ArticlePDF Available

Enhancement of oral bioavailability of atorvastatin calcium by self-emulsifying drug delivery systems (SEDDS)

Taylor & Francis
Pharmaceutical Development and Technology
Authors:

Abstract and Figures

The aim of the present study was to formulate a self-emulsifying drug delivery system of atorvastatin calcium and its characterization including in vitro and in vivo potential. The solubility of atorvastatin calcium was determined in various vehicles such as Captex 355, Captex 355 EP/NF, Ethyl oleate, Capmul MCM, Capmul PG-8, Gelucire 44/14, Tween 80, Tween 20, and PEG 400. Pseudoternary phase diagrams were plotted on the basis of solubility data of drug in various components to evaluate the microemulsification region. Formulation development and screening was carried out based on results obtained from phase diagrams and characteristics of resultant microemulsion. Prepared formulations were tested for microemulsifying properties and evaluated for clarity, precipitation, viscosity determination, drug content and in vitro dissolution. The optimized formulation further evaluated for particle size distribution, zeta potential, stability studies and in vivo potential. In vivo performance of the optimized formulation was evaluated using a Triton-induced hypercholesterolemia model in male Albino Wistar rats. The formulation significantly reduced serum lipid levels as compared with atorvastatin calcium. Thus studies illustrated the potential use for the delivery of hydrophobic drug such as atorvastatin calcium by oral route.
Content may be subject to copyright.
Introduction
Approximately 40% of new drug candidates are
hampered for further development because of their
high lipophilicity and poor water solubility and thus
leads to low bioavailability, high intra and inter-subject
variability and a lack of dose proportionality.[1] Drug
discovery strategies based on the technology of com-
binatorial chemistry information, high throughput
screening, genomics, robotic technology and mini-
aturization have increased the drug libraries of many
pharmaceutical companies to millions. However, during
drug discovery process attempts which are being made
to enhance pharmacological activities of New Chemical
Entity (NCE), very basic physico-chemical properties like
solubility always suer a great deal. Drug molecules with
restricted aqueous solubility are becoming more and
more common in the research and development port-
folios of discovery- focused pharmaceutical companies.
To triumph over these problems, various formulation
strategies are exploited such as use of surfactants, lipids,
permeation enhancers, micronization, salt formation,
cyclodextrins, nanoparticles and solid dispersions. Each
and every method for bioavailability enhancement has
its own merits and demerits. Recently, much attention
has been paid to lipid-based formulations with particu-
lar emphasis on self-emulsifying drug delivery systems
(SEDDS) to improve the oral bioavailability of lipophilic
drugs.[2] SEDDS are dened as an isotropic mixture
of natural or synthetic oils, solid or liquid surfactants
or alternatively one or more hydrophilic solvents and
co-solvents/surfactants. Upon peroral administration,
Pharmaceutical Development and Technology
Pharmaceutical Development and Technology, 2011; 16(1): 65–74
2011
16
1
65
74
Address for Correspondence: Dr Surendra G. Gattani, Prof. and Head, Department of Pharmaceutics and Quality assurance, R. C. Patel Institute of
Pharmaceutical Education and Research, Near Karwand Naka, Shirpur 425 405, Dhule, Maharashtra, India. Tel: +91 099708 16927. Fax: +91 02563 255189.
Email: sggattani@redimail.com
21 March 2009
13 November 2009
13 November 2009
1083-7450
1097-9867
© 2011 Informa Healthcare USA, Inc.
10.3109/10837450903499333
RESEARCH ARTICLE
Enhancement of oral bioavailability of atorvastatin
calcium by self-emulsifying drug delivery systems
(SEDDS)
Pawan J. Kadu, Sachin S. Kushare, Dhaval D. acker, and Surendra G. Gattani
R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule, Maharashtra, India
Abstract
The aim of the present study was to formulate a self-emulsifying drug delivery system of atorvastatin calcium
and its characterization including in vitro and in vivo potential. The solubility of atorvastatin calcium was
determined in various vehicles such as Captex 355, Captex 355 EP/NF, Ethyl oleate, Capmul MCM, Capmul
PG-8, Gelucire 44/14, Tween 80, Tween 20, and PEG 400. Pseudoternary phase diagrams were plotted on
the basis of solubility data of drug in various components to evaluate the microemulsification region.
Formulation development and screening was carried out based on results obtained from phase diagrams
and characteristics of resultant microemulsion. Prepared formulations were tested for microemulsifying
properties and evaluated for clarity, precipitation, viscosity determination, drug content and in vitro dis-
solution. The optimized formulation further evaluated for particle size distribution, zeta potential, stability
studies and in vivo potential. In vivo performance of the optimized formulation was evaluated using a
Triton-induced hypercholesterolemia model in male Albino Wistar rats. The formulation significantly reduced
serum lipid levels as compared with atorvastatin calcium. Thus studies illustrated the potential use for the
delivery of hydrophobic drug such as atorvastatin calcium by oral route.
Keywords: Atorvastatin calcium; self emulsifying drug delivery system; bioavailability enhancement;
Triton-induced hypercholesterolemic in vivo evaluation
PHD
450351
(Received 21 March 2009; revised 13 November 2009; accepted 13 November 2009)
ISSN 1083-7450 print/ISSN 1097-9867 online © 2011 Informa Healthcare USA, Inc.
DOI: 10.3109/10837450903499333 http://www.informahealthcare.com/phd
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by Laurentian University on 02/01/11
For personal use only.
66 P.J. Kadu et al
these systems shape ne oil-in-water emulsions (or
microemulsions) in the gastrointestinal (GI) tract with
mild agitations provided by gastric mobility. ese
systems advantageously present the drug in dissolved
form and the small droplet size provides a large inter-
facial area for solvent drug interaction and ultimately
drug absorption.[1,3] For eective in vivo performance of
SEDDS, the formulation should form droplets of size <
5 µ. e smaller oil droplets oer a large interfacial area
for pancreatic lipase to hydrolyze triglycerides and thus
promote the rapid release of the drug.[4] Selection of a
suitable self emulsifying formulation depends upon the
assessment of solubility of the drug in various compo-
nent, area of the self-emulsifying region as obtained in
the phase diagram, and droplet size distribution of the
resultant emulsion following self-emulsication.[4,5]
Atorvastatin calcium (ATR) is a selective and com-
petitive inhibitor of hydroxyl methyl glutaryl-coenzyme
A (HMG-CoA) reductase. is enzyme is responsible
for converting HMG-CoA to mevalonate, which is a
precursor of cholesterol synthesis. As a result of inhibi-
tion of the enzyme by ATR there occurs a decrease in
mevalonate level, and a subsequent decrease in hepatic
cholesterol levels and increase in uptake of low density
lipoprotein cholesterol (LDL-CH). us ATR is used
to lower cholesterol and triglycerides in patients with
hypercholesterolemia and mixed dyslipidemia and in the
treatment of homozygous familial hypercholesterolemia.
ATR has a unique structure, long half-life, and hepatic
selectivity, explain its greater LDL-CH lowering potency
compared to other HMG-CoA reductase inhibitors.
ATR-[R-(R*,R*)]-2-(4-uorophenyl)- b,d-dihydroxy-5-
(1-methylethyl)-3-phenyl-4-[(phenyl amino) carbonyl]-
1H-pyrrole-1-heptanoic acid, calcium salt (2:1) trihydrate,
is a white to o-white crystalline powder that is insoluble
in aqueous solutions of pH 4 and below, which are the
conditions typically present in the stomach of a subject.
ATR is very slightly soluble in distilled water, pH 7.4
phosphate buer and acetonitrile, slightly soluble in etha-
nol, and freely soluble in methanol, also rapidly absorbed
after oral administration. e absolute bioavailability of
ATR is approximately 14% and as per Biopharmaceutical
Classication System; the drug is classied as a class
II drug.[6–8] For such drugs, dissolution in GI lumen is
a rate controlling step for absorption and subsequent
therapeutic action. Improved absorption can be achieved
by the use of delivery systems, which can enhance drug
dissolution from its dosage form and maintains drug
in dissolved state in GI uids. erefore, the current
scenario demands a need for a delivery strategy that can
improve its therapeutic ecacy. SEDDS recently have
gained great interest in drug delivery research for their
potential in improving oral bioavailability of poorly water
soluble drugs as compared to conventional approaches.
e main objective of the study was to develop and
evaluate an optimal SEDDS formulation containing ATR
and to assess its pharmacodynamic eect in terms of lipid
lowering potential.
