Sie sind auf Seite 1von 11

J Pharm Innov (2014) 9:95–105

DOI 10.1007/s12247-014-9176-3

RESEARCH ARTICLE

Optimization of Chronomodulated Delivery System Coated


with a Blend of Ethyl Cellulose and Eudragit L100 by Central
Composite Design: In Vitro and In Vivo Evaluation
Om Prakash Ranjan & Usha Y. Nayak & M. Sreenivasa Reddy &
Swapnil J. Dengale & Prashant B. Musmade &
Nayanabhirama Udupa

Published online: 27 March 2014


# Springer Science+Business Media New York 2014

Abstract increase in water uptake and polymer leaching. As ex-


Introduction In this study, we present the development pected, with increasing coating level, lag time increased
of a chronomodulated delivery system consisting of a and release rate decreased due to the increased diffusion
fast-swelling tablet core containing montelukast sodium pathways. In vivo study revealed comparative pharmaco-
coated with a blend of different ratios of ethyl cellulose kinetic profiles of core tablets and pulsatile release tablets
(gastrointestinal tract (GIT)-insoluble polymer) and (PRTs); however, Tmax of 2 h for core tablets and 6 h for
Eudragit L100 (enteric polymer). Montelukast sodium PRTs were observed.
is a leukotriene receptor antagonist commonly pre- Conclusion Thus, designed PRTs were found to be suitable in
scribed for patients of asthma and allergic rhinitis. Asth- treating episodic attack of asthma in early morning and asso-
ma and allergic rhinitis share a common core patho- ciated allergic rhinitis.
physiology and have almost similar temporal pattern in
their occurrence or exacerbation of their respective
symptoms, suggesting a role for chronotherapy. Keywords Chronotherapy . Pulsatile release . Lag time .
Methods The developed formulation was optimized statisti- Enteric polymer . Central composite design
cally using central composite design to achieve desired release
profile. The coated tablets were studied for water uptake,
bursting time, and in vitro release study.
Results The bursting time (lag time) of coated tablet was Introduction
affected by the pH of buffer, molarity of ions, and con-
centration of different types of surfactant in dissolution The temporal control of drug delivery has been of attention to
media. With increasing percentage of Eudragit L100 in achieve improved drug therapies [1] for many diseased con-
coating composition, the lag time decreased and release ditions, such as asthma, hypertension, rheumatoid arthritis,
rates significantly increased—could be attributed due to etc. [13]. Chronotherapeutics is the discipline concerned with
administration of medicines according to inherent activities of
a disease over a certain period of time [3]. Bronchial asthma
O. P. Ranjan : U. Y. Nayak : M. S. Reddy (*)
Department of Pharmaceutics, Manipal College of Pharmaceutical
and allergic rhinitis share a common core pathophysiology
Sciences, Manipal University, Manipal 576104, Karnataka, India and exhibit similar temporal pattern in the occurrence or
e-mail: ms.reddy@manipal.edu exacerbation of their respective symptoms suggesting a role
for chronotherapy [8]. In patients with asthma, symptoms
S. J. Dengale : P. B. Musmade
generally worsen during the early hours of the morning,
Department of Pharmaceutical Quality Assurance, Manipal College
of Pharmaceutical Sciences, Manipal University, and pulmonary function often deteriorates at the same
Manipal 576104, Karnataka, India time [7, 8]; also, the symptoms of allergic rhinitis such
as sneezing, nasal rhinorrhea, red itchy eyes, nasal pru-
N. Udupa
ritus, and nasal congestion were found to occur most
Department of Pharmaceutical Management, Manipal College of
Pharmaceutical Sciences, Manipal University, recurrently before breakfast in the morning and least
Manipal 576104, Karnataka, India frequently in the middle of the day [8].
96 J Pharm Innov (2014) 9:95–105

