Está en la página 1de 13

Journal of Liposome Research

ISSN: 0898-2104 (Print) 1532-2394 (Online) Journal homepage: http://www.tandfonline.com/loi/ilpr20

Preparation and optimization of tablets containing


a self-nano-emulsifying drug delivery system
loaded with rosuvastatin

Heba F. Salem, Rasha M. Kharshoum, Abdel Khalek A. Halawa & Demiana M.


Naguib

To cite this article: Heba F. Salem, Rasha M. Kharshoum, Abdel Khalek A. Halawa & Demiana
M. Naguib (2018) Preparation and optimization of tablets containing a self-nano-emulsifying drug
delivery system loaded with rosuvastatin, Journal of Liposome Research, 28:2, 149-160, DOI:
10.1080/08982104.2017.1295990

To link to this article: https://doi.org/10.1080/08982104.2017.1295990

Accepted author version posted online: 15


Feb 2017.
Published online: 13 Mar 2017.

Submit your article to this journal

Article views: 77

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=ilpr20
http://informahealthcare.com/lpr
ISSN: 0898-2104 (print), 1532-2394 (electronic)

J Liposome Res, 2018; 28(2): 149–160


! 2017 Informa UK Limited, trading as Taylor & Francis Group. DOI: 10.1080/08982104.2017.1295990

RESEARCH ARTICLE

Preparation and optimization of tablets containing a


self-nano-emulsifying drug delivery system loaded with rosuvastatin
Heba F. Salem1, Rasha M. Kharshoum1, Abdel Khalek A. Halawa2, and Demiana M. Naguib2
1
Department of Pharmaceutics, Faculty of Pharmacy, Beni-Suef University, Beni Suef, Egypt and 2Department of Pharmaceutics, Faculty of
Pharmacy, Nahda University, Beni Suef, Egypt

Abstract Keywords
Background: Rosuvastatin (ROS) calcium is the latest synthetic drug in the statin group that has Nano-silica, pharmacokinetics, pseudo ternary
an anti-hyperlipidemic activity. It is available as tablets, and its poor aqueous solubility, slow diagram, rosuvastatin calcium, self-nano-
dissolution rate and low-absorption extent result in less than 20% bioavailability and about emulsifying system
80% being excreted unchanged in the feces without absorption.
Objective: To utilize nanotechnology to reformulate ROS as a self-nano-emulsifying drug History
delivery system (SNEDDS), and utilizing design optimization to fabricate the SNEDDS as a tablet.
Methods: The solubility of ROS in different oils, surfactants and co-surfactants was tested. Received 30 November 2016
Pseudo-ternary phase diagrams were developed and various SNEDDS formulations were Revised 5 February 2017
prepared and evaluated regarding globule size, self-emulsification, viscosity and transmittance. Accepted 12 February 2017
The optimized system was examined using transmission electron microscopy. The self-
nano-emulsifying tablets were prepared using two types of nano-silica and different
percentages of Avicel as a binder and Ac-Di-Sol as a disintegrant. The prepared tablets were
evaluated for their physicochemical properties. Bioavailability in human volunteers was assessed.
Results: A SNEDDS system was successfully developed with a droplet size range of 15 nm and a
composition of 10% Labrafac, 80% Cremophore RH40 and 10% Propylene glycol. The optimized
tablet formula contained: hydrophilic nano-silica, 3% Ac-Di-Sol and 30% Avicel. The
pharmacokinetic study revealed that the bioavailability was enhanced by more than 2.4-fold
compared with the commercially available tablet.
Conclusions: Tablets containing SNEDDS loaded with ROS represent a promising novel formula
that has higher gastrointestinal absorption and enhanced systemic bioavailability.

Introduction of solvent, co-solvents or surfactant as a solubilizing agent


(Chena et al., 2015a; Filippa & Gasull, 2013), reduction in
Rosuvastatin (ROS) is an anti-hyperlipidemic drug commonly
particle size (Xu & Luo, 2014), microemulsion (Chena et al.,
used to treat hypercholesterolemia that produces a consider-
2015b), self-emulsification (Qureshi et al., 2015), complex-
able dose-dependent reduction in low-density lipoprotein
ation (Chia et al., 2015; He & Ho, 2015; Poon et al., 2005),
cholesterol (Schachter, 2004). It is the latest synthetic drug in
the pro-drug approach (Poon et al., 2005) and solid dispersion
the statin group, which acts as an inhibitor of 3-hydroxy-3-
(He & Ho, 2015; Mohanachandran et al., 2010).
methylglutaryl coenzyme A (HMG-CoA) reductase (the rate-
SNEDDS is an isotropic combination of surfactant, oil,
limiting enzyme in cholesterol biosynthesis) (Seker et al.,
co-surfactant and the drug, that produces oil in water nano-
2015). It was also reported to treat benign prostatic hyper-
emulsion upon mild agitation with the GI fluids. This nano-
plasia, osteoporosis and Alzheimer’s disease (Schachter,
emulsion, containing the solubilized drug, can accordingly be
2004). It is a Class II drug in the Biopharmaceutics
absorbed by the lymphatic system, thus bypassing the hepatic
Classification System that shows low dissolution because of
first-pass metabolism (Kohli et al., 2010). SNEDDS was
its crystalline nature and, thus, poor oral bioavailability of
selected as to enhance the dissolution of ROS over the other
20%. It is also extensively metabolized by the liver (Fritz
solubilization techniques, as SNEDDS requires very simple
et al., 2006; Schachter, 2004).
and economical manufacturing facilities, such as simple
Improvement of the dissolution characteristics of poorly
mixer (Date et al., 2010).
water soluble drugs has been achieved using different
The liquid SNEDDS (L-SNEDDS) have various limita-
techniques, such as salt formation (Ewing et al., 2015), use
tions: low-drug stability, interaction between capsule shell
and drug precipitation (Qureshi et al., 2015). Conversion of
Address for correspondence: Demiana M. Naguib, Department of L-SNEDDS to solid SNEDDS using a suitable adsorbent has
Pharmaceutics, Faculty of Pharmacy, Nahda University, Beni Suef,
Egypt. Tel: +00 201289241024. E-mail: demianamonier@gmail.com been commonly employed to overcome the problems related
150 H. F. Salem et al. J Liposome Res, 2018; 28(2): 149–160

