In Vivo Dissolution and Systemic Absorption of Immediate Release

Sep 22, 2017 - *Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Room 3353, Building 520,1600 Huron Parkway, Ann Ar...
0 downloads 9 Views 2MB Size
Subscriber access provided by University of Sussex Library

Article

In Vivo Dissolution and Systemic Absorption of Immediate Release Ibuprofen in Human Gastrointestinal Tract Under Fed and Fasted Conditions Duxin Sun, Jason R. Baker, Bo Wen, Ann Frances, Huixia Zhang, Alex Yu, Ting Zhao, Yasuhiro Tsume, Manjunath Pai, Barry Bleske, Xinyuan Zhang, R. Lionberger, Allen Lee, Gordon Amidon, William L. Hasler, and Mark Koenigsknecht Mol. Pharmaceutics, Just Accepted Manuscript • DOI: 10.1021/acs.molpharmaceut.7b00425 • Publication Date (Web): 22 Sep 2017 Downloaded from http://pubs.acs.org on September 24, 2017

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 32

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 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

Molecular Pharmaceutics

In Vivo Dissolution and Systemic Absorption of Immediate Release Ibuprofen in Human Gastrointestinal Tract Under Fed and Fasted Conditions Mark J. Koenigsknecht1, Jason R. Baker2, Bo Wen1, Ann Frances1, Huixia Zhang1, Alex Yu1, Ting Zhao1, Yasuhiro Tsume1, Manjunath P. Pai3, Barry E Bleske4, Xinyuan Zhang5§, Robert Lionberger5§, Allen Lee2, Gordon L. Amidon1, William L. Hasler2*, Duxin Sun1* Affiliations 1Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, USA 2Department

of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan 48109, USA

3Department

of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, USA

4 Department

of Pharmacy Practice and Administrative Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico, 87120, USA

5Office

of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland 20993, USA

§Disclaimer:

The views expressed in this article are those of the authors and not necessarily those of the Food and Drug Administration (FDA).

*Corresponding

authors:

Duxin Sun, Ph.D., Professor, Department of Pharmaceutical Sciences, College of Pharmacy. University of Michigan, Room 3353, Building 520,1600 Huron Parkway, Ann Arbor, Michigan 48105. Phone: 734-615-8740, Email: [email protected] William L. Hasler, M.D., Professor, Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan 48109. Phone: 734936-4780, Email: [email protected]

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

41

Page 2 of 32

Graphical Abstract:

42 43

Abstract:

44

Purpose: In vivo drug dissolution in the gastrointestinal (GI) tract is largely

45

unmeasured. The purpose of this clinical study was to evaluate the in vivo drug

46

dissolution and systemic absorption of the BCS Class IIa drug ibuprofen under fed

47

and fasted conditions by direct sampling of stomach and small intestinal luminal

48

content. Expanding current knowledge of drug dissolution in vivo will help to

49

establish physiologically relevant in vitro models predictive of drug dissolution.

50 51

Methods: A multi-lumen GI catheter was orally inserted into the GI tract of healthy

52

human subjects. Subjects received a single oral dose of ibuprofen (800 mg tablet)

53

with 250 mL of water under fasting and fed conditions. The GI catheter facilitated

54

collection of GI fluid from the stomach, duodenum, and jejunum. Ibuprofen

55

concentration in GI fluid supernatant and plasma was determined by LC-MS/MS.

56 57

Results: A total of 23 subjects completed the study, with 11 subjects returning for

58

an additional study visit (a total of 34 completed study visits). The subjects were

ACS Paragon Plus Environment

1

Page 3 of 32

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

59

primarily white (61%), male (65%) with an average age of mean of 30 years. The

60

subjects had a median [minutes, max] weight of 79 [52, 123] kg and body mass

61

index of 25.7 [19.4, 37.7] kg/m2. Ibuprofen plasma levels were higher under fasted

62

conditions and remained detectable for 28 hours under both conditions. The AUC0-24

63

and Cmax were lower in fed subjects vs. fasted subjects and Tmax was delayed in fed

64

subjects vs. fasted subjects. Ibuprofen was detected immediately after ingestion in

65

the stomach under fasting and fed conditions until 7 hours after dosing. Higher

66

levels of ibuprofen were detected in the small intestine soon after dosing in fasted

67

subjects compared to fed. In contrast to plasma drug concentration, overall gastric

68

concentrations remained higher under fed conditions due to increased gastric pH vs.

69

fasting condition. The gastric pH increased to near neutrality after feeding before

70

decreasing to acidic levels after 7 hours.

71 72

Conclusions: Induction of the fed state reduced systemic levels but increased

73

gastric levels of ibuprofen, which suggest slow gastric emptying and transit

74

dominate effect for plasma drug concentration. High levels of ibuprofen in stomach

75

and small intestine 7 hours post dosing was an unexpected finding. Future work is

76

needed to better understand the role of various GI parameters, such as motility and

77

gastric emptying, on systemic ibuprofen levels in order to improve in vitro

78

predictive models.

79 80

Key Words:

81

Ibuprofen; in vivo dissolution; pharmacokinetics; immediate release; local

ACS Paragon Plus Environment

2

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

82

Page 4 of 32

gastrointestinal concentration; clinical study

83 84

List of abbreviations:

85

Gastrointestinal (GI), biopharmaceutical classification system (BCS), nonsteroidal

86

anti-inflammatory drug (NSAIDs), Food and Drug Administration (FDA).

87 88 89

Introduction: Designing oral drug formulations with predictable gastrointestinal (GI)

90

absorption profiles is a major objective during drug development. Understanding

91

the dissolution properties of oral formulations using in vitro systems can support

92

this endeavor. Current in vitro models are useful for in vivo prediction of absorption

93

of high solubility drugs. In contrast the absorption of low solubility drugs with high

94

GI permeability, referred to as biopharmaceutical classification system (BCS) II

95

drugs, are especially difficult to predict1, 2. This prediction problem arises from the

96

inability to optimally mimic the dynamic physiologic parameters such as pH,

97

buffering conditions, and shifting GI volumes that impart changes dissolution and

98

absorption of BCS II drugs in the GI tract. Designing a predictive model that is able

99

to address this complex drug-GI behavior may play a major role in drug

100

development. Importantly, such a system has the potential to help qualify waivers

101

of bioavailability and bioequivalence studies for BCS II drugs during generic drug

102

development.