Montelukast sodium (MKS) is leukotriene receptor antag- with the least number of experiments and selection of
onists most commonly used in the treatment of asthma and the optimal composition for achieving the presetting
allergic rhinitis [4]. Presently, montelukast sodium is available target [2].
in market as conventional immediate release film-coated tab-
lets. It is indicated for the treatment of asthma, exercise-
induced bronchoconstriction, and for the relief of allergic Material and Methods
rhinitis. As the marketed formulation releases the drug imme-
diately within an hour of intake, so there is a need for Materials
chronomodulated drug delivery system of montelukast sodi-
um, which will lag the drug release till midnight and then MKS was obtained from the Lupin Limited, (Pune, India).
releases the drug, which will take care of asthma as well as Cross-carmellose sodium (Ac-di-sol) was supplied by FMC
allergic rhinitis. BioPolymer. Low-substituted hydroxypropyl cellulose (L-
Polymeric film coatings are frequently used to control HPC; LH 11) and microcrystalline cellulose (Avicel 112) were
and modify drug release profile from solid pharmaceu- obtained from the Signet. EC (Ethocel standard 10 premium)
tical dosage forms. Fan et al. [1] used polymeric blend supplied by Dow Chemical Company, USA and Eudragit
of gastrointestinal tract (GIT)-insoluble polymer (ethyl L100 (Eud L100) by Evonic Degussa were used for film
cellulose, EC) and enteric polymer (Eudragit L) to coat coating. Dibutyl pthalate (DBP) was obtained from Sigma
diltiazem HCl tablets to achieve pulsatile drug delivery Aldrich, magnesium stearate from Himedia Laboratories Pvt.
and rapid drug release after a pre-determined lag time in Ltd., Mumbai, and talc from Lobachemie Pvt. Ltd., Mumbai,
the intestine [5]. Soni et al. [9] attempted to achieve the India. All other chemicals were of analytical grade.
chronotherapy for bronchial asthma by utilizing dual
approach for the effective colonic delivery of theophyl- Preformulation studies
line using pH-dependent solubility behavior of Eudragit
and susceptibility of guar gum to colonic environment. Fourier Transform Infrared Spectroscopy
Yassin et al. [12] designed a chronotherapeutic delivery
system of theophylline with high potential benefits in Fourier transform infrared spectroscopy (FTIR) was carried
treating nocturnal asthma. out using a Shimadzu FT-IR 8300 Spectrophotometer
In this paper, we present the development of a (Shimadzu, Tokyo, Japan) in the wavelength region of 4,000
chronomodulated delivery system consisting of a fast- to 400 cm−1. The process consisted of dispersing a sample
swelling tablet core containing montelukast sodium coated (drug alone or mixture of drug and excipients) in KBr and
with a blend of different ratios of EC (GIT-insoluble compressing into discs by applying pressure in a hydraulic
polymer) and Eudragit L100 (enteric polymer). Eudragit L as press. The pellet was placed in the light path, and the spectrum
a polymer, only dissolved at pH above 6, was selected to be the was obtained [6].
coating material along with ethyl cellulose to fit the mentioned
purpose [1]. The lag time before drug release is mainly Differential Scanning Calorimetry
controlled by the properties of the outer polymeric coat-
ing, and the water uptake and the swelling behavior of Differential scanning calorimetry (DSC) was performed using
the swelling core [13]. The major objectives of the a DSC-60 (Shimadzu, Tokyo, Japan) calorimeter. The instru-
present study were: (i) to study the effect of polymer ment comprised of a calorimeter (DSC 60), flow controller
blend ratio and coating level of a GIT-insoluble and an (FCL 60), thermal analyzer (TA 60), and operating software
enteric/polymer on lag time of drug release and (ii) to (TA 60). The samples (drug alone or drug–polymer mixture)
optimize the polymer blend ratio in coating to get a were heated in sealed aluminum pans under nitrogen flow
desired lag time with wide range of drug release profile. (30 mL/min) at a scanning rate of 5 °C min up to 250 °C.
A comprehensible approach for fruitful appraisal and Indium was used as a reference [6].
optimization of the formulation parameters is necessary.
Central composite design (CCD), one of the techniques Solubility Studies
in response surface methodology (RSM), is used for
optimization of formulation development. RSM, support- Solubility studies of MKS were performed in different media
ed by statistical software, is a well-established approach (with or without Tween 80) such as water, 0.1 N HCl, acetate
for pharmaceutical formulation development and optimi- buffer pH 4.5, and phosphate buffer pH 6.8 and pH 7.4. An
zation allowing extraction of maximal information out excess amount of drug was added in different vials with
of few well-designed experiments. CCD is suitable for different media and shaken in water bath shaker for 24 h at
pharmaceutical blending problems allowing investigation 37 °C (Remi Equipments Ltd., Bangalore, India). The media
J Pharm Innov (2014) 9:95–105 97

were filtered using a 0.45-μm filter paper and were estimated h  i


by validated high-performance liquid chromatography Carr0 s indexð%Þ ¼ ρtapped −ρbulk =ρtapped  100
(HPLC) method.
Hausnerratio ¼ ρtapped =ρbulk

