You are on page 1of 10

Colloids and Surfaces B: Biointerfaces 162 (2018) 236–245

Contents lists available at ScienceDirect

Colloids and Surfaces B: Biointerfaces


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

Nanostructured lipid carriers for improved oral delivery and


prolonged antihyperlipidemic effect of simvastatin
Heba A. Fathi a , Ayat Allam b , Mahmoud Elsabahy a,b,c,d,∗ , Gihan Fetih a,b ,
Mahmoud El-Badry a,b
a
Assiut International Center of Nanomedicine, Al-Rajhy Liver Hospital, Assiut University, Assiut 71515, Egypt
b
Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
c
Laboratory for Synthetic-Biologic Interactions, Department of Chemistry, Texas A&M University, College Station, TX, USA
d
Misr University for Science and Technology, 6th of October City, Egypt

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

Article history: The purpose of the current study is to develop nanostructured lipid carriers (NLCs) for the delivery of
Received 18 September 2017 the antihyperlipidemic drug simvastatin (SIM) to increase its extremely low oral bioavailability (<5%)
Received in revised form and prolong its antihyperlipidemic effect. NLCs were prepared via emulsification-solvent evaporation
17 November 2017
technique followed by ultrasonication, and the effect of composition of the nanocarriers on the particle
Accepted 27 November 2017
size, size distribution, surface charge, entrapment efficiency, drug release kinetics and physical stability
Available online 28 November 2017
was extensively studied. NLCs exhibited nanosized (<200 nm) spherical morphologies with narrow size
distribution and high drug entrapment efficiency (>75%), sustained drug release pattern, and negative
Keywords:
Nanostructured lipid carriers
surface charge (zeta potential of −35–40 mV) that imparts sufficient electrostatic physical stability. When
Simvastatin tested in vivo, SIM-NLCs of the optimal composition demonstrated improved and prolonged reduction
Hyperlipidemia in the total cholesterol and non-high density lipoprotein cholesterol levels, as compared to the drug
Drug delivery suspension. After oral administration of a single dose of SIM-NLC, 4-fold increase in bioavailability was
Nanoparticles observed, as compared to the SIM suspension. Hence, NLCs might provide efficient nanodevices for the
management of hyperlipidemia and promising drug delivery systems to enhance SIM oral bioavailability.
© 2017 Elsevier B.V. All rights reserved.

1. Introduction and high permeability) with short plasma half-life and variable
absorption [5]. SIM is exposed to extensive first pass metabolism
Elevated blood cholesterol level (i.e. hypercholesterolemia) by cytochrome P3A (CYP3A) in the intestinal guts and the liver
leads to development and progression of atherosclerosis, and con- [6]. The oral bioavailability of SIM in its intact form is ca. 5% due
sequently cardiovascular diseases, and it is estimated to result in 2.6 to the slow dissolution rate in the intestinal fluid and significant
million deaths (4.5% of total mortality) globally [1]. Successful treat- first-pass metabolism [7]. Various approaches have been utilized
ment of hypercholesterolemia substantially reduces morbidity and to enhance the bioavailability of SIM, such as self microemulsi-
mortality from cardiovascular diseases [2,3]. Statins have the ability fication [8], nanoemulsification [9], nanoparticles [10], solid lipid
to retard the accelerated atherosclerosis in hyperlipidemic indi- nanoparticles [11], nanocrystals [12] and nanosuspensions [13].
viduals. Simvastatin (SIM) is one of the statins family, which is Nanocarriers including biodegradable polymeric nanoparticles
considered as the first-line treatment of hypercholesterolemia, [14], polymeric micelles [15], nanocrystals [16], nanosuspensions
dyslipidemia and coronary heart diseases [4]. SIM is a potent [17] and lipid colloidal nanocarriers have received significant con-
inhibitor of 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG-CoA) sideration as delivery systems for several drugs. Lipid nanocarriers
reductase which converts HMG-CoA into mevalonate, a precur- composed of natural or synthetic lipids have the advantage of
sor in cholesterol synthesis. The biopharmaceutical classification controlling drug release with high biocompatibility. Solid lipid
system classifies SIM as class 2 drug (i.e., low aqueous solubility nanoparticles (SLNs) have emerged in the early nineties as an alter-
native carrier system to the existing traditional lipid carriers due
to the feasibility of large scale production, low toxicity and avail-
ability of their excipients [18]. However, the solid lipid matrix of
∗ Corresponding author at: Assiut International Center of Nanomedicine, Al-Rajhy
SLNs results in the formation of a relatively perfect crystal lat-
Liver Hospital, Assiut University, Assiut 71515, Egypt.
E-mail address: mahmoud.elsabahy@chem.tamu.edu (M. Elsabahy). tice leaving a limited space for drug incorporation, which limits

https://doi.org/10.1016/j.colsurfb.2017.11.064
0927-7765/© 2017 Elsevier B.V. All rights reserved.
H.A. Fathi et al. / Colloids and Surfaces B: Biointerfaces 162 (2018) 236–245 237

the loading capacity and leads to drug leakage during storage effects of oleic acid-to-stearic acid ratio and surfactant (Pluronic
[19]. Nanostructured lipid carriers (NLCs), the second-generation F-68) concentration on the physicochemical characteristics of the
of lipid nanoparticles, are composed of a mixture of spatially dif- formulations were evaluated.
ferent lipid molecules, i.e., a solid lipid is blended with a liquid
lipid. The addition of liquid lipid distorts the formation of per- 2.3. Characterizations of the prepared SIM-NLCs
fect lipid crystals, and, thus increasing drug loading capacity, and
decreasing particle size and risk of gelation and drug leakage dur- 2.3.1. Determination of the entrapment efficiency
ing storage [20,21]. Additionally, NLCs promote oral absorption NLCs containing SIM were separated from the free drug via dial-
of encapsulated drugs via selective uptake through the lymphatic ysis. The drug was then released from the NLCs samples by addition
route or the payer’s patches [22]. NLCs have been previously uti- of sufficient volume of methanol. The drug content was assessed
lized for enhancing the oral bioavailability of various drugs [23,24]. spectrophotometrically at 238 nm using UV–vis spectrophotome-
Moreover, SIM-NLCs have been reported earlier to improve SIM ter (Shimadzu Seisakusho, Ltd., Kyoto, Japan). The entrapment
efficiency (EE, %) was then calculated according to Eq. (1):
amount of drug entrapped into the nanoparticles
Entrapment efficiency (%) = × 100 (1)
amount of drug initially added

