1
|
Maeda Y, Goto Y, Ohnishi F, Koga S, Kawano S, Hieda Y, Goromaru T, Murakami T. 5-Aminosalicylic Acid Distribution into the Intestinal Membrane Along the Gastrointestinal Tract After Oral Administration in Rats. Pharmaceutics 2024; 16:1567. [PMID: 39771546 PMCID: PMC11677752 DOI: 10.3390/pharmaceutics16121567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND 5-Aminosalicylic acid (5-ASA), the first-line therapy for ulcerative colitis, is a poorly soluble zwitterionic drug. Unformulated 5-ASA is thought to be extensively absorbed in the small intestine. METHODS The pH-dependent solubility of 5-ASA in vitro and the intestinal membrane distribution of 5-ASA and its N-acetyl metabolite (AC-5-ASA) after the oral administration of 5-ASA were examined in fed rats. 5-ASA was administered as a suspension in water, 0.1 M HCl, or 0.1 M NaOH to untreated rats or as a solution in 5% NaHCO3 to lansoprazole-pretreated rats. RESULTS 5-ASA solubility in vitro was higher at pH < 2 and pH > 7. In rats, the 5-ASA and AC-5-ASA were detected mostly in the small intestine at 3 h and in the colonic region at 8 h after administration. The dosing vehicle (suspension or solution) and lansoprazole pretreatment did not significantly affect the pH of the luminal fluid in rats or the 5-ASA distribution in membranes. CONCLUSIONS The 5-ASA distribution in membranes in the proximal intestine was found to be restricted by the intrinsic regional luminal pH, low solubility, and saturable membrane permeability. Unabsorbed 5-ASA in the proximal intestine was delivered to the distal intestine. The higher the oral dose of 5-ASA, the more 5-ASA may be delivered to the distal intestine due to the restricted absorption in the small intestine.
Collapse
Affiliation(s)
- Yorinobu Maeda
- Laboratory of Drug Information Analytics, Faculty of Pharmacy & Pharmaceutical Sciences, Fukuyama University, Hiroshima 729-0292, Japan; (Y.G.); (F.O.); (S.K.); (S.K.); (T.G.)
| | - Yuta Goto
- Laboratory of Drug Information Analytics, Faculty of Pharmacy & Pharmaceutical Sciences, Fukuyama University, Hiroshima 729-0292, Japan; (Y.G.); (F.O.); (S.K.); (S.K.); (T.G.)
| | - Fumiya Ohnishi
- Laboratory of Drug Information Analytics, Faculty of Pharmacy & Pharmaceutical Sciences, Fukuyama University, Hiroshima 729-0292, Japan; (Y.G.); (F.O.); (S.K.); (S.K.); (T.G.)
| | - Syoutarou Koga
- Laboratory of Drug Information Analytics, Faculty of Pharmacy & Pharmaceutical Sciences, Fukuyama University, Hiroshima 729-0292, Japan; (Y.G.); (F.O.); (S.K.); (S.K.); (T.G.)
| | - Satoshi Kawano
- Laboratory of Drug Information Analytics, Faculty of Pharmacy & Pharmaceutical Sciences, Fukuyama University, Hiroshima 729-0292, Japan; (Y.G.); (F.O.); (S.K.); (S.K.); (T.G.)
| | - Yuhzo Hieda
- Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, 1 Sanzo, Fukuyama 729-0292, Japan;
| | - Takeshi Goromaru
- Laboratory of Drug Information Analytics, Faculty of Pharmacy & Pharmaceutical Sciences, Fukuyama University, Hiroshima 729-0292, Japan; (Y.G.); (F.O.); (S.K.); (S.K.); (T.G.)
| | - Teruo Murakami
- Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hiro-koshingai, Kure 737-0112, Japan
| |
Collapse
|
2
|
Ikuta S, Nakagawa H, Kai T, Sugano K. Bicarbonate buffer dissolution test with gentle mechanistic stress for bioequivalence prediction of enteric-coated pellet formulations. Eur J Pharm Sci 2024; 192:106622. [PMID: 37884100 DOI: 10.1016/j.ejps.2023.106622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/19/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
This study aimed to develop a dissolution test that can predict the bioequivalence (BE) of enteric-coated pellet formulations. The original duloxetine hydrochloride capsule (reference formulation (RF); Cymbalta® 30 mg capsule) and four generic test formulations (two capsules (CP) and two orally disintegrating tablets (OD)) were used as model formulations. Clinical BE studies were conducted on 24-47 healthy male subjects under fasting conditions. Dissolution tests were performed using a compendial paddle method (PD) (paddle speed: 50 rpm) and a flow-through cell method (FTC) (flow rate: 4 mL/min). For a further test, cotton balls were added to the vessel to apply gentle mechanistic stress to the formulations, and paddle speed was reduced to 10 rpm (paddle with cotton ball method (PDCB)).All the dissolution tests were conducted with 0.01 M HCl (pH 2.0) for 0.5 h followed by 10 mM bicarbonate buffer solutions (pH 6.5) for 4 h. One each of the two CP and two OD showed BE with RF. PDCB was able to discriminate between BE and non-BE formulations, while this was not possible with PD and FTC. In PDCB, the cotton balls intermittently moved the pellets near the vessel bottom. PDCB is useful for predicting BE during formulation development.
