You are on page 1of 9

Journal of Colloid and Interface Science 605 (2022) 146–154

Contents lists available at ScienceDirect

Journal of Colloid and Interface Science


journal homepage: www.elsevier.com/locate/jcis

Comparison of cubosomes and hexosomes for the delivery of phenytoin


to the brain
Younus Mohammad a,1, Richard N. Prentice a,1, Ben J. Boyd b, Shakila B. Rizwan a,⇑
a
School of Pharmacy, University of Otago, Dunedin, New Zealand
b
Drug Delivery, Disposition and Dynamics and ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences,
Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia

g r a p h i c a l a b s t r a c t

a r t i c l e i n f o a b s t r a c t

Article history: The ability to formulate cubosomes and hexosomes with a single lipid by changing only the colloidal sta-
Received 13 April 2021 biliser presents a unique opportunity to directly compare the biological performance of these uniquely
Revised 8 July 2021 structured nanoparticles. This was explored here via the encapsulation and brain delivery of a model
Accepted 9 July 2021
anti-seizure drug, phenytoin, in selachyl alcohol cubosomes and hexosomes. Nanoparticles were pre-
Available online 18 July 202118 July 2021
pared with PluronicÒ F127 or Tween 80Ò as the stabiliser and characterised. The internal nanostructure
of nanoparticles shifted from hexosomes when using PluronicÒ F127 as the stabiliser to cubosomes when
Keywords:
using Tween 80Ò and was conserved following loading of phenytoin, with high encapsulation efficiencies
Phenytoin
Cubosomes
(>97%) in both particle type. Cytotoxicity towards brain endothelial cells using the hCMEC/D3 line was
Hexosomes comparable regardless of stabiliser type. Finally, in vivo brain delivery of phenytoin encapsulated in cubo-
hCMEC/D3 somes and hexosomes after intravenous administration to rats was studied over a period of 60 min,
Tween 80Ò showing cubosomes to be superior to hexosomes, both in terms of brain concentrations and brain to
PluronicÒ F127 plasma ratio. While the role of stabiliser and/or internal nanostructure remains to be conclusively deter-
Brain mined, this study is the first in vivo comparison of cubosomes and hexosomes for the delivery of a ther-
apeutic drug molecule across the BBB and into the brain.
Ó 2021 Published by Elsevier Inc.

1. Introduction

⇑ Corresponding author. Delivery of therapeutics to the brain is a formidable challenge


E-mail address: shakila.rizwan@otago.ac.nz (S.B. Rizwan). due to the strict regulation of molecular trafficking across the
1
Younus M. and R.N. Prentice contributed equally as first author of the manuscript. blood-brain barrier (BBB). Targeted drug-loaded nanoparticles

https://doi.org/10.1016/j.jcis.2021.07.070
0021-9797/Ó 2021 Published by Elsevier Inc.
Y. Mohammad, R.N. Prentice, B.J. Boyd et al. Journal of Colloid and Interface Science 605 (2022) 146–154

have been suggested as a promising strategy to circumvent this like severe pain [28,29], tissue necrosis [30,31] and ‘‘purple glove”
[1,2]. The approach is largely based on the idea of a biocompatible syndrome (oedema, discoloration, and pain distal to the site of
particle coated with a surface ligand that either interacts with intravenous administration) [32,33]. Such complications could
endothelial cells at the BBB to facilitate entry into the brain or potentially be avoided by encapsulation of phenytoin in aqueous
adsorbs plasma proteins which make the particle appear to be car- dispersible LCNs and delivery in a particulate formulation may also
rying endogenous cargo, thereby mediating entry. A prominent facilitate targeting of drug to the brain, thereby reducing off-target
example of the latter is the surfactant Tween 80Ò, which has been side effects.
shown to adsorb apolipoprotein E and increase brain delivery of Whilst dispersions of selachyl alcohol stabilised with PluronicÒ
therapeutics such as dalargin, doxorubicin and loperamide [3–5]. F127 or Tween 80Ò result in the formation of cubosomes and hex-
Amphiphilic lipids when in contact with aqueous environments osomes [14,19], respectively, the addition of guest molecules are
can self-assemble to produce a variety of thermodynamically known to disrupt the structure of liquid crystalline systems. There-
stable lyotropic liquid crystalline systems. Of these systems, non- fore, we first sought to encapsulate phenytoin in selachyl alcohol
lamellar phases, such as reverse bicontinuous cubic (V2) and and then investigate its effect on particle nanostructure. The
reverse hexagonal (H2) phases, have received considerable interest well-characterised and commonly studied synthetic lipid phy-
in drug delivery research due to their compartmentalised, ordered tantriol was used for comparison in these studies (Fig. 1). Cubo-
internal structure and for their ability to incorporate and sustain somes and hexosomes of selachyl alcohol and encapsulating
the release of a wide range of molecules [6–9]. Dispersing the V2 comparable amounts of phenytoin, where only the surface sta-
phase and the H2 phase in the presence of a colloidal stabiliser biliser differed were successfully prepared. Following on from the
results in the formation of liquid crystalline nanoparticles (LCNs) formulation studies, selachyl alcohol cubosome and hexosome for-
termed cubosomes and hexosomes, respectively [10,11]. Phy- mulations with respect to in vitro cellular toxicity in a human cere-
tantriol (Fig. 1) or glyceryl monooleate (GMO) are two lipids that bral microvascular endothelial cell line (hCMEC/D3) and in vivo
are typically used as building blocks in the production of LCNs brain delivery of phenytoin in rats were then compared in order
[12,13]. More recently, endogenous lipids such as selachyl alcohol to gain insight into the effect of internal structure on drug delivery
have also shown promise in this regard [14,15]. across the BBB.
Selachyl alcohol (Fig. 1) belongs to the family of alkylglycerols,
which are natural ether lipids that exhibit several biological activ- 2. Materials and methods
ities with distinct mechanisms. Of particular interest, co-
administration of alkylglycerols with chemotherapeutic drugs has Selachyl alcohol was obtained from Haihang Industry Co., Ltd,
been shown to dramatically increase their transport across the (China). Phytantriol was purchased from A & E Connock (Hamp-
BBB in both healthy and tumour-afflicted brains, an effect which shire, England). PluronicÒ F127 was obtained from BASF (Lud-
has been attributed to transient opening of tight junctions [16– wigshafen, Germany). TweenÒ 80, and phenytoin were purchased
18] and has been well-established with short-chain alkylglycerols. from Sigma-Aldrich (Australia). Propylene glycol (99.0%), chloro-
Most other biological effects of alkylglycerols have been estab- form, methanol (HPLC grade) and acetonitrile (HPLC grade) were
lished using long-chain molecules, however, neither the effect on sourced from Merck (Germany). All water used in this study was
mediating drug delivery across the BBB, nor their potential as ion exchanged distilled and passed through a Milli-Q water purifi-
self-assembling particulate brain delivery vehicles have yet been cation system (Millipore, Bedford, MA, USA). All the chemicals
investigated for long-chain alkylglycerols. were used as sourced, without any further purification.
Selachyl alcohol is a long-chain alkylglycerol that forms the H2
phase when in excess water, which can be dispersed into hexo-
2.1. Preparation of dispersions
somes with the commonly used stabiliser PluronicÒ F127 [14,19].
Recently we have shown that when selachyl alcohol dispersions
The liquid precursor technique [34] was used to prepare the
are stabilised with Tween 80Ò the nanoparticles possess an inter-
dispersions. Briefly, selachyl alcohol (250 mg), stabiliser (PluronicÒ
nal V2 phase structure [15]. Whilst a lot has been published in
F127 or Tween 80Ò) (37.5 mg) and a co-solvent, propylene glycol
the area of formulation and physicochemical characterisation of
(625 mg) with or without phenytoin (2.5 mg) were weighed into
LCNs, studies of the biological performance of cubosomes and hex-
glass vials. The mixture was dissolved in excess chloroform, which
osomes as drug delivery systems in vivo is lacking. Moreover, to the
was then removed by evaporation under vacuum at 45 °C leaving
best of our knowledge, there are no reports in the literature that
behind a lipid mixture. The lipid mixture was then dispersed in
directly compare the biological performance of these two LCNs.
excess water (5 mL) by vortex mixing with glass beads typically
The need to alter the lipid composition of the particles to induce
for 15 min. All formulations were stored at room temperature.
a V2 to H2 phase change makes this a challenging task [20]. There-
fore, the ability to formulate cubosomes and hexosomes as with
selachyl alcohol by changing only the stabiliser presents a unique 2.2. Particle size analysis
opportunity to directly compare the in vivo performance of these
differently structured nanoparticles. Consequently, the overall Dynamic light scattering (DLS) was used to measure the size
aim of the current study was to compare the performance of sela- (expressed as Z-average) and polydispersity index (PDI) of disper-
chyl alcohol cubosomes and hexosomes, by comparing their encap- sions using a Malvern ZetaSizer NanoÒ (ATA Scientific, Australia)
sulation and brain delivery of a model anti-seizure drug, phenytoin instrument. To adjust the signal level a 5 lL aliquot of the disper-
(Fig. 1). sions was diluted to 1 mL with water. Three sets of measurements,
Phenytoin is used intravenously [21,22] as a first-line treatment each comprising 10 runs of 10 s were performed for each sample at
for status epilepticus [23], a life-threatening, prolonged state of 25 °C and a scattering angle of 173°.
seizure associated with high morbidity and mortality [24,25].
Given its poor water solubility (weak acid, pKa 8.33 and aqueous 2.3. Small angle X-ray scattering (SAXS)
solubility of 7.53  105 M) [26], the commercial parenteral form
of phenytoin is dissolved in 40% propylene glycol and 10% ethanol Phase structure of dispersions was investigated using the SAXS/
and adjusted to a pH of 12 to maintain solubility [27]. Conse- WAXS beamline at the Australian Synchrotron using a previously
quently, intravenous administration often results in complications described method [35]. A 96-well plate pre-loaded with samples
147
Y. Mohammad, R.N. Prentice, B.J. Boyd et al. Journal of Colloid and Interface Science 605 (2022) 146–154

