Impact of polymer type on bioperformance and physical stability of hot melt extruded formulations of a poorly water soluble drug

Impact of polymer type on bioperformance and physical stability of hot melt extruded formulations of a poorly water soluble drug

Accepted Manuscript Title: IMPACT OF POLYMER TYPE ON BIOPERFORMANCE AND PHYSICAL STABILITY OF HOT MELT EXTRUDED FORMULATIONS OF A POORLY WATER SOLUBLE...

536KB Sizes 7 Downloads 69 Views

Accepted Manuscript Title: IMPACT OF POLYMER TYPE ON BIOPERFORMANCE AND PHYSICAL STABILITY OF HOT MELT EXTRUDED FORMULATIONS OF A POORLY WATER SOLUBLE DRUG Author: Amitava Mitra Li Li Patrick Marsac Brian Marks Zhen Liu Chad Brown PII: DOI: Reference:

S0378-5173(16)30236-8 http://dx.doi.org/doi:10.1016/j.ijpharm.2016.03.036 IJP 15634

To appear in:

International Journal of Pharmaceutics

Received date: Revised date: Accepted date:

23-10-2015 13-3-2016 20-3-2016

Please cite this article as: Mitra, Amitava, Li, Li, Marsac, Patrick, Marks, Brian, Liu, Zhen, Brown, Chad, IMPACT OF POLYMER TYPE ON BIOPERFORMANCE AND PHYSICAL STABILITY OF HOT MELT EXTRUDED FORMULATIONS OF A POORLY WATER SOLUBLE DRUG.International Journal of Pharmaceutics http://dx.doi.org/10.1016/j.ijpharm.2016.03.036 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 2 3 4 5 6 7 8 9 10 11 12

IMPACT OF POLYMER TYPE ON BIOPERFORMANCE AND PHYSICAL STABILITY OF HOT MELT EXTRUDED FORMULATIONS OF A POORLY WATER SOLUBLE DRUG Amitava Mitraa* [email protected], Li Lib, Patrick Marsacc,e, Brian Marksb, Zhen Liuc, Chad Brownd a Biopharmaceutics, Pharmaceutical Sciences and Clinical Supply, Merck & Co. Inc. b Analytical Sciences, Pharmaceutical Sciences and Clinical Supply, Merck & Co. Inc. c Preformulation, Pharmaceutical Sciences and Clinical Supply, Merck & Co. Inc. d Formulation Sciences, Pharmaceutical Sciences and Clinical Supply, Merck & Co. Inc. e Current Affiliation: College of Pharmacy, University of Kentucky * Corresponding Author at: Merck & Co., Inc, West Point, PA-19486, USA; Tel.: +1 215 652 8551; Fax: +1 215 993 1245

1

12 13 14

Graphical abstract

15 16 17 18 19

2

19

Abstract

20

Amorphous solid dispersion formulations have been widely used to enhance bioavailability of poorly

21

soluble drugs. In these formulations, polymer is included to physically stabilize the amorphous drug by

22

dispersing it in the polymeric carrier and thus forming a solid solution. The polymer can also maintain

23

supersaturation and promote speciation during dissolution, thus enabling better absorption as compared to

24

crystalline drug substance. In this paper, we report the use of hot melt extrusion (HME) to develop

25

amorphous formulations of a poorly soluble compound (FaSSIF solubility = 1 μg/mL). The poor

26

solubility of the compound and high dose (300 mg) necessitated the use of amorphous formulation to

27

achieve adequate bioperformance. The effect of using three different polymers (HPMCAS-HF,

28

HPMCAS-LF and copovidone), on the dissolution, physical stability, and bioperformance of the

29

formulations was demonstrated. In this particular case, HPMCAS-HF containing HME provided the

30

highest bioavailability and also had better physical stability as compared to extrudates using HPMCAS-

31

LF and copovidone. The data demonstrated that the polymer type can have significant impact on the

32

formulation bioperformance and physical stability. Thus a thorough understanding of the polymer choice

33

is imperative when designing an amorphous solid dispersion formulation, such that the formulation

34

provides robust bioperformance and has adequate shelf life.

35 36 37

Keywords: hot melt extrusion; dissolution; pharmacokinetics; stability; solid dispersion; anti-nucleation

38

3

38

1. INTRODUCTION

39

The use of amorphous solid dispersion (ASD) formulations to enhance bioavailability of poorly soluble

40

drugs has been widely published (Serajuddin, 1999; Newman et al. 2012; Paudel et al. 2013; Lang et al.

41

2014). The ASDs typically enhance bioavailability due to higher kinetic solubility of the drug substance

42

and increased dissolution rate of the formulation, by the virtue of the fact that the drug molecule exists in

43

the formulation in a high energy amorphous state. The hot melt extrusion (HME) process has been

44

successfully used in pharmaceutical applications to produce ASDs and several of these products have

45

been approved by the FDA including Noxafil®, Kaletra™, and Norvir™ (Lang et al. 2014; Crowley et al.

