Accepted Manuscript Title: Formulation and process optimization of naproxen nanosuspensions stabilized by hydroxy propyl methyl cellulose Author: Bibaswan Mishra Jagannath Sahoo Prasanna Kumar Dixit PII: DOI: Reference:

S0144-8617(15)00290-8 http://dx.doi.org/doi:10.1016/j.carbpol.2015.03.077 CARP 9819

To appear in: Received date: Revised date: Accepted date:

4-2-2015 19-3-2015 25-3-2015

Please cite this article as: Mishra, B., Sahoo, J., and Dixit, P. K.,Formulation and process optimization of naproxen nanosuspensions stabilized by hydroxy propyl methyl cellulose, Carbohydrate Polymers (2015), http://dx.doi.org/10.1016/j.carbpol.2015.03.077 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

1

TITLE PAGE

2 3 4 5

Formulation and process optimization of naproxen nanosuspensions stabilized by hydroxy propyl methyl cellulose

a

6 b

Gayatri College of pharmacy, Sambalpur, Odisha 768200, India

Shri Ram Murti Smarak College of Engg. and Technology (Pharmacy), Bareilly, U.P. 243202, India

8 c

Department of Zoology, Berhampur University, Ganjam, Odisha 760007,India

us

9

an

10

E-mail address of authors:

12

Bibaswan Mishra – [email protected]

13

Jagannath Sahoo - [email protected]

14

Prasanna Kumar Dixit- [email protected]

d

M

11

te

15

18 19

Ac ce p

16 17

cr

7

ip t

Bibaswan Mishraa*, Jagannath Sahoob, Prasanna Kumar Dixitc

20

* Corresponding author.

21

Bibaswan Mishra

22

Gayatri College of pharmacy, Sambalpur, 768200, Odisha, India

23

Ph no- +91 98613 38249

24

E-mail address: [email protected]

1 Page 1 of 33

25 26 27 28 29 30

In present study precipitation-ultrasonication was used to obtain nanosuspensions of poorly

31

water-soluble drug, Naproxen (NPX). We investigated the effects of HPMC concentration (X1)

32

and time of ultrasonication (X2) on imperative attributes like mean particle size (Y1), % drug

33

content (Y2), and time required to 90% drug release (Y3) via 32 factorial design. The morphology

34

of nanosuspensions was found almost spherical by SEM observation. DSC and XRD studies

35

suggested slight crystalline change in drug. FT-IR revealed lack of significant interactions

36

between NPX and HPMC. Nanosuspensions of mean particle size 530.55 nm was achieved.

37

Dissolution rate obtained from all nanosuspensions were markedly higher than pure NPX.

38

Response surface methodology and optimized polynomial equations were used to select optimal

39

formulation i.e. 1.36 % W/V of X1 and 13.9 mins of X2 to get desired response Y1; 727.97 nm,

40

Y2; 95.59 % and Y3; 8.67 mins that were in reasonable agreement with observed value.

Formulation and process optimization of naproxen nanosuspensions stabilized by hydroxy propyl methyl cellulose

te

d

M

an

us

cr

ip t

Abstract

Ac ce p

41 42

Key words: Naproxen; HPMC; Nanosuspensions; Ultrasonication; Response surface

43

methodology.

44 45 46 47 48 49 50 51 52 53

2 Page 2 of 33

Formulation and process optimization of naproxen nanosuspensions stabilized by hydroxy propyl methyl cellulose

57

1. Introduction

58

In the pharmaceutical drug delivery system poor solubility of drugs is a major challenge due to

59

non optimal physicochemical and pharmaceutical properties. About 40% of active

60

pharmaceutical ingredients (API) discovered through different drug discovery program have

61

dissolution related bioavailability problem (Lipinski, 2002). Such drugs show variable

62

bioavailability and very erratic absorption profile. Low dissolution velocity of such drugs in both

63

water and organic media prevents to formulate intravascular preparations for getting sufficiently

64

high blood level.

65

Many approaches have been developed to overcome such issues like inclusion complexes with

66

B-cyclodextrin (Yang et al., 2014), co crystal (Schultheiss & Newman, 2009), salt formation

67

(Serajuddin, 2007), pH micro environment modifier (Stephenson, Aburub, & Woods, 2011), Self

68

emulsified drug delivery system (SEDDS) (Neslihan & Benita 2004), solid lipid nanoparticles

69

(Muchow, Maincent, & Müller, 2008), liposome (Fenske, Chonn, & Cullis, 2008),

70

microemulsion (Stephenson, Aburub, & Woods, 2011), and so on. Because of various obstacles

71

such as poor drug loading, poor stability, high economical cost, complex manufacturing

72

approaches and high toxicity, none of these techniques can be applied to all categories of drug.

73

Nanosuspension refers to the reduction in drug particle size down to submicron range suitably

74

stabilized by polymer and/or surfactants (Müller, Peters, Becker, & Kruss, 1995a.; Müller,

75

Becker, Kruss, & Peters, 1998). The contributing role of nanosized drugs to the dissolution

76

velocity is described by Nernst Brunner equation and increase in solubility illustrated by

77

Ostwald-Freundlich equation (Brough & Williams III 2013). Nanosuspensions are advantageous

Ac ce p

te

d

M

an

us

cr

ip t

54 55 56

3 Page 3 of 33

in achieving quick onset of action, reduce fed/fasted state variability, improved dose

79

proportionality, and improve the saturation solubility and hence enhancing bioavailability. The

80

methods of producing nanosuspension are of two types; ‘bottom up’ and ‘top down’. Top down

81

approaches such as high pressure homogenization (Xiong et al., 2008) and media milling

82

(Vander et al., 2007) based on disintegration approach from large particle, microparticles to

83

nanoparticles and bottom up techniques include microprecipitation, microemulsion, melt

84

emulsification and so on, based on assembling method from molecules to nano sized particle

85

have been successfully developed to obtain stable nanosuspension (Grau, Kayser, & Müller,

86

2000).