Phytantriol Tween 80®


(w+x+y+z) = 20

Selachyl alcohol

Pluronic® F127
Phenytoin

Fig. 1. Chemical structures of the lipids (selachyl alcohol and phytantriol), drug (phenytoin) and colloidal stabilisers (Tween 80Ò and PluronicÒ F127) used in this study.

(200 lL) and sealed with an adhesive film was mounted vertically ysis for quantification of phenytoin using a method modified after
in the beam path. The samples were exposed to the X-rays (13 keV) [38,39]. An Agilent HPLC (Agilent Technologies 1200 series)
for 1 s at 25 °C and a Pilatus 1 M detector (total area: equipped with UV detector was used. Samples were subjected to
169  180 mm; pixel size: 172 lm  170 lm) placed at a distance a separation on a Phenomenex Gemini C18 column (150 mm  4.
of 1532 mm from the samples was used to generate two dimen- 6 mm, 5 lm) column maintained at 25 °C. A mobile phase contain-
sional SAXS patterns. Scatterbrain software was used to integrate ing 60% v/v methanol in water in isocratic mode with a flow rate of
two-dimensional diffraction patterns into one-dimensional scat- 1 mL/min was used. A 25 lL of sample was injected into system
tering functions and were plotted as intensity (I) versus the magni- from the auto sampler which was maintained at 25 °C. Validation
tude of the scattering vector (q) plots. The relative positions of the data for the HPLC assay used to quantify the phenytoin is provided
Bragg peaks, which correspond to their Miller indices were used to in Supplementary Information 2. The standard concentration range
define the phase space groups and the lattice parameters were cal- of phenytoin was 0.5 to 50 lg/mL (Supplementary Fig. 2). The col-
culated using the appropriate expressions [36]. umn eluents were monitored at a wavelength of 225 nm. Pheny-
toin eluted at 4.5 min (Supplementary Fig. 3).
2.4. Cryogenic transmission electron microscopy (cryo-TEM)
2.6. In vitro cytotoxicity
Our previously reported cryo-TEM method was used to image
the morphology of the nanoparticles [37]. A 5 lL aliquot of the dis-
Human cerebral microvascular endothelial cells (hCMEC/D3)
persion was applied to a glow discharged 400 mesh R2/2 Quantifoil
(Cedarlane, Canada) between passage 23 to 34 were grown in
grid (Quantifoil GmbH, Germany). After allowing 10 s adsorption
flasks pre-coated with collagen type 1 (FalconTM) and in EBM-2
time, excess sample was blotted with filter paper (Whatman grade
medium (Lonza, Basel, Switzerland) supplemented with 5% fetal
1) for 3 s to obtain a thin liquid film. The grid was then vitrified by
bovine serum (Life Technologies, NZ), 1% penicillin-streptomycin
rapidly plunging into liquid ethane maintained at 180 °C (Reich-
(Life Technologies, NZ), 1.4 lM hydrocortisone (Sigma-Aldrich),
ert KF80 cryo-fixation device) and stored in liquid nitrogen until
5 lg/mL ascorbic acid (Sigma-Aldrich), 0.001% chemically defined
analysis. Samples were viewed using a JEOL 2200FS TEM with a
lipid concentrate (Life Technologies, NZ), 10 mM HEPES (Life Tech-
Gatan side-entry cryo-stage equipped with a TVIPS F416 CMOS
nologies, NZ) and 1 ng/mL human basic fibroblast growth factor
camera at 20 000 magnification. IMOD software (University of
(Sigma-Aldrich). Cells were grown at a density of 5  104 cells
Colorado, Boulder, CO, USA) was used for image analysis.
per well at 37 °C with 5% CO2 and saturated humidity. Cell culture
medium was replaced every 2 to 3 days and studies were con-
2.5. Quantification of phenytoin encapsulated in nanoparticles ducted when the cells reached 70% confluency, typically 3 to 4 days
after seeding.
Our previously described gel-permeation chromatography A propidium iodide (PI) assay was used to determine cell viabil-
method [37] was used to determine the encapsulation of pheny- ity. Cells were washed three times with Dulbecco’s phosphate buf-
toin in the dispersions. Briefly, dispersions were diluted in water fered saline (Invitrogen Corporation) and incubated with
(0.5–2 mL) and passed through PD-10 columns filled with Sepha- formulations diluted in media at various concentrations of lipid
dex G-25 (GE Healthcare Bio-Sciences Corp, Piscataway, NJ, USA). (12.5, 25, 37.5 and 50 lg/mL without phenytoin; 10, 15, 20, 25
The column was eluted with 24 mL of water and eluents were col- and 30 lg/mL with phenytoin) for 2 h at 37 °C. The cells were then
lected in 3 mL aliquots. Subsequently, methanol (20 mL) was used washed with flow assisted cell sorting (FACS) buffer (1% bovine
to elute the column and 5 mL fractions were collected. The elution serum albumin and 0.1% sodium azide in phosphate buffered sal-
profile of unencapsulated (free) phenytoin was obtained by passing ine) to remove any remaining formulation. TrypLE (Gibco, Den-
the phenytoin solution (2.5 mg of phenytoin, 625 mg of propylene mark) was added to each well and left for 10 min at 37 °C to
glycol were dissolved in chloroform, chloroform was evaporated allow the cells to detach. The cells were re-suspended with FACS
and mixture was suspended in 5 mL water) through the column. buffer and centrifuged for 10 min at 160 g. Supernatants were dis-
The eluents were dissolved in 2:1 acetonitrile: methanol (Merck, carded and the cells were re-suspended in 100 lL of FACS buffer
Germany) (to solubilise nanoparticle components and phenytoin) containing PI. Cells were then analysed using flow cytometry. PI
and were filtered through 0.2 lm nylon filters prior to HPLC anal- fluorescence was quantified using a FACS CantoTM II flow cytome-
148
Y. Mohammad, R.N. Prentice, B.J. Boyd et al. Journal of Colloid and Interface Science 605 (2022) 146–154