46

2007; Repka et al. 2007). Briefly, in the HME process the drug substance and stabilizing polymer are melt

47

compounded in an extruder forming a solid solution. Upon exiting the extruder the molten mixture is

48

quickly quenched such that the temperature drops below its glass transition temperature thus kinetically

49

inhibiting recrystallization. These extrudates are then processed to produce the final tablet product. While

50

the HME process has certainly been a great addition in the pharmaceutical scientist’s repertoire to

51

formulate poorly soluble drugs, the successful development of a product using HME is determined by

52

careful consideration of material properties (drug and polymer), process (temperature, shear) and

53

equipment design. For a more detailed description of the HME process and operations, interested readers

54

are referred to several in-depth reviews on this topic (Lang et al. 2014; Crowley et al. 2007; Repka et al.

55

2007). Most often, unique formulations yield unique physical stability, dissolution performance, and

56

ultimately bioperformance.

57

particularly complex – each formulation may require unique processing conditions given the differences

58

in the material properties and the associated phase diagrams. A balance must be struck between

59

processing, physical stability, and measures of in vitro performance without compromising

60

bioperformance. Although there are reports which focus on physical stability, dissolution, and other

The number of formulation options makes the production of ASDs

4

61

measures of in-vitro performance (Ilevbare et al. 2013; Sarode et al. 2014), relatively few reports

62

highlight the influence of formulation on bioperformance.

63 64

The amorphous nature of the active pharmaceutical ingredient (API) could lead to physical instability in

65

the drug product such as conversion to the crystalline state. One common approach to physically stabilize

66

the amorphous drug is to disperse the API in a polymeric carrier and thus form a solid solution of the drug

67

and the polymer. Another goal of using the polymer matrix is to maintain the supersaturation achieved

68

during dissolution over an extended period of time so as to better enable absorption of the solubilized API

69

i.e. the higher energy amorphous form of the drug substance transiently increases solubility relative to that

70

of the stable crystalline form and the polymer inhibits nucleation and crystal growth and maintains

71

supersaturation for an extended time period (Guzman et al. 2007; Brouwers et al. 2009; Augustijns et al.

72

2012). The polymer can also promote speciation during dissolution, which also would enhance

73

bioperformance of the formulation (Friesen et al. 2008). Several polymers have been reported in literature

74

for use in pharmaceutical ASDs, interested readers are referred to the following references (Paudel et al.

75

2013; Lang et al. 2014; Konno et al. 2008; Curatolo et al. 2009; Rumondor et al. 2009; Tajarobi et al.

76

2011).

77 78

In this paper, we report the development of an HME formulation, in-vitro characterization including

79

dissolution and physical stability, as well as preclinical pharmacokinetics (PK) data for Merck compound

80

A. In addition, we also report the impact of three different polymers used in the HME formulation-

81

Copovidone, HPMCAS-HF, and HPMCAS-LF, on the physical stability and bioperformance. Compound

82

A is a low solubility and high permeability (BCS class 2) compound (Table 1) with a fairly high

83

efficacious dose projection of approximately 300 mg. This results in a very high dose to volume ratio i.e.

84

high dose number (Do = dose/FaSSIF solubility/250 mL) of 1200 indicating significant solubility limited 5

85

absorption for this compound (Oh et al. 1993). Hence there was a need to develop an enabled formulation

86

such as an ASD so as to transiently increase the concentration in solution and the dissolution rate. Further,

87

the data shown in this paper also demonstrates that the choice of polymer can have a significant impact on

88

the performance (physical stability and bioavailability) of the ASD formulation. It is the aim of this

89

publication to highlight the influence that formulation selection has on bioperformance and physical

90

stability of amorphous formulation so as to facilitate improved approaches and methodologies employed

91

in the careful balance between process, formulation, and performance.

92 93

2. MATERIALS and METHODS

94

2.1 Preparation of Hot Melt Extrusion (HME) formulations of compound A:

95

The melting point (Tm) of compound A is approximately 140°C and it is thermally stable up to

96

approximately 220°C by thermal gravimetric analysis (TGA), making the compound a prime candidate

97

for HME. Thus, formulations of compound A were extrusion compounded at a 20% drug load in a custom

98

built co-rotating 7.5 mm twin screw extruder with L/D=15 and 1 cm slit die (MP&R, Hackensack, NJ)

99

with three individual polymers- copovidone (Kollidon VA-64™, BASF), hydroxypropyl methyl cellulose

100

LF grade (HPMCAS-LF, Shin Etsu), hydroxypropyl methyl cellulose HF grade (HPMCAS-HF, Shin

101

Etsu). These three polymers were chosen based on high-throughput screening to assess compatibility of

102

the drug and the polymer. This was achieved by film-casting of the drug with several polymers, and

103

analysis of the film casts by XRD, DSC and dissolution studies (data not shown). The extruder was

104

equipped with all conveying screws and heated to target a product temperature of 145°C to ensure facile

105

processing of each polymer. This temperature was above the Tm of compound A thus making this a facile

106

compounding process as the drug was completely melted. The screw speed was set at 50 revolutions per

107

minute. Approximately 7.5g of feed stock for each formulation was pre-blended in a turbula blender for

108

10 minutes prior to extrusion to help ensure compositional homogeneity. A VIBRI (SympaTec, Germany) 6

109

vibratory feeder was used to convey the formulation into the extruder. The gap width was set to 8 mm

110

and a V-shaped tray was used to convey the material to the feed port on the extruder. The vibration setting

111

was set at 35% to provide a feed rate of approximately 1 g/minute. Initial breakthrough of the extruded

112

formulation through the slit die (1 mm x 10 mm) was approximately 3.5 minutes after the start of feeding.

