Physiologically Based Absorption Modeling for Amorphous Solid


Physiologically Based Absorption Modeling for Amorphous Solid...

0 downloads 85 Views 2MB Size

Subscriber access provided by CARLETON UNIVERSITY

Article

Physiologically Based Absorption Modeling for Amorphous Solid Dispersion Formulations Amitava Mitra, Wei Zhu, and Filippos Kesisoglou Mol. Pharmaceutics, Just Accepted Manuscript • DOI: 10.1021/acs.molpharmaceut.6b00424 • Publication Date (Web): 21 Jul 2016 Downloaded from http://pubs.acs.org on July 23, 2016

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Molecular Pharmaceutics is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

Molecular Pharmaceutics

Physiologically Based Absorption Modeling for Amorphous Solid Dispersion Formulations

2

3

Amitava Mitra*#, Wei Zhu#, Filippos Kesisoglou#

4

Biopharmaceutics

5

Pharmaceutical Sciences and Clinical Supply, Merck & Co. Inc. #

6

All authors had equal contribution

7 8 9 10 11 12

*Corresponding Author

13

Amitava Mitra, PhD

14

Merck & Co., Inc

15

West Point, PA-19486, USA

16

Tel.: +1 215 652 8551

17

Fax: +1 215 993 1245

18

E-mail: [email protected]

19

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

20

Abstract

21

Amorphous solid dispersion (ASD) formulations are routinely used to enable the delivery of

22

poorly soluble compounds. This type of formulations can enhance bioavailability due to higher

23

kinetic solubility of the drug substance and increased dissolution rate of the formulation, by the

24

virtue of the fact that the drug molecule exists in the formulation in a high energy amorphous

25

state. In this paper we report the application of physiologically based absorption models to

26

mechanistically understand the clinical pharmacokinetics of solid dispersion formulations. Three

27

case studies are shown here to cover a wide range of ASD bioperformance in human and

28

modeling to retrospectively understand their in-vivo behavior. Case study 1 is an example of

29

fairly linear PK observed with dose escalation and the use of amorphous solubility to predict

30

bioperformance. Case study 2 demonstrates the development of a model that was able to

31

accurately predict the decrease in fraction absorbed (%Fa) with dose escalation thus

32

demonstrating that such model can be used to predict the clinical bioperformance in the scenario

33

where saturation of absorption is observed. Finally, case study 3 shows the development of an

34

absorption model with the intent to describe the observed incomplete and low absorption in

35

clinic with dose escalation. These case studies highlight the utility of physiologically based

36

absorption modeling in gaining a thorough understanding of ASD performance and the critical

37

factors impacting performance to drive design of a robust drug product that would deliver the

38

optimal benefit to the patients.

39 40 41

Keywords - Absorption Modeling, PBPK, Pharmacokinetics, Solid Dispersion, Amorphous, Dissolution

42

ACS Paragon Plus Environment

Page 2 of 38

Page 3 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

43

INTRODUCTION

44

The prediction of clinical performance of the formulations, based on preclinical data, is an

45

integral component of formulation development and a key deliverable for biopharmaceutics

46

scientists. In the last decade, physiologically based absorption modeling has attracted attention as

47

a means to achieve this preclinical to clinical translation. (1, 2, 3) Along with the advancement of

48

dissolution methods towards biorelevant measurements (4), efforts have been undertaken in the

49

field of physiologically based pharmacokinetic (PBPK) modeling to enable incorporation of

50

physicochemical and formulation characterization information into the models to simulate

51

absorption of drug compounds. (1) The principles of oral absorption PBPK models have been

52

reviewed in the literature along with a description of some of the models used in some of the

53

most common software such as such as GastroPlus (2), simCYP (3), PK-Sim (5) and

54

Intellipharm (6).

55

A review of the published literature on PBPK models for characterization or prediction of

56

formulation performance shows that the majority of publications have focused on immediate

57

release dosage forms or modified release (MR) dosage forms containing crystalline form of the

58

drug substance. For the former, a typical strategy is to describe the dissolution data using models

59

such as the Noyes-Whitney equation, and subsequent incorporation of the relevant information

60

into the absorption models. For example, a good number of publications have demonstrated the

61

successful application of these models to study the effect of particle size (7-10) or the impact of

62

dissolution rate as measured in-vitro (11-13), on absorption of these “conventional” dosage

63

forms. Similarly for MR formulations, given that the release rate from the dosage form is

64

typically the rate limiting step to absorption, direct incorporation of the measured in-vitro release

65

rates in the absorption model is employed. (10, 14-15) In addition, the development of more

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

66

robust dissolution models that accurately predict dissolution rates of low solubility molecules

67

have been recently reported. (16)

68

With the increase in the number of insoluble compounds in drug development pipelines (17),

69

often new formulation approaches are employed to achieve adequate bioavailability. Such

70

approaches may include the utilization of nanosuspensions, lipids or co-solvents, and amorphous

71

solid dispersions among others. (18) Among these approaches, amorphous solid dispersions

72

(ASD) have recently received increased attention with several new drugs on the market. (19, 20)

73

Amorphous solid dispersion (ASD) holds the promise of superior performance because of the

74

inherently higher energy state of active pharmaceutical ingredients (API) over the crystalline

75

counterpart. (21) The ASDs typically enhance bioavailability due to higher kinetic solubility of

76

the drug substance and increased dissolution rate of the formulation, by the virtue of the fact that

77

the drug molecule exists in the formulation in a high energy amorphous state. However, a higher