ter (BD Biosciences, California, USA) and data acquired using Cell- using unpaired two-tailed t-tests. All the data was analysed in
QuestPro software (BD Biosciences). Data analysis was conducted GraphPad PrismÒ with P < 0.05 considered statistically significant.
using FlowJoÒ 7.6 software (Tree Star, Oregon, USA). Cell viability
was defined as the percentage of PI-negative cells within the single
3. Results
cell population.

3.1. Physicochemical properties of phenytoin containing dispersions


2.7. Intravenous administration of LCNs to animals

Phenytoin is a poorly water-soluble crystalline drug which can


Plasma and brain concentrations of phenytoin and its major
be seen as anisotropic, birefringent crystals under the cross
metabolite, 4-HPPH (5-(4-hydroxyphenyl)-5-phenylhydantoin),
polarised light microscope (CPLM), where crystals as small as
were measured in rats up to 60 min after intravenous administra-
0.2 lm can be observed [41]. CPLM was used to estimate the max-
tion of selachyl alcohol LCNs. The study was approved by the
imum loading of phenytoin in selachyl alcohol and phytantriol by
University of Otago Animal Ethics Committee. Male Wistar rats
the detection of residual drug crystals (method and results are
(average weight ~280 g) were sourced from the Hercus Taieri
summarised in Supplementary Information 1, Supplementary
Resource Unit. LCNs were administered as an intravenous bolus
Table 1 and Supplementary Fig. 1). Based on CPLM studies, concen-
to rats at a dose of 1 mL/kg (equivalent to 0.5 mg/kg phenytoin)
trations of 1% w/w (relative to lipid) of phenytoin was selected and
via a lateral tail vein using a 0.5 mL Lo-dose U-100 insulin syringe
selachyl alcohol and phytantriol dispersions containing phenytoin
with 29 G  12.7 mm needle (BD Biosciences).
were formulated. Table 1 summarises the resulting particle size
distribution and encapsulation efficiency of phenytoin in the dif-
2.8. Tissue collection
ferent dispersions. Encapsulation of phenytoin into nanoparticles
prepared with either selachyl alcohol or phytantriol did not affect
Animals were euthanased by guillotine decapitation after one of
the mean particle size and PDI (P > 0.5) when compared to their
three time points (15, 30 or 60 min) following the injection (n = 4
blank counterparts.
to 5 animals per group for each formulation). Trunk blood was col-
The effect of phenytoin on the nanostructure of the LCNs was
lected in a 6 mL tube coated with sodium heparin (BD Biosciences)
investigated by SAXS (Table 1 and Fig. 2) and cryo-TEM (Fig. 3).
at the time of euthanasia and centrifuged at 2000 g for 10 min at
Phytantriol dispersions stabilised with PluronicÒ F127, and con-
ambient temperature (Heraeus Multifuge X3FR, Thermo Scientific),
taining no phenytoin displayed four Bragg peaks with relative posi-
after which the plasma supernatant was extracted. Brains were p p p p
tions at ratios of 2: 3: 4: 6 indicating the presence of V2 phase
also dissected and rinsed in PBS. Both plasma and brain samples
particles with Pn3m space group. The lattice parameter of 70.5 ± 0.
were frozen at 80 °C until required for LC-MS analysis.
12 Å (Table 1) calculated from the relative peak positions was in
agreement with our previous findings [15]. Phytantriol cubosomes
2.9. Quantification of phenytoin and 4-HPPH in plasma and brain
stabilised with Tween 80Ò displayed three Bragg peaks with rela-
tissue by LC-MS p p p
tive positions at ratios of 2: 4: 6 indicating the presence V2
phase particles but with Im3m space group. The lattice parameter
Quantification of phenytoin and its major metabolite 4-HPPH in
of 120.8 ± 0. 6 Å for these particles is also consistent with our pre-
plasma and brain tissue was performed in accordance with a vali-
vious findings [37].
dated LC-MS method modified after Tanaka et al. [40]. Briefly, ali-
Incorporation of phenytoin in either selachyl alcohol or phy-
quots of 100 mL plasma or brain (homogenised in 2 mL/g of water)
tantriol LCNs did not cause dramatic changes in the phase struc-
were spiked with d10-phenytoin (internal standard). Phenytoin and
ture with Bragg peaks appearing at similar relative positions to
4-HPPH were extracted by adding 200 mL of chilled acetonitrile and
their blank counterparts. However, a decrease in the lattice param-
800 mL of tert-butyl methyl ether (TBME) (Merck, Germany), cen-
eter (Table 1) of selachyl alcohol LCNs prepared in the presence of
trifuged (17,200 g for 20 min at 4 °C) and then the supernatant
both stabilisers was observed with the addition of phenytoin. In
transferred to a new tube which was then evaporated to dryness
contrast, incorporation of phenytoin in phytantriol LCNs showed
in a centrifugal evaporator and reconstituted in methanol.
a mixed effect in the presence of the two stabilisers used. PluronicÒ
Samples were analysed using an Agilent 1290 HPLC system con-
F127 stabilised dispersions showed an increase in the lattice
nected to an AbSciex QTRAP 5500 mass spectrometer with Turbo
parameter whilst a decrease in the lattice parameter was observed
Spray ion source in positive mode. The ion pairs monitored were
for dispersions stabilised with Tween 80Ò (Table 1), when com-
253/182 (phenytoin), 263/192 (d10-phenytoin) and 269/198 (4-
pared to their blank counterparts.
HPPH). Mobile phase A consisted of 0.1% formic acid in water.
Selachyl alcohol dispersions containing phenytoin were
Mobile phase B was 0.1% formic acid in 2:1 acetonitrile:methanol.
selected for ultrastructural analysis by cryo-TEM. Representative
Analysis was performed at a flow rate of 0.25 mL/min by injecting
electron micrographs are shown in Fig. 3. In Panel A onion type
5 mL of sample into a Kinetex EVO 5 mm 100 Å C18 (150  2.1 mm)
vesicles with curved striations typical of hexosomes were observed
column maintained at 40 °C. The gradient was started at 80% A, 20%
for dispersions prepared in the presence of PluronicÒ F127 [15]. In
B, where it was held for 30 s before shifting to 5% A, 95% B over
contrast, dispersions stabilised with Tween 80Ò (Panel B) showed
7 min to elute the analytes. The auto-sampler was maintained at
the characteristic internal cubic structure of cubosomes when
a temperature of 20 °C during analysis. Data was collected in Ana-
observed under TEM.
lystÒ software and analyte/internal standard ratio was used to con-
struct calibration curves and analyse the data in Microsoft Excel.
3.2. In vitro cell viability studies using human brain endothelial cells
2.10. Statistical analysis
Selachyl alcohol LCNs not only showed significantly higher
Encapsulation efficiencies and DLS data were compared statisti- encapsulation of phenytoin compared to phytantriol LCNs, but
cally between selachyl alcohol and phytantriol nanoparticles using the nanostructure of the particles (i.e. cubosome and hexosome)
two-way ANOVA combined with Sidak’s multiple comparison test. was retained upon the addition of the model drug. As a result, sela-
Tissue concentration and cell culture data was compared statisti- chyl alcohol cubosomes and hexosomes were selected for further
cally between formulations at each concentration or time point biological testing in vitro and in vivo. The effect of cubosomes
149
Y. Mohammad, R.N. Prentice, B.J. Boyd et al. Journal of Colloid and Interface Science 605 (2022) 146–154