103 104

Currently there are very limited data available analyzing in vivo BCS II drug dissolution and dosage form performance in the GI tract, however recent studies by

ACS Paragon Plus Environment

3

Page 5 of 32

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

105

Hens et al. and Van Den Abeele et al. have begun to shed light on drug dissolution

106

and absorption of weakly acidic BCS II drugs3-5. Drug absorption in the GI tract is a

107

complex process that remains to be fully elucidated. Constructing a comprehensive

108

translational model requires clinical measurement of GI variables such as pH, buffer

109

capacity, motility, etc. in order to develop better in vitro models. The goal of this

110

clinical study was to obtain in vivo drug dissolution of BCS Class IIa drug ibuprofen

111

in the GI tract in fed and fasted conditions and correlate with systemic drug

112

concentration. We chose to evaluate a model BCS IIa drug, ibuprofen, one of the

113

most commonly used nonsteroidal anti-inflammatory drug (NSAIDs) to treat acute

114

pain, fever, and inflammation6.

115

The potential benefits of ibuprofen are hampered by side-effects7.

116

Specifically, the chronic use of NSAIDs like ibuprofen is associated with GI adverse

117

events such as gastroduodenal ulceration8. This adverse event occurs due to topical

118

epithelial irritant effects, suppression of GI prostaglandin synthesis, alteration of

119

local GI blood flow, and interference of injury repair mechanisms. To improve GI

120

tolerability, ibuprofen is often taken with food9. Food can delay the rate of ibuprofen

121

absorption and lower its pharmacological effects10. While we understand the effect

122

food has on plasma levels we have yet to understand the effect of food on local GI

123

ibuprofen concentrations.

124

In this study a novel multi-lumen GI catheter was orally placed into the

125

stomach and small intestine of healthy human volunteers to obtain in vivo samples

126

of the GI tract after dosage of an ibuprofen tablet. This study was performed in

127

volunteers under both fed and fasted conditions in order to characterize in vivo drug

ACS Paragon Plus Environment

4

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 6 of 32

128

disintegration and dissolution under each condition. Corresponding plasma samples

129

and GI fluid samples were collected for seven hours following study drug

130

administration. The concentration of ibuprofen in plasma and GI fluid supernatant

131

was determined via LC-MS/MS and correlated with the human physiology data

132

(such as pH) to better understand the effect of food on drug absorption. Correlating

133

in vivo drug concentrations with GI physiology data under fasting and fed conditions

134

from healthy subjects will help support mechanistic absorption model development

135

in the future. This information may also be helpful to improve our understanding of

136

local ibuprofen concentrations that may influence gastroduodenal ulceration with

137

NSAIDs use.

138 139

Methods:

140

Ethics Statement

141

Samples collected in this study were part of clinical trial NCT02806869. The

142

institutional review boards at the University of Michigan (IRBMED, protocol number

143

HUM00085066) and the Department of Health and Human Services, Food and Drug

144

Administration (Research Involving Human Subjects Committee/RIHSC, protocol

145

number14-029D) both approved the study protocol. All subjects provided written

146

informed consent in order to participate. The study was carried out accordance with

147

the protocol, International Conference on Harmonization Good Clinical Practice

148

guidelines, and applicable local regulatory requirements.

149 150

Ibuprofen Formulation

ACS Paragon Plus Environment

5

Page 7 of 32

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

151

Ibuprofen tablets (800 mg) were purchased from Dr. Reddy’s Laboratories

152

(Shreveport, LA). The ibuprofen tablet was administered with approximately 250

153

mL of water, actual volumes of water consumed for each subject is listed in

154

Supplemental Table 1. The study drug was dispensed by the Investigational Drug

155

Service (IDS) at the University of Michigan.

156 157

Catheter Design

158

A customized multi-lumen catheter was manufactured by Mui Scientific.

159

(Mississauga, ON, Canada). The catheter was 292 cm long and consisted of four

160

independent aspiration ports located 30-20 cm apart and 16 manometry ports

161

located proximal to the aspiration ports. The outer diameter of the catheter was 3.3

162

mm while the single lumen tubes used for aspiration had an inner diameter of 1.2

163

mm. Additionally, the catheter had a channel to fit a guidewire (0.035 in x 450 cm,

164

Boston Scientific, Marlborough, MA) as well as a channel connected to a balloon that

165

could be filled with 7 mL of water to assist in catheter placement. Finally, the end of

166

the catheter was weighted with 7.75 grams of tungsten weights.

167 168 169

Inclusion and Exclusion Criteria for the Study Male and female healthy human volunteers between the ages of 18 and 55

170

were eligible for inclusion in the study. All subjects completed a physical exam and

171

had their previous medical history reviewed by the study physician to confirm the

172

subjects were healthy volunteers able to participate in the study. All subjects

173

enrolled in the study had normal values for the following laboratory tests: vital

ACS Paragon Plus Environment

6

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 8 of 32

174

signs, electrocardiogram (ECG), urine drug screen, serum pregnancy test (for

175

women of child-bearing potential), comprehensive metabolic panel, complete blood

176

count with platelet and differential, and lactate dehydrogenase. There were no

177

exclusionary criteria in regards to BMI, however all tests listed above had to be in

178

normal range and the subject had to overall be deemed healthy by the study

179

physician.

180

Exclusionary criteria for the study were as follows: Adults unable to consent

181

for themselves or mentally incapacitated; prisoners; significant clinical illness

182

within 3 weeks prior to screening; use of concomitant medications within 2 weeks

183

prior to receiving study drug, including but not limited to prescription drugs, herbal

184

and dietary supplements, over the counter medications, and vitamins (oral

185

contraceptive was permitted); received an investigational drug within 60 days prior

186

to receiving the study drug; history of gastrointestinal surgery; surgery within the

187

past 3 months; history of allergy to ibuprofen or other non-steroidal anti-

188

inflammatory drugs (NSAIDs); pregnant or lactating females; history of severe

189

allergic diseases including drug allergies, with the exception of seasonal allergies;

190

history of drug addiction or alcohol abuse within the past 12 months; any clinically

191

significant abnormal lab values during screening’ any other factor, condition, or

192

disease, including, but not limited to, cardiovascular, renal, hepatic, or

193

gastrointestinal disorders that may, in the opinion of the investigator, jeopardize the

194

safety of the patient or impact the validity of the study results.

