You are on page 1of 9

Powder Technology 256 (2014) 336–344

Contents lists available at ScienceDirect

Powder Technology
journal homepage: www.elsevier.com/locate/powtec

Improved oral delivery of clonazepam through liquisolid powder


compact formulations: In-vitro and ex-vivo characterization
Krishna Sanka, Sravanthi Poienti, Abdul Bari Mohd, Prakash V. Diwan ⁎
Department of Pharmaceutics, School of Pharmacy, (Formerly Lalitha College of Pharmacy) Anurag Group of Institutions, Hyderabad, 500088 AP, India

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

Article history: The liquisolid powder compacts (LSPCs) proved to be the potential solubility improvement strategy for efficient
Received 30 August 2013 oral delivery of BCS class II and IV drugs. Henceforth, an attempt was made to improve the oral delivery of BCS
Received in revised form 3 February 2014 class II drug clonazepam (CLZ) by formulating into a novel LSPCs. Solubility studies were conducted in different
Accepted 9 February 2014
liquid vehicles, namely propylene glycol, span 20 and span 80. The LSPCs were formulated using propylene glycol
Available online 17 February 2014
as non volatile solvent. The effect of different formulation variables on LSPCs performance was evaluated using 32
Keywords:
factorial design. The selected independent variables were % of clonazepam in propylene glycol (X1) and % of
Clonazepam sodium starch glycolate (X2) and dependent variables were disintegration time (YDT) and % cumulative drug
Factorial design release at 15th minute (YQ15). LSPCs of CLZ formulated with propylene glycol at optimum drug concentration
Liquisolid powder compacts produced high dissolution profile with acceptable tablet properties. Fourier transform infra-red spectroscopy
Solubility (FTIR) studies revealed that there was no interaction between drug and polymers, differential scanning calorim-
Dissolution rate etry (DSC) and X-Ray Diffraction (XRD) indicated conversion of crystalline to amorphous form of the CLZ. Further
the permeation studies carried out in isolated rat intestine revealed that potential of LSPCs for enhanced perme-
ation of CLZ across rat intestinal barrier. The increase in permeation of clonazepam from LSPCs formulation across
rat intestine suggests the potential of LSPC formulation for improved oral delivery of CLZ.
© 2014 Elsevier B.V. All rigths reserved.

1. Introduction described by Spireas [34,35] are composed of a non-volatile, water mis-


cible liquid vehicle, solid drug particles, and selected excipients (carrier
Clonazepam (CLZ) is an anticonvulsant chloro-nitrobenzodiazepine and coating materials). The liquid portion, which can be a liquid drug, a
that enhances the gamma-amino butyric acid (GABA) receptor response. drug suspension, or a drug solution in suitable non-volatile liquid vehi-
CLZ exerts its action by binding to the benzodiazepine site of the GABA cles, is incorporated into the porous carrier material. Once the carrier is
receptors, causing an enhancement of the electric effect of GABA on the saturated with liquid, a liquid layer is formed on the particle surface,
neuron when it binds to the receptor. It is poorly soluble in water (at which is instantly adsorbed by the fine coating particles. Thus, an appar-
25 °C b 0.1 mg/ml) [1]. According to the biopharmaceutical classification ently dry, free flowing, and compressible powder is obtained. Besides
(BCS), CLZ is in class II, which consists of drugs of low solubility and drug release enhancement, the liquisolid approach is a promising tech-
high permeability. The rate of oral absorption of these class II drugs is nique because of the simple manufacturing process, low production
often controlled by the dissolution rate in the gastrointestinal (GI) tract costs, and the possibility of industrial manufacture due to the good
[2]. Therefore, together with permeability, the solubility and dissolution flow and compaction properties of the liquisolid formulations. To
behavior of a drug are key determinants of its oral bioavailability. calculate the required amount of powder excipients (carrier and coating
Different approaches have been attempted to increase water solubility materials), a mathematical approach for the formulation of liquisolid
of poorly soluble drugs, such as conversion of a crystalline molecule to its systems has been developed by Spireas [36]. Depending on the excipient
amorphous state [3,4] particle size reduction via micronization [5–10], co- ratio R of the powder substrate, an acceptably flowing and compressible
grinding [11–13], inclusion complexation [14–16], solid dispersions liquisolid powder can be obtained only if a maximum liquid load, named
[17–19], self emulsifying drug delivery system [20–22], nanosuspensions “liquid load factor” (Lf), is not exceeded. The terms “acceptable flow”
[23–25] and adsorption of drugs to hydrophilic silica aerogels [26–28]. and “acceptable compressibility” imply the desired and thus preselected
One of the most promising strategies for release enhancement is the flow and compaction properties, which must be met by the final
Liquisolid Powder Compacts (LSPCs) [29–35]. Liquisolid systems as liquisolid formulation. R represents the ratio between the mass of the
carrier (Q) and the coating (q) materials present in the formulation:
⁎ Corresponding author. Tel.: +91 9849005903 (Cell).
E-mail addresses: sreekrishnaspips@gmail.com (K. Sanka), diwanpv@gmail.com
(P.V. Diwan). R ¼ Q =q: ð1Þ

http://dx.doi.org/10.1016/j.powtec.2014.02.026
0032-5910/© 2014 Elsevier B.V. All rigths reserved.
K. Sanka et al. / Powder Technology 256 (2014) 336–344 337

Lf represents the ratio between the mass of the liquid portion W and Table 1
the carrier materials Q: Observed responses from 32 factorial design.