Table 1
Physicochemical properties (Z-average, PDI, encapsulation efficiency (EE%) of phenytoin, lattice parameters and phase structure obtained from SAXS measurements) of selachyl
alcohol and phytantriol dispersions with (+) and without () phenytoin. Dispersions contained 15% w/w stabiliser (relative to lipid). Data presented are the mean ± SD of three
independent experiments. #P < 0.0001.

Formulation Composition Z-average (nm) PDI EE (%) Lattice parameter (Å) Phase structure
Lipid Stabiliser Phenytoin
Selachyl alcohol PluronicÒ F127  174 ± 3 0.14 ± 0.01 58.0 ± 0.1 H2
þ 162 ± 7 0.13 ± 0.02 97.5 ± 2.4# 56.6 ± 0.1
Tween 80Ò  155 ± 6 0.12 ± 0.02 131.6 ± 0.6 V2 (Im3m)
þ 144 ± 4 0.12 ± 0.03 96.7 ± 2.4# 128.3 ± 0.8
Phytantriol PluronicÒ F127  152 ± 5 0.15 ± 0.02 70.5 ± 0.1 V2 (Pn3m)
þ 150 ± 10 0.20 ± 0.03 68.3 ± 3.1 74.1 ± 0.6
Ò
Tween 80  131 ± 5 0.18 ± 0.01 120.8 ± 0.6 V2 (Im3m)
þ 143 ± 14 0.15 ± 0.05 72.6 ± 2.7 116.7 ± 0.5

Phytantriol 10–30 lg/mL was then investigated (Fig. 4). The results show that
2 incorporation of phenytoin at the tested concentrations into cubo-
3 somes or hexosomes did not alter its cytotoxicity towards hCMEC/
4
6
D3 cells.
3
2
4 + Phenytoin
6 3.3. In vivo brain and plasma profiles of phenytoin administered in
Pluronic® F127 selachyl alcohol cubosomes and hexosomes
2

4
6
Brain and plasma concentrations of phenytoin after administra-
tion in cubosomes and hexosomes are shown in Fig. 5. Plasma con-
Arbitrary intensity units

2 centrations (Panel A) of phenytoin was significantly higher when


4
6 administered in cubosome formulations as compared to hexo-
+ Phenytoin somes at 15 min (184.1 ng/mL vs 102.9 ng/mL, p = 0.0239), but
not at 30 and 60 min, where plasma concentrations decreased with
1
Tween 80®
both formulations. Brain concentrations (Panel B) appeared mark-
edly higher after administration of the cubosome formulations at
Selachyl alcohol 15 min (159.0 ng/g vs 58.5 ng/g) and 30 min (151.9 ng/g vs
74.1 ng/g), but the difference was only statistically significant at
3
1
4 + Phenytoin 30 min (p = 0.0308), and not 15 min (p = 0.0705). At 60 min, the
difference in plasma concentration of phenytoin for the cubosome
treated group as compared to the hexosome group (64.6 ng/g vs
3
Pluronic® F127 44.7 ng/g) was no longer significant.
2
4
Brain to plasma ratio for each individual animal and the average
4
6
was calculated and is shown in Panel C. Both formulations had a
2 ratio below 1.0 at 15 min, but at 30 and 60 min, the average ratio
4
6
+ Phenytoin
for cubosomes was 1.23 and 1.20, respectively, which exceeded 1.0
and indicated accumulation of phenytoin in the brain. The ratio for
hexosomes also continued to rise after 15 min but remained below
Tween 80®
1.0 (0.82 at 30 min and 0.91 at 60 min) and was significantly lower
than the cubosomes (p = 0.0115 and 0.0097 for 30 and 60 min,
0.05 0.10 0.15 0.20 0.25 0.30 0.35 0.40 respectively).
q(Å-1)
To gain further insight into the delivery of phenytoin encapsu-
Fig. 2. SAXS diffractograms of selachyl alcohol and phytantriol dispersions lated in cubosomes and hexosomes, the concentration of its major
stabilised with PluronicÒ F127 or Tween 80Ò (15% w/w to lipid) without phenytoin metabolite 4-HPPH was simultaneously measured in the plasma
(black curve) and dispersions containing 1% w/w phenytoin (blue curves). Relative and brain tissue. Fig. 5D shows that 4-HPPH was present in plasma
peak positions for each formulation are shown. Note that the scattering profiles are
at 15 min (44.1 ng/mL (cubosomes), 27.6 ng/mL (hexosomes)),
displaced on the vertical axis for clarity of presentation.
appeared to rise slightly at 30 min (60.7 ng/mL (cubosomes),
47.6 ng/mL (hexosomes)) and fall again at 60 min (41.8 ng/mL (cu-
bosomes), 44.7 ng/mL (hexosomes)), but did not vary significantly
and hexosomes on cell viability as a function of the concentration
between the formulations at any time point (P > 0.05). No 4-HPPH
of selachyl alcohol and the internal structure of the LCNs, was
was detected in the brain tissue within the quantifiable range of
investigated using a PI staining flow cytometry assay (Fig. 4). Cell
the assay.
viability of greater than 80% of untreated control was considered
non-cytotoxic as previously described [42,43]. Selachyl alcohol
cubosomes and hexosomes were found to be non-cytotoxic to 4. Discussion
hCMEC/D3 up to a concentration of 37.5 lg/mL of selachyl alcohol.
No differences (P > 0.05) in cytotoxicity between cubosomes and The aim of this study was to explore the encapsulation and
hexosomes was observed. brain delivery of phenytoin in LCNs composed of the endogenous
As the hCMEC/D3 cells could tolerate up to 37.5 lg/mL of sela- alkylglycerol lipid, selachyl alcohol. Qualitatively, CPLM studies
chyl alcohol, the cytotoxicity of selachyl alcohol cubosomes and indicated greater solubility of phenytoin in selachyl alcohol than
hexosomes containing phenytoin within a concentration range of in phytantriol. This is consistent with previous studies using the
150
Y. Mohammad, R.N. Prentice, B.J. Boyd et al. Journal of Colloid and Interface Science 605 (2022) 146–154

Fig. 3. Representative cryo-TEM micrographs of selachyl alcohol dispersions containing phenytoin (1% w/w) and stabilised with 15% w/w PluronicÒ F127 (A) or Tween 80Ò
(B). Panel A shows onion-type vesicles with curved striations (hexosomes) whereas in Panel B nanoparticles with distinct cubic symmetry (cubosomes) are evident. Scale
bar = 200 nm.