2.3.2. Particle size and zeta potential


oral bioavailability, although pharmacodynamic profile was not Mean particle size (nm) and polydispersity index (PDI) of the
studied [7]. Pharmacokinetic parameters are not sufficient to pre- prepared formulations were measured by dynamic light scattering
dict the pharmacodynamic activity of statins [25]. In the current using a Zetasizer Nano ZS (Malvern Instruments, Worcestershire,
study, SIM-NLCs are prepared via emulsification-solvent evapo- UK) equipped with a backscattered light detector operating at 173◦ .
ration technique followed by ultrasonication, and the effect of The zeta potentials (mV) were measured by laser Doppler anemom-
composition on the physicochemical characteristics of the formu- etry using the same Malvern Zetasizer Nanoseries instrument. All
lations has been extensively studied. In vitro release kinetics and samples were diluted ten times with double distilled water prior
stability of SIM-NLCs were also evaluated. In vivo pharmacokinetics to measurements. All the measurements were performed at 25 ◦ C
and pharmacodynamics of SIM-NLCs have been investigated in nor- in triplicates.
mal rats and poloxamer-induced hyperlipidemic Wister rat model,
respectively, to evaluate the ability of the developed nanocarriers 2.3.3. In vitro release study
to improve the oral bioavailability and antihyperlipidemic activity In vitro release of SIM from the NLCs was studied under sink
of the drug. conditions using the dialysis method [5]. The dialysis bag (molec-
ular weight cut off: 12–14 kDa) was soaked in distilled water for
2. Materials and methods 12 h before use. Nanocarriers (equivalent to 1 mg of the drug) were
placed into the dialysis bag and immersed in 100 mL of phosphate-
2.1. Materials buffered saline (PBS, pH 7.4) in a beaker. The beaker was placed
in a thermostatic shaker (Gesellschaft für Labortechnik mbH, Burg-
Stearic acid was purchased from El-Nasr Chemicals Co. (Abu Zaa- wedel, Germany) at 37 ◦ C and 100 rpm. Five milliliter aliquots were
bal, Egypt). Oleic acid was purchased from Alpha Chemicals (Cairo, withdrawn at pre-determined time intervals (0.5, 1, 2, 4, 6, 8 and
Egypt). Simvastatin was purchased from El-Ryad Pharma (El-Ryad, 24 h), and replaced immediately with equal volumes of fresh PBS to
KSA). Pluronic F-68 was provided by BASF (Luwigshafen, Germany). maintain the same volume. The release of free SIM (SIM suspended
Lecithin was purchased from Sigma-Aldrich Co. (St. Louis, MO). in PBS) was examined as a control. The withdrawn aliquot at each
Other chemicals and reagents were of analytical grades. time interval was analyzed for the amount of SIM spectrophoto-
metrically at a wavelength of 238 nm. The cumulative amount of
2.2. Preparation of simvastatin-nanostructured lipid carriers drug release was calculated as a function of time [27]. The release
(SIM-NLCs) kinetics were analyzed by curve fitting to different kinetic models
of zero order, first order, Higuchi and Korsemeyer-Peppas model
SIM-NLCs were prepared via emulsification-solvent evapora- [28–30].
tion technique followed by ultrasonication as reported earlier with
slight modifications [26]. Briefly, certain amount of SIM, stearic 2.3.4. Stability studies
acid, oleic acid, and lecithin were dissolved in chloroform and Samples of the prepared formulations were stored in sealed
heated at 70 ◦ C. Aqueous phase containing the surfactant (Pluronic glass vials at room temperature or at 4 ◦ C for 30 days. Samples from
F-68) in 10 mL of deionized water was heated to 70 ◦ C. The aqueous each batch were withdrawn at definite time intervals and evaluated
phase was then added to the lipid phase under stirring at 1200 rpm for physical appearance, particle size and zeta potential. In addition,
and 70 ◦ C, and mixed for 5 min. The obtained pre-emulsion was further experiments were performed to study the changes of parti-
sonicated by probe-type sonicator (Cole-Parmer, Vernon Hills, IL) cle characteristics (size, PDI, zeta potential) for the selected optimal
for 7 min (40 W). Then, the dispersions were cooled down to the formulation (SIM-NLCs, F4) in media of different pHs (1.2, 6.8, 7 and
room temperature under agitation at 300 rpm for 1 h to obtain the 7.4). The particle characteristics were assessed immediately after
aqueous NLC dispersions. Purification of SIM-NLCs was performed mixing with the buffers (t0 ) and 2 h after the incubation (t2 ).
via dialysis into 250 mL of double distilled water and stirred at
300 rpm for 24 h, to remove the free drug. The dialysis water was 2.3.5. The particle morphology
replaced with fresh double distilled water after 12 h. The concen- Morphology of the formed particles was studied by transmis-
tration of the drug was adjusted to ensure that the purification sion electron microscopy (TEM). Samples were prepared by placing
process is carried out under sink conditions (i.e. the dialysis vol- a drop of SIM-NLCs onto a 400-mesh copper grid coated with a car-
ume exceeds the volume required to solubilize the total amount bon film, and left for air drying. TEM images were then observed at
of the drug). Eight different formulations of different compositions an acceleration voltage of 80 kV by TEM (JEM 100 CX11, Japan). The
were prepared (F1-F8, Supplementary information, Table S1). The surface morphology of SIM-NLCs was visualized by scanning elec-
238 H.A. Fathi et al. / Colloids and Surfaces B: Biointerfaces 162 (2018) 236–245