195 196

Study Procedure

ACS Paragon Plus Environment

7

Page 9 of 32

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

197

Molecular Pharmaceutics

All clinical procedures were performed at the Michigan Clinical Research

198

Unit (MCRU) or the Medical Procedures Unit (MPU) in the University of Michigan

199

hospital. The subjects were instructed to fast beginning 13 hours prior to dosing and

200

to avoid consuming water for 10 hours prior to dosing. Upon arrival a physical exam

201

was performed by the study doctor to ensure the health of the subject prior to the GI

202

catheter insertion procedure. The multi-lumen GI catheter was orally inserted into

203

the GI tract of the subject under the supervision of the study doctor. Lubricating jelly

204

was applied to the GI catheter prior to insertion. To lessen the gag reflex, subjects

205

received a topical anesthetic (1 mL of 4% lidocaine) before the catheter insertion.

206

Once the catheter was swallowed, catheter placement was continued under

207

abdominal fluoroscopy to ensure proper positioning in the GI tract. When catheter

208

placement was completed the GI catheter was secured to the cheek of the subject

209

using medical tape. The subject remained in bed for one hour while the GI tube was

210

equilibrated to the manometry instrument, then for 3-5 hours while baseline GI

211

motility data was collected. Additionally, GI motility was measured continuously for

212

7 hours following study drug administration. The rate of water perfused into the

213

subject to measure motility was approximately 100 mL per hour for the 11-13 hours

214

that motility was recorded.

215

Prior to study drug administration, an intravenous (IV) catheter was placed

216

and was kept open with saline solution throughout the study visit. A blood sample

217

and GI sample was collected immediately prior to study drug administration and

218

food consumption. The study subjects were randomized to one of two arms for this

219

study: the study drug dosed in the fasting state or the study drug dosed immediately

ACS Paragon Plus Environment

8

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 10 of 32

220

after consuming up to 710 calories (473 mL) of a 55% fat liquid meal to induce the

221

fed state (Pulmocare (pH 6.6), Abbott Nutrition, Lake Forest, IL). Randomization

222

was performed using Research Randomizer software version 4.0. The maximum

223

time allowed to consume the liquid meal was 15 minutes and then the subjects were

224

immediately dosed with ibuprofen.

225

The subject was given an 800 mg tablet of ibuprofen to be swallowed with

226

250 mL of water. After administration of the study drug the subjects did not

227

consume any additional food or water. The study drug, water, and/or Pulmocare

228

were swallowed by the subject and were not administered via the GI catheter. Blood

229

samples were collected at 0 (immediately prior to food consumption and/or study

230

drug administration), 10, 20, 30, and 45 minutes, then at 1, 1.5, 2, 2.5, 3, 4, 5, 6, 7, 8,

231

11 or 12, and approximately 28 hours following study drug administration. Blood

232

samples were added to venous blood collections tubes (K2 EDTA (spray-dried),

233

7.2mg) and plasma was separated from blood samples by centrifugation and stored

234

at -80⁰C.

235

GI fluid samples were collected from available ports by aspiration from

236

individual tubes within the GI catheter. Before collection the contents in the GI

237

catheter remaining from the previous aspiration were collected and discarded. This

238

volume ranged from 3.2 mL for the most distal (jejunal) tube to 1.7 mL for the most

239

proximal (stomach) tube. If the tube was filled with air bubbles then at least 30 cc of

240

the air/fluid mixture (containing no more than 3.2 mL of liquid) was collected and

241

discarded. Collection times included 0 (immediately prior to study drug

242

administration), 15, 30, and 45 minutes, then at 1, 1.5, 2, 2.5, 3, 4, 5, 6, and 7 hours

ACS Paragon Plus Environment

9

Page 11 of 32

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

243

following study drug administration. Immediately after collection the samples were

244

pH using a calibrated micro pH electrode (Thermo Scientific Orion pH probe

245

9810BN). Then the GI fluid samples were centrifuged at 21,000 x g for 5 minutes

246

and the supernatant was collected and stored at -80⁰C.

247

At 7 hours following study drug administration, the GI catheter was removed

248

orally from the GI tract and the subject was served a meal. Following the 8 hour

249

blood draw the IV catheter was removed and the remaining blood draws were

250

performed with a butterfly needle and syringe. The subject then returned

251

approximately 28 hours post dosing for the final blood draw.

252

To gain an understanding of intrasubject variability, each subject was asked

253

to complete two GI catheter insertion procedures under the same conditions, either

254

fasting state or fed state. A minimum of 7 days separated each GI catheter insertion

255

procedure. The screening visit was repeated prior to the second study visit if

256

scheduled more than 4 weeks after the first study visit.

257 258 259

LC-MS/MS Analysis of Ibuprofen in GI Fluid and Plasma All samples were analyzed by the Pharmacokinetics Core at the University of

260

Michigan. LC-MS/MS analyses was performed using a Shimadzu HPLC system

261

interfaced to an AB SCIEX QTRAP 5500 mass spectrometer by a TurboV

262

electrospray ionization (ESI) source (Applied Biosystems/MDS Sciex, Toronto,

263

Canada). Ibuprofen-D3 was used as an internal standard (IS) to normalize variation

264

during sample preparation and LC-MS/MS analyses. Chromatographic separation

265

was achieved using a 2.1 x 50 mm, 3.5 µm Agilent ZORBAX Extend-C18 column. The

ACS Paragon Plus Environment

10

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 12 of 32

266

injection volume was 5 µL and the flow rate was kept constantly at 0.4 mL/minutes.

267

Mobile phase A and B were water and acetonitrile, respectively. Both water and

268

acetonitrile contained 0.1% acetic acid (v/v). The flow gradient was initially 98:2

269

v/v of A:B for 0.5 minutes, linearly ramped to 5:95 A:B over 1 minute. To completely

270

wash the column, the gradient was held at 5:95 A:B for 2 minutes and then returned

271

to 98:2 over 0.5 minutes. This condition was held for 3 minutes prior to the injection

272

of another sample. The mass spectrometer was operated at ESI negative ion mode

273

and multiple reaction monitoring (MRM) was used for monitoring the transitions of

274

m/z 205.1→159.1 and m/z 208.2→161.2 for ibuprofen and ibuprofen-D3 (IS),

275

respectively.