110 docetaxel, a less dramatic effect on phase structure resulting only


in a decrease in the lattice parameter was observed in the current
100 study with phenytoin (which is smaller in size). With phytantriol,
encapsulation of phenytoin in the presence of the two stabilisers
Cell viability (%)

90 had different effects on the lattice parameter suggesting that sta-


bilisers also play a role in accommodating guest molecules in the
80
lipid bilayer. Tween 80Ò is believed to be more internalised into
70 the lipid bilayer compared to PluronicÒ F127 which is believed to
be mostly surface associated [15,37,46,47]. Incorporation of pheny-
60 toin into phytantriol dispersions stabilised with PluronicÒ F127,
where it is assumed that the stabilizer is surface-associated
50 resulted in greater swelling of the lipid bilayer, thereby leading
10
0 to an increase in the lattice parameter by ~4 Å. In contrast, in the
presence of the more internalised Tween 80Ò, encapsulation of
0 10 20 30 40 50 phenytoin in the phytantriol lipid bilayer resulted in an increase
Selachyl alcohol ( g/mL) in the negative curvature, with a corresponding ~4 Å reduction in
the lattice parameter.
Fig. 4. Viability (%) of hCMEC/D3 cells after exposure to selachyl alcohol cubosomes Cellular toxicity of selachyl alcohol cubosomes and hexosomes
(squares) and hexosomes (triangles) containing: no phenytoin (black curves) or 1%
towards brain endothelial cells has not been previously reported.
w/w phenytoin (black curves). Data presented are the mean ± SD of three
independent experiments. LCNs with different internal structures have been reported to show
differences in their interaction with lipid membranes of cells by
membrane fusion and lipid exchange, resulting in membrane dis-
poorly water-soluble drug, cinnarizine [9]. Additionally, encapsula- ruption and cell death [43,48,49]. Despite this, comparable cell via-
tion efficiency of phenytoin was significantly greater in selachyl bility following exposure to selachyl alcohol hexosomes and
alcohol LCNs compared to phytantriol LCNs, collectively suggesting cubosomes in this study suggested a similar mode of interaction
higher solubility in selachyl alcohol than phytantriol. The with cells. This was an interesting finding considering that a recent
hydrophobic tail group chain architecture in selachyl alcohol and review of LCNs by Tan et al. [50] concluded that nanostructures
in phytantriol are likely to be responsible for the differences in sol- with higher negative curvature (i.e. the H2 phase with respect to
ubilisation of phenytoin. The more rigid phytanyl chain in phy- our study) demonstrated better tolerability across different cell
tantriol may have permitted less accommodation of phenytoin in lines. It was postulated that this might be due to a difference in vis-
its structure (cubic), compared to the oleate chain (with a kink) coelastic behaviour, with hexosomes being less rigid than cubo-
in selachyl alcohol, resulting in lower solubilisation of phenytoin somes leading to a lower degree of lipid mixing with cell
(and thus lower encapsulation). membranes. However, the authors did acknowledge that incorpo-
The decrease in the lattice parameter observed in selachyl alco- ration of additional dopant lipids, changes in the stabiliser or mod-
hol LCNs containing phenytoin compared to their non-drug con- ification of the surface charge in the reviewed studies meant that
taining counterparts, suggests that encapsulation of hydrophobic the effect of internal structure on cellular interactions is by no
phenytoin in the lipid bilayer leads to an increase in mean negative means clear-cut. For instance, Tran et al. [51] studied GMO-based
curvature. Encapsulation of other hydrophobic drugs such as pacli- LCNs and reported that less curved V2 phase nanoparticles were
taxel and docetaxel in GMO and phytantriol based LCNs, respec- more toxic than H2 phase nanoparticles towards fibroblasts. This
tively, have also resulted in an increase in the negative curvature was tested by adding increasing amounts of the endogenous lipid
leading to a complete phase transformation from a V2 to an H2 capric acid to induce the structural transformation from V2 to H2
phase [44,45]. Compared to the effects seen with paclitaxel and phase, however the capric acid control solution showed the highest

151
Y. Mohammad, R.N. Prentice, B.J. Boyd et al. Journal of Colloid and Interface Science 605 (2022) 146–154

400 400
A B

PhenytoinPlasma (ng/mL)

PhenytoinBrain (ng/g)
300 * 300 *

200 200

100 100

0 0
15 30 60 15 30 60
Time (min) Time (min)

2.0 400
C D

4-HPPHPlasma (ng/mL)
* *
Brain:Plasma ratio

1.5 300

1.0 200

0.5 100

0.0 0
15 30 60 15 30 60
Time (min) Time (min)

Fig. 5. Plasma (A) and brain (B) concentrations of phenytoin, brain to plasma ratio of phenytoin (C) and plasma concentrations of 4-HPPH (D) in rats 15, 30 and 60 min after
intravenous administration of selachyl alcohol cubosomes (circles) or hexosomes (triangles) encapsulating phenytoin (1% w/w relative to lipid). *P < 0.05, n = 4 to 5 rats per
group.

cellular viability, so the effect of internal structure could not easily biliser differs. The question remaining, however, is whether the
be dissociated from the potentially protective effect of the increas- internal structure or stabiliser, or both, are responsible for the
ing amount of capric acid in the system. In contrast, in this study apparently increased circulation time in plasma and drug delivery
we were able to alter the phase from V2 to H2 by only modifying to the brain. Previous literature on the proposed apolipoprotein-
the stabiliser. While previous studies have suggested that stabiliser mediated brain uptake mechanism associated with Tween 80Ò
modification can alter the cell toxicity profile depending on the [5,52], which formed the basis of the hypothesis, lends weight to
balance of surface coverage and interaction of the stabiliser with the stabiliser being the responsible factor. The internal nanostruc-
the bilayer [50], we did not find a difference. Whatever the case, tures of cubosomes and hexosomes may also impart different drug
our findings offered a point of confidence for progressing towards release profiles which could have played a role, but no direct com-
in vivo comparisons of brain uptake of phenytoin from these differ- parison of these with a hydrophobic drug was found in the litera-
ently structured nanoparticles without the confounding effect of ture which leans heavily towards investigating the release of
differences in cytotoxicity of the formulation components. hydrophilic compounds only [53–55]. Furthermore, there are
The final element of this study was to investigate in vivo brain potential interactions of the LCNs with blood plasma components
delivery of phenytoin using the two differently structured selachyl leading to changes in internal structure, as has been previously
alcohol LCNs, cubosomes and hexosomes. Average concentrations reported [56,57], which must also be considered when correlating
of phenytoin in the brain and plasma were consistently higher internal structure with drug delivery and investigations into this
when administered in cubosomes compared with hexosomes. are currently ongoing in our lab.
The differences were statistically significant at 15 min in plasma The major metabolite of phenytoin, 4-HPPH, was also quantified
and 30 min in the brain, which can be interpreted as a greater in plasma and brain in order to give some extra insight into the
retention of cubosomes in the plasma relative to hexosomes, fol- delivery of phenytoin in LCNs. There are a few interesting points
lowed by a higher relative accumulation in the brain. This finding that can be taken from the results. Firstly, for phenytoin to be
is further supported by the average brain to plasma ratio of pheny- effective, it must be released from the LCNs which the presence
toin which exceeded 1.0 (indicating accumulation in the brain rel- of the metabolite in the plasma indicates happened for both for-
ative to plasma) at 30 min after cubosome administration, but not mulations. Given the absence of 4-HPPH in the brain, which is con-
hexosomes, a difference that was maintained up to 60 min. These sistent with previous studies [58,59], this metabolism must have
results therefore support the hypothesis that selachyl alcohol occurred due to free drug in the plasma. Whether the release of
cubosomes enhance drug delivery to the brain compared with hex- the participating free phenytoin occurred in the plasma before
osomes prepared using the same lipid. the particles reached the brain, or occurred in the brain, followed
As touched on above, comparing different types of LCNs, most by an equilibration-related movement out into plasma where the
commonly cubosomes and hexosomes as in this study, is a chal- metabolism occurred is not clear just by tracking the concentra-
lenging task given that the lipid composition usually needs to be tions of 4-HPPH. When phenytoin concentration is considered in
modified in order to alter bilayer curvature enough to induce a conjunction with this, however, it can be seen that the proportion
phase change [50]. The data presented here is unique in that it of 4-HPPH relative to phenytoin in the plasma and brain is signif-
compares cubosomes and hexosomes composed entirely of one icantly lower in the cubosome group at 15 and 30 min, respec-
lipid, selachyl alcohol, and encapsulating the same amount of drug tively, given that no significant difference was found between the
(phenytoin) with 97–98% efficiency, where only the surface sta- 4-HPPH concentrations after each formulation, but was with