Collapse
Affiliation(s)
- Shotaro Ikuta
- Molecular Pharmaceutics Lab., College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1, Noji-higashi, Kusatsu, Shiga 525-8577, Japan; Pharmaceutical Research Laboratories, Pharmaceutical Department, Nipro Corporation, 3023, Noji-Cho, Kusatsu, Shiga 525-0055, Japan.
| | - Hidetoshi Nakagawa
- Pharmaceutical Research Laboratories, Pharmaceutical Department, Nipro Corporation, 3023, Noji-Cho, Kusatsu, Shiga 525-0055, Japan
| | - Toshiya Kai
- Pharmaceutical Research Laboratories, Pharmaceutical Department, Nipro Corporation, 3023, Noji-Cho, Kusatsu, Shiga 525-0055, Japan
| | - Kiyohiko Sugano
- Molecular Pharmaceutics Lab., College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1, Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| |
Collapse
|
3
|
Koziolek M, Augustijns P, Berger C, Cristofoletti R, Dahlgren D, Keemink J, Matsson P, McCartney F, Metzger M, Mezler M, Niessen J, Polli JE, Vertzoni M, Weitschies W, Dressman J. Challenges in Permeability Assessment for Oral Drug Product Development. Pharmaceutics 2023; 15:2397. [PMID: 37896157 PMCID: PMC10609725 DOI: 10.3390/pharmaceutics15102397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/12/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
Drug permeation across the intestinal epithelium is a prerequisite for successful oral drug delivery. The increased interest in oral administration of peptides, as well as poorly soluble and poorly permeable compounds such as drugs for targeted protein degradation, have made permeability a key parameter in oral drug product development. This review describes the various in vitro, in silico and in vivo methodologies that are applied to determine drug permeability in the human gastrointestinal tract and identifies how they are applied in the different stages of drug development. The various methods used to predict, estimate or measure permeability values, ranging from in silico and in vitro methods all the way to studies in animals and humans, are discussed with regard to their advantages, limitations and applications. A special focus is put on novel techniques such as computational approaches, gut-on-chip models and human tissue-based models, where significant progress has been made in the last few years. In addition, the impact of permeability estimations on PK predictions in PBPK modeling, the degree to which excipients can affect drug permeability in clinical studies and the requirements for colonic drug absorption are addressed.
Collapse
Affiliation(s)
- Mirko Koziolek
- NCE Drug Product Development, Development Sciences, AbbVie Deutschland GmbH & Co. KG, 67061 Ludwigshafen, Germany
| | - Patrick Augustijns
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Constantin Berger
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070 Würzburg, Germany;
| | - Rodrigo Cristofoletti
- Department of Pharmaceutics, University of Florida, 6550 Sanger Road, Orlando, FL 32827, USA
| | - David Dahlgren
- Department of Pharmaceutical Biosciences, Uppsala University, 75124 Uppsala, Sweden (J.N.)
| | - Janneke Keemink
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, 4070 Basel, Switzerland;
| | - Pär Matsson
- Department of Pharmacology and SciLifeLab Gothenburg, University of Gothenburg, 40530 Gothenburg, Sweden;
| | - Fiona McCartney
- School of Veterinary Medicine, University College Dublin, D04 V1W8 Dublin, Ireland;
| | - Marco Metzger
- Translational Center for Regenerative Therapies (TLZ-RT) Würzburg, Branch of the Fraunhofer Institute for Silicate Research (ISC), 97082 Würzburg, Germany
| | - Mario Mezler
- Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, 67061 Ludwigshafen, Germany;
| | - Janis Niessen
- Department of Pharmaceutical Biosciences, Uppsala University, 75124 Uppsala, Sweden (J.N.)