276

Protein precipitation with methanol was used to extract ibuprofen from the

277

human plasma. 60 µL of each plasma sample was mixed with 180 µL of methanol

278

containing 500 ng/mL ibuprofen-D3 in a 96-well plate. The mixture was vortexed

279

for 60 seconds at a high speed. The 96-well plate was centrifuged at 3000 x g for 20

280

minutes to precipitate proteins at 4 ⁰C. The clear supernatants were collected and 5

281

µL of the supernatant was injected for LC-MS/MS analysis.

282

Sample preparation of GI fluid was similar to the plasma samples. Due to the

283

inadequate amount of blank GI fluid for preparation of calibration standards and QC

284

samples, the plasma calibration curve was used to quantify ibuprofen in GI fluid. In

285

order to reduce matrix effect, all of the GI fluid samples were diluted 10 fold with

286

blank human plasma before protein precipitation in methanol containing 500

287

ng/mL ibuprofen-D3.

ACS Paragon Plus Environment

11

Page 13 of 32

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

288

Molecular Pharmaceutics

Stock solutions of ibuprofen (2 mg/mL) were prepared in methanol. An

289

ibuprofen-D3 (IS) stock solution (5 mg/mL) was prepared in dimethyl sulfoxide. To

290

prepare the calibration standards, the 2 mg/mL ibuprofen stock solution was

291

diluted to 500 µg/mL using methanol, which was spiked with blank human plasma

292

to provide a final ibuprofen plasma concentration of 10 µg/mL. Then serial dilution

293

with the blank human plasma was carried out to provide a series of calibration

294

standards from 2.5-10,000 ng/mL. Quality control (QC) plasma solutions at 5, 25,

295

250, 5000, and 10000 ng/mL were prepared similarly using separately weighed

296

methanol stock solutions of ibuprofen (2 mg/mL). Methanol was used to further

297

dilute the stock ibuprofen-D3 solution to 500 ng/mL for protein precipitation

298

during sample preparation, also to minimize the variation.

299

According to Food and Drug Administration (FDA) guidelines, validation

300

procedures were performed in human plasma and GI fluid diluted with human

301

plasma. These procedures included: (A) specificity and selectivity; (B) recovery and

302

matrix effects at low (0.015 µg/mL), medium (0.1 and 10 µg/mL), and high (35

303

µg/mL) concentrations; (C) calibration curve with a correlation coefficient (r) of

304

more than 0.98; (D) precision and accuracy, with the intra-day and inter-day assay

305

precision and accuracy estimated by analyzing six replicates at three QC levels; (E)

306

stability, with all stability studies conducted at three concentration levels in the

307

biomatrix at room temperature, 4 °C, −20 °C, and −80 °C; and (F) dilution integrity,

308

with experiments carried out with blank biomatrix.

309 310

Pharmacokinetic Analysis and Data Presentation

ACS Paragon Plus Environment

12

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

311

Page 14 of 32

Given the intensive plasma sampling procedure (up to 17 samples/subject),

312

plasma pharmacokinetics were characterized by non-compartmental analyses.

313

These analyses included computation of the area under the curve from time zero to

314

24 hours (AUC0-24) based on the linear-logarithm trapezoidal rule, maximum

315

concentration (Cmax) and time to Cmax (Tmax). Statistical comparisons of these

316

between group comparisons of pharmacokinetic parameters were performed using

317

the Kruskal-Wallis test. Matched comparisons by subjects who had repeated

318

measures was performed using the Wilcoxon-signed rank test. A p value of < 0.05

319

was qualified as statistically significant. Pharmacokinetic and statistical analysis

320

were performed using StataIC version 14.2 (StataCorp, College Station, TX) . All

321

figures were generated with the R 3.2.3 software package utilizing ggplot2 or with

322

Prism 6 (GraphPad Software, Inc.). Average values were shown with the standard

323

error of the mean (SEM). Significance was determined using nonparametric

324

statistical tests.

325 326

Results:

327

Study enrollment numbers and demographics

328

To determine eligibility for this study, 65 subjects were screened and 46 met

329

eligibility criteria. These 46 subjects participated in 60 GI catheter placement

330

procedures. Of these 60 catheter placement studies, 24 were discontinued. There

331

was 1 study visit that was discontinued after study drug administration due to

332

discomfort from the GI catheter. There were 23 study visits that were discontinued

333

prior to study drug administration due to the following reasons: inability to advance

ACS Paragon Plus Environment

13

Page 15 of 32

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

334

the GI catheter (n=1), vomiting (n=2), or discomfort caused by the GI catheter after

335

insertion (n=20). One subject experienced an adverse event during the study visit.

336

The subject became hypotensive and reported feeling hot and was clammy to the

337

touch during the study procedures. The subject was immediately placed into the

338

Trendelenburg position while cooling the forehead and neck with ice packs. The

339

subject’s blood pressure returned to baseline after 8 minutes and remained at that

340

level throughout the remainder of the study.

341

The plasma concentration-time information from all 25 subjects that

342

completed the study were reviewed individually and revealed two subjects as

343

extreme values. One subject had a rising concentration time profile over the 8 to 28-

344

hour period with concentrations at the last time-point that were two-fold higher

345

than at the mid-point. Non-adherence with the study protocol such as self-

346

administration of another dose of ibuprofen was assumed and as a precaution

347

samples collected from this subject were not included in subsequent analysis. The

348

second subject had plasma concentrations that were approximately ten-fold lower

349

on two study visits (fed conditions) than values measured in comparable subjects

350

with no reasonable explanation for these observations. Given the small sample size,

351

inclusion of these extreme value data from this subject would contribute to a larger

352

difference in the central tendency estimates of ibuprofen exposure in fed compared

353

to fated subjects (detailed in next section). As a precaution we removed this subject

354

from subsequent data analyses to not skew the effects of food on ibuprofen

355

dissolution and absorption.