Materials and methods
Materials
ATR was a generous gift from FDC Ltd, India. Triglycerides
of caprylic/capric acids (Captex® 355), Triglycerides of
caprylic/capric acids (Captex® 355 EP/NF), C8/C10
mono-/diglycerides (Capmul® MCM) and propylene glycol
caprylate (Capmul PG-8), were generous gifts from Abitec
Corporation USA. Gelucire 44/14 was received as a gift
from Colorcon Asia Pvt. Ltd, Goa, India. Polyethylene
glycol 400 was purchased from Merck Specialties Pvt.
Ltd, Mumbai. Tween 80, Tween 20 and Ethyl oleate
LR were purchased from SD Fine Chem. Ltd, Mumbai.
Empty hard gelatin capsules were obtained as gift from
Associated Capsules Pvt. Ltd, Mumbai. Triton WR 1339
and Sigma® Dialysis Tubing (seamless cellulose tubing,
MWCO 12000) was purchased from Sigma Chemical Co,
USA. In vitro diagnostic kits (Cholesterol LS, LDL-CH
and Triglycerides) were purchased from RFCL Pvt. Ltd,
Haridwar, India. All other chemicals and reagents used
were of analytical grades.
Methods
Solubility studies
e objective of solubility is to determine the solubiliza-
tion capacity for drug in given vehicles. Vehicles which
show highest solubility are then used for formulation
of SEDDS. e solubility of ATR in various vehicles, i.e.
oils (Captex 355, Captex 355 EP/NF, Ethyl oleate LR),
surfactants (Capmul MCM, Capmul PG-8, Gelucire
44/14, Tween 80, Tween 20) and cosurfactant (PEG 400)
was determined initially. A total of 5 mL of each of the
selected vehicles were added to each cap vial containing
an excess of ATR. Vials were then shaken for 48 h in an
orbital shaker (Remi Instruments, Mumbai, India) main-
tained at 37 ± 1°C. After reaching equilibrium, each vial
was centrifuged at 3000 g for 5 min, and excess insoluble
ATR was discarded by ltration using Whatman lter
paper (No. 41). e concentration of solubilized ATR
was quantied by UV spectrophotometer at 241 nm
(Shimadzu-1700, Japan).[5]
Pseudoternary phase diagram study
Pseudoternary phase diagrams of oil, surfactants/
cosurfactant or cosolvents (S/CoS), and water were
developed using the water titration method. e mixtures
of oil and S/CoS at certain weight ratios were diluted with
water in a drop wise manner. For each phase diagram at
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by Laurentian University on 02/01/11
For personal use only.
Enhancement of oral bioavailability of atorvastatin calcium 67
a specic ratio of surfactants, i.e. (1:1, 2:1) and S/CoS (i.e.
1:1, 2:1, 3:1, 4:1 and 5:1 wt/wt), a transparent and homog-
enous mixture of oil and S/CoS was formed. en, each
mixture was titrated with water and visually observed for
phase clarity and ow ability.[5] After the identication of
microemulsion region in the phase diagrams, the micro-
emulsion formulations were selected at desired compo-
nent ratios. In order to form the stable microemulsion, a
series of SEDDS formulations were prepared (Figure 2).
Preparation of SEDDS formulations
A series of SEDDS formulations were prepared using
Captex 355 as the oil, Capmul MCM and Tween 80 as
surfactants mixture (Smix), and PEG 400 as cosolvent
(Table 1). In all the formulations, the level of ATR was
kept constant (i.e. 1.9% wt/wt of the total formulation
weight). Accurately weighed ATR was placed in a glass
vial, and surfactant (Capmul MCM) was added and mixed
by gentle stirring on a magnetic stirrer at 40°C until ATR
was completely dissolved. e remaining components,
i.e. Tween 80, Captex 355 and PEG 400 were added with
constant stirring at 40°C until stable mixture was formed.
60
50
40
30
20
10
0
mg/ml
Tween 80
Tween 20
Ethyle oleate
Captex 355
Captex 355 NF
Capmul MCM
Gelucire 44/14
PEG400
Components
Figure 1. Solubility of ATR in various components.
10
10
10
20
20
20
30
30
30
40
40
40
50 50
50
70
70
70
60
60
60
80
80
80
90
90
90
S/Cos
S/Cos
Smix 1:1, S/Cos 2:1 Smix 1:1, S/Cos 3:1
Register CHEMIX Now!
10
10
10
20
20
20
30
30
30
40
40
40
50
50
50
70
70
70
60
60
60
80
80
80
90
90
90
Register CHEMIX Now!
Water Water
Oil
10
10
10
20
20
20
30
30
30
40
40
40
50
50
50
70
70
70
60
60
60
80
80
80
90
90
90
S/Cos
Smix 1:1, S/Cos 4:1
Register CHEMIX Now!
Water
Oil
10
10
10
20
20
20
30
30
30
40
40
40
50 50
50
70
70
70
60
60
60
80
80
80
90
90
90
S/Cos
Smix 1:1, S/Cos 5:1
Register CHEMIX Now!
Water
Oil
Oil
a
Figure 2. (a) Pseudoternary phase diagrams of oil, water and S/Cos ratio for Smix 1:1. (b) Pseudoternary phase diagrams of oil, water and S/Cos
ratio for Smix 2:1.
Figure 2. continued on next page
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by Laurentian University on 02/01/11
For personal use only.
68 P.J. Kadu et al
e mixture was stored at room temperature until further
use.[5]
Characterization and evaluation of the formulation
Dilution study
SEDDS formulations containing 10 mg of ATR (1 part)
were diluted with 10 parts of distilled water, 0.1 N HCl
and Phosphate buer of pH 6.8 and visually observed
(Table 2).[9]
Drug content
ATR from preweighed SEDDS formulations was extracted
by dissolving the formulations in 25 ml of methanol. ATR
content in the methanolic extract was analyzed spec-
trophotometrically (Shimadzu 1700, Japan) at 241 nm,
against the standard methanolic solution of ATR.[10,11]
Disintegration test
e test was performed according to procedure and spec-
ication mentioned in ocial compendia (USP 2005).
Self-emulsication and precipitation assessment
Assessment of the self-emulsifying properties of SEDDS
formulations were performed by visual assessment.[5,12]
Dierent formulations were categorized on the basis of
speed of emulsication, clarity, and apparent stability of
the resultant emulsion. Visual assessment was performed
by dropwise addition of the SEDDS into 250 mL of dis-
tilled water at room temperature, and the contents were
gently stirred magnetically at 100 rpm. Precipitation
was evaluated by visual assessment of the resultant emul-
sion after 24 h. e formulations were then categorized
as clear (transparent or transparent with bluish tinge),
non-clear (turbid), stable (no precipitation at the end of
24 h), or unstable (showing precipitation within 24 h).
Figure 2. Continued.
10
10
10
20
20
20
30
30
30
40
40
40
50
50
50
70
70
70
60
60
60
80
80
80
90
90
90
S/Cos S/Cos
Smix 2:1, S/Cos 2:1 Smix 2:1, S/Cos 3:1
Register CHEMIX Now!
10
10
10
20
20
20
30
30
30
40
40
40
50
50
50
70
70
70
60
60
60
80
80
80
90
90
90
Register CHEMIX Now!
Water Water
Oil
10
10
10
20
20
20
30
30
30
40
40
40
50 50
50
70
70
70
60
60
60
80
80
80
90
90
90
S/Cos
Smix 2:1, S/Cos 4:1
Register CHEMIX Now!
Water
Oil
10
10
10
20
20
20
30
30
30
40
40
40
50 50
50
70
70
70
60
60
60
80
80
80
90
90
90
S/Cos
Smix 2:1, S/Cos 5:1
Register CHEMIX Now!
Water
Oil
Oil
b
Table 1. Composition of various SEDDS formulations.