78

energy alone does not guarantee higher bioavailability. Amorphous materials have unique

79

challenges as compared to other more common approaches for increasing bioavailability due to

80

an increased risk of crystallization to a more stable crystalline form. Thus it would appear

81

beneficial to expand the application of physiologically based absorption modeling to these

82

formulations. However, there are not many published reports available on simulation of

83

absorption from these “enabled” dosage forms and from solid dispersions in particular. While the

84

principles of absorption from supersaturated solutions have been studied (22), publications on

85

applications of such models during product development are very limited. Gao et al. (23)

86

described the prediction of performance of a solid dispersion, among other formulations, for rat

87

toxicology studies while more recently Zheng et al, (24) discussed the utility of PBPK models in

88

the discovery space to drive utilization of a solid dispersion. To the best of our knowledge, there

ACS Paragon Plus Environment

Page 4 of 38

Page 5 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

89

have been no detailed reports of clinical data from solid dispersion formulations and application

90

of absorption models to describe the clinical data.

91

In this manuscript, we report three case studies attempting to highlight successes and challenges

92

in utilizing physiologically based absorption models for solid dispersion formulations. Given the

93

complexity of the dissolution behavior of solid dispersions (25), we focused on formulations that

94

could be categorized as supersaturating systems without evidence of complicated species

95

formation such as nanoparticles in in-vitro dissolution studies. The compounds exhibited varied

96

absorption and pharmacokinetic behavior ranging from complete absorption with linear

97

pharmacokinetics for case study 1 to incomplete and low absorption for case study 3. We discuss

98

the development of models either based on the in-vitro solubility and dissolution data or based

99

on extrapolation of models developed to first describe formulation performance in a preclinical

100

dog model. Extensive physical characterization of these ASDs was carried out to ensure that they

101

maintained their amorphous nature prior to in-vitro and in-vivo studies. However, discussion of

102

those data is outside the scope of this paper, since the focus here is on development of absorption

103

models.

104 105

MATERIALS and METHODS

106

Software Used

107

GastroPlus v 8.5 (Simulations Plus, Lancaster, CA, USA) was used for absorption modeling in

108

all the case studies.

109 110 111

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

112

Case Study 1: Linear pharmacokinetics with high fraction absorbed

113

Merck compound A physicochemical properties: Compound A is a BCS class II free base

114

molecule. The following key compound properties were used in building the model – molecular

115

weight 442, log P 4.21, pKa 3.53, density 1.2 g/mL, calculated human effective permeability

116

(based on Caco-2 data) was 3.2 x 10-4 cm/sec and diffusion coefficient was calculated based on

117

molecular weight. The amorphous solubility values from dissolution study were input as pH

118

solubility profile, as described below. The solubility of the crystalline form in simulated gastric

119

fluid (SGF) and fasted state simulated intestinal fluid (FaSSIF) was 0.02 mg/mL and 0.004

120

mg/mL, respectively. Diffusion coefficient was adjusted in GastroPlus to account for bile salt

121

concentration changes across the gastrointestinal (GI) tract based on diffusion coefficient and in-

122

vivo dissolution rate. The precipitation time was fixed at the default value of 900 seconds. The

123

blood to plasma ratio was 0.83. Fraction unbound in plasma was 0.9%.

124

Dosage form and dissolution data input: The compound was formulated as an amorphous solid

125

dispersion using HPMC-AS as the polymer matrix. Immediate release tablet was chosen as the

126

formulation option in GastroPlus. The dissolution data for this formulation was generated in 2-

127

stage dissolution (SGF followed by FaSSIF). The samples from the dissolution studies were

128

ultracentrifuged at 80,000 rpm before analysis to remove any undissolved API. Furthermore the

129

dissolution data did not show any indication of crystallization during the time-frame of the study.

130

Hence this data represented amorphous solubility of the compound. This dissolution data was

131

input in the model as a pH- amorphous solubility profile – 0.43 mg/mL (pH 1.3), 0.2 mg/mL (pH

132

6.0), 0.013 mg/mL (pH 6.2), and 0.015 mg/mL (pH 6.5). The solubility profile was input to

133

maintain supersaturation in the small in the small intestine, as there were no signs of

134

crystallization in the dissolution study. Based on particle size measurement of the dissolving

ACS Paragon Plus Environment

Page 6 of 38

Page 7 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

135

solid dispersion in the dissolution study, the mean size of the dissolving particle was set to be 1

136

µm (diameter) in the model. Similar particle size was assumed for the crystalline form of the

137

API, so that there is direct comparison of the effect of solubility difference of the API forms on

138

PK.

139

Physiology: The default human fasted physiological model in GastroPlus (Opt logD SA/v6.1)

140

was used in these simulations.

141

Pharmacokinetic (PK) parameters: Human PK parameters were estimated by fitting the 2 mg oral

142

data from first-in-human (FIH) study (Merck data on file) to a two-compartment model in PK-

143

plus. The mean PK parameters used in these simulations were CL/F = 0.235 L/hr/kg and V/F =

144

0.398 L/kg, k12 = 0.653 hr-1, and k21 = 0.25 hr-1. Based on the low dose and linear

145

pharmacokinetics it was assumed that F=1 (i.e. CL/F and V/F were used directly to simulate

146

systemic disposition)

147

Simulation: Single simulations were conducted to predict the mean PK profiles and parameters

148

for several doses from the FIH study using the amorphous solubility data from the dissolution

149

study. Additional simulations were conducted using the crystalline solubility of the compound to

150

investigate whether these solubility data were able to predict the observed FIH data. Parameter

151

sensitivity analysis (PSA) was conducted to assess the impact of precipitation time (90 – 9000

152

seconds) on fraction absorbed as function of dose (2 – 400 mg).