ACS Paragon Plus Environment

14

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

356

Page 16 of 32

Details regarding study participants who completed the study procedure and

357

were included in our analysis are included in Table 1. There were a total of 23

358

subjects that completed the study, with 11 subjects returning for an additional study

359

visit (a total of 34 completed study visits) in our analysis. The subjects were

360

primarily white (61%), male (65%) with an average age of 30 years. The subjects

361

had a median [minutes, max] weight of 79 [52, 123] kg and body mass index of 25.7

362

[19.4, 37.7] kg/m2.

363

Table 1: Demographics of study subjects. Study Arm

Number of Subjects

Age (years)

N

Mean ± SD (MinutesMax) 30 ± 9 (18-54)

Total

23

Fed

10

32 ± 9 (22-49)

Fasted

13

29 ± 9 (18-54)

Body Mass Index (BMI) Mean ± SD (MinutesMax) 26.2 ± 4.6 (19.437.7) 27.0 ± 4.5 (21.635.9) 25.7 ± 4.7 (19.435.8)

Sex

Race

Ethnicity

Male

Female

Caucasian

AfricanAmerican

Asian

Hispanic or Latino

2

Not Hispanic or Latino 22

15

8

14

7

6

4

6

4

0

9

1

9

4

8

3

2

13

0

1

364 365 366

Plasma concentration after administration of ibuprofen The mean and standard deviation concentration time profiles from the 23

367

included subjects are displayed in Figure 1 as a semi-logarithm plot and as absolute

368

values by fasted and fed states. Individual concentration-time profiles of subjects by

369

fasted and fed conditions are shown in Figure 2. In both the fasted and fed

370

conditions ibuprofen was detected in the plasma 10 minutes after administration of

371

ibuprofen. As shown in Figures 1 and 2, the Cmax was lower and Tmax longer in the

372

fed versus fasted state.

ACS Paragon Plus Environment

15

Page 17 of 32

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

373

374 375

Figure 1. Average plasma concentration vs. time profiles of ibuprofen. Fasted

376

(n=20, green line) and fed (n=14, black line) conditions plotted in A) logarithmic

377

and B) linear scale. Error bars indicate the standard error of the mean (SEM). Data

378

from fasted subjects (n=20) and fed subjects (n=14) are shown.

379

380 381

Figure 2. Individual plasma concentration vs. time profiles of ibuprofen under

382

(A) fasted, and (B) fed conditions. Each line represents an individual subject.

ACS Paragon Plus Environment

16

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 32

383 384

Fed subjects had a significantly (p= 0.020) lower AUC0-24 value than those in

385

the fasted condition, though this difference was a mean of 5.42% lower. In contrast,

386

the difference in Cmax was 35.2% lower (p=0.025) in the fed versus fasted

387

conditions. This lower Cmax was also associated with a longer Tmax that was a mean

388

of 1.71 hours longer in the fed condition. The specific values and statistical

389

comparison of these parameters are provided in Table 2. To quantify intra-subject

390

variability, we analyzed the AUC0-24 from the 11 subjects that completed two

391

separate study visits under the same treatment conditions. The geometric mean

392

[90% confidence interval] plasma AUC0-24 ratio was 1.056 [0.929, 1.193].

393

Table 2. Plasma pharmacokinetic parameters of ibuprofen (mean +/-

394

standard deviation), including area under the curve from time zero to 24

395

hours (AUC0-24), maximum concentration (Cmax) and time to Cmax (Tmax). Study Arm

Number of Observations

Fasted

20

Fed

14

Statistical Significance

AUC0-24 (µg•hr/mL) 241.888 ± 88.907 229.452 ± 76.879 p = 0.020

Cmax (µg/mL) 58.247 ± 18.400 43.051 ± 14.312 p = 0.025

Tmax (hr) 2.980 ± 1.613 4.694 ± 1.994 p = 0.010

396

Significance was determined using the nonparametric Kruskal-Wallis equality-of-

397

populations rank test.

398 399 400 401

GI pH vales in the GI tract The average pH values in the GI tract are shown in Figure 3. Immediately after feeding the gastric pH increased from acidic values (2.3 ± 0.3) closer to

ACS Paragon Plus Environment

17

Page 19 of 32

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

402

neutrality (4.6 ± 0.58). The pH levels gradually decreased until the final

403

measurement 7 hours post dosing, returning to values seen prior to feeding (2.8 ±

404

0.64). After feeding, the pH of the small intestine increased from approximately 5 to

405

6. Fasted subjects had a relatively stable pH compared to the fed subjects

406

throughout the study. The pH values from each subject are shown in Figure 4.

407

Regardless of fed or fasted conditions, the pH in the jejunum showed much less

408

variability than the duodenum and stomach. Both of these regions exhibited large

409

variably between subjects.

410

411 412

Figure 3. Average pH values vs. time profiles throughout the GI tract. A) Fasted

413

and B) Fed study visits are shown. Mid jejunum (black line), Proximal jejunum (red

414

line), Duodenum (green line) and Stomach (blue line). Error bars indicate the

415

standard error of the mean (SEM). Data from fasted subjects (stomach: n=15,

416

duodenum: n=16, proximal jejunum: n=10, mid jejunum: n=12) and fed subjects

417

(stomach: n=13, duodenum: n=11, proximal jejunum: n=6, mid jejunum: n=7) are

418

shown.

419

ACS Paragon Plus Environment

18

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 32

420 421

Figure 4. Individual pH values vs. time profiles for each subject throughout the

422

GI tract. Each line represents an individual subject.

423 424

In vivo GI concentration of ibuprofen in the GI tract

425

The average concentration of ibuprofen detected in the GI tract is shown in

426

Figure 5 while the ibuprofen concentration in the GI tract for each individual study

427

visit is shown in Figure 6. GI fluid was aspirated 15 minutes post dosing and

428

continued at specified intervals until 7 hours post dosing. As shown in Figure 5,

429

initial samples had a higher concentration of ibuprofen in the stomach for fed

430

subjects while fasted subjects had a higher concentration of ibuprofen in the small

431

intestine. All subjects still had high levels of ibuprofen throughout the GI tract until

432

the last sampling time point, 7 hours post dosing.

433

ACS Paragon Plus Environment

19

Page 21 of 32

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

434 435

Figure 5. Average concentration of ibuprofen in luminal GI fluid supernatant

436

throughout the GI tract. A) fasted and B) fed conditions. Mid jejunum (black line),

437

Proximal jejunum (red line), Duodenum (green line) and Stomach (blue line). Error

438

bars indicate the standard error of the mean (SEM). Data from fasted subjects

439

(stomach: n=15, duodenum: n=16, proximal jejunum: n=10, mid jejunum: n=12) and

440

fed subjects (stomach: n=13, duodenum: n=11, proximal jejunum: n=6, mid jejunum:

441

n=6) are shown.