Analytical Method for Estimation of Montelukast Sodium

Analysis of montelukast sodium was done by HPLC (LC- Preparation of Core Tablets
2010CHT, Shimadzu, Kyoto, Japan), equipped with low-
pressure quaternary gradient pump along with dual wave- Direct compression method was employed for preparation of
length UV detector, column oven, and autosampler. Chro- fast disintegrating core tablet. The blends for compression
matographic data were recorded and processed using LC were prepared by mixing drug, cross-carmellose sodium,
solution 1.24SP1 software. Phenomenex®Luna C18 (250.0× and L-HPC with Avicel pH 112 for 10 min. Purified talc,
4.6 mm, 5 μm) column was used for the estimation of and magnesium stearate, were added to above blend and
montelukast sodium. The mobile phase consisted of acetoni- further mixed for 5 min. The final blend was compressed to
trile and 20 mM ammonium acetate buffer (pH 5±0.02) with a 100 mg weight tablet using 6.4 mm concave punch on rotary
volumetric ratio of 80:20 at a flow rate of 1.0 mL/min. The tablet press (10 station tablet compression machine, Cadmach,
detector was set at 345 nm, and injection volume was 25 μL. Ahmedabad, India). The hardness of the tablets (n=6) was
The calibration curve was generated for concentrations rang- determined by using the Monsanto hardness tester Electrolab,
ing from 0.5 to 25.0 μg/mL. Mumbai, India. The thickness and diameter of the tablets (n=
3) were measured using Vernier caliper. The friability (%) of
the tablets was determined using Friabilator (USP EF-2),
Preparation of Pulsatile Release Tablets Electrolab, Mumbai, India. The disintegration test of core
tablets was carried out by the disintegration tester USP (ED-
Micromeritic Properties 2AL), Electrolab, Mumbai, India. The drug content was de-
termined after crushing core tablet (n=3); 100 mg of powder
The composition of the core tablets is given in Table 1. The was dissolved in 100 mL of methanol. The solution was
blends for core tablets were prepared by mixing drug, cross- filtered through a 0.45-μm filter and analyzed by validated
carmellose sodium, L-HPC with Avicel pH 112, and lactose HPLC method after sufficient dilution.
anhydrous (with or without) for 10 min. Purified talc, magne-
sium stearate, and silicon dioxide were added to each blend
and further mixed for 5 min. The final blend ready for com- Coating of Core Tablets
pression was characterized for angle of repose, Carr’s index,
and Hausner ratio. The angle of repose of formulation blend The Pharma R&D coater (model: Delux, Ideal Cures Pvt. Ltd.,
was determined by the fixed funnel method [6]. The bulk Mumbai, India) was used to coat core tablets. The 4 % (w/v)
density (BD) and tapped densities (TD) were determined by solution of EC/Eudragit L100 in different ratios (60:40, 70:30,
using a density apparatus (Serwell Instruments, Bangalore, 80:20, and 90:10) was prepared in solvent mixture of isopro-
India). The Carr's index (%) and the Hausner ratio were pyl alcohol, acetone, and water. To avoid sticking, talc (20 %
calculated as follows [6]: (w/w) based on total polymer mass) was added to the coating
solutions and DBP (20 % (w/w) based on total polymer mass)
as plasticizer. The process parameters for the coatings were as
Table 1 Composition of core tablets follows: product temperature 35+2 °C, pan speed 40 rpm,
pump speed 1–2 rpm, and atomization pressure 1.0 bar. Sub-
Ingredients Quantity/tablet (mg) sequent to the coating, the tablets were further cured for
Batch No. C-1 C-2 C-3 C-4
30 min in coating pan at 40 °C to remove residual solvent.
Montelukast sodium 10.40 10.40 10.40 10.40
Microcrystalline cellulose 50.00 84.00 82.00 72.00 Optimization Using Experimental Design
Lactose anhydrous 34.00 – – –
Cross-carmellose sodium 3.00 3.00 5.00 5.00 For systematic optimization of developed formulations, the
L-Hydroxypropyl cellulose – – – 10.00 experimental design methodology was employed by CCD
Purified talc 1.60 1.60 1.60 1.60 with the help of Design Expert software 8.0.5 (Stat-Ease
Magnesium stearate 1.00 1.00 1.00 1.00 Inc., Minneapolis, MN, USA). A CCD with α = 1 was
employed in the development of montelukast pulsatile release
98 J Pharm Innov (2014) 9:95–105