153 154

Case Study 2: Saturation of absorption

155

Merck compound B physicochemical properties: Compound B is a BCS class II molecule. The

156

following key compound properties were used in building the model – molecular weight 624.8,

157

log D (pH 7) 2.63, density 1.2 g/mL, calculated human effective permeability (based on Caco-2

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

158

data) was 2.38 x 10-4 cm/sec and diffusion coefficient was calculated based on molecular weight.

159

The solubility in buffer was 0.021 mg/mL and there was no pH dependent solubility profile. The

160

amorphous solubility in SGF and in FaSSIF was 0.021 mg/mL and 0.035 mg/mL, respectively.

161

The amorphous solubility values were obtained from the dissolution data as described in case

162

study 1. The solubility and diffusion coefficient was adjusted in GastroPlus to account for

163

changes across the gastrointestinal (GI) tract based on bile salt concentration changes, by fitting

164

the SGF and FaSSIF solubility data. The size of the dissolving particle was assumed to be 1 µm

165

(diameter), based on particle size measurement of the dissolving solid dispersion in the

166

dissolution study. The precipitation time was fixed at the default value of 900 seconds because

167

there is no pH dependent solubility of this compound and dissolution studies have shown no

168

indication of precipitation, therefore precipitation rate is expected to have minimal impact on

169

fraction absorbed. The blood to plasma ratio was set as 1. Fraction unbound in plasma was set as

170

5%.

171

Dosage form input: The compound was formulated as an amorphous solid dispersion using

172

HPMC-AS as the polymer matrix. Immediate release tablet was chosen as the formulation option

173

in GastroPlus.

174

Physiology: The default human fasted physiological model in GastroPlus (Opt logD SA/v6.1)

175

was modified by reducing absorption from caecum and ascending colon by manually reducing

176

the ASF to 1.0 in both caecum and ascending colon. This was done due to low buffer solubility

177

of the compound and no evidence of prolonged absorption in preclinical studies. Based on

178

experience with similar compounds, it was assumed that this compound would have minimal

179

absorption in the lower GI tract in human.

ACS Paragon Plus Environment

Page 8 of 38

Page 9 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

180

Pharmacokinetic (PK) parameters: Human PK parameters were estimated by fitting the 25 mg,

181

which was the starting dose, oral data from first-in-human (FIH) study (Merck data on file) to a

182

two-compartment model in WinNonin v5.3. The mean PK parameters used in these simulations

183

were CL = 0.66 L/hr/kg and V = 1.84 L/kg, k12 = 0.139 hr-1, and k21 = 0.190 hr-1. Based on the

184

low starting dose, linear pharmacokinetics up to 500 mg dose and lack of food effect at 125 mg

185

dose, it was assumed that fraction absorbed (Fa) = 1 at 25 mg. The first pass extraction (FPE)

186

was back-calculated as 51% using hepatic blood flow and blood/plasma ratio.

187

Simulation: Single simulations were conducted to predict the mean PK profiles and parameters

188

for several doses from the FIH study using the amorphous solubility data from the dissolution

189

study.

190 191

Case Study 3: Incomplete and low absorption

192

Merck compound C physicochemical properties: Compound C is a BCS IV compound with low

193

solubility (~0.1 µg/mL) across the physiological pH range and moderate permeability (LLCPK1

194

Papp = 7.9 x 10-6 cm/sec). Given the very low pKa ( 90%) across all the dose levels. Some fluctuations in the PK profile in the

258

elimination phase at the higher doses were likely an artifact of the model and have no bearing on

259

the overall outcome.

260 261

To investigate whether the use of amorphous solubility was critical to be able to accurately

262

predict the observed clinical data at all the doses, additional simulations were carried out using

263

the crystalline solubility of compound A. As shown in figure 2A, the low dose (2 mg) was

264

simulated accurately. However at 50 mg and above, underprediction of AUC and Cmax was

265

observed along with a right shift in Tmax. Figure 2B shows the simulations at 200 mg. These

266

simulations clearly demonstrated that solubility of the amorphous form of the API was needed to

267

predict the observed PK in the full FIH dose range. It is possible at the intermediate and higher

268

doses some mixture of amorphous and crystalline forms co-exists. However the current version

269

of the software doesn’t have the ability to handle two solubility values simultaneously hence

270

such a scenario cannot be simulated without significant assumptions. Finally, parameter

ACS Paragon Plus Environment

Page 13 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

271

sensitivity analysis demonstrated that there was minimal impact on fraction absorbed (90-100%)

272

over a wide range of precipitation time in the FIH dose range.

273 274

Case Study 2: Saturation of absorption

275

The simulated plasma concentration vs. time profiles and the PK parameters are shown in Figure

276

3 and Table 2, respectively, for doses evaluated in FIH study. The observed data are shown for

277

comparison. Overall, the model was able to predict observed human PK data with reasonable

278

accuracy across the doses, except that delayed Tmax was predicted at 125 mg dose and above,

279

which was not consistent with the observed Tmax across doses.

