Download as pdf or txt
Download as pdf or txt
You are on page 1of 38

Subscriber access provided by UNIV OF NEWCASTLE

Article
Low Buffer Capacity and Alternating Motility Along
The Human Gastrointestinal Tract: Implications for In
Vivo Dissolution and Absorption of Ionizable Drugs
Bart Hens, Yasuhiro Tsume, Marival Bermejo, Paulo Paixao, Mark Koenigsknecht, Jason R.
Baker, William L. Hasler, R. Lionberger, Jianghong Fan, Joseph Dickens, Kerby A. Shedden,
Bo Wen, Jeffrey Wysocki, Raimar Löbenberg, Allen Lee, Ann F. Fioritto, Gregory E. Amidon,
Alex Yu, Gail Benninghoff, Niloufar Salehi, Arjang Talattof, Duxin Sun, and Gordon L. Amidon
Mol. Pharmaceutics, Just Accepted Manuscript • DOI: 10.1021/acs.molpharmaceut.7b00426 • Publication Date (Web): 24 Jul 2017
Downloaded from http://pubs.acs.org on July 27, 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 37 Molecular Pharmaceutics

1
2
3 Submitted to Molecular Pharmaceutics ((featured topic issue, Industry-Academic Collaboration in Oral
4 Biopharmaceutics: The European IMI OrBiTo Project)
5
6 Low Buffer Capacity and Alternating Motility Along the Human
7
8
Gastrointestinal Tract: Implications for in vivo Dissolution and
9 Absorption of Ionizable Drugs
10

11 Bart Hens1, ‡Yasuhiro Tsume1, ‡Marival Bermejo2, ‡Paulo Paixao3, ‡Mark J. Koenigsknecht1, ‡Jason
12 R. Baker4, ‡William L. Hasler4, ‡Robert Lionberger5, ‡Jianghong Fan5, Joseph Dickens6, Kerby
13 Shedden6, Bo Wen1, Jeffrey Wysocki1, Raimar Loebenberg7, Allen Lee2, Ann Frances1, Greg
14 Amidon1, Alex Yu1, Gail Benninghoff1, Niloufar Salehi8, Arjang Talattof1, Duxin Sun1, ‡Gordon L.
15 Amidon1*
16
17
18

19 Bart Hens, Yasuhiro Tsume, Marival Bermejo, Paulo Paixao, Mark J. Koenigsknecht, Jason R.
20 Baker, William L. Hasler, Ann Frances Robert Lionberger, Jianghong Fan, Duxin Sun and Gordon L.
21 Amidon are the primary authors.
22
23
24 1
Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor,
25
26 Michigan 48109, USA
27 2
Department Engineering Pharmacy Section, Miguel Hernandez University, San Juan de Alicante,
28
03550 Alicante, Spain
29
30 3
Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av.
31 Professor Gama Pinto, 1649-003 Lisboa, Portugal
32
4
33 Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor,
34 Michigan 48109, USA
35
5
36 Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug
37 Administration, Silver Spring, MD, USA
38 6
39 Department of Statistics, University of Michigan 48109, Ann Arbor, USA
40 7
Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
41
42 8
Center for the Study of Complex Systems and Department of Chemical Engineering, University of
43 Michigan, Ann Arbor, Michigan 48109-2136, USA
44
45 Corresponding Author
46 * Prof. Dr. Gordon L. Amidon
47
48 428 Church St.
49
50 College of Pharmacy
51
52
University of Michigan
53 Ann Arbor, MI 48109-1065
54
55 Phone: +(1) 734-764-2226
56
57
Fax: +(1) 734-764-6282
58 Email: glamidon@med.umich.edu
59
60 1
ACS Paragon Plus Environment
Molecular Pharmaceutics Page 2 of 37

1
2
3 Graphical Abstract
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24 Keywords
25 clinical study; in vivo dissolution; local drug concentration in the GI tract; ibuprofen; immediate
26
27 release; bioequivalence; bioavailability; oral absorption; buffer capacity
28
29
30 Abstract
31
In this study, we determined the pH and buffer capacity of human gastrointestinal (GI) fluids
32
33
(aspirated from the stomach, duodenum, proximal jejunum and mid/distal jejunum) as a function of
34
35
time, from 37 healthy subjects after oral administration of an 800 mg immediate-release tablet of
36
37
38 ibuprofen (reference listed drug; RLD) under typical prescribed bioequivalence (BE) study protocol
39
40 conditions in both fasted and fed state (simulated by ingestion of a liquid meal). Simultaneously,
41
42 motility was continuously monitored using water-perfused manometry. The time to appearance of
43
44 phase III contractions (i.e. housekeeper wave) was monitored following administration of the
45
46 ibuprofen tablet. Our results clearly demonstrated the dynamic change in pH as a function of time and,
47
48 most significantly, the extremely low buffer capacity along the GI tract. The buffer capacity on
49
50 average was 2.26 µmol/mL/∆pH in fasted state (range: 0.26 and 6.32 µmol/mL/∆pH) and 2.66
51
52 µmol/mL/∆pH in fed state (range: 0.78 and 5.98 µmol/mL/∆pH) throughout the entire upper GI tract
53
54 (stomach, duodenum, proximal and mid/distal jejunum). The implication of this very low buffer
55
56 capacity of the human GI tract is profound for the oral delivery of both acidic and basic active
57
58 pharmaceutical ingredients (APIs). An in vivo predictive dissolution method would not only require a
59
60 2
ACS Paragon Plus Environment
Page 3 of 37 Molecular Pharmaceutics

1
2
3 bicarbonate buffer, but more significantly, a low buffer capacity of dissolution media to reflect in vivo
4
5 dissolution conditions.
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 3
ACS Paragon Plus Environment
Molecular Pharmaceutics Page 4 of 37

1
2
3 Introduction
4 Predicting in vivo outcomes with computational software today, including variation in oral
5
6 absorption, requires accurate and biorelevant input functions reflecting the dynamic environment of
7
8 the gastrointestinal (GI) tract. The in vitro USP 1 and 2 dissolution tests are very useful in a quality
9
10 control (QC) setting and, in some cases, these tests can be used for a biowaiver of in vivo
11
12 bioequivalence (BE) studies for immediate release (IR) dosage forms (BCS class I (high solubility,
13
14 high permeability) and III (high solubility, low permeability) compounds) since gastric emptying is on
15
16 average slower than the in vitro dissolution of the API. Thus, gastric emptying would control plasma
17
18 variation of two oral products (i.e. marketed and generic) with the same API. Nevertheless, these QC
19
20 methods do not capture the inter- and intra-individual differences of GI variables as well as in
21
22 systemic outcomes (plasma AUC, Cmax and Tmax) after oral administration of oral IR drug products.1,2
23
24 In the case of slower dissolution and/or particulary lower solubility, various physiological GI variables
25
26 will impact the disintegration and dissolution of the formulation and drug, and thus the time-dependent
27
28 presentation of the drug to the intestinal absorbing membrane along the intestinal tract.3
29
30
31 As recently demonstrated, the impact of gastric motility and the variation in the fasted state on
32
33 oral drug absorption may be one of the major determinants of variability in bioavailability, particularly
34
35 Cmax among normal subjects in the fasted state.4 The time of intake of the oral dosage form relative to
36
37 the fasted state cyclic motility pattern (i.e. migrating motor complex (MMC)5) will cause significant
38
39 variation in gastric emptying and presentation of the drug to the absorbing membrane and thus,
40
41 ultimately, systemic availability of the drug. In fasting conditions, the GI muscular system contracts
42
43 with a cyclical periodic interdigestive MMC, in which the phases of quiescence (phase I) are followed
44
45 up by a phase of intermittent contractions (phase II) and a final phase of very strong contractions
46
47 (phase III) resulting in a motility varying gastric emptying rate.5,6
48
49
When no digestible food is administered, this cyclical fasted state motility pattern will be
50
51
repeated periodically. In 1973, Heading and colleagues found a strong correlation between plasma
52
53
Cmax, Tmax and the rate of gastric emptying of acetaminophen (expressed as gastric half-life; T1/2,G), an
54
55
56
early demonstration that gastric emptying is the rate-limiting step for absorption of acetaminophen
57
58 (which is a BCS class I compound).7 More recently, similar results were noted for BCS class III
59
60 4
ACS Paragon Plus Environment
Page 5 of 37 Molecular Pharmaceutics

1
2
3 compounds, i.e. fluorouracil and diethylcarbamazine, when applying periodic gastric emptying rates
4
5 with computational modeling software developed to reflect the GI motility variation based on
6
7 available published data.4 These results suggest that strong phase III contractions stimulate gastric
8
9 emptying of drug content in such way that a large amount of drug is directly available for absorption
10
11 in the small intestine, being a main determinant of the plasma Cmax and Tmax of the drug. Most
12
13 significantly, in BE studies, we usually dose randomly relative to the this motility cycle and, therefore,
14
15 introduce this random variable, independent of dosage form, into normal BE studies.
16
17 Further, the dynamic nature of the pH along the GI tract, and most significantly the very low
18
19 buffer capacity of the GI fluids, will substantially alter drug dissolution for ionizable drug
20
21 substances.8,9 In the case of carboxylic acids (e.g. ibuprofen, diclofenac, ketoprofen, flurbiprofen,
22
23 naproxen, etc.), the dissolved fraction of drug substance along the GI tract depends on the existing
24
25 luminal pH and buffer capacity.10–12 The pKa values reported for this group of medicines range from
26
27 3.9 to 5.8, where solubility and dissolution rate are highly dependent on the duodenal pH and buffer
28
29 capacity at the time of gastric emptying.13 At low physiological pH values (e.g. stomach), these drug
30
31 substances are poorly soluble in gastric fluids (pH 1-2) and become much more soluble in the dynamic
32
33
pH range of the intestinal fluids. However, while a static intestinal pH value of 6.5 (fasted state
34
35
simulated intestinal fluid (FaSSIF)) and 6.8 (USP simulated intestinal fluid (SIF)) are frequently used
36
37
in in vitro dissolution experiments, the GI environment is known to be much more dynamic with a pH
38
39
ranging from 2.4 to 7.5 in the duodenum.14,15 Duodenal pH and buffer capacity were described as
40
41
42 highly variable depending on the active pancreatic and mucosal cell secretion of bicarbonate to
43
44 neutralize gastric content variably emptying from the stomach, depending on the fasted state motility
45
46 cycle.9,16–19 As stated by Dooley and colleagues, the contribution of each phase to the cycle length in
47
48 the antrum is 55% for phase I, 41% for phase II and 4% for phase III.20 The pancreatic bicarbonate
49
50 secreted into the small intestine varies in the fasted state with the motility cycle (4-21 mM bicarbonate
51
52 concentration)16,21. In addition to pancreatic secretions, duodenal epithelium cells secrete bicarbonate
53
54 too and protect the duodenal epithelium from the gastric fluids discharged from the stomach.9
55
56
57
58
59
60 5
ACS Paragon Plus Environment
Molecular Pharmaceutics Page 6 of 37