Formula Independent Dependent variables


code variables
L f ¼ W=Q : ð2Þ X1 X2 YQ15 (%) YDT (sec)

F1 −1 −1 55.59 ± 4.10 110 ± 5


F2 −1 0 77.49 ± 2.48 55.4 ± 4
With the desired amount of liquid, the amount of carrier and coating F3a −1 +1 90.27 ± 7.07 30.9 ± 2
material can be calculated if the liquid load factor Lf is known. F4 0 −1 52.63 ± 5.46 108.00 ± 6
The aim of this study was to increase the solubility and dissolution F5 0 0 62.84 ± 10.24 21.00 ± 2
F6 0 +1 63.11 ± 9.46 11.50 ± 3
properties of CLZ using liquisolid technique. In this study CLZ, a poorly F7 +1 −1 19.94 ± 0.67 95.00 ± 8
water-soluble, anticonvulsant drug was formulated into liquisolid com- F8 +1 0 48.45 ± 0.44 24.90 ± 6
pacts consisting of similar powder excipients and propylene glycol with F9 +1 +1 51.78 ± 1.10 11.00 ± 4
different drug concentrations in their liquid medications and super
Coded values Actual values
disintegrating agent in the final LSPCs. Further ex-vivo studies have
been carried out to assess the permeation of CLZ from LSPCs using iso- X1 (%) X2 (%)

lated rat intestine. −1 10 0


0 50 2
+1 90 4
2. Materials and methods
a
Indicates optimized formulation; X1% of CLZ in non-volatile solvent; X2% of sodium
2.1. Materials starch glycolate; YQ15% Cumulative drug release at 15th min; YDT disintegration time in
sec.

CLZ was a gift sample from Centaur Pharmaceuticals, India. Micro-


crystalline cellulose PH 102 (MCC PH 102) and sodium starch glycolate
were obtained from SD-Fine Chemicals Ltd., Mumbai, India. AEROSIL®
200 was obtained from Southern Chemicals, Hyderabad, India. Propylene statistical model incorporating interactive and polynomial terms was
glycol was provided by the Universal Laboratories Pvt., Ltd., Mumbai, used to evaluate the response (Eq. (3))
India. HPLC grade methanol and acetonitrile were obtained from Merck
Specialities Pvt., Ltd., Mumbai, India. HPLC grade orthophosphoric acid Y ¼ b0 þ b1 X1 þ b2 X2 þ b12 X1 X2 þ b11 X1 X1 þ b22 X2 X2 ð3Þ
and triethanolamine (TEA) are obtained from Thermo Fisher Scientific
India Pvt., Ltd., Mumbai. India. All other reagents and chemicals used
where Y is the dependent variable, b0 is the arithmetic mean of the 9
were of analytical grade.
trials, and bi is the estimated coefficient for the factor Xi. The X1 and X2
are the main effects; represent the average results of changing one factor
2.2. Methods
at a time from its low to high value. The interaction terms (X1X2) show
how the response changes when two factors are simultaneously varied.
2.2.1. HPLC analysis of clonazepam
The polynomial terms (X1X1 and X2X2) are included to investigate
The samples were assayed for clonazepam by using RP-HPLC method
nonlinearity.
reported earlier [37]. HPLC was equipped LC-20AD pumps, DGU-20A3
degasser, SPD-M20A Photo Diode Array Detector, LC Solutions 1.25 soft-
2.2.4. Preparation of liquisolid powder compacts
ware, Rheodyne injector fitted with 20 μL capacity (Shimadzu, Nakagyo-
LSPCs were formulated according to the 32 factorial design (Table 1).
Ku, Kyoto, Japan) and a Luna (Phenomenex®, USA) C18 column (5 μm,
The weighed amount of drug substance was dispersed in the calculated
4.6 mm × 250 mm). Isocratic elution was carried out at a flow rate
amount of non-volatile solvent (propylene glycol) termed as liquid
1 ml/min. The mobile phase consisted of 60:40 (%v/v) acetonitrile and
vehicle. The mixing procedure was conducted in three stages as
0.1%v/v aqueous TEA (pH 5.5), respectively and the detection wave-
described by Spireas et al. [42]. Firstly, the weighed quantity of carrier
length was set at 254 nm. Aliquots of 20 μL of each sample were injected
material (Microcrystalline cellulose PH 102) was blended with liquid
onto the column.
medication in order to evenly distribute the liquid medication into the
powder. Then, the calculated amount of coating material (AEROSIL®
2.2.2. Solubility studies
200) was added to the system under continuous triturating in a mortar.
Solubility studies were conducted by placing an excess amount of
Finally, to the above binary mixture super disintegrating agent
drug in each vehicle in a 2 ml Clicklock microcentrifuge tubes (Tarsons
i.e., sodium starch glycolate was added and mixed for a period of 10 to
Products Pvt. Ltd., Kolkata, India) containing 1.5 ml of the vehicle. The
20 min producing the final liquisolid powder which was compressed
mixture was vortexed and kept for 48 h at 25 °C in an orbital shaking
using table top pilot scale 10 station rotary tablet Rimek mini press-I
incubator (Remi Electrotechnik Ltd, CIS-2413L, Mumbai, India) to
(M/S Karnavati Engineering Ltd., Gujarat, India)
facilitate the solubilization [34,38]. The samples were centrifuged at
3000 rpm for 15 min to remove the undissolved drug. The supernatant
2.2.5. Micromeritic properties of prepared pre compressed liquisolid
was taken and the concentration of drug in each vehicle was quantified
powder systems
by RP-HPLC method.
A fixed funnel method was used to study the angle of repose (θ). A
weighed quantity of samples was transferred in to graduated cylinder
2.2.3. Experimental design for designing liquisolid powder compacts
from each batch to determine the bulk and tap density using USP-I
A 32 factorial design consists of two independent variables at three
tapped density tester (TD 1025, Labindia Instruments, Mumbai, India).
levels. According to this design, 9 runs were conducted in total [39–41].
The experiments were performed in triplicate. The parameters selected
The independent variables selected for this study were X1, % of CLZ
to study flow properties were determined using Eqs. (4), (5), (6), (7), (8)
in non-volatile solvent (propylene glycol); X2, % of superdisintegrant
(sodium starch glycolate). The dependent variables were Y1, amount of
drug release at 15th min (YQ15); and disintegration time (YDT). The levels h
Tan θ ¼ ð4Þ
of independent and dependent variables were listed in Table 1. A r
338 K. Sanka et al. / Powder Technology 256 (2014) 336–344