| | - James E. Polli
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD 21021, USA;
| | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, 157 84 Zografou, Greece;
| | - Werner Weitschies
- Institute of Pharmacy, University of Greifswald, 17489 Greifswald, Germany
| | - Jennifer Dressman
- Fraunhofer Institute of Translational Medicine and Pharmacology, 60596 Frankfurt, Germany
| |
Collapse
|
4
|
Doggwiler V, Lanz M, Paredes V, Lipps G, Imanidis G. Tablet formulation with dual control concept for efficient colonic drug delivery. Int J Pharm 2023; 631:122499. [PMID: 36529358 DOI: 10.1016/j.ijpharm.2022.122499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Aim of this study was to develop a tablet formulation for targeted colonic drug release by implementing two control mechanisms: A pH-sensitive coating layer based on Eudragit® FS 30 D to prevent drug release in the upper gastrointestinal tract, combined with a matrix based on plant-derived polysaccharide xyloglucan to inhibit drug release after coating removal in the small intestine and to allow microbiome triggered drug release in the colon. In vitro dissolution tests simulated the passage through the entire gastrointestinal tract with a four-stage protocol, including microbial xyloglucanase addition in physiologically relevant concentrations as microbiome surrogate to the colonic dissolution medium. Matrix erosion was monitored in parallel to drug release by measurement of reducing sugar equivalents resulting from xyloglucan hydrolysis. Limited drug release in gastric and small intestinal test stages and predominant release in the colonic stage was achieved. The xyloglucan matrix controlled drug release after dissolution of the enteric coating through the formation of a gummy polysaccharide layer at the tablet surface. Matrix degradation was dependent on enzyme concentration in the colonic medium and significantly accelerated drug release resulting in erosion-controlled release process. Drug release at physiologically relevant enzyme concentration was completed within the bounds of colonic transit time. The dual control concept was applicable to two drug substances with different solubility, providing similar release rates in colonic environment containing xyloglucanase. Drug solubility mechanistically affected release, with diffusion of caffeine, but not of 5-ASA, contributing to the overall release rate out of the matrix tablet.
Collapse
Affiliation(s)
- Viviane Doggwiler
- School of Life Sciences, University of Applied Sciences Northwestern Switzerland, Hofackerstrasse 30, 4132 Muttenz, Switzerland; Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Michael Lanz
- School of Life Sciences, University of Applied Sciences Northwestern Switzerland, Hofackerstrasse 30, 4132 Muttenz, Switzerland
| | - Valeria Paredes
- School of Life Sciences, University of Applied Sciences Northwestern Switzerland, Hofackerstrasse 30, 4132 Muttenz, Switzerland
| | - Georg Lipps
- School of Life Sciences, University of Applied Sciences Northwestern Switzerland, Hofackerstrasse 30, 4132 Muttenz, Switzerland
| | - Georgios Imanidis
- School of Life Sciences, University of Applied Sciences Northwestern Switzerland, Hofackerstrasse 30, 4132 Muttenz, Switzerland; Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| |
Collapse
|
5
|
O'Farrell C, Stamatopoulos K, Simmons M, Batchelor H. In vitro models to evaluate ingestible devices: Present status and current trends. Adv Drug Deliv Rev 2021; 178:113924. [PMID: 34390774 DOI: 10.1016/j.addr.2021.113924] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/03/2021] [Accepted: 08/09/2021] [Indexed: 12/14/2022]
Abstract
Orally ingestible medical devices offer significant opportunity in the diagnosis and treatment of gastrointestinal conditions. Their development necessitates the use of models that simulate the gastrointestinal environment on both a macro and micro scale. An evolution in scientific technology has enabled a wide range of in vitro, ex vivo and in vivo models to be developed that replicate the gastrointestinal tract. This review describes the landscape of the existing range of in vitro tools that are available to characterize ingestible devices. Models are presented with details on their benefits and limitations with regards to the evaluation of ingestible devices and examples of their use in the evaluation of such devices is presented where available. The multitude of models available provides a suite of tools that can be used in the evaluation of ingestible devices that should be selected on the functionality of the device and the mechanism of its function.
Collapse
Affiliation(s)
- Connor O'Farrell
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Konstantinos Stamatopoulos
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; Biopharmaceutics, Pharmaceutical Development, PDS, MST, RD Platform Technology & Science, GSK, David Jack Centre, Park Road, Ware, Hertfordshire SG12 0DP, UK
| | - Mark Simmons
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Hannah Batchelor
- Strathclyde Institute of Pharmacy and Biomedical Sciences, 161 Cathedral Street, Glasgow G4 0RE, UK.
| |
Collapse
|
6
|
Quantification of Fluid Volume and Distribution in the Paediatric Colon via Magnetic Resonance Imaging. Pharmaceutics 2021; 13:pharmaceutics13101729. [PMID: 34684022 PMCID: PMC8540766 DOI: 10.3390/pharmaceutics13101729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 12/16/2022] Open
Abstract
Previous studies have used magnetic resonance imaging (MRI) to quantify the fluid in the stomach and small intestine of children, and the stomach, small intestine and colon of adults. This is the first study to quantify fluid volumes and distribution using MRI in the paediatric colon. MRI datasets from 28 fasted (aged 0-15 years) and 18 fluid-fed (aged 10-16 years) paediatric participants were acquired during routine clinical care. A series of 2D- and 3D-based software protocols were used to measure colonic fluid volume and localisation. The paediatric colon contained a mean volume of 22.5 mL ± 41.3 mL fluid, (range 0-167.5 mL, median volume 0.80 mL) in 15.5 ± 17.5 discreet fluid pockets (median 12). The proportion of the fluid pockets larger than 1 mL was 9.6%, which contributed to 94.5% of the total fluid volume observed. No correlation was detected between all-ages and colonic fluid volume, nor was a difference in colonic fluid volumes observed based on sex, fed state or age group based on ICH-classifications. This study quantified fluid volumes within the paediatric colon, and these data will aid and accelerate the development of biorelevant tools to progress paediatric drug development for colon-targeting formulations.