Components (% W/W) F1 F2 F3 F4 F5 F6
ATR 1.9 1.9 1.9 1.9 1.9
1.9
Captex-355 39.3 49.1 44.11 53.9 37.2
50.9
Capmul-MCM 31.4 25.5 28.4 23.5 33.3
24.5
Tween-80 15.7 12.7 13.7 11.7 15.7
11.7
PEG-400 11.7 9.8 10.8 8.8 11.9
9.4
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by Laurentian University on 02/01/11
For personal use only.
Enhancement of oral bioavailability of atorvastatin calcium 69
Viscosity determination
SEDDS (1 mL) was diluted 10 and 100 times with the
distilled water in beaker with constant stirring on mag-
netic stirrer.[9] Viscosity of the resultant microemulsion
and undiluted SEDDS was measured using viscometer
(Brookeld-DV-E) (Table 3).
Droplet size analysis
Droplet size of SEDDS diluted with water was determined
using a photon correlation spectrometer (Zetasizer 3000
HAS, Malvern Ltd, UK) based on the laser light scattering
phenomenon.[1,3,10] Optimized formulation was diluted
200 times with puried water. Diluted samples were
directly placed into the module and measurements were
made in triplicate after 2-min stirring (Table 3).
Zeta-potential determination
SEDDS (1 mL) was diluted 10 times and 100 times with dis-
tilled water in beaker with constant stirring on a magnetic
stirrer.[9,13] Zeta-potential of the resulting microemulsion
was determined using the Zetasizer (3000 HAS, Malvern
Ltd, UK) (Figure 3).
In vitro dissolution studies
e purpose of in vitro dissolution study was to check the
dissolution rate of SEDDS.
e quantitative in vitro release test was performed
in 900 mL of phosphate buer pH 6.8 maintained at
37 ± 0.5°C using USP XXIV type II dissolution appara-
tus (Electrolab TDT-08L Plus, India). e paddles were
rotated at 100 rpm. e SEDDS formulations were lled
into transparent hard gelatin capsules (0 sizes) and used
for drug release studies; results were compared with
marketed ATR tablet (ZIVAST). Five mL aliquots were
collected periodically and replaced with fresh dissolution
medium. Aliquots, after ltration through Whatman lter
paper (No. 41), were analyzed spectrophotometrically at
241 nm for ATR content.[14]
In vitro drug diusion studies
e purpose of in vitro diusion study was to check the
permeation of drug through biological membrane. In
vitro diusion studies were carried out by using the dialy-
sis technique.[10] One end of pretreated cellulose dialysis
tubing (7 cm in length) was tied with thread and 0.5 mL
of self-emulsifying formulation (equivalent to 10 mg ATR)
was placed in it along with 0.5 mL of dialyzing medium
(phosphate buer pH 6.8). e other end of tubing was
also secured with thread and was allowed to rotate freely
in the dissolution vessel of a USP XXIV type II dissolu-
tion test apparatus (Electrolab TDT-08L Plus, India) that
contained 900 mL dialyzing medium (phosphate buer
pH 6.8) maintained at 37 ± 0.5°C and stirred at 100 rpm.
Placebo formulation (blank SEDDS, without drug) was
also tested simultaneously under identical conditions so
as to check interference, if any. Aliquots were collected
periodically and replaced with fresh dissolution medium
and analyzed spectrophotometrically at 241 nm for ATR
content.
In vivo studies
In vivo study was approved and performed in accord-
ance with the guideline of the animal ethics committee.
e study was conducted in three groups consisting of
six male albino rats weighing 150–200 g. Animals were
grouped as:
Group I: Six rats for pure ATR drug solution
(Reference);
Table 3. Characterization of SEDDS formulations.
Parameters F1 F2 F3 F4 F5 F6
Drug content (%) 92.8 ± 1.23 91.9 ± 2.83 92.5 ± 1.91 94.7 ± 2.15 96.98 ± 2.78 93.3 ± 2.43
Disintegration time (sec) 2.45 ± 1.72 2.33 ± 1.2 1.49 ± 0.9 3.5 ± 0.35 1.33 ± 1.1 2.13 ± 1.34
S.E.T. (sec) 49 ± 2 98±3 67±5 53±6 59±7 51 ± 1.8
Precipitation Unstable Stable Unstable Stable Stable Stable
Clarity Turbid Bluish Turbid Bluish Bluish Bluish
Viscosity (cps) 0 Times dilution 315 314 312 315 317 319
10 Times dilution 1.12 1.13 1.21 1.11 1.24 1.31
100 Times dilution 0.887 0.884 0.87 0.869 0.845 0.841
Droplet size (nm) 231.11 191.24 135.23 229.11 56.14 231.19
n = 3.
Table 2. Dilution study.
Vehicles F1 F2 F3 F4 F5 F6
Distilled water Stable up to 1 h Unclear within
30 min
Stable up to 3 h Unclear within
30 min
Stable up to 5 h
Unclear within
30 min
0.1 N HCl Stable up to 1 h Unclear within
30 min
Stable up to 3 h Unclear within
30 min
Stable up to 5 h
Unclear within
30 min
Phosphate buer
6.8
Stable up to 1 h Unclear within
30 min
Stable up to 3 h Unclear within
30 min
Stable up to 5 h
Unclear within
30 min
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by Laurentian University on 02/01/11
For personal use only.
70 P.J. Kadu et al
Group II: Six rats for self-microemulsifying drug
delivery system (SMEDDS) formulation of ATR (Test);
Group III: Six rats for placebo of SEDDS formulation
(Placebo);
Group IV: Six rats for Control (No formulation)
(Control)
e rats were fasted overnight and injected intraperi-
toneally 250 mg/kg Triton WR 1339 (Tyloxapol, Sigma
Chemical Co, St Louis, MO, USA) dissolved in 0.5% car-
boxy methyl cellulose (CMC) solution. Control groups
of rats were given the vehicle (CMC solution 0.5%), and
experimental groups were given pure ATR (25 mg/kg
body weight) or the SEDDS formulation (equivalent to
25 mg/kg ATR). e oral dosing was performed by intu-
bation using an 18-gauge feeding needle (the volume
to be fed was 1.0 mL in all cases). Blood samples were
drawn at 0, 24, and 48 h and serum was separated by
centrifugation at 10,000 g. Serum cholesterol, triglyc-
erides, low-density lipoprotein cholesterol (LDL- CH)
were estimated in all groups. Statistical analysis of the
collected data was performed using one way analysis of
variance (ANOVA).[5]
Stability studies
Chemical and physical stability of optimized ATR
SEDDS formulation was assessed at 40 ± 2°C/75 ± 5%
RH as per ICH Guidelines. SEDDS equivalent to 10 mg
ATR was lled in size ‘0’ hard gelatin capsules, packed in
aluminum strips and stored for three months in stabil-
ity chamber (CHM 10S, REMI Instruments Ltd, India).
Samples were analyzed at 0, 30, 60 and 90 days for drug
content, disintegration time and in vitro dissolution
prole.[15]
Results and discussion
Solubility studies
Solubility is the most important criteria for selecting the
vehicle for formulating a self emulsifying formulation.
erefore, the components used in the system should
have high solubilization capacity for the drug, ensuring
the solubilization of the drug in the resultant dispersion
(Figure 1). As seen from the results, Captex 355, Capmul
MCM, Tween 80 and PEG 400 showed the highest solu-
bilization capacity for ATR, as compared to other com-
ponents. us, for present study Captex 355 selected as
an oil, Capmul MCM and Tween 80 as surfactants and
PEG 400 used as co-surfactant.
Low HLB surfactants may also be an important com-
ponent of oral lipid-based
formulations by behaving as a coupling agent for the
high HLB surfactant and lipophilic solvent components
as well as contributing to solubilization by remaining
associated with the lipophilic solvent post dispersion.
Furthermore, using a blend of low and high HLB sur-
factants may also lead to more rapid dispersion and ner
emulsion droplet size on addition to an aqueous phase.