tron microscopy (SEM, JSM-5400 LV; JEOL, Japan). Samples were enzyme, which is involved in cholesterol biosynthesis [19–21].
prepared by placing a droplet onto an aluminum specimen stub, Twenty rats were divided into five groups, four animals in each
dried overnight, and sputter-coated with gold prior to imaging. An group, namely, normal group receiving normal saline, hyperlipi-
acceleration voltage of 15 kV was utilized for SEM visualization. demic control group receiving water, standard group receiving
free drug in a form of suspension, and two test groups receiving
2.3.6. Fourier transform-infrared spectroscopy the selected formulations (F4 and F6). The drug suspension was
Prior to characterizations by the Fourier transform-infrared prepared by suspending 10 mg of SIM in 10 mL of distilled water
(FT-IR) spectroscopy, differential scanning calorimetry and x-ray containing 0.5% w/v sodium carboxymethyl cellulose. The rats were
diffractometry, SIM-NLCs were frozen at – 80 ◦ C overnight prior fasted overnight prior to the study with free access to water. Hyper-
to the lyophilization process. Freeze-drying was performed using lipidemia was induced by a single intraperitoneal injection of 1 g/kg
Alpha 2–4 LD plus freeze-dryer (Martin Christ GmbH, Osterode, poloxamer F-127 solution (20% w/v). The rats were orally dosed
Germany) and the operating conditions were set at a temperature with multiple doses of drug suspension and formulations after
of − 60 ◦ C and a pressure of 0.011 mbar. The FT-IR spectra of the 12 h of the poloxamer injection for three days (dose = 5 mg/kg/day).
selected formulations (F4 and F6), corresponding physical mix- Blood samples were withdrawn at 0, 12, 36 and 60 h.
tures and the individual solid components were recorded using For further investigation of the sustained antihyperlipidemic
FT-IR spectrophotometer (IR-470; Shimadzu, Kyoto, Japan). Sam- activity of SIM-NLCs in comparison to SIM-suspension, pharmaco-
ples were mixed with potassium bromide (spectroscopic grade) dynamic study during the first 24 h after single oral administration
and compressed into disks using hydraulic press before scanning of the different treatments was also examined. Eight rats were
from 4000 to 500 cm−1 . divided into two groups (four rats in each group) for the oral admin-
istration of a single dose of the selected SIM-NLCs to one group and
2.3.7. Differential scanning calorimetry the drug suspension to the second group (dose = 5 mg/kg). Multi-
The thermal behaviors of the selected formulations (F4 and F6), ple blood samples were collected before drug administration and
corresponding physical mixtures and the individual solid com- 2-, 6-, 12- and 24 h following drug administration. Blood samples
ponents were investigated by differential scanning calorimetry were allowed to clot for 15–30 min at room temperature, and serum
(DSC-60; Shimadzu Corporation, Tokyo, Japan). Samples of 4–7 mg was separated by centrifugation at 3000 rpm for 10 min (Sigma
were sealed in aluminum pans and heated over a temperature range Laborzentrifugen GmbH, Osterode am Harz, Germany). The serum
of 40–200 ◦ C at a constant rate of 10 ◦ C/min. Thermal analysis data samples were then frozen at − 20 ◦ C and stored for further analysis
were recorded using a TA 50I PC system with Shimadzu software [33].
programs. Indium standard was used to calibrate the DSC temper- Blood serum was analyzed for total cholesterol (TC) and high
ature and enthalpy scale. N2 was used as purging gas at a rate of density lipoprotein cholesterol (HDL-C) levels. TC levels were
50 mL/min. An empty sealed aluminum pan was used as a reference. determined by enzymatic, colorimetric assay, and HDL-C levels
were measured by homogeneous enzymatic colorimetric assay
2.3.8. X-ray diffraction on Roche/Hitachi cobas c 311 analyzer (Roche Ltd. Mannheim,
X-ray diffraction (XRD) patterns were studied to identify the Germany). The non-HDL-cholesterol (non-HDL-C) concentrations
crystal form of SIM dispersed in the lipid matrix. XRD patterns of the (i.e. cholesterol concentrations in the lower density lipoproteins
selected formulations (F4 and F6), corresponding physical mixtures (chylomicrons, low and very low density lipoproteins)) were cal-
and the individual solid components were recorded by Philips X-ray culated by subtracting HDL-C from the TC. Moreover, the% of initial
diffractometer model PW 1710 (Philips, Amsterdam, Netherlands) TC level and% of initial non-HDL-C level were also calculated at each
with CuK ␣ radiation ( = 1.5405 Å), at a voltage of 40 kV and 30 mA time point according to Eqs. (2) and (3):
current. Samples were scanned at room temperature from 2␪ = 5◦
Measured TC level
to 2␪ = 50◦ with a step of 0.06◦ /min. % of initial TC level = × 100 (2)
Baseline TC level
2.4. In vivo studies Measured non − HDL − C level
% of initial non − HDL − C level = × 100 (3)
Baseline non − HDL − C level
In vivo experiments were carried out to study the pharmaco-
dynamics and pharmacokinetics of SIM-NLCs as compared to SIM 2.4.3. Pharmacokinetic study
suspension after oral administration in rats. Pharmacokinetic study was performed to investigate the ability
of NLCs to improve SIM oral bioavailability. The rats were randomly
2.4.1. Experimental animals divided into two groups (three animals per group) and received a
The research protocol was reviewed and approved by the Insti- single oral dose of SIM (20 mg/kg) in the form of free drug sus-
tutional Animal Ethical Committee of the Faculty of Pharmacy, pension for the first group and SIM-NLCs for the second group.
Assiut University, and it adheres to the Guide for the Care and Use of Although this dose is higher than the clinically used dose, phar-
Laboratory Animals, 8th Edition, National Academies Press, Wash- macokinetic studies in rodents indicate that higher statin doses
ington, DC. Male Wister rats (200–250 g) were obtained from the (as compared to human) are required to achieve similar effec-
Animal House at the Assiut University Faculty of Medicine. Rats tive concentrations [13,14]. Hence, we have selected the dose of
were allowed to acclimatize to the experimental conditions of tem- 20 mg/kg/day, which is consistent with doses that have been used
perature and humidity one week prior to the experiments. The rats in other reported pharmacokinetic studies for SIM [15]. Also, a
were fed a standard rat pellet diet and allowed free access to water. higher dose (20 mg/kg) was used in the pharmacokinetic studies,
The rats were maintained at a temperature of 25 ± 2 ◦ C with a 12 h as compared to the pharmacodynamic studies (5 mg/kg), to allow
dark/light cycles throughout the study. for the detection of low concentrations of the drug in the plasma
(on the nanogram scale). All formulations were freshly prepared.
2.4.2. Pharmacodynamic study of SIM-NLCs All rats were fasted overnight prior to the administration of the
The anti-cholesterolemic effects of different SIM-NLCs and SIM formulations and were fed again 4 h after treatments. Blood sam-
suspension were studied using poloxamer-induced hyperlipidemic ples (0.5 mL) were obtained via vein puncture from the caudal vein
rat model [31,32]. Poloxamer is a non-ionic surfactant that induces at designated time points (1, 2, 4, 8, 12 and 24 h after adminis-
hyperlipidemia via indirect stimulation of HMG-CoA reductase tration) and were transferred into heparinized tubes. The plasma
H.A. Fathi et al. / Colloids and Surfaces B: Biointerfaces 162 (2018) 236–245 239