442

ACS Paragon Plus Environment

20

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 22 of 32

443 444

Figure 6. Individual ibuprofen concentrations in plasma and luminal GI fluid

445

supernatant. Each line represents an individual subject.

ACS Paragon Plus Environment

21

Page 23 of 32

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

446 447

Figure 7 shows the ibuprofen concentration profiles broken down into each

448

site of the stomach and small intestine. In the stomach under fasted condition, the

449

ibuprofen concentration (19,442 ng/ mL) was detected at 15 minutes post dosing.

450

Surprisingly, the high ibuprofen concentration in stomach remained for 7 hours

451

after dosing (20,055 ng/ mL). In the stomach under fed condition, the ibuprofen

452

concentration (90,637 ng/mL) was detected at 15 minutes post dosing. This was 4.7

453

fold higher than that of fasted condition and this increase of ibuprofen dissolution in

454

the stomach may be due to the higher pH under fed condition.

455

ACS Paragon Plus Environment

22

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

456

Page 24 of 32

Figure 7. Site comparison of the average concentration of ibuprofen in

457

luminal GI fluid supernatant in fed and fasting conditions. A) Stomach, B)

458

Duodenum, C) Proximal Jejunum, and D) Mid Jejunum. Error bars indicate the

459

standard error of the mean (SEM). Data from fasted subjects (stomach: n=15,

460

duodenum: n=16, proximal jejunum: n=10, mid jejunum: n=12) and fed subjects

461

(stomach: n=13, duodenum: n=11, proximal jejunum: n=6, mid jejunum: n=6) are

462

shown.

463 464

It is worth noting that although ibuprofen has higher dissolution in the

465

stomach under fed condition (due to higher pH), the ibuprofen concentration in the

466

duodenum is delayed (due to slow gastric emptying and transit). This delay resulted

467

lower plasma ibuprofen concentration. These data strongly suggest the gastric

468

emptying and transit (at fed state) is dominant to determine the plasma drug

469

concentration.

470 471

Discussion:

472

The goal of this clinical study was to directly measure drug dissolution in the

473

stomach and small intestine of healthy humans in order to better understand the in

474

vivo conditions that affect drug disintegration and dissolution. We chose to evaluate

475

the in vivo drug dissolution and systemic absorption of the BCS Class IIa drug

476

ibuprofen by in vivo sampling of stomach and small intestinal luminal content of

477

fasted and fed subjects. In this study we were able to characterize in vivo the

478

dissolution pattern of ibuprofen through to the mid-jejunum. In addition, this study

ACS Paragon Plus Environment

23

Page 25 of 32

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

479

was to determine influence of gastric pH and a fed state on dissolution. These in vivo

480

findings improved our understanding of the luminal profile of this poorly soluble

481

drug and can help to support future models of agents within this class.

482

The effect of food on the pharmacokinetics of immediate release NSAIDs has

483

recently been reviewed10. Having increased plasma levels of ibuprofen results in

484

increased analgesic effect of the drug11, however consumption of food can decrease

485

plasma levels of ibuprofen10. Ibuprofen is generally taken with food to improve

486

tolerability therefore we sought to understand the effect of food on in vivo drug

487

disintegration and dissolution of ibuprofen. Our results were consistent with

488

previous studies10 showing an overall decrease in the Cmax, delay of the Tmax, and

489

overall decrease in the AUC0-24 in the plasma of fed subjects compared to fasted

490

subjects. By utilizing direct in vivo sampling of the GI tract we were able to detect

491

ibuprofen 15 minutes after dosing in both fed and fasted conditions. In fasted

492

subjects ibuprofen was readily detected in the small intestine, which reflected in

493

higher plasma concentrations compared to fed subjects. There were high

494

concentrations of ibuprofen in the stomachs of fed subjects compared to fasted

495

subjects. The increase in pH increased drug dissolution in the stomach, however,

496

that did not increase the rate of systemic drug absorption. Ibuprofen is thought to

497

be poorly absorbed in the stomach of humans, with the small intestine being the

498

main site of absorption12. Our data suggests that feeding increases the stomach pH

499

which in turn increases drug dissolution, however this does not result in higher

500

plasma concentrations. The fed condition likely delay of gastric emptying and transit

501

of drug from stomach to duodenum which correlated with lower Cmax, lower AUC0-24,

ACS Paragon Plus Environment

24

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 26 of 32

502

and longer Tmax. These data strongly suggest that the GI transit a major determinant

503

for lower plasma drug concentration under fed state.

504

Our results, as well as results from others13, 14, show that there is remarkable

505

intersubject variability in the pH of the gastrointestinal tract. The low pH in the

506

stomach below the pKa of 4.4 for ibuprofen15, 16 correlates with low measurable

507

concentrations in this part of the GI tract. Consumption of food that leads to an

508

initial increase in stomach pH also leads to measurable stomach concentrations of

509

ibuprofen. However, food slows the rate of absorption leading to a lower Cmax and

510

longer Tmax but marginally alters the extent of absorption (as indicated by a similar

511

AUC0-24). This alteration in the concentration-time profile relevant for ibuprofen

512

because the analgesic effects of this drug are related to increased plasma levels of

513

the drug11. The observed extension in the Tmax by approximately 1.7 hours also

514

implies an expected delay in the therapeutic effects of this drug.

515

A major unexpected finding of this work was the prolonged high levels of

516

ibuprofen in the GI tract. Our results show that ibuprofen was detected in the GI

517

tract over the course of the seven hours of sampling. Although this was limited to 7

518

hours based on our sampling schema, it is likely that some subjects may have local

519

concentrations in the GI tract for longer periods of time. The implications of

520

prolonged GI retention of ibuprofen have not been previously characterized.

521

Previously, the transit in stomach is estimated from 30 minutes to 2 hours and

522

transit time in small intestine is average of 3 hours and these estimates were used in

523

drug absorption model prediction. The high drug concentration in the stomach and

524

small intestine for 7 hours may challenge these previous assumptions in drug

ACS Paragon Plus Environment

25

Page 27 of 32

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

525

absorption prediction. Better models may need to be developed for better in vitro

526

and in vivo correlation and oral absorption prediction.