Mass 2.2.9. Solid state characterization


Bulk density ðσ bÞ ¼ ð5Þ
Poured volume
2.2.9.1. Differential scanning calorimetry (DSC). DSC thermograms of Pure
CLZ, MCC PH 102, AEROSIL® 200, sodium starch glycolate and opti-
mized CLZ loaded LSPC were analyzed by DSC (Model: SIIO-6300,
Mass Japan). Samples (3–5 mg weighed to precision of 0.005 mg) were
Tap density ðσtÞ ¼ ð6Þ
Tapped volume placed in aluminium pans and the lids were crimped using a crimper.
Thermal behavior of the samples was investigated at a scanning rate
of 10 °C/min, using nitrogen as blanket gas covering a temperature
range of 0 to 300 °C [45].
ðσ t – σ bÞ
Carr’s Indexð%Þ ¼  100 ð7Þ
σt 2.2.9.2. X-ray diffraction (XRD). For further characterization of the crys-
talline state, the X-ray diffraction (XRD) patterns were determined
for Pure CLZ, MCC PH 102, AEROSIL® 200, sodium starch glycolate and
optimized CLZ loaded LSPC. The X-ray powder diffraction was obtained
0 ðσ tÞ
Hausner s ratio ¼ ð8Þ using X-RD (Model: Stereoscans S120, Cambridge, UK). The samples
ðσ bÞ
are loaded on to the diffractometer, then exposed to Cu-Kα radiation
(40 kV × 30 mA) and scanned over a range of 2θ, values from 10 to
80°at a scan rate of 0.05°/0.4 s [45].

2.2.6. Post compression studies of the clonazepam liquisolid powder 2.2.10. Ex-vivo permeation studies
compacts Male Wistar rats weighed between 180 and 200 g used in the study.
The prepared LSPCs further evaluated for drug content (n = 6) The animals were housed in separate cages and maintained under con-
thickness (n = 20), hardness (n = 20), friability (n = 20) and weight trolled condition of temperature and the rats had free access to water
variation (n = 20). Drug content in each batch was determined by trit- and food until they were sacrificed. The study was conducted with the
urating 20 tablets in a mortar with the help of pestle and the amount prior approval of the Institutional Animal Ethical Committee, School of
equivalent to one average tablet was weighed and dissolved in 250 ml Pharmacy, Anurag Group of Institutions, Hyderabad, India.
volumetric flask containing 100 ml of solvent mixture of methanol The rats were sacrificed with excess ether inhalation. The abdomen
and water (1:1). The flask was placed in orbital shaking instrument was opened and a segment of the ileum was removed and flushed
(Remi, Electrotechnik Ltd., Vasai, India). Temperature and rpm were with Krebs–Ringer solution to remove the mucus and adhered intestinal
adjusted to 27 °C and 150, respectively. Shaking was continued for contents. One end of the intestine segment was tied and dose equivalent
72 h, and later was filtered through a 0.45 μm Millipore membrane filter crushed LSPCs of optimized formulation were introduced into the
paper. The few ml of initial filtrate was discarded and sufficient volume lumen and were tightly closed. The tissue was placed in a 250 ml beaker
of filtrate was collected. The amount of drug was estimated by injecting with continuous aeration and maintained at a temperature of 37 °C.
appropriated diluted samples in to HPLC. The thickness of CLZ liquisolid The beaker consists of 100 ml of phosphate buffer (pH 6.8). At predeter-
powder compacts was determined by using digital Vernier calipers mined time intervals (5, 10, 15, 30, 45, 90 and 120 min) an aliquot of 1
(CD-6″CSX, Mitutoyo Corp., Kawasaki, Japan) which permits accurate ml was collected and replenished with equal volume of medium. The
measurements and provides information on the variation between samples were treated with an equal volume of methanol, centrifuged
tablets. The hardness of the LSPCs was evaluated using Monsanto hard- and the supernatant was quantified for CLZ using HPLC. Control (pure
ness tester (MHT-20, Kshitij International, Ambala, India), the mean CLZ) and marketed formulation were also included in the study for
hardness of each formula was determined. The friability of prepared comparison [46,47].
formulations was determined using Roche friabilator (FT 1020, Labindia, The cumulative amount of CLZ permeated (Q) was plotted against
Mumbai, India). The disintegration time of the LSPCs was measured time (t). The steady state flux (Jss) was calculated from the slope of
using disintegration tester (DT 1000, Labindia, Mumbai, India). Weight linear portion of the cumulative amount permeated per unit area vs.
variation test was performed according to the official method (USP) time plot. The permeability coefficient (Kp) of the CLZ through intestine
using an electronic balance (ATX224, Shimadzu, Japan). was calculated by dividing steady state flux with initial concentration of
CLZ donor compartment. The enhancement ratio (ER) was calculated by
2.2.7. In vitro dissolution studies using the following equation:
In vitro dissolution was carried out using USP II dissolution apparatus
(DS 8000, Labindia, Mumbai, India). LSPCs were placed in the vessel Jss of liquisolid formulation
ER ¼ : ð9Þ
containing 900 ml of HPLC water as suitable dissolution media at Jss of control
37 ± 0.5 °C and paddle speed of 75 rpm. At predetermined time
intervals (5, 10, 15, 30, 45, 60, 90 and 120 min), an aliquot of 5 ml was
withdrawn and replaced with an equal volume of fresh dissolution 2.2.11. Stability studies
medium to maintain constant volume [43,44]. The samples were filtered Stability studies were carried out according to the ICH guidelines by
through the 0.45 μm Millipore membrane filter (NYLON 66, Axiva, Lab storing the CLZ loaded LSPC formulation at 40 °C/75% ± 5% RH for a
filters, Delhi, India) and assessed for drug release using HPLC. period of three months. The samples were withdrawn after 90 days
and analyzed for assay, disintegration time and in vitro release studies
2.2.8. Fourier transforms infrared (FTIR) spectroscopy [48–51].
Fourier transform infrared spectra of the Pure CLZ, MCC PH 102,
AEROSIL® 200, sodium starch glycolate and optimized CLZ loaded LSPC 3. Results and discussion
were obtained using Fourier Transform Infrared Spectrophotometer
(Bruker, Alpha-T, Ettlingen, Germany). Samples were prepared using 3.1. Solubility studies
KBr disks by means of hydraulic pellet press at a pressure of 10 t. The
samples were scanned from 4000 to 400 Cm−1. Spectra are analyzed The solubility of CLZ in propylene glycol, span 80, span 20 and water
by OPUS-6.5 software. is given in Table 2. The results show that the solubility of the CLZ is very
K. Sanka et al. / Powder Technology 256 (2014) 336–344 339

Table 2 Table 4
Solubility of CLZ in various non-volatile solvents. Regression coefficients for the responses.