to the L-SNEDDS in addition to its ability to maintain all of Table 1. Composition of the prepared systems.
the benefits of L-SNEDDS (Jain et al., 2014; Seo et al., 2013).
System Surfactant Oil Co-surfactant
These adsorbents are hydrophobic and hydrophilic solid
carriers, such as colloidal silica, microcrystalline cellulose System I Tween 80 Labrafil Propylene glycol
(Avicel), Sylysia (350, 550 and 750), magnesium trisilicate System II Cremophore RH40 Labrafil Propylene glycol
System III Tween 80 Labrafac Propylene glycol
and NeusilinÕ US2, etc. (Seo et al., 2013). Different System IV Cremophore RH40 Labrafac Propylene glycol
techniques are used to convert them into powders, pellets or System V Cremophore RH40 Oleic acid Propylene glycol
other solid dosage forms, which could be filled as capsules or System VI Tween 80 Oleic acid Propylene glycol
compressed into tablets (e.g. granulation, freeze drying,
extrusion-spheronization and spray drying, etc.) (Date et al., to form six different systems (Table 1) with each system
2010). containing 36 different ratios ranging from 10 to 80% for each
This study is intended to design a tablet composed of vehicle. For any ratio, the total of the three vehicles always
SNEDDS loaded with ROS for enhancement of its solubility amounted to 100%.
and bioavailability. Each ratio (1 ml) was prepared and mixed using a vortex
mixer to form a homogenous mixture. Each mixture was
Materials and methods diluted with 100 ml distilled water, and mixed using a
magnetic stirrer at 500 rpm for 2 min. The prepared emulsions
Materials
were examined visually for clarity against a dark background,
ROS was a kind gift from Eipico, Egypt. Tween 80 and the clarity was confirmed by measuring the percentage of
(Polyoxyethylene sorbitan monooleate, HLB ¼ 15), transmittance at 638.2 nm using distilled water as the blank.
Cremophore RH 40 (Polyoxyethylene 40 hydrogenated The obtained emulsions were nominated as clear or turbid and
castor oil, HLB, 14–16), Microcrystalline cellulose (AvicelÕ the systems with large nano-emulsion regions were selected
PH 102), Lactose monohydrate, Ac-Di-SolÕ (croscarmellose (Kommuru et al., 2001). The prepared emulsions were
sodium) and propylene glycol were a kind gift from considered nano-emulsions with droplet sizes of 200 nm or
Arabcomide, Egypt. Labrafil M 1944 CS (Oleoyl macro- smaller only when they are clear (Zhang et al., 2008). Pseudo-
golglycerides), Labrafac lipophile WL 1349 (Medium chain ternary phase diagrams were plotted for each system using the
triglycerides) were a kind gift from Gattefosse, France. ProSim software (Labège, France) and the system that
Soybean oil and oleic acid (Octadecenoic acid) were contained wider nano-emulsion region was selected for
purchased from Lab Chemicals Trading Co., Egypt. optimization.
Hydrophilic and hydrophobic nano-silica were purchased
from Nano Tech Co., Egypt. Methanol and Acetonitrile Optimization of the selected system
(HPLC grade) were purchased from Romil, London, UK.
Saturated solubility study in liquid SNEDDS (L-SNEDDS)
Triethylamine was purchased from Adwic Co., Cairo, Egypt.
Formic acid (HPLC grade) was purchased from Sigma Excess amount of the drug was added to 1 ml of each of the
chemicals, St. Louis, MO. clear ratios (L-SNEDDS) in the selected system (Cremophore
RH40/Labrafac/Propylene glycol) and mixed using a vortex
mixer, moved to a thermodynamic water bath shaker for 72 h
Methods
at room temperature and then centrifuged at 5000 rpm for
Solubility studies 20 min. The supernatant was diluted with phosphate buffer at
The solubility of ROS in different vehicles, such as oils a pH 6.8 using a magnetic stirrer at 500 rpm for 2 min at room
(Labrafil M 1944 CS, Labrafac lipophile WL 1349, Oleic acid temperature (25 ± 1  C) (Beg et al., 2012). The amount of
and Soybean oil), surfactants (Cremophore RH40, Tween 80, dissolved drug was determined spectrophotometrically at max
Cremophore EL, Span 80 and Span20) and co-surfactants 241 nm.
(Propylene glycol, Ethanol and polyethylene glycol (PG)) was
assessed by addition of an excess amount of ROS (300 mg) to Globule size using Malvern Zetasizer. SNEDDS were diluted
1 ml of each vehicle in screw-capped glass vials and mixing with deionized water and subjected to particle size examin-
using a vortex mixer. The mixtures were moved to a ation. Average particle size and poly dispersity index (PDI)
thermodynamic water bath shaker for 72 h at room tempera- were determined using dynamic light scattering on a Malvern
ture and then centrifuged at 5000 rpm for 20 min. The Zetasizer (Goddeeris et al., 2006; Hassan et al., 2014; Panda
supernatant was diluted with methanol using magnetic stirrer et al., 2001).
at 500 rpm for 2 min at room temperature (25 ± 1  C). The
Viscosity measurement
amount of dissolved drug was determined at max 243 nm
using UV spectrophotometer (Shimadzu, UV-1800 PC, Viscosity of 1 ml of ROS loaded SNEDDS was measured
Japan), the test was repeated in triplicate (Beg et al., 2012). using a Brookfield viscometer without dilution (Gupta et al.,
2011).
Construction of pseudo ternary diagram
Self-emulsification time & optical clarity measurement
Based on the solubility studies, the selected vehicles were:
surfactants (Tween 80 and Cremophore RH 40), oils (Oleic Determination of self-emulsification time was carried out by
acid, Labrafac, Labrafil) and co-surfactant (propylene glycol), dilution of 0.5 ml of SEDDS in a glass beaker with 100 ml
DOI: 10.1080/08982104.2017.1295990 Preparation and optimization of tablets 151