1
2
3 The reported differences in buffer capacity/pH values of human intestinal fluids are highly
4
5 variable.22 This dynamic, intestinal environment is, however, not reflected in conventional dissolution
6
7 media (e.g. FaSSIF and USP SIF). FaSSIF (pH 6.5) and USP SIF (pH 6.8) retaining a strong buffer
8
9 capacity of 12 and 18.4 µmol/mL/∆pH, respectively, equal to a 5 and 7.7 times higher buffer capacity
10
11 compared to human intestinal fluid (ranging from 2.4-5.6 µmol/mL/∆pH).12,23 Further, much larger
12
13 dissolution volumes of simulated GI fluids are used in current in vitro dissolution methodologies than
14
15 are present in the human GI tract based on magnetic resonance imaging (MRI) studies.24 This may
16
17 certainly contribute as one of the major factors for poor in vivo dissolution predictions. The applied
18
19 phosphate buffer is not a biorelevant reflection of the more complex bicarbonate/CO2 buffer25, the
20
21 predominant buffer in the GI tract.9 In vitro dissolution studies demonstrated the added value of using
22
23 bicarbonate buffers instead of phosphate buffers.9,16,26 For example, physiological bicarbonate buffers
24
25 proved to be more discriminative of the drug release behavior of enteric-coated formulations for
26
27 ileocolonic delivery, resulting in better reflections of in vivo disintegration-dissolution times than
28
29 observed for conventional phosphate buffers.26,27 As stated by Tsume and colleagues, the adoption of
30
31 inappropriate buffer media for in vitro BE studies would likely cause a failure in achieving an in vitro-
32
33
in vivo correlation (IVIV-C) and BE/bioavailability (BA) for oral drug products.12
34
35
36 The broad aim of this project was to map the link between the drug product disintegration and
37
38 drug dissolution in vivo, the presentation of the drug to the intestinal mucosal absorbing surface, and
39
40 systemic availability of an orally administered ibuprofen tablet (800 mg; RLD)28. Ibuprofen is highly
41
42 permeable and shows no limits in dissolving at the neutral pH of the small intestine (BCS class 2a
43
44 drug), as demonstrated by Tsume et al.12 However, we hypothesize that inter-individual variability in
45
46 systemic outcome for ibuprofen may be caused by differences in drug release and dissolution of the
47
48 drug along the entire GI tract. Therefore, this research proposal wants to explore how (i) alternating
49
50 motility patterns and (i) alternating buffer capacity and pH changes along the GI tract contribute to
51
52
inter-individual variability in systemic response of ibuprofen (in terms of Cmax, Tmax and AUC).
53
54
55
56
57
58
59
60 6
ACS Paragon Plus Environment
Page 7 of 37 Molecular Pharmaceutics

1
2
3 Materials and Methods
4 Chemicals
5
6 Acetonitrile and pH buffer solutions (pH 4.00, 7.00 and 10.00) were purchased from VWR
7
8 International (West Chester, PA). Methanol and HCl were obtained from Fisher Scientific (Pittsburgh,
9
10 PA). Ibuprofen, ibuprofen-D3 and NaOH were received from Sigma-Aldrich (St. Louis, MO). Mineral
11
12 oil was purchased from Acros Organics (Morris Plains, NJ). Immediate-release tablets of ibuprofen
13
14 administered in the clinical study were obtained from Dr. Reddy’s Laboratories Limited (Shreveport,
15
16 LA; IBU™ – Ibuprofen Tablets, USP, 800 mg, Lot number L400603). Pulmocare® was obtained from
17
18 Abbott Nutrition (Lake Forest, IL). Purified water (i.e. filtrated and deionized) was used for the
19
20 analysis methods (Millipore, Billerica, Massachusetts).
21
22
23 Intraluminal and Systemic Profiling of Ibuprofen in Healthy Volunteers
24
25 Samples collected in this study were part of clinical trial NCT02806869. The institutional review
26
27 boards at the University of Michigan (IRBMED, protocol number HUM00085066) and the
28
29 Department of Health and Human Services, Food and Drug Administration (Research Involving
30
31 Human Subjects Committee/RIHSC, protocol number 14-029D) both approved the study protocol. All
32
33 subjects provided written informed consent in order to participate. The study was carried out
34
35 accordance with the protocol, International Conference on Harmonization Good Clinical Practice (ICH
36
37 GCP) guidelines, and applicable local regulatory requirements. The protocol of the clinical study has
38
39 recently been described by Koenigsknecht et al.15 Briefly, two experimental treatment arms were
40
41 tested in 25 subjects: intake of one IR tablet of ibuprofen (IBU™ – Ibuprofen Tablets, USP, 800 mg,
42
43 Dr. Reddy’s Laboratories Limited) in fasted state with water or in fed state conditions simulated by
44
45 intake of a liquid meal (Pulmocare®) prior to drug administration with approximately 250 mL of
46
47 water. Of all 25 subjects, 12 individuals performed a second study visit in order to generate intra-
48
49 subject variability data (Table 1).
50
51
52
53
54
55
56
57
58
59
60 7
ACS Paragon Plus Environment
Molecular Pharmaceutics Page 8 of 37

1
2
3
4
5 Table 1: Overview of the study course in terms of (i) the ID of all subjects, (ii) test condition that was performed (fasted or fed state) and
6 (iii) the number of study visits. (*) Subject B003 withdrew from study 2.5 h after dose administration. (*1) Only plasma samples and motility
7 data were collected for subject B044; no GI fluid collection. Data included in the table are derived from Koenigsknecht et al.15
8
9 Subject ID Treatment Arm Study Visit #1 Study Visit #2
10
11 B002 Fed X X
12 *B003 Fasted X
13 B004 Fasted X X
14
15 B005 Fasted X X
16 B006 Fasted X
17 B008 Fed X
18
B017 Fasted X X
19
20 B020 Fed X X
21 B022 Fed X X
22 B026 Fed X
23
24 B031 Fed X
25 B034 Fed X
26 B041 Fed X
27
B042 Fasted X X
28
29 B043 Fed X X
30 1
* B044 Fasted X
31 B046 Fed X
32
33 B049 Fasted X X
34 B052 Fasted X
35 B053 Fasted X
36
B055 Fasted X X
37
38 B060 Fed X X
39 B063 Fasted X
40 B065 Fasted X X
41
42 B066 Fed X
43
44
45 In total, 20 fasted and 17 fed state test conditions were performed. Upon arrival in the hospital,
46
47 a customized aspiration multi-channel catheter (body length 292 cm; MUI Scientific, Mississauga,
48
49 ON) was intubated via the mouth and positioned in the mid-jejunum, proximal jejunum, duodenum
50
51 and stomach. Each segment contains aspiration and motility channels to aspirate GI fluids and to
52
53 monitor motility patterns along the GI tract, respectively. Positioning and guiding of the catheter were
54
55 verified by fluoroscopy (Figure 1).
56
57
58
59
60 8
ACS Paragon Plus Environment
Page 9 of 37 Molecular Pharmaceutics

1
2
3
4
5
6
7
8
9
10
Stomach
11
12
13
14 Duodenum
15 aspirates
16
17
18
19 Proximal
20 jejunum
21
22
23
24 Mid
25 jejunum
26
27
Figure 1: Fluoroscopic image of the position of a 23-channel catheter in the GI tract of subject ID B022, study visit #2. The arrows indicate
28 the different ports present in each segment of the GI tract.
29
30
31 After checking the positioning of the catheter, volunteers were asked to remain in a hospital bed in an
32
33 upright sitting position. After performing a baseline motility test of 3-5 h (see next paragraph), the
34
35 ibuprofen tablet was administered together with 250 mL of water containing 25 mg of USP grade
36
37 phenol red as a non-absorbable marker for monitoring GI fluid changes related to dilution, secretion,
38
39 and absorption. To induce the fed state condition, volunteers were asked to drink two cans of
40
41 Pulmocare® (total volume of 474 mL, containing 29.6 g of proteins, 44.2 g of fat, 25 g of
42
43 carbohydrates and a total amount of 710 calories) prior to dose administration. Volunteers were not
44
45 obliged to drink the total amount of administered water and/or liquid meal to avoid any feeling of
46
47
nausea at the start of the study. The study drug, water, and/or Pulmocare was swallowed by the
48
49
50 subject and was not administered via the GI catheter. After study drug administration, GI fluids were
51
52 aspirated at specific predetermined time points for 7 h. Immediately after aspiration of GI fluids, pH
53
54 was measured ex vivo using a pH electrode (Mettler InLab® Micro Pro, Mettler-Toledo LLC,
55
56 Columbus, OH), suitable for measuring pH in small or large volumes. After oral administration of the
57
58 drug, GI fluids were aspirated at specific predetermined time points for 7 h. The GI fluid samples were
59
60 9
ACS Paragon Plus Environment
Molecular Pharmaceutics Page 10 of 37

1
2
3 centrifuged at 21,000 x g for 5 minutes and the supernatant was collected and stored at -80°C until
4
5 analysis. Blood samples were collected for up to 24 h to monitor ibuprofen systemically. Blood
6
7 samples were added to venous blood collections tubes (K2EDTA (spray-dried), 7.2 mg) and plasma
8
9 was separated from blood samples by centrifugation and stored at -80°C until analysis. Blood samples
10
11 were collected for 24 h to monitor ibuprofen systemically. LC-MS/MS analyses were performed using
12
13 a Shimadzu HPLC system interfaced to an AB SCIEX QTRAP 5500 mass spectrometer by a TurboV
14
15 electrospray ionization (ESI) source (Applied Biosystems/MDS Sciex, Toronto, Canada). Ibuprofen-
16
17 D3 was used as an internal standard (IS) to normalize variation during sample preparation and LC-
18
19 MS/MS analyses. Chromatographic separation was achieved using a 2.1 x 50 mm, 3.5 µm Agilent
20
21 ZORBAX Extend-C18 column. The injection volume was 5 µL and the flow rate was kept constantly
22
23 at 0.4 mL/min. Mobile phase A and B were water and acetonitrile, respectively. Both water and
24
25 acetonitrile contained 0.1% acetic acid (v/v). The flow gradient was initially 98:2 v/v of A:B for 0.5
26
27 min, linearly ramped to 5:95 A:B over 1 min. To completely wash the column, the gradient was held
28
29
at 5:95 A:B for 2 min and then returned to 98:2 over 0.5 min. This condition was held for 3 min prior
30
31
to the injection of the sample and ibuprofen was detected at 2.48 min. The mass spectrometer was
32
33
operated at ESI negative ion mode ([M-H]-) and multiple reaction monitoring (MRM) was used for
34
35
36
monitoring the transitions of m/z 205.1→159.1 and m/z 208.2→161.2 for ibuprofen and ibuprofen-D3
37
38 (IS), respectively. Protein precipitation with methanol was used to extract ibuprofen from the human
39
40 plasma. 60 µL of each plasma sample was mixed with 180 µL of methanol containing 500 ng/mL
41
42 ibuprofen-D3 in a 96-well plate. The mixture was vortexed for 60 seconds at a high speed. The 96-
43
44 well plate was centrifuged at 3000 x g for 20 min to precipitate proteins at 4 °C. The clear supernatants
45
46 were collected and 5 µL of the supernatant was injected for LC-MS/MS analysis. Sample preparation
47
48 of GI fluid was similar to the plasma samples. All of the GI fluid samples were diluted 10-fold with
49
50 the blank human plasma before protein precipitation in methanol containing 500 ng/mL ibuprofen-D3
51
52 in order to reduce matrix effect on GI fluid samples and ibuprofen in GI fluid was quantified with the
53
54 plasma calibration curve. Stock solutions of ibuprofen (2 mg/mL) and ibuprofen-D3 (IS) (5 mg/mL)
55
56 were prepared in methanol and dimethyl sulfoxide, respectively. To prepare the calibration standards,
57
58 the 2 mg/mL ibuprofen stock solution was diluted to 500 µg/mL using methanol, which was spiked
59
60 10
ACS Paragon Plus Environment
Page 11 of 37 Molecular Pharmaceutics