152
Y. Mohammad, R.N. Prentice, B.J. Boyd et al. Journal of Colloid and Interface Science 605 (2022) 146–154

phenytoin concentrations at these time points. This suggests a Acknowledgements


preferential distribution of phenytoin to the brain while encapsu-
lated in the cubosomes and a subsequently lower proportion of The present work was supported by funding from Health
drug being metabolised to 4-HPPH, in turn supporting the conclu- Research Council of New Zealand (SBR). RP and YM were supported
sions drawn earlier from the phenytoin concentration data. Collec- by a Doctoral Scholarship from the University of Otago. Authors
tively, our data suggest that selachyl alcohol cubosomes can would like to acknowledge Mr. Richard Easingwood from the Otago
deliver drug cargo to the brain. However, precisely how the drug Centre for Electron Microscopy for help with acquisition and anal-
cargo is transported across the BBB will need to be elucidated in ysis of the electron microscopy data. The SAXS studies were per-
future studies. Whilst no toxic effects were observed via beha- formed on the SAXS/WAXS beamline at the Australian
vioural observations in animals after intravenous injection of sela- Synchrotron (BJB).
chyl alcohol LCNs, a detailed toxicity study (including haemolytic
studies) is warranted in the future as LCNs formulated with other
liquid crystal forming lipids such GMO/PluronicÒ F127 have shown Appendix A. Supplementary material
evidence of cytotoxicity [54]. Finally, the therapeutic efficacy of the
new cubosomes formulations which repurpose old drugs such as Supplementary data to this article can be found online at
phenytoin in the case of this study, should also be compared https://doi.org/10.1016/j.jcis.2021.07.070.
against marketed formulations.

5. Conclusions References

[1] J.V. Georgieva, D. Hoekstra, I.S. Zuhorn, Smuggling drugs into the brain: an
Even though cubosomes and hexosomes are two uniquely
overview of ligands targeting transcytosis for drug delivery across the blood-
structured lipid nanoparticles with immense potential to deliver brain barrier, Pharmaceutics 6 (4) (2014) 557–583.
therapeutics, they have not been compared for their ability to deli- [2] S. Wohlfart, S. Gelperina, J. Kreuter, Transport of drugs across the blood-brain
ver drugs in vivo [60]. Studies on the cytotoxicity and cellular barrier by nanoparticles, J. Control. Release 161 (2) (2012) 264–273.
[3] A.E. Gulyaev, S.E. Gelperina, I.N. Skidan, A.S. Antropov, G.Y. Kivman, J. Kreuter,
uptake of cubosomes and hexosomes in different cell lines have Significant transport of doxorubicin into the brain with polysorbate 80-coated
been reported, however, often differing conclusions are reported nanoparticles, Pharm. Res. 16 (10) (1999) 1564–1569.
[43,48–51]. A direct comparison of cubosomes and hexosomes is [4] J. Kreuter, P. Ramge, V. Petrov, S. Hamm, S.E. Gelperina, B. Engelhardt, R.
Alyautdin, H. von Briesen, D.J. Begley, Direct evidence that polysorbate-80-
a challenging task as the lipid composition often needs to be coated poly(butylcyanoacrylate) nanoparticles deliver drugs to the CNS via
altered to induce a phase change which likely contributed to the specific mechanisms requiring prior binding of drug to the nanoparticles,
differences observed in the in vitro studies. Herein is the first Pharm. Res. 20 (3) (2003) 409–416.
[5] J. Kreuter, D. Shamenkov, V. Petrov, P. Ramge, K. Cychutek, C. Koch-Brandt, R.
reported study of the comparison of cubosomes and hexosomes Alyautdin, Apolipoprotein-mediated transport of nanoparticle-bound drugs
composed entirely of a single lipid, selachyl alcohol, for the deliv- across the blood-brain barrier, J. Drug Target. 10 (4) (2002) 317–325.
ery of a therapeutic molecule in vivo. Of interest for our research [6] B. Boyd, S. Khoo, D. Whittaker, G. Davey, C. Porter, A lipid-based liquid
crystalline matrix that provides sustained release and enhanced oral
group is using LCNs to deliver therapeutics across the BBB as there bioavailability for a model poorly water soluble drug in rats, Int. J. Pharm.
remains a pressing need to find better ways to deliver molecules at 340 (1-2) (2007) 52–60.
therapeutic concentrations to the brain to treat the many types of [7] C.J. Drummond, C. Fong, Surfactant self-assembly objects as novel drug
delivery vehicles, Curr. Opin. Colloid Interface Sci. 4 (6) (1999) 449–456.
destructive neurological disorders currently affecting human
[8] C. Guo, J. Wang, F. Cao, R.J. Lee, G. Zhai, Lyotropic liquid crystal systems in drug
health. Consequently, cubosomes and hexosomes were compared delivery, Drug Discovery Today 15 (23-24) (2010) 1032–1040.
for their ability to deliver the model drug phenytoin to the brain. [9] T.-H. Nguyen, T. Hanley, C.J.H. Porter, B.J. Boyd, Nanostructured reverse
We demonstrate for the first time that cubosomes are superior to hexagonal liquid crystals sustain plasma concentrations for a poorly water-
soluble drug after oral administration, Drug Deliv. Translat. Res. 1 (6) (2011)
hexosomes for delivery of a therapeutic molecule in the context 429–438.
of drug delivery to the brain. Our findings demonstrate the tremen- [10] J. Gustafsson, H. Ljusberg-Wahren, M. Almgren, K. Larsson, Cubic lipidwater
dous drug delivery potential of cubosomes. However, there phase dispersed into submicron particles, Langmuir 12 (20) (1996) 4611–
4613.
remains much to discover about the link, not only between the [11] X. Mulet, B.J. Boyd, C.J. Drummond, Advances in drug delivery and medical
internal nanostructure but also the surface architecture, and the imaging using colloidal lyotropic liquid crystalline dispersions, J. Colloid
performance of these complex colloids in vivo. Interface Sci. 393 (2013) 1–20.
[12] B.J. Boyd, Y.D. Dong, T. Rades, Nonlamellar liquid crystalline nanostructured
particles: advances in materials and structure determination, J. Liposome Res.
CRediT authorship contribution statement 19 (1) (2009) 12–28.
[13] A. Yaghmur, O. Glatter, Characterization and potential applications of
nanostructured aqueous dispersions, Adv. Colloid Interface Sci. 147-148
Younus Mohammad: Conceptualization, Methodology, Data (2009) 333–342.
curation, Formal analysis, Validation, Visualization, Writing – orig- [14] J. Barauskas, M. Johnsson, T. Nylander, F. Tiberg, Hexagonal liquid-crystalline
nanoparticles in aqueous mixtures of glyceryl monooleyl ether and pluronic
inal draft, Writing - review & editing. Richard N. Prentice: Concep- F127, Chem. Lett. 35 (8) (2006) 830–831.
tualization, Methodology, Data curation, Formal analysis, [15] M. Younus, A. Hawley, B.J. Boyd, S.B. Rizwan, Bulk and dispersed aqueous
Validation, Visualization, Writing – original draft, Writing - review behaviour of an endogenous lipid, selachyl alcohol: Effect of Tween 80 and
Pluronic F127 on nanostructure, Colloids Surf., B 169 (2018) 135–142.
& editing. Ben J. Boyd: Methodology, Writing - review & editing, [16] H. Eibl, V. Jendrossek, B. Erdlenbruch, M. Lakomek, Transient and controllable
Funding acquisition, Supervision, Writing - review & editing. Shak- opening of the blood-brain barrier to cytostatic and antibiotic agents by
ila B. Rizwan: Conceptualization, Data curation, Formal analysis, alkylglycerols in rats, Exp. Brain Res. 135 (3) (2000) 417–422.
[17] B. Erdlenbruch, V. Jendrossek, W. Kugler, H. Eibl, M. Lakomek, Increased
Validation, Visualization, Funding acquisition, Supervision, Project delivery of erucylphosphocholine to C6 gliomas by chemical opening of the
administration, Methodology, Writing - review & editing. blood-brain barrier using intracarotid pentylglycerol in rats, Cancer
Chemother. Pharmacol. 50 (4) (2002) 299–304.
[18] B. Erdlenbruch, W. Kugler, C. Schinkhof, H. Neurath, H. Eibl, M. Lakomek,
Declaration of Competing Interest Blood-brain barrier opening with alkylglycerols: Biodistribution of 1-O-
pentylglycerol after intravenous and intracarotid administration in rats, J.
The authors declare that they have no known competing finan- Drug Target. 13 (3) (2005) 143–150.
[19] J. Barauskas, I. Svedaite, E. Butkus, V. Razumas, K. Larsson, F. Tiberg, Synthesis
cial interests or personal relationships that could have appeared and aqueous phase behavior of 1-glyceryl monooleyl ether, Colloids Surf., B 41
to influence the work reported in this paper. (1) (2005) 49–53.