Pseudo ternary phase diagram
Self-microemulsifying systems form ne oil-water emul-
sions with only gentle agitation, upon their introduction
Results
Zeta Potential (mV):
Zeta Deviation (mV):
Conductivity (mS/cm):
Result quality: See result quality report
8.97
22.1 18.0
30.8
0.000.0995
Mean (mV)
Peak 1:
Peak 2:
Peak 3:
63.4
36.6
0.0
Area (%) Width (mV)
6.75
4.02
0.00
Zeta Potential Distribution
100000
80000
60000
40000
20000
0
Total Counts
200 100 0 100 200
Zeta Potential (mV)
Record 2: SMEDDS 1
Figure 3. Zeta potential analysis of optimized formulation.
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by Laurentian University on 02/01/11
For personal use only.
Enhancement of oral bioavailability of atorvastatin calcium 71
into aqueous media. Surfactant and co-surfactant get
preferentially adsorbed at the interface, reducing the
interfacial energy as well as providing a mechanical barrier
to coalescence. e decrease in the free energy required
for the emulsion formation consequently improves the
thermodynamic stability of the microemulsion formula-
tion. erefore, the selection of oil and surfactant, and
the mixing ratio of oil to S/CoS, play an important role in
the formation of the microemulsion.[16] After performing
solubility studies, components in which drug showed
more solubility put forwarded for phase behavior study.
In the present study, combinations of surfactants (Smix)
with high and low HLB values were used. Capmul MCM
has low HLB value (5–6) and Tween 80 having higher
(15). e combination of low and high HLB surfactants
leads to more rapid dispersion and ner emulsion droplet
size on addition to aqueous phase.[17] Capmul MCM and
Tween 80 in the ratio of 2:1 showed wider microemul-
sion existence area and rapid emulsications compared
with1:1 and 3:1. us 2:1 Smix ratio along with cosur-
factant designated as S/Cos used in combination with
constant amount of Captex-355 in the ratio of 2:1, 3:1,
4:1 and 5:1 of S/Cos and Captex-355, respectively. Among
these 4:1 and 5:1 showed wider microemulsion existence
area. Figure 2 shows an increase in the microemulsion
area as increase in S/Cos ratio from 2:1 to 5:1. But in 5:1
ratio the concentration of surfactants goes beyond limit,
hence authors selected 4:1 S/Cos ratio for formulation.
PEG 400 is reported to be incompatible with hard gelatin
capsules when used in high concentrations (> 15% W/W
of total formulation).[5] us PEG 400 concentration was
kept below 15% w/w in formulation.
Dilution study
e objective of the dilution study was to study the degree
of emulsication and recrystallization of the drug, if any.
Viscosity of diluted and undiluted SMEDDS was meas-
ured to study the eect on emulsication time. Dilution
may better mimic conditions in the stomach following
oral administration of SMEDDS pre- concentrate.
Dilution study was carried out to access the eect of dilu-
tion on SMEDDS pre-concentrates. Accurate mixture of
emulsier is necessary to form stable microemulsion, for
the development of SEDDS formulation when one part
of each SEDDS formulation was diluted with 10 parts
of distilled water, 0.1 HCl and phosphate buer 6.8 pH
(Table 2). It implies that the formulation F5 was more
stable because there was no precipitation or cryalalliza-
tion of drug.[9]
Drug content
Drug content of the SEDDS formulations are shown in
Table 3, which was in the limit (98–102%).
Disintegration test
e disintegration time of SEDDS formulation is shown
in Table 3. e results demonstrate that formulation F5
showed less disintegration time (1.33 ± 1.1 sec) compared
with the other formulations.
Self-emulsication and precipitation assessment
It was found that the self-emulsication time (SET)
decreases with an increase in concentration of surfactants
up to 49%, beyond and below which there was turbid and
unstable dispersion. is may be due to excess penetra-
tion of water into the bulk oil causing massive interfacial
disruption and ejection of droplets into the bulk aqueous
phase. However, a higher level of surfactant decreases the
solubility limit of the drug and may subsequently lead
to precipitation. e decrease in self-emulsication time
can be assumed due to the relative increase in surfactants
concentration. Hence the combination of high and low
HLB value surfactants were used because low HLB sur-
factants were behaving as coupling agent for high HLB
surfactants. Furthermore, using a blend of low and high
HLB surfactants may also lead to more rapid dispersion
and ner emulsion droplet size on addition to an aque-
ous phase.[17]
As concentration of surfactant increases the molec-
ular volume increases which aects penetration at
the interface hence SET decreases. e ratio of Smix
of 2:1 and S/CoS of 4:1 was kept constant for initial
formulations.
Viscosity determination
Viscosity of SEDDS without dilution was found to be in
between 312 and 319 cP, which was suitable for lling in
hard gelatin capsule without risk of leaking problem. As
SEDDS was diluted 10 and 100 times with water, viscosity
of the system was decreased, which indicates that oral
administration of SEDDS formulation will be diluted
with the stomach uid and viscosity will be decreased
and therefore absorption from the stomach will be fast
(Table 3).
Droplet size analysis
e droplet size of the emulsion is a crucial factor in self-
emulsication performance because it determines the
rate and extent of drug release as well as drug absorption.
Also, it has been reported that the smaller particle size of
the emulsion droplets may lead to more rapid absorption
and improve the bioavailability.[13] It was observed that
increasing the S/CoS ratio led to decrease in mean drop-
let size. It is well known that in microemulsion systems
the addition of surfactants stabilize and condense the
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by Laurentian University on 02/01/11
For personal use only.
72 P.J. Kadu et al
interfacial lm, while the addition of cosurfactant causes
the lm to expand; thus, the relative proportion of sur-
factant to cosurfactant has varied eects on the droplet
size. e SEDDS was found to be clear transparent after
the 100 times dilution with distilled water and remained
stable (Table 3).
Zeta-potential determination
e magnitude of the zeta potential gives an indication
of the potential stability of the colloidal system. If all the
particles have a large negative or positive zeta potential
they will repel each other and there is dispersion stability.
If the particles have low zeta potential values then there
is no force to prevent the particles coming together and
there is dispersion instability. A dividing line between
stable and unstable aqueous dispersions is generally
taken at either +30 or −30mV. Particles with zeta poten-
tials more positive than +30mV are normally considered
stable. Particles with zeta potentials more negative than
−30mV are normally considered stable. Zeta potential of
the system was found to be −22.1 mV, which indicated
the droplets of microemulsion having negative charge,
which is closer to range.
In vitro dissolution studies
Drug release from the SEDDS formulation (F5) was
found to be signicantly higher (P) as compared with
that of Marketed ATR tablet (ZIVAST, FDC Ltd, India).
e result indicates that (F5) shows 70% of drug release
within 10 min and 97% of drug release in 60 min, which
is higher than the marketed formulation (39% and 61%
in 10 and 60 min, respectively). It could be suggested
that the SEDDS formulation (F5) resulted in spontane-
ous formation of a microemulsion with a small droplet
size, which permitted a faster rate of drug release into
the aqueous phase, much faster than that of Marketed
ATR tablet. e dramatic increase in the rate of release of
ATR from SMEDDS compared to marketed formulation
can be attributed to its quick dispersability and ability to
keep drug in solubilized state. us, this greater availabil-
ity of dissolved ATR from the SEDDS formulation could
lead to higher absorption and higher oral bioavailability
(Figure 4).
In vitro drug diusion studies
Conventional dissolution testing of SEDDS has a limita-
tion in mimicking its real time in vivo dissolution and
such a technique can only provide a measure of dispers-
ibility of SEDDS in the dissolution medium. Alternatively,
for evaluating the in vitro performance of SEDDS, drug
diusion studies using the dialysis technique are very
popular and well documented in many literatures.[10,16]
Diusion studies were performed for SEDDS F2, F4,
and F5, as these formulations show smaller droplet size
among other formulations. ough SEDDS F3 has less
droplet size than F2 it found unstable and turbid on
precipitation and clarity test, respectively. e release of
ATR from these dosage forms was evaluated in phosphate
buer pH 6.8; the release percentage of F5 was signi-
cantly higher than that of F2 and F4 (Figure 5). It could
suggest that ATR dissolved perfectly in SEDDS form could
be released due to the small droplet size, which permits a
faster rate of drug release into aqueous phase. e release
rate of ATR from SEDDS F5 (mean droplet size: 56.86 nm)
was faster than SEDDS F2 and F4 (mean droplet size: 191
and 231 nm, respectively). In this study, diusion proles
of all three formulations (F2, F4, and F5) did not show
any dierences during initial 2 h, however, at the end
of 12 h, formulation F5 showed about 97.07% diusion
against F2 and F4 shows 85.64% and 87.31% diusion,
respectively (Figure 5). Results clearly indicate the eect
of mean droplet size on drug diusion across dialyzing
membrane. Hence increasing the particle size of micro-
emulsion could decrease the release rate of drug and it
might suggest that release rate of drug could be control-
led by regulating the mean particle size.
