51
|
Turner DC, Yin F, Kindt JT, Zhang H. Understanding pharmacokinetic food effects using molecular dynamics simulation coupled with physiologically based pharmacokinetic modeling. Biopharm Drug Dispos 2012; 33:510-21. [PMID: 23065939 DOI: 10.1002/bdd.1818] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 08/22/2012] [Accepted: 10/09/2012] [Indexed: 12/23/2022]
Abstract
In this study, a molecular dynamics (MD) method is assessed as a new front-end tool for deriving relevant drug-micelle partitioning rates for use in conjunction with a compartmental-style gastrointestinal absorption model. A refined mechanistic approach for handling micelle-associated vs unbound drug is presented and examined in terms of its utility for projecting human oral pharmacokinetic food/formulation effects. Similar to predecessor oral absorption models, the intestinal drug absorption rate is formulated as a function of the combined permeability through the unstirred water layer and the epithelial membrane, however, an additional diffusion coefficient adjustment is applied to account for the viscosity changes of the postprandial small intestine. Bulk passage of drug particles through the GI tract is simulated by compartmental transit through a network of nine compartments comprising the stomach, small intestine and colon. The tandem MD simulation/compartmental absorption algorithm is applied to identify factors influencing the fed vs fasted absorption ratios of a structurally diverse set of orally administered drugs. The data illustrate the interplay and apparent compromise between micelle solubilization and permeability.
Collapse
Affiliation(s)
- David C Turner
- Department of Pharmaceutical Sciences, Mercer University, Atlanta, GA 30341, USA
| | | | | | | |
Collapse
|
52
|
Ozaki S, Kushida I, Yamashita T, Hasebe T, Shirai O, Kano K. Evaluation of Drug Supersaturation by Thermodynamic and Kinetic Approaches for the Prediction of Oral Absorbability in Amorphous Pharmaceuticals. J Pharm Sci 2012; 101:4220-30. [PMID: 22930424 DOI: 10.1002/jps.23306] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 07/25/2012] [Accepted: 08/08/2012] [Indexed: 11/10/2022]
Affiliation(s)
- Shunsuke Ozaki
- Physical Chemistry, Analytical Research Laboratories, Eisai Product Creation Systems, Eisai Company Ltd., Tsukuba, Ibaraki 300-2635, Japan.
| | | | | | | | | | | |
Collapse
|
53
|
Predicting feasibility and characterizing performance of extended-release formulations using physiologically based pharmacokinetic modeling. Ther Deliv 2012; 3:1047-59. [PMID: 23035591 DOI: 10.4155/tde.12.81] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
This review presents nine case studies where physiologically based pharmacokinetic modeling has been used in the design and development of extended-release formulations. While the approaches for creating the models were similar, in each case a product-development or drug-delivery problem unique to each compound was solved so that the drug-release rate could be optimized to achieve the best clinical performance. Examples presented include understanding the relationship between colonic absorption and efflux, effect of drug release and gastric emptying on maximum achieved drug concentration in plasma and area under the plasma concentration-time curve for a Biopharmaceutics Classification System class 3 compound, feasibility of an extended-release product for a prodrug, feasibility of an extended-release product for a biopharmaceutics classification system class 4 compound and predicting the pharmacokinetics in humans based on a primate model and coupling the physiologically-based pharmacokinetic model with a pharmacodynamic model so that the clinical efficacy of the formulations could be predicted based on the simulated plasma concentrations. The use of physiologically based pharmacokinetic models in the development of extended-release formulations is rapidly becoming an acceptable part of the knowledge management and design space components of a quality by design approach to product development. As the use of these in silico tools increase and examples become available through scientific presentations and literature, the inclusion of this approach will become a necessary part of the development process rather than the exception.