153
Y. Mohammad, R.N. Prentice, B.J. Boyd et al. Journal of Colloid and Interface Science 605 (2022) 146–154

[20] Y.D. Dong, I. Larson, T. Hanley, B.J. Boyd, Bulk and dispersed aqueous phase tetrachloride-intoxicated rats by high-performance liquid chromatography, J.
behavior of phytantriol: effect of vitamin E acetate and F127 polymer on liquid Chromatogr. B Biomed. Appl. 673 (1) (1995) 147–151.
crystal nanostructure, Langmuir 22 (23) (2006) 9512–9518. [41] S.P. Collins, I. Dolbnya, B.A. Palmer, G.R. Edwards-Gau, A. Morte-Ródenas, B.M.
[21] C. Kellinghaus, S. Berning, M. Besselmann, Intravenous lacosamide as Kariuki, G.K. Lim, K.D.M. Harris, Y. Joly, X-ray birefringence in highly
successful treatment for nonconvulsive status epilepticus after failure of anisotropic materials, J. Phys. Conf. Ser. 425 (13) (2013) 132015, https://doi.
first-line therapy, Epilepsy Behav. 14 (2) (2009) 429–431. org/10.1088/1742-6596/425/13/132015.
[22] G.M. Brophy, R. Bell, J. Claassen, B. Alldredge, T.P. Bleck, T. Glauser, S.M. [42] T.M. Hinton, F. Grusche, D. Acharya, R. Shukla, V. Bansal, L.J. Waddington, P.
LaRoche, J.J. Riviello, L. Shutter, M.R. Sperling, D.M. Treiman, P.M. Vespa, Monaghan, B.W. Muir, Bicontinuous cubic phase nanoparticle lipid chemistry
Guidelines for the evaluation and management of status epilepticus, Neurocrit. affects toxicity in cultured cells, Toxicol. Res. 3 (1) (2014) 11–22.
Care 17 (1) (2012) 3–23. [43] H.-H. Shen, J.G. Crowston, F. Huber, S. Saubern, K.M. McLean, P.G. Hartley, The
[23] E.M. Manno, Status epilepticus: current treatment strategies, The influence of dipalmitoyl phosphatidylserine on phase behaviour of and cellular
Neurohospitalist 1 (1) (2011) 23–31. response to lyotropic liquid crystalline dispersions, Biomaterials 31 (36)
[24] D.M. Treiman, M.C. Walker, Treatment of seizure emergencies: convulsive and (2010) 9473–9481.
non-convulsive status epilepticus, Epilepsy Res. 68 (Suppl 1) (2006) S77–S82. [44] V. Jain, N.K. Swarnakar, P.R. Mishra, A. Verma, A. Kaul, A.K. Mishra, N.K. Jain,
[25] J.W. Chen, C.G. Wasterlain, Status epilepticus: pathophysiology and Paclitaxel loaded PEGylated gleceryl monooleate based nanoparticulate
management in adults, Lancet Neurol. 5 (3) (2006) 246–256. carriers in chemotherapy, Biomaterials 33 (29) (2012) 7206–7220.
[26] Y.H. Samuel H. Yalkowsky, Parijat Jain, Hand Book of Aqueous Solubility Data, [45] S. Jain, N. Bhankur, N.K. Swarnakar, K. Thanki, Phytantriol based ‘‘stealth”
second ed., CRC Press, Taylor and Francis Group, 2010. lyotropic liquid crystalline nanoparticles for improved antitumor efficacy and
[27] O.P.M. Rajniti Prasad, A. Gupta, Gangrene of left hand following accidental reduced toxicity of docetaxel, Pharm. Res. 32 (10) (2015) 3282–3292.
intra-arterial injection of phenytoin sodium, Pediatric Oncall 9 (5) (2012) 41– [46] H. Azhari, M. Strauss, S. Hook, B.J. Boyd, S.B. Rizwan, Stabilising cubosomes
42. with Tween 80 as a step towards targeting lipid nanocarriers to the blood–
[28] W.M. Coplin, D.H. Rhoney, J.A. Rebuck, E.A. Clements, M.S. Cochran, B.J. O’Neil, brain barrier, Eur. J. Pharm. Biopharm. 104 (2016) 148–155.
Randomized evaluation of adverse events and length-of-stay with routine [47] A.J. Tilley, C.J. Drummond, B.J. Boyd, Disposition and association of the steric
emergency department use of phenytoin or fosphenytoin, Neurol. Res. 24 (8) stabilizer PluronicÒ F127 in lyotropic liquid crystalline nanostructured particle
(2002) 842–848. dispersions, J. Colloid Interface Sci. 392 (2013) 288–296.
[29] K.L. Fuller, Y.Y. Wang, M.J. Cook, M.A. Murphy, W.J. D’Souza, Tolerability, [48] S. Murgia, A.M. Falchi, M. Mano, S. Lampis, R. Angius, A.M. Carnerup, J. Schmidt,
safety, and side effects of levetiracetam versus phenytoin in intravenous and G. Diaz, M. Giacca, Y. Talmon, M. Monduzzi, Nanoparticles from lipid-based
total prophylactic regimen among craniotomy patients: a prospective liquid crystals: Emulsifier influence on morphology and cytotoxicity, J. Phys.
randomized study, Epilepsia 54 (1) (2013) 45–57. Chem. B 114 (10) (2010) 3518–3525.
[30] C.A. Twardowschy, L. De Paola, F.M. Germiniani, L.C. Werneck, C. Silvado, [49] J. Barauskas, C. Cervin, M. Jankunec, M. Špandyreva, K. Ribokaitė, F. Tiberg, M.
Pearls & oy-sters: soft-tissue necrosis as a result of intravenous leakage of Johnsson, Interactions of lipid-based liquid crystalline nanoparticles with