Collapse
|
7
|
Vinarov Z, Abrahamsson B, Artursson P, Batchelor H, Berben P, Bernkop-Schnürch A, Butler J, Ceulemans J, Davies N, Dupont D, Flaten GE, Fotaki N, Griffin BT, Jannin V, Keemink J, Kesisoglou F, Koziolek M, Kuentz M, Mackie A, Meléndez-Martínez AJ, McAllister M, Müllertz A, O'Driscoll CM, Parrott N, Paszkowska J, Pavek P, Porter CJH, Reppas C, Stillhart C, Sugano K, Toader E, Valentová K, Vertzoni M, De Wildt SN, Wilson CG, Augustijns P. Current challenges and future perspectives in oral absorption research: An opinion of the UNGAP network. Adv Drug Deliv Rev 2021; 171:289-331. [PMID: 33610694 DOI: 10.1016/j.addr.2021.02.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/12/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023]
Abstract
Although oral drug delivery is the preferred administration route and has been used for centuries, modern drug discovery and development pipelines challenge conventional formulation approaches and highlight the insufficient mechanistic understanding of processes critical to oral drug absorption. This review presents the opinion of UNGAP scientists on four key themes across the oral absorption landscape: (1) specific patient populations, (2) regional differences in the gastrointestinal tract, (3) advanced formulations and (4) food-drug interactions. The differences of oral absorption in pediatric and geriatric populations, the specific issues in colonic absorption, the formulation approaches for poorly water-soluble (small molecules) and poorly permeable (peptides, RNA etc.) drugs, as well as the vast realm of food effects, are some of the topics discussed in detail. The identified controversies and gaps in the current understanding of gastrointestinal absorption-related processes are used to create a roadmap for the future of oral drug absorption research.
Collapse
Affiliation(s)
- Zahari Vinarov
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium; Department of Chemical and Pharmaceutical Engineering, Sofia University, Sofia, Bulgaria
| | - Bertil Abrahamsson
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden
| | - Per Artursson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Hannah Batchelor
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Philippe Berben
- Pharmaceutical Development, UCB Pharma SA, Braine- l'Alleud, Belgium
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - James Butler
- GlaxoSmithKline Research and Development, Ware, United Kingdom
| | | | - Nigel Davies
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Gøril Eide Flaten
- Department of Pharmacy, UiT The Arctic University of Norway, Tromsø, Norway
| | - Nikoletta Fotaki
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | | | | | | | | | | | - Martin Kuentz
- Institute for Pharma Technology, University of Applied Sciences and Arts Northwestern Switzerland, Basel, Switzerland
| | - Alan Mackie
- School of Food Science & Nutrition, University of Leeds, Leeds, United Kingdom
| | | | | | - Anette Müllertz
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Petr Pavek
- Faculty of Pharmacy, Charles University, Hradec Králové, Czech Republic
| | | | - Christos Reppas
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Kiyohiko Sugano
- College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan
| | - Elena Toader
- Faculty of Medicine, University of Medicine and Pharmacy of Iasi, Romania
| | - Kateřina Valentová
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Saskia N De Wildt
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Clive G Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Patrick Augustijns
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
8
|
Drug Disposition in the Lower Gastrointestinal Tract: Targeting and Monitoring. Pharmaceutics 2021; 13:pharmaceutics13020161. [PMID: 33530468 PMCID: PMC7912393 DOI: 10.3390/pharmaceutics13020161] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/20/2022] Open
Abstract
The increasing prevalence of colonic diseases calls for a better understanding of the various colonic drug absorption barriers of colon-targeted formulations, and for reliable in vitro tools that accurately predict local drug disposition. In vivo relevant incubation conditions have been shown to better capture the composition of the limited colonic fluid and have resulted in relevant degradation and dissolution kinetics of drugs and formulations. Furthermore, drug hurdles such as efflux transporters and metabolising enzymes, and the presence of mucus and microbiome are slowly integrated into drug stability- and permeation assays. Traditionally, the well characterized Caco-2 cell line and the Ussing chamber technique are used to assess the absorption characteristics of small drug molecules. Recently, various stem cell-derived intestinal systems have emerged, closely mimicking epithelial physiology. Models that can assess microbiome-mediated drug metabolism or enable coculturing of gut microbiome with epithelial cells are also increasingly explored. Here we provide a comprehensive overview of the colonic physiology in relation to drug absorption, and review colon-targeting formulation strategies and in vitro tools to characterize colonic drug disposition.