samples were separated by centrifugation at 3000 rpm for 10 min. tions of 1.5, 1, 3 and 5% w/v (coded as F4, F6, F7 and F8, respectively)
The plasma samples were then stored frozen at −20 ◦ C for further while maintaining the oleic acid-to-stearic acid ratio constant. Eval-
analysis. uation of the prepared NLCs was carried out by measuring the
The SIM concentrations in plasma were analyzed using a particle size, PDI, zeta potential and entrapment efficiency.
reversed phase-HPLC as described previously, with some modifi- Table S2 (Supplementary Information) shows the particle size,
cations [34]. The HPLC method was performed on a Dionex Ultimat PDI and zeta potential of the freshly prepared NLCs at different oleic
3000 UHPLC system (Thermo Scientific, Waltham, MA) equipped acid and surfactant concentrations. All formulations were in the
with a HPG-3200 RS pump, a DAD-3000 RS detector, a WPS- nanosized range, and demonstrated a highly significant decrease
3000TRS analytical autosampler, and a Hypersil BDS C18 analytical (p < 0.001) in both the particle size and PDI upon increasing oleic
column (dimensions of 150 mm × 4.6 mm ID × 5 ␮m). The mobile acid concentration from 15% to 70%. The decrease in size is due
phase composed of acetonitrile:deionized water (65:35 v/v) and to the difference in viscosity between the liquid lipid (oleic acid)
was adjusted to pH 3.5 by phosphoric acid. The flow rate was set and solid lipid (stearic acid) [19,36]. The high oleic acid content
at 1.0 mL/min, the injection volume was 20 ␮L and elute was ana- reduced the viscosity inside the NLCs, and, thus reducing the inter-
lyzed with DAD detector set at a wavelength of 239 nm. Acetonitrile facial tension and afford the formation of smaller particles with
(1 mL) was added to the plasma sample (200 ␮L) to precipitate the homogeneous size distribution and smooth surface [36,37]. On the
plasma proteins, and the samples were then vortexed and cen- contrary, there was an increase in particle size upon increasing the
trifuged at 4000 rpm for 15 min [35]. The supernatant was collected, concentration of solid lipid (stearic acid) due to the increased vis-
filtered through 0.45 ␮m syringe filter (Millipore, Billerica, MA), cosity and interfacial tension, which is consistent with previously
and analyzed using the HPLC. A calibration curve of SIM in the reported data [37,38]. The small particle size and high negative
plasma was constructed using blank plasma spiked with standard zeta potential values of NLCs indicate the stability of the formed
SIM solutions to obtain a concentration range of 0.01–10 ␮g/mL. colloidal dispersion. No change in the zeta potential values was
The spiked plasma samples were then subjected to the same extrac- observed upon increasing the oleic acid content of nanoparticles,
tion procedure described previously. which is consistent with previously reported data for clobetasol-
The pharmacokinetic parameters were calculated by fitting the loaded NLCs [36].
plasma concentration–time data to a suitable model using Win Regarding the effect of surfactant concentration, highly sig-
Nonlin Professional Edition software version 2.0 (Science Consult- nificant reduction (p < 0.001) in particle size and increase in the
ing, Apex, NC). The area under the plasma concentration-time curve zeta potential values (p < 0.001) were observed upon increasing the
from zero to infinity (AUC0-∞ ) was calculated using the trapezoidal surfactant concentration from 1 to 5% w/v (Supplementary informa-
rule method. The maximum concentration (Cmax ), time to reach tion, Table S2). The reduction of particle size might be attributed to
the maximum concentration (Tmax ), elimination half-life (t½ ), elim- the decreased interfacial tension between the lipid matrix and the
ination rate constant (Kel ), mean residence time (MRT), clearance aqueous phase upon increasing the surfactant concentration. The
(CL) and volume of distribution (Vd ) were calculated. The relative nonionic nature of the surfactant (Pluronic F-68) provides steric
bioavailability (Fr , %) of SIM-NLCs after oral administration was stabilization, and, thus preventing aggregation of the particles,
calculated according to Eq. (4): and consequently preventing the coalescence of the droplets and
preserving the physical stability of the lipid nanoparticles. Since
(AUC)SIM−NLCs
Relative bioavailability (Fr, %) = × 100 (4) Pluronic F-68 is nonionic, the increase of zeta potential values that
(AUC)SIM suspension
is observed upon increasing the surfactant concentration can be
attributed to the coating layers of the surfactant that shield the
2.5. Statistical analysis negative surface charge of stearic acid [39,40]. It was observed that
there was no significant change in the PDI of NLCs upon increasing
Experiments were performed in triplicates unless otherwise surfactant concentration from 1 to 3% (p > 0.05). On the other hand,
indicated. Statistical significance for differences between exper- further increase of surfactant concentration to 5% led to a highly
imental groups was assessed by one-way analysis of variance significant increase (p < 0.01) of PDI of NLCs from 0.18 to 0.30.
or two-sided Student’s t-test for pairwise comparison (Graph-
Pad Prism 6.0, GraphPad, San Diego, CA). Differences between
means were considered statistically non-significant for p values
>0.05. The differences were considered as statistically significant 3.2. Entrapment efficiency
for 0.05> p ≥ 0.01, and highly significant for p < 0.01.
The effect of concentrations of oleic acid and Pluronic F-68 on
3. Results and discussion the EE of the drug in the prepared NLCs was also studied (Sup-
plementary information, Table S2). Increasing the concentration of
3.1. Preparation and characterizations of the SIM-NLCs oleic acid from 15 to 80 wt% resulted in a highly significant increase
(p < 0.001) in the EE of the drug into the NLCs from 40.44 ± 2.7 to
SIM-NLCs were prepared via emulsification-solvent evapora- 78.40 ± 2.9%, respectively. The resulted improve in EE upon increas-
tion technique followed by ultrasonication. The effects of oleic ing oleic acid content is in agreement with a previous study, which
acid-to-stearic acid ratio and Pluronic F-68 concentration were reported that incorporation of liquid lipids into solid lipids led to a
evaluated for further optimization of the formulations (Supplemen- massive crystal order disturbance that left enough space to accom-
tary Information, Table S1). Since oleic acid concentration affects modate drug molecules, thus leading to improved drug entrapment
the viscosity inside NLCs, different formulations were prepared efficiency [36]. On the other hand, no significant change (p > 0.05) in
using constant Pluronic F-68 concentration (1.5% w/v) and various EE was detected upon increasing surfactant concentration from 1 to
oleic acid-to-stearic acid ratios (15, 30, 50, 70 and 80 wt% of oleic 3%. Moreover, further increase of surfactant concentration to 5% led
acid/total lipid), and coded as F1, F2, F3, F4 and F5, respectively. to a highly significant decrease (p < 0.01) in EE from 75.77 ± 4.9 to
On the other hand, the surfactant at the aqueous/organic interface 54.58 ± 3.7%, which might be attributed to the hydrophobic nature
governs the effectiveness of the emulsification process and sta- of the drug that tends to diffuse out from the oil nanodroplets
bilization of the oil nanodroplets during the solvent evaporation and solubilize in the micelles formed in the aqueous phase upon
process. Therefore, Pluronic F-68 was used at different concentra- increasing the surfactant concentration [41]. Drug loading did not
240 H.A. Fathi et al. / Colloids and Surfaces B: Biointerfaces 162 (2018) 236–245

Fig. 1. In vitro release profiles of simvastatin from nanostructured lipid carriers, as compared to the drug suspension: (A) Different oleic acid concentrations and (B) Different
surfactant (Pluronic F-68) concentrations. Data are presented as means ± SD (n = 3).