113

Strands of clear, glassy extrudate were collected on a custom built take-off belt equipped with a dual

114

nozzle cold air gun Vortec™ (AiRTX, Cincinnati, OH) to provide rapid quenching. Extrudates of each

115

composition (20% compound A and 80% polymer) were milled in a coffee grinder (Krups, Milville, NJ)

116

on the fine setting for approximately 30 seconds. The particle size of the extrudates was approximately

117

200 μm and 100 μm for the HPMCAS and copovidone based extrudates, respectively. Approximately 300

118

mg of extrudate (60 mg potency) were hand-filled into hard gelatin capsules (size 00) for dosing to beagle

119

dogs and for biorelevant dissolution testing. An overall yield of approximately 55% for the process was

120

achieved. The low yield from this process is primarily because of the small batch size (~7.5g), which

121

results in fixed losses such as approximately 2g loss in the extruder due to free volume and approximately

122

1g loss during milling. This yield is not representative of large scale HME process.

123 124

2.2 Physical characterization of the extrudates:

125

Milled extrudate were tested by X-ray diffraction (XRD) and differential scanning calorimetry (DSC) to

126

ensure a single phase, amorphous solid dispersion was formed. XRD was performed on a Philips X’Pert

127

with a 1 hour scan over a 2Ө of 2-40 (PANalytical, Westborough, MA). Modulated differential scanning

128

calorimetry (DSC) was conducted on a TA instrument Q2000 over a temperature range of 0˚C to 130˚C or

129

145˚C (TA Instruments, New Castle, DE). The heating rate was 2˚C /min with a modulation frequency of

130

±0.5˚C every 60 seconds. Solid dispersions prepared with HPMCAS-L and HPMCAS-H were placed on

131

stability at 40°C/35%RH and 40°C/75%RH in open containers and analyzed after 4 weeks of storage. The

132

copovidone systems were stored at 30°C/65%RH and 40°C/35%RH. For the stability studies a 7

133

combination of temperature and moisture was used to trigger physical instability in the HME formulation.

134

The reason of selecting those temperature and relative humidity combinations was to expedite instability

135

kinetics (phase separation and/or drug recrystallization) as well as ranking the physical stability of various

136

HME formulations. The differences in temperature and relative humidity conditions selected for

137

HPMCAS and copovidone formulations were deliberate, since copovidone is known to be significantly

138

hygroscopic so much so that it absorbs enough water at the 40°C/75%RH condition that the entire

139

dispersion coalesces into a single liquid mass (data not shown).

140 141

2.3 Dissolution studies of the formulations:

142

The dissolution studies were conducted in an USP apparatus II (Vankel VK 7000) using 500 mL fasted

143

state simulated intestinal fluid (FaSSIF, pH 6.5) at 37 °C and paddle speed of 100 rpm. Samples were

144

manually collected at pre-determined time intervals (15, 30, 60 and 120 minutes). 1 mL of sample was

145

ultracentrifuged (Beckman Coulter Optima TLX Ultracentrifuge) at 80,000 rpm for 15 minutes and was

146

diluted immediately with 500 uL of 50:50 v/v water: acetonitrile to prevent any further precipitation of

147

compound A from the dissolution medium. These samples were analyzed by reverse phase HPLC using a

148

mobile phase of 30% 0.1% H3PO4 and 70% acetonitrile at a flow rate of 5 mL/minute, UV detection at

149

260 nm and a Merck KGaA Chromolith SpeedROD 18e monolithic column. The compound A retention

150

time was 0.4 minutes.

151 152

2.4 Dog study and pharmacokinetic analysis:

153

In order to investigate the bioperformance of the HME formulations, pharmacokinetic studies were

154

conducted in male beagle dogs at a dose of 5 mg/kg (equivalent to the projected human dose of 300 mg).

155

After an overnight fast, the dogs were dosed with one capsule each containing HPMCAS-HF, HPMCAS-

156

LF or copovidone based HME formulation or one conventional dry filled capsule (DFC) formulation 8

157

containing the crystalline form of compound A, followed by 3.5 mL/kg water rinse. Water was restricted

158

for 1 hour post dose. Food was returned at 4 hours after dosing. 1 mL blood sample was drawn from a 21g

159

catheter placed in the cephalic vein into EDTA tubes at pre-dose and 0.25, 0.5, 1, 2, 4, 6, 8, and 24 hours

160

after dosing. The plasma was separated by centrifugation (10 minutes at 2500 rpm) and kept frozen at -

161

70ºC until analysis. Concentrations of compound A in dog plasma were quantified by LC-MS/MS

162

analysis. All studies were conducted under a protocol approved by the Merck IACUC.

163 164

Area under the curve (AUC0-24hr), observed maximum plasma concentration (Cmax), and time of Cmax

165

(Tmax) were calculated using the linear trapezoidal, non-compartmental model in WinNonLin v5.2

166

(Certara, Princeton, NJ). Plasma concentration values below LOQ were set at zero for PK calculation

167

purposes.