Collapse
|
9
|
Pepin XJH, Huckle JE, Alluri RV, Basu S, Dodd S, Parrott N, Emami Riedmaier A. Understanding Mechanisms of Food Effect and Developing Reliable PBPK Models Using a Middle-out Approach. AAPS JOURNAL 2021; 23:12. [PMID: 33398593 DOI: 10.1208/s12248-020-00548-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022]
Abstract
Over the last 10 years, 40% of approved oral drugs exhibited a significant effect of food on their pharmacokinetics (PK) and currently the only method to characterize the effect of food on drug absorption, which is recognized by the authorities, is to conduct a clinical evaluation. Within the pharmaceutical industry, there is a significant effort to predict the mechanism and clinical relevance of a food effect. Physiologically based pharmacokinetic (PBPK) models combining both drug-specific and physiology-specific data have been used to predict the effect of food on absorption and to reveal the underlying mechanisms. This manuscript provides detailed descriptions of how a middle-out modeling approach, combining bottom-up in vitro-based predictions with limited top-down fitting of key model parameters for clinical data, can be successfully used to predict the magnitude and direction of food effect when it is predicted poorly by a bottom-up approach. For nefazodone, a mechanistic clearance for the gut and liver was added, for furosemide, an absorption window was introduced, and for aprepitant, the biorelevant solubility was refined using multiple solubility measurements. In all cases, these adjustments were supported by literature data and showcased a rational approach to assess the factors limiting absorption and exposure.
Collapse
Affiliation(s)
- Xavier J H Pepin
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK.
| | - James E Huckle
- Drug Product Technology, Amgen, Thousand Oaks, California, USA
| | - Ravindra V Alluri
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Sumit Basu
- Pharmacokinetic, Pharmacodynamic and Drug Metabolism-Quantitative Pharmacology and Pharmacometrics (PPDM-QP2), Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Stephanie Dodd
- Chemical & Pharmaceutical Profiling, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | - Neil Parrott
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | | |
Collapse
|
10
|
Riedmaier AE, DeMent K, Huckle J, Bransford P, Stillhart C, Lloyd R, Alluri R, Basu S, Chen Y, Dhamankar V, Dodd S, Kulkarni P, Olivares-Morales A, Peng CC, Pepin X, Ren X, Tran T, Tistaert C, Heimbach T, Kesisoglou F, Wagner C, Parrott N. Use of Physiologically Based Pharmacokinetic (PBPK) Modeling for Predicting Drug-Food Interactions: an Industry Perspective. AAPS JOURNAL 2020; 22:123. [PMID: 32981010 PMCID: PMC7520419 DOI: 10.1208/s12248-020-00508-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/01/2020] [Indexed: 12/19/2022]
Abstract
The effect of food on pharmacokinetic properties of drugs is a commonly observed occurrence affecting about 40% of orally administered drugs. Within the pharmaceutical industry, significant resources are invested to predict and characterize a clinically relevant food effect. Here, the predictive performance of physiologically based pharmacokinetic (PBPK) food effect models was assessed via de novo mechanistic absorption models for 30 compounds using controlled, pre-defined in vitro, and modeling methodology. Compounds for which absorption was known to be limited by intestinal transporters were excluded in this analysis. A decision tree for model verification and optimization was followed, leading to high, moderate, or low food effect prediction confidence. High (within 0.8- to 1.25-fold) to moderate confidence (within 0.5- to 2-fold) was achieved for most of the compounds (15 and 8, respectively). While for 7 compounds, prediction confidence was found to be low (> 2-fold). There was no clear difference in prediction success for positive or negative food effects and no clear relationship to the BCS category of tested drug molecules. However, an association could be demonstrated when the food effect was mainly related to changes in the gastrointestinal luminal fluids or physiology, including fluid volume, motility, pH, micellar entrapment, and bile salts. Considering these findings, it is recommended that appropriately verified mechanistic PBPK modeling can be leveraged with high to moderate confidence as a key approach to predicting potential food effect, especially related to mechanisms highlighted here.