120
100
80
60
40
20
0
% Cumulative Release
F1
F2
F3
F4
F5
F6
ZIVAST
010203040506070
Time (min)
Figure 4. In vitro dissolution studies of various developed and mar-
keted (ZIVAST) formulations.
120
100
80
60
40
20
0
% Diffusion
F2
F4
F5
02468101214
Time (h)
Figure 5. In vitro diusion studies of F2, F4 and F5.
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by Laurentian University on 02/01/11
For personal use only.
Enhancement of oral bioavailability of atorvastatin calcium 73
In vivo studies
e study was performed to evaluate the pharmacody-
namic potential of an optimized formulation (F5) against
pure ATR using a Triton-induced hyperlipidemia model.
Triton is a non-ionic surfactant that induces hyper-
lipidemia by inhibiting peripheral lipoprotein lipase
enzymes responsible for removal of lipid particles from
the body. e administration of Triton leads to transient
elevation of lipid levels. Hypolipidemic activity of ATR
causes reduction in elevated total CH, LDL-CH and TG
levels in blood. At the same time, it causes elevation
of plasma HDL-CH level, which promotes the removal
of cholesterol from peripheral cells and facilitates its
delivery back to the liver (Figure 6). As ATR has longer
half-life of 14 h, it is known to have a longer stay in
the blood circulation. us there is longer duration of
action due to an optimal initial plasma drug level. On
comparison with pure ATR, optimized SEDDS formu-
lation (F5) shows better results. Results indicates that
in 48 h ATR lowered cholesterol (78.61% inhibition),
triglyceride (63.77% inhibition), and LDL (49.65% inhi-
bition), and SEDDS formulation resulted in a greater
reduction of cholesterol (95.93% inhibition), triglycer-
ide (95.47% inhibition), and LDL (100.14% inhibition).
e higher lipid-lowering activity of the SEDDS is due
to complete dissolution of ATR in SEDDS, which could
have increased absorption and thereby led to a higher
plasma drug concentration (higher bioavailability).
e low bioavailability of ATR is attributed to its poor
aqueous solubility. e results of statistical analysis
of collected data using one way analysis of variance
(ANOVA) shows signicant dierence between pure
ATR and optimized SEDDS formulation (P < 0.001). e
above dierence in pharmacodynamic activity and the
results from in vitro dissolution studies suggest that the
SEDDS formulation shows better lipid lowering activity
than pure ATR.
Stability studies
Optimized formulation (F5) lled into hard gelatin cap-
sules as the nal dosage form. However liquid-lled hard
gelatin capsules are prone to leakage, and the entire sys-
tem has a very limited shelf life owing to its liquid char-
acteristics and the possibility of precipitation of the drug
from the system. us, the optimized formulation (F5)
was subjected to stability studies to evaluate its stability
and the integrity of the dosage form. No change in the
physical parameters such as homogeneity and clarity
was observed during the stability studies. ere was no
major change in the drug content, disintegration time,
and in vitro dissolution prole. It was also observed that
the formulation was compatible with the hard gelatin
capsule shells. Also, there was no phase separation, and
drug precipitation was found at the end of three-month
stability studies indicating that ATR remained chemically
stable in SEDDS (Table 4).
Conclusions
SEDDS appeared to be an interesting approach to
improve problems associated with oral delivery of ATR.
ATR SEDDS formulation was superior to marketed
formulation in respect to in vitro dissolution prole. It
also shows better in vivo hypolipidimic activity than
pure ATR. us, SEDDS can be regarded as a novel
and commercially feasible alternative to current ATR
formulations
Acknowledgements
e authors are thankful to R.C. Patel Institute of
Pharmaceutical Education and Research for providing
120
100
80
60
40
20
0
120
100
80
60
40
20
0
120
100
80
60
40
20
0
% Inhibition% Inhibition% Inhibition
24hrs 48hrs
Time(Hr)
Triglyceride
LDL Cholesterol
Cholesterol
ATR
F5
**
** **
**
**
**
**
**
**
**
**
**
Figure 6. In vivo study of ATR and optimized formulation F5.
Table 4. Stability studies.
Sampling points
Disintegration
time (sec) % Drug content % Drug release
0 day 1.33 ± 1.1 95.1 ± 1.83 96.98 ± 2.78
30 days 1.49 ± 0.9 94.7 ± 2.15 95.31 ± 3.35
60 days 2.33 ± 1.2 93.3 ± 2.43 93.7 ± 2.85
90 days 2.45 ± 1.7 91.9 ± 2.83 92.5 ± 1.91
n = 3.
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by Laurentian University on 02/01/11
For personal use only.
74 P.J. Kadu et al
facilities to carry out the research work. e authors are
also thankful to FDC Ltd (India), Abitec Corporation
(USA), Colorcon Asia Pvt. Ltd (Goa, India), and Associated
capsules (India) for providing gift samples of ATR, oils
and surfactants, Gelucire 44/14 and empty hard gelatin
capsules, respectively.
Declaration of interest
e authors report no conicts of interest. e authors
alone are responsible for the content and writing of the
pap e r.
References
1. Gursoy R, Benita S. Self-emulsifying drug delivery systems
(SEDDS) for improved oral delivery of lipophilic drugs. Biomed
Pharmacother 2004;58:173–182.
2. Tang J, Sun J, He Z. Self-emulsifying drug delivery systems:
Strategy for improving oral delivery of poorly soluble drugs. Curr
Drug er 2007;2:85–93.
3. Pouton C. Self-emulsifying drug delivery systems:
Assessment of the eciency of emulsication. Int J Pharm
1985;27:335–348.
4. Kommuru T, Gurley B, Khan M, Reddy I. Self-emulsifying
drug delivery systems (SEDDS) of coenzyme Q10: Formulation
development and bioavailability assessment. Int J Pharm
2001;212:233–246.
5. Patel A, Vavia P. Preparation and in vivo evaluation of SMEDDS
(Self-microemulsifying drug delivery system) containing
fenobrate. AAPS J 2007;9(3):344–352.
6. Kim M, Jin S, Kim J, Park H, Song H, Neubert R, Hwang S.
Preparation, characterization and in vivo evaluation of amor-
phous atorvastatin calcium nanoparticles using supercritical
antisolvent (SAS) process. Eur J Pharm Biopharm 2008;
69:454–465.
7. Kim J, Kim M, Park H, Jin S, Lee S, Hwang S. Physicochemical
properties and oral bioavailability of amorphous atorvastatin
hemi-calcium using spray-drying and SAS process. Int J Pharm
2008;359:211–219.
8. Dollery C. erapeutic drugs. 2nd ed. Vol. 1. London: Churchill
Livingstone; 1999:A228–232.
9. Patel D, Sawant K. Oral bioavailability enhancement of acyclovir
by self-microemulsifying drug delivery systems (SMEDDS). Drug
Develop Ind Pharm 2007;33:1318–1326.
10. Paradkar A , Patil P, Patil V. Formulation of a self-emulsifying
system for oral delivery of simvastatin: In vitro and in vivo evalu-
ation. Acta Pharm 2007;57:111–122.
11. Patil P, Praveen S, Shobha Rani R, Paradkar A. Bioavailability
assessment of ketoprofen incorporated in gelled self-
emulsifying formulation: A technical note. AAPS PharmSciTech
2005;6(1):9–13.
12. Khooa S, Humberstonea A, Portera C, Edwardsb G, Charmana W.
Formulation design and bioavailability assessment of lipidic
self-emulsifying formulations of halofantrine. Int J Pharm 1998;
167:155–164.
13. Liu L, Pang X, Zhang W, Wang S. Silymarin-loaded self-
microemulsifying drug delivery systems. Asian J Pharm Sci
2007;2(4):150–160.