1
2
3 with blank human plasma to provide a final ibuprofen plasma concentration of 10 µg/mL. Then serial
4
5 dilution with the blank human plasma was carried out to provide a series of calibration standards from
6
7 2.5-10,000 ng/mL. Quality control (QC) plasma solutions at 5, 25, 250, 5000, and 10000 ng/mL were
8
9 prepared similarly using separately weighed methanol stock solutions of ibuprofen (2 mg/mL).
10
11 Methanol was used to further dilute the stock ibuprofen-D3 solution to 500 ng/mL for protein
12
13 precipitation during sample preparation, also to minimize the variation. According to the FDA
14
15 guidelines, validation procedures were performed in human plasma and GI fluid diluted with human
16
17 plasma.15,29 These procedures included: (A) specificity and selectivity; (B) recovery and matrix effects
18
19 at low (0.015 µg/mL), medium (0.1 and 10 µg/mL), and high (35 µg/mL) concentrations; (C)
20
21 calibration curve with a correlation coefficient (r) of more than 0.98; (D) precision and accuracy, with
22
23 the intra-day and inter-day assay precision and accuracy estimated by analyzing six replicates at three
24
25 QC levels; (E) stability, with all stability studies conducted at three concentration levels in the
26
27 biomatrix at room temperature, 4 °C, −20 °C, and −80 °C; and (F) dilution integrity, with experiments
28
29 carried out with blank biomatrix. Accuracy and precision errors (< 15%) met the FDA requirements
30
31 for bioanalytical method validation. All samples were analyzed by the PK Core at the University of
32
33
Michigan.
34
35
36
37
Intraluminal Profiling of Periodic Motility Patterns along the GI Tract in Healthy Volunteers
38
39
After positioning, the catheter was connected to a computer console that generated real-time
40
41
42 manometry recordings in the different segments of the GI tract (Medical Measurement Systems,
43
44 Dover, NH). The manometric channels attached to the catheter were perfused with sterilized water at a
45
46 rate up to 2 mL/min and served as intestinal pressure recording ports to assess intestinal motility. Each
47
48 segment contained four motility channels to monitor pressure events. Baseline intestinal motility was
49
50 evaluated for 3-5 h prior to study drug administration of the tablet. Subsequently, GI motility was
51
52 measured continuously for 7 h. In fasting conditions, periods of active GI motility (phase II-III)
53
54 alternated between periods of quiescence (phase I). After food intake, the proximal stomach (fundus)
55
56 serves as a reservoir and will initiate tonic contractions/relaxations similar in strength as phase II
57
58
59
60 11
ACS Paragon Plus Environment
Molecular Pharmaceutics Page 12 of 37

1
2
3 contractions (approximately 3 contractions per min).30,31 The strength and number of pressure events
4
5 were displayed as a function of time (Figure 2).
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 Figure 2: Water-perfused manometry recording of the MMC in subject B049, study visit #2. The indicated area demonstrates the initiation
39 of antral phase III activity, propagating more distally towards the intestinal segments as a function of time.
40
41
42 We identify MMC phase III motility periods using spectral density estimation and penalized
43
44 logistic regression. The spectral density estimate of a manometry sample decomposes the sample into
45
46 a series of periodic components of decreasing frequencies and associated energies for each periodic
47
48 component.32 Figure 2 demonstrates the start of a powerful antral phase III contractions defined as the
49
50 occurrence of regular 2-3 contractions per minute for at least 2 minutes with an average amplitude of
51
52 75 mmHg.5,33 Duodenal phase III contractions are characterized by 11-12 contractions per minute with
53
54 an average amplitude of 33 mmHg which can last for at least 3 minutes.5 As the contractile activity
55
56 propagates, it becomes less spatiotemporally organized resulting in slower propulsion rates in the
57
58 distal small bowel.34,35 The corresponding spectral density estimate of a phase III period will have high
59
60 12
ACS Paragon Plus Environment
Page 13 of 37 Molecular Pharmaceutics

1
2
3 energy levels in the 10-12 cycles/min components, leading to a concentrated spectrum. During non-
4
5 phase III motility, the spectral density will have a more diffuse spectrum. Using penalized logistic
6
7 regression, we find that the proportion of energy in the 9-12 cycle/min frequencies are an important
8
9 predictor of phase III motility.
10
11
12 Measurement of Buffer Capacity in Aspirated Human Intestinal Fluids Ex Vivo
13 Briefly, buffer capacity was measured by adding accurate amounts of HCl to 100 µL of fasted
14
15
or fed duodenal/jejunal fluid and measuring the pH change.36 In case of gastric samples, fasted state
16
17
gastric fluids were titrated with accurate amounts of NaOH, whereas fed state gastric fluids were
18
19
titrated with accurate amounts of HCl due to the elevation of pH by liquid meal intake. For each
20
21
22
sample, 50 µL of pure mineral oil was added in order to prevent the loss of carbon dioxide (CO2)
23
24 during buffer capacity measurements.9 Accurate and precise pH measurements were assured by
25
26 calibration of the pH electrode (Mettler InLab® Micro Pro, Mettler-Toledo LLC, Columbus, OH)
27
28 using three different pH buffer solutions (pH 4.00, pH 7.00, and pH 10.00) prior to the start of each
29
30 experimental day, resulting in a Pearson Coefficient of Determination (R2) of 95 ± 0.9% (mean ± SD).
31
32 The sample has a buffer capacity value of 1 when one equivalent of strong acid or alkali is required to
33
34 change the pH value of 1 L by one pH unit.37
35
36
37
38 Neutralizing Effect of Bicarbonate on Gastric Acid and the Effect of Ibuprofen Dissolution on pH
39 The available bicarbonate (HCO3-) present in the intestinal tract reacts with hydrogen ions (H+)
40
41 to neutralize the gastric acid (HCl) emptied from the stomach. The formation of carbonic acid (H2CO3)
42
43
produces, in turn, CO2 and H2O through a dehydration reaction:
44
45
46  - +   ⇌ 
 ⇌ 
 + 
(Reaction 1)
47
48 This mentioned pathway is a reversible process and concentrations of each of the component are
49
50 depending on the corresponding equilibrium constants. As gastric fluids (pH 2=0.01 M HCl) would
51
52 empty the stomach as a first order kinetic process, a mass transport analysis was performed that
53
54 describes the appearing duodenal concentrations of H+:38
55
56

   
 =    −  
57 
 (Equation 1)
58    

59
60 13
ACS Paragon Plus Environment
Molecular Pharmaceutics Page 14 of 37

1
2
3 where  is the concentration of HCl that appears in the duodenum as a function of time t , Q  stands
4
5 for the amount of HCl present in the stomach (0.01 M HCl in 35 mL of fluids = 350 µmol)39, V# is the
6
7 constant duodenal volume (40 mL)40, k % is the first order gastric emptying rate constant (0.046 min-1)
8
9 and F is the constant flow rate (1.6 mL/min) from duodenal compartment to the more distal segments
10
11 of the intestinal tract.38 The maximum concentration of HCl that was observed in the duodenum was
12
13 used to react with HCO3- (Reaction 1) present in the duodenal compartment (minimum concentration
14
15 of 4 mM and maximum concentration of 21 mM, as reported in literature).16,21 Based on the simulated
16
17 concentrations of HCl in the duodenum, the reaction with HCO3- and corresponding duodenal pH can
18
19 be carried out by the following calculation:
20
21
() * +(),-. / +0 23 ),-. / ()1 ,-. +0
22 &'  -) = ()1 ,-. +0
⇌ (  + = (),-. / +0
⇌ −456 ( + = 7 (Equation 2)
23
24
25
26 where Ka (HCO3-) is equal to 4.47x10-7. [HCO3-]f and [H2CO3]f are the final concentrations of HCO3-
27
28 and H2CO3 after reaction of H+ (maximal simulated H+ concentration) and HCO3- (0.2 M).
29
30 Besides the fluctuations that may occur due to differences in H+ stomach secretions and
31
32 duodenal HCO3- secretions, pH along the intestinal tract can be altered by the present drug. Dissolution
33
34
of a weak acid or base may decrease or increase intestinal pH, depending on the strength of the acid
35
36
(Ka) or base (Kb). Dissolution of ibuprofen (weak acid (HA); Ka = 1.23 x 10 -5) in the intestinal fluids
37
38
can be described by following reaction:
39
40
41 8 + 
 ⇌ 8- +   (Reaction 2)
42
43
44 Based on the amount (X) of ibuprofen that dissociates as A- and H+, the drop in pH can be perceived
45
46 by the following calculation:
47
48
() * +(F/ + G1 G1
49 &' 9:;7<5=> = 1,23 × 10E = = = ⇌ (  + = H = I &' × (8+ ⇌ −456 (  + = 7
()F+ ()F+ ()F+
50
51 (Equation 3)
52
53
54 The value that will be applied for [HA] is equal to the average solution concentrations as observed in
55
56 the fasted duodenum as a function of time in this study.
57
58
59
60 14
ACS Paragon Plus Environment
Page 15 of 37 Molecular Pharmaceutics