increase the size of NLCs, which is consistent with a previous report were no significant differences (p > 0.05) in either particle size or
[42]. zeta potential values for all the formulations except for F5, F7 and
F8, which had a significant increase (p < 0.001) in the mean particle
3.3. In vitro drug release size after 30 days of storage. Among all NLC formulations, formula-
tions F4 and F6 were selected for further investigations due to their
The release profiles of SIM from the lipid nanocarriers of vari- small particle sizes, higher entrapment efficiencies, slower in vitro
ous compositions were studied over 24 h (Fig. 1). The drug release cumulative drug release, higher stability upon storage, and their
patterns demonstrated initial burst release followed by sustained lower surfactant contents. Moreover, neither F4 nor F6 exhibited
release at a constant rate over 24 h. The initial rapid release may any significant change (p > 0.05) in the EE after 30 days of stor-
be attributed to the fast release of drug entrapped near the sur- age either at 4 ◦ C (F4: 70.64 ± 7.6%, F6: 66.30 ± 8.2%) or at room
face of the nanoparticles. The sustained release of drug after the temperature (F4: 68.62 ± 6.61%, F6: 51.30 ± 15%).
initial burst release could be related to the lipophilic nature of SIM The effect of pH on the particle size, PDI and zeta potential was
that is entrapped deeply in the core matrix of the NLCs. SIM in also studied (Supplementary information, Tables S5 and S6). At low
the core of the nanoparticles has a longer diffusion path to reach pH, there was no significant change (p > 0.05) in size and PDI of the
the surface, as compared to the drug entrapped near the surface NLCs in comparison to the original sample (NLCs in water), which
[43]. Faster drug release patterns were observed upon increasing may be attributed to the steric stabilization effect of the nonionic
the concentrations of oleic acid (Fig. 1A) and the surfactant con- surfactant (Pluronic F-68) [12]. At pH 7.4, there was a slight increase
centration (Fig. 1B). This enhanced drug release from the NLCs in particle size, which is consistent with earlier studies [12,13]. The
might be attributed to the significant decrease in particle size, and zeta potential of the SIM-NLCs reached 1.27 ± 1.67 mV when the pH
thus increasing the specific surface area and, subsequently, the was decreased to 1.2, which might be attributed to protonation of
release rate of the drug [36]. Table S3 (Supplementary informa- the anionic groups of lecithin and oleic acid, thereby reducing the
tion) summarizes the release kinetics and correlation coefficients net negative charge [14].
(R2 ) calculated for the tested formulations. The in vitro release data
indicated that the release of SIM from NLC formulations followed 3.5. Morphology of the SIM-NLCs
first order kinetics except for F1 (15 wt% OA) and F2 (30 wt% OA),
which were fitted to a diffusion–controlled mechanism (Higuchi Fig. 2A shows the transmission electron micrograph of SIM-
model). Korsmeyer-Peppas was utilized to provide further informa- NLCs. The lipid nanocarriers appeared as spherical or elliptical
tion on drug release mechanisms, whether it is Fickian (diffusion), vesicles with smooth surfaces. There were several dark spots on
non-Fickian (anomalous), or erosion-mediated (zero order) release. the spherical structures of the NLCs, which might be attributed to
The values of the diffusional release exponents for all formulations the liquid lipids sticking on the surface of the lipid nanoparticles
were <0.5 except for F6 (1% w/v Pluronic F-68), which indicates that [44,45]. The average particle size was ca. 169.4 ± 33.9 nm (n = 20),
Fickian mechanism is the dominant mechanism that controls the which is in agreement with the DLS results (Fig. 2C). Scanning elec-
drug release from the nanoparticles. tron micrograph clearly demonstrates the spherical shape of the
prepared NLCs with characteristic smooth surface (Fig. 2B).
3.4. Stability studies
3.6. Fourier transform-infrared spectroscopy
The stability of the various NLC formulations was monitored at
4 ◦ C and at room temperature for 30 days to assess their long-term The FT-IR spectra of the pure drug, the selected formulations (F4
stability (Supplementary Information, Table S4). The visual obser- and F6), physical mixtures, and the individual solid components,
vation of all the prepared NLCs did not indicate any sign of gelation, were recorded (Fig. 3). The spectrum of pure SIM drug showed
creaming, color change or particle aggregation either at 4 ◦ C or at characteristic peaks at 3012 cm−1 (C CH), 1698.9 cm−1 (aromatic
room temperature. After 4 weeks, it could be noticed that there C O), 3551.4 cm−1 (aliphatic O-H) and 1466.7 cm−1 (benzene ring).
H.A. Fathi et al. / Colloids and Surfaces B: Biointerfaces 162 (2018) 236–245 241

Fig. 2. Transmission electron micrograph (A), scanning electron micrograph (B) and Intensity-, volume- and number-averaged hydrodynamic diameter histograms of
the SIM-NLCs (F4) (C). The intensity-, volume- and number-averaged hydrodynamic diameters of SIM-NLCs (F4) demonstrated on Fig. 2C are 185.2, 175.9 and 120.9 nm,
respectively.

peared, shifted or replaced by the peak of stearic acid, and thus


indicating the drug entrapment in the lipid matrix [47].

3.7. Differential scanning calorimetry

DSC thermograms of the pure drug, the selected formula-


tions (F4 and F6), physical mixtures, and solid components, were
recorded (Fig. 4I). The thermogram of the pure SIM showed a sharp
single melting peak at ca. 138.5 ◦ C, which corresponds to the melt-
ing of the crystalline drug [7,11,34]. In the physical mixture of SIM
and stearic acid, the characteristic peak of the drug disappeared
with slight increase in the onset temperature of the stearic acid
(62.2 ◦ C). The disappearance of the drug characteristic peak can
be ascribed to solubilization of the drug within the lipid matrix
Fig. 3. FT-IR spectra of the pure drug (SIM) (A), stearic acid (B), Pluronic F-68 (C),
physical mixture (SIM/stearic acid) (D), physical mixture (SIM/Pluronic F-68) (E), F4 [47,48]. DSC thermograms of F4 and F6 showed a decrease in the
(F) and F6 (G). onset of melting peak of the used lipid and disappearance of the
characteristic melting peak of the drug, which may be attributed
to drug solubilization within the lipid matrix in amorphous and/or
This pattern is in agreement with previously recorded spectrum molecularly dispersed form [49]. On the other hand, the decrease
for the same drug [46]. The characteristic peaks of the drug were in the melting temperature of the used lipid might be due to the
also observed in the FT-IR spectrum of the physical mixture of pure interactions between the solid lipid and the liquid lipid and the sur-
drug/Pluronic F-68 due to the absence of predominant chemical factant. Also, it may be attributed to the small particle size of the
interactions between the drug and the surfactant. However, in the prepared NLCs, as a result of the kelvin effect, which is described by
spectrum of SIM-NLCs, the characteristic peaks of the drug disap- the Thomson equation. According to this equation, small isolated
242 H.A. Fathi et al. / Colloids and Surfaces B: Biointerfaces 162 (2018) 236–245