Collapse
|
54
|
Mathias NR, Crison J. The use of modeling tools to drive efficient oral product design. AAPS J 2012; 14:591-600. [PMID: 22644702 PMCID: PMC3385810 DOI: 10.1208/s12248-012-9372-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 05/10/2012] [Indexed: 11/30/2022] Open
Abstract
Modeling and simulation of drug dissolution and oral absorption has been increasingly used over the last decade to understand drug behavior in vivo based on the physicochemical properties of Active Pharmaceutical Ingredients (API) and dosage forms. As in silico and in vitro tools become more sophisticated and our knowledge of physiological processes has grown, model simulations can provide a valuable confluence, tying-in in vitro data with in vivo data while offering mechanistic insights into clinical performance. To a formulation scientist, this unveils not just the parameters that are predicted to significantly impact dissolution/absorption, but helps probe explanations around drug product performance and address specific in vivo mechanisms. In formulation, development, in silico dissolution-absorption modeling can be effectively used to guide: API selection (form comparison and particle size properties), influence clinical study design, assess dosage form performance, guide strategy for dosage form design, and breakdown clinically relevant conditions on dosage form performance (pH effect for patients on pH-elevating treatments, and food effect). This minireview describes examples of these applications in guiding product development including those with strategies to mitigate observed clinical exposure liability or mechanistically probe product in vivo performance attributes.
Collapse
Affiliation(s)
- Neil R Mathias
- Drug Product Science & Technology Department, Bristol-Myers Squibb Co, New Brunswick, New Jersey 08903, USA.
| | | |
Collapse
|
55
|
Use of physiologically based pharmacokinetic modeling for assessment of drug-drug interactions. Future Med Chem 2012; 4:681-93. [PMID: 22458685 DOI: 10.4155/fmc.12.13] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Interactions between co-administered medicines can reduce efficacy or lead to adverse effects. Understanding and managing such interactions is essential in bringing safe and effective medicines to the market. Ideally, interaction potential should be recognized early and minimized in compounds that reach late stages of drug development. Physiologically based pharmacokinetic models combine knowledge of physiological factors with compound-specific properties to simulate how a drug behaves in the human body. These software tools are increasingly used during drug discovery and development and, when integrating relevant in vitro data, can simulate drug interaction potential. This article provides some background and presents illustrative examples. Physiologically based models are an integral tool in the discovery and development of drugs, and can significantly aid our understanding and prediction of drug interactions.
Collapse
|
56
|
Budha NR, Frymoyer A, Smelick GS, Jin JY, Yago MR, Dresser MJ, Holden SN, Benet LZ, Ware JA. Drug absorption interactions between oral targeted anticancer agents and PPIs: is pH-dependent solubility the Achilles heel of targeted therapy? Clin Pharmacol Ther 2012; 92:203-13. [PMID: 22739140 DOI: 10.1038/clpt.2012.73] [Citation(s) in RCA: 241] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A majority of the novel orally administered, molecularly targeted anticancer therapies are weak bases that exhibit pH-dependent solubility, and suppression of gastric acidity with acid-reducing agents could impair their absorption. In addition, a majority of cancer patients frequently take acid-reducing agents to alleviate symptoms of gastroesophageal reflux disease, thereby raising the potential for a common but underappreciated drug-drug interaction (DDI) that could decrease the exposure of anticancer medication and result in subsequent failure of therapy. This article is a review of the available clinical literature describing the extent of the interaction between 15 orally administered, small-molecule targeted anticancer therapies and acid-reducing agents. The currently available clinical data suggest that the magnitude of this DDI is largest for compounds whose in vitro solubility varies over the pH range 1-4. This range represents the normal physiological gastric acidity (pH ~1) and gastric acidity while on an acid-reducing agent (pH ~4).