527

In addition, it is known that prolonged use of NSAIDs are associated with an

528

increase in gastroduodenal ulceration. From a theoretical perspective, sustained

529

exposure of local GI tissue to ibuprofen may contribute to the peptic ulcer disease

530

associated with this drug. Our results suggest that there is prolonged exposure to

531

ibuprofen in the GI tract, especially after taking ibuprofen with food. Additionally,

532

food increases the initial ibuprofen convention approximately 5 fold compared to

533

fasted subjects 15 minutes after dosing. Our findings suggest we may need to

534

reconsider taking ibuprofen with food. While taking with food might alleviate acute

535

symptoms of GI discomfort it may lead to long term consequences by delaying

536

gastric emptying and leading to prolonged exposure of the stomach lining to

537

ibuprofen.

538

One potential limitation for this study is that with any aspiration catheter going

539

past the pylorus in the stomach there is a possibility of altering gastric emptying by

540

altering how the pylorus opens and closes. Additionally, the presence of a catheter

541

could have an effect on duodenogastric reflux and this in turn could affect drug

542

dissolution and absorption. However, this was examined in work by Muller-Lissner

543

et al. using a catheter of the similar size (3.0 mm compared to our 3.3 mm catheter

544

diameter)17. They found no effect on gastric emptying or duodenogastric reflux

545

when the catheter was placed past the pylorus. No effect of the catheter was found

546

in both fed and fasted conditions. Therefore, the method of using an aspiration

ACS Paragon Plus Environment

26

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 28 of 32

547

catheter to study drug dissolution and absorption should be similar compared to the

548

catheter no being present.

549

Another potential limitation is that the use of aspiration catheters can be

550

considered uncomfortable to the subject due to the presence of a foreign object

551

placed down the subject’s throat. This caused generalized anxiety and

552

uncomfortable feeling in the subject’s throat. We did have 36 out of 60 subjects

553

complete the entire study that had the tube placed for approximately 15 hours. This

554

success rate was similar to a previous study using a similar multi-lumen catheter

555

where the tube was placed for a similar length of time14. While the success rate is

556

lower than other studies5, this could be due to the length of time the tube was

557

placed. For example, work from Hens et al. had a higher success rate for the study

558

but the tube was in place for much less time (approximately 4 hours)5. The

559

generalized anxiety of having the tube placed for 15 hours in our study may have

560

decreased the success rate. However, having the tube placed for this long allowed us

561

to obtain manometry data as well has aspirate fluid for 7 hours after dosing that

562

resulted in a very large data set.

563

The use on aspiration catheters allows us to better understand the in vivo

564

conditions involved in to study drug dissolution and absorption. In this study we

565

presented the data obtained for the gastric and small intestinal pH as well as the

566

concentrations of ibuprofen in the GI tract and plasma. Further analysis of the data

567

set obtained in this clinical study is described in the manuscript by Hens et al.18. In

568

this work, Hens et al. further described how ibuprofen concentration in the stomach

569

and small intestine correlate to buffer capacity as well as motility. Additionally Hens

ACS Paragon Plus Environment

27

Page 29 of 32

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

570

et al. looks at the effect of pH and drug dissolution by analyzing the levels of

571

undissolved ibuprofen in the GI tract.

572

The ability to directly sample small intestinal luminal samples allowed us to

573

follow drug dissolution in vivo. The use of multi-lumen catheters to aspirate GI fluid

574

to study drug dissolution has been pioneered over 30 years ago, however relatively

575

few studies have been performed since19 (reviewed by Hens20 and Brouwers21). In

576

the past few years multiple groups have begun to utilize this technique to aspirate

577

samples from the stomach and GI tract5, 14, 20, 22. Because of this we are beginning to

578

get a better understanding of the complex GI physiological parameters involved in

579

drug dissolution and absorption. A multi-lumen GI catheter is a powerful tool to be

580

able to independently sample multiple sites in the GI tract to compare regional and

581

temporal drug disintegration and dissolution. Future work is needed to connect

582

motility, gastric emptying, buffer capacity, viscosity to better understand in vivo

583

drug dissolution and absorption. We hope the in vivo results obtained in this study

584

can be used to validate in vitro dissolution methodologies in order to support future

585

computational and mathematical modeling efforts. This will aid in the development

586

of an oral drug product optimization process that can be applied to future drugs in

587

order to maximize oral drug safety and efficacy.

588 589 590

Conclusion: In this study we used a novel multi-lumen catheter with the ability to aspirate

591

luminal GI fluid from the stomach and small intestine. Using this catheter, we were

592

able to identify that ibuprofen remains in stomach and small intestine for 7 hours.

ACS Paragon Plus Environment

28

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 30 of 32

593

The fed state delayed transit of ibuprofen into the duodenum (presumably due to a

594

delay in gastric emptying) compared to the fasted state. The fed state increased drug

595

dissolution in stomach (presumably due to increase of pH in the fed state) compared

596

to the fasted state however this did not result in an overall increase in plasma levels

597

of ibuprofen.

598 599 600

Acknowledgments: This research was funded by FDA grant HHSF223201310144C. Clinical

601

samples collected with help from Michigan Institute for Clinical & Health Research

602

(MICHR) NIH grant UL1TR000433. We also thank Bart Hens for thoughtful

603

discussion of this manuscript.