87

Antisolvent precipitation has been successfully employed as an effective and simple technique to

88

get stable nanosuspension of nitrendipine (Xia et al., 2010). In this method the drug was

89

dissolved in a suitable solvent mixture which was then incorporated in to a precooled antisolvent

90

solution containing stabilizer. The antisolvent solution must be maintained at low temperature to

91

restrict the formation of larger particle due to Ostwald ripening which is a function of rapid

92

disolvation technique. A number of stabilizers such as hydroxy propyl methyl cellulose & methyl

93

cellulose (Douroumis & Fahr, 2007), Povidone (Lindfors, Forssén, Westergren, & Olsson, 2008),

94

and poly vinyl alcohol (Xia et al., 2010) in aqueous solution showed a tremendous result in

95

stabilizing the formulation as a result of hydrogen bonding to the hydrophobic core of drug.

96

Naproxen (NPX), a propionic acid derivative, is used extensively in inflammatory diseases such

97

as acute gout and rheumatoid arthritis. The absorption of drugs with poor aqueous solubility like

98

NPX is dissolution rate limited and therefore, they exhibit poor bioavailability resulting in

99

multiple dosing of drug as well as fluctuation in blood concentrations (Correa, Scarpa, Franzini,

100

& Oliveira, 2005; Guo et al., 2010). The oral administration of the drug is the most common

Ac ce p

te

d

M

an

us

cr

ip t

78

4 Page 4 of 33

route of delivery due to convenience and patient compliance which make it more effective when

102

compared with other routes of administration. But the important problem associated in obtaining

103

a solid oral dosage forms for NPX has been its poor aqueous solubility and wettability.

104

Pharmaceutical formulators often face the challenge of finding the appropriate combination of

105

process variables to obtain a product with desired quality attributes. However, statistical design

106

of experiment tools such as factorial designs can be used to analyze and understand these

107

variables to get optimum properties. All factors are studied in all possible combinations with

108

minimum experimentation and time to estimate the influence of individual process variables.

109

This optimization technique encompasses the generation of model equations for the investigated

110

responses over the experimental design to obtain the optimum formulation(s) (Acharya, Patra, &

111

Pani, 2014).

112

In the present study precipitation ultrasonication method has been employed to develop and

113

fabricate nanosuspensions of naproxen. The effect of process variables i.e., stabilizer

114

concentration and time length of sonication (two independent factors) on mean particle size, %

115

drug content and time required to 90% of drug release (t90) (three dependent variables) were

116

systematically investigated through response surface methodology (RSM) via 32 factorial design.

117

The dependent variables are so chosen in order to get enhanced bioavailability with minimum

118

dose. The in-vitro dissolution study was considered to access the solubility enhancement of drug

119

in different formulations.

120

2. Materials

121

Naproxen (mean particle size; 67.32 µm) was obtained as a gift sample from Dr. Reddy’s

122

Laboratories Ltd. (Hyderabad, Andhra Pradesh, India). Hydroxy Propyl Methyl Cellulose

123

(Methocel K4M) was procured from Corolcon Asia Pvt. Ltd. and Polyethylene Glycol 200 (PEG

Ac ce p

te

d

M

an

us

cr

ip t

101

5 Page 5 of 33

200) and acetone were purchased from Ranbaxy fine chemicals Ltd., New Delhi, India. All the

125

reagents were of analytical grade and used without further purification.

126

3. Methods

127

3.1. Preparation of naproxen nanosuspensions

128

Nanosuspensions of naproxen (NPXN) stabilized by HPMC were prepared by controlled

129

precipitation-ultrasonication method. NPX (50 mg) was dissolved in 2mL of solvent mixture of

130

acetone and PEG 200 (1:1, V/V). Antisolvents containing different percentage of HPMC (0.5%,

131

1% and 1.5%, W/V) were prepared with water. Both solutions are filtered through 0.45 micron

132

filter (Nylon 66 membrane filter). HPMC solution of 40mL was placed in a bath ultrasonicator

133

(Sonica, Spincotech Pvt. Ltd. 2200 MH, 305 W, Soltech Sri Milano, Italy) and the water inside

134

the ultrasonicator was precooled and maintained at temperature of 3°C. The organic drug

135

solution was quickly incorporated to the later with the help of a 22 gauze syringe after the

136

application of ultrasonic wave. Ultrasonication was applied for a time period of 5, 10 and 15

137

mins for each combination of the antisolvent solutions. The obtained nanosuspension were

138

subjected to cooling ultracentrifuge (C-24 BL, Remi, Mumbai, India) at 20 000 rpm for 40 mins.

139

The supernatant was discarded and replaced by same quantity of fresh antisolvent. The solid

140

residue is redispersed by the application of sonication. The study design is presented in Table-1.

cr

us

an

M

d

te

Ac ce p

141

ip t

124

142 143 144 145 6 Page 6 of 33

ip t

Table-1. 32 full factorial design with observed response values of Naproxen nanosuspensions.

cr

146

148 149 150

R.E (%)

Avg. drug content (%)

R.E (%)

Avg. time to 90% drug release (mins) Obs. Pred. value value

R.E (%)

Z.P. (mV)

Batch code

HPMC (%W/V)

U. Sonic time (Mins)

NPXN-1

0.5(-1)

5(-1)

2519.61

2527.99

0.33

95.98

95.58

-0.42

23.9

23.73

-0.71

-32.56

NPXN-2

1.0(0)

5(-1)

1372.12

1412.88

2.88

96.32

96.58

0.26

13.92

13.84

-0.55

-29.94

NPXN-3

1.5(1)

5(-1)

2178.78

2184.61

0.27

97.89

97.57

-0.33

19.5

19.71

1.08

-35.77

NPXN-4

0.5(-1)

10(0)

2311.35

2264.17

-2.08

93.11

93.63

0.56

21.68

22.17

2.20

-32.49

NPXN-5

1.0(0)

10(0)

ed

Mean Particle size(nm)

us

147

982.42

961.69

-2.16

95.11

95.21

0.11

11.77

11.47

-2.58

-29.96

NPXN-6

1.5(1)

10(0)