168 169

3. RESULTS

170

3.1 Physical characterization:

171

The XRD and DSC profiles of the extrudates are shown in Figures 1 and 2, respectively. All three HME

172

formulations were found to be x-ray amorphous at the initial time-point, indicating the successful

173

formation of amorphous solid dispersions. The modulated DSC profiles also showed a single glass

174

transition temperature (Tg) of approximately 73°C, 74°C and 81°C for the HME formulations using

175

HPMCAS-HF, HPMCAS-LF and copovidone polymers, respectively. These were well above the Tg of

176

the pure drug which resides at about 24°C. As confirmed by the XRPD and DSC results, the ASDs

177

produced by HME were x-ray amorphous and displayed no amorphous phase separation for any of these

178

formulations at the initial time-point.

179

After stressing these HME formulations for four weeks under different conditions clear distinction

180

between physical stability of each formulation was observed. The HPMCAS-HF based HME maintained 9

181

a single amorphous phase at 40°C/75% relative humidity (RH) open dish condition (Figures 1 and 2).

182

The HPMCAS-LF based HME showed subtle signs of crystallization at 40°C/75% RH open dish

183

condition but was amorphous at 40°C/35% RH open dish. Peaks at 2θ of 17.78 to 18.40 were used to

184

determine drug recrystallization for HPMCAS-LF based HME. For better visualization, a close up of this

185

region is shown as inset in Figure 1. The copovidone based HME showed signs of crystallization at

186

30°C/65% RH open dish condition and showed signs of amorphous-amorphous phase separation (by

187

modulated DSC) at 40°C/35% RH open dish condition. These data highlights the need to consider both

188

crystallization and amorphous phase behavior since it is reasonable to consider the amorphous phase

189

separation of multi-component amorphous systems as a precursor to crystallization. It should be noted

190

that the crystalline anhydrous form of compound A used to make the HME formulations was shown to be

191

highly crystalline, non-hygroscopic, physically and chemically stable at room temperature and the

192

relevant processing conditions (data not shown).

193 194

3.2 Dissolution data:

195

The in vitro dissolution showed that the crystalline compound A DFC formulation only achieved a

196

concentration of approximately 1.4 μg/mL at 60 minutes (Figure 3), which was consistent with the

197

FaSSIF solubility of the crystalline form (1 μg/mL). In contrast, at 60 minutes the HME formulations

198

showed dissolution of approximately 13.3, 4.7 and 5.3 μg/mL for HPMCAS-HF, HPMCAS-LF and

199

copovidone, respectively (Figure 3). The dissolution data also clearly demonstrated that HPMCAS-HF

200

can achieve and maintain a higher level of free drug concentration than HPMCAS-LF and copovidone

201

polymers with highest concentrations achieved of approximately 16.4, 14.7, 7.1 μg/mL at 15 minutes,

202

respectively.

203 204 10

205

3.3 Pharmacokinetics in dogs:

206

Table 2 and figure 4 summarize the mean pharmacokinetic parameters and plasma concentration profiles,

207

respectively, after oral administration of compound A formulations in beagle dogs. These data clearly

208

demonstrated that the HME formulations provided significantly higher bioavailability as compared to the

209

dry filled capsule (DFC) formulation containing crystalline form of compound A. The PK data were

210

consistent with the biorelevant dissolution data shown in figure 3. For example in the dissolution study,

211

compound A concentration at 60 minutes was 0.3-fold for the DFC, as compared to the copovidone based

212

HME formulation (Table 3). Similarly, the relative bioavailability of compound A from the dog PK study

213

was 0.2 for DFC as compared to copovidone HME. Of the three HME formulations evaluated in this PK

214

study, the copovidone based HME showed the worst bioperformance, with the HPMCAS-HF and

215

HPMCAS-LF based formulations showing approximately 2.2 and 1.4-fold higher bioavailability,

216

respectively, when compared to the copovidone formulation. These PK data were also in agreement with

217

the biorelevant dissolution data, which showed that at 60 minutes compound A concentration was 2.5-fold

218

for HPMCAS-HF and 0.9-fold for HPMCAS-LF as compared to copovidone (Table 3).

219 220

4. DISCUSSION

221

ASDs are a very valuable formulation tool to enhance bioavailability of poorly soluble drugs and enable

222

development of compounds, which otherwise would not achieve adequate exposures in human. However

223

due to the high energy state of the amorphous drug, these formulations have an inherent physical stability

224

liability (due to crystallization of the amorphous drug or drug-polymer phase separation) during storage

225

and/or during dissolution. Hence, there is a lot of interest in identifying and selecting polymers that can

226

afford sufficient shelf-life to these formulations (Ilevbare et al. 2012; Ilevbare et al. 2013; Rumondor et al.

227

2009) as well as be able to maintain supersaturation (Konno et al. 2008; Curatolo et al. 2009; Sarode et al.

228

2014) and promote speciation (Friesen et al. 2008) during dissolution so that greater bioavailability can be 11

229

achieved. In addition, mechanistic understanding of the interaction of the polymer with the drug substance

230

is also critical in gaining an understanding of the formulation behavior as well as for rational formulation

231

design to achieve adequate performance (Curatolo et al. 2009; Ilevbare et al. 2012; Friesen et al. 2008). In

232

this paper, we report the comparison of three different polymer types on physical stability and

233

bioperformance of HME formulations of an extremely poorly soluble compound. These results highlight

234

the importance of robust formulation design to ensure adequate product bioperformance and shelf life.