14. Arayne S. In vitro availability of atorvastatin in presence of losa-
rtan. Pak J Pharm Sci 2006;19(2):134–141.
15. Borhade V, Nair H, Hegde D. Design and evaluation of self
microemulsifying drug delivery system (SMEDDS) of tacrolimus.
AAPS PharmSciTech 2008;9(1):13–21.
16. Kang B, et al.Development of self-microemulsifying drug deliv-
ery systems (SMEDDS) for oral bioavailability enhancement of
simvastatin in beagle dogs. Int J Pharm 2004;274:65–73.
17. Liu R. Water insoluble formulations. 2nd ed. Boca Raton,
FL: CRC Press; 2008.
Pharmaceutical Development and Technology Downloaded from informahealthcare.com by Laurentian University on 02/01/11
For personal use only.
... However, poor drug solubility limits its bioavailability [24]. Therefore, SNEDDS formulation was used in the literature with a remarkable increment in AT bioavailability and therapeutic activity [25][26][27][28][29][30][31]. Moreover, various studies have shown that AT suffers from chemical degradation when exposed to acid microenvironments [32,33]. ...
Article
Full-text available
Atorvastatin (AT) is widely prescribed by physicians during the treatment of hyperlipidemia. The self-nanoemulsifying drug delivery system (SNEDDS) is used to overcome its low drug solubility and bioavailability. However, the presence of free fatty acids in SNEDDS formulation resulted in remarkable AT degradation. This study explores innovative carbonated SNEDDS to enhance the stability of AT within SNEDDS formulation. Various types of SNEDDS formulations were prepared and evaluated. In vitro dissolution was performed to examine the ability of SNEDDS formulation to enhance AT dissolution. The solidified SNEDDS formation was prepared using Syloid adsorbent (AT-SF6). In addition, sodium bicarbonate was loaded within the best formulation at various concentrations to prepare carbonated SNEDDS (AT-CF6). Kinetics of drug degradation were studied over 45 days to assess AT stability in SNEDDS formulations. It was found that the SNEDDS formulation was able to enhance the dissolution of AT by about 1.5-fold compared with the pure drug formulation. AT-SF6 did not reduce the degradation rate of the drug compared with AT-F6. However, AT-CF6 formulations showed that increasing the concentration of incorporated sodium bicarbonate significantly reduced the degradation rate of AT. It was found that sodium bicarbonate in AT-CF6 significantly reduced the degradation rate of AT (0.00019) six-fold compared with AT-F6 (0.00115). The obtained results show that carbonated SNEDDS is a promising approach to enhance the stability of acid-labile drugs and their pharmaceutical application.
... The use of non-ionic surfactant reduces the magnitude of the zeta potential as a result of the decrease in the surface charge of the droplets [59]. For the stability of disperse systems, it is aimed that the zeta potential is greater than + 30 mV or less than − 30 mV [60], but thermodynamically stable microemulsions are stable even if the zeta potential is quite close to zero [61][62][63]. In addition, the high negative surface charge (close to zero) means that the formulation is stable, and aggregation will not occur [64]. ...
Article
Full-text available
Modern drug carrier technologies, such as microemulsions with small droplet sizes and high surface areas, improve the ability of low water solubility active ingredients to permeate and localize. The goal of this study was to create microemulsion formulations for wound healing that contained both fusidic acid (FA), an antibacterial agent, and benzocaine (BNZ), a local anesthetic. Studies on characterization were carried out, including viscosity, droplet size, and zeta potential. The drug-loaded microemulsion had a stable structure with –3.014 ± 1.265 mV of zeta potential and 19.388 ± 0.480 nm of droplet size. In both in vitro release and ex vivo permeability studies, the microemulsion was compared with Fucidin cream and oily BNZ solution. According to the drug release studies, BNZ release from the microemulsion and the BNZ solution showed a similar profile ( p > 0.05), while FA release from the microemulsion had a higher drug release compared to Fucidin cream ( p < 0.001). The microemulsion presented lower drug permeation ( p > 0.05) for both active ingredients, on the other hand, provided higher drug accumulation compared to the control preparations. Moreover, according to the results of in vitro wound healing activity, the microemulsion indicated a dose-dependent wound healing potential with the highest wound healing activity at the highest concentrations. To the best of our knowledge, this developed BNZ- and FA-loaded microemulsion would be a promising candidate to create new opportunities for wound healing thanks to present the active ingredients, which have low water solubility, in a single formulation and achieved higher accumulation than control preparations. Graphical Abstract
... In general, it is thought that a ZP greater than +30 mV or less than −30 mV offers enough repulsion forces to avoid particle aggregation [12]. The colloidal dispersion was guaranteed to be stable for the optimized SXE-NTF formulation, which exhibited a zeta potential of −29.69 mV ( Figure 3). ...
Article
Full-text available
The goal of this investigation is to improve the topical delivery of medicine by preparing and maximizing the potential of a nanotransferosome gel infused with Solanum xanthocarpum methanolic extract (SXE) to provide localized and regulated distribution. Thin-film hydration was used to create SXE-infused nanotransferosomes (SXE-NTFs), and a Box-Behnken design was used to improve them. Phospholipon 90G (X1), cholesterol (X2) and sodium cholate (X3) were chosen as the independent variables, and their effects on vesicle size (Y1), polydispersity index (PDI) (Y2) and the percentage of entrapment efficiency (EE) (Y3) were observed both individually and in combination. For the SXE-NTFs, the vesicle size was 146.3 nm, the PDI was 0.2594, the EE was 82.24 ± 2.64%, the drug-loading capacity was 8.367 ± 0.07% and the drug release rate was 78.86 ± 5.24%. Comparing the antioxidant activity to conventional ascorbic acid, it was determined to be 83.51 ± 3.27%. Ex vivo permeation tests revealed that the SXE-NTF gel (82.86 ± 2.38%) considerably outperformed the SXE gel (35.28 ± 1.62%) in terms of permeation. In addition, it seemed from the confocal laser scanning microscopy (CLSM) picture of the Wistar rat's skin that the rhodamine-B-loaded SXE-NTF gel had a higher penetration capability than the control. Dermatokinetic studies showed that the SXE-NTF gel had a better retention capability than the SXE gel. According to the experimental results, the SXE-NTF gel is a promising and successful topical delivery formulation.
... Berbagai teknik telah dilakukan untuk meningkatkan disolusi kalsium atorvastatin, antara lain modifikasi kristal (Gozali et al., 2014;Wicaksono et al., 2017;Wicaksono et al., 2019), co-grinding (Prabhu & Patravale, 2015), dispersi padat (Khan & Dehghan, 2011;Panghal et al., 2014;Rodde et al., 2014;Gozali et al., 2015), formulasi berbasis lipid seperti mikro dan nanoemulsi (Chouksey et al., 2011;Kadu et al., 2011;Snela et al., 2019) dan teknik likuisolid (Sanjeev et al., 2010;Baskaran et al., 2016;Windriyati et al., 2020;Patil, et al., 2021). Metode yang relatif murah dan cukup mudah diterapkan dalam skala industri farmasi adalah teknik likuisolid karena tidak membutuhkan energi besar seperti pemanasan dan tidak menggunakan pelarut organik yang mudah menguap (Yadavb & Yardava, 2009;Lu et al., 2016). ...