eters are not sufficient to predict the pharmacodynamic activity


[52]. This issue has been adequately discussed in a previous study
which reported that plasma concentrations of atorvastatin did not
correlate with the reduction in LDL-C [25]. Therefore, the pharma-
codynamics and pharmacokinetics of the selected SIM-NLCs and
the drug suspension were evaluated.
After 12h of poloxamer injection into the rats, there was ca. 5-
fold increase in the TC level of the hyperlipidemic control group
and the different treatment groups as compared to normal group
(data not shown). The rats were orally dosed with multiple doses
of drug suspension and formulations after 12 h of the poloxamer
injection for three days (dose = 5 mg/kg/day). Percentages of the
initial (before drug administration) TC and non-HDL-C levels were
estimated to evaluate the efficacy of the treatment in each group.
Percentages of initial TC and non-HDL-C levels after oral admin-
istration of the SIM-NLCs (F4 and F6) and SIM suspension are
presented on Fig. 5I. The TC level after 12 h was significantly higher
(p < 0.001) in the control group as compared to the treated groups,
which indicate the ability of the SIM-NLCs and the drug suspension
to prevent the increase in TC levels (Fig. 5IA). After 36 h of the treat-
ment, there was a further increase in the TC levels of the control
rats, and rats treated with the SIM suspension and SIM-NLCs-F6.
Interestingly, in the case of F4 formulation, the TC significantly
decreased (p < 0.001). After 60h of the treatment, F4 reduced the
TC level to a highly significant level (p < 0.01), as compared to F6
and the drug suspension. Similar significant decrease (p < 0.001)
in the non-HDL-C level was also observed for F4-treated group, as
compared to F6 and the SIM suspension-treated groups (Fig. 5IB).
Fig. 4. DSC analysis (I) and X-ray diffraction patterns (II) of the pure drug (SIM) (A),
The greater lipid lowering activity of the F4 (1.5% w/v Pluronic F-
stearic acid (B), Pluronic F-68 (C), physical mixture (SIM/stearic acid) (D), physical 68) as compared to F6 (1% w/v Pluronic F-68) might be attributed
mixture (SIM/Pluronic F-68) (E), F4 (F) and F6 (G). to the higher Pluronic F-68 concentration of F4. The presence of
Pluronic F-68 might enhance the absorption of NLCs, due to its
effect on the intestinal epithelial permeability [53]. Furthermore,
particles (SIM-NLCs) would melt at a lower temperature than the
F4 has smaller particle size that might increase the absorption and
bulk material (stearic acid) [38].
dissolution rate, and thus improving the drug bioavailability [34].
For further investigation of the sustained antihyperlipidemic
3.8. X-ray diffraction effect of the SIM-NLCs in comparison to the SIM suspension, single
dose pharmacodynamic study was evaluated and the percentages
X-ray diffractograms of the pure drug, F4, F6, physical mixtures, of the initial TC and non-HDL-C levels were measured (Fig. 5II).
and the individual solid components are presented in Fig. 4II. The After 2 h of the treatment, the TC and non-HDL-C levels were sig-
crystalline nature of pure SIM was demonstrated upon detection nificantly higher (p < 0.001) in the SIM suspension-treated group,
of the diffraction peaks in the XDR pattern. These characteristic as compared to the SIM-NLCs-treated group (F4). Moreover, the
diffraction peaks of the pure drug were also detected on the XDR enhanced efficacy of the F4 remained detectable during the 24 h of
pattern of the physical mixtures, however, they disappeared in the the treatment period. On the contrary, SIM suspension resulted in
diffractogram of SIM-NLCs, which clearly confirm that SIM did not a detectable increase in TC and non-HDL-C levels above the base-
crystallize in the lipid matrix. The crystalline nature of the pure line level, and this increase was continued during the 24 h of the
lipid and surfactant was also confirmed by the diffraction peaks treatment. The sustained antihyperlipidemic activity of the SIM-
observed in the XDR. However the disappearance of the character- NLCs might lower the dose and frequency of administration, and
istic peaks of the surfactant on diffractogram of SIM-NLCs indicates thus improving patient tolerability and decreasing side effects of
that the crystallinity of Pluronic F-68 is lost when it is adsorbed on the drug.
the stearic acid nanoparticles, due to the change in the surfactant
conformation [47]. 3.10. Pharmacokinetic study

3.9. Pharmacodynamic study of SIM-NLCs Selected SIM-NLCs (F4) and SIM suspension were orally admin-
istrated as a single dose to male Wistar rats. SIM plasma
Hypercholesterolemia is usually managed by lowering the concentrations were measured using a previously reported HPLC
serum concentration of TC and low density lipoprotein choles- method, with some modifications [34]. SIM was completely sep-
terol (LDL-C) or non-HDL-C. Non-HDL-C contains more atherogenic arated as a sharp peak at a retention time of ca. 8.2 min without
cholesterol than LDL-C, and is considered as a more accurate any interfering peaks. SIM plasma concentrations after oral admin-
measurement of the total amount of atherogenic particles in the istration of SIM-NLCs and the drug suspension are presented on
circulation [50]. Moreover, recent investigations indicate that non- Fig. 6. The plasma concentration-time data were subjected to a non-
HDL-C ≥ LDL-C is a predictor of coronary artery diseases [51]. SIM is compartmental analysis to determine the mean pharmacokinetic
a potent lipid-lowering drug that suppresses the formation of bad parameters (Supplementary information, Table S7).
cholesterol in the liver through inhibiting the HMG-CoA reductase The SIM plasma concentrations were remarkably higher after
enzyme, thus resulting in substantial decrease in mortality from administration of SIM-NLCs, as compared to those observed for
cardiovascular diseases. For statins, the pharmacokinetic param- SIM suspension. The Cmax for SIM-NLCs was significant higher than
H.A. Fathi et al. / Colloids and Surfaces B: Biointerfaces 162 (2018) 236–245 243

Fig. 5. (I) Changes in cholesterol levels in rats after administration of simvastatin suspension and simvastatin-nanostructured lipid carriers (F4 and F6) over 3 days. (II)
Changes in the cholesterol levels in rats after single dose administration of simvastatin suspension and simvastatin-nanostructured lipid carriers (F4). (A) Total cholesterol
and (B) Non-high density lipoprotein-cholesterol. *p < 0.05 when compared to the drug suspension; **p < 0.001 when compared to the drug suspension. Data are presented
as means ± SD (n = 4).