235 236

The physical stability, dissolution and PK data we have shown here clearly indicates that the polymers

237

used have an influence on the performance of the HME formulations, with HPMCAS-HF showing the

238

best overall performance in this particular case. It is obvious from the dissolution data that amorphous

239

compound A has a strong tendency to crystallize during dissolution (Figure 3). Hence the selection of

240

polymer to be used in the HME formulation is of critical importance to sustain supersaturation of the

241

metastable amorphous drug, which in turn is a key driver for enhancing bioperformance. The dissolution

242

profile is characterized by an initial drug supersaturation, followed by nucleation and/or recrystallization,

243

resulting in a decrease in free drug concentration. The dissolution data indicated that the crystallization

244

inhibition performance was in the order of HPMCAS-HF > HPMCAS-LF > copovidone. The HPMCAS-

245

HF and LF systems were able to achieve an apparent solubility of 16.4 and 15.7 ug/ml at 15 minutes,

246

respectively, representing a degree of supersaturation (Ds = solubility of an amorphous drug/solubility of

247

crystalline drug) of approximately 15. In contrast, the copovidone based system showed lower degree of

248

supersaturation of approximately 7. While all the HME formulations showed indications of initial

249

supersaturation followed by precipitation, HPMCAS-HF was able to maintain higher degree of drug

250

supersaturation during the entire dissolution process as compared to the other two polymer systems. These

251

dissolution data also indicated that in this particular case the extrudate particle size had minimal impact on

252

their performance. This was demonstrated by the fact that the copovidone extrudates had smaller particle 12

253

size (~100 μm) as compared to the HPMCAS extrudates (~200 μm), however HPMCAS extrudates

254

showed better dissolution than copovidone. The dissolution data was also corroborated with PK data in

255

dogs. The XRD and DSC data also demonstrated that the HPMCAS based HME had superior physical

256

stability than the copovidone HME (Figures 1 and 2). These results are in agreement with previous

257

studies that have shown that HPMCAS in general is superior to copovidone at both achieving high levels

258

of supersaturation as well as inhibiting the recrystallization of dissolved drugs in solution (Konno et al.

259

2008; Yin et al. 2014; Ilevbare et al. 2012; Ilevbare et al. 2013; Sarode et al. 2014), which can

260

significantly enhance bioperformance of these formulations. In addition HPMCAS is better at resisting

261

moisture-induced crystallization of amorphous solid dispersions during storage, as compared to other

262

polymers such as PVP (Friesen et al. 2008; Rumondor et al. 2009). The better antinucleation properties of

263

HPMCAS as compared to copovidone are often attributed to its unique physicochemical characteristics

264

(Ting et al. 2015; Curatolo et al. 2009; Ilevbare et al. 2012). HPMCAS is a cellulose derivative of two

265

ethers - methoxy and hydroxypropoxy as well as two esters - acetate and succinate. The hydrophobic

266

acetate groups are thought to facilitate the molecular dispersions of hydrophobic drugs within the

267

polymeric matrix through hydrophobic-hydrophobic interactions, and the small amount of unreacted

268

hydrophilic hydroxyls allows sufficient hydration of the resulting ASDs in solution. In addition, the

269

carboxylic acids from succinates (pKa ~ 5) are ionized at intestinal pH (approximately pH 6-7) and these

270

negative charges can stabilize polymer - drug nanostructures that are formed during dissolution, through

271

repulsive surface charge, which can be important in preventing crystallization of the dissolved drug

272

(Friesen et al. 2008). This amphiphilicity is not present in a hydrophilic polymer such as copovidone.

273

Hence it cannot provide sufficient stabilization of supersaturation through hydrophobic interactions or

274

repulsive charge (Ting et al. 2015). In addition, copovidone has the lowest glass transition temperature at

275

around 105°C and it is the most hygroscopic of the polymers studied, these properties also reduce its anti-

276

nucleation abilities. 13

277 278

The dissolution and physical stability data also clearly showed that HPMCAS-HF was further

279

differentiated from HPMCAS-LF in its ability to better inhibit recrystallization of compound A, in spite

280

of the structural similarity between the two polymers. We believe that enhanced hydrophobicity of

281

HPMCAS-HF is responsible for the superior crystallization inhibition of compound A. In fact, Ueda et al.

282

(Ueda et al. 2013; Ueda et al. 2014) have shown that HPMCAS-HF inhibited the recrystallization of

283

several poorly soluble drugs (e.g. carbamazepine) more strongly than HPMCAS-LF. Solution NMR

284

studies showed that the molecular mobility of these drugs were clearly suppressed in the HPMCAS-HF

285

solution compared to that in the HPMCAS-LF solution. These studies also revealed strong interaction of

286

the drug with the acetate substituent of HPMCAS, however interaction with the succinate substituent was

287

quite small. Since HPMCAS-HF is more hydrophobic than HPMCAS-LF due to its higher ratio of acetate

288

to succinate, this might be responsible for the stronger hydrophobic interaction between HPMCAS-HF

289

and poorly soluble drugs. Similar intermolecular hydrophobic interactions between the drug and

290

HPMCAS-HF are most likely responsible for substantially better inhibition of recrystallization of

291

compound A during dissolution and better physical stability, as compared to HPMCAS-LF.

292 293

Finally, PK studies in dogs demonstrated that bioperformance of these formulations were in agreement

294

with the in-vitro dissolution data. A valid question when using preclinical animal models for formulation

295

screening prior to human studies is whether metabolic and/or physiological differences between species

296

can lead to different formulation performance. However this will appear unlikely for Compound A.