S.No. Solvent Solubility (mg/ml) S.No. Coefficients of regression parameters

1 Propylene glycol 6.48 ± 0.081 b0 b1 b2 b12 b11 b22 R2-value p-value


2 Span 80 3.46 ± 0.022
YQ15 58.01 −17.2 12.83 – – – 0.8958 0.0011
3 Span 20 0.44 ± 0.010
YDT 28.63 −10.98 −43.27 −1.22 7.7 27.3 0.9810 0.0087
4 Water 0.14 ± 0.070
YQ15% cumulative drug release at 15th min; YDT disintegration time in sec.

poor in water (0.14 mg/ml) whereas, propylene glycol enhanced the polynomial equation can be used to draw a conclusion after considering
solubility of CLZ to the level of 6.48 mg/ml. Solubility in span 80 and the magnitude of coefficient and the mathematical sign it carries (posi-
span 20 was found to be 3.46 and 0.44, respectively. Thus, from the tive or negative). Results of ANOVA were depicted in Table 5 and the in-
above results of solubility studies among the non-volatile solvents vitro dissolution release profile of 9 trials vs pure CLZ was shown in
tested, propylene glycol could be a better choice as a solvent for the Fig. 1. To demonstrate the effect of the % of CLZ in propylene glycol
formulation of LSPCs. and % of sodium starch glycolate, the response surface plots were
generated for the dependent variables YQ15, YDT using Design-Expert®
Software (Stat-Ease Inc., Minneapolis).
3.2. Precompression studies of the prepared liquisolid systems
3.3.1. Effect of formulation variables on release profile (YQ15)
The flowability of powder material plays a major role in the produc-
The in vitro performance of CLZ loaded LSPCs showed release profile
tion of pharmaceutical dosage forms in order to get a uniform feed as
depends upon the formulation variables i.e., % of CLZ in propylene glycol
well as reproducible filling of tablet dies, otherwise dose variations
(X1) and % of sodium starch glycolate (X2). The results showed that
will occur. Flow properties of the liquisolid powder systems were
optimized formulation of CLZ loaded LSPC demonstrated more than
studied by determining angle of repose (θ), Carr's index, Hausner's
85% drug release at Q15, when compared with that of pure drug which
ratio, bulk density (σ b) and tapped density (σ t) [52] and their results
demonstrated less than 10% drug release at Q15 at P b 0.05. The effect
are presented in Table 3.
of formulation variables on the amount of drug release at 15th minute
As angle of repose (θ) is a characteristic of internal friction or
(YQ15), is given in Eq. (10).
cohesion of the particles, the value of the angle of repose will be high
if the powder is cohesive and low if the powder is non-cohesive. As
presented in Table 3, angle of repose was in the range from 25.15 ± YQ 15 ¼ 58:01−17:20X1 þ 12:83X2 : ð10Þ
0.14 to 28.07 ± 0.08 which indicates good flow ability of the powder
from all formulations. The values of bulk and tapped density were
found to be in the range from 0.511 ± 0.001 to 0.573 ± 0.003 g/cm3, In Eq. (10), b1 bears negative sign indicating an increase in the % of
0.596 ± 0.02 to 0.657 ± 0.011 g/cm3, respectively. The Carr's index CLZ in propylene glycol (X1) decreased the amount of drug release at
(%) and Hausner's ratio was found to be in the range from 11.10 ± 15th min, b2 bears positive sign in the same equations indicating an
0.10 to 17.16 ± 0.15, 1.11 ± 0.09 to 1.19 ± 0.0, respectively. These increase in the % of sodium starch glycolate (X2) increase in amount of
values indicate that the micromeritic properties of all powder blends drug release at 15th min.
from each batch are within the limits and they exhibit good flow prop-
erty and compressibility. 3.3.2. Effect of formulation variables on disintegration time (YDT)
The effect of formulation variables i.e., CLZ % propylene glycol (X1)
and % of sodium starch glycolate (X2) on disintegration time (YDT), is
3.3. Application of experimental design for designing liquisolid tablets
given in Eq. (11).

In the present investigation, the effect of % of CLZ in propylene glycol


(X1) and % of sodium starch glycolate (X2) on % cumulative drug release YDT ¼ 28:63−10:98X1 −43:27X2 −1:22X1 X2 þ 7:7 X1 X1 þ 27:30 X2 X2 :
at 15th min (YQ15) and disintegration time (YDT) is studied using 32 ð11Þ
factorial design, revealed wide variation (Table 1). The data clearly
indicates that the dependent variables are strongly dependent on the
independent variables. The fitted equation relating the response YQ15, In the above equation, b1 and b2 bear negative sign indicating an
YDT to the transformed factor are shown in Eqs. (10) and (11). The increase in the % of CLZ in propylene glycol (X1) and % of sodium starch
value of correlation coefficient (Table 4) indicates good fit. The glycolate (X2) decreased the disintegration time, b12 bears negative sign

Table 3
Micromeritic properties of prepared pre-compression liquisolid powders.

Formula code Angle of repose (θ) Bulk density (gm/cm3) Tapped density (gm/cm3) Carr's index (%) Hausner's ratio

F1 27.31 ± 0.3 0.573 ± 0.003 0.647 ± 0.006 11.10 ± 0.10 1.11 ± 0.09
F2 26.55 ± 0.49 0.541 ± 0.002 0.656 ± 0.003 17.16 ± 0.15 1.17 ± 0.06
F3 25.64 ± 0.56 0.563 ± 0.005 0.657 ± 0.011 13.91 ± 0.03 1.15 ± 0.005
F4 27.59 ± 0.09 0.524 ± 0.014 0.59 ± 0.0015 12.07 ± 0.11 1.126 ± 0.005
F5 26.51 ± 0.04 0.526 ± 0.005 0.596 ± 0.02 11.39 ± 0.01 1.123 ± 0.005
F6 28.07 ± 0.08 0.526 ± 0.015 0.630 ± 0.001 15.95 ± 0.066 1.19 ± 0.01
F7 26.61 ± 0.03 0.522 ± 0.001 0.627 ± 0.002 17.02 ± 0.06 1.17 ± 0.05
F8 27.36 ± 0.32 0.563 ± 0.029 0.655 ± 0.009 11.57 ± 0.15 1.13 ± 0.005
F9 25.15 ± 0.14 0.511 ± 0.001 0.601 ± 0.007 14.24 ± 0.22 1.15 ± 0.005
340 K. Sanka et al. / Powder Technology 256 (2014) 336–344

Table 5
Results of analysis of variance for measured response.