distilled water and mixing gently using a magnetic stirrer at (S-4100, Hitachi, Japan). The powders were placed in a
room temperature with a rotation speed of 100 rpm. The time brass specimen holder using two-sided adhesive tape and
taken by the SEDDS to form a homogenous nano-emulsion coated with platinum in a vacuum evaporator (6 Pa) prior to
was measured. The clarity of the nano-emulsions was investigation.
observed visually and confirmed by measurement of the
percentage of transmittance at 638.2 nm, using distilled water Formulation of SNEDD tablets
as the blank on a UV–Vis spectrophotometer. As the Nine formulations were prepared by direct compression
percentage transmittance comes closer to 100%, it indicates technique using different percentages of Avicel as a binder
that the formed systems are clear and the size of the prepared and Ac-Di-Sol as a disintegrant, as shown in Table 2. The
globules is in the nanometric range (Obitte et al., 2013). components of each formula were mixed to obtain free
flowing powder by geometrical dilution method and the
Development of liquid ROS-SNEDDS
obtained powder mixture was filled manually and compressed
ROS (10 mg) added to the optimized SNEDDS (0.1 ml) in a under a fixed pressure to obtain a 500-mg tablet.
glass vial was mixed using a sonicator for 20 min until the
drug was dissolved and subjected to surface morphology Evaluation of the prepared SNEDDS tablets
determination (Elnaggar et al., 2009). Flow properties
Surface morphology determination of the liquid The flow properties of the powdered mixtures determined
ROS-SNEDDS using the Carr’s index and the Hausner ratio were evaluated
by measuring the bulk and tapped densities of the powdered
Transmission electron microscopy (TEM) was used to study
mixtures as explained (Carr, 1965).
the morphology of the prepared liquid ROS-SNEDDS (Beg
et al., 2012; Elnaggar et al., 2009; Zhao et al., 2010). The L-
Tablet friability. The friability of the tablets was measured
SNEDDS samples were diluted 100 times with water to form
using the Erweka friabilator (Germany) (Frederick Osei-
a nano-emulsion. In the TEM technique, a drop of the formed
Yeboah CCS, 2015).
nano-emulsion was applied to a copper grid and left for
30 min to allow the particles to bind to collodion and the Drug content
excess was removed. A drop of 2% uranyl acetate solution was
applied. The excess was removed and the sample was air dried The content uniformity in the prepared tablets was assessed
for 1 min and observed under a transmission electron by milling the tablets. The powder was diluted with methanol,
microscope (Jeol, 1200 EXII, Tokyo, Japan) (Abdelrahman with the use of a sonicator to dissolve the drug. The solutions
et al., 2015; Khallaf et al., 2016). obtained were filtered, and the supernatants were measured
spectrophotometrically for the drug content at max 243 nm
Development of solid SNEDDS (Ali et al., 2015).
The prepared liquid ROS-SNEDDS was solidified by adsorp- Tablet hardness
tion on two types of nano-silica (hydrophilic and lipophilic
nano-silica). The adsorption capacity of each type of nano- The crushing strength of the tablets was calculated using a
silica was determined by dropwise addition of a fixed amount hardness tester and the mean hardness was expressed as kg
of liquid ROS-SNEDDS to different weights of nano-silica in (±SD) (Elbary et al., 2015).
a small glass dish and was mixed well. The weight of each
type of silica that formed free flowing self-nano-emulsifying Drug release
granules (SNEGs) was determined (Weerapol et al., 2014). The in-vitro study of dissolution of ROS from the prepared
SNEDDS tablets was carried out in 500 ml of phosphate
Morphological analysis of solid SNEDDS buffer (pH 6.8) using a USP Dissolution Tester with a paddle
The outer morphologic structures of ROS powder and solid speed of 100 rpm at 37  C. Samples were withdrawn at 5, 10,
SNEDDS of both hydrophilic and lipophilic nano-silica were 15, 30, 45, 60, 90 and 120 min. The samples were then
observed using a scanning electron microscope (SEM) analyzed spectrophotometrically for ROS content by

Table 2. The composition of SNEDDS tablets.

Formula no.
Tablet composition F1 F2 F3 F4 F5 F6 F7 F8
ROS-loaded SNEDDS (mg/0.1 ml) 10 10 10 10 10 10 10 10
Hydrophilic nano-silica (mg) 150 150 150 150 – – – –
Lipophilic nano-silica (mg) – – – – 130 130 130 130
Avicel PH-102 (%) 30 30 40 40 30 30 40 40
Ac-Di-Sol (%) 1 3 1 3 1 3 1 3

Lactose monohydrate was used as filler to bring the total weight to 500 mg.
Magnesium stearate (1%) was added to each formula as lubricant.
152 H. F. Salem et al. J Liposome Res, 2018; 28(2): 149–160