1588.13

1546.05

-2.72

96.44

96.79

0.36

16.76

16.57

-1.16

-31.85

NPXN-7

0.5(-1)

15(1)

1961.58

2000.35

1.94

91.79

91.68

-0.12

17.18

16.82

-2.15

-34.56

NPXN-8

1.0(0)

15(1)

530.55

510.5

-3.93

94.17

93.85

-0.35

5.27

5.35

1.50

-33.09

NPXN-9

1.5(1)

15(1)

871.27

907.49

3.99

96.04

96.01

-0.03

9.68

9.67

-0.12

-31.1

NPXN-O

1.36

13.9

757.16

727.97

-4.01

94.91

95.59

0.71

8.89

8.67

-2.54

-34.73

Ac

Obs. value

M an

Pred. value

ce pt

Obs. value

Pred. value

Note: NPXN, Naproxen Nanosuspensions; NPXN -O, Optimized batch; HPMC, Hydroxy Propyl Methyl Cellulose; U. Sonic Time, Ultasonication Time; Obs. Value, Observed Value; Pred. Value, Predicted Value; R.E, Relative Error; Z.P, Zeta Potential

151 7 Page 7 of 33

3.2. Isolation of dried nanosuspensions.

153

The solidified state is preferred as compared with aqueous nanosuspensions because aggregation

154

and other instability factors are significantly decreased. Nanosuspension of 20 mL was filled in

155

flint colored vials with rubber stoppers and frozen using deep freezer at −75 °C for 24 hr. These

156

frozen semisolids were freeze-dried using lyophilizer (Virtis benchtop, Bombay, India) at a

157

vacuum degree of 200 Pascal for 36 hr to produce free flowing dry powder.

158

3.3. Characterization of naproxen nanosuspensions

159

3.3.1. Mean Particle size, zeta-potential and particle morphology

160

The particle size and Zeta-potential of nanosuspensions were measured using dynamic light

161

scattering zetasizer (Zetasizer Nano ZS 90, Malvern Ltd., UK). All formulations were adequately

162

diluted to obtained final drug particle concentration of 1 mg/mL and introduced into the

163

electrophoretic cell. In addition, nanosuspensions were also stored to assess the short term and

164

accelerated stability and their particle size were analyzed. The surface topography was studied

165

using scanning electron microscope (JEOL, JSM-6390, Tokyo, Japan). Morphological

166

examination of surface was performed at 3000X and 10000X magnifications.

167

3.3.2. Differential Scanning Calorimetry

168

NPX pure drug and freeze dried NPXN were analyzed by differential scanning calorimetry

169

(DSC-60, Shimadzu Co., Japan) at a heating rate of 10°C /min from 30 to 300°C to study the

170

phase transition. This study was done under dry nitrogen atmosphere using Al2O3 as a reference.

171

3.3.3. Fourier transform infrared spectroscopy (FT-IR)

172

The molecular structures of the samples were recorded through obtaining FT-IR spectra using

173

FT-IR spectrometer (Shimadzu Corporation, Japan). The IR spectra were obtained in a KBr disc.

Ac ce p

te

d

M

an

us

cr

ip t

152

8 Page 8 of 33

Pure drug, HPMC and nanosuspensions were mixed with potassium bromide separately. Scans

175

were obtained at a resolution of 2 cm−1, from 4500 to 400 cm−1.

176

3.3.4. X-ray diffraction analysis

177

The pure NPX, HPMC and freeze dried NPXN were analyzed by X-ray diffractometer (DX-

178

2700, China) in symmetrical reflection mode using Cu Kα radiation generated at 30mA and 40

179

kV. The scanning speed was 10°/min from 0° to 55°of 2θ.

180

3.4. Drug content study

181

Drug equivalent to 10 mg of NPX was dissolved in 50 ml ethanol. The samples were stirred for

182

4h and subsequently centrifuged at 6000 rpm for 10 min. The contents of NPX in sample were

183

analyzed after suitable dilution by double beam UV spectrophotometer (model UV-1700,

184

Shimadzu, Japan) at λmax272 nm. The procedure was performed in triplicate and the averages

185

were calculated (Adibkia et al., 2013). Drug content was calculated by the following equation:

186

Drug content (%) = (Observed drug content/Theoretical drug content) ×100

187

3.5. Dissolution study

188

In vitro release tests were performed for the prepared nanosuspension as well as pure NPX. The

189

study was performed in 900 mL of hydrochloric acid solution (pH 3) using USP apparatus II

190

(TDT-08 L, Electrolab, Mumbai, India) with constant temperature maintained at 37 ± 0.5°C and

191

50 rpm. An aliquot of 5 mL was withdrawn at various time intervals and passed through a 0.1µm

192

syringe filter (Whatman Ltd, Middlesex, UK). The prewarmed fresh dissolution medium was

193

replaced immediately. The contents of NPX in sample were analyzed by double beam UV

194

spectrophotometer (model UV-1700, Shimadzu, Japan) at λmax 272 nm (The United States

195

Pharmacopeia, 2011). Then the corresponding concentrations were determined from the standard

Ac ce p

te

d

M

an

us

cr

ip t

174

9 Page 9 of 33

curve of NPX prepared in hydrochloric acid solution (pH 3) at λmax 272nm. The mean values

197

were calculated from 3 replicates. The cumulative % of drug released as a function of time was

198

plotted as a measure of dissolution velocity.

199

3.6. Experimental design and optimization

200

A 32 randomized full factorial design was used to optimize the variables in the present study. In

201

this design, 2 factors: percentages of HPMC and time length of ultrasonication were evaluated,

202

each at 3 levels: low, medium and high (Table-1) and experimental trials were performed at all 9

203

possible combinations as depicted (Acharya et al., 2014). Various percentage of HPMC (X1)

204

such as 0.5%, 1% and 1.5% W/V, and time length of ultrasonication (X2), such as 5, 10, and 15

205

mins were selected as independent variables. Mean particle size (Y1), percentage drug content

206

(Y2) and t90 (Y3) were selected as dependent variables. Data obtained from all formulations were

207

analyzed using Design Expert software (Stat-Ease Inc., Minneapolis, Minnesota) and were used

208

to generate the study design and the response surface plots. The matrix of the design including

209

investigated factors and responses are shown in Table-1. Polynomial models, including linear,

210

interaction and quadratic terms were generated for all the response variables using the software.