Collapse
Affiliation(s)
| | - Kevin DeMent
- Global DMPK, Takeda Pharmaceutical Co., Ltd., San Diego, California, USA
| | - James Huckle
- Drug Product Technology, Amgen, Thousand Oaks, California, USA
| | - Phil Bransford
- Modeling & Informatics, Vertex Pharmaceuticals, Boston, Massachusetts, USA
| | - Cordula Stillhart
- Pharmaceutical R&D, Formulation & Process Sciences, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Richard Lloyd
- Computational & Modelling Sciences, Platform Technology Sciences, GlaxoSmithKline R&D, Ware, Hertfordshire, UK
| | - Ravindra Alluri
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Sumit Basu
- Pharmacokinetic, Pharmacodynamic and Drug Metabolism-Quantitative Pharmacology and Pharmacometrics (PPDM-QP2), Merck & Co, Inc., West Point, Pennsylvania, USA
| | - Yuan Chen
- Department of Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California, USA
| | - Varsha Dhamankar
- Formulation Development, Vertex Pharmaceuticals, Boston, Massachusetts, USA.,Formulation Development, Cyclerion Therapeutics Inc., Cambridge, Massachusetts, USA
| | - Stephanie Dodd
- Chemical & Pharmaceutical Profiling, Novartis Institutes for Biomedical Research, Cambridge, Massachusetts, USA
| | - Priyanka Kulkarni
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Cambridge, Massachusetts, USA
| | - Andrés Olivares-Morales
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Chi-Chi Peng
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Cambridge, Massachusetts, USA.,Drug Metabolism and Pharmacokinetics, Theravance Biopharma, South San Francisco, California, USA
| | - Xavier Pepin
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| | - Xiaojun Ren
- Modeling & Simulation, PK Sciences, Novartis Institutes of Biomedical Research, East Hanover, New Jersey, USA
| | - Thuy Tran
- Computational & Modelling Sciences, Platform Technology Sciences, GlaxoSmithKline R&D, Collegeville, Pennsylvania, USA
| | | | - Tycho Heimbach
- PBPK & Biopharmaceutics, Novartis Institutes of Biomedical Research, Wayne, New Jersey, USA
| | | | - Christian Wagner
- Pharmaceutical Technologies, Chemical and Pharmaceutical Development, Merck Healthcare KGaA, Darmstadt, Germany
| | - Neil Parrott
- Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| |
Collapse
|
11
|
Riccio BVF, Spósito L, Carvalho GC, Ferrari PC, Chorilli M. Resveratrol isoforms and conjugates: A review from biosynthesis in plants to elimination from the human body. Arch Pharm (Weinheim) 2020; 353:e2000146. [PMID: 32886393 DOI: 10.1002/ardp.202000146] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/22/2020] [Accepted: 07/29/2020] [Indexed: 12/18/2022]
Abstract
The natural isomers of resveratrol, cis- and trans-resveratrol, are natural phenolic substances synthetized via the shikimate pathway and found in many sources, including grapes, peanuts, blackberries, pistachios, cacao, cranberries, and jackfruits. They have functional and pharmacological properties such as anticarcinogenic, antidiabetic, anti-inflammatory, and cardioprotective activities. The aim of this article is to review the data published on resveratrol and its isomers, and their biosynthesis in plants, food sources, health and toxic effects, and the excretion of their metabolites. Due to its contribution to the promotion of human health, it is convenient to gather more knowledge about its functional properties, food sources, and the interactions with the human body during the processes of eating, digestion, absorption, biotransformation, and excretion, to combine this information to improve the understanding of these substances.
Collapse
Affiliation(s)
- Bruno V F Riccio
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Larissa Spósito
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Gabriela C Carvalho
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Priscileila C Ferrari
- Department of Pharmaceutical Sciences, Ponta Grossa State University (UEPG), Ponta Grossa, Paraná, Brazil
| | - Marlus Chorilli
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| |
Collapse
|
12
|
Dissolution testing of modified release products with biorelevant media: An OrBiTo ring study using the USP apparatus III and IV. Eur J Pharm Biopharm 2020; 156:40-49. [PMID: 32882421 DOI: 10.1016/j.ejpb.2020.08.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/15/2020] [Accepted: 08/24/2020] [Indexed: 11/24/2022]
Abstract
During the OrBiTo project, our knowledge on the gastrointestinal environment has improved substantially and biorelevant media composition have been refined. The aim of this study was to propose optimized biorelevant testing conditions for modified release products, to evaluate the reproducibility of the optimized compendial apparatus III (USP apparatus III) and compendial apparatus IV (USP apparatus IV, open-loop mode) dissolution methods and to evaluate the usefulness of these methods to forecast the direction of food effects, if any, based on the results of two «ring» studies and by using two model modified release (MR) products, Ciproxin / Cipro XR and COREG CR. Six OrBiTo partners participated in each of the ring studies. All laboratories were provided with standard protocols, pure drug substance, and dose units. For the USP apparatus III, the dissolution methods applied to Ciproxin / Cipro XR, a monolithic MR product of an active pharmaceutical ingredient (API) with moderate aqueous solubility, were robust with low intra- and inter-laboratory data variability. Data from all partners were in line on a qualitative basis with food effect data in humans. For the USP apparatus IV, the dissolution methods applied to COREG CR, a multiparticulate, pH dependent, MR product of an API with low and pH dependent solubility led to high intra- and inter- laboratory data variability. Data from all partners were in line, on a qualitative basis, with the previously observed food effects in humans.