measuring their absorbance at max 241 nm, and the percent solubilize marked amounts of the drug, as it controls the
of cumulative drug released was determined. drug loading efficiency (Sallam & Boscá, 2015). Among
the tested vehicles, oleic acid, Labrafac and Labrafil were
Bioavailability study selected as the oil phase, Cremophore RH40 and Tween 80
This in-vivo study was carried out in healthy male volunteers, were selected as surfactants and propylene glycol was
to compare the ROS pharmacokinetics of the optimized SNE- selected as co-surfactant as shown in Figure 1. The
tablet (F2) with the commercially available tablet CrestorÕ mentioned vehicles were selected for further investigation
(10 mg of AstraZeneca Pty Ltd.). The human volunteers were of their nano-emulsification ability for final selection of the
divided into two groups (n ¼ 6) with average age of SNEDDS.
32 ± 5 years, average height of 173.5 ± 5.5 cm and average
Construction of pseudo ternary diagram
body weight of 71.5 ± 6.3 kg. The human in-vivo study was
approved by the Ethics Committee of the Bani-Suef Pseudo ternary phase diagram was constructed to identify the
University, and the protocol complied with the Declaration ability of different oils, surfactants and co-surfactants to form
of Helsinki (Declaration of Helsinki, 2000). a clear nano-emulsion, and also to identify the range of the
The volunteers were instructed to refrain from smoking or nano-emulsion region. The result indicated that Labrafac and
taking medicines for 7 d before the study and were fasted for Labrafil have better self-nano-emulsification ability with both
at least 10 h before treatment was administered. Two formu- surfactants than oleic acid, which did not form any clear
lations, including the marketed Crestor tablet and the emulsions; only translucent and turbid emulsions were
optimized SNEDDS tablet were administered orally with formed. Oils and triglycerides of medium length carbon
200 ml of water for each group of volunteers separately at a chain are more favored to form nano-emulsions than those of
dose of 10 mg each (FDA, 2002). The volunteers’ safety and longer carbon chain (e.g. oleic acid); and our results were
sample collection during the trial were under physician similar to earlier reported observations (Beg et al., 2012).
supervision and any spontaneously noticed adverse events Systems that showed visual clarity on dilution with distilled
were recorded and evaluated. Venous blood samples (5 ml) water are shown in Figures 2–5.
were collected at the predetermined time points (0, 0.5, 1, 1.5, The surfactants used (Tween 80 and Cremophore RH 40)
2, 2.5, 3, 4, 6, 8, 24 and 36 h) into heparinized tubes. are non-ionic hydrophilic surfactants and were selected due to
Separation of plasma was carried out by centrifugation of the lower oral toxicity. They were also reported to have some
blood at 5000 rpm for 10 min at 4  C. The plasma was stored biological activity: Tween 80 has lymphotropic character and
at 20  C until drug analysis. Cremophore RH40 has an inhibitory effect on p-gp and CYP
A previously reported validated and sensitive HPLC enzymes (Elnaggar et al., 2009). Surfactants of high-hydro-
method for determination of ROS in plasma was adopted philic lipophilic balance (HLB) that produce oil in water
(Kumar et al., 2006; Trivedi et al., 2005). The mobile phase nano-emulsions also enhance drug solubility, preventing its
consisting of methanol: formic acid (0.1 M): acetonitrile in a precipitation when dispersed in the GI fluids. HLB have an
ratio of 1:3:6 was employed at a flow rate of 0.8 ml/min and effect on the globule size of the emulsion, as the globule size
injection volume of 20 ml. The detection wavelength was decreases with the increase in the HLB of the surfactant
241 nm and the internal standard used was Atorvastatin (Weerapol et al., 2014). Tween 80 and Cremophore RH 40
(ATOR). All assays were performed at ambient conditions have nearly similar high-HLB values, i.e. HLB 15 and HLB
(Krishnamoorthy et al., 2013). 14–16, respectively. The choice of surfactant was according to
Pharmacokinetic data of ROS were measured using its capacity to form nano-emulsion with the selected oil and to
WinNonlinÕ (version 1.5; Scientific Consulting Inc., Cary, solubilize ROS (Beg et al., 2012). It was observed that
NC). After oral administration, the maximum ROS concen- systems I and III can form clear emulsions up to 30% v/v of
tration in human plasma (Cmax) and the time required for both oils, while systems II and IV formed clear emulsions up
reaching Cmax (Tmax) were computed from the obtained to 40% v/v of both oils, which indicates slightly higher
plasma concentration/time curves. The area under the curve capacity of Cremophore RH40 to form nano-emulsions with
was extrapolated from zero to infinity (AUC0–1) and to 24 h the selected oils than Tween 80, as reported by (Elnaggar
(AUC0–24); they were obtained using the linear trapezoidal et al., 2009). System IV, which consisted of Cremophore
rule. The formula t1/2 ¼ 0.693/Kel was used to calculate the RH40, Labrafac and propylene glycol as surfactant, oil and
half-life (t1/2). The SPSS version 16.0 software (SPSS Inc., co-surfactant, respectively, was selected due to the wider
Chicago, IL) was used for the statistical analysis of the data. nano-emulsion region.
The results were expressed as the mean values of six
volunteers ± SD (Yamaoka et al., 1978). Optimization of the selected system
Saturated solubility study in liquid SNEDDSs (L-SNEDDSs)
Results and discussion
The goal was to obtain clear nano-emulsions, which have
Solubility studies
optimum drug loading with the maximum solubilizing effect;
Solubility studies were performed to select suitable oils, hence, saturated solubility of the drug in L-SNEDDSs was
surfactants and co-surfactants for the development of tested. System IV showed high-solubilizing effect for the drug
SNEDDS with high-solubilizing capacity for the drug. The up to 253.52 ± 5.12 mg/ml which could solubilize a single
most vital element in nano-emulsion is the oil; it should dose (10 mg) of ROS and reduce the potential for drug
DOI: 10.1080/08982104.2017.1295990 Preparation and optimization of tablets 153

Figure 1. Solubility studies of ROS in each oil, surfactant and co-surfactant.

Figure 2. Pseudo ternary phase diagram for system I. Figure 4. Pseudo ternary phase diagram for system III.

Figure 5. Pseudo ternary phase diagram for system IV.

Figure 3. Pseudo ternary phase diagram for system II.

Globule size using Malvern Zetasizer. It was observed that the


precipitation. The results of saturated solubility test of ROS z-average of the formed nano-emulsions decreased with the
are shown in Table 3. We noticed that the higher the increase in the surfactant concentration. Nano-emulsions of
percentage of Cremophore RH40 in the L-SEDDS, the higher the smallest globule size have rapid absorption and hence,
the solubility of the drug. improved bioavailability (39). The smallest size with the best
154 H. F. Salem et al. J Liposome Res, 2018; 28(2): 149–160

Table 3. Optimization studies of the selected system.