280 281

Figure 4 depicts the observed dose normalized AUC and simulated fraction absorbed (Fa) for

282

compound B at individual doses. The trend of saturation of absorption was observed in human

283

PK data, as dose normalized AUC0-t decreases from 0.108 to 0.037 µg/mL*hr/mg with increase

284

in dose. Similarly, a 3~4 fold decrease in Fa (%) was predicted in the absorption model across

285

the doses from 25 mg to 750 mg. This demonstrated that the model was able to capture the

286

observed saturation of absorption with reasonable accuracy.

287 288

Case Study 3: Incomplete and low absorption

289

The results of the dog pharmacokinetic study at 2 mg/kg dose and the modeling of the data are

290

shown in Figure 5. The overall bioavailability was low (approximately 10-15%). As shown in

291

Figure 5 an initial simulation based on the amorphous API solubility resulted in significant

292

underprediction of the data. An increase in solubility by 4-fold (implemented in the software by

293

increasing the bile salt solubilization ratio (SR); very comparable results are obtained by

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 38

294

adjusting the baseline solubility and maintaining the initial SR value) resulted in a better

295

description of the data; the overall extent of absorption was reasonably well captured although

296

the absorption phase was not fully captured. Since the intent of this dog simulation was not to

297

fully describe the dog data but to get an approximation of in vivo solubility with minimal model

298

changes, this solubility estimate was considered acceptable in an early development setting for

299

human PK projections. It is likely that the permeability in the dog was somewhat underpredicted

300

from the current model. However since there is no direct translation of dog to human

301

permeability that aspect of the model was not modified. This 4-fold higher in-vivo solubility

302

was subsequently used in the human simulations, with the assumption that the higher in-vivo

303

solubility observed in dogs is representative of human in-vivo solubility. It should be noted that

304

compound C plasma concentration at 24 hours post dose was unexpectedly high, but such

305

observations have been reported previously. (27) It is acknowledged that an increase in solubility

306

due to better bile salt solubilization in vivo may not represent the only solution to obtaining an

307

improved data fit. Thus additional simulations were conducted exploring alternative permeability

308

values or different particle size. An effective permeability of 6.7 × 10-4 cm/sec provided a similar

309

fit as that to the increase of in-vivo solubility (data not shown). This is a very high permeability

310

value (only a couple of compounds such as ketoprofen and piroxicam have been reported to have

311

such high permeability), that would appear unlikely given the cell line permeability and the

312

molecular weight (~600 Da) of compound C. Simulations conducted using small in vivo particle

313

size could not account for the observed exposures. Even when 0.2 µm (diameter) particle size

314

was used for the simulation, plasma concentrations were significantly underpredicted

315

(approximately 3-fold), similar to the simulations with the original solubility.

316

ACS Paragon Plus Environment

Page 15 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

317

Figure 6 depicts the simulation of clinical data at doses of 5, 15, 50 and 150 mg. Higher doses

318

(400 mg and 800 mg) are not shown as will be explained below. While simulations based on the

319

amorphous API solubility accurately predicted the plasma concentration vs time profile for the

320

first two doses (5 and 15 mg – Figure 6 panels A and B), they significantly underpredicted the

321

pharmacokinetics at the higher doses. Given the significant underprediction at 150 mg (Figure 6

322

panel D), simulations of higher doses are not shown. Incorporation of the in-vivo solubility

323

estimate from the dog model (i.e. 4 fold higher in-vivo solubility) significantly improved the

324

prediction at the 150 mg dose, which was a similar dose to that used in the dog study (2 mg/kg).

325

Although similar to the simulations of the dog data some underprediction of the absorption phase

326

was observed.

327 328

DISCUSSION

329

ASDs enable higher bioavailability as compared to formulation made from crystalline drug

330

substance due to higher solubility and dissolution rate of the amorphous product. However, it has

331

been observed that rendering a material amorphous does not guarantee success. (28) Hence

332

mechanistic understanding of the factors providing enhanced bioperformance (or lack of) is

333

imperative. PBPK modeling to prospectively and/or retrospectively understand the absorption

334

and bioperformance of ASDs in clinic based on in-vitro dissolution data and/or preclinical PK

335

data could be very important to enable successful development of these formulations. The three

336

case studies shown here were chosen to cover a wide range of ASD bioperformance in human

337

and modeling to retrospectively understand their in-vivo behavior. Case study 1 is an example of

338

fairly linear PK observed in the FIH study and the use of amorphous solubility to predict

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

339

bioperformance, case study 2 exemplifies saturation of absorption observed in FIH and case

340

study 3 demonstrates a formulation showing in-complete and low absorption.

Page 16 of 38

341 342

Compound A (case study 1) was formulated as a spray dried dispersion using HPMC-AS as the

343

polymer matrix. In single ascending dose FIH study, reasonable dose proportionality in AUC

344

was observed throughout the dose range of 2-400 mg but Cmax showed less than dose

345

proportionality at the highest dose (400 mg). Dissolution of this formulation in SGF followed by

346

FaSSIF, showed that in FaSSIF the amorphous formulation was able to provide approximately 4-

347

fold higher solubility as compared to the crystalline form of the API and also this supersaturation

348

was maintained throughout the duration (120 minutes) of the dissolution experiment. An

349

absorption model built using these amorphous solubility data from the dissolution experiment

350

was able to predict the PK profile (Figure 1) and parameters (Table 1) with reasonable accuracy.