Collapse
|
13
|
Six years of progress in the oral biopharmaceutics area – A summary from the IMI OrBiTo project. Eur J Pharm Biopharm 2020; 152:236-247. [DOI: 10.1016/j.ejpb.2020.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/10/2020] [Indexed: 12/18/2022]
|
14
|
On the usefulness of compendial setups and tiny-TIM system in evaluating the in vivo performance of oral drug products with various release profiles in the fasted state: Case example sodium salt of A6197. Eur J Pharm Biopharm 2020; 149:154-162. [DOI: 10.1016/j.ejpb.2020.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/13/2020] [Accepted: 02/05/2020] [Indexed: 11/24/2022]
|
15
|
Combining biorelevant in vitro and in silico tools to simulate and better understand the in vivo performance of a nano-sized formulation of aprepitant in the fasted and fed states. Eur J Pharm Sci 2019; 138:105031. [DOI: 10.1016/j.ejps.2019.105031] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/01/2019] [Accepted: 08/01/2019] [Indexed: 11/21/2022]
|
16
|
Successful oral delivery of poorly water-soluble drugs both depends on the intraluminal behavior of drugs and of appropriate advanced drug delivery systems. Eur J Pharm Sci 2019; 137:104967. [PMID: 31252052 DOI: 10.1016/j.ejps.2019.104967] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/27/2019] [Accepted: 06/21/2019] [Indexed: 12/11/2022]
Abstract
Poorly water-soluble drugs continue to be a problematic, yet important class of pharmaceutical compounds for treatment of a wide range of diseases. Their prevalence in discovery is still high, and their development is usually limited by our lack of a complete understanding of how the complex chemical, physiological and biochemical processes that occur between administration and absorption individually and together impact on bioavailability. This review defines the challenge presented by these drugs, outlines contemporary strategies to solve this challenge, and consequent in silico and in vitro evaluation of the delivery technologies for poorly water-soluble drugs. The next steps and unmet needs are proposed to present a roadmap for future studies for the field to consider enabling progress in delivery of poorly water-soluble compounds.
Collapse
|
17
|
Impact of regional differences along the gastrointestinal tract of healthy adults on oral drug absorption: An UNGAP review. Eur J Pharm Sci 2019; 134:153-175. [DOI: 10.1016/j.ejps.2019.04.013] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/03/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023]
|
18
|
Butler J, Hens B, Vertzoni M, Brouwers J, Berben P, Dressman J, Andreas CJ, Schaefer KJ, Mann J, McAllister M, Jamei M, Kostewicz E, Kesisoglou F, Langguth P, Minekus M, Müllertz A, Schilderink R, Koziolek M, Jedamzik P, Weitschies W, Reppas C, Augustijns P. In vitro models for the prediction of in vivo performance of oral dosage forms: Recent progress from partnership through the IMI OrBiTo collaboration. Eur J Pharm Biopharm 2019; 136:70-83. [DOI: 10.1016/j.ejpb.2018.12.010] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 12/19/2018] [Indexed: 02/08/2023]
|
19
|
Physiologically based absorption modeling to predict bioequivalence of controlled release and immediate release oral products. Eur J Pharm Biopharm 2019; 134:117-125. [DOI: 10.1016/j.ejpb.2018.11.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 11/20/2018] [Accepted: 11/21/2018] [Indexed: 11/23/2022]
|
20
|
Koziolek M, Kostewicz E, Vertzoni M. Physiological Considerations and In Vitro Strategies for Evaluating the Influence of Food on Drug Release from Extended-Release Formulations. AAPS PharmSciTech 2018; 19:2885-2897. [PMID: 30155808 DOI: 10.1208/s12249-018-1159-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 08/18/2018] [Indexed: 02/08/2023] Open
Abstract
Food effects on oral drug bioavailability are a consequence of the complex interplay between drug, formulation and human gastrointestinal (GI) physiology. Accordingly, the prediction of the direction and the extent of food effects is often difficult. With respect to novel formulations, biorelevant in vitro methods can be extremely powerful tools to simulate the effect of food-induced changes on the physiological GI conditions on drug release and absorption. However, the selection of suitable in vitro methods should be based on a thorough understanding not only of human GI physiology but also of the drug and formulation properties. This review focuses on in vitro methods that can be applied to evaluate the effect of food intake on drug release from extended release (ER) products during preclinical formulation development. With the aid of different examples, it will be demonstrated that the combined and targeted use of various biorelevant in vitro methods can be extremely useful for understanding drug release from ER products in the fed state and to be able to forecast formulation-associated risks such as dose dumping in early stages of formulation development.
Collapse
|
21
|
Pentafragka C, Symillides M, McAllister M, Dressman J, Vertzoni M, Reppas C. The impact of food intake on the luminal environment and performance of oral drug products with a view to in vitro and in silico simulations: a PEARRL review. J Pharm Pharmacol 2018; 71:557-580. [DOI: 10.1111/jphp.12999] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 08/04/2018] [Indexed: 01/24/2023]
Abstract
Abstract
Objectives
Using the type of meal and dosing conditions suggested by regulatory agencies as a basis, this review has two specific objectives: first, to summarize our understanding on the impact of food intake on luminal environment and drug product performance and second, to summarize the usefulness and limitations of available in vitro and in silico methodologies for the evaluation of drug product performance after food intake.
Key findings
Characterization of the luminal environment and studies evaluating product performance in the lumen, under conditions suggested by regulatory agencies for simulating the fed state, are limited. Various in vitro methodologies have been proposed for evaluating drug product performance in the fed state, but systematic validation is lacking. Physiologically based pharmacokinetic (PBPK) modelling approaches require the use of in vitro biorelevant data and, to date, have been used primarily for investigating the mechanisms via which an already observed food effect is mediated.