297

Preclinical studies using liver microsome suggested similar primary metabolic route (CYP3A4) in both

298

dogs and humans and the compound exhibited low in vivo clearance in dogs (plasma clearance = 3.1

299

mL/min/kg). While no intravenous data is available in the clinic to allow for calculation of clearance, the

300

predicted clearance in human is also low (< 5 mL/min/kg). These data suggested that there are minimal 14

301

metabolic differences for compound A in dogs and human. It should be also noted that given the high

302

permeability (BCS 2) of the compound, differences in intestinal permeability between species is also

303

unlikely to contribute to differential formulation behavior in the preclinical model and in the clinic. Thus

304

it could be assumed that dog is an appropriate species to investigate the formulation bioperformance and

305

guide formulation development for clinical evaluation. In addition, dogs are generally considered to be an

306

appropriate species for screening formulations for clinical evaluation due to similarities in gastrointestinal

307

physiology (Kararli 1995; Hasiwa et al. 2011) and there are numerous literature examples of use of dogs

308

to successfully guide formulation development (Miller et al. 2015; Jang & Kang, 2014; Zheng et al.,

309

2007). As is seen in table 3, relative bioavailability of the formulations in dog were very close to the

310

relative concentrations achieved at 60 minutes in the biorelevant dissolution study. These data showed

311

that the ASD formulations achieved significantly higher exposures of compound A as compared to the

312

DFC formulation containing crystalline form of the drug substance. These results are similar to several

313

previous publications (Lakshman et al. 2008; He et al. 2010; Newman et al. 2012; Lang et al. 2014),

314

which have demonstrated the use of HME based ASD formulations to enhance bioavailability of poorly

315

soluble drugs. In addition, clear PK (AUC0-24hr and Cmax) differentiation was observed between the three

316

ASD formulations in these dog studies (Table 2), with the HPMCAS-HF based HME showing the highest

317

bioavailability. This indicated that the dissolution differences observed between the ASD formulations

318

were significant enough to result in differences in the extent and rate of absorption of compound A in

319

dogs. The prolonged Tmax observed for HPMCAS-LF and copovidone based systems as compared to

320

HPMCAS-HF (4 hours vs. 1 hour) cannot be explained with the current data and might indicate the need

321

for further mechanistic understanding of the in-vivo behavior of these formulations. The data presented

322

here demonstrate that not only the type of polymer used (e.g. HPMCAS vs. copovidone) but also the

323

substituents on HPMCAS (e.g. HPMCAS-HF vs. HPMCAS-LF) can have a profound effect on the

15

324

performance (physical stability and bioavailability) of an ASD formulation. As a result, careful

325

consideration should be given on the choice of polymer used as a matrix in ASD formulations.

326 327

5. CONCLUSION

328

The data shown here demonstrate the effect of polymer type (e.g. HPMCAS vs. copovidone) on

329

bioperformance and physical stability of a hot melt extruded formulation. It was further shown that the

330

substituents on HPMCAS (e.g. HPMCAS-HF vs. HPMCAS-LF) had profound effect on the performance

331

of the HME. In this particular case HPMCAS-HF containing HME provided the highest bioavailability in

332

dogs and also had better physical stability as compared to extrudates with HPMCAS-LF and copovidone.

333

These data highlight the influence that polymer selection can have on the performance of amorphous

334

formulations. Thus it is imperative that a thorough understanding of the interaction of polymer with drug

335

substance is gained during formulation design. This will enable rational formulation design to achieve

336

adequate product bioperformance and shelf life.

337 338

ACKNOWLEDGEMENTS

339

The authors would like to thank Kim Manser and Becky Nissley for conducting the dog studies.

340

16

340

REFERENCES

341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384

Augustijns, P., Brewster, M.E. 2012. Supersaturating drug delivery systems: fast is not necessarily good enough. J. Pharm. Sci. 101, 7-9. Brouwers, J., Brewster, M.E., Augustijns, P. 2009. Supersaturating drug delivery systems: the answer to solubility-limited oral bioavailability? J. Pharm. Sci. 98, 2549-2572. Crowley, M.M., Zhang, F., Repka, M.A., Thumma, S., Upadhye, S.B., Battu, S.K., McGinity, J.W., Martin, C. 2007. Pharmaceutical applications of hot-melt extrusion: part I. Drug Dev. Ind. Pharm. 33, 909-926. Curatolo, W., Nightingale, J.A., Herbig, S.M. 2009. Utility of hydroxypropyl methyl cellulose acetate succinate (HPMCAS) for initiation and maintenance of drug supersaturation in the GI milieu. Pharm. Res. 26, 1419-1431. Friesen, D.T., Shanker, R., Crew, M., Smithey, D.T., Curatolo, W.J., Nightingale, J.A. 2008. Hydroxypropyl methylcellulose acetate succinate-based spray-dried dispersions: an overview. Mol. Pharm. 5, 1003-1019. Guzmán, H.R., Tawa, M., Zhang, Z., Ratanabanangkoon, P., Shaw, P., Gardner, C.R., Chen, H., Moreau, J.P., Almarsson, O., Remenar, J.F. 2007. Combined use of crystalline salt forms and precipitation inhibitors to improve oral absorption of celecoxib from solid oral formulations. J. Pharm. Sci. 96, 26862702. Hasiwa, N., Bailey, J., Clausing, P., Daneshian, M., Eileraas, M., Farkas, S., Gyertyán, I., Hubrecht, R., Kobel, W., Krummenacher, G., Leist, M., Lohi, H., Miklósi, A., Ohl, F., Olejniczak, K., Schmitt, G., Sinnett-Smith, P., Smith, D., Wagner, K., Yager, J.D., Zurlo, J., Hartung, T. 2011. Critical evaluation of the use of dogs in biomedical research and testing in Europe. ALTEX. 28, 326-340. He, H., Yang, R., Tang, X. 2010. In vitro and in vivo evaluation of fenofibrate solid dispersion prepared by hot melt extrusion. Drug Dev. Ind. Pharm. 36, 681-687. Ilevbare, G.A., Liu, H., Edgar, K.J., Taylor, L.S. 2012. Understanding polymer properties important for crystal growth inhibition impact of chemically diverse polymers on solution crystal growth of ritonavir. Cryst. Growth Des.12, 3133–3143. Ilevbare, G.A., Liu, H., Edgar, K.J., Taylor, L.S. 2013. Impact of polymers on crystal growth rate of structurally diverse compounds from aqueous solution. Mol. Pharm. 10, 2381-2393. Jang, S.W., Kang, M.J. 2014. Improved oral absorption and chemical stability of everolimus via preparation of solid dispersion using solvent wetting technique. Int J Pharm. 473, 187-193. Kararli, T. 1995. Comparison of the gastrointestinal anatomy, physiology, and biochemistry of humans and commonly used laboratory animals. Biopharm. & Drug Disposition. 16, 351-380.