Collapse
Affiliation(s)
- N R Budha
- Department of Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
57
|
McClements DJ. Crystals and crystallization in oil-in-water emulsions: implications for emulsion-based delivery systems. Adv Colloid Interface Sci 2012; 174:1-30. [PMID: 22475330 DOI: 10.1016/j.cis.2012.03.002] [Citation(s) in RCA: 200] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 02/29/2012] [Accepted: 03/01/2012] [Indexed: 02/08/2023]
Abstract
Many bioactive components intended for oral ingestion (pharmaceuticals and nutraceuticals) are hydrophobic molecules with low water-solubilities and high melting points, which poses considerable challenges to the formulation of oral delivery systems. Oil-in-water emulsions are often suitable vehicles for the encapsulation and delivery of this type of bioactive component. The bioactive component is usually dissolved in a carrier lipid phase by either dilution and/or heating prior to homogenization, and then the carrier lipid and water phases are homogenized to form an emulsion consisting of small oil droplets dispersed in water. The successful development of this kind of emulsion-based delivery system depends on a good understanding of the influence of crystals on the formation, stability, and properties of emulsions. This review article addresses the physicochemical phenomena associated with the encapsulation, retention, crystallization, release, and absorption of hydrophobic bioactive components within emulsions. This knowledge will be useful for the rational formulation of effective emulsion-based delivery systems for oral delivery of crystalline hydrophobic bioactive components in the food, health care, and pharmaceutical industries.
Collapse
|
58
|
Takano R, Kataoka M, Yamashita S. Integrating drug permeability with dissolution profile to develop IVIVC. Biopharm Drug Dispos 2012; 33:354-65. [PMID: 22581486 DOI: 10.1002/bdd.1792] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 04/11/2012] [Accepted: 05/06/2012] [Indexed: 11/07/2022]
Abstract
In this review article, three different approaches to predict in vivo oral absorption based on the in vitro data of drug permeability, solubility and dissolution were introduced. At the drug discovery stage, the absorption potential of each candidate is most important to select better compounds for further development. The concept of maximum absorbable dose is applied widely, not only to evaluate the absorption potential but also to elucidate the rate-limiting process of oral absorption that helps us to understand the cause of poor absorption. To integrate the permeability of the drug with its dissolution profile, two different approaches, in vitro dissolution/permeation system (D/P system) and in silico model and simulation method, are proposed. In the D/P system, by mimicking the in vivo process of drug absorption, the permeated amount of drugs, that is the total output of dissolution and permeation processes, are correlated with the fraction absorbed in human (F(a)). This system is powerful for evaluating the improved absorption by various formulations and the effect of food intake. On the other hand, in the model and simulation approach, an intrinsic dissolution parameter of drug particle, z, was extracted from the small scale in vitro test and the process of intestinal absorption was re-constructed in silico by incorporating the physiological parameters in human. The effective use of these approaches for the development of oral drug products is discussed through various case studies.
Collapse
Affiliation(s)
- Ryusuke Takano
- Pre-clinical Research Department, Chugai Pharmaceutical Co.,Ltd, 1-135 Komakado, Gotemba, Shizuoka, 412-8513, Japan
| | | | | |
Collapse
|
59
|
Bridging solubility between drug discovery and development. Drug Discov Today 2012; 17:486-95. [DOI: 10.1016/j.drudis.2011.11.007] [Citation(s) in RCA: 295] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 10/25/2011] [Accepted: 11/18/2011] [Indexed: 11/22/2022]
|
60
|
Ozaki S, Minamisono T, Yamashita T, Kato T, Kushida I. Supersaturation–Nucleation Behavior of Poorly Soluble Drugs and its Impact on the Oral Absorption of Drugs in Thermodynamically High-Energy Forms. J Pharm Sci 2012; 101:214-22. [PMID: 21918988 DOI: 10.1002/jps.22760] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 07/28/2011] [Accepted: 08/25/2011] [Indexed: 11/09/2022]
Affiliation(s)
- Shunsuke Ozaki
- Physical Chemistry, Analytical Research Laboratories, Eisai Product Creation Systems, Eisai Company Ltd, Tsukuba, Ibaraki 300-2635, Japan.