211

Design Expert software provides the best fitting model was selected based on comparisons of

212

several statistical parameters, including the regression coefficient (R2), coefficient of variation

213

(CV), and adjusted regression coefficient (adj. R2). To identify significant effects of factors on

214

response regression coefficients, analysis of variance (ANOVA) was used by calculating the F

215

test and P values. For optimization, the mathematical model equation involving independent

216

variables and their interactions for various measured responses generated by 32 factorial design

217

is:

218

Y = b0 + b1X1 + b2X2 + b12X1X2 + b11X12 + b22X22

Ac ce p

te

d

M

an

us

cr

ip t

196

10 Page 10 of 33

Where Y is the dependent variable, b0 is the arithmetic mean response of the 9 runs, and bi is the

220

estimated coefficient for the factor Xi. The main effects (X1and X2), the interaction terms (X1X2),

221

and the polynomial terms (X12and X22) represent the average result of changing one factor at a

222

time from its low to high value, how the response changes when two factors are simultaneously

223

changed and non-linearity respectively. Response surface plots are used to elucidate the

224

relationship between the dependent and independent variables. Subsequently, a numerical

225

optimization technique using the desirability approach and a graphical optimization technique

226

using overlay plots were used to generate new formulations with the desired attributes.

227

3.7. Validation of the experimental design

228

To validate the chosen experimental design, the experimental observed values of the responses

229

were quantitatively compared with predicted values and the relative error (%) was calculated

230

using the following equation:

231

Relative error (%) = {(Predicted value − Experiment value) / Predicted value} × 100

232

3.8. Stability testing

233

Stability is the first requisite for nanosuspension based product to be a commercial drug product.

234

Both short term and long term stress testing were investigated to ensure the stability of the

235

prepared formulation during storage.

236

3.8.1. Short-term physical stability test

237

NPXN-8 was stored at 25°C for up to 3 weeks. The nanosuspensions were kept in a 10 mL

238

closed glass vial. At the predetermined times, aliquots were taken and subjected to particle size

239

analysis as described before (Yao et al., 2014).

240

3.8.2. Long term accelerated stability test

Ac ce p

te

d

M

an

us

cr

ip t

219

11 Page 11 of 33

To ensure the long term stability, NPXN-8 was tested during storage (40°C, RH 75%). Samples

242

were sealed tightly in 10 mL closed glass vial and the stability was evaluated for the increase in

243

particle size after 1, 3 and 6 months of storage (Yao et al., 2014).

244

4. Result and discussion

245

4.1. Particle morphology

246

To characterize the surface morphology of nanosuspension formulations, optical microscopy

247

(Olympus, CH2 model) of pure drug was compared with SEM images, taken directly after

248

preparation, as depicted in Figure-1. The micrographs clearly showed the great differences

249

between NPX raw crystal and nanosuspensions. The former were very large and irregular

250

(Figure-1.a), while in nanosuspension, the particle size reduced significantly and found to be

251

spherical in shape perhaps because the particles were coated with a HPMC polymeric layer

252

(Figure-1.b and c.).

Ac ce p

te

d

M

an

us

cr

ip t

241

12 Page 12 of 33

ip t cr us an M d te Ac ce p 253 254

Figure-1. Optical microscopic photograph of NPX crystal (a); SEM of NPX

255

nanosuspensions (b and c). 13 Page 13 of 33

4.2. Differential scanning calorimetry

257

In order to determine, whether amorphization had occurred at particle surfaces and to explore

258

potential transformations of the structure, DSC analysis was performed on the freeze dried

259

nanosuspension formulations (Figure-2.a). DSC of pure naproxen displayed single sharp

260

endothermic fusion peak at 152.7°C, which is corresponding to the melting point of drug. In

261

order to further confirm the physical state, DSC of NPXN-8 and NPXN-9 were also performed to

262

analyze nanosuspensions. The DSC spectra of nanosuspensions also exhibited endothermic

263

melting peak at 151.9°C and 152.3°C respectively but with lower intensity which could be

264

explained by lower amount of NPX in nanosuspensions comparing to pure NPX. In formulations

265

with high ratio of polymers, lower intensity is as a result of dilution effect of the polymers

266

(Cafaggi et al., 2008). The study inferred that there was no interaction between drug and

267

excipients (Oguchi, Sasaki, Hara, Tozuka, &Yamamoto, 2003).

268

4.3. X-ray diffraction

269

X-ray diffraction has been used to analyze potential changes in the inner structure of NPX

270

crystals. XRD findings, as shown in Figure-2.b, were in good agreement with the DSC results.

271

Nanosuspension showed distinctive patterns of peaks that were consistent with the peak pattern

272

of the NPX raw crystal. The height of the peaks in the nanosuspension demonstrated the

273

reduction in magnitude of peaks due to the dilution of drug in the inner core of stabilizer and a

274

decrease of NPX crystallinity in nanosuspension (Adibkia et al., 2013). During the addition of

275

the anti-solvent, the newly formed crystal nuclei are most likely amorphous (Lindfors et al.,

276

2006a) but it increases the bulk drug concentration and facilitates particle growth by Ostwald

277

ripening (Lindfors et al., 2006b).

278

4.4. Fourier transform infrared spectroscopy (FT-IR)

Ac ce p

te

d

M

an

us

cr

ip t

256

14 Page 14 of 33

FT-IR studies were done to find out the possible intermolecular interactions between the NPX

280

and HPMC. The characteristic peaks of NPX, HPMC and nanosuspensions are depicted in

281

Figure-2.c. Absorption peaks of 1155 cm−1and 1174 cm−1 are correlated to etheric bonds and

282

peaks of 1724 cm−1 and 1681 cm−1 are related to the carboxylic acid bond and benzene ring,

283

respectively. Absence of any other new peaks and also no differences in the positions of the

284

absorption bands in the nanosuspensions, indicate the lack of significant interactions between

285

NPX and HPMC.