SEDDS no. (S:O:Co-S) Drug solubility in (mg/ml) Particle size (nm) PDI Transmittance (%) Emulsificationtime (Sec) Viscosity (Cp)
1 3:1:6 91.25 ± 1.56 23.8 0.353 100 19 168.44
2 4:1:5 121.41 ± 2.34 21.2 0.386 99.77 22 183.62
3 4:2:4 119.73 ± 2.62 62.0 0.527 98.40 155 181.51
4 5:1:4 153.59 ± 1.81 17.6 0.127 100 36 194.46
5 5:2:3 148.27 ± 2.73 25.1 0.239 99.77 67 190.17
6 5:3:2 144.35 ± 4.31 68.4 0.353 97.95 183 188.36
7 5:4:1 139.81 ± 3.12 106.7 0.533 90.78 191 186.24
8 6:1:3 184.22 ± 2.15 15.9 0.177 100 54 215.62
9 6:2:2 176.45 ± 3.71 25.8 0.160 100 107 211.43
10 6:3:1 171.26 ± 2.31 20.9 0.291 99.77 145 209.84
11 7:1:2 218.74 ± 4.21 13.506 0.253 100 83 224.17
12 7:2:1 211.61 ± 3.15 18.1 0.141 100 101 222.51
13 8:1:1 253.52 ± 5.12 15.2 0.022 100 92 247.33

Figure 6. (a,b) Intensity size distribution curves for the selected system.
DOI: 10.1080/08982104.2017.1295990 Preparation and optimization of tablets 155

Figure 6. Continued.

PDI was obtained using SNEDDS no. 13; (10% oil, 80% from 168.44 to 247.33 cp. The viscosity was increased with
surfactant and 10% co-surfactant) as showed in Figure 6 (a,b) the decrease in oil proportion and the increase in surfactant,
and Table 3; however, higher surfactant concentration may which resulted in an obvious increase in the emulsification
lead to gastric irritation (Lawrence & Rees, 2000). Therefore, time of SNEDDS.
the least amount of SNEDDS (0.1 ml) was used to solubilize
the ROS dose (10 mg). Self-emulsification time & optical clarity measurement
Spontaneous emulsification was suggested to occur when
Viscosity measurement
water quickly penetrates the gel phases formed on the surface
The geometry and shape of SNEDDS could be attributed to of the droplet by the gentle agitation of the peristaltic activity
the viscosity results (Shakeel et al., 2013). The viscosity of of GIT (Pouton, 1997; Rang & Miller, 1999). Self-emulsifi-
selected SNEDDS (Chena et al., 2015a,b; Chia et al., 2015; cation time was increased by the increase in the concentra-
Ewing et al., 2015; Filippa & Gasull, 2013; Fritz et al., 2006; tions of oil and surfactant and decreased by the increase in the
He & Ho, 2015; Poon et al., 2005; Qureshi et al., 2015; co-surfactant concentration. These observations may be due
Schachter, 2004; Seker et al., 2015; Xu & Luo, 2014) ranged to the increase in the viscosity and the free surface energy of
156 H. F. Salem et al. J Liposome Res, 2018; 28(2): 149–160

system by the surfactant, while the increase in oil concentra-


tion affected the emulsification time, as it let the system take
more time to disperse in water (Parmar et al., 2011).
The percentage of transmittance of all the tested systems
was above 90%, which point out the clarity of the formed
emulsions and indicate the formation of emulsions with
globule sizes within the nano-range.
Based on the previous results, SNEDDS no (Poon et al.,
2005) was used for the formulation of tablets and further
investigation. SNEDDS no (Poon et al., 2005) showed the
maximum solubilizing effect for ROS (253.52 ± 5.12 mg/ml)
with particle size of 15.2 nm and PDI of 0.022.

Development of liquid ROS-SNEDDS


Surface morphology determination of the liquid
ROS-SNEDDS
The morphology of the formed nano-emulsion droplets was
observed as dark sphere-shaped droplets, with each droplet
being surrounded by a thick bright frame (Figure 7). We can Figure 7. TEM of the optimized liquid ROS-SNEDDS (magnification
conclude that the thick frame acts as a barrier to coalescence 50 000).
and reduces the interfacial energy, which indicates the

Figure 8. Scanning electron micrographs of (A) & (B) ROS powder, (C) & (D) solid SNEDDS prepared with hydrophilic nano-silica, (E) & (F) solid
SNEDDS prepared with lipophilic nano-silica at two different magnifications.
DOI: 10.1080/08982104.2017.1295990 Preparation and optimization of tablets 157
Table 4. Physical properties of the prepared ROS-SNE powders.

Formula Angle of repose Hausner ratio Carr’s index Friability (%) Hardness (kg/cm2) Drug content (%)
F1 33.64 1.13 11.11 0.88 3.93 106.32 ± 2.21
F2 34.78 1.19 16.00 0.75 4.92 102.55 ± 0.73
F3 36.15 1.47 32.14 0.82 4.74 101.71 ± 1.12
F4 35.87 1.25 20.00 0.71 5.22 97.37 ± 3.11
F5 35.13 1.24 19.51 0.54 4.17 99.62 ± 1.44
F6 34.51 1.18 15.00 0.52 3.76 100.55 ± 1.58
F7 34.16 1.25 20.00 0.78 6.32 95.71 ± 1.83
F8 31.48 1.36 26.67 0.63 5.43 98.25 ± 1.43