Table 2 Formulation trials as per experimental design and coded values of pH, types, and concentration of surfactant and ionic con-
Batch No. X1: % of Eud L100 X2: % coat centration of media on bursting time was studied.
weight The media of different pH 1.2, 4.5, 6.8, and 7.4 were
prepared as per USP. The bursting time of coated tablet in
F1 0 1 different pH media was determined by visual observation.
F2 0 −1 Different media were prepared in molar concentrations (20,
F3 −1 1 50, and 100 mM) containing the electrolyte stated, and pH of
F4 1 0 the media was adjusted to 6.8 either with 1 M HCl or 1 M
F5 −1 −1 NaOH. The bursting time of coated tablet in different media
F6 0 0 was determined by visual observation.
F7 0 0 The media of pH 6.8 were prepared as per USP. The effect
F8 −1 0 of different surfactants (SLS and Tween 80) in different con-
F9 0 0 centrations (0.1, 0.2, 0.5, and 1.0 % (w/v)) on bursting time
F 10 1 −1 was observed by visual observation. The bursting time of
F 11 1 1 optimized formulation in dissolution media was determined
F 12 0 0 [medium 0.1 N HCl (with 0.5 % (w/v) Tween 80) for 2 h and
F 13 0 0 phosphate buffer pH 6.8 (with 0.5 % (w/v) Tween 80)] in USP
Factors Coded values type II (paddle) apparatus at 37 °C, rotation speed 75 rpm.
−1 0 1
X1: Eudragit L100 (%, w/w) 20 30 40 In Vitro Dissolution Studies
X2: coat weight (%, w/w) 8 10 12
Responses Constraints The in vitro dissolution study was carried out using paddle
Y1 =lag time (h) 4.5 type (USP type II) dissolution apparatus (TDT-06P,
Y2 =release at 6 h (%) Maximize Electrolab, Mumbai, India. The in vitro dissolution study of
core tablets was performed in 900 mL of phosphate buffer
pH 6.8 with 0.5 % (w/v) Tween 80, whereas for coated tablet
tablet formulations. Eudragit L100 (X1) and total coat weight in 500 mL 0.1 N HCl (with 0.5 % (w/v) Tween 80) for the first
(X2) were selected as factors (independent variables), while 2 h, followed by 900 mL of phosphate buffer pH 6.8 (with
release lag time (Y1) and percentage drug release at 6 h (Y2) 0.5 % (w/v) Tween 80). The temperature of dissolution medi-
were selected as obtained responses (dependent variables). um was maintained at 37±0.5 °C and paddle speed of 75 rpm.
Each factor was studied at three different levels (−1, 0, and At different time intervals, 5 mL of sample was withdrawn
+1). Table 2 summarizes an account of the 13 formulation and analyzed by UV–visible spectrophotometer. At each time
batches studied, their factor combinations, and the trans- of withdrawal, 5 mL of fresh corresponding medium was
lation of the coded levels to the experimental units replaced into the dissolution vessel. The MKS has shown
employed during the study. maximum absorbance at wavelength of 385 and 345 nm in
0.1 N HCl and phosphate buffer pH 6.8, respectively, so
Characterization of Pulsatile Release Tablets corresponding wavelength was used for sample analysis.

Water Uptake Study Scanning Electron Microscopy

Water uptake by the pulsatile release tablets was examined at the Photographs of the outer surface of the coating film of pulsa-
condition of drug release test. The media were 0.1 N HCl (with tile release tablets were taken using a scanning electron mi-
0.5 % (w/v) Tween 80) for 2 h and phosphate buffer pH 6.8 croscope (Zeiss, EVO 18, Carl Zeiss SMT Ltd, UK) at initial
(with 0.5 % (w/v) Tween 80) for further till just before the and after predetermined time points (2, 3, and 4 h) in dissolu-
rupture time. The weight of pulsatile release tablets was mea- tion media [1].
sured with elapse of time at predetermined time interval (n=3).
Pharmacokinetic Studies
In Vitro Bursting Time Study
The pharmacokinetic study was carried out in male rabbits to
The bursting time is time point, when the outer coating rup- compare the pharmacokinetic parameters of optimized PRTs
tured and was noted (n=3) by visual observation of the of montelukast sodium with immediate release core tablet.
pulsatile release tablets in a USP type II (paddle) apparatus Study protocol was approved by the Institutional Animal
at 37 °C, rotation speed 75 rpm in different media. The effect Ethical Committee (IAEC/KMC/75/2011-2012). The
J Pharm Innov (2014) 9:95–105 99