Ac ce p

te

d

M

an

us

cr

ip t

279

15 Page 15 of 33

ip t cr us an M d te Ac ce p 286 287

Figure-2. DSC thermograms of the original NPX powder and NPX nanosuspensions (a); X-

288

ray diffractograms of the original NPX powder, HPMC, and NPX nanosuspensions (b)

289

and FTIR of original NPX powder, HPMC, and NPX nanosuspensions (c). 16 Page 16 of 33

4.5. Analysis of data and optimization of design

291

As ‘stabilizer concentration’ and ‘time length of ultrasonication’ are independent to each other

292

and controlled in experiments, both were considered as independent variable in this study. Mean

293

particle size, % drug content, and time required to 90% drug release (t90) obtained were

294

considered as dependent variables due to their significant contribution in formulation

295

development. The software provided suitable polynomial model equations involving individual

296

main factors and interaction factors after fitting these data. The co-efficients in the optimized

297

model equations relating Y1, Y2 and Y3 as responses are given in (Table-2). The results of

298

ANOVA, as shown in table-2, indicated that all models were significant (p < 0.05) for all

299

response parameters investigated. Model simplification was carried out by eliminating non-

300

significant terms (p > 0.05) in polynomial equations (Nayak & Pal, 2011).

M

an

us

cr

ip t

290

304 305 306 307

te

303

Ac ce p

302

d

301

308 309 310 17 Page 17 of 33

ip t

Co-efficient value

df

Sum square

Mean square

F-Value

p-value Prob > F

961.70 -359.06 -451.19 -187.37 943.42

4 1 1 1 1

3915477.17 773544.50 1221407.43 140430.07 1780095.17

978869.29 773544.50 1221407.43 140430.07 1780095.17

416.14 328.85 519.25 59.70 756.76

< 0.0001 0.0001 < 0.0001 0.0015 0.0001

3

27.56

9.19

54.12

0.0003

1 1 1

15.01 11.18 1.37

15.01 11.18 1.37

88.42 65.86 8.06

0.0002 0.0005 0.0363

5 1 1 1 1 1

283.57 47.15 105.76 2.40 121.58 6.69

56.71 47.15 105.76 2.40 121.58 6.69

325.98 271.02 607.86 13.81 698.79 38.43

0.0003 0.0005 0.0001 0.0339 0.0001 0.0085

For Mean Particle Size Model X1 X2 X1X2 X12

95.21

X1 X2 X1X2

1.58 -1.37 0.59

ce pt

Model

ed

For Percent drug Content

us

Source

cr

Table-2. Analysis of variance table for dependent variables from full factorial design.

M an

311

For time to 90% drug release

11.54 -2.80 -4.20 -0.78 7.80 -1.83

Ac

Model X1 X2 X1X2 X12 X22 312 313

18 Page 18 of 33

4.5.1 Effect of independent factors on Mean particle size and Zeta potential

315

Mean Particle sizes and Zeta potentials of all the formulations were depicted in Table-1. The

316

relationship between the formulation ingredients (independent variables) and dependent

317

variables) was elucidated using response surface plots presented in Figure-3.a for Y1. The effect

318

of X1 and X2 and their interaction on Y1 by fixing one factor at constant level was determined.

319

As seen in figure, at fixed levels of X2, Y1 was decreased when the amount of HPMC

320

concentration added was increased from 0.5 to 1.0 % W/V and was increased when the HPMC

321

concentration was increased from 1.0 to 1.5 % W/V. The decrease in particle size may be

322

attributed to the following reasons. As HPMC is a nonionic polymer, potential agglomeration

323

was less liable to occurred. Due to the presence of a hydrodynamic boundary layer surrounding

324

the nanosuspensions as well as adsorption of the polymer molecules on the growing crystal

325

faces, HPMC provides a steric hindrance to agglomeration and growth of nanosuspensions

326

(Raghavan, Trividic, Davis, & Hadgraft, 2001; Zimmermann et al., 2009). The drug-stabilizer

327

system can be explained on the basis of hydrogen bonding between the drug molecules and the

328

stabilizer with abundance of hydroxyl groups (Figure-4). Amongst all formulation NPXN-8

329

showed the least mean particle size i.e. 530.55 nm.

331 332

cr

us

an

M

d

te

Ac ce p

330

ip t

314

333 334 335 19 Page 19 of 33

ip t cr us an M d te Ac ce p 336 337

Figure-3. Three-dimensional response curve of mean particle size (a); % drug content (b);

338

and t90 (c).

20 Page 20 of 33

ip t cr us an

339

Figure-4. Molecular structures of Naproxen (a) and HPMC (b).

341

The increase in the particle size with increase in HPMC concentration can be explained as

342

follows. Enhance viscosity of the solution hinder the transmission of ultrasonic energy and

343

thereby obstruct the diffusion between the solvent and antisolvent during precipitation formation.

344

This resulted in the formation of larger particle due to Ostwald ripening. Moreover the

345

deposition of concentric layers of HPMC on the drug surface resulted in larger particle size.

346

However in formulations with 1.0 % W/V of HPMC concentration found to be the optimal value

347

that sufficiently cover the crystal surface and provide steric repulsion between crystals.

348

At low levels of X1, the Y1 was decreased from 2519.61 nm to 1961.58 nm as X2 was increasing

349

from 5 to 15 minutes. Similarly, at high levels of X1, Y1 was decreasing from 2178.78 nm to

350

871.27 nm when X2 was increasing from 5 to 15 minutes. The decrease in the particle size with

351

decrease in X2 can be attributed to (a) the increase time length of the erosion effect on the surface

Ac ce p

te

d

M

340

21 Page 21 of 33

of large crystal and crystal agglomerates, (b) the HPMC molecule get completely adsorbed onto

353

the crystal surface to provide steric hindrance.