Summary
Better understanding of the impact of changes induced by the meal administration conditions suggested by regulatory agencies on the luminal fate of the drug product is needed. Relevant information will be useful for optimizing the in vitro test methods and increasing the usefulness of PBPK modelling methodologies.
Collapse
Affiliation(s)
- Christina Pentafragka
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Mira Symillides
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Jennifer Dressman
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt/Main, Germany
| | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Christos Reppas
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
22
|
Tsume Y, Patel S, Fotaki N, Bergstrӧm C, Amidon GL, Brasseur JG, Mudie DM, Sun D, Bermejo M, Gao P, Zhu W, Sperry DC, Vertzoni M, Parrott N, Lionberger R, Kambayashi A, Hermans A, Lu X, Amidon GE. In Vivo Predictive Dissolution and Simulation Workshop Report: Facilitating the Development of Oral Drug Formulation and the Prediction of Oral Bioperformance. AAPS JOURNAL 2018; 20:100. [PMID: 30191341 DOI: 10.1208/s12248-018-0260-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 08/23/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Yasuhiro Tsume
- College of Pharmacy, The University of Michigan, 428 Church Street, Ann Arbor, Michigan, 48109, USA. .,Merck & Co., Inc., 126 E Lincoln Ave, Rahway, New Jersey, 07065, USA.
| | - Sanjaykumar Patel
- Merck & Co., Inc., 126 E Lincoln Ave, Rahway, New Jersey, 07065, USA
| | - Nikoletta Fotaki
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | | | - Gordon L Amidon
- College of Pharmacy, The University of Michigan, 428 Church Street, Ann Arbor, Michigan, 48109, USA
| | - James G Brasseur
- Aerospace Engineering Sciences, University of Colorado, Boulder, Colorado, USA
| | | | - Duxin Sun
- College of Pharmacy, The University of Michigan, 428 Church Street, Ann Arbor, Michigan, 48109, USA
| | | | - Ping Gao
- Abbvie, Inc., Chicago, Illinois, USA
| | - Wei Zhu
- Merck & Co., Inc., West Point, Pennsylvania, 19486, USA
| | - David C Sperry
- Eli Lilly and Company, Indianapolis, Indiana, 46285, USA
| | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Neil Parrott
- F. Hoffmann-La Roche, Ltd., Roche Innovation Center, Basel, Switzerland
| | | | | | - Andre Hermans
- Merck & Co., Inc., West Point, Pennsylvania, 19486, USA
| | - Xujin Lu
- Bristol-Myers Squibb Company, New Brunswick, New Jersey, 08903, USA
| | - Gregory E Amidon
- College of Pharmacy, The University of Michigan, 428 Church Street, Ann Arbor, Michigan, 48109, USA
| |
Collapse
|
23
|
Introduction to the OrBiTo decision tree to select the most appropriate in vitro methodology for release testing of solid oral dosage forms during development. Eur J Pharm Biopharm 2018; 130:207-213. [DOI: 10.1016/j.ejpb.2018.07.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 06/28/2018] [Accepted: 07/03/2018] [Indexed: 11/19/2022]
|
24
|
Murray K, Hoad CL, Mudie DM, Wright J, Heissam K, Abrehart N, Pritchard SE, Al Atwah S, Gowland PA, Garnett MC, Amidon GE, Spiller RC, Amidon GL, Marciani L. Magnetic Resonance Imaging Quantification of Fasted State Colonic Liquid Pockets in Healthy Humans. Mol Pharm 2017. [DOI: 10.1021/acs.molpharmaceut.7b00095] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Kathryn Murray
- Nottingham Digestive Diseases Centre and NIHR Nottingham Biomedical Research Centre at Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, United Kingdom
| | - Caroline L. Hoad
- Nottingham Digestive Diseases Centre and NIHR Nottingham Biomedical Research Centre at Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Sir
Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | | | - Jeff Wright
- Nottingham Digestive Diseases Centre and NIHR Nottingham Biomedical Research Centre at Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, United Kingdom
| | - Khaled Heissam
- Nottingham Digestive Diseases Centre and NIHR Nottingham Biomedical Research Centre at Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, United Kingdom
| | - Nichola Abrehart
- Nottingham Digestive Diseases Centre and NIHR Nottingham Biomedical Research Centre at Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, United Kingdom
| | - Susan E. Pritchard
- Sir
Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Salem Al Atwah
- Nottingham Digestive Diseases Centre and NIHR Nottingham Biomedical Research Centre at Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, United Kingdom
| | - Penny A. Gowland
- Sir
Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Martin C. Garnett
- School
of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Gregory E. Amidon
- College
of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109-1065, United States
| | - Robin C. Spiller
- Nottingham Digestive Diseases Centre and NIHR Nottingham Biomedical Research Centre at Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, United Kingdom
| | - Gordon L. Amidon
- College
of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109-1065, United States
| | - Luca Marciani
- Nottingham Digestive Diseases Centre and NIHR Nottingham Biomedical Research Centre at Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, United Kingdom
| |
Collapse
|