the SIM suspension (89.86 ± 4.11 vs. 45.73 ± 12.15 ng/mL, p < 0.05). was previously reported using other drug delivery systems such as
After 24 h of oral administration of SIM–NLCs, the SIM plasma solid lipid nanocarriers [7] and self microemulsifying drug deliv-
concentration was 34.54 ± 1.52 ng/mL, whereas SIM suspension ery system [54]. In the case of SIM-NLCs, the pre-systemic hepatic
resulted in a plasma concentration of 9.71 ± 1.55 ng/mL. The higher effect can be largely avoided via absorption through the lymphatic
plasma concentrations observed following the oral administra- pathway [7]. Improved solubilization of SIM due to the transforma-
tion of SIM-NLCs as compared to the drug suspension reflect the tion from crystalline to amorphous form is another reason for the
enhanced systemic absorption of SIM when incorporated into the increased systemic absorption [55]. The other factors that facilitate
NLCs. Similar enhancement of systemic absorption of the same drug the absorption in the intestinal milieu are the high dispersibility of
244 H.A. Fathi et al. / Colloids and Surfaces B: Biointerfaces 162 (2018) 236–245

to enhance the oral delivery and prolong the therapeutic effect of


SIM, and thus improving the patient compliance by eliminating the
need for frequent dosing of the drug.

Conflicts of interest

The authors report no conflicts of interest in this work.

Appendix A. Supplementary data

Supplementary data associated with this article can be found,


in the online version, at https://doi.org/10.1016/j.colsurfb.2017.11.
064.

References
Fig. 6. Plasma concentration–time profiles of SIM after single oral dose (20 mg/kg)
of SIM suspensions and SIM-NLCs. Values are presented as means ± SD (n=3). [1] M. Casula, E. Tragni, A.L. Catapano, Patient Prefer Adherence 6 (2012) 805.
[2] J.G. Robinson, B. Smith, N. Maheshwari, H. Schrott, J. Am. Coll. Cardiol. 46
(2005) 1855.
[3] S.M. Grundy, J.I. Cleeman, C.N. Merz, H.B. Brewer Jr., L.T. Clark, D.B.
the NLCs and small particle size of the NLC formulation (<200 nm) Hunninghake, R.C. Pasternak, S.C. Smith Jr., N.J. Stone, J. Am. Coll. Cardiol. 44
that resulted in a great increase in the surface area of the particles. (2004) 720.
Furthermore, NLCs could protect the drug from enzymatic degrada- [4] K. Sharma, K. Kumar, N. Mishra, Drug Deliv. 23 (2016) 694.
[5] Z. Zhang, H. Bu, Z. Gao, Y. Huang, F. Gao, Y. Li, Int. J. Pharm. 394 (2010) 147.
tion by reducing the exposure of the drug to the intestinal bacteria, [6] G. De Angelis, Int. J. Clin. Pract. 58 (2004) 945.
as well as, protecting the drug from the enzymatic degradation dur- [7] R. Tiwari, K. Pathak, Int. J. Pharm. 415 (2011) 232.
ing the absorption process, and thus allowing for a longer residence [8] P. Patil, V. Patil, A. Paradkar, Acta Pharm. 57 (2007) 111.
[9] R.P. Dixit, M.S. Nagarsenker, Drug Dev. Ind. Pharm. 34 (2008) 1285.
time in the gastrointestinal tract [56]. In general, improved phar-
[10] J. Lai, J.M. Chen, Y. Lu, J. Sun, F.Q. Hu, Z.N. Yin, W. Wu, AAPS PharmSciTech 10
macokinetic profile was observed when SIM was loaded into the (2009) 960.
NLCs (Supplementary Information, Table S7). The SIM-NLCs exhib- [11] S.G. Padhye, M.S. Nagarsenker, Indian J. Pharm. Sci. 75 (2013) 591.
[12] T. Jiang, N. Han, B. Zhao, Y. Xie, S. Wang, Drug Dev. Ind. Pharm. 38 (2012) 1230.
ited a delayed Tmax (2 h), as compared to that of SIM suspension
[13] M. Shah, K. Chuttani, A.K. Mishra, K. Pathak, Drug Dev. Ind. Pharm. 37 (2011)
(1 h), which could be attributed to the slow and sustained release 526.
of the drug from the NLCs. Longer elimination half-life of SIM-NLCs [14] H. Elzeny, F. Zhang, E.N. Ali, H.A. Fathi, S. Zhang, R. Li, M.A. El-Mokhtar, M.A.
(35.04 ± 2.86 h) was also observed as compared to the SIM sus- Hamad, K.L. Wooley, M. Elsabahy, Drug Des. Dev. Ther. 11 (2017) 483.
[15] M. Elsabahy, M.H. Dufresne, J.C. Leroux, Handbook of Materials for
pension (10.20 ± 0.47 h), which might aid in reducing the dose and Nanomedicine, 1, 2010, pp. 169.
frequency of administration. The extent of SIM absorption from NLC [16] L. Gao, D.R. Zhang, M.H. Chen, J. Nanopart. Res. 10 (2008) 845.
(AUC0-∞ ) was significantly higher (p < 0.001) than the AUC0-∞ of the [17] B.E. Rabinow, Nat. Rev. Drug Discov. 3 (2004) 785.
[18] R.H. Muller, K. Mader, S. Gohla, Eur. J. Pharm. Biopharm. 50 (2000) 161.
SIM suspension. [19] V. Jenning, A.F. Thunemann, S.H. Gohla, Int. J. Pharm. 199 (2000) 167.
The oral bioavailability of SIM after its incorporation into [20] J. Pardeike, K. Schwabe, R.H. Muller, Int. J. Pharm. 396 (2010) 166.
solid lipid nanoparticles was improved by 3.37-fold [5]. Similarly, [21] V. Teeranachaideekul, E.B. Souto, V.B. Junyaprasert, R.H. Muller, Eur. J. Pharm.
Biopharm. 67 (2007) 141.
increase in SIM oral bioavailability from self microemulsifying drug [22] C.J.H. Porter, W.N. Charman, Adv. Drug Deliv. Rev. 50 (2001) 61.
delivery system (1.5-fold increase) [54] and nanosuspension (2.2- [23] C.Y. Zhuang, N. Li, M. Wang, X.N. Zhang, W.S. Pan, J.J. Peng, Y.S. Pan, X. Tang,
fold increase) [13] have been also reported previously. Our group Int. J. Pharm. 394 (2010) 179.
[24] T.H. Tran, T. Ramasamy, D.H. Truong, H.G. Choi, C.S. Yong, J.O. Kim, AAPS
has utilized various strategies to improve stability and delivery effi-
PharmSciTech 15 (2014) 1509.
ciency of several drugs [14,57–61]. In the current study, the relative [25] H. Lennernas, Clin. Pharmacokinet. 42 (2003) 1141.
bioavailability of SIM-NLCs was 4-fold higher than for the drug sus- [26] D. Pooja, L. Tunki, H. Kulhari, B.B. Reddy, R. Sistla, Data Brief. 6 (2016) 15.
[27] S.M. Hiremath, D. Sarudkar S, Indian J. Nov. Drug Deliv. 1 (2009) 18.
pension, indicating significant enhancement of oral delivery and
[28] A. Allam, G. Fetih, Drug Des. Devel Ther. 10 (2016) 2421.
bioavailability of SIM when incorporated into the NLCs. The rate and [29] F.G. El-Badry, D. Fathalla, F. Shakeel, Pharm. Dev. Technol. 20 (2015) 820.
extent of systemic absorption of SIM were significantly increased, [30] G.R. Korsmeyer, E. Doelker, P. Buri, N.A. Peppas, Int. J. Pharm. 15 (1983) 25.
and thus improving the oral bioavailability of SIM. It is worth men- [31] J. Zhou, D. Zhou, Drug Des. Dev. Ther. 9 (2015) 5269.
[32] H.R. Chaudhary, D.R. Brocks, J. Pharm. Pharm. Sci. 16 (2013) 65.
tioning that no mortalities have been observed in the treatment [33] J.I. Ihedioha, O.A. Noel-Uneke, T.E. Ihedioha, Comp. Clin. Pathol. 22 (2011) 93.
group and the hyperlipidemic control group in the in vivo experi- [34] G.I. Harisa, A.H. Alomrani, M.M. Badran, Eur. J. Pharm. Sci. 96 (2017) 62.
ments. [35] M. Elmowafy, A. Samy, M.A. Raslan, A. Salama, R.A. Said, A.E. Abdelaziz, W.
El-Eraky, S. El Awdan, T. Viitala, AAPS PharmSciTech 17 (2016) 663.
[36] F.Q. Hu, S.P. Jiang, Y.Z. Du, H. Yuan, Y.Q. Ye, S. Zeng, Colloids Surf. B
Biointerfaces 45 (2005) 167.
4. Conclusions
[37] L.J. Jia, D.R. Zhang, Z.Y. Li, F.F. Feng, Y.C. Wang, W.T. Dai, C.X. Duan, Q. Zhang,
Drug Deliv. 17 (2010) 11.
In the present study, SIM-NLCs were successfully prepared and [38] V. Teeranachaideekul, P. Boonme, E.B. Souto, R.H. Muller, V.B. Junyaprasert, J.
evaluated for their abilities to enhance oral delivery of SIM as com- Control. Release 128 (2008) 134.
[39] L. Wu, J. Zhang, W. Watanabe, Adv. Drug Deliv. Rev. 63 (2011) 456.
pared to the drug suspension. Several in vitro characterizations of [40] R. Gref, A. Domb, P. Quellec, T. Blunk, R.H. Muller, J.M. Verbavatz, R. Langer,
SIM-NLCs were carried out. SIM-NLCs of small particle size, high Adv. Drug Deliv. Rev. 16 (1995) 215.
entrapment efficiency, sustained in vitro drug release, high stability [41] N. Sharma, P. Madan, S.S. Lin, Asian J. Pharm. Sci. 11 (2016) 404.
[42] N.P. Aditya, A.S. Macedo, S. Doktorovov, E.B. Souto, S. Kim, P.S. Chang, S. Ko,
and low surfactant concentration were selected for further charac- Food Sci. Tchnol. 59 (2014) 115.
terizations. The in vivo pharmacodynamic study of the SIM-NLCs [43] A.R. El-Helw, U.A. Fahmy, Int. J. Nanomed. 10 (2015) 5797.
demonstrated a sustained and greater lipid lowering activity as [44] A. Saupe, K.C. Gordon, T. Rades, Int. J. Pharm. 314 (2006) 56.
[45] K. Jores, W. Mehnert, M. Drechsler, H. Bunjes, C. Johann, K. Mader, J. Control.
compared to the free drug suspension. There was a 4-fold increase Release 95 (2004) 217.
in the oral bioavailability of SIM-NLCs in comparison to the SIM sus- [46] H. Singh, B. Philip, K. Pathak, Iran J. Pharm. Res. 11 (2012) 433.
pension. The developed NLCs might provide efficient nanodevices [47] S. Kumar, J.K. Randhawa, RSC Adv. 5 (2015) 68743.
H.A. Fathi et al. / Colloids and Surfaces B: Biointerfaces 162 (2018) 236–245 245