1
2
3 Results and Discussion
4 Oral Drug Behavior of Ibuprofen Along the GI tract: Monitoring of Intraluminal Concentrations,
5 Post-dose Phase III Contractions, pH and Buffer Capacity
6
7 Several studies have determined the ibuprofen plasma levels after oral administration of
8
9 different dose strengths of the drug in fasting and fed state conditions.41–44 Generally, all studies
10
11 demonstrated a decrease in systemic Cmax/AUC (ratio Cmax fed/fasted: 0.81; ratio AUCfed/fasted: 0.90) and a
12
13 delay in Tmax (ratio Tmax fed/fasted: 1.33) after oral administration of ibuprofen in postprandial conditions.
14
15 Similar ratios fed/fasted state were observed in this study (0.69, 0.77 and 1.67 for Cmax, AUC, and Tmax
16
17 respectively). A more controlled and delayed gastric emptying of ibuprofen in the fed state conditions
18
19 may explain the lower and delayed exposure of ibuprofen in plasma as compared to the fasting state
20
21 conditions as it has been demonstrated that the intake of a liquid meal delays or slows gastric
22
23 emptying significantly.38 The results of this study with simultaneous intestinal sampling and
24
25 manometry recording of GI motility are in line with earlier published systemic data on ibuprofen. In
26
27 this study, we investigated the causes of both inter- and intra-subject variability. For example, in fasted
28
29 state conditions, plasma Cmax ranged from 28100 to 89700 ng/mL, while plasma Tmax varied from 1 to
30
31 8 hour. In fed state conditions, plasma Cmax and Tmax varied from 5700 to 64572 ng/mL and 1 to 8
32
33 hour, respectively. Thus we hypothesized that the underlying cause of this variability in plasma levels
34
35 is due to (i) the time of drug intake relative to the present interdigestive cyclic motility in the stomach
36
37 (i.e. phase I, II or III), both subject to subject and within the subject and (ii) the dynamic change in pH
38
39 as well as the base pH difference along the GI tract.
40
41
42 The time to appearance of the maximal concentration of ibuprofen in plasma (plasma Tmax) is
43
44 plotted as a function of the time to appearance of the maximal solution concentration of ibuprofen in
45
46 the duodenum (duodenal Tmax; Figure 3A) and in the jejunum (jejunal Tmax; Figure 3B).
47
48
49
50
51
52
53
54
55
56
57
58
59
60 15
ACS Paragon Plus Environment
Molecular Pharmaceutics Page 16 of 37

1
2
3
4
5 A B
9 9
6
7 8 8
8 7 7

Plasma Tmax (h)


Plasma Tmax (h)

9 6 6
10
5 5
11
12 4 4
13 3 y = 0.6932x + 1.506 3 y = 1.008x + 0.6247
14 (0.158 - 0.612)
2 2 (0.139 - 0.497)
R² = 0.4062
15 R² = 0.6534
1 1
16
0 0
17
0 1 2 3 4 5 6 7 8 9 0 1 2 3 4 5 6 7 8 9
18
Duodenal Tmax (h) Jejunal Tmax (h)
19
20 Fasted State Fed State Fasted State Fed State
21
22
23 Figure 3: Plot of plasma Tmax (h) as a function of (A) duodenal Tmax (h) and as a function of (B) jejunal Tmax. Fasted and fed state results are depicted by the blue and orange dots, respectively. Trendlines for both graphs
24 are given by the black line and expressed by the slope and intercept. The Pearson Coefficient of Determination is expressed as R2. Standard errors of slope and intercept, respectively, of the linear regression are
25 indicated in parenthesis. Regression for both plots was significant with p < 0.05 (Analysis of Variance, ANOVA).
26
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42 16
43
44
45
46 ACS Paragon Plus Environment
47
48
Page 17 of 37 Molecular Pharmaceutics

1
2
3 Figure 3A and Figure 3B clearly illustrate the link between the time of maximal concentrations of
4
5 ibuprofen in solution (dissolved) appearing in the intestine and in blood. Ibuprofen is a weak acid
6
7 (pKa ~4.85) and dissolution of ibuprofen is favored at pH > 4.85, while very low intrinsic solubility
8
9 concentrations are expected in the stomach in fasting state conditions (pH 1-2). The pH is highly
10
11 variable and fluctuating along the GI tract3,8,22,45,46, and this will have a major influence on the
12
13 dissolution of the drug.12,47 Figure 4A and Figure 4B shows the pH values as measured in this study at
14
15 the time of the duodenal/jejunal Tmax appeared in both fasted and fed state conditions, respectively,
16
17 with the red horizontal line approximating the pKa of ibuprofen.
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 17
ACS Paragon Plus Environment
Molecular Pharmaceutics Page 18 of 37

1
2
3
4
Fasted State Fed State
5A B
6
7 8 8
8 7 7
9
6 6
10
11 5 5
12

pH
pH

4 4
13
14 3 3
15 2 2
16
1 1
17
18 0 0
19
20
21
22 pH at Duodenal Tmax pH at Jejunal Tmax pH at Duodenal Tmax pH at Jejunal Tmax
23
24
25 Figure 4: (A) pH values as measured at the time Tmax of ibuprofen appeared in duodenum (blue bars) and jejunum (yellow bars) in fasted state conditions. (B) pH values as measured at the time Tmax of ibuprofen
26 appeared in duodenum (blue bars) and jejunum (yellow bars) in fed state conditions. The red line indicates the pKa value of ibuprofen (pKa ~4.85). F1 stands for ‘Fasted State – Visit #1’; F2 stands for ‘Fasted State –
Visit #2’; P1 stands for ‘Fed State – Visit #1’; P2 stands for ‘Fed State – Visit #2’.
27
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42 18
43
44
45
46 ACS Paragon Plus Environment
47
48
Page 19 of 37 Molecular Pharmaceutics

1
2
3 The pH values measured at the time of appearance of maximal solution concentrations of
4
5 ibuprofen in the duodenum/jejunum were higher than the pKa of the drug (~4.85) and favorable for
6
7 dissolution of ibuprofen at the duodenal/jejunal Tmax. This was observed for the majority of the
8
9 subjects (30 out of 36 subjects). Furthermore, based on the individual plots (data not shown), it was
10
11 indeed shown that at the time when the plasma Cmax appeared, solution concentrations in the intestinal
12
13 tract were associated with pH values higher than the pKa of ibuprofen, sufficient to ensure complete
14
15 dissolution of ibuprofen (i.e. solution and total concentrations are equal) for almost all subjects (10 out
16
17 of 11 for the fasted state; 7 out of 11 for the fed state). Remarkably, whenever the first wave of phase
18
19 III contractions was observed after administration of the tablet, the appearance of the plasma Cmax was
20
21 on average 1 h later in fasting state conditions: the strong burst of phase III contractions, often referred
22
23 to as ‘the housekeeper wave’48,49, would likely stimulate gastric emptying of ibuprofen such that large
24
25 quantities of ibuprofen enter the small intestine and dissolve. If this arrival in the small intestine is
26
27 accompanied with a sufficient pH to promote ibuprofen’s dissolution (as observed in Figure 4), we
28
29 suggest that motility and pH are the major determinants for the appearance of plasma Cmax and Tmax of
30
31 the drug.
32
33
Based on research reports, there is a clear link observed between the rate of duodenal
34
35
secretions (i.e. bile, pancreatic enzymes and bicarbonate) and the appearance of the interdigestive
36
37
cyclic motility phases of the MMC: pancreatic enzyme and bile secretion peak in the late phase II
38
39
contractions (emptying of approximately 25% of gallbladder contents), whereas gastric acid and
40
41
42 bicarbonate secretion into the duodenum peak during the initiation of phase III contractions.19,50–52
43
44 This link between GI motility and secretions has been defined as the ‘secretomotor complex’, which
45
46 appears to be regulated by hormonal (e.g. motilin and pancreatic peptide) and neural (e.g. vagus nerve
47
48 stimulation mediated by acetylcholine) stimuli.53,54 As confirmed by Vantrappen and colleagues, a rise
49
50 in duodenal pH in humans was observed following the start of duodenal phase III contractions, which
51
52 would be expected to be reflected in the pH values we observed (> 4.85) at the time of appearance of
53
54 the duodenal/jejunal Tmax (Figure 4).17
55
56 In this study, the pH along the GI tract was highly fluctuating as a function of time and
57
58 ibuprofen was present in the GI tract up to 7 hours based on aspiration of luminal samples. Figure 5
59
60 19
ACS Paragon Plus Environment
Molecular Pharmaceutics Page 20 of 37

1
2
3 shows the average pH profiles as a function of time for the different GI segments in fasted and fed
4
5 state conditions.
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
ACS Paragon Plus Environment
Page 21 of 37 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 Figure 5: Mean pH versus time profiles in fasting (n=20) and fed state (n=17) conditions as measured in the stomach, the duodenum and the jejunum (mean + SD). Data obtained from Koenigsknecht et al.15
35
36
37
38
39
40
41
42 21
43
44
45
46 ACS Paragon Plus Environment
47
48
Molecular Pharmaceutics Page 22 of 37

1
2
3 Gastric pH was observed to be highly variable in fasted (range 1.1-7.47) and fed (range 1.1-
4
5 7.39) states. The average and median pH observed in fasted state were 2.50 and 2.25, respectively. The
6
7 average and median pH in fed state were 4.04 and 3.95, respectively. Ingestion of the liquid meal (pH
8
9 6.66) resulted in an initial increase in gastric pH, which slowly decreased as a function of time
10
11 returning to fasted state conditions.15 Both the average and pH range were in line as observed in other
12
13 studies where gastric pH was measured after aspiration of gastric fluids in healthy volunteers.14,55,56
14
15 Duodenogastric reflux and/or saliva swallowed by the volunteer may contribute to higher gastric pH
16
17 values in fasted state conditions, as discussed in the literature and as observed in this study.14
18
19
20 Duodenal pH values ranged from 1.71 to 7.57 in fasted state, with a mean value of 4.93 and a
21
22 median value of 4.91. Jejunal pH values ranged from 2.2 to 6.75 in fasted state, with a mean value of
23
24 5.55 and a median value of 5.62. Less variability in pH was observed in the jejunal segment compared
25
26 to the duodenum. As the main function of the duodenum is alkalizing and mixing the gastric content
27
28 with pancreatic secretions (e.g. bicarbonate, amylase, lipase, trypsinogen), variability in pH would be
29
30 more expected in this part of the intestine compared to the more distal segments (i.e. jejunum and
31
32 ileum).57 Recently published data demonstrated that pH values for aspirated duodenal fluids as a
33
34 function of time in a wide range from 3.4 to 8.3 and 4.7 to 7.1 in fasted and fed state, respectively.45
35
36 Regarding fed state conditions, duodenal pH values ranged from 1.27 to 7.21 (mean: 5.26; median:
37
38 5.35), whereas jejunal pH values ranged from 4.46 to 7.77 (mean: 6.04; median: 6.10).
39
40
41 The pH along the GI tract is a dynamic physiological variable. The wide range of pH that has
42
43 been measured in all volunteers can significantly affect drug dissolution of ibuprofen along the
44
45 intestinal tract and thus the amount of drug in solution and available for absorption. Figure 6 depicts
46
47 the average profiles of solution concentrations of ibuprofen, total concentrations and pH as a function
48
49 of time in fasted state duodenum (A) and jejunum (B), as well as in fed state duodenum (C) and
50
51 jejunum (D). Moreover, the average time of appearance of post-dose phase III contractions (2.02 and
52
53 4.64 h in fasted and fed state, respectively) is indicated by the green line, while the average plasma
54
55 Tmax (2.98 and 4.88 h in fasted and fed state, respectively) is indicated by the dark blue line.
56
57
58
59
60 22
ACS Paragon Plus Environment
Page 23 of 37 Molecular Pharmaceutics