17

385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432

Konno, H., Handa, T., Alonzo, D.E., Taylor, L.S. 2008. Effect of polymer type on the dissolution profile of amorphous solid dispersions containing felodipine. Eur. J. Pharm. Biopharm. 70, 493-499. Lakshman, J.P., Cao, Y., Kowalski, J., Serajuddin, A.T. 2008. Application of melt extrusion in the development of a physically and chemically stable high-energy amorphous solid dispersion of a poorly water-soluble drug. Mol. Pharm. 5, 994-1002. Lang, B., McGinity, J.W., Williams, R.O. 3rd. 2014. Hot-melt extrusion-basic principles and pharmaceutical applications. Drug Dev. Ind. Pharm. 40, 1133-1155. Miller, D.A., Keen, J.M., Brough, C., Ellenberger, D.J., Cisneros, M., Williams, R.O. 3rd, McGinity, J.W. 2015. Bioavailability enhancement of a BCS IV compound via an amorphous combination product containing ritonavir. J Pharm Pharmacol. 1-14. Newman, A., Knipp, G., Zografi, G. 2012. Assessing the performance of amorphous solid dispersion. J. Pharm. Sci. 101, 1355-1377. Oh, D.M., Curl, R.L., Amidon, G.L. 1993. Estimating the fraction dose absorbed from suspensions of poorly soluble compounds in humans: a mathematical model. Pharm. Res. 10, 264-270. Paudel, A., Worku, Z.A., Meeus, J., Guns, S., Van den Mooter, G. 2013. Manufacturing of solid dispersions of poorly water soluble drugs by spray drying: formulation and process considerations. Int. J. Pharm. 453, 253-284. Repka, M.A., Battu, S.K., Upadhye, S.B., Thumma, S., Crowley, M.M., Zhang, F., Martin, C., McGinity, J.W. 2007. Pharmaceutical applications of hot-melt extrusion: Part II. Drug Dev Ind Pharm. 33, 10431057. Rumondor, A.C., Stanford, L.A., Taylor, L.S. 2009. Effects of polymer type and storage relative humidity on the kinetics of felodipine crystallization from amorphous solid dispersions. Pharm. Res. 26, 25992606. Sarode, A.L., Wang, P., Obara, S., Worthen, D.R. 2014. Supersaturation, nucleation, and crystal growth during single and biphasic dissolution of amorphous solid dispersions: polymer effects and implications for oral bioavailability enhancement of poorly water soluble drugs. Eur. J. Pharm. Biopharm. 86, 351-360. Serajuddin, A. T. 1999. Solid dispersion of poorly water-soluble drugs: early promises, subsequent problems, and recent breakthroughs. J. Pharm. Sci. 88, 1058-1166. Tajarobi, F., Larsson, A., Matic, H., Abrahmsén-Alami, S. 2011. The influence of crystallization inhibition of HPMC and HPMCAS on model substance dissolution and release in swellable matrix tablets. Eur. J. Pharm. Biopharm. 78, 125-133. Ting, J. M., Navale, T. S., Jones, S. D., Bates, F. S., Reineke, T. M. 2015. Deconstructing HPMCAS: Excipient design to tailor polymer–drug interactions for oral drug delivery. ACS Biomater. Sci. Eng. 1, 978-990. 18

433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448

Ueda, K., Higashi, K., Yamamoto, K., Moribe, K. 2013. Inhibitory effect of hydroxypropyl methylcellulose acetate succinate on drug recrystallization from a supersaturated solution assessed using nuclear magnetic resonance measurements. Mol. Pharm. 10, 3801-3811. Ueda, K., Higashi, K., Yamamoto, K., Moribe, K. 2014. The effect of HPMCAS functional groups on drug crystallization from the supersaturated state and dissolution improvement. Int. J. Pharm. 464, 205213. Yin, L., Hillmyer, M.A. 2014. Preparation and performance of hydroxypropyl methylcellulose esters of substituted succinates for in vitro supersaturation of a crystalline hydrophobic drug. Mol. Pharm. 11, 175185. Zheng, X., Yang, R., Zhang, Y., Wang, Z., Tang, X., Zheng L. 2007. Part II: bioavailability in beagle dogs of nimodipine solid dispersions prepared by hot-melt extrusion. Drug Dev Ind Pharm. 33, 783-789.