physical stability of the formed nano-emulsion (Parmar et al., comply with the limits of British pharmacopeia, 2013. All
2011). formulae showed percentage of friability of51% and hardness
between 3.76 and 6.32 kg/cm2.
Development of solid SNEDDS
The adsorption capacity of two types of nano-silica (hydro- Drug content uniformity
philic and lipophilic nano-silica) was determined by the weight ROS content ranged from 106.32 ± 2.21 to 95.71 ± 1.83
of each type of silica to adsorb 0.1 ml of ROS-SNEDDS. It was (Table 3). The results were found to comply with the limits
observed that the lipophilic nano-silica showed higher adsorp- of British pharmacopeia, 2013 (85–115%).
tion capacity than hydrophilic nano-silica (130 mg/0.1 ml and
150 mg/0.1 ml, respectively). These results agree with those of Drug release
Kang JH, et al. (Kanga et al., 2012), which suggests the higher The release profiles of the prepared SNE-tablets and L-
adsorption capacity of lipophilic carriers than that of hydro- SNEDDS compared with conventional Crestor tabletsÕ are
phobic carriers for the SNEDDS. shown in Figure 9. The SNE-tablet (F1–F4) prepared with
hydrophilic nano-silica showed higher drug release than the
Morphological analysis of solid SNEDDS
SNEDD tablet (F5–F8) prepared with lipophilic nano-silica.
The scanning electron micrographs (SEM) of ROS powder The drug release results may be attributed to the ease of
and solid SNEDDS of both hydrophilic and lipophilic nano- penetration of the aqueous gastric solutions into the hydro-
silica are shown in Figure 8. ROS powder (Figure 8A) was philic nano-silica than the hydrophilic nano-silica followed by
shaped like irregular crystals (Zhou et al., 2013). The SEM of facilitated self-nano-emulsification (El-Say et al., 2010).
solid SNEDDS prepared with both hydrophilic and lipophilic Additionally, the higher absorption of L-SNEDDS on the
nano-silica (Figures 8B and 7C) show rough granular outer surface of hydrophilic nano-silica, compared with the lipo-
surface for both, but the surface of the solid SNEDDS philic nano-silica, facilitates its release. L-SNEDDS showed
prepared with hydrophilic nano-silica was rougher with a high ROS release (100.19 ± 0.65%) while SNE-tablet F2
more aggregated granular surface. showed the highest ROS release (97.05 + 1.51%).
The results indicate that the L-SNEDDS was absorbed SNE-tablets F2 showed the highest ROS release with good
inside the pores of both types of nano-silica (and) or coated compressibility and friability and, therefore, were used in the
the surface (Kanga et al., 2012). As the lipophilic nano-silica bioavailability studies.
shows higher porosity than that of the hydrophilic nano-silica
(Sarawade et al., 2011), its absorption of the L-SNEDDS Bioavailability
inside the pores is higher than the surface, while the The plasma concentration-time profiles for ROS-SNE tablet
hydrophilic nano-silica shows more aggregated surface with (F2) and Crestor following oral administration are presented
L-SNEDDS than the lipophilic nano-silica. These observa- in Figure 10. The pharmacokinetic parameters of ROS are
tions explain the rougher surface of the hydrophilic nano- tabulated in Table 5. The results demonstrated that the
silica and concur with the previously mentioned results of the AUC(0–1) of ROS in SNE tablet increased 2.45 times and
adsorption capacity. Cmax increased 2.78 times when compared with Crestor.
Similarly, tmax also decreased for SNE tablet (2 ± 0.216 h)
Evaluation of the prepared SNEDDS tablets compared with Crestor (3 ± 0.347 h), which is consistent with
Flow properties the differences in ROS dissolution and absorption in both
tablets. The results of all of the pharmacokinetic data were
The flow properties of the powdered mixtures are considered found to be highly significant (p50.05) for SNE tablet
good, when the Hausner ratio is below 1.25, Carr’s index is compared with Crestor indicating that oral absorption of ROS
lower than 20% and the angle of repose is between 25 and 40 was significantly increased by SNEDDS. Another pharmaco-
(Abdelbary et al., 2012; El-Say et al., 2010). F1, F2, F5 and kinetic study in male Wistar rats by Dudhipala and
F6 showed reasonable flow properties (Table 4). Veerabrahma; showed improvement in the oral bioavailability
of solid lipid nanoparticles by 4.6-fold when compared to that
Tablet friability & hardness
of ROS suspension (Dudhipala & Veerabrahma, 2017). From
The results of the prepared ROS-SNE tablets regarding the bioavailability results, it may be predicted that after oral
hardness and the percentage of friability were found to administration of ROS-SNE tablet, a nano-emulsion will form
158 H. F. Salem et al. J Liposome Res, 2018; 28(2): 149–160

Figure 9. Dissolution of ROS from L-SNEDDS and SNE-tablets compared with CrestorÕ tablet (pH6.8).

Table 5. Pharmacokinetic parameters and relative bioavailability of SNE


tablet and Crestor (mean ± SD, n ¼ 6).

Parameters CrestorÕ SNE tablet


AUC0–36 (ng h/ml) 241 747 ± 22.041 591 406 ± 68.235
Cmax (ng/ml) 23 885 ± 3.124 66 521 ± 2.824
Tmax (h) 3 ± 0.347 2 ± 0.216
t1/2 11 351 ± 1.932 11 497 ± 1.872
AUC0–1 264 210 ± 25.311 648 219 ± 73.219
Relative BA% based on AUC(0–1) _ 245%
MRT(0–1) (h) 13 916 ± 0.615 13 664 ± 0.416