354

If particles possess sufficient zeta potential to provide enough electric repulsion or steric

355

hindrance, particle aggregation is less likely to occur. It has to be noted that adsorption of a steric

356

stabilizer such as the HPMC in all the formulations in our study, leads to a reduction of the

357

measured zeta potential. This is because the adsorption layer of the HPMC on the drug surface

358

increases its distance to the plane of shear, at which the zeta potential is measured.

359

4.5.2. Effect of independent factors on % Drug content

360

Similar trend was observed for the effect of X1 and X2 and their interaction on Y2. As seen from

361

Figure-3.b, at fixed levels of X2, Y2 was increased when the amount of HPMC concentration

362

added (X1) was increasing from 0.5 to 1.0 % W/V. The increase in the percentage drug content

363

with increase in HPMC concentration can be ascribed to the formation of more concentric layers

364

of HPMC molecule on the drug surface that restricts the drug to diffuse back in to the bulk. At

365

fixed levels of X1, Y2 was decreased as X2 was increasing from 5 to 15 minutes. The decrease in

366

the particle size with decrease in X2 may be as follows. The insufficient time of ultrasonic energy

367

at lower value of X2 resulted in the incomplete adsorption of HPMC molecule on the newly

368

formed crystal nuclei as a result of agglomeration that occurred rapidly (Xia et al., 2010).

369

4.5.3. Effect of independent factors on t90

370

Dissolution profiles of NPX pure drug and nanosuspensions are presented in Figure-5. The slow

371

dissolution rate of NPX may be due to its large crystal size while the nanosuspensions exhibited

372

considerably faster dissolution rates which may be ascribed to the following reasons (a) high

373

hydrophilicity of the polymers bring about aggregation reduction, wettability improvement and

Ac ce p

te

d

M

an

us

cr

ip t

352

22 Page 22 of 33

local solubilization by the carrier in the diffusion layer and thereby increasing in the dissolution

375

rate (Jalali et al., 2012; Tiong & Elkordy, 2009), (b) the crystallinity reduction in the NPX loaded

376

nanosuspensions prepared by HPMC as the amorphous form of a drug has a higher

377

thermodynamic activity than its crystalline form, leading to rapid dissolution of the drug (Paudel,

378

Worku, Meeus, Guns, & Van den, 2013; Mohammadi et al., 2010), (c) the reduced particle size

379

and accordingly elevated surface area could enhance the dissolution rate of NPX in the

380

formulation (Adibkia et al., 2013).

us

cr

ip t

374

382

Ac ce p

te

d

M

an

381

383

Figure-5. Comparison of dissolution profiles of Nanosuspensions formulation with pure

384

Naproxen

385

As seen in figure-3.c, at fixed levels of X2, Y1 was decreased when the amount of HPMC

386

concentration added was increased from 0.5 to 1.0 % W/V and was increased when the HPMC

387

concentration was increased from 1.0 to 1.5 % W/V. The variation in the dissolution rate in

388

different formulations is attributed to variation in diffusion layer thickness due to gel formation 23 Page 23 of 33

effect of HPMC since the drug molecules were entrapped completely within the gel structure.

390

This effect was probably affected by hydrophilicity of HPMC in the higher drug-HPMC ratios.

391

Although, a direct relationship between the amount of stabilizer and NPX dissolution rate could

392

not be established from the dissolution profiles of the different nanosuspensions, but dissolution

393

rate obtained from all nanosuspension were much higher than the pure NPX.

394

To optimize all the responses with different targets, a multi-criteria decision approach, a

395

numerical optimization technique by the desirability and a graphical optimization technique by

396

the overlay plot was used (Figure- 6.a and 6.b). The optimized formulation was obtained by

397

applying constraints on independent variables and dependent responses. These constrains for

398

independent variable were X1; 0.5%, 1% and 1.5% W/V, and X2; 5, 10, and 15 mins and that of

399

dependant variable were: Y1; 530.55-1000 nm, Y2; 95- 97.89 % and Y3; 5.27-10 mins. The

400

recommended values of the independent variables were calculated by the Design Expert software

401

from the above plots which has the highest desirability near to 1.0. The optimum values of

402

selected variables obtained using Design Expert software were 1.36 % W/V of X1 and 13.9 min

403

of X2 to get the desired response Y1; 727.97 nm, Y2; 95.59 % and Y3; 8.67 mins in the final

404

composition of NPXN-O.

Ac ce p

te

d

M

an

us

cr

ip t

389

24 Page 24 of 33

ip t cr us an M d te Ac ce p

405 406

Figure-6. Optimization of nanosuspensions through desirability plot (a); and overlay plot

407

(b).

408

4.6. Evaluation and validation of the optimized formulation

25 Page 25 of 33

The statistically optimized formulation (NPXN-O) was prepared, evaluated and Y1, Y2, and Y3

410

were calculated to verify with the theoretical prediction. It was found that the observed value of

411

Y1 (757.16), Y2 (94.91) and Y3 (8.89) were in close agreement with the model predictions Y1

412

(727.97), Y2 (95.59) and Y3 (8.67) of in NPXN-O. The relative errors (%) between the predicted

413

and experimental values for each response were calculated and the values found to be within ±

414

5% (Table-1). The experimental values were in agreement with the predicted values confirming

415

the predictability and validity of the model.

416

4.7. Stability study

417

The representative diameters of the nanosuspensions were plotted as a function of the storage

418

time. It was found that the particle size of the NPXN-8 increased from 530.55 nm to 553.14 nm

419

after three week, and then they changed insignificantly over the subsequent period. The shift in

420

the particle size distribution to a higher range could be attributed to Oswald ripening. So it can be

421

concluded nanosuspensions were able to maintain the particle size during storage. In long term

422

stability testing, though the mean particle size of nanoparticles had a slight increasing trend from

423

530.55 to 562.69 during the test, it is still reasonable to believe the nanosuspension revealed high

424

stability within accelerated testing. It is well documented that HPMC have sufficient affinity for

425

the particle surface and possess an adequately high diffusion rate to cover the generated surface

426

rapidly with the application of ultrasonic energy. In addition, the amount of stabilizer was found

427

to be sufficient for full coverage of the particle surface in order to provide enough steric

428

repulsion between the particles (Gao, Zhang, & Chen, 2008).