Parameters Degree of freedom Sum square Mean square f-Value p-Value

For YQ15
Regression 2 2762.52 1381.26 25.79 0.0011
Residual 6 321.33 53.56
Total 8 3083.85

For YDT
Regression 5 13560.05 2712.01 31.01 0.0087
Residual 3 262.37 87.46
Total 8 1322.42

YQ15% cumulative drug release at 15th min; YDT disintegration time in sec.

in the same equation indicating the interaction effect of X1X2 decreased


the disintegration time, b11 and b22 bear positive sign which indicates
that the interaction effect of X1X1 and X2X2 increased the disintegration
time.
The relationship between dependent and independent variables
was further elucidated using contour plots and 3D plots. The effects of
X1 and X2 and their interaction on YQ15 and YDT are given in Fig. 2. It
could be seen that increase in % of CLZ in propylene glycol and % of
sodium starch glycolate had a negative effect on YDT.

3.4. Post compression studies of the clonazepam liquisolid tablets

The CLZ loaded LSPCs were evaluated to determine the post com-
pression parameters, the results were shown in Table 6. The thickness
of the LSPCs was found to be ranged from 4.98 ± 0.1067 to 5.15 ±
0.125 mm. The hardness of the liquisolid compacts was measured by
Monsanto hardness tester and was found to be ranged from 4.76 ± Fig. 2. Response surface plot (A) and contour plot (B) showing effect of X1 and X2 on
response YDT.
0.28 to 5.16 ± 0.35 kg/cm2. The friability was found to be ranged from
0.498 ± 0.166 to 0.691 ± 0.193% which was below 1% for all the
formulations, which is an indication of good mechanical resistance of
the compacts. The weight variation for different formulations (F1–F9)
showed satisfactory results as per the United States Pharmacopeia CLZ loaded LSPC formulations, assay studies were performed which
(USP) limit (average weight ± 5%). For estimation of drug in different showed satisfactory results as per the USP i.e., CLZ tablets must
contain not less than 90% and not more than 110% of the labeled amount
of CLZ.

3.5. Fourier transform infrared spectroscopy

Drug-excipient interactions play a crucial role with respect to


the stability and potency of the drug. Fourier transform infrared
spectroscopy (FTIR) techniques have been used to study the physical
and chemical interaction between drug and excipients used. In the
spectra of optimized CLZ loaded formulation of LSPCs (Fig. 3, Table 7),
the peak characteristics to the excipients were present at almost
same positions, whereas CLZ peaks were also present, but at a reduced
intensity of absorption, indicating the trapping of CLZ inside the carrier
matrix. None of the spectra showed any peaks other than those
assigned to CLZ and excipients, which indicate that there is no differ-
ence between the IR patterns of the optimized formulation of CLZ and
pure drug.

3.6. Solid state characterization

3.6.1. Differential scanning calorimetry


Differential scanning calorimetry (DSC) curve for CLZ showed a
sharp endothermic (melting) peak at 241 °C (Fig. 4, curve A). The CLZ
peak was absent in the DSC curve of Optimized formulation of CLZ
liquisolid powder compacts, indicating that the drug in final formula-
Fig. 1. Dissolution release profiles of pure CLZ vs CLZ loaded LSPC formulations (F1–F9). tion was less crystalline (more amorphous). Conversion of crystalline
K. Sanka et al. / Powder Technology 256 (2014) 336–344 341

Table 6
Physical evaluation parameters of CLZ liquisolid powder compacts.

Formula code Thickness (mm) Hardness (kg/cm2) Friability (%) Weight variation (mg) Assay (%)

F1 5.11 ± 0.1067 5.03 ± 0.05 0.691 ± 0.193 448.2 ± 1.62 95 ± 3.12


F2 5.15 ± 0.125 5.06 ± 0.11 0.648 ± 0.165 447.5 ± 1.94 97 ± 2.84
F3 5.06 ± 0.124 5 ± 0.2 0.55 ± 0.157 450 ± 0.67 98.00 ± 2.02
F4 5.03 ± 0.047 5.16 ± 0.35 0.498 ± 0.166 448.2 ± 1.83 100 ± 1.03
F5 5.05 ± 0.095 4.93 ± 0.11 0.683 ± 0.186 448.7 ± 1.28 99.25 ± 3.41
F6 5.15 ± 0.047 4.83 ± 0.28 0.647 ± 0.194 447.9 ± 1.92 97.82 ± 2.03
F7 4.98 ± 0.1067 4.76 ± 0.25 0.594 ± 0.224 446.8 ± 1.75 98.33 ± 1.22
F8 5.11 ± 0.106 5 ± 0.2 0.645 ± 0.163 448.1 ± 1.55 99.88 ± 2.09
F9 5.08 ± 0.134 4.83 ± 0.28 0.610 ± 0.154 449 ± 1.38 97.26 ± 1.71

form of drug to amorphous form indicates enhanced solubility, which 3.6.2. X-Ray powder diffraction analysis
led to improved dissolution release profile of the drug by formulation X-ray powder diffraction (XRD) analysis was used to assess the
of LSPCs. degree of crystallinity of the LSPC constituents. CLZ showed major
peaks at 2θ values of 11.86, 14.79, 15.06, 18.27, 18.54, 20.07, 20.49,
22.84, 23.95, 24.34, 26.11, 27.16, 27.48, 27.81, 30.23° (Fig. 5, curve A).
Analysis of XRD patterns of the CLZ loaded optimized formulation
(Fig. 5, curve E) indicated that all the major peaks corresponding to
CLZ disappeared except peak at 22.84°, which show conversion of
crystalline form of drug to amorphous form due to addition of the
excipients to the formulation. A fall in degree of crystallinity means an
improvement in the amorphousness of a sample. Hence, from the
above discussion it was concluded that the LSPC technique resulted in
an amorphous form of CLZ with suitable excipients, which led to
improved dissolution release profile.