solid SNEDDS, this aim was obtained by converting the


selected L-SNEDDS into tablets using two types of nano-
Figure 10. Plasma concentration profiles of ROS after oral administra-
Õ
tion of SNE tablet and Crestor to healthy human volunteers silica as carriers. The use of hydrophilic nano silica as carrier
(mean ± SD, n ¼ 6). increased the loading of L-SNEDDS, and enhanced the
release of ROS than the lipophilic nano-silica. The in-vitro
spontaneously presenting the active components in a dissolution study showed that both L-SNEDDS and F2
solubilized state. The formed nano-emulsion will have showed the maximum drug release with no significant
droplets with small size that provide a large surface area for difference. After in-vivo study, the enhancement in bioavail-
extensive ROS absorption (Cho et al., 2013). The enhance- ability was more than 2.45 fold indicating that; F2 is
ment in bioavailability of more than 2.45-fold could be considered a promising novel formula that has higher
attributed to the size of ROS-NE, which have the advantages gastrointestinal absorption and improved systemic
of nanoparticles, and improve the adhesion to and absorption bioavailability.
into the intestinal epithelial cells (Xia et al., 2006). In
addition, the nano-emulsion vesicles may promote uptake by
the M cells in the Peyer’s patches and increase absorption Declaration of interest
through the lymphatic pathway (Guo et al., 2001; Khan et al., The authors report no declaration of interests.
2013). Permeation of intact NE through the intestinal
epithelia is considered as an additional possible mechanism
for the enhancement in bioavailability of the prepared ROS- References
NE formula (Chen et al., 2009). Abdelbary G, Amin M, Salah S. (2012). Self nano-emulsifying
simvastatin based tablets: design and in vitro/in vivo evaluation.
Pharm Dev Technol 18:1294–304.
Conclusions Abdelrahman AA, Salem HF, Khallaf RA, Ali AMA. (2015). Modeling,
optimization, and in vitro corneal permeation of chitosan lomeflox-
This research aimed to enhance the dissolution and the acin HCl nanosuspension intended for ophthalmic delivery. J Pharm
bioavailability of ROS by using an optimum formulation of Innov 10:254–68.
DOI: 10.1080/08982104.2017.1295990 Preparation and optimization of tablets 159
Ali MFM, Salem HF, Abdelmohsen HF, Attia SK. (2015). Preparation part I. Formulation development, statistical optimization, and in vitro
and clinical evaluation of nano-transferosomes for treatment of characterization. Pharm Res 31:923–45.
erectile dysfunction. Drug Des Devel Ther 9:2431–47. Jill BS, Sachin JP. (2010). Formulation and evaluation of self micro
Beg S, Swain S, Singh HP, et al. (2012). Development, optimization, and emulsifyingsystem of Candesartan cilexetil. Int J Pharm Pharm Sci 2:
characterization of solid self-nanoemulsifying drug delivery systems 143–6.
of valsartan using porous carriers. AAPS PharmSciTech 13:1416–27. Kanga JH, Oha DH, Ohc YK, et al. (2012). Effects of solid carriers on
Carr RL. (1965). Evaluating flow properties of solids. Chem Eng 18: the crystalline properties, dissolution and bioavailability of flurbipro-
163–8. fen in solid self-nanoemulsifying drug delivery system (solid
Chen Y, Lu Y, Chen J, et al. (2009). Enhanced bioavailability of the SNEDDS). Eur J Pharm Biopharm 80:289–97.
poorly water-soluble drug fenofibrate by using liposomes containing a Khallaf RA, Salem HF, Abdelbary A. (2016). 5-Fluorouracil shell-
bile salt. Int J Pharm 376:153–60. enriched solid lipid nanoparticles (SLN) for effective skin carcinoma
Chena X, Faddaa HM, Aburubd A, et al. (2015a). Cosolvency approach treatment. Drug Deliv 23:3452–60.
for assessing the solubility of drugs in poly(vinylpyrrolidone). Int J Khan AA, Mudassir J, Mohtar N, Darwis Y. (2013). Advanced drug
Pharm 494:346–56. delivery to the lymphatic system: lipid-based nanoformulations. Int J
Chena Y, Tuoa J, Huanga H, et al. (2015b). Optimized mixed oils Nanomedicine 8:2733–44.
remarkably reduce the amount of surfactants in microemulsions Kohli K, Chopra S, Dhar D, et al. (2010). Self-emulsifying drug delivery
without affecting oral bioavailability of ibuprofen by simultaneously systems: an approach to enhance oral bioavailability. Drug Discov
enlarging microemulsion areas and enhancing drug solubility. Int J Today 15:958–65.
Pharm 487:17–24. Kommuru TR, Gurley B, Khan MA, Reddy IK. (2001). Self-emulsifying
Chia L, Liua R, Guoa T, et al. (2015). Dramatic improvement of the drug delivery systems (SEDDS) of coenzyme Q 10 : formulation
solubility of pseudolaric acid B by cyclodextrin complexation: development and bioavailability assessment. Int J pharm 212:233–46.
preparation, characterization and validation. Int J Pharm 479:349–56. Krishnamoorthy B, Vijaya CR, Selvan Natarajan T, et al. (2013). Self
Cho W, Kim MS, Kim JS, et al. (2013). Optimized formulation of solid nanoemulsifying drug delivery system (SNEDDS) of rosuvastatin
self-microemulsifying sirolimus delivery systems. Int J Nanomedicine calcium: design, formulation, bioavailability and pharmacokinetic
8:1673–82. evaluation. Colloids Surf B Biointerfaces 112:337–43.
Date AA, Dixit R, Nagarsenker M. (2010). Self-nanoemulsifying drug Kumar TR1, Shitut NR, Kumar PK, et al. (2006). Determination of
delivery systems?: formulation insights, applications and advances. rosuvastatin in rat plasma by HPLC: validation and its application to
Nanomedicine (Lond) 5:1595–616. pharmacokinetic studies. Biomed Chromatogr 20:881–7.
Declaration of Helsinki. (2000). As amended by the 52nd World Medical Lawrence MJ, Rees GD. (2000). Microemulsion-based media as novel
Assembly (WMA). Edinburgh, Scotland: World Medical Association. drug delivery systems. Adv Drug Deliv Rev 45:89–121.
Dudhipala N, Veerabrahma K. (2017). Improved anti-hyperlipidemic Mohanachandran PS, Sindhumol PG, Kiran TS. (2010). Enhancement of
activity of rosuvastatin calcium via lipid nanoparticles: pharmacoki- solubility and dissolution rate: an overview. Pharmacie Globale Int J
netic and pharmacodynamic evaluation. Eur J Pharm Biopharm 110: Compr Pharm 1:1–10.
47–57. Obitte NC, Rohan LC, Adeyeye CM, et al. (2013). The utility of self-
Elbary AA, Salem HF, Ali AMA, Maher EM. (2015). Formulation and emulsifying oil formulation to improve the poor solubility of the anti
in-vitro evaluation of fast dissolving tablets containing a poorly HIV drug CSIC. AIDS Res Ther 10:14–19.
soluble antipsychotic drug. Int J Drug Delivery 7:113–125. Panda AK, Moulik SP, Bhowmik BB, Das AR. (2001). Dispersed
Elnaggar YSR, El-Massik MA, Abdallah OY, et al. (2009). Self- molecular aggregates: II. synthesis and characterization of nanopar-
nanoemulsifying drug delivery systems of tamoxifen citrate: design ticles of tungstic acid in H2O/(TX-100 + alkanol)/n-heptane W/O
and optimization. Int J Pharm 380:133–41. microemulsion media. J Colloid Interface Sci 235:218–26.
El-Say KM, Samy AM, Fetouh MI. (2010). Formulation and evaluation Parmar N, Singla N, Amin S, et al. (2011). Study of cosurfactant effect
of rofecoxib liquisolid tablets. Int J Pharm Sci Rev Res 3:135–42. on nanoemulsifying area and development of lercanidipine loaded
Ewing AV, Wray PS, Clarke GS, Kazarian SG. (2015). Evaluating drug (SNEDDS) self nanoemulsifying drug delivery system. Colloids Surf
delivery with salt formation: drug disproportionation studied in situ by B Biointerfaces 86:327–38.
ATR-FTIR imaging and Raman mapping. J Pharm Biomed Anal 111: Poon SF, Stock N, Payne JE, et al. (2005). Novel approach to pro-drugs
248–56. of lactones: water soluble imidate and ortho-ester derivatives of a
FDA. (2002). Guidance of industry, Food-effect bioavailability and furanone-based COX-2 selective inhibitor. Bioorg Med Chem Lett 15:
bioequivalence studies. Rockville (MD): Food and Drug 2259–63.
Administration. Pouton CW. (1997). Formulation of self-emulsifying drug delivery
Filippa MA, Gasull EI. (2013). Ibuprofen solubility in pure organic systems. Adv Drug Del Rev 25:47–58.
solvents and aqueous mixtures of cosolvents: interactions and Qureshi MJ, Mallikarjun C, Kian WG. (2015). Enhancement of solubility
thermodynamic parameters relating to the solvation process. Fluid and therapeutic potential of poorly soluble lovastatin by SMEDDS
Phase Equilibria 354:185–90. formulation adsorbed on directly compressed spray dried magnesium
Frederick Osei-Yeboah CCS. (2015). Validation and applications of an aluminometasilicate liquid loadable tablets: a study in diet induced
expedited tablet friability method. Int J Pharm 484:146–55. hyperlipidemic rabbits. Asian J Pharm Sci 10:40–56.
Fritz B, Paul A, Van Der S, M S. (2006). Crystalline forms of Rang MJ, Miller CA. (1999). Spontaneous emulsification of oils
rosuvastatin calcium salt. US Patent, No.20080194604A1. containing hydrocarbon, nonionic surfactant, and oleyl alcohol.
Goddeeris C, Cuppo F, Reynaers H, et al. (2006). Light scattering
J Colloid Interface Sci 209:179–92.
measurements on microemulsions: estimation of droplet sizes. Int J Sallam MA, Boscá MTM. (2015). Optimization, ex vivo permeation, and
Pharm 312:187–95.
stability study of lipid nanocarrier loaded gelatin capsules for
Guo JX, Ping QN, Chen Y. (2001). Pharmacokinetic behavior of
treatment of intermittent claudication. Int J Nanomedicine 10:
cyclosporin A in rabbits by oral administration of lecithin vesicle and
4459–78.
sandimmun neoral. Int J Pharm 216:17–21.
Sarawade PB, Kim JK, Hilonga A, et al. (2011). Synthesis of hydrophilic
Gupta S, Chavhan S, Sawant KK. (2011). Self-nanoemulsifying drug
and hydrophobic xerogels with superior properties using sodium
delivery system for adefovir dipivoxil?: design, characterization, in
silicate. Micropor Mesopor Mat 139:138–47.
vitro and ex vivo evaluation. Colloids Surf A Physicochem Eng Asp
Schachter M. (2004). Chemical, pharmacokinetic and pharmacodynamic
392:145–55.
properties of statins: an update. Fundam Clin Pharmacol 19:117–25.
Hassan TH, Metz H, Mäder K. (2014). Novel semisolid SNEDDS based
Seker FB, Kilic U, Caglayan B, et al. (2015). HMG-CoA reductase
on PEG-30-dipolyhydroxystearate: development and characterization.
inhibitor rosuvastatin improves abnormal brain electrical activity via
Int J Pharm 477:506–18.
mechanisms involving eNOS. Neuroscience 284:349–59.
He Y, Ho C. (2015). Amorphous solid dispersions: utilization and
Seo YG, Kim DH, Ramasamy T, et al. (2013). Development of
challenges in drug discovery and development. J Pharm Sci 104:
docetaxel-loaded solid self-nanoemulsifying drug delivery system
3237–58.
(SNEDDS) for enhanced chemotherapeutic effect. Int J Pharm 452:
Jain AK, Thanki K, Jain S. (2014). Solidified self-nanoemulsifying
formulation for oral delivery of combinatorial therapeutic regimen: 412–20.
160 H. F. Salem et al. J Liposome Res, 2018; 28(2): 149–160