351

Thus demonstrating that in-vivo the formulation was able to achieve similar amorphous

352

solubility as shown in the in-vitro dissolution experiment and also maintain supersaturation

353

throughout the dose levels. This was further confirmed by the results of simulations conducted

354

using solubility of the crystalline API, which showed that while at low dose (2 mg) the model

355

predicted the observed data well (Figure 2A), there was significant underprediction at 200 mg

356

(Figure 2B). Thus demonstrating that the formulation was able to maintain amorphous solubility

357

in-vivo at all doses in-order to achieve the observed dose proportional exposure. Since

358

compound A is a weakly basic amorphous compound, precipitation in the GI tract might impact

359

its bioavailability. However, sensitivity analysis (Figure 2C) showed that precipitation time had

360

minimal impact on fraction absorbed. This is likely because the amorphous solubility of the

361

compound is high enough across the GI tract that it maintains supersaturation throughout the

ACS Paragon Plus Environment

Page 17 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

362

dose range studied in the clinic. These simulations clearly demonstrate that a thorough

363

understanding of behavior of these amorphous formulations in appropriately designed dissolution

364

experiments and subsequently incorporating that data in the PBPK model is needed to be able to

365

predict their bioperformance.

366 367

Compound B (case study 2) was formulated as a spray dried amorphous solid dispersion using

368

HPMC-AS as the polymer matrix. In single ascending dose FIH study, both AUC and Cmax

369

showed less than dose proportionality PK as the dose increases from 25 mg to 750 mg, indicating

370

saturation of absorption at higher doses. Dissolution of this formulation showed that the

371

dissolved drug of solid dispersion formulation was able to provide approximately 2-fold higher

372

solubility as compared to a formulation with crystalline form of the API, and the supersaturation

373

was maintained throughout the duration (90 minutes) of the dissolution experiment in FaSSIF.

374

An absorption model was built using the amorphous solubility data from the dissolution

375

experiment to predict the clinical PK profiles (Figure 3) and PK parameters (Table 2) with

376

reasonable accuracy, although a somewhat delayed Tmax was predicted especially at higher doses.

377

The reason for lack of prediction of the absorption phase at higher doses could be due to factors

378

such as inadequate estimation of in-vivo solubility even with use of bile salt based solubilization

379

or inadequate effective permeability estimation or a combination of these factors. To further

380

illustrate the saturation of absorption in FIH study, the observed dose normalized AUC and

381

simulated %Fa for compound B at individual doses were plotted in Figure 4, which showed that

382

the degree of saturation of absorption in observed human PK data are in agreement with the

383

decrease of %Fa predicted by the absorption model across the doses from 25 mg to 750 mg. The

384

less than dose proportional PK could be an indication that the amorphous solubility limit has

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 18 of 38

385

been reached at the higher doses resulting in a solubility-limited absorption. In addition, it is

386

possible that the supersaturation achieved in-vivo in the GI fluid may not be maintained as well

387

as that suggested by the in-vitro dissolution system, which could also contribute to a less-than-

388

dose proportional increase in PK. While it is acknowledged that in this particular case the Tmax

389

was not precisely captured, the model was able to accurately predict the decrease in fraction

390

absorbed with dose escalation. Thus demonstrating that such physiologically based oral

391

absorption model can be used to model the clinical bioperformance in the scenario where

392

saturation of absorption is observed.

393 394

Compound C (case study 3) represents an interesting and challenging case study for oral

395

absorption modeling. Based on our experience, the behavior of compound C would have been

396

difficult to model with the currently available absorption models even if this formulation was a

397

simple crystalline API suspension. Specifically, compound C appears to exhibit relatively low

398

bioavailability (based on preclinical estimates and experience with other compounds in the

399

series) across all doses, however saturation of absorption is not seen until relatively high dose

400

(400 mg and higher). This relatively constant bioavailability across a wide dose range 5-400 mg

401

is very difficult to model based on the known compound and formulation properties. It is likely

402

that the low permeability of the compound is playing a role in the behavior and may be coupled

403

with regiodependent absorption (no data were available during development of Compound C to

404

assess this possibility). It is generally acknowledged that BCS IV compounds are the most

405

difficult to handle from absorption modeling standpoint. (1, 29)

406

ACS Paragon Plus Environment

Page 19 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

407

The better fit at 150 mg (Figure 6, panel D) based on extrapolated in-vivo solubility from the dog

408

data suggests that this preclinical to clinical translation may be one way to inform simulations for

409

these challenging compounds. Others have also suggested this preclinical to clinical translation

410

step as an initial step in absorption modeling. Recently Zhang et al. used estimates of in-vivo

411

solubilities from rats and dogs to successfully predict FIH pharmacokinetics for eighteen

412

compounds. (30) However one limitation with this approach is that extrapolation to doses outside

413

of those that have been tested in preclinical species may not be straightforward. It is also possible

414

that for solid dispersions (and other solubilizing formulations) the dose to excipient ratio also

415

dictates in-vivo solubilization capacity (or crystallization potential), which can make preclinical

416

to human translation complicated. In the case of compound C, the extrapolation at a comparable

417

mg/kg dose (where dose/excipient/dosing volume ratios were quite similar) appeared to work,

418

although based on the simulations shown in Figure 6 it is evident that the same higher solubility

419

would overpredict exposures at lower doses (data not shown). In a typical drug/formulation

420

development setting, prior to FIH studies would be conducted at a dose representing the

421

projected clinical dose and/or the upper range of the FIH study. This was the case for compound

422

C. While this approach works to project the human PK at the intended dose, it is acknowledged

423

that more studies may be required to project the behavior across the entire FIH dose range (if that

424

is of interest). The final projection at the clinical dose is within 2-fold of the observed data,

425

which would be considered a generally acceptable prediction error for these types of models at

426

early development stage.