overnight fasted rabbits of weight 2.5 kg were divided Differential Scanning Calorimetry
into two groups (n=3) and treated orally (10 mg MKS
per tablets) as follows: The thermal curve of MKS showed absence of sharp endo-
thermic peak up to 250 °C (Fig. 2) which confirms that MKS
Group I Core tablets is amorphous in nature. MKS has shown glass transition (Tg)
Group II PRTs (F 14) at 62.1 °C. There was no significant change in glass transition
Tg (MKS 62.1 °C and drug–polymer mixture 60.2 °C) or
After a single oral administration of core tablet and PRTs, absence of sharp endothermic peak of MKS in drug polymer
0.6 mL of blood samples was collected from the marginal ear mixture, confirming no interaction.
vein at different time-points into tubes containing EDTA. The
plasma was separated immediately using spinwin centrifugation Solubility Study
at 10,000 rpm for 10 min and stored at −70 °C until analysis.
The solubility study result is shown in Table 3. The solubility
of MKS is observed to be pH dependent. It has shown higher
Bioanalysis of MKS in Rabbit Plasma
solubility at higher pH, and significant improvement in solu-
bility was observed along with surfactant, Tween 80 in differ-
The RP-HPLC method was used for bioanalysis of MKS in
ent pH media.
rabbit plasma. The HPLC system used was HPLC LC-
2010CHT (Shimadzu, Kyoto, Japan) equipped with low-
Preparation of Pulsatile Release Tablets
pressure quaternary gradient pump along with dual wave-
length UV detector. The mobile phase consisting of a mixture
Micromeritic Properties
of acetonitrile and ammonium acetate buffer (20 mM, pH
adjusted to 5.5±0.02) in the ratio 80:20 (v/v) was delivered
Angle of repose, Carr’s index, and Hausner ratio have been
isocratically at a flow rate of 1.0 mL min−1. The detection
used most popularly in predicting flow characteristics of pow-
wavelength was 345 nm.
der. For direct compression of materials, it is required to
Extraction was accomplished by liquid–liquid extraction
possess good flow and compacting properties. Values for
method adding 1.5 mL MTBE following gentle vortex for
angle of repose 31–35° (USP) generally indicate good flow
15 min on spinwin. The mixture was then centrifuged for
property. Hausner ratio of less than 1.25 and Carr's index of
10 min at 10,000 rpm at 4 °C. The organic supernatant was
less than 20 indicate fair to good flow (USP). The prepared
transferred to a clean glass vial and evaporated using nitrogen
formulation mixtures showed good flow properties.
gas, TurboVap® LV (15 psi) at 50 °C for 5 min. The residue
was then reconstituted with 150 μL mobile phase mixture, and
Evaluation of Core Tablets
50 μL was injected to HPLC. Lercanidipine HCL was used as
internal standard. Calibration curve was plotted in the concen-
Different tablet formulations of MKS were prepared by direct
tration range of 20–5,000 ng/mL. The developed method was
compression. Tablets were studied for hardness, disintegration,
validated as per USFDA guidelines.
friability, and weight variation. To improve disintegration,
super disintegrant was added in the formulation. Ac-di-sol is
a water insoluble fibrous nature cross-carmellose sodium type
Result and Discussion super-disintegrant exhibiting good water uptake, high capillary
activity, and rapid hydration properties. The fibrous nature of
Preformulation Studies Ac-di-sol provides many sites for fluid uptake and gives it
excellent wicking capabilities. Core tablet (C-4) was consid-
Fourier Transform Infrared Spectroscopy ered as optimized formulation based on rapid disintegration
and was selected for further studies and coating process. The
The FTIR spectra of pure MKS, MKS–polymer mixture, and hardness of optimized formulation (C-4) was found to be 30–
PRTs are shown in Fig. 1. Pure MKS has shown a broad peak 35 N, and disintegration time was less than 1 min. The
of tertiary hydroxyl group at around 3,443 cm−1 and a strong friability was less than 0.5 % which is within the acceptance
peak of carboxylic acid group near 1,568 and 1,600 cm−1. limit.
Numbers of aromatic C–H peaks are also observed between
2,900 to 3,000 cm−1. These are the characteristic absorption Statistical Analysis of Experimental Data
peak of MKS. These bands are of indicative value to elucidate
drug–polymer interactions which are appeared unchanged in The results of the experimental design indicated that this
MKS–polymer mixture and PRTs confirming no interaction. system was highly influenced by the % Eudragit L100 in
100 J Pharm Innov (2014) 9:95–105

Fig 1 FTIR of pure MKS and


PRTs
J Pharm Innov (2014) 9:95–105 101

Table 4 Presentation of values and responses in CCD (13 possible


combinations)

Batch No. X1: % of X2: coat weight Y1: lag Y2: release
Eud L100 in % time (h) at 6 h (%)

F1 30 12 5.2 78.45
F2 30 8 3.4 96.54
F3 20 12 6.3 0.92
F4 40 10 2.8 98.35
F5 20 8 4.2 89.39
F6 30 10 4.3 91.41
F7 30 10 4.2 88.73
F8 20 10 5.3 79.92
F9 30 10 4.4 84.29
F 10 40 8 2.3 97.65
F 11 40 12 3.4 97.65
F 12 30 10 4.3 89.15
F 13 30 10 4.3 92.35

The regression coefficients for each term in the regression


Fig 2 DSC thermogram of pure MKS and PRTs model are summarized as follows:

Y 1 ¼ 4:30−1:22X 1 þ 0:83X 2 −0:25X 1 X 2 −0:25X 21


coating composition and total weight gain in % which resulted Y 2 ¼ þ91:40 þ 20:57X 1 −17:76X 2 þ 22:12X 1 X 2 −7:80X 21 −9:44X 22
in desired lag time and release profile. Responses obtained
from all 13 formulations (Table 4) were fed into Design-
A positive value in the regression equation indicates direct
Expert® trial version 8 software for design of experiments
relationship, while a negative value an inverse relationship
using CCD.
between the factor and the response [11, 2]. For estimation of
A polynomial equation with six coefficients was produced
the significance of the model, the analysis of variance
for the account of the measured responses as a function of the
was determined as per the provision of Design Expert
process variables and expressed in as follows:
software. A model is considered significant if the p
value (significance probability value) is less than 0.05
Y i ¼ A0 þ A1 X 1 þ A2 X 2 þ A3 X 1 X 2 þ A4 X 21 þ A5 X 22 [10, 2]. Based on the largest r2 value for all responses,
quadratic model was suggested for both lag time and
release at 6 h. The three-dimensional response surface
where Y is the measured response, A0 is an intercept, and A1– plots for the effect of factors (X1: % Eudragit L100; X2:
A5 are the regression coefficients; X1, X2 represents the main % Coat weight) on responses related to lag time (Y1)
effect; X12, X22 the quadratic effect and X1X2 interaction effect. and drug release at 6 h (Y2) are shown Fig. 3.

Table 3 Solubility study of MKS in different media


Optimization and Validation
Media Solubility (mg/
mL)
After generating the polynomial equations relating the
0.1 N HCl pH 1.2 0.0040 factors and responses, a further optimization process
Acetate buffer pH 4.5 0.0132 was undertaken with desirable characteristics to probe
Phosphate buffer pH 6.8 0.0145 the optimal formula solution of PRTs which depended
Phosphate buffer pH 7.4 0.0212 on the prescriptive criteria of 4.5 h of drug release lag
0.1 N HCl pH 1.2 with 0.5 % (w/v) Tween 80 0.0069 time and maximize the release at 6 h. The list of
Acetate buffer pH 4.6 with 0.5 % (w/v) Tween 80 0.0235 solutions was sorted with the highest desirability first;
Phosphate buffer pH 6.8 with 0.5 % (w/v) Tween 80 3.70 solutions that meet the criteria are reported in Table 5.
Phosphate buffer pH 7.4 with 0.5 % (w/v) Tween 80 6.79 Desirability for optimization of PRTs of MKS coated
with EC/Eudragit L100 is shown in Fig. 4.
102 J Pharm Innov (2014) 9:95–105

Fig 4 Desirability for optimization of PRTs of MKS coated with EC/


Eudragit L100

Characterization of Pulsatile Release Tablets

Water Uptake Study

The water imbibition rate and extent were strongly affected by


polymer blend ratio. With increasing Eudragit L content, the
water uptake increased in both media (Fig. 5), which can be
attributed to the higher hydrophilicity of Eud L100 compared
to ethyl cellulose [5].

In Vitro Bursting Time Study


Fig 3 Response surface plots for the effect of percentage Eudragit L100
and coat weight on responses related to a lag time and b drug release at The burst time of the PRTs in dissolution media was
6h
investigated and could be mainly controlled by the
coating level (%) and EC/Eudragit ratio of outer poly-
mer coating (Fig. 6a). The bursting time increased with
The optimum formulation was selected based on the higher coating levels and decreasing Eudragit content in
drug release lag time of 4.5 h, maximize % drug release outer coating composition because of the increased me-
at 6 h; satisfying these parameters, the first solution chanical strength of the coating membrane and the re-
from Table 5 was chosen as optimized formulation with duced medium permeation rate.
the highest desirability of 0.947 (F 14). The optimum The bursting time was affected by the pH of buffer
formulation was evaluated for all the evaluation param- (Fig. 6b), molarity of ions (Fig. 6c) and concentration
eters. The observed values for the optimized formulation of different types of surfactant (Fig. 6d) in dissolution
were compared with the predicted values. The results
were found to be close to the predicted values, which
confirm the practicability of the model. The comparative Table 6 Comparison of predicted and observed responses for the statis-
tically optimized formulation F 14
table is shown in Table 6.
Formulation Response Observed Predicted Relative
error (%)