[48] R.H. Muller, S.A. Runge, V. Ravelli, A.F. Thunemann, W. Mehnert, E.B. Souto, [56] M. Uner, Pharmazie 61 (2006) 375.
Eur. J. Pharm. Biopharm. 68 (2008) 535. [57] M. Elsabahy, M. Zhang, S. Gan, K.C. Waldron, J.C. Leroux, Soft Matter 4 (2008)
[49] G.I. Harisa, M.M. Badran, J. Mol. Liq. 208 (2015) 202. 294.
[50] R. Garg, C.R. Vasamreddy, R.S. Blumenthal, Prev. Cardiol. 8 (2005) 173. [58] M. Elsabahy, K.L. Wooley, Chem. Soc. Rev. 41 (2012) 2545.
[51] S.M. Grundy, Am. J. Cardiol. 81 (1998) 18B. [59] M. Elsabahy, R. Shrestha, C. Clark, S. Taylor, J. Leonard, K.L. Wooley, Nano Lett.
[52] A.A. Date, M.S. Nagarsenker, J. Pharm. Pharmacol. 59 (2007) 1583. 13 (2013) 2172.
[53] G. Wu, K.Y. Lee, Langmuir 25 (2009) 2133. [60] S. Samarajeewa, R. Shrestha, M. Elsabahy, A. Karwa, A. Li, R.P. Zentay, J.G.
[54] B.K. Kang, J.S. Lee, S.K. Chon, S.Y. Jeong, S.H. Yuk, G. Khang, H.B. Lee, S.H. Cho, Kostelc, R.B. Dorshow, K.L. Wooley, Mol. Pharm. 10 (2013) 1092.
Int. J. Pharm. 274 (2004) 65. [61] F. Zhang, S. Khan, R. Li, J.A. Smolen, S. Zhang, G. Zhu, L. Su, A.A. Jahnke, M.
[55] G. Suresh, K. Manjunath, V. Venkateswarlu, V. Satyanarayana, AAPS Elsabahy, X. Chen, et al., Nanoscale 9 (2017) 15773.
PharmSciTech 8 (2007) 24.

You might also like