| | | | | | | | | |
Collapse
|
61
|
McClements DJ, Xiao H. Potential biological fate of ingested nanoemulsions: influence of particle characteristics. Food Funct 2012; 3:202-20. [DOI: 10.1039/c1fo10193e] [Citation(s) in RCA: 229] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
62
|
Jones HM, Dickins M, Youdim K, Gosset JR, Attkins NJ, Hay TL, Gurrell IK, Logan YR, Bungay PJ, Jones BC, Gardner IB. Application of PBPK modelling in drug discovery and development at Pfizer. Xenobiotica 2011; 42:94-106. [DOI: 10.3109/00498254.2011.627477] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
63
|
Shono Y, Jantratid E, Dressman JB. Precipitation in the small intestine may play a more important role in the in vivo performance of poorly soluble weak bases in the fasted state: Case example nelfinavir. Eur J Pharm Biopharm 2011; 79:349-56. [DOI: 10.1016/j.ejpb.2011.04.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 04/06/2011] [Accepted: 04/11/2011] [Indexed: 11/16/2022]
|
64
|
Young RJ, Green DV, Luscombe CN, Hill AP. Getting physical in drug discovery II: the impact of chromatographic hydrophobicity measurements and aromaticity. Drug Discov Today 2011; 16:822-30. [DOI: 10.1016/j.drudis.2011.06.001] [Citation(s) in RCA: 212] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 05/08/2011] [Accepted: 06/07/2011] [Indexed: 11/24/2022]
|
65
|
Rowland M, Peck C, Tucker G. Physiologically-based pharmacokinetics in drug development and regulatory science. Annu Rev Pharmacol Toxicol 2011; 51:45-73. [PMID: 20854171 DOI: 10.1146/annurev-pharmtox-010510-100540] [Citation(s) in RCA: 428] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The application of physiologically-based pharmacokinetic (PBPK) modeling is coming of age in drug development and regulation, reflecting significant advances over the past 10 years in the predictability of key pharmacokinetic (PK) parameters from human in vitro data and in the availability of dedicated software platforms and associated databases. Specific advances and contemporary challenges with respect to predicting the processes of drug clearance, distribution, and absorption are reviewed, together with the ability to anticipate the quantitative extent of PK-based drug-drug interactions and the impact of age, genetics, disease, and formulation. The value of this capability in selecting and designing appropriate clinical studies, its implications for resource-sparing techniques, and a more holistic view of the application of PK across the preclinical/clinical divide are considered. Finally, some attention is given to the positioning of PBPK within the drug development and approval paradigm and its future application in truly personalized medicine.
Collapse
Affiliation(s)
- Malcolm Rowland
- Centre for Pharmacokinetic Research, University of Manchester, United Kingdom.
| | | | | |
Collapse
|
66
|
D'Arcy DM, Persoons T. Mechanistic Modelling and Mechanistic Monitoring: Simulation and Shadowgraph Imaging of Particulate Dissolution in the Flow-Through Apparatus. J Pharm Sci 2011; 100:1102-15. [DOI: 10.1002/jps.22337] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Revised: 07/15/2010] [Accepted: 07/30/2010] [Indexed: 11/08/2022]
|
67
|
Sugano K. Computational oral absorption simulation of free base drugs. Int J Pharm 2010; 398:73-82. [DOI: 10.1016/j.ijpharm.2010.07.027] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 07/15/2010] [Accepted: 07/17/2010] [Indexed: 11/25/2022]
|
68
|
Willmann S, Thelen K, Becker C, Dressman JB, Lippert J. Mechanism-based prediction of particle size-dependent dissolution and absorption: Cilostazol pharmacokinetics in dogs. Eur J Pharm Biopharm 2010; 76:83-94. [DOI: 10.1016/j.ejpb.2010.06.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 05/10/2010] [Accepted: 06/02/2010] [Indexed: 11/26/2022]
|
69
|
Predicting Intestinal Precipitation—A Case Example for a Basic BCS Class II Drug. Pharm Res 2010; 27:2119-30. [DOI: 10.1007/s11095-010-0213-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 07/06/2010] [Indexed: 10/19/2022]
|
70
|
Takano R, Takata N, Saito R, Furumoto K, Higo S, Hayashi Y, Machida M, Aso Y, Yamashita S. Quantitative Analysis of the Effect of Supersaturation on in Vivo Drug Absorption. Mol Pharm 2010; 7:1431-40. [DOI: 10.1021/mp100109a] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Ryusuke Takano
- Discovery Platform Technology Department, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, and Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan
| | - Noriyuki Takata