Shakeel F, Haq N, El-Badry M, et al. (2013). Ultra fine super Xu J, Luo KQ. (2014). Enhancing the solubility and bioavailability of
self-nanoemulsifying drug delivery system (SNEDDS) enhanced isoflavone by particle size reduction using a supercritical carbon
solubility and dissolution of indomethacin. J Mol Liquids 180: dioxide-based precipitation process. Chem Eng Res Des 92:2542–9.
89–94. Yamaoka K, Nakagawa T, Uno T. (1978). Application of Akaike’s
Trivedi RK1, Kallem RR, Mullangi R, Srinivas N. (2005). Simultaneous information criterion (AIC) in the evaluation of linear pharmacoki-
determination of rosuvastatin and fenofibric acid in human plasma by netic equations. J Pharmacokinet Biopharm 6:165–75.
LC-MS/MS with electrospray ionization: assay development, valid- Zhang P, Liu Y, Feng N, Xu J. (2008). Preparation and evaluation of self-
ation and application to a clinical study. J Pharm Biomed Anal 39: microemulsifying drug delivery system of oridonin. Int J Pharm 355:
661–9. 269–76.
Weerapol Y, Limmatvapirat S, Nunthanid J, Sriamornsak P. (2014). Self- Zhao Y, Wang C, Chow AHL, et al. (2010). Self-nanoemulsifying drug
nanoemulsifying drug delivery system of nifedipine: impact of delivery system (SNEDDS) for oral delivery of Zedoary essential oil:
hydrophilic-lipophilic balance and molecular structure of mixed formulation and bioavailability studies. Int J Pharm 383:170–7.
surfactants. AAPS PharmSciTech 15:456–64. Zhou C, Gao W, Lu G, et al. (2013). Preparation, characterization and in
Xia S, Xu S, Zhang X. (2006). Optimization in the preparation of vitro release of based on dextran-rosuvastatin conjugate. Carbohydr
coenzyme Q10 nanoliposomes. J Agric Food Chem 54:6358–66. Polym 96:156–62.

También podría gustarte