Article
Full-text available
Background: Marketed tablets of drugs must fulfill the required standards of dissolution to guarantee the equivalent to reference. Atorvastatin calcium needs to be enhanced dissolution since this compound included BCS Class II drugs with low solubility and high permeability. This meaning that the dissolution affects the bioavailability of drugs.Objective: This research aimed to develop a formulation of a liquisolid tablet using propylene glycol as a solvent and some carrier materials in various compositions to increase the dissolution of atorvastatin calcium.Method: Different formulations of liquisolid tablets were conducted using different quantities of carrier material like Avicel PH 101, Avicel PH 102, and Neusilin US2, while Aerosil 200 as the coating material. The liquisolid powder was compressed into tablets by the direct compressing method. The liquisolid tablets were characterized for their properties and possible drug-excipient interaction by XRD and FTIR analysis.Results: The liquisolid tablets of atorvastatin calcium were within the acceptable limits criteria. The dissolution of AA4 tablets was higher compared to marketed tablets. Based on the XRD and FTIR analysis, no chemical interactions between drug and excipient.Conclusion: The liquisolid formulation can then be developed as an alternative for the production of atorvastatin calcium tablets in the pharmaceutical industry.Keywords: Atorvastatin calcium, Dissolution, Liquisolid, propylene glycol Intisari Latar belakang: Sediaan tablet yang dipasarkan harus memenuhi standar disolusi yang ditetapkan untuk menjamin ekivalensinya dengan produk standar. Kalsium atorvastatin perlu ditingkatkan disolusinya karena termasuk dalam kelas II Sistem Klasifikasi Biofarmasetik dengan sifat kelarutan yang rendah dan permeabilitas yang tinggi. Disolusi kalsium atorvastatin mempengaruhi bioavailabilitasnya.Tujuan: Penelitian ini bertujuan mengembangkan formulasi tablet likuisolid menggunakan pelarut propilenglikol dan beberapa bahan pembawa dengan variasi komposisi untuk meningkatkan disolusi kalsium atorvastatin.Metode: Tablet likuisolid dibuat dengan menambahkan berbagai komposisi bahan pembawa seperti Avicel PH 101, Avicel PH 102, dan Neusilin US2, serta Aerosil 200 sebagai pelapis pada suspensi kalsium atorvastatin dalam propilenglikol. Serbuk likuisolid yang terbentuk lalu dikempa menjadi tablet dengan cetak langsung. Tablet likuisolid kalsium atorvastatin dievaluasi karakteristiknya dan dianalisis dengan XRD dan FTIR untuk mengetahui adanya interaksi antara zat aktif dengan eksipien yang digunakan. Hasil: Tablet likuisolid kalsium atorvastatin memenuhi kriteria tablet dalam literatur dan kompendia. Disolusi tablet likuisolid AA4 lebih tinggi dibandingkan dengan tablet yang beredar di pasaran. Hasil analisis XRD dan FTIR menunjukkan tidak ada interaksi kimia antara zat aktif dengan eksipien.Kesimpulan: Formulasi tablet likuisolid dapat dikembangkan sebagai alternatif untuk produksi tablet kalsium atorvastatin di industri farmasi.
Article
Oral drug delivery is noninvasive, has a high rate of patient compliance, is easy to handle, and doesn't require any special sterile settings, it is the most popular method of administration. Nevertheless, a number of physical, biological, and pharmacological obstacles that certain medications must overcome in order to be absorbed into the systemic circulation reduce their therapeutic efficiency. The use of nanocarriers to deliver drugs orally has proven to be an effective solution to the aforementioned problems and is being explored as a potential replacement for oral medication administration. This chapter provides an overview of the latest developments in the use of nanocarriers for oral medication delivery in the treatment of different disorders. The chapter also describes how diverse nanocarrier designs and technologies improve therapeutic potential by overcoming physical, biological, and biochemical obstacles. it is easy to produce, has few sterility limitations, is less expensive, has flexible dosage form design, and has high patient compliance, oral drug delivery (ODD) is the most convenient and favored method of drug administration. However, low medication bioavailability—which is influenced by three crucial factors—is one of the difficulties associated with oral drug delivery. The other is solubility. Many mathematical models that predict the medication's rate of solubility and dissolution have been developed in order to achieve efficient drug absorption in vivo. Similarly, models that are noncellular and cellular determine permeability. Furthermore, the medication's behavior in the gastrointestinal tract (GIT) is influenced by physiological parameters, such as pH, microbial colonization, and enzymes, as well as intrinsic drug properties. A drug's dosage form is a method of getting it into a living organism. The medicine must be administered to the site of action at a rate and concentration that will maximize therapeutic benefit and minimize side effects in order to provide the intended result. Although the oral method is still commonly used, swallowing tablets and capsules can be a regular problem. As a result, numerous studies on cutting-edge drug delivery methods have been conducted. This review focuses on oral dispersible tablets, a novel approach to drug delivery systems that are currently more focused on formulation and set a new course that not only helped patients increase their level of therapy compliance.
Article
The influence of resonance wave effects on the sedimentation stability of potato and corn starch nanoparticle dispersions obtained by coprecipitation was studied. It has been established that the proportion of the dispersed phase of potato starch nanoparticle dispersions formed using traditional mixing remains unchanged for two days. For corn starch nanoparticle dispersions, this indicator remains at the initial level only for the first five minutes. The use of wave action at the stage of coprecipitation leads to an increase in the values of the ξ-potential of the obtained nanoparticles by 4.5 and 3.5 times for corn and potato starches, respectively. Due to this, the dispersion stability of corn starch nanoparticles increases up to two days and dispersions of potato starch nanoparticles increase up to forty days. The results presented in this article are the basis for the development of a resource-saving technology for obtaining highly stable dispersions of biopolymer nanoparticles for food, medical, pharmaceutical, and other industries.
Article
Full-text available
Development of coating technologies for electrochemical sensors that consistently exhibit antifouling activities in diverse and complex biological environments over extended time is vital for effective medical devices and diagnostics. Here, we describe a micrometer-thick, porous nanocomposite coating with both antifouling and electroconducting properties that enhances the sensitivity of electrochemical sensors. Nozzle printing of oil-in-water emulsion is used to create a 1 micrometer thick coating composed of cross-linked albumin with interconnected pores and gold nanowires. The layer resists biofouling and maintains rapid electron transfer kinetics for over one month when exposed directly to complex biological fluids, including serum and nasopharyngeal secretions. Compared to a thinner (nanometer thick) antifouling coating made with drop casting or a spin coating of the same thickness, the thick porous nanocomposite sensor exhibits sensitivities that are enhanced by 3.75- to 17-fold when three different target biomolecules are tested. As a result, emulsion-coated, multiplexed electrochemical sensors can carry out simultaneous detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) nucleic acid, antigen, and host antibody in clinical specimens with high sensitivity and specificity. This thick porous emulsion coating technology holds promise in addressing hurdles currently restricting the application of electrochemical sensors for point-of-care diagnostics, implantable devices, and other healthcare monitoring systems.
Article
Full-text available
Conclusion Based on the results of the present study, it is apparent that the gelled SEF containing KPF did not significantly alter its bioavailability as compared with that of an immediate release solid dosage form when administered to human volunteers by the oral route.
Article
Purpose: To develop a stable formulation for self-microemulsifying drug delivery systems (SMEDDS) in order to enhance the solubility, release rate, and oral absorption of the poorly soluble drug, silymarin. Methods: A series of solubility studies using various vehicles were carried out using HPLC. Two types of pseudo-ternary phase diagrams were constructed to study the phase behaviors and the most effi cient self-microemulsifying regions when the system was diluted with 0.1 mol/l hydrochloric acid solution. In vitro release was investigated using a bulk-equilibrium reverse dialysis bag method. Results: A smaller gel-like domain was found in pseudo-ternary phase diagrams when diluted with 0.1 mol/l hydrochloride solution, and the fi nal formulation with good self-emulsifying ability as well as a high solubilization capacity was established. The mean particle size of the resultant emulsions was about 127 nm, with a singlet-peak in the volume-weighted particle size distribution obtained by the particle size analyzer regardless of the dispersion medium; the ζ-potential of the silymarin SMEDDS in distilled water was –10.2 mV, determined by a Coulter counter, model DELSA 440. Conclusions: Differences in the release medium signifi cantly infl uenced the drug release from SMEDDS and the release profi les of silymarin from SMEDDS was higher than that for commercial capsules (Legalon, Germany), and signifi cantly higher than that for commercial tablets (Yiganling, China). The optimal formulation of SMEDDS is an alternative oral dosage form for improving the oral absorption of silymarin.
Article
Self-emulsifying formulations have potential uses as vehicles for the administration of lipophilic drugs by the oral route. In this study model oil-surfactant mixtures were allowed to self-emulsify under conditions of gentle agitation. During emulsification the relative intensity of light scattered by the dispersion was monitored continuously which enabled the rates of emulsification to be compared. The particle sizes of resultant emulsions were compared by light microscopy and using a Coulter Nano-Sizer. Efficient self-emulsifying formulations were produced by the oils Miglyol 812 or Miglyol 840 in combination with the surfactant Tween 85. The properties of these systems have been studied over a range of mixture compositions. The finest dispersions were produced rapidly and in reproducible time by a mixture of 30% w/w Tween 85 and 70% w/w Miglyol 812.