- Discovery Platform Technology Department, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, and Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan
| | - Ryoichi Saito
- Discovery Platform Technology Department, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, and Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan
| | - Kentaro Furumoto
- Discovery Platform Technology Department, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, and Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan
| | - Shoichi Higo
- Discovery Platform Technology Department, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, and Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan
| | - Yoshiki Hayashi
- Discovery Platform Technology Department, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, and Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan
| | - Minoru Machida
- Discovery Platform Technology Department, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, and Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan
| | - Yoshinori Aso
- Discovery Platform Technology Department, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, and Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan
| | - Shinji Yamashita
- Discovery Platform Technology Department, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, Chugai Pharmaceutical Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan, and Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge-cho, Hirakata, Osaka 573-0101, Japan
| |
Collapse
|
71
|
Avdeef A, Tam KY. How well can the Caco-2/Madin-Darby canine kidney models predict effective human jejunal permeability? J Med Chem 2010; 53:3566-84. [PMID: 20373811 DOI: 10.1021/jm901846t] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The study aimed to predict effective human jejunal permeability (P(eff)) using a biophysical model based on parametrized paracellular, aqueous boundary layer, and transcellular permeabilities, and the villus-fold surface area expansion factor (k(VF)). Published human jejunal data (119 P(eff), 53 compounds) were analyzed by a regression procedure incorporating a dual-pore size paracellular model. Transcellular permeability, scaled by k(VF), was equated to that of Caco-2 at pH 6.5. The biophysical model predicted human jejunal permeability data within the experimental uncertainty. This investigation revealed several surprising predictions: (i) many molecules permeate predominantly (but not exclusively) by the paracellular route, (ii) the aqueous boundary layer thickness in the intestinal perfusion experiments is larger than expected, (iii) the mucosal surface area in awake humans is apparently nearly entirely accessible to drug absorption, and (iv) the relative "leakiness" of the human jejunum is not so different from that observed in a number of published Caco-2 studies.
Collapse
Affiliation(s)
- Alex Avdeef
- pION Inc., 5 Constitution Way, Woburn, Massachusetts 01801, USA.
| | | |
Collapse
|
72
|
Avdeef A. Leakiness and size exclusion of paracellular channels in cultured epithelial cell monolayers-interlaboratory comparison. Pharm Res 2010; 27:480-9. [PMID: 20069445 DOI: 10.1007/s11095-009-0036-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Accepted: 12/10/2009] [Indexed: 11/30/2022]
Abstract
PURPOSE To determine and compare the paracellular characteristics of permeability (Papp) of Caco (-2), MDCK, and 2/4/A1 cell lines. METHODS The Papp data from 14 studies were analyzed by weighted nonlinear regression in terms of the paracellular parameters: porosity-pathlength (epsilon/delta), pore radius (R), and electrostatic potential drop (deltaphi). Aqueous diffusivities, Daq, for the analysis, were empirically determined. The required hydrodynamic radii, rHYD, were estimated without knowledge of compound density. Mannitol iso-paracellular profiles allowed comparisons of "leakiness" across labs. RESULTS Daq (37 degreeC) was predicted as 9.9x10(-5) MW(-0.453); rHYD=(0.92+21.8 MW(-1))xrSE, where rSE is the Stokes-Einstein radius. Values of pore radius ranged from 4.0(+/-0.1) to 18(+/-3) A, with the 2/4/A1 indicating the largest pores. The epsilon/delta capacity factor ranged from 0.2 (+/-0.1) to 69 (+/-5) cm(-1), with most values <1.5 cm(-1). The average potential drop for Caco-2 models was deltaphi(wt avg) Caco(-2)=(-43)+/-20 mV. The paracellular model predicted measured log Papp values with pooled r2=0.93 and s=0.17 (n=108). CONCLUSION R and epsilon/delta are negatively correlated to a large extent. Papp can be rate-limited by either factor, with a wide range of possible combinations still indicating nearly constant leakiness for a given marker.