1 Fasted State Duodenum Fasted State Jejunum


2
3 3000000 8 3000000 8

4 2000000 2000000
5 1000000 6 1000000 6

Conc IBU (ng/mL)

Conc IBU (ng/mL)


6
7 600000 600000

pH

pH
4 4
8
400000 400000
9
2 2
10 200000 200000
11
12 0 0 0 0
13 0 1 2 3 4 5 6 7 8 0 1 2 3 4 5 6 7 8
Time (h) Time (h)
14
15 Total Conc Duodenum Post-dose Phase III Contr Total Conc Jejunum Post-dose Phase III Contr
16 pH Duodenum pH Jejunum
17 Solution Conc Duodenum
Plasma Tmax
Solution Conc Jejunum
Plasma Tmax
18
19
20 Fed State Duodenum Fed State Jejunum
21 8 8
1200000 1200000
22
23 800000 6 800000 6
Conc IBU (ng/mL)

Conc IBU (ng/mL)


24 300000
25 600000

pH
26
4 pH 4
200000
27 400000

28 2 2
100000
200000
29
30 0 0 0 0
31 0 1 2 3 4 5 6 7 8 0 1 2 3 4 5 6 7 8

32 Time (h) Time (h)

33 Total Conc Duodenum Total Conc Jejunum


Post-dose Phase III Contr Post-dose Phase III Contr
34 pH Jejunum
pH Duodenum
35 Plasma Tmax Solution Conc Jejunum Plasma Tmax
Solution Conc Duodenum
36
37
Figure 6: Average solution concentrations of ibuprofen (IBU) (blue line), total concentrations of IBU (gray area) and pH profiles (red line) of all subjects as measured in fasted state duodenum (A), fasted state jejunum
38 (B), fed state duodenum (C) and fed state jejunum (D). The green line indicates the average time when phase III contractions occurred after intake of the tablet. The dark blue line represents the average plasma Tmax.
39 Data presented as mean + SD.
40
41
42 23
43
44
45
46 ACS Paragon Plus Environment
47
48
Molecular Pharmaceutics Page 24 of 37

1
2
3 Based on the average and individual profiles, the present pH determined the fraction dissolved
4
5 of ibuprofen along the intestinal tract, indicated by the same profile pattern between solution
6
7 concentrations and pH in fasted state (Figure 6A and 6B). In postprandial conditions, duodenal pH
8
9 (mean pH 5.26) was decreasing as a function of time, resulting in more solid ibuprofen in the collected
10
11 samples in the last three hours of aspiration (Figure 6C), whereas jejunal pH (mean pH 6.04) remained
12
13 relatively constant as a function of time, resulting in dissolution of ibuprofen along the jejunal tract
14
15 (Figure 6D). Gastric emptying of ibuprofen will likely be slowed down in postprandial conditions,
16
17 which is observed in the delayed plasma Tmax in fed state conditions compared to fasted state
18
19 conditions (2.97 h versus 4.88 h, respectively). This is due to the later maximal concentrations of
20
21 ibuprofen in the intestinal tract (Figure 3; orange dots). Further, as depicted in Figure 7C and 7D, the
22
23 onset of phase III contractions was delayed in fed state conditions relative to the fasted state,
24
25 indicating a slow release of ibuprofen from the stomach to the small intestine. The plasma Cmax values
26
27 of all volunteers are depicted as a function of the post-dose appearance of phase III contractions in
28
29 Figure 7.
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 24
ACS Paragon Plus Environment
Page 25 of 37 Molecular Pharmaceutics

1
2
3
4
5 100000
6
7 90000
8 y = - 6713.467 + 69802.07
80000
9

Plasma Cmax (ng/mL)


(1702.21 – 6298.96)
10 70000
11 R² = 0.3204
60000
12
50000
13
14 40000
15 30000
16
17 20000
18 10000
19
0
20 0 1 2 3 4 5 6 7 8
21
Time to Phase III contractions post-dose (h)
22
23
Fasted State Fed State
24
25
26 Figure 7: Plot of plasma Cmax (h) as a function of time of appearance of phase III contractions after oral intake of ibuprofen. Fasted and fed state results are depicted by the blue and orange dots, respectively. The
trendline is given by the black line and the Pearson Coefficient of Determination is expressed as R2. Standard errors of slope and intercept, respectively, of the linear regression are indicated in parenthesis. Regression
27 was significant with p < 0.05 (Analysis of Variance, ANOVA).
28
29
30
31
32
33
34
35
36
37
38
39
40
41
42 25
43
44
45
46 ACS Paragon Plus Environment
47
48
Molecular Pharmaceutics Page 26 of 37

1
2
3 The delayed appearance of phase III contractions in fed state conditions was positively linked
4
5 to a decreased systemic exposure of ibuprofen (in terms of Cmax), which clearly illustrates the effect of
6
7 motility on systemic drug exposure. It should be noted that subject B041 - visit #1 in fed state
8
9 conditions, ingested only 7 mL of Pulmocare® and, therefore, demonstrated a fast onset of phase III
10
11 contractions, occurring in one hour after intake of the tablet. The time to onset of phase III
12
13 contractions is associated with the rate of gastric emptying of the liquid meal which, in turn, will
14
15 depend on the caloric intake of the meal. Moreover, the amplitude and origin (i.e. antral versus
16
17 duodenal) of these contractions observed in fasted and fed state may reveal how they affect
18
19 formulation disintegration and drug release along the GI tract and is also subject of interest for future
20
21 research.
22
23
24 Where most in vitro predictive dissolution tests focus on a constant pH and high buffer
25
26 capacity during dissolution testing, it should be noted that this is not an adequate reflection of the in
27
28 vivo situation. In the case of ionized drugs, depending on pKa, the fluctuating pH along the intestinal
29
30 tract is one of the major causes in explaining inter- and intra-individual variability in drug exposure
31
32 since it affects the dissolved fraction of a drug. The changing intestinal pH is determined by the buffer
33
34 capacity of these fluids. Figure 8 depicts buffer capacity of the aspirated GI fluids as a function of time
35
36 in fasted (A) and fed (B) state.
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60 26
ACS Paragon Plus Environment
Page 27 of 37 Molecular Pharmaceutics

1
2
3
4
5
Fasted State Fed State
6
7
8 A 12 B 12
Buffer Capacity (µmol/mL/∆pH)

Buffer Capacity (µmol/mL/∆pH)


10 12 10
10

Buffer Capacity (µmol/mL/∆pH)


11 10
8 8
12
13 6 8
6
14
15 4 6
4
16
2 4
17 2
18 2
0 0
19 0 1 2 3 4 5 6 7 8
20 0 0 1 2 3 4 5 6 7 8
Time (h) Time (h)
21 0 1 2 3 4 5 6 7 8
22 Time (h)
23
24
Buffer Capacity Stomach Buffer Capacity Duodenum Buffer Capacity Jejunum
25
26
27
28
29 Figure 8: Buffer capacity values of the aspirated fluids in (A) fasting state conditions and (B) fed state conditions as a function of time (h). The orange line represents buffer capacity data of the aspirated gastric fluids;
30 the yellow line represents buffer capacity data of the aspirated duodenal fluids; the blue line represents buffer capacity data of the aspirated jejunal fluids. Data presented as mean + SD.
31
32
33
34
35
36
37
38
39
40
41
42 27
43
44
45
46 ACS Paragon Plus Environment
47
48
Molecular Pharmaceutics Page 28 of 37

1
2
3 Surprisingly, the buffer capacity of the luminal fluid along the entire GI tract is extremely low.
4
5 This is likely the reason why ibuprofen was still present in the luminal samples throughout the entire 7
6
7 hours of aspiration, as a dynamic pH was observed that will determine the fraction dissolved (Figure
8
9 6). Only a few publications in the literature report buffer capacity of GI fluids. To the extent of our
10
11 knowledge, buffer capacity has never been measured for GI fluids at several sites in the upper GI tract
12
13 based on aspirated samples as a function of time; usually this was only measured for pooled samples.
14
15 The measured buffer capacity in the intestinal fluids derived from this study is in line with reported
16
17 results in literature: Fadda et al. reported an average of 3.23 µmol/mL/∆pH for aspirated fasted state
18
19 jejunal fluids, whereas Persson and colleagues measured a buffer capacity of 2.8 µmol/mL/∆pH for
20
21 fasted human intestinal fluids (HIF).36,58 In fed HIF, Persson et al. reported a buffer capacity of 14.6
22
23 µmol/mL/∆pH (single value), remarkably higher than the average buffer capacity measured in this
24
25 study for all fed state aspirates in the duodenum and jejunum (2.16 µmol/mL/∆pH and 1.81
26
27 µmol/mL/∆pH, respectively).58 Based on the two plots (Figure 8A and 8B), values in fed state were on
28
29 average slightly higher than fasted state values as the intake of the liquid meal will stimulate
30
31 pancreatic secretions (e.g. amylase, lipase, and bicarbonate secretion) to facilitate food digestion.57
32
33
The buffer capacity of gastric fluids is reported to range between 7-18 µmol/mL/∆pH after intake of
34
35
250 mL of water14 and 4.7-27.6 µmol/mL/∆pH after intake of 240 mL of water.59 When a liquid meal
36
37
(Ensure Plus®) was administered, a rise in gastric buffer capacity was observed: reported values
38
39
between 14-28 µmol/mL/∆pH14 and 22.5-30.0 µmol/mL/∆pH60, which are higher than observed in our
40
41
42 clinical study (ranging between 2.33 and 5.98 µmol/mL/∆pH on average for all subjects). This
43
44 observation is likely due to differences in administered volumes/types of liquid meals between studies.
45
46 In general, the measured buffer capacity values are extremely low on average and five times
47
48 lower compared to the buffer capacity of simulated intestinal fluids, frequently applied in dissolution
49
50 testing. For example, FaSSIF (pH 6.5) and USP SIF (pH 6.8) both demonstrate retaining a strong
51
52
buffer capacity of 12 and 18.4 µmol/mL/∆pH, which is not suitable for in vitro predictive BE
53
54
dissolution testing, maintaining a constant pH value during the entire dissolution experiment.
55
56
Furthermore, besides the low buffer capacity of human intestinal fluids, differences in secretion rates
57
58
59
60 28
ACS Paragon Plus Environment
Page 29 of 37 Molecular Pharmaceutics