3.7. Ex-vivo intestinal permeation study

The rate and extent of absorption are not only dependent on the
dissolution rate rather than permeation across the gastrointestinal
tract but are also limiting factors for absorption. Therefore to have an
insight on the ability of liquisolid tablet formulations for improved
absorption, ex-vivo permeation study was carried out using the rat
intestinal segment. Since majority of the drugs get absorbed in vivo
from the small intestine it is more relevant to use the rat tissue [28].
Henceforth, we have used ileum portion to assess the potential of
LSPCs for improving the permeation of CLZ across the intestinal barrier.
Ex-vivo permeation data did not show significant (p N 0.05) differ-
ence between marketed formulation and free CLZ. The cumulative
amount permeated (CAP) across the intestine was calculated and repre-
sented in Fig. 6. The cumulative amount of CLZ permeated from control
to marketed formulation was 511 ± 28 and 405 ± 6 μg, respectively
and was significantly increased to 1492 ± 88 μg with optimized formu-
lation (P b 0.05) (Table 8). The flux was also increased significantly

Table 7
Characteristic peaks of free CLZ and CLZ in optimized LSPC.

S.No Compound Type of vibration Peaks (cm−1)

1 Pure drug CLZ N\H stretching 3106


Ar\H stretch 2971
C\H stretch
Ar\C\C stretch 1695
(in–ring)
Aromatics 1615
2 Optimized LSPC N\H stretching 3383
Ar\H stretch 2904
C\H stretch 1637
Aromatic
C\C stretch 1614
(in–ring)
Fig. 3. FTIR Spectra of A) Pure CLZ B) MCC PH 102 C) AEROSIL® 200 D) Sodium starch
Aromatics
glycolate E) Optimized CLZ loaded LSPCs.
342 K. Sanka et al. / Powder Technology 256 (2014) 336–344

Fig. 4. DSC thermograms of A) Pure CLZ B) MCC PH 102 C) AEROSIL® 200 D) Sodium
starch glycolate E) Optimized CLZ loaded LSPCs.

with optimized CLZ loaded LSPC formulation compared to control and


marketed formulation (P b 0.05). The ER was above 1 (3.80 ± 0.1) for
optimized CLZ loaded LSPCs (F3) and indicates enhanced permeation
compared to pure CLZ.

3.8. Stability study


Fig. 5. XRD patterns of A) pure CLZ B) MCC PH 102 C) AEROSIL® 200 D) Sodium starch
glycolate E) Optimized CLZ loaded LSPCs.
Stability study of optimized CLZ loaded LSPC formulations was
performed at 40 °C/75% ± 5% RH for three months. Samples were with-
drawn after 3 months and showed no significance (p N 0.05) change in
assay (%), and disintegration time (YDT). The similarity factor (f2) for the
optimized formulation was found to be 67.80 (Table 9), which indicates
good similarity of dissolution release profile, before and after stability
study.

4. Conclusion

In conclusion, the present study showed that LSPC technique could


be a promising strategy in improving dissolution of poorly water soluble
CLZ and wettability was improved by making a suspension in propylene
glycol, the water soluble, nonvolatile solvent. LSPCs could be prepared
using MCC PH 102 as a carrier, and AEROSIL® 200 as a coating material.
The FTIR studies revealed that excipients were compatible with the
drug. DSC and XRD studies showed that there is a decrease in crystallin-
ity of the CLZ in liquisolid compact formulation. A fall in crystallinity
means improved dissolution release profile. The optimized formulation
(F3) showed higher dissolution rate when compared with that of Fig. 6. Ex-vivo permeation of CLZ across rat intestine from Free CLZ, LSPCs and market
pure drug. formulation.
K. Sanka et al. / Powder Technology 256 (2014) 336–344 343

Table 8 [13] M. Barzegar-Jalali, H. Valizadeh, M.R.S. Shadbad, K. Adibkia, G. Mohammadi, A.