604

ACS Paragon Plus Environment

29

Page 31 of 32

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

605

References:

606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648

1. Amidon, G. L.; Lennernas, H.; Shah, V. P.; Crison, J. R. A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharmaceutical research 1995, 12, (3), 41320. 2. Tsume, Y.; Mudie, D. M.; Langguth, P.; Amidon, G. E.; Amidon, G. L. The Biopharmaceutics Classification System: subclasses for in vivo predictive dissolution (IPD) methodology and IVIVC. Eur J Pharm Sci 2014, 57, 152-63. 3. Van Den Abeele, J.; Brouwers, J.; Mattheus, R.; Tack, J.; Augustijns, P. Gastrointestinal Behavior of Weakly Acidic BCS Class II Drugs in Man--Case Study of Diclofenac Potassium. Journal of pharmaceutical sciences 2016, 105, (2), 687-96. 4. Van Den Abeele, J.; Schilderink, R.; Schneider, F.; Mols, R.; Minekus, M.; Weitschies, W.; Brouwers, J.; Tack, J.; Augustijns, P. Gastrointestinal and Systemic Disposition of Diclofenac under Fasted and Fed State Conditions Supporting the Evaluation of in Vitro Predictive Tools. Mol Pharm 2017. 5. Hens, B.; Corsetti, M.; Brouwers, J.; Augustijns, P. Gastrointestinal and Systemic Monitoring of Posaconazole in Humans After Fasted and Fed State Administration of a Solid Dispersion. Journal of pharmaceutical sciences 2016, 105, (9), 2904-12. 6. Rainsford, K. D. Ibuprofen: pharmacology, efficacy and safety. Inflammopharmacology 2009, 17, (6), 275-342. 7. Davies, N. M. Clinical pharmacokinetics of ibuprofen. The first 30 years. Clin Pharmacokinet 1998, 34, (2), 101-54. 8. Wolfe, M. M.; Lichtenstein, D. R.; Singh, G. Gastrointestinal toxicity of nonsteroidal antiinflammatory drugs. The New England journal of medicine 1999, 340, (24), 1888-99. 9. Singh, B. N. Effects of food on clinical pharmacokinetics. Clin Pharmacokinet 1999, 37, (3), 213-55. 10. Moore, R. A.; Derry, S.; Wiffen, P. J.; Straube, S. Effects of food on pharmacokinetics of immediate release oral formulations of aspirin, dipyrone, paracetamol and NSAIDs - a systematic review. Br J Clin Pharmacol 2015, 80, (3), 381-8. 11. Laska, E. M.; Sunshine, A.; Marrero, I.; Olson, N.; Siegel, C.; McCormick, N. The correlation between blood levels of ibuprofen and clinical analgesic response. Clin Pharmacol Ther 1986, 40, (1), 1-7. 12. Adams, S. S.; Bough, R. G.; Cliffe, E. E.; Lessel, B.; Mills, R. F. Absorption, distribution and toxicity of ibuprofen. Toxicol Appl Pharmacol 1969, 15, (2), 310-30. 13. Koziolek, M.; Grimm, M.; Becker, D.; Iordanov, V.; Zou, H.; Shimizu, J.; Wanke, C.; Garbacz, G.; Weitschies, W. Investigation of pH and Temperature Profiles in the GI Tract of Fasted Human Subjects Using the Intellicap((R)) System. Journal of pharmaceutical sciences 2015, 104, (9), 2855-63. 14. Yu, A.; Baker, J. R.; Fioritto, A. F.; Wang, Y.; Luo, R.; Li, S.; Wen, B.; Bly, M.; Tsume, Y.; Koenigsknecht, M. J.; Zhang, X.; Lionberger, R.; Amidon, G. L.; Hasler, W. L.; Sun, D. Measurement of in vivo Gastrointestinal Release and Dissolution of Three

ACS Paragon Plus Environment

30

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

649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683

Page 32 of 32

Locally Acting Mesalamine Formulations in Regions of the Human Gastrointestinal Tract. Mol Pharm 2017, 14, (2), 345-358. 15. Levis, K. A.; Lane, M. E.; Corrigan, O. I. Effect of buffer media composition on the solubility and effective permeability coefficient of ibuprofen. Int J Pharm 2003, 253, (1-2), 49-59. 16. Krieg, B. J.; Taghavi, S. M.; Amidon, G. L.; Amidon, G. E. In Vivo Predictive Dissolution: Comparing the Effect of Bicarbonate and Phosphate Buffer on the Dissolution of Weak Acids and Weak Bases. Journal of pharmaceutical sciences 2015, 104, (9), 2894-904. 17. Muller-Lissner, S. A.; Fimmel, C. J.; Will, N.; Muller-Duysing, W.; Heinzel, F.; Blum, A. L. Effect of gastric and transpyloric tubes on gastric emptying and duodenogastric reflux. Gastroenterology 1982, 83, (6), 1276-9. 18. Hens, B.; Tsume, Y.; Bermejo, M.; Paixao, P.; Koenigsknecht, M. J.; Baker, J. R.; Hasler, W. L.; Lionberger, R.; Fan, J.; Dickens, J.; Shedden, K.; Wen, B.; Wysocki, J.; Loebenberg, R.; Lee, A.; Frances, A.; Amidon, G.; Yu, A.; Benninghoff, G.; Salehi, N.; Talattof, A.; Sun, D.; Amidon, G. L. Low Buffer Capacity and Alternating Motility along the Human Gastrointestinal Tract: Implications for in Vivo Dissolution and Absorption of Ionizable Drugs. Mol Pharm 2017. 19. Jobin, G.; Cortot, A.; Godbillon, J.; Duval, M.; Schoeller, J. P.; Hirtz, J.; Bernier, J. J. Investigation of drug absorption from the gastrointestinal tract of man. I. Metoprolol in the stomach, duodenum and jejunum. Br J Clin Pharmacol 1985, 19 Suppl 2, 97S-105S. 20. Hens, B.; Corsetti, M.; Spiller, R.; Marciani, L.; Vanuytsel, T.; Tack, J.; Talattof, A.; Amidon, G. L.; Koziolek, M.; Weitschies, W.; Wilson, C. G.; Bennink, R. J.; Brouwers, J.; Augustijns, P. Exploring gastrointestinal variables affecting drug and formulation behavior: Methodologies, challenges and opportunities. Int J Pharm 2017, 519, (12), 79-97. 21. Brouwers, J.; Augustijns, P. Resolving intraluminal drug and formulation behavior: Gastrointestinal concentration profiling in humans. Eur J Pharm Sci 2014, 61, 2-10. 22. Litou, C.; Vertzoni, M.; Goumas, C.; Vasdekis, V.; Xu, W.; Kesisoglou, F.; Reppas, C. Characteristics of the Human Upper Gastrointestinal Contents in the Fasted State Under Hypo- and A-chlorhydric Gastric Conditions Under Conditions of Typical Drug - Drug Interaction Studies. Pharmaceutical research 2016, 33, (6), 1399-412.

ACS Paragon Plus Environment

31