phenytoin, Neurology 73 (19) (2009) e94–e95. model and cell membranes, Int. J. Pharm. 391 (1-2) (2010) 284–291.
[31] N. Hasija, A.J. Hazarika, N. Sokhal, S. Kumar, Tissue necrosis of hand caused by [50] A. Tan, L. Hong, J.D. Du, B.J. Boyd, Self-assembled nanostructured lipid systems: is
phenytoin extravasation: an unusual occurrence, Saudi J. Anaesthesia 8 (2) there a link between structure and cytotoxicity?, Adv Sci. 6 (3) (2019) 1801223.
(2014) 309–310. [51] N. Tran, X. Mulet, A.M. Hawley, T.M. Hinton, S.T. Mudie, B.W. Muir, E.C.
[32] A.J. Scumpia, J. Yahsou, J. Cajina, C. Cao, Purple glove syndrome after Giakoumatos, L.J. Waddington, N.M. Kirby, C.J. Drummond, Nanostructure and
intravenous phenytoin administration presenting in the emergency cytotoxicity of self-assembled monoolein-capric acid lyotropic liquid
department, The Journal of Emergency Medicine 44 (2) (2013) e281–e283. crystalline nanoparticles, RSC Adv. 5 (34) (2015) 26785–26795.
[33] R. Chokshi, J. Openshaw, N.N. Mehta, E. Mohler, Purple glove syndrome [52] E. Garcia-Garcia, K. Andrieux, S. Gil, P. Couvreur, Colloidal carriers and blood–
following intravenous phenytoin administration, Vascular Med. 12 (1) (2007) brain barrier (BBB) translocation: a way to deliver drugs to the brain?, Int J.
29–31. Pharm. 298 (2) (2005) 274–292.
[34] S.B. Rizwan, D. Assmus, A. Boehnke, T. Hanley, B.J. Boyd, T. Rades, S. Hook, [53] I. Martiel, N. Baumann, J.J. Vallooran, J. Bergfreund, L. Sagalowicz, R. Mezzenga,
Preparation of phytantriol cubosomes by solvent precursor dilution for the Oil and drug control the release rate from lyotropic liquid crystals, J. Control.
delivery of protein vaccines, Eur. J. Pharm. Biopharm. 79 (1) (2011) 15–22. Release 204 (2015) 78–84.
[35] N.M. Kirby, S.T. Mudie, A.M. Hawley, D.J. Cookson, H.D.T. Mertens, N. [54] A. Otte, B.-K. Soh, G. Yoon, K. Park, Liquid crystalline drug delivery vehicles for
Cowieson, V. Samardzic-Boban, A low-background-intensity focusing small- oral and IV/subcutaneous administration of poorly soluble (and soluble) drugs,
angle X-ray scattering undulator beamline, J. Appl. Crystallogr. 46 (6) (2013) Int. J. Pharm 539 (1-2) (2018) 175–183.
1670–1680. [55] A. Zabara, R. Mezzenga, Controlling molecular transport and sustained drug
[36] S.T. Hyde, Identification of lyotropic liquid crystalline mesophases, Handbook release in lipid-based liquid crystalline mesophases, J. Control. Release 188
of Applied Surface and Colloid Chemistry, John Wiley & Sons, Ltd, Great Britain, (2014) 31–43.
2001, pp. 299–332. [56] I.D. Mat Azmi, L. Wu, P.P. Wibroe, C. Nilsson, J. Østergaard, S. Stürup, B.
[37] M. Younus, R.N. Prentice, A.N. Clarkson, B.J. Boyd, S.B. Rizwan, Incorporation of Gammelgaard, A. Urtti, S.M. Moghimi, A. Yaghmur, Modulatory effect of
an endogenous neuromodulatory lipid, oleoylethanolamide, into cubosomes: human plasma on the internal nanostructure and size characteristics of liquid-
nanostructural characterization, Langmuir 32 (35) (2016) 8942–8950. crystalline nanocarriers, Langmuir 31 (18) (2015) 5042–5049.
[38] V. Barillaro, P.P. Pescarmona, M. Van Speybroeck, T.D. Thi, J. Van Humbeeck, J. [57] J.C. Bode, J. Kuntsche, S.S. Funari, H. Bunjes, Interaction of dispersed cubic
Vermant, P. Augustijns, J.A. Martens, G. Van Den Mooter, High-throughput phases with blood components, Int. J. Pharm. 448 (1) (2013) 87–95.
study of phenytoin solid dispersions: formulation using an automated solvent [58] C.L. DeVane, J.W. Simpkins, S.A. Stout, Distribution of phenobarbital and
casting method, dissolution testing, and scaling-up, J. Comb. Chem. 10 (5) phenytoin in pregnant rats and their fetuses, Epilepsia 32 (2) (1991) 250–256.
(2008) 637–643. [59] Y.G. Kim, M.K. Cho, J.W. Kwon, S.G. Kim, S.J. Chung, C.-K. Shim, M.G. Lee, Effects
[39] R. Baumgartner, A. Eitzlmayr, N. Matsko, C. Tetyczka, J. Khinast, E. Roblegg, of cysteine on the pharmacokinetics of intravenous phenytoin in rats with
Nano-extrusion: a promising tool for continuous manufacturing of solid nano- protein-calorie malnutrition, Int. J. Pharm. 229 (1-2) (2001) 45–55.
formulations, Int. J. Pharm. 477 (1–2) (2014) 1–11. [60] A. Yaghmur, H. Mu, Recent advances in drug delivery applications of
[40] E. Tanaka, N. Sakamoto, M. Inubushi, S. Misawa, Simultaneous determination cubosomes, hexosomes, and solid lipid nanoparticles, Acta Pharm. Sin. B 11
of plasma phenytoin and its primary hydroxylated metabolites in carbon (4) (2021) 871–885.

154

You might also like