429

5. Conclusion

430

This study discussed a positive application of statistical optimization techniques to predict the

431

compositions of a set of formulations that gives optimum quality attributes for the development

Ac ce p

te

d

M

an

us

cr

ip t

409

26 Page 26 of 33

of naproxen nanosuspension of formulated by controlled diffusion-ultrasonication technique. The

433

optimized formulation fulfilled all the requirements of the target set and exhibited suitable values

434

of HPMC concentration and time length of ultrasonication. With the optimized process

435

parameters, nanosuspensions with diameter of about 757.16 nm could be obtained. There is

436

slight crystalline change found during nanosuspension preparation. Marked enhancement of

437

dissolution velocity with excellent % drug content was achieved by the nanoprecipitation

438

method. Hence the successful results of the present research establish the significant role of

439

HPMC as a stabilizer capable of maintain the particle size even after long term stability. It may

440

contribute in the development of an ideal pharmaceutical formulation with high drug content and

441

superior dissolution velocity to overcome the poor bioavailability of naproxen.

442

6. Acknowledgement

443

Authors are acknowledged the Director and Principal of Gayatri College of Pharmacy,

444

Sambalpur, Odisha for providing the research facilities.

447 448 449 450

cr

us

an

M

d

te

446

Ac ce p

445

ip t

432

451 452 453 454

27 Page 27 of 33

REFERENCES

456

The United States Pharmacopeia 34: The National Formulary 29. (2011). The United States

457

Pharmacopoeial Convention, Inc., Rockville.

458

Acharya, S., Patra, S., & Pani, N. R. (2014). Optimization of HPMC and carbopol

459

concentrations in non-effervescent floating tablet through factorial design. Carbohydrate

460

Polymers, 102, 360– 368.

461

Adibkia, K., Jalali, M. B., Esfanjani, H. M., Ghanbarzadeh, S., Shokri, J., Sabzevari, A., &

462

Javadzadeh, Y. (2013). Physicochemical characterization of naproxen solid dispersions

463

prepared via spray drying technology. Powder Technology, 246, 448–455.

464

Brough, C., & Williams III, R. O. (2013). Amorphous solid dispersions and nano-crystal

465

technologies for poorly water-soluble drug delivery. International Journal of Pharmaceutics,

466

453, 157– 166

467

Cafaggi, S., Russo, E., Caviglioli, G., Parodi, B., Stefani, R., Sillo, G., Leardi, R., &

468

Bignardi, G. (2008). Poloxamer 407 as a solubilising agent for tolfenamic acid and as a base

469

for a gel formulation. European Journal of Pharmaceutical Sciences, 35, 19–29.

470

Correa, M. A., Scarpa, M. V., Franzini, M. C., & Oliveira, A. G. (2005). On the

471

incorporation of the non-steroidal anti-inflammatory naproxen into cationic O/W

472

microemulsions. Colloids and Surfaces B: Biointerfaces, 43, 108–114.

473

Douroumis, D., & Fahr, A. (2007). Stable carbamazepine colloidal systems using the

474

cosolvent technique. European Journal of Pharmaceutical Sciences, 30, 367–374.

475

Fenske, D. B., Chonn, A., & Cullis, P.R. (2008). Liposomal nanomedicines: an emerging

476

field. Toxicologic Pathology, 36, 21–29.

Ac ce p

te

d

M

an

us

cr

ip t

455

28 Page 28 of 33

Gao, L., Zhang, D., & Chen, M. (2008). Drug nanocrystals for the formulation of poorly

478

soluble drugs and its application as a potential drug delivery system. Journal of Nanoparticle

479

Research, 10, 845–862.

480

Grau, M. J., Kayser, O., & Müller, R. H. (2000). Nanosuspensions of poorly soluble drugs -

481

reproducibility of small-scale production. International Journal of Pharmaceutics, 196, 155-

482

157.

483

Guo, Z., Liu, X. M., Ma, L., Li, J., Zhang, H., Gao, Y. P., & Yuan, Y. (2010). Effects of

484

particle morphology, pore size and surface coating of mesoporous silica on Naproxen

485

dissolution rate enhancement. Colloids and Surfaces B: Biointerfaces, 101, 228–235.

486

Jalali, M. B., Beirami, M. A., Javadzadeh, Y., Mohammadi, G., Hamidi, A., Andalib, S., &

487

Adibkia, K. (2012). Comparison of physicochemical characteristics and drug release of

488

diclofenac sodium-eudragit® RS100 nanoparticles and solid dispersions. Powder

489

Technology, 219, 211–216.

490

Lindfors, L., Forssén, S., Westergren, J., & Olsson, U. (2008). Nucleation and crystal growth

491

in supersaturated solutions of a model drug. Journal of Colloid and Interface Science, 325,

492

404–413.

493

Lindfors, L., Forssn, S., Skantze, P., Skantze, U., Zackrisson, A., & Olsson, U. (2006a).

494

Amorphous drug nanosuspensions. 2. Experimental determination of bulk monomer

495

concentrations. Langmuir, 22, 911–916.

496

Lindfors, L., Skantze, P., Skantze, U., Rasmusson, M., Zackrisson, A., & Olsson, U. (2006b).

497

Amorphous drug nanosuspensions. 1. Inhibition of Ostwald ripening. Langmuir, 22, 906–

498

910.

Ac ce p

te

d

M

an

us

cr

ip t

477

29 Page 29 of 33

Lipinski, C. A. (2002). Poor aqueous solubility—an industry wide problem in drug delivery.

500

American Pharmaceutical Review, 5, 82–85.

501

Mohammadi, G., Jalali, M. B., Valizadeh, H., Nazemiyeh, H., Jalali, A. B., Shadbad, M.R.S.,

502

Adibkia, K., & Zare, M. (2010). Reciprocal powered time model for release kinetic analysis

503

of ibuprofen solid dispersions in oleaster powder, microcrystalline cellulose and

504

crospovidone. Journal of Pharmacy and Pharmaceutical Sciences, 13, 152–161.