1
2
3 between H+ (approximately 17 mmol/h)61 from the parietal cells in the stomach and HCO3- (8
4
5 mmol/h)62 from the pancreas and mucosal cells in the duodenum, will likely contribute to the observed
6
7 pH fluctuation in the upper small intestine. Moreover, the low luminal concentrations of bicarbonate in
8
9 combination with the limited available duodenal fluid volume (i.e. less than 50 mL free fluid available
10
11 for drug dissolution), is likely to be an additional reason for this dynamic change in pH.24 It should be
12
13 noted that the length and intensity of motor contractions, e.g. phase III, and secretory rates are variable
14
15 between and within humans due to the time of the day, time of dosing, and the length of time after
16
17 food ingestion.50
18
19
20 Therefore, the important role of buffer capacity needs to be reflected in preclinical in vitro/in
21
22 silico models in order to make a good prediction of the systemic appearance of an oral drug product.
23
24 This in vivo predictive dissolution method would likely be more complex than the current QC
25
26 methods. The iPD method would be useful for developing optimized formulations and it may be more
27
28 appropriately to refer to it as formulation predictive dissolution (fPD) testing. This method would be
29
30 able to discriminate between inter- and intra-individual differences in systemic exposure of a drug
31
32 product and would be extremely useful for oral drug product development and optimization. The use
33
34 of more complex, multicompartmental models (e.g. gastrointestinal simulator (GIS)63, artificial
35
36 stomach-duodenum (ASD) model64 and the two-phase dissolution model65,66) attempt to integrate the
37
38 variables of the dynamic GI environment, capturing the critical formulation and product variables,
39
40 such as gastric emptying, intestinal secretions and the potential for drug absorption, resulting in a
41
42 better understanding of formulation and/or drug behavior for certain classes of drugs and/or
43
44 formulations. While much public policy debate is required, these fPD models may eventually be useful
45
46 in BE determinations (e.g. BCS biowaiver extensions) and/or may lead to a reduction in the number of
47
48 human subjects required for in vivo BE trials and minimize the number of failed BE studies.47
49
50
51 Neutralizing Effect of Bicarbonate on Gastric Acid and the Effect of Ibuprofen Dissolution on pH
52
53 Measured bicarbonate concentrations in the upper small intestine have been reported to be 4-
54
55 21 mM with an average of 15 mM.16,21 If we assume that gastric fluids (pH 2=0.01 M HCl) would
56
57 empty the stomach as first kinetic order process (Equation 1), the maximum concentration of protons
58
59
60 29
ACS Paragon Plus Environment
Molecular Pharmaceutics Page 30 of 37

1
2
3 (H+) that would enter the small intestine is 3.4 mM. As this concentration would interact with the
4
5 present bicarbonate (a minimum of 4 mM and a maximum of 21 mM), a measured intestinal pH of
6
7 5.46 and 6.21 would be observed for both situations (Equation 2). Regarding the dynamic environment
8
9 of the GI tract, concentrations of HCl entering the duodenum and concentrations of bicarbonate
10
11 present in the duodenum may vary, resulting in fluctuations of pH levels up to 5 units which would not
12
13 be uncommon in the upper part of the intestinal tract.61
14
15
16 In addition, the dissolution of an acidic or basic drug in these fluids would be expected to alter
17
18 the pH of the intestinal fluids. Based on the measured solution concentrations of ibuprofen in the
19
20 duodenum, a drop in pH of approximately two units can be estimated due to the drug (Equation 3) in
21
22 an aqueous environment with a negligible buffer capacity (Figure 9).
23
24
25 12
26 10
27
28 8
29
pH

6
30
31 4
32 2
33
34 0
0 1 2 3 4 5 6 7 8
35
36 Time (h)
37
38 Figure 9: Impact of drug dissolution of ibuprofen on pH in an aqueous environment with a negligible buffer capacity. Data presented as
39 mean + SD.
40
41
42
43 A similar drop in pH was observed when a dissolution experiment was performed for one
44
45 tablet of ibuprofen (400 mg) in 500 mL of 10 mM phosphate buffer dissolution media at a pH of 6.0
46
47 and 6.8 (buffer capacity 1.6-2.8 µmol/mL/∆pH).12 Dissolution of ibuprofen resulted in a drop in pH
48
49 from 6.0 to 5.1 and from 6.8 to 6.1, respectively, not as outspoken as observed in our study. Based on
50
51 the administered dose, the drop in pH along the intestinal tract may have a significant effect on drug’s
52
53 dissolution. Performing standard QC dissolution methods didn’t seem to affect the in silico systemic
54
55 outcome of lesinurad (weak acid, pKa 3.2) for different batches of the drug, differing in particle size.67
56
57 However, selection of the right buffer and applying biorelevant buffer concentrations is of utmost
58
59
60 30
ACS Paragon Plus Environment
Page 31 of 37 Molecular Pharmaceutics

1
2
3 importance in developing a predictive dissolution test to link GI variability with the systemic exposure
4
5 of the drug, which could explain the possible failures in BE studies. It should be noted that the
6
7 frequently applied phosphate (pKa 7.2) buffer (e.g. FaSSIF and USP SIF) is not a biorelevant
8
9 reflection of the bicarbonate (pKa 6.35) buffer as observed in vivo. Differences in drug release
10
11 behavior have been reported for both buffers during in vitro dissolution experiments.27 Further, the
12
13 dynamic change in buffer capacity along the intestinal tract is due to the CO2 concentrations in the GI
14
15 fluids, both in solution (CO2(aq)) and the luminal gas phase (CO2(g)) as these concentrations directly
16
17 determine the formation of bicarbonate. The concentration of CO2 dissolved in water follows Henry’s
18
19 Law constant (KH) and the partial pressure of gaseous CO2 (i.e. J,-1 :
20
21
22 
6 ⇌ 
KL (Reaction 3)
23
24
(,-1 'N +
25 &)M OPQ1
(Reaction 4)
26
27
28 The formation of bicarbonate can be produced by sparging gaseous CO2 equilibrated with
29
30 water. At different pH values, the formation of bicarbonate will differ and so will the buffer capacity,
31
32 as experimentally demonstrated by Krieg and colleagues (Table 2).25
33
34
35 Table 2: Effect of sparged CO2 on the formation of bicarbonate in water at different pH values. Data derived from Krieg et al.25
36
37 Bicarbonate Buffer Concentration (mM)
38
39 Buffer Capacity in Parentheses (µmol/mL/∆pH)
40
41 %CO2
42
43 pH 5% 10% 15% 20% 40% 60%
44
45
5.5 0.19 0.38 0.57 0.76 1.52 2.29 (0.61)
46
47
6 0.6 1.20 1.81 2.41 4.82 7.23
48
49
50 6.5 1.9 3.81 (2.10) 5.71 (3.10) 7.62 (4.20) 15.23 22.85
51
52 7 6.02 12.04 18.07 24.10 48.17 72.26
53
54 7.5 19.04 (2.50) 38.08 57.13 76.17 152.34 228.51
55
56
57
58
59
60 31
ACS Paragon Plus Environment
Molecular Pharmaceutics Page 32 of 37

1
2
3 At lower pH values (pH 5.5), the equilibrium solubility of CO2 is too low, even at high partial
4
5 pressures, to form meaningful bicarbonate buffer concentrations. Under atmospheric pressure
6
7 conditions, the percentage of CO2 is equal to 400 ppm (0.04% ~ 0.304 mmHg). In the stomach of
8
9 healthy volunteers, measured luminal percentages of CO2 ranged between 4-10%. In the proximal
10
11 duodenum, this value was significantly higher: 66%.68,69 This means that the partial pressure of CO2 in
12
13 the upper small intestine can be 1650 times higher than the partial pressure measured in the
14
15 atmosphere likely explained by the formed CO2 after the neutralizing reaction of HCl and bicarbonate
16
17 (Reaction 1). As derived from Table 2, a maximal concentration of bicarbonate (and thus a maximal
18
19 buffer capacity) can be obtained at a neutral pH (pH 7-7.5) along the intestinal tract. As CO2 (g) may
20
21 evaporate after intestinal aspiration, it should be noted that values mentioned in literature for aspirates
22
23 may underestimate the effective buffer capacity. However, as reported by Fadda et al., buffer capacity
24
25 measurements immediately performed on fluid collection and after storage did not significantly
26
27 differ.36 It can be stated that the formation of bicarbonate along the intestinal tract is a dynamic and
28
29 complex phenomenon that depends on the rate of bicarbonate secretion (secretomotor complex), the
30
31 circulating CO2 (aq)/ CO2 (g) along the GI tract and the dehydration reaction of H2CO3 which is
32
33
formed by the reaction of H+ and HCO3- itself.
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 32
ACS Paragon Plus Environment
Page 33 of 37 Molecular Pharmaceutics

1
2
3 Conclusion and Future Directions
4
5
6 The fluctuating pH and buffer capacity along the GI tract have been shown to be a major
7
8 determinant of inter- and intra-subject variability in systemic exposure of ibuprofen. The pH presented
9
10 in the different segments of the intestinal tract determine the solution concentrations, total
11
12 concentrations and absorption of ibuprofen from the intestinal lumen. This study clearly demonstrated
13
14 the link between the plasma Tmax and duodenal/jejunal Tmax of ibuprofen after the administration of the
15
16 800 mg ibuprofen tablet to healthy volunteers. At the time these maximal duodenal/jejunal
17
18 concentrations of ibuprofen were reached, pH values in these aspirated samples were higher than the
19
20 pKa value of ibuprofen, supporting the dissolution of the drug. The dynamic change in pH along the
21
22 GI tract was explained by the limited buffer capacity of the aspirated fluids. The plasma Cmax was, in
23
24 turn, positively linked with the time to appearance of the first phase III contractions after oral
25
26 administration of ibuprofen. These results demonstrate how the dynamic pH, explained by the limited
27
28 buffer capacity, and the time of drug intake relative to the cyclic motility pattern will result in not only
29
30 inter-subject variability but also intra-subject variability in systemic availability (Cmax and Tmax) for a
31
32 BCS class 2a drug.70
33
34
35 Based on the results of this study, we expect that this very low buffer capacity and dynamic
36
37 pH are also essential and determining for the absorption of basic drugs, where the levels of
38
39 supersaturation and rates of precipitation will be affected by the dynamic change of pH and buffer
40
41 capacity along the intestinal tract.71 Future in vivo studies should focus on how (i) the amplitude of
42
43 contractions, (ii) the origin of contractions (i.e. antral versus duodenal), (iii) the gastric emptying rate
44
45 (linked to the ingested amount of calories) and (iv) intestinal transit times (based on concentrations of
46
47 a non-absorbable marker), may account for variability in systemic outcome of the administered drug
48
49 product. Finally, based on the results of this study, adjustments to the GIS model (e.g. use of
50
51 bicarbonate buffer) will be performed in order to adequately predict the in vivo outcome of oral drug
52
53 absorption and to capture the GI and plasma variability as observed in the clinical study.
54
55
56
57
58
59
60 33
ACS Paragon Plus Environment
Molecular Pharmaceutics Page 34 of 37

