PEG-
liquid crystalline nanoparticles (LCNPs) for improved bioavailability, Int. J. Pharm.
424 (2012) 5866.
[206] M. Almgren, S. Rangelov, Polymorph dispersed particles from the bicontinuous cubic
phase of glycerol monooleate stabilized by PEG-copolymers with lipid-mimetic
hydrophobic anchors, J. Dispers. Sci. Technol. 27 (2006) 599609.
[207] J.Y.T. Chong, et al., Novel RAFT amphiphilic brush copolymer steric stabilisers for
cubosomes: poly(octadecyl acrylate)-block-poly(polyethylene glycol methyl ether
acrylate), Soft Matter 10 (2014) 66666676.
[208] J. Gustafsson, et al., Phase behavior and aggregate structure in aqueous mixtures of
sodium cholate and glycerol monooleate, J. Colloid Interface Sci. 211 (1999) 326335.
[209] P.T. Spicer, et al., Dry powder precursors of cubic liquid crystalline nanoparticles
(cubosomes), J. Nanopart. Res. 4 (2002) 297311.
[210] A. Salonen, F.o. Muller, O. Glatter, Internally self-assembled submicrometer emul-
sions stabilized by spherical nanocolloids: finding the free nanoparticles in the aqueous
continuous phase, Langmuir 26 (2010) 79817987.
[211] P. Alexandridis, Amphiphilic copolymers and their applications, Curr. Opin. Colloid
Interface Sci. 1 (1996) 490501.
[212] A. Mori, et al., Influence of the steric barrier activity of amphipathic
poly(ethyleneglycol) and ganglioside GM1 on the circulation time of liposomes and
on the target binding of immunoliposomes in vivo, FEBS Lett. 284 (1991) 263266.
ARTICLE IN PRESS

Steric Stabilizers for Cubosomes 57

[213] M.-S. Martina, et al., The in vitro kinetics of the interactions between PEG-ylated
magnetic-fluid-loaded liposomes and macrophages, Biomaterials 28 (2007)
41434153.
[214] M.-S. Martina, C. Wilhelm, S. Lesieur, The effect of magnetic targeting on the uptake
of magnetic-fluid-loaded liposomes by human prostatic adenocarcinoma cells,
Biomaterials 29 (2008) 41374145.
[215] J. Patton, M. Carey, Watching fat digestion, Science 204 (1979) 145148.
[216] M. Lindstrom, et al., Aqueous lipid phases of relevance to intestinal fat digestion and
absorption, Lipids 16 (1981) 749754.
[217] W. Buchheim, K. Larsson, Cubic lipidproteinwater phases, J. Colloid Interface Sci.
117 (1987) 582583.
[218] M. Almgren, et al., Dispersed lipid liquid crystalline phases stabilized by a
hydrophobically modified cellulose, Langmuir 23 (2007) 27682777.
[219] Y. Luo, et al., Dry coating, a novel coating technology for solid pharmaceutical dosage
forms, Int. J. Pharm. 358 (2008) 1622.
[220] C.D. Kablitz, M. Kappl, N.A. Urbanetz, Parameters influencing polymer particle lay-
ering of the dry coating process, Eur. J. Pharm. Biopharm. 69 (2008) 760768.
[221] H. Konno, et al., Effect of polymer type on the dissolution profile of amorphous solid
dispersions containing felodipine, Eur. J. Pharm. Biopharm. 70 (2008) 493499.
[222] C.D. Driever, et al., Layer-by-layer polymer coating on discrete particles of cubic
lyotropic liquid crystalline dispersions (cubosomes), Langmuir 29 (2013)
1289112900.
[223] B. Angelov, et al., DNA/fusogenic lipid nanocarrier assembly: millisecond structural
dynamics, J. Phys. Chem. Lett. 4 (2013) 19591964.
[224] J.D. Du, et al., A novel approach to enhance the mucoadhesion of lipid drug
nanocarriers for improved drug delivery to the buccal mucosa, Int. J. Pharm.
471 (2014) 358365.

Vous aimerez peut-être aussi