505

Muchow, M., Maincent, P., & Müller, R. H. (2008). Lipid nanoparticles with a solid matrix

506

(SLN®, NLC®, LDC®) for oral drug delivery. Drug Development and Industrial Pharmacy,

507

34, 1394–1405.

508

Müller, R. H., Becker, R., Kruss, B., & Peters, K. (1998). Pharmaceutical nanosuspensions

509

for medicament administration as system of increased saturation solubility and rate of

510

solution. US Patent, No. 5858410.

511

Müller, R. H., Peters, K., Becker, R., & Kruss, B. (1995 a). Nanosuspensions — a novel

512

formulation for the i.v. administration of poorly soluble drugs. 7th International Conference

513

on Pharmaceutical Technology, (APV: APGI), Budapest, pp. 491-492.

514

Nayak, A. K., & Pal, D., (2011). Development of pH-sensitive tamarind seed

515

polysaccharide–alginate composite beads for controlled diclofenac sodium delivery using

516

response surface methodology. International Journal of Biological Macromolecules, 49,

517

784–793.

518

Neslihan, G. R., & Benita, S. (2004). Self-emulsifying drug delivery systems (SEDDS) for

519

improved oral delivery of lipophilic drugs. Biomedicine & Pharmacotherapy, 58, 173–182.

Ac ce p

te

d

M

an

us

cr

ip t

499

30 Page 30 of 33

Oguchi, T., Sasaki, N., Hara, T., Tozuka, Y., & Yamamoto, K. (2003). Differentiated thermal

521

crystallization from amorphous chenodeoxycholic acid between the ground specimens

522

derived from the polymorphs. International Journal of Pharmaceutics, 253, 81–88.

523

Paudel, A., Worku, Z.A., Meeus, J., Guns, S., & Van den, M. G. (2013). Manufacturing of

524

solid dispersions of poorly water soluble drugs by spray drying: Formulation and process

525

considerations. International Journal of Pharmaceutics, 453, 253-84.

526

Raghavan, S. L., Trividic, A., Davis, A. F., & Hadgraft, J. (2001). Crystallization of

527

hydrocortisone acetate: influence of polymers. International Journal of Pharmaceutics, 212,

528

213–221.

529

Schultheiss, N., & Newman, A. (2009). Pharmaceutical cocrystals and their physicochemical

530

properties. Crystal Growth & Design, 9, 2950–2967.

531

Serajuddin, A. T. M. (2007). Salt Formation to Improve Drug Solubility. Advance Drug

532

Delivery Reviews. 59, 603–616.

533

Stephenson, G. A., Aburub, A., & Woods, T. A. (2011). Physical stability of salts of weak

534

bases in the solid-state. Journal of Pharmaceutical Sciences, 100, 1607–1617.

535

Tiong, N., & Elkordy, A. A. (2009). Effects of liquid solid formulations on dissolution of

536

naproxen. European Journal of Pharmaceutics and Biopharmaceutics, 73, 373–384.

537

Van, E. B., Froyen, L., Martens, J. A., Blaton, N., Augustijns, P., Brewster, M., & Vanden,

538

M. G. (2007). Characterization of physico-chemical properties and pharmaceutical

539

performance of sucrose co-freeze-dried solid nanoparticulate powders of the anti-HIV agent

540

loviride prepared by media milling. International Journal of Pharmaceutics, 338, 198–206.

541

Xia, D., Quan, P., Piao, H., Piao, H., Sun, S., Yin, Y., & Cui, F. (2010). Preparation of stable

542

nitrendipine

Ac ce p

te

d

M

an

us

cr

ip t

520

nanosuspensions

using

the

precipitation–ultrasonication

method

for

31 Page 31 of 33

543

enhancement

544

of Pharmaceutical Sciences, 40, 325–334

545

Xiong, R., Lu, W., Li, J., Wang, P., Xu, R., & Chen, T. (2008). Preparation and

546

characterization of intravenously injectable nimodipine nanosuspension. International

547

Journal of Pharmaceutics, 350, 338–343.

548

Yang, R., Chen, J. B., Xiao, C. F., Liu, Z. C., Gao, Z. Y., Yan, S. J., Zhang, J. H., Zhang, H.

549

B., & Lin, J. (2014). Inclusion complex of GA-13316 with β-cyclodextrin: Preparation,

550

characterization, molecular modeling, and in vitro evaluation. Carbohydrate Polymers, 111,

551

655-662.

552

Yao, Q., Tao, X., Tian, B., Tang, Y., Shao, Y., Kou, L., Gou, J., Li, X., Yin, T., & Tang, X.

553

(2014). Improved oral bioavailability of core–shell structured beads by redispersion of the

554

shell-forming nanoparticles: Preparation, characterization and in vivo studies. Colloids and

555

Surfaces B: Biointerfaces, 113, 92– 100.

556

Zimmermann, A., Millqvist-Fureby, A., Elema, M. R., Hansen, T., Müllertz, A., &

557

Hovgaard, L. (2009). Adsorption of pharmaceutical excipients onto microcrystals of

558

siramesine hydrochloride: effects on physicochemical properties. European Journal of

559

Pharmaceutics and Biopharmaceutics, 71, 109–116.

oral

bioavailability.

European Journal

ip t

and

te

d

M

an

us

cr

dissolution

Ac ce p

560

of

32 Page 32 of 33

560

Highlights  HPMC stabilized nanosuspensions have optimized to improve dissolution of Naproxen.

562

 Controlled precipitation-ultrasonication technique adopted.

563

 Mean particle size, % drug content, and dissolution time correlated with HPMC.

564

 Response surface methodology have adopted for formulation optimization.

ip t

561

Ac ce p

te

d

M

an

us

cr

565

33 Page 33 of 33

Formulation and process optimization of naproxen nanosuspensions stabilized by hydroxy propyl methyl cellulose.

In present study precipitation-ultrasonication was used to obtain nanosuspensions of poorly water-soluble drug, naproxen (NPX). We investigated the ef...
755KB Sizes 0 Downloads 11 Views