1
2
3 Acknowledgments
4 This work was supported by grant # HHSF223201510157C and grant # HHSF223201310144C by the
5
6 U.S. Food and Drug Administration (FDA). This report represents the scientific views of the authors
7
8 and not necessarily that of the FDA.
9
10
11 References
12 (1) Food & Drug Administration Waiver of In Vivo Bioavailability and Bioequivalence Studies for
13 Immediate-Release Solid Oral Dosage Forms Based on a Biopharmaceutics Classification System
14 Guidance for Industry 2015.
15 (2) Food & Drug Administration Guidance for Industry Bioequivalence Studies with Pharmacokinetic
16 Endpoints for Drugs Submitted Under an ANDA 2013.
17 (3) Hens, B.; Corsetti, M.; Spiller, R.; Marciani, L.; Vanuytsel, T.; Tack, J.; Talattof, A.; Amidon, G. L.;
18 Koziolek, M.; Weitschies, W.; Wilson, C. G.; Bennink, R. J.; Brouwers, J.; Augustijns, P. Exploring
19 Gastrointestinal Variables Affecting Drug and Formulation Behavior: Methodologies, Challenges and
20 Opportunities. Int J Pharm 2016, 59, 79–97.
21 (4) Talattof, A.; Price, J. C.; Amidon, G. L. Gastrointestinal Motility Variation and Implications for
22 Plasma Level Variation: Oral Drug Products. Mol. Pharm. 2016, 13, 557–567.
23 (5) Deloose, E.; Janssen, P.; Depoortere, I.; Tack, J. The migrating motor complex: control mechanisms
24 and its role in health and disease. Nat Rev Gastroenterol Hepatol 2012, 9, 271–285.
25 (6) Van Den Abeele, J.; Rubbens, J.; Brouwers, J.; Augustijns, P. The dynamic gastric environment and
26 its impact on drug and formulation behaviour. Eur J Pharm Sci 2017, 96, 207–231.
27 (7) Heading, R. C.; Nimmo, J.; Prescott, L. F.; Tothill, P. The dependence of paracetamol absorption on
28
the rate of gastric emptying. Br. J. Pharmacol. 1973, 47, 415–421.
29
(8) Fuchs, A.; Dressman, J. B. Composition and Physicochemical Properties of Fasted-State Human
30
Duodenal and Jejunal Fluid: A Critical Evaluation of the Available Data. J. Pharm. Sci. 2014, 103,
31
3398–3411.
32
(9) McNamara, D. P.; Whitney, K. M.; Goss, S. L. Use of a physiologic bicarbonate buffer system for
33
dissolution characterization of ionizable drugs. Pharmaceutical Research 2003, 20, 1641–6.
34
35
(10) Mooney, K. G.; Mintun, M. A.; Himmelstein, K. J.; Stella, V. J. Dissolution kinetics of carboxylic
36 acids I: effect of pH under unbuffered conditions. J Pharm Sci 1981, 70, 13–22.
37 (11) Mooney, K. G.; Mintun, M. A.; Himmelstein, K. J.; Stella, V. J. Dissolution kinetics of carboxylic
38 acids II: effect of buffers. J Pharm Sci 1981, 70, 22–32.
39 (12) Tsume, Y.; Langguth, P.; Garcia-Arieta, A.; Amidon, G. L. In silico prediction of drug dissolution and
40 absorption with variation in intestinal pH for BCS class II weak acid drugs: ibuprofen and ketoprofen.
41 Biopharm Drug Dispos 2012, 33, 366–377.
42 (13) Fillet, M.; Bechet, I.; Piette, V.; Crommen, J. Separation of nonsteroidal anti-inflammatory drugs by
43 capillary electrophoresis using nonaqueous electrolytes. Electrophoresis 1999, 20, 1907–1915.
44 (14) Kalantzi, L.; Goumas, K.; Kalioras, V.; Abrahamsson, B.; Dressman, J. B.; Reppas, C.
45 Characterization of the human upper gastrointestinal contents under conditions simulating
46 bioavailability/bioequivalence studies. Pharm. Res. 2006, 23, 165–176.
47 (15) Koenigsknecht, M.; Jason, B.; Fioritto, A.; Yu, A.; Tsume, Y.; Wen, B.; Zhao, T.; Pai, M.; Bleske, B.;
48 Zhang, X.; Lionberger, R.; Lee, A.; Amidon, G.; Hasler, W.; Sun, D. In Vivo Dissolution and
49 Systemic Absorption of Immediate Release Ibuprofen in Human Gastrointestinal Tract Under Fed and
50 Fasted Conditions. Mol. Pharm. In Submission.
51 (16) Sheng, J. J.; McNamara, D. P.; Amidon, G. L. Toward an in vivo dissolution methodology: a
52 comparison of phosphate and bicarbonate buffers. Mol. Pharm. 2009, 6, 29–39.
53 (17) Vantrappen, G. R.; Peeters, T. L.; Janssens, J. The Secretory Component of the Interdigestive
54 Migrating Motor Complex in Man. Scandinavian Journal of Gastroenterology 1979, 14, 663–667.
55 (18) Vantrappen, G.; Janssens, J.; Hellemans, J.; Ghoos, Y. The interdigestive motor complex of normal
56 subjects and patients with bacterial overgrowth of the small intestine. J. Clin. Invest. 1977, 59, 1158–
57 1166.
58
59
60 34
ACS Paragon Plus Environment
Page 35 of 37 Molecular Pharmaceutics

1
2
3 (19) Sjövall, H.; Hagman, I.; Abrahamsson, H. Relationship between interdigestive duodenal motility and
4 fluid transport in humans. Am. J. Physiol. 1990, 259, G348-354.
5 (20) Dooley, C. P.; Di Lorenzo, C.; Valenzuela, J. E. Variability of migrating motor complex in humans.
6 Dig. Dis. Sci. 1992, 37, 723–728.
7 (21) Karr, W. G.; Abbott, W. O.; Sample, A. B. INTUBATION STUDIES OF THE HUMAN SMALL
8 INTESTINE. IV. CHEMICAL CHARACTERISTICS OF THE INTESTINAL CONTENTS IN THE
9 FASTING STATE AND AS INFLUENCED BY THE ADMINISTRATION OF ACIDS, OF
10 ALKALIES AND OF WATER. J. Clin. Invest. 1935, 14, 893–900.
11 (22) Perez de la Cruz Moreno, M.; Oth, M.; Deferme, S.; Lammert, F.; Tack, J.; Dressman, J.; Augustijns,
12 P. Characterization of fasted-state human intestinal fluids collected from duodenum and jejunum. J.
13 Pharm. Pharmacol. 2006, 58, 1079–1089.
14 (23) Jantratid, E.; Janssen, N.; Reppas, C.; Dressman, J. B. Dissolution media simulating conditions in the
15 proximal human gastrointestinal tract: an update. Pharm. Res. 2008, 25, 1663–1676.
16 (24) Mudie, D. M.; Murray, K.; Hoad, C. L.; Pritchard, S. E.; Garnett, M. C.; Amidon, G. L.; Gowland, P.
17 A.; Spiller, R. C.; Amidon, G. E.; Marciani, L. Quantification of gastrointestinal liquid volumes and
18 distribution following a 240 mL dose of water in the fasted state. Mol. Pharm. 2014, 11, 3039–3047.
19 (25) Krieg, B. J.; Taghavi, S. M.; Amidon, G. L.; Amidon, G. E. In vivo predictive dissolution: transport
20 analysis of the CO2 , bicarbonate in vivo buffer system. J Pharm Sci 2014, 103, 3473–3490.
21 (26) Fadda, H. M.; Merchant, H. A.; Arafat, B. T.; Basit, A. W. Physiological bicarbonate buffers:
22 stabilisation and use as dissolution media for modified release systems. International Journal of
23 Pharmaceutics 2009, 382, 56–60.
24 (27) Garbacz, G.; Kołodziej, B.; Koziolek, M.; Weitschies, W.; Klein, S. A dynamic system for the
25 simulation of fasting luminal pH-gradients using hydrogen carbonate buffers for dissolution testing of
26 ionisable compounds. European Journal of Pharmaceutical Sciences 2014, 51, 224–231.
27 (28) Food & Drug Administration Referencing Approved Drug Products in ANDA Submissions: Guidance
28 for Industry 2017.
29 (29) Food & Drug Administration Guidance for Industry: Bioanalytical Method Validation 2001.
30 (30) Marciani, L.; Gowland, P. A.; Fillery-Travis, A.; Manoj, P.; Wright, J.; Smith, A.; Young, P.; Moore,
31 R.; Spiller, R. C. Assessment of antral grinding of a model solid meal with echo-planar imaging. Am.
32 J. Physiol. Gastrointest. Liver Physiol. 2001, 280, G844-849.
33
(31) Marciani, L.; Gowland, P. A.; Spiller, R. C.; Manoj, P.; Moore, R. J.; Young, P.; Al-Sahab, S.; Bush,
34
D.; Wright, J.; Fillery-Travis, A. J. Gastric response to increased meal viscosity assessed by echo-
35
planar magnetic resonance imaging in humans. J. Nutr. 2000, 130, 122–127.
36
(32) Shumway, R. H.; Stoffer, D. S. Time Series Analysis and Its Applications - With R Examples; Springer
37
Science & Business Media, 2010.
38
(33) Tomomasa, T.; Kuroume, T.; Arai, H.; Wakabayashi, K.; Itoh, Z. Erythromycin induces migrating
39
motor complex in human gastrointestinal tract. Dig. Dis. Sci. 1986, 31, 157–161.
40
41 (34) Sarna, S. K.; Otterson, M. F. Small intestinal physiology and pathophysiology. Gastroenterol. Clin.
42 North Am. 1989, 18, 375–404.
43 (35) Kerlin, P.; Zinsmeister, A.; Phillips, S. Relationship of motility to flow of contents in the human small
44 intestine. Gastroenterology 1982, 82, 701–706.
45 (36) Fadda, H. M.; Sousa, T.; Carlsson, A. S.; Abrahamsson, B.; Williams, J. G.; Kumar, D.; Basit, A. W.
46 Drug Solubility in Luminal Fluids from Different Regions of the Small and Large Intestine of
47 Humans. Mol. Pharmaceutics 2010, 7, 1527–1532.
48 (37) Van Slyke, D. D. On the Measurement of Buffer Values and on the Relationship of Buffer Value to
49 the Dissociation Constant of the Buffer and the Concentration and Reaction of the Buffer Solution. J.
50 Biol. Chem. 1922, 52, 525–570.
51 (38) Hens, B.; Brouwers, J.; Anneveld, B.; Corsetti, M.; Symillides, M.; Vertzoni, M.; Reppas, C.; Turner,
52 D. B.; Augustijns, P. Gastrointestinal transfer: in vivo evaluation and implementation in in vitro and in
53 silico predictive tools. Eur J Pharm Sci 2014, 63, 233–242.
54 (39) Culen, M.; Rezacova, A.; Jampilek, J.; Dohnal, J. Designing a dynamic dissolution method: a review
55 of instrumental options and corresponding physiology of stomach and small intestine. J Pharm Sci
56 2013, 102, 2995–3017.
57 (40) Kourentas, A.; Vertzoni, M.; Stavrinoudakis, N.; Symillidis, A.; Brouwers, J.; Augustijns, P.; Reppas,
58 C.; Symillides, M. An in vitro biorelevant gastrointestinal transfer (BioGIT) system for forecasting
59
60 35
ACS Paragon Plus Environment
Molecular Pharmaceutics Page 36 of 37