427 428 429

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 20 of 38

430

CONCLUSION

431

For amorphous solid dispersion formulations understanding the factors that impact their

432

bioperformance is critical because by design these formulations are thermodynamically unstable

433

with propensity to crystalize and they have complex dissolution characteristics. Physiologically

434

based oral absorption modeling can play an integral part in advancing biopharmaceutics

435

knowledge of these formulations and help in robust design of formulation per the quality by

436

design (QbD) paradigm. The case studies discussed in this manuscript demonstrate the potential

437

application of absorption modeling in such context, with three examples showing increasing

438

levels of biopharmaceutic complexity. Case study 1 is an example of successful application of

439

amorphous solubility obtained from dissolution experiment to predict the linear PK observed

440

with dose escalation in the FIH study. Case study 2 demonstrates that these models can be used

441

to simulate the clinical bioperformance in the scenario where saturation of absorption is observed

442

with dose escalation. Finally, case study 3 exemplifies the application of such model to describe

443

the observed in-complete and low absorption with dose escalation. The biopharmaceutics

444

knowledge gained from these absorption models would maximizes the mechanistic

445

understanding of critical factors affecting the bioperformance of solid dispersion formulations

446

and thereby help in development of a final product that would deliver the optimal benefit to the

447

patients.

448 449

ACKNOWLEDGEMENTS

450

The authors would like to thank the project teams for providing the critical data needed to build

451

the models.

452

ACS Paragon Plus Environment

Page 21 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

453 454

Molecular Pharmaceutics

REFERENCES 1. Kostewicz, E.S.; Aarons, L.; Bergstrand, M.; Bolger, M.B.; Galetin, A.; Hatley, O.;

455

Jamei, M.; Lloyd, R.; Pepin, X.; Rostami-Hodjegan, A.; Sjögren, E.; Tannergren, C.;

456

Turner, D.B.; Wagner, C.; Weitschies, W.; Dressman, J. PBPK Models for the Prediction

457

of in vivo Performance of Oral Dosage Forms. Eur J Pharm Sci. 2014, 57, 300-321.

458

2. Agoram, B.; Woltosz, W.S.; Bolger, M.B. Predicting the Impact of Physiological and

459

Biochemical Processes on Oral Drug Bioavailability. Adv Drug Deliv Rev. 2001, 50

460

Suppl 1, S41-67.

461

3. Jamei, M.; Turner, D.; Yang, J.; Neuhoff, S.; Polak, S.; Rostami-Hodjegan, A.; Tucker,

462

G. Population-based Mechanistic Prediction of Oral Drug Absorption. AAPS J. 2009, 11,

463

225-237.

464

4. Kostewicz, E.S.; Abrahamsson, B.; Brewster, M.; Brouwers, J.; Butler, J.; Carlert, S.;

465

Dickinson, P.A.; Dressman, J.; Holm, R.; Klein, S.; Mann, J.; McAllister, M.; Minekus,

466

M.; Muenster, U.; Müllertz, A.; Verwei, M.; Vertzoni, M.; Weitschies, W.; Augustijns, P.

467

In vitro Models for the Prediction of in vivo Performance of Oral Dosage Forms. Eur J

468

Pharm Sci. 2014, 57, 342-366.

469

5. Thelen, K.; Coboeken, K.; Willmann, S.; Dressman, J.B.; Lippert, J. Evolution of a

470

Detailed Physiological Model to Simulate the Gastrointestinal Transit and Absorption

471

Process in Humans, Part II: Extension to Describe Performance of Solid Dosage Forms. J

472

Pharm Sci. 2012, 101, 1267-1280.

473

6. Johnson, K.C. Dissolution and Absorption Modeling: Model Expansion to Simulate the

474

Effects of Precipitation, Water Absorption, Longitudinally Changing Intestinal

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

475

Permeability, and Controlled Release on Drug Absorption. Drug Dev Ind Pharm. 2003,

476

29, 833-842.

477 478 479 480

Page 22 of 38

7. Kesisoglou, F.; Mitra, A. Application of Absorption Modeling in Rational Design of Drug Product Under Quality-by-Design Paradigm. AAPS J. 2015, 17, 1224-1236. 8. Kesisoglou, F.; Wu, Y. Understanding the Effect of API Properties on Bioavailability Through Absorption Modeling. AAPS J. 2008, 10, 516-525.

481

9. Parrott, N.; Hainzl, D.; Scheubel, E.; Krimmer, S.; Boetsch, C.; Guerini, E.; Martin-

482

Facklam, M. Physiologically based Absorption Modelling to Predict the Impact of Drug

483

Properties on Pharmacokinetics of Bitopertin. AAPS J. 2014, 16, 1077-1084.

484

10. Kesisoglou, F.; Chung, J.; Van Asperen,

J.; Heimbach, T. Physiologically Based

485

Absorption Modeling to Impact Biopharmaceutics and Formulation Strategies in Drug

486

Development—Industry Case Studies. J Pharm Sci. 2016, (article in press)

487

11. Mitra, A.; Kesisoglou, F.; Dogterom, P. Application of Absorption Modeling to Predict

488

Bioequivalence Outcome of Two Batches of Etoricoxib Tablets. AAPS PharmSciTech.

489

2015, 16, 76-84.

490

12. Crison, J.R.; Timmins, P.; Keung, A.; Upreti, V.V.; Boulton, D.W.; Scheer, B.J.

491

Biowaiver Approach for Biopharmaceutics Classification System Class 3 Compound

492

Metformin Hydrochloride Using In Silico Modeling. J Pharm Sci. 2012, 101, 1773-1782.