Table 5 Solutions suggested by Design Expert that meet the criteria F 14 Drug release lag 4.60 4.5 2.22
required for PRTs time in h (Y1)
% drug release at 83.52 88.33 5.75
S. No. HPMC Coat wt. Lag time Release at Desirability 6 h (Y2)
coded/actual coded/actual (h) 6 h (%) F 15 Drug release lag 4.75 4.5 5.55
time in h (Y1)
F 14 −0.48 −0.34 4.5 88.33 0.947 % drug release at 81.26 88.33 8.00
F 15 −0.47 −0.33 4.5 88.33 0.947 6 h (Y2)
J Pharm Innov (2014) 9:95–105 103

Fig 7 Release profile of core tablet (C-4) and PRTs (F-14)


Fig 5 Water uptake study by PRTs coated with EC/Eud L100 in different
ratios at 10 % coating level

media. In media of different pH, the coated tablets (F 8) In Vitro Dissolution Study
have shown significant difference in bursting time may-
be due to pH-dependent solubility of Eudragit polymer. The drug release of core tablet (C-4) and optimized
At higher pH, decrease in bursting time was observed batch (F 14) is shown in Fig. 7. The release study of
maybe because of pH-dependent solubility of Eudragit different batches was carried to study the effect of EC/
polymer used in coating composition. It was observed Eudragit L100 blend ratio at different coating levels.
that at higher molar concentration, tablets burst fast as The effects of the EC/Eudragit L100 blend ratio
shown in Fig. 6c. Surfactant plays a major role in (90:10, 80:20, 70:30, and 60:40) and coating level (8,
release profile; SLS has shown less bursting time com- 10, and 12 % w/w) on the in vitro release study of
pared to Tween 80 may be due to fast wettability of PRTs are shown in Fig. 8a, b, respectively. A large
tablet surface leading to fast erosion of polymer coating range of drug release profile with different lag times
because of greater reduction in surface tension of media. can be obtained by varying the Eud L100 content in
As concentration of surfactant (SLS and Tween 80) polymer blend and thickness of coating membrane. As
increased from 0.1 to 1.0 % (w/v), there was significant expected, the release rate decreased with increasing
reduction in bursting time observed (Fig. 6d). coating level, due to the increased diffusion pathways.

Fig 6 Effect of a % of Eudragit L100 and coat weight, b pH of media, c molarity of ions, and d surfactant concentration on bursting time of PRTs coated
with EC/Eud L100 (80:20) at 10 % (w/w) coating level
104 J Pharm Innov (2014) 9:95–105

Fig 10 Pharmacokinetic parameters of core tablet (C-4) and PRTs (F-14)

Eudragit L100 [5] leading to formation of a large num-


ber of pores on polymeric coating membrane.

Scanning Electron Microscopy

The optimized formulations were subjected to scanning elec-


tron microscopy (SEM) studies, and resulting images are
Fig 8 Release profile of a EC/Eud L100 coated tablet in different ratios
shown below in Fig. 9. From these studies, it is clearly evident
of 10 % (w/w) coating level and b EC/Eud L100 (80:20) coated tablet at
different weight gain levels that tablet surface was smooth as initial even after 2 h disso-
lution in pH 0.1 N HCl because of no solubility of Eudragit
L100 in acidic media, whereas after 3 and 4 h tablet has shown
With increasing Eudragit L100 content, the release rate a considerable change in surface morphology due to formation
increased (irrespective of the coating level), which can of pores and cracks maybe because of leaching of Eudragit
be attributed to the higher permeability and leaching of L100 in buffer pH 6.8 from outer coating membrane.

Fig 9 Scanning electron


microscopy images of optimized
formulation at a 0 h, b 2 h, c 3 h,
and d 4 h in dissolution media
J Pharm Innov (2014) 9:95–105 105

Table 7 Pharmacokinetic parameters of MKS after oral administration of of the coating film. The pharmacokinetic study indicates that
core tablet and PRTs (n=3)
the developed system can release the drug in the GI tract in a
Parameters Core tablet (C-4) PRTs (F 14) manner similar to that in vitro as supported by pharmacoki-
netic study. Thus, the designed device can be considered as
Cmax (ng/mL) 607.985+29.11 632.6+39.16 promising delivery system in management of asthma and
Tmax (h) 2 6 associated allergic rhinitis.
AUC0–t (ng h/mL) 5,438.43+317.63 6,677.78+511.25*
AUC0–∞ (ng h/mL) 5,732.39+376.83 7,612.91+551.47*
Acknowledgments The authors are grateful to acknowledge Manipal
MRT (h) 9.0+0.96 13.9+1.43* College of Pharmaceutical Sciences, Manipal University, Manipal for
Vd (L) 4.5+0.71 4.5+0.76 providing infrastructure facility and ICMR, New Delhi, India for financial
CL (L/h) 0.698+0.065 0.525+0.056* support.
T1/2 (h) 4.5+0.44 5.96+0.56*
Conflicts of Interest Authors have no conflict of interest.
Ke (1/h) 0.154+0.041 0.116+0.037