Permeation parameters of CLZ from LSPC formulation across rat intestine. Farahani, Z. Arash, A. Nokhodchi, Cogrinding as an approach to enhance dissolution
rate of a poorly water soluble drug (gliclazide), Powder Technol. 197 (2010)
Formulation Jss (μg/cm−4/h) Kp(cm/h) × 10−7 ER 150–158.
[14] L.P. Ruan, B.Y. Yu, G.M. Fu, D.N. Zhu, Improving the solubility of ampelopsin by solid
Pure CLZ 0.2041 ± 0.220 0.1020 ± 0.0024 – dispersions and inclusion complexes, J. Pharm. Biomed. Anal. 38 (2005) 457–464.
F3 formulation 0.7750 ± 0.0096 0.3875 ± 0.0026 3.8 ± 0.1 [15] N.P. Sapkal, V.A. Kilor, K.P. Bhusari, A.S. Daud, Evaluation of some methods for
Market formulation 0.2212 ± 0.0016 0.1106 ± 0.0091 1.08 ± 0.03 preparing gliclazide-ß-cyclodextrin inclusion complexes, Trop. J. Pharm. Res. 6
(2007) 833–840.
[16] B. Yang, J. Lin, Y. Chen, Y. Liu, Artemether/hydroxy propyl β-cyclodextrin host–guest
system: characterization, phase solubility and inclusion mode, Bioorg. Med. Chem.
17 (2009) 6311–6317.
[17] M. Barzegar-Jalali, S. Dastmalchi, Kinetic analysis of chlorpropamide dissolution
Table 9 from solid dispersions, Drug Dev. Ind. Pharm. 33 (2007) 63–70.
Results of stability study of optimized formulation. [18] O.I. Corrigan, Mechanisms of dissolution of fast release solid dispersions, Drug Dev.
Ind. Pharm. 11 (1985) 697–724.
Time (min) % cumulative drug releasea
[19] H. Valizadeh, P. Zakeri-Milani, M. Barzegar-Jalali, G. Mohammadi, M.A. Danesh-
Initial After storage at 40 °C/75 ± 5% RH Bahreini, K. Adibkia, A. Nokhodchi, Preparation and characterization of solid disper-
for 90 days sions of piroxicam with hydrophilic carriers, Drug Dev. Ind. Pharm. 33 (2007)
45–56.
0 0 0 [20] J.D. Wei, H.O. Ho, C.H. Chen, W.T. Ke, E.T. Chen, M.T. Sheu, Characterisation of
5 85.79 ± 1.05 83.38 ± 1.12 fenofibrate dissolution delivered by a self-microemulsifying drug delivery system,
10 88.75 ± 4.60 86.28 ± 2.23 J. Pharm. Pharmacol. 62 (2010) 1685–1696.
15b 90.27 ± 7.07 91.33 ± 6.06 [21] D. Patel, K.K. Sawant, Self micro-emulsifying drug delivery system: formulation
30 89.19 ± 0.5 90.22 ± 1.65 development and biopharmaceutical evaluation of lipophilic drugs, Curr. Drug
45 82.09 ± 11.5 89.58 ± 5.22 Delivery 6 (2009) 419–424.
60 82.39 ± 3.17 89.43 ± 1.88 [22] C.W. Pouton, Lipid formulations for oral administration of drugs: nonemulsifying,
self-emulsifying and ‘self-microemulsifying’ drug delivery systems, Eur. J. Pharm.
90 83.21 ± 3.18 88.24 ± 2.53
Sci. 11 (2000) S93–S98.
120 77.69 ± 8.01 85.26 ± 4.54
[23] R.H. Muller, B.H.L. Bohm, J. Grau, Nanosuspensions: a formulation approach for
Assay (%)c 98.00 ± 2.02 99.04 ± 2.07
poorly soluble and poorly bioavailable drugs, in: D. Wise (Ed.), Handbook of
YDT (sec)c 30.90 ± 2.00 32.22 ± 3.00 Pharmaceutical Controlled Release Technology, Marcel Dekker Inc., New York, NY,
a
f2 N 50 (67.80). 2000, pp. 345–357.
b
YQ15% cumulative drug release at 15th min; YDT disintegration time in sec. [24] E. Merisko-Liversidge, G.G. Liversidge, E.R. Cooper, Nanosizing: a formulation
c approach for poorly-water-soluble compounds, Eur. J. Pharm. Sci. 18 (2003)
P N 0.05.
113–120.
[25] J.I. Jinno, N. Kamada, M. Miyake, K. Yamada, T. Mukai, M. Odomi, H. Toguchi, G.G.
Liversidge, K. Higaki, T. Kimura, Effect of particle size reduction on dissolution and
oral absorption of a poorly water-soluble drug, cilostazol, in beagle dogs, J. Control.
Acknowledgments Release 111 (2006) 56–64.
[26] I. Smirnova, M. Tuerk, R. Wischumerski, M.A. Wahl, Comparison of different
methods for enhancing the dissolution rate of poorly soluble drugs: case of griseo-
All authors are thankful to Dr. Rajeshwar Reddy, Chairman, School of fulvin, Eng. Life Sci. 5 (2005) 277–280.
Pharmacy (Formerly Lalitha College of Pharmacy), Anurag Group of [27] I. Smirnova, S. Suttiruengwong, W. Arlt, Feasibility study of hydrophilic and hydro-
Institutions for providing research facilities. Appreciation is also extended phobic silica aerogels as drug delivery systems, J. Non-Cryst. Solids 350 (2004) 54–60.
[28] U. Maver, A. Godec, M. Bele, O. Planinsek, M. Gaberscek, S. Srcic, J. Jamnik, Novel
to Dr. A. Madhu Babu and A. Padmanabha Rao for their invaluable hybrid silica xerogels for stabilization and controlled release of drug, Int. J. Pharm.
assistance. 330 (2007) 164–174.
[29] Y. Javadzadeh, M.R. Siahi-Shadbad, M. Barzegar-Jalali, A. Nokhodchi, Enhancement
of dissolution rate of piroxicam using liquisolid compacts, Farmaco 60 (2005)
References 361–365.
[30] A. Nokhodchi, Y. Javadzadeh, M.R. Siahi-Shadbad, M. Barzegar-Jalali, The effect of
[1] M. Moneghini, I. Kikie, D. Vomovich, B. Perissutti, J. Filipovie-Grei, Processing of type and concentration of vehicles on the dissolution rate of a poorly soluble drug
carbamazepine-PEG 4000 solid dispersions with supercritical carbon dioxide: prep- (indomethacin) from liquisolid compacts, J. Pharm. Pharm. Sci. 8 (2005) 18–25.
aration, characterization, and in vitro dissolution, Int. J. Pharm. 222 (2001) 129–138. [31] S.K. Emmadi, K. Sanka, A.P. Potu, R. Jukanti, S. Bandariand, P.R. Veerareddy, Formu-
[2] G.L. Amidon, H. Lennernas, J.R. Crison, A theoretical basis for biopharmaceutics lation and pharmacodynamic evaluation of meloxicam liquisolid compacts, Lat. Am.
drugs classification: the correlation of in-vitro drug product dissolution and in-vivo J. Pharm. 29 (8) (2010) 1303–1310.
bioavailability, Pharm. Res. 12 (1999) 413–420. [32] N. Tiong, A.A. Elkordy, Effects of liquisolid formulations on dissolution of naproxen,
[3] B.C. Hancock, Disordered drug delivery: destiny, dynamics and the Deborah num- Eur. J. Pharm. Biopharm. 73 (2009) 373–384.
ber, J. Pharm. Pharmacol. 54 (2002) 737–746. [33] A. Nokhodchi, C.M. Hentzschel, C.S. Leopold, Drug release from liquisolid systems:
[4] A. Nokhodchi, The effect of type and concentration of vehicles on the dissolution speed it up, slow it down, Expert Opin. Drug Deliv. 8 (2011) 1–15.
rate of a poorly soluble drug (indomethacin) from liquisolid compacts, J. Pharm. [34] S. Spireas, S. Sadu, Enhancement of prednisolone dissolution properties using
Pharm. Sci. 8 (2005) 18–25. liquisolid compacts, Int. J. Pharm. 166 (1998) 177–188.
[5] N. Rasenack, H. Hartenhauer, B.W. Muller, Microcrystals for dissolution rate [35] S. Spireas, S. Sadu, R. Grover, In vitro release evaluation of hydro-cortisone liquisolid
enhancement of poorly water-soluble drugs, Int. J. Pharm. 254 (2003) 137–145. tablets, J. Pharm. Sci. 87 (1998) 867–872.
[6] T.L. Rogers, K.P. Johnston, R.O. Williams, Micronised powders of a poorly water- [36] S. Spireas, Liquisolid Systems and Methods of Preparing Same, US Patent
soluble drug produced by a spray-freezing into liquid-emulsion process, Eur. 6423339B1, 2002.
J. Pharm. Biopharm. 55 (2003) 161–172. [37] R.H.C. Queiroz, V.L. Lanchote, P.S. Bonato, D. Carvalho, Simultaneous HPLC analysis of
[7] D. Kayrak, U. Akman, O. Hortacsu, Micronisation of ibuprofen by RESS, J. Supercrit. tricyclic antidepressants and metabolites in plasma samples, Pharm. Acta Helv. 70
Fluids 26 (2002) 17–31. (1995) 181–186.
[8] M. Sarkari, J. Brown, X.X. Chen, S. Swinnea, R.O. Williams, K.P. Johnston, Enhanced [38] Y. Javadzadeh, L. Musaalrezaei, A. Nokhodchi, Liquisolid technique as a new
drug dissolution using evaporative precipitation into aqueous solution, Int. approach to sustain propranolol hydrochloride release from tablet matrices, Int.
J. Pharm. 243 (2002) 17–31. J. Pharm. 362 (2008) 102–108.
[9] M.D. Ticehurst, P.A. Basford, C.I. Dallman, T.M. Lukas, P.V. Marshall, G. Nichols, D. [39] L. Kürtia, Á. Kukoveczb, G. Kozmab, R. Ambrusa, M.A. Delic, P. Szabó-Révésza, Study
Smith, Characterisation of the influence of micronisation on the crystallinity and of the parameters influencing the co-grinding process for the production of
physical stability of revatropate hydrobromide, Int. J. Pharm. 193 (2000) 247–259. meloxicam nanoparticles, Powder Technol. 212 (2011) 210–217.
[10] J.C. Feely, P. York, B.S. Sumby, Determination of surface properties and flow charac- [40] A. Bodea, S.E. Leucuta, Optimization of propranolol hydrochloride sustained release
teristics of salbutamol sulphate, before and after micronisation, Int. J. Pharm. 172 pellets using a factorial design, Int. J. Pharm. 154 (1997) 49–57.
(1998) 89–96. [41] K. Sanka, S. Bandari, R. Jukanti, P.R. Veerareddy, Colon-specific microparticles of
[11] M. Sugimoto, T. Okagaki, S. Narisawa, Y. Koida, K. Nakajima, Improvement of piroxicam: formulation and optimization using 32 factorial design, J. Dispers. Sci.
dissolution characteristics and bioavailability of poorly water soluble drugs by Technol. 32 (2011) 1396–1403.
novel cogrinding method using water-soluble polymer, Int. J. Pharm. 160 (1998) [42] S. Spireas, (2002). Liquisolid Systems and Methods of Preparing Same, U.S. Patent
11–19. 6,423,339 B1.
[12] M. Fujii, H. Okada, Y. Shibata, H. Teramachi, M. Kondoh, Y. Watanabe, Preparation, char- [43] M.A. Roni, M.S. Islam, G. Kibria, S.M.A. Sadat, M.R. Rony, M.H. Rahman, R.U. Jalil,
acterization and tableting of a solid dispersion of indomethacin with crospovidone, Effects of poloxamer and HPMC on the dissolution of CLZ-polyethylene glycol
Int. J. Pharm. 293 (2005) 145–153. solid dispersions and tablets, Indian J. Pharm. Educ. 45 (2011) 139–144.
344 K. Sanka et al. / Powder Technology 256 (2014) 336–344