Article
Drugs are most often administered by the oral route. However, more than 40% of new chemical entities exhibit poor aqueous solubility, resulting in unsatisfactory oral drug delivery. Recently, much attention has been focused on selfemulsifying drug delivery systems (SEDDS) to improve the oral bioavailability of poorly aqueous soluble drugs. SEDDS are isotropic mixtures of oil, surfactants, solvents and co-solvents/surfactants. The principal characteristic of these systems is their ability to form fine oil-in-water (o/w) emulsions or microemulsions upon mild agitation following dilution by an aqueous phase. For lipophilic drugs, which display dissolution rate-limited absorption, SEDDS may be a promising strategy to improve the rate and extent of oral absorption. This article gives an overview of the new excipients used in SEDDS and biopharmaceutical aspects of SEDDS. The application of SEDDS and closely related lipid-based systems as drug delivery vehicles is also introduced, with particular emphasis being placed on the application of SEDDS in traditional Chinese medicine (TCM).
Article
The potential for lipidic self-emulsifying drug delivery systems (SEDDS) and self-microemulsifying drug delivery systems (SMEDDS) to improve the oral bioavailability of a poorly absorbed, antimalarial drug (Halofantrine, Hf) was investigated in fasted beagles. Hf free base, rather than the commercially available hydrochloride salt (Hf.HCl), was studied due to its much higher solubility in lipidic triglyceride solvents. The multi-component delivery systems were optimised by evaluating their ability to self-emulsify when introduced to an aqueous medium under gentle agitation, and by determination of particle size of the resulting emulsion. Optimised formulations selected for bioavailability assessment were medium-chain triglyceride SEDDS and SMEDDS, and a long-chain triglyceride SMEDDS. The relevant pharmacokinetic parameters of Hf, and its desbutyl metabolite, were determined relative to an intravenous formulation. The lipid-based formulations of Hf base afforded a six- to eight-fold improvement in absolute oral bioavailability relative to previous data of the solid Hf.HCl tablet formulation. These data indicate the utility of dispersed lipid-based formulations for the oral delivery of Hf free base, and potentially other lipophilic drugs.
Article
Formulation of a self-emulsifying system for oral delivery of simvastatin: In vitro and in vivo evaluation The objective of the present work was to formulate a self-emulsifying drug delivery system (SEDDS) for simvastatin, which is widely used in the treatment of hypercholesterolemia and dyslipidemia as an adjunct to diet. Simvastatin SEDDS were formulated using a 1:1 ( V/V ) mixture of diesters of caprylic/capric acids and polyglycolyzed glycerides with varying concentrations of polyoxy castor oil and C8/C10 mono-/diglycerides. The developed SEDDS were evaluated for turbidimetry, droplet size analysis, drug content and in vitro diffusion profiles. In vivo performance of the optimized formulation was evaluated in rats using pharmacodynamic marker parameters like plasma total cholesterol (CH), triglycerides (TG) and high-density lipoprotein (HDL-CH) for 21 days. SEDDS containing 9.1% ( m/m ) simvastatin and 23.0% ( m/m ) of each excipient showed minimum mean droplet size (124 nm) and optimal drug diffusion. This test formulation showed significant reduction in plasma CH and TG (around 5-fold and 4-fold, respectively), while HDL-CH concentration was markedly higher (2-fold) compared a reference simvastatin suspension formulation after oral administration for 21 days of study. Test formulation has shown enhanced pharmacodynamic performance compared to reference formulation in rats. The study illustrated the potential of simvastatin SEDDS for oral administration and its biopharmaceutic performance.
Article
The goals of our investigations are to develop and characterize self-emulsifying drug delivery systems (SEDDS) of coenzyme Q10 (CoQ10), using polyglycolyzed glycerides (PGG) as emulsifiers and to evaluate their bioavailability in dogs. Solubility of CoQ10 was determined in various oils and surfactants. SEDDS consisted of oil, a surfactant and a cosurfactant. Four types of self-emulsifying formulations were prepared using two oils (Myvacet 9-45 and Captex-200), two emulsifiers (Labrafac CM-10 and Labrasol) and a cosurfactant (lauroglycol). In all the formulations, the level of CoQ10 was fixed at 5.66% w/w of the vehicle. The in vitro self-emulsification properties and droplet size analysis of these formulations upon their addition to water under mild agitation conditions were studied. Pseudo-ternary phase diagrams were constructed identifying the efficient self-emulsification region. From these studies, an optimized formulation was selected and its bioavailability was compared with a powder formulation in dogs. Medium chain oils and Myvacet 9-45 provided higher solubility than long chain oils. Efficient and better self-emulsification processes were observed for the systems containing Labrafac CM-10 than formulations containing Labrasol. Addition of a cosurfactant improved the spontaneity of self-emulsification. From these studies, an optimized formulation consisting of Myvacet 9-45 (40%), Labrasol (50%) and lauroglycol (10%) was selected for its bioavailability assessment. A two-fold increase in the bioavailability was observed for the self-emulsifying system compared to a powder formulation. SEDDS have improved the bioavailability of CoQ10 significantly. The data suggest the potential use of SEDDS to provide an efficient way of improving oral absorption of lipophilic drugs.
Article
The main purpose of this work is to prepare self-microemulsifying drug delivery system (SMEDDS) for oral bioavailability enhancement of a poorly water soluble drug, simvastatin. Solubility of simvastatin was determined in various vehicles. SMEDDS is mixture of oils, surfactants, and cosurfactants, which are emulsified in aqueous media under conditions of gentle agitation and digestive motility that would be encountered in the gastro-intestinal (GI) tract. Pseudo-ternary phase diagrams were constructed to identify the efficient self-emulsification region and particle size distributions of the resultant microemulsions were determined using a laser diffraction sizer. Optimized formulations for in vitro dissolution and bioavailability assessment were Carpryol 90 (37%), Cremophor EL (28%), and Carbitol (28%). The release rate of simvastatin from SMEDDS was significantly higher than the conventional tablet. The prepared SMEDDS was compared with the conventional tablet (Zocor) by administering the prefilled hard capsules to fasted beagle dogs. The absorption of simvastatin acid from SMEDDS form resulted in about 1.5-fold increase in bioavailability compared with the conventional tablet. Our studies illustrated the potential use of SMEDDS for the delivery of hydrophobic compounds, such as simvastatin by the oral route.
Article
The oral delivery of hydrophobic drugs presents a major challenge because of the low aqueous solubility of such compounds. Self-emulsifying drug delivery systems (SEDDS), which are isotropic mixtures of oils, surfactants, solvents and co-solvents/surfactants, can be used for the design of formulations in order to improve the oral absorption of highly lipophilic drug compounds. SEDDS can be orally administered in soft or hard gelatin capsules and form fine relatively stable oil-in-water (o/w) emulsions upon aqueous dilution owing to the gentle agitation of the gastrointestinal fluids. The efficiency of oral absorption of the drug compound from the SEDDS depends on many formulation-related parameters, such as surfactant concentration, oil/surfactant ratio, polarity of the emulsion, droplet size and charge, all of which in essence determine the self-emulsification ability. Thus, only very specific pharmaceutical excipient combinations will lead to efficient self-emulsifying systems. Although many studies have been carried out, there are few drug products on the pharmaceutical market formulated as SEDDS confirming the difficulty of formulating hydrophobic drug compounds into such formulations. At present, there are four drug products, Sandimmune and Sandimmun Neoral (cyclosporin A), Norvir (ritonavir), and Fortovase (saquinavir) on the pharmaceutical market, the active compounds of which have been formulated into specific SEDDS. Significant improvement in the oral bioavailability of these drug compounds has been demonstrated for each case. The fact that almost 40% of the new drug compounds are hydrophobic in nature implies that studies with SEDDS will continue, and more drug compounds formulated as SEDDS will reach the pharmaceutical market in the future.