Data were represented as the mean±SD


*p<0.05, significant difference was observed References

Pharmacokinetic Study 1. Fan TY, Wei SL, Yan WW, Chen DB, Li J. An investigation of
pulsatile release tablets with ethylcellulose and Eudragit L as film
The developed bioanalytical method was validated as per coating materials and cross-linked polyvinylpyrrolidone in the core
USFDA guidelines. The method was found to be linear in tablets. J Control Release. 2001;77:245–51.
2. Hao J, Wang F, Wang X, Zhang D, Bi Y, Gao Y, et al. Development
concentration range of 20–5,000 ng/mL with a correlation and optimization of baicalin-loaded solid lipid nanoparticles prepared
coefficient (r2) of 0.9981. The plasma concentration of MKS by coacervation method using central composite design. Eur J Pharm
against time is shown Fig. 10 following the oral administra- Sci. 2012;47:497–505.
tion of the core tablet and PRTs (F-14). The pharmacokinetic 3. Jha N, Bapat S. Chronobiology and chronotherapeutics. Kathmandu
Univ Med J. 2004;2:384–8.
parameters were calculated from the plasma concentration– 4. Lane SJ. Leukotriene antagonism in asthma and rhinitis. Respir Med.
time curves using pk solution and presented in Table 7. Ab- 1998;92:795–809.
sorption of MKS from core tablet (Tmax, 2 h) after oral admin- 5. Lecomte F, Siepmann J, Walther M, MacRae RJ, Bodmeier R.
istration was immediate and rapid as compared to PRTs (Tmax, Blends of enteric and GIT-insoluble polymers used for film coating:
physicochemical characterization and drug release patterns. J Control
6 h) which is desirable for chronotherapy. The PRTs has Release. 2003;89:457–71.
shown in vivo lag time of 4 h. The AUC values of the PRTs 6. Nayak UY, Shavi GV, Nayak Y, Averinen RK, Mutalik S, Reddy SM,
were slightly higher than that of core tablet indicating better et al. Chronotherapeutic drug delivery for early morning surge in
extent of absorption. There was no significant difference blood pressure: a programmable delivery system. J Control Release.
2009;136:125–31.
observed for maximum absorption concentration (Cmax) be- 7. Nainwal N. Chronotherapeutics—a chronopharmaceutical approach
tween core tablets and PRTs. The mean residential time to drug delivery in the treatment of asthma. J Control Release.
(MRT) and half-life (T1/2) were observed more for PRTs than 2012;163(3):353–60.
core tablet because of slow elimination rate (Ke) and clearance 8. Smolensky MH, Lemmer B, Reinberg AE. Chronobiology and chro-
notherapy of allergic rhinitis and bronchial asthma. Adv Drug Deliv
(CL) which is attributed due to extended release pattern of Rev. 2007;59:852–82.
PRTs. Statistical testing of differences between the mean 9. Soni ML, Namdeo KP, Jain SK, Gupta M, Dangi JS, Kumar M, et al.
values was performed by t test with p<0.05 as the minimum pH-enzyme di-dependent chronotherapeutic drug delivery sys-
level of significance. tem of theophylline for nocturnal asthma. Chem Pharm Bull.
2011;59:191–5.
10. Varshosaz J, Ghaffari S, Khoshayand MR, Atyabi F, Azarmi S,
Kobarfard F. Development and optimization of solid lipid nanopar-
Conclusion ticles of amikacin by central composite design. J Liposome Res.
2010;20:97–104.
11. Verma S, Lan Y, Gokhale R, Burgess DJ. Quality by design approach
In the present study, pulsatile release tablets of montelukast to understand the process of nanosuspension preparation. Int J Pharm.
sodium were developed and optimized statistically using CCD 2009;377:185–98.
to release the drug after a desired lag period of time. The 12. Yassin AE, Aodah AH, Al-Suwayeh S, Taha EI. Theophylline colon
designed systems consist of fast disintegrating core coated specific tablets for chronotherapeutic treatment of nocturnal asthma.
Pharm Dev Technol. 2012;17:712–8.
with a blend of GIT-insoluble (ethyl cellulose) and enteric 13. Zhu Y, Zheng L. Development and mathematical simulation of
polymer (Eudragit L100). The lag time could be controlled by theophylline pulsatile release tablets. Drug Dev Ind Pharm.
Eudragit L100 content in coating composition and thickness 2005;31:1009–17.

Das könnte Ihnen auch gefallen