493

13. Sperry, D.C.; Thomas, S.J.; Lobo, E. Dissolution Modeling of Bead Formulations and

494

Predictions of Bioequivalence for a Highly Soluble, Highly Permeable Drug. Mol Pharm.

495

2010, 7, 1450-1457.

496 497

14. Mathias, N.R.; Crison, J. The Use of Modeling Tools to Drive Efficient Oral Product Design. AAPS J. 2012, 14, 591-600.

ACS Paragon Plus Environment

Page 23 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

498

15. Chen, W.; Desai, D.; Good, D.; Crison, J.; Timmins, P.; Paruchuri, S.; Wang, J.; Ha, K.

499

Mathematical Model-Based Accelerated Development of Extended-release Metformin

500

Hydrochloride Tablet Formulation. AAPS PharmSciTech. 2015, Oct 19. [Epub ahead of

501

print]

502

16. Wang, Y.; Abrahamsson, B.; Lindfors, L.; Brasseur, J.G. Analysis of Diffusion-

503

Controlled Dissolution from Polydisperse Collections of Drug Particles with an Assessed

504

Mathematical Model. J Pharm Sci. 2015, 104, 2998-3017.

505 506

17. Venkatesh, S.; Lipper R. A. Role of the Development Scientist in Compound Lead Selection and Optimization. J Pharm Sci. 2000, 89, 145-154.

507

18. Williams, H.D.; Trevaskis, N.L.; Charman, S.A.; Shanker, R.M.; Charman, W.N.;

508

Pouton, C.W.; Porter, C.J. Strategies to Address Low Drug Solubility in Discovery and

509

Development. Pharmacol Rev. 2013, 65, 315-499.

510

19. Baghel, S.; Cathcart, H.; O'Reilly, N.J.; Polymeric Amorphous Solid Dispersions: A

511

Review of Amorphization, Crystallization, Stabilization, Solid-State Characterization,

512

and Aqueous Solubilization of Biopharmaceutical Classification System Class II Drugs. J

513

Pharm Sci. 2016 [Epub ahead of print].

514

20. Rumondor, A.C..; Dhareshwar, S.S.; Kesisoglou, F. Amorphous Solid Dispersions or

515

Prodrugs: Complementary Strategies to Increase Drug Absorption, J Pharm Sci. 2016

516

[Epub ahead of print]

517

21. Serajuddin, A.T.M. Solid dispersion of Poorly Water-Soluble Drugs: Early Promises,

518

Subsequent Problems, and Recent Breakthroughs. J Pharm Sci. 1999, 88, 1058-1066.

519

22. Miller, J.M.; Beig, A.; Carr, R.A.; Spence, J.K.; Dahan, A. A Win-Win Solution in Oral

520

Delivery of Lipophilic Drugs: Supersaturation via Amorphous Solid Dispersions

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

521

Increases Apparent Solubility without Sacrifice of Intestinal Membrane Permeability.

522

Mol Pharm. 2012, 9, 2009-2016.

Page 24 of 38

523

23. Gao, Y.; Carr, R.A.; Spence, J.K.; Wang, W.W.; Turner, T.M.; Lipari, J.M.; Miller, J.M.

524

A pH-dilution Method for Estimation of Biorelevant Drug Solubility along the

525

Gastrointestinal Tract: Application to Physiologically based Pharmacokinetic Modeling.

526

Mol Pharm. 2010, 7, 1516-1526.

527

24. Zheng, W.; Jain, A.; Papoutsakis, D.; Dannenfelser, R.M.; Panicucci, R.; Garad, S.

528

Selection of Oral Bioavailability Enhancing Formulations during Drug Discovery. Drug

529

Dev Ind Pharm. 2012, 38, 235-247.

530

25. Friesen, D.T.; Shanker, R.; Crew, M.; Smithey, D.T.; Curatolo, W.J.; Nightingale, J.A.

531

Hydroxypropyl Methylcellulose Acetate Auccinate-based Spray-Dried Dispersions: An

532

Overview. Mol Pharm. 2008, 5, 1003-1019.

533

26. Kesisoglou, F. Use of Preclinical Dog Studies and Absorption Modeling to Facilitate Late

534

Stage Formulation Bridging for a BCS II Drug Candidate. AAPS PharmSciTech. 2014,

535

15, 20-28.

536

27. Benincosa, L.J.; Audet, P.R.; Lundberg, D.; Zariffa, N.; Jorkasky, D.K. Pharmacokinetics

537

and Absolute Bioavailability of Epristeride in Healthy Male Subjects. Biopharm Drug

538

Dispos. 1996, 17, 249-258.

539 540

28. Newman, A.; Knipp, G.; Zografi, G. Assessing the Performance of Amorphous Solid Dispersions. J Pharm Sci. 2012, 101, 1355-1377.

541

29. Lennernas, H.; Aarons, L.; Augustijns, P.; Beato, S.; Bolger, M.; Box, K.; Brewster, M.;

542

Butler, J.; Dressman, J.; Holm, R.; Julia Frank, K.; Kendall, R.; Langguth, P.; Sydor, J.;

543

Lindahl, A.; McAllister, M.; Muenster, U.; Mullertz, A.; Ojala, K.; Pepin, X.; Reppas, C.;

ACS Paragon Plus Environment

Page 25 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

544

Rostami-Hodjegan, A.; Verwei, M.; Weitschies, W.; Wilson, C.; Karlsson, C.;

545

Abrahamsson, B. Oral Biopahrmaceuotcs Tools – Time for a New Initiative – An

546

Introduction to the IMI Project OrBito. Eu J Pharm Sci. 2014, 57, 292-299.