[44] Ravi Ravichandran, USP Monographs: CLZ tablets, (MDPP05) Monograph [49] S. Madaka, K. Sanka, P.R. Veerareddy, Design and evaluation of hydrochlorothiazide
development-psychiatrics and psychoactives, USP 29-NF 24, page 556; Pharmaco- gastroretentive floating drug delivery system, Asian, J. Pharm. Sci. 6 (2011) 208–217.
peial forum: 29 (3) page 261. [50] K. Rapolu, K. Sanka, P.K. Vemula, V. Aatipamula, A.B. Mohd, P.V. Diwan, Optimization
[45] R.H. Fahmy, M.A. Kassem, Enhancement of famotidine dissolution rate through and characterization of gastroretentive floating drug delivery system using Box–
liquisolid tablets formulation: in vitro and in vivo evaluation, Eur. J. Pharm. Behnken design, Drug Dev. Ind. Pharm. 39 (12) (2013) 1928–1935.
Biopharm. 69 (2008) 993–1003. [51] K. Sanka, V.S. Munjulury, A.B. Mohd, P.V. Diwan, Enhancement of solubility, dissolu-
[46] G. Alekhya, J. Raju, B.S. Reddy, K. Swetha, B.J. Jyothi, Improved oral delivery of tion release profile and reduction in ulcerogenicity of piroxicam by inclusion
valsartan from maltodextrin based proniosome powders, Adv. Powder Technol. 23 complex with skimmed milk, Drug Delivery (2013), http://dx.doi.org/10.3109/
(2012) 583–590. 10717544.2013.856964.
[47] K.G. Pradip, J.M. Rita, L.U. Manish, S.R.M. Rayasa, Design and development of [52] G.S. Banker, N.L. Anderson, Tablets, in: L. Lachman, H.A. Liberman, J.L. Kanig (Eds.),
microemulsion drug delivery system of acyclovir for improvement of oral bioavail- The Theory and Practice of Industrial Pharmacy, Varghese Publishing House,
ability, AAPS PharmSciTech 7 (2006) E1–E5. Bombay India, 1987, pp. 293–345.
[48] R.J. Sanjeev, J. Ravindra, Formulation and characterization of atorvastatin calcium
liquisolid compacts, Asian, J. Pharm. Sci. 5 (2010) 50–60.

You might also like