19

449 450 451

FIGURES CAPTION

452

Figure 1: X-ray diffraction of HME formulations of compound A, at initial time-point and after stressing

453

for 4 weeks.

454

Figure 2: Modulated DSC thermograms of HME formulations of compound A, at initial time-point and

455

after stressing for 4 weeks.

456

Figure 3: Dissolution of compound A formulations in fasted state simulated intestinal fluid (FaSSIF)

457

Figure 4: Mean plasma concentration versus time profiles of compound A in fasted male beagle dogs

458

(n=3, mean ± SE).

459 460

20

460 461

TABLE CAPTION

462

Table 1: Physicochemical properties of compound A

463

Table 2: Pharmacokinetic parameters (n=3, mean ± SE) of compound A in fasted male beagle dogs

464

following oral administration of several formulations.

465

Table 3: Comparison of dissolution of compound A formulations to their bioperformance in dogs.

466 467 468

21

468 469

470 471 472 473 474 475 476 477 478 479 480 481 482 483 484

Figure 1:

From bottom to top: HPMCAS-HF initial and after 4 weeks of storage at 40°C/75%RH open, HPMCAS-LF initial and after 4 weeks of storage at 40°C/35%RH open and 40°C/75%RH open, and copovidone initial and after 4 weeks of storage at 40°C/35%RH open and 30°C/65%RH open. All samples are at a drug loading of 20 wt% of compound A in polymer. Inset figure: Close-up of X-ray spectrum image focusing on crystalline peaks of compound A at 17.78 and 18.40 2θ angles (arrows), indicating significant drug crystallization in the copovidone based HME after 4 weeks at 30°C/65%RH open (top), subtle signs of drug crystallization in the HPMCAS-LF based HME formulation after 4 weeks at 40°C/75%RH open (middle) and no drug crystallization in the HPMCAS-HF based HME formulation after 4-week at 40°C/75%RH open (bottom).

22

484

485 486 487 488 489 490 491 492 493 494

Figure 2:

From bottom to top: HPMCAS-HF initial and after storage for 4 weeks at 40°C/75%RH open, HPMCASLF initial and after storage for 4 weeks at 40°C/35%RH open, and copovidone initial and after storage at 4 weeks at 40°C/35%RH open. All samples are at a drug loading of 20 wt% of compound A in polymer.

23

495 496 497

Figure 3:

498 499 500 501 502 503 504 505 506 507

24

507 508

Figure 4:

509 510 511 512 513 514 515 516

25

516 517 518 519

Tables Table 1: Physicochemical properties of compound A

520 Melting point (crystalline anhydrous form II) = 140°C Caco-2 permeability = 14.6 x 10-6 cm/sec Solubility (crystalline anhydrous form II): Simulated Gastric Fluid (SGF, pH 1.2) = 0.001 mg/mL Fasted State Simulated Intestinal Fluid (FaSSIF, pH 6.5) = 0.001 mg/mL Fed State Simulated Intestinal Fluid (FeSSIF, pH 5.0) = 0.002 mg/mL Water = 0.001 mg/mL 521 522 523 524 525 526

26

Table 2: Pharmacokinetic parameters (n=3, mean ± SE) of compound A in fasted male beagle dogs following oral administration of several formulations.

a

Formulation

Dose (mg/kg)

AUC0-24hr (µM*hr)

Dose normalized AUC0-24hr (µM*hr/mg/kg)

Cmax (µM)

Tmax (hr) a

Crystalline API in Dry Filled Capsule (DFC) c

15

4.76 ± 1.12

0.32

0.58 ± 0.11

2.0 (1.0-2.0)

HME (HPMCAS-HF)

5

18.1 ± 2.93

3.62

1.51 ± 0.34

HME (HPMCAS-LF)

5

11.2 ± 2.07

2.24

0.73 ± 0.08

HME (copovidone)

5

8.19 ± 2.91

1.64

0.59 ± 0.26

1.0 (0.5-4.0) 4.0 (1.0-24.0) 4.0 (2.0-6.0)

Tmax is reported as median with range in parenthesis Calculated using dog IV data (AUC0-24hr = 4.3 µM*hr at a dose of 1 mg/kg) c Crystalline jet milled compound A with mean particle size of 4.9 μm was used in the DFC formulation b

27

A Bioav

Table 3: Comparison of dissolution of compound A formulations to their bioperformance in dogs. Dissolution data Formulation

Crystalline API in Dry Filled Capsule (DFC) HME (HPMCAS-HF) HME (HPMCAS-LF) HME (copovidone) a

Relative bioavailability from dog PK study a

Concentration at 60 minutes (ug/mL)

Relative concentration a

1.46

0.3

0.2

13.3 4.7 5.3

2.5 0.9 --

2.2 1.4 --

Relative ratios were calculated with respect to copovidone based HME

28