547 548

30. Zhang, T.; Heimbach, T.; Lin, W.; Zhang, J.; He, H. Prospective Predictions of Human Pharmacokinetics for Eighteen Compounds. J Pharm Sci. 2015, 104, 2795-2806.

549

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

550

TABLE LEGENDS

551

Table 1: Simulated and observed pharmacokinetic parameters for compound A at dose range of

552

2 - 400 mg.

553

Table 2: Simulated and observed pharmacokinetic parameters for compound B at dose range of

554

25 – 750 mg.

555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572

ACS Paragon Plus Environment

Page 26 of 38

Page 27 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

573

FIGURE LEGENDS

574

Figure 1: (A) Simulated and observed plasma concentration vs. time profiles for compound A at

575

2 mg, 4 mg, 8 mg and 15 mg doses. (B) Simulated and observed plasma concentration vs. time

576

profiles for compound A at 25 mg, 50 mg, 100 mg, 200 mg and 400 mg doses.

577 578

Figure 2: Simulated plasma concentration vs. time profiles for compound A at 2 mg dose (A),

579

200 mg dose (B) using solubility of the crystalline form of the compound. The observed plasma

580

concentration vs. time profiles at both doses is shown for comparison. (C) Parameter sensitivity

581

analysis showing the impact of precipitation time and dose on fraction absorbed.

582 583

Figure 3: Simulated and observed plasma concentration vs. time profiles for compound B at 25

584

mg, 50 mg, 125 mg, 250 mg, 500 mg and 750 mg doses.

585 586

Figure 4: Observed dose normalized AUC0-t (µg/mL*hr/mg) and simulated fraction absorbed

587

(%Fa) for compound B at 25 mg, 50 mg, 125 mg, 250 mg, 500 mg and 750 mg doses.

588 589

Figure 5 Simulated (lines) and observed (squares) plasma concentration vs. time profiles for

590

compound C in dogs at a 2 mg/kg dose. Initial simulation based on amorphous API solubility

591

(dashed line) and final model fit to 4-fold higher in-vivo solubility (solid line) are shown.

592 593

Figure 6 Simulated (dashed lines) and observed (squares) plasma concentration vs. time profiles

594

for compound C at 5 mg, 15 mg, 50 mg and 150 mg doses (panels A-D, respectively). For the

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

595

150 mg dose an additional simulation based on the in-vivo solubility obtained from dog data is

596

depicted (solid line).

597

ACS Paragon Plus Environment

Page 28 of 38

Page 29 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

598

Molecular Pharmaceutics

Table 1 Observed Dose (mg)

Predicted

2

AUC0-24hr (µg/mL*hr) 0.12

Cmax (µg/mL) 0.018

Tmax (hr) 1.0

AUC0-24hr (µg/mL*hr) 0.13

Cmax (µg/mL) 0.022

Tmax (hr) 1.4

4

0.25

0.037

2.0

0.23

0.047

1.4

8

0.49

0.069

2.0

0.46

0.094

1.4

15

1.03

0.163

2.0

0.86

0.177

1.4

25

1.61

0.216

2.0

1.43

0.295

1.7

50

2.89

0.551

2.0

2.86

0.559

1.7

100

6.40

1.106

2.0

5.72

0.978

2.0

200

11.5

1.92

1.0

11.4

1.79

1.0

400

19.1

1.94

2.0

20.3

2.30

1.2

599 600 601 602 603

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

604

Page 30 of 38

Table 2 Observed Dose

Predicted

AUC0-t (µg/mL*hr)

Cmax (µg/mL)

Tmax (hr)

AUC0-t (µg/mL*hr)

Cmax (µg/mL)

Tmax (hr)

Fa (%)

25

0.270

0.043

2.0

0.256

0.051

1.8

100

50

0.398

0.058

1.5

0.528

0.095

2.0

100

125

0.861

0.088

1.5

1.155

0.144

3.5

87

250

1.123

0.113

1.0

1.563

0.161

3.5

59

500

2.131

0.180

2.0

1.908

0.175

4.5

36

750

2.810

0.249

2.0

2.094

0.181

4.8

26

(mg)

605 606 607

ACS Paragon Plus Environment

Page 31 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

608

Molecular Pharmaceutics

Figure 1 (A)

609

(B)

610

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

611

Figure 2

(A)

612

(B)

613 614

ACS Paragon Plus Environment

Page 32 of 38

Page 33 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

615 616

Molecular Pharmaceutics

Figure 2

617

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

618

Figure 3

619 620

ACS Paragon Plus Environment

Page 34 of 38

Page 35 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

621

Molecular Pharmaceutics

Figure 4

622 623

ACS Paragon Plus Environment

Molecular Pharmaceutics

624

Figure 5

625 Observed data

160

Simulation based on amorphous API solubility Simulation with optimized 4-fold higher in vivo solubility

140 Plasma Concentration (ng/mL)

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 36 of 38

120 100 80 60 40 20 0 0

4

8

12 Time (hr)

626 627

ACS Paragon Plus Environment

16

20

24

Page 37 of 38

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

628

Molecular Pharmaceutics

Figure 6 (A)

629

(B)

630 631 632

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

633

Figure 6 (C)

634

(D)

635 636 637 638 639

ACS Paragon Plus Environment

Page 38 of 38