1
2
3 concentrations in the fasted upper small intestine: Design, implementation, and evaluation. Eur J
4 Pharm Sci 2016, 82, 106–114.
5 (41) Geisslinger, G.; Dietzel, K.; Bezler, H.; Nuernberg, B.; Brune, K. Therapeutically relevant differences
6 in the pharmacokinetical and pharmaceutical behavior of ibuprofen lysinate as compared to ibuprofen
7 acid. Int J Clin Pharmacol Ther Toxicol 1989, 27, 324–328.
8 (42) Kapil, R.; Nolting, A.; Roy, P.; Fiske, W.; Benedek, I.; Abramowitz, W. Pharmacokinetic properties of
9 combination oxycodone plus racemic ibuprofen: two randomized, open-label, crossover studies in
10 healthy adult volunteers. Clin Ther 2004, 26, 2015–2025.
11 (43) Tanner, T.; Aspley, S.; Munn, A.; Thomas, T. The pharmacokinetic profile of a novel fixed-dose
12 combination tablet of ibuprofen and paracetamol. BMC Clin Pharmacol 2010, 10, 10.
13 (44) Levine, M. A.; Walker, S. E.; Paton, T. W. The effect of food or sucralfate on the bioavailability of
14 S(+) and R(-) enantiomers of ibuprofen. J Clin Pharmacol 1992, 32, 1110–1114.
15 (45) Riethorst, D.; Mols, R.; Duchateau, G.; Tack, J.; Brouwers, J.; Augustijns, P. Characterization of
16 Human Duodenal Fluids in Fasted and Fed State Conditions. J Pharm Sci 2016, 105, 673–681.
17 (46) Bergström, C. A. S.; Holm, R.; Jørgensen, S. A.; Andersson, S. B. E.; Artursson, P.; Beato, S.; Borde,
18 A.; Box, K.; Brewster, M.; Dressman, J.; Feng, K.-I.; Halbert, G.; Kostewicz, E.; McAllister, M.;
19 Muenster, U.; Thinnes, J.; Taylor, R.; Mullertz, A. Early pharmaceutical profiling to predict oral drug
20 absorption: current status and unmet needs. Eur J Pharm Sci 2014, 57, 173–199.
21 (47) Yu, L. X.; Amidon, G. L.; Polli, J. E.; Zhao, H.; Mehta, M. U.; Conner, D. P.; Shah, V. P.; Lesko, L.
22 J.; Chen, M.-L.; Lee, V. H. L.; Hussain, A. S. Biopharmaceutics classification system: the scientific
23 basis for biowaiver extensions. Pharm. Res. 2002, 19, 921–925.
24 (48) Cassilly, D.; Kantor, S.; Knight, L. C.; Maurer, A. H.; Fisher, R. S.; Semler, J.; Parkman, H. P. Gastric
25 emptying of a non-digestible solid: assessment with simultaneous SmartPill pH and pressure capsule,
26 antroduodenal manometry, gastric emptying scintigraphy. Neurogastroenterology & Motility 2008, 20,
27 311–319.
28 (49) Conklin, J. In Color Atlas of High Resolution Manometry; Soffer, E.; Pimentel, M.; Conklin, J., Eds.;
29 Springer US, 2009; pp. 59–70.
30 (50) DiMagno, E. P. Regulation of interdigestive gastrointestinal motility and secretion. Digestion 1997,
31 58, 53–5.
32 (51) Sjövall, H. Meaningful or redundant complexity - mechanisms behind cyclic changes in
33
gastroduodenal pH in the fasting state. Acta Physiol (Oxf) 2011, 201, 127–131.
34
(52) Dalenbäck, J.; Abrahamson, H.; Björnson, E.; Fändriks, L.; Mattsson, A.; Olbe, L.; Svennerholm, A.;
35
Sjövall, H. Human duodenogastric reflux, retroperistalsis, and MMC. Am. J. Physiol. 1998, 275,
36
R762-769.
37
(53) Mellander, A.; Sjövall, H. Indirect evidence for cholinergic inhibition of intestinal bicarbonate
38
absorption in humans. Gut 1999, 44, 353–360.
39
(54) Layer, P.; Chan, A. T.; Go, V. L.; Zinsmeister, A. R.; DiMagno, E. P. Cholinergic regulation of phase
40
41 II interdigestive pancreatic secretion in humans. Pancreas 1993, 8, 181–188.
42 (55) Pedersen, B. L.; Müllertz, A.; Brøndsted, H.; Kristensen, H. G. A Comparison of the Solubility of
43 Danazol in Human and Simulated Gastrointestinal Fluids. Pharm Res 2000, 17, 891–894.
44 (56) Pedersen, P. B.; Vilmann, P.; Bar-Shalom, D.; Müllertz, A.; Baldursdottir, S. Characterization of
45 fasted human gastric fluid for relevant rheological parameters and gastric lipase activities. European
46 Journal of Pharmaceutics and Biopharmaceutics 2013, 85, 958–965.
47 (57) Barrett, K. Gastrointestinal Physiology; McGraw-Hill Education, 2005.
48 (58) Persson, E. M.; Gustafsson, A.-S.; Carlsson, A. S.; Nilsson, R. G.; Knutson, L.; Forsell, P.; Hanisch,
49 G.; Lennernäs, H.; Abrahamsson, B. The Effects of Food on the Dissolution of Poorly Soluble Drugs
50 in Human and in Model Small Intestinal Fluids. Pharm Res 2005, 22, 2141–2151.
51 (59) Litou, C.; Vertzoni, M.; Goumas, C.; Vasdekis, V.; Xu, W.; Kesisoglou, F.; Reppas, C. Characteristics
52 of the Human Upper Gastrointestinal Contents in the Fasted State Under Hypo- and A-chlorhydric
53 Gastric Conditions Under Conditions of Typical Drug - Drug Interaction Studies. Pharm. Res. 2016,
54 33, 1399–1412.
55 (60) Diakidou, A.; Vertzoni, M.; Dressman, J.; Reppas, C. Estimation of intragastric drug solubility in the
56 fed state: comparison of various media with data in aspirates. Biopharm. Drug Dispos. 2009, 30, 318–
57 325.
58
59
60 36
ACS Paragon Plus Environment
Page 37 of 37 Molecular Pharmaceutics

1
2
3 (61) Kaunitz, J. D.; Akiba, Y. Review article: duodenal bicarbonate - mucosal protection, luminal
4 chemosensing and acid-base balance. Aliment. Pharmacol. Ther. 2006, 24 Suppl 4, 169–176.
5 (62) Hogan, D. L.; Ainsworth, M. A.; Isenberg, J. I. Review article: gastroduodenal bicarbonate secretion.
6 Aliment. Pharmacol. Ther. 1994, 8, 475–488.
7 (63) Matsui, K.; Tsume, Y.; Amidon, G. E.; Amidon, G. L. The Evaluation of In Vitro Drug Dissolution of
8 Commercially Available Oral Dosage Forms for Itraconazole in Gastrointestinal Simulator With
9 Biorelevant Media. J Pharm Sci 2016, 105, 2804–2814.
10 (64) Carino, S. R.; Sperry, D. C.; Hawley, M. Relative bioavailability estimation of carbamazepine crystal
11 forms using an artificial stomach-duodenum model. J Pharm Sci 2006, 95, 116–125.
12 (65) Shi, Y.; Erickson, B.; Jayasankar, A.; Lu, L.; Marsh, K.; Menon, R.; Gao, P. Assessing
13 Supersaturation and Its Impact on In Vivo Bioavailability of a Low-Solubility Compound ABT-072
14 With a Dual pH, Two-Phase Dissolution Method. J Pharm Sci 2016, 105, 2886–2895.
15 (66) Hens, B.; Brouwers, J.; Corsetti, M.; Augustijns, P. Gastrointestinal behavior of nano- and microsized
16 fenofibrate: In vivo evaluation in man and in vitro simulation by assessment of the permeation
17 potential. Eur J Pharm Sci 2015, 77, 40–47.
18 (67) Pepin, X. J. H.; Flanagan, T. R.; Holt, D. J.; Eidelman, A.; Treacy, D.; Rowlings, C. E. Justification of
19 Drug Product Dissolution Rate and Drug Substance Particle Size Specifications Based on Absorption
20 PBPK Modeling for Lesinurad Immediate Release Tablets. Mol. Pharmaceutics 2016, 13, 3256–3269.
21 (68) Kivelä, A.-J.; Kivelä, J.; Saarnio, J.; Parkkila, S. Carbonic anhydrases in normal gastrointestinal tract
22 and gastrointestinal tumours. World J. Gastroenterol. 2005, 11, 155–163.
23 (69) McGee, L. C.; Hastings, A. B. The carbon dioxide tension and acid-base balance of jejunal secretions
24 in man. Journal of Biological Chemistry 1942, 142, 893–904.
25 (70) Tsume, Y.; Mudie, D. M.; Langguth, P.; Amidon, G. E.; Amidon, G. L. The Biopharmaceutics
26 Classification System: subclasses for in vivo predictive dissolution (IPD) methodology and IVIVC.
27 Eur J Pharm Sci 2014, 57, 152–163.
28 (71) Berben, P.; Mols, R.; Brouwers, J.; Tack, J.; Augustijns, P. Gastrointestinal behavior of itraconazole in
29 humans - Part 2: The effect of intraluminal dilution on the performance of a cyclodextrin-based
30 solution. Int J Pharm 2017, 526, 235–243.
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 37
ACS Paragon Plus Environment

You might also like