Collapse
Affiliation(s)
- Alex Avdeef
- pION INC, 5 Constitution Way, Woburn, Massachusetts 01801,USA.
| |
Collapse
|
73
|
Sugano K, Kataoka M, da Costa Mathews C, Yamashita S. Prediction of food effect by bile micelles on oral drug absorption considering free fraction in intestinal fluid. Eur J Pharm Sci 2010; 40:118-24. [DOI: 10.1016/j.ejps.2010.03.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Revised: 02/18/2010] [Accepted: 03/12/2010] [Indexed: 11/30/2022]
|
74
|
Pieper IA, Bertau M. Predictive tools for the evaluation of microbial effects on drugs during gastrointestinal passage. Expert Opin Drug Metab Toxicol 2010; 6:747-60. [DOI: 10.1517/17425251003769859] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
75
|
Abstract
Bile micelles play an important role in oral absorption of low-solubility compounds. Bile micelles can affect solubility, dissolution rate, and permeability. For the pH-solubility profile in bile micelles, the Henderson-Hasselbalch equation should be modified to take bile-micelle partition into account. For the dissolution rate, in the Nernst-Brunner equation, the effective diffusion coefficient in bile-micelle media should be used instead of the monomer diffusion coefficient. The diffusion coefficient of bile micelles is 8- to 18-fold smaller than that of monomer molecules. For permeability, the effective diffusion coefficient in the unstirred water layer adjacent to the epithelial membrane, and the free fraction at the epithelial membrane surface should be taken into account. The importance of these aspects is demonstrated here using several in vivo and clinical oral-absorption data of low-solubility model compounds. Using the theoretical equations, the food effect on oral absorption is further discussed.
Collapse
Affiliation(s)
- Kiyohiko Sugano
- Global Research & Development, Sandwich Laboratories, Research Formulation, Pfizer Inc., CT13 9NJ, Sandwich, Kent, UK.
| |
Collapse
|
76
|
Sugano K. Possible reduction of effective thickness of intestinal unstirred water layer by particle drifting effect. Int J Pharm 2009; 387:103-9. [PMID: 20006692 DOI: 10.1016/j.ijpharm.2009.12.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 12/03/2009] [Accepted: 12/04/2009] [Indexed: 11/25/2022]
Abstract
According to the present theory of oral absorption, in the case of solubility limited absorption, the absorbed amount would not increase despite an increase in dose or a decrease in particle size. However, many experimental observations suggested that the absorbed amount was often increased (though sub-proportionally) as the dose strength increased. In addition, the particle size reduction was often effective to increase the absorbed amount even in the case of solubility limited absorption. Since an increase of the dose strength and a decrease of the particle size cause no or little change in solubility and the mean intestinal transit time, effective intestinal membrane permeability (P(eff)) should have changed. The previous theory postulated that drug particles do not exist in the unstirred water layer (UWL) which is adjacent to the intestinal membrane. However, many reports suggested that nano- to micro-scale drug particles existed in the UWL. In this case, the effective thickness of the UWL (h(eff)) could be smaller than the nominal thickness, resulting in an increase of P(eff). In the present study, h(eff) was simply calculated assuming that the reduction of h(eff) is in proportion to the surface area of drug particles in the UWL. When the particle drifting effect was taken into account, the discrepancy between the theoretical calculation and experimental observations was reduced. It was suggested that when the dose (mg)/particle diameter (microm) ratio exceeds 20, the particle drifting effect would become significant.
Collapse
Affiliation(s)
- Kiyohiko Sugano
- Global Research & Development, Sandwich Laboratories, Research Formulation, Pfizer Inc., CT13 9NJ, Sandwich, Kent, UK.
| |
Collapse
|
77
|
Sugano K. A simulation of oral absorption using classical nucleation theory. Int J Pharm 2009; 378:142-5. [DOI: 10.1016/j.ijpharm.2009.05.056] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Revised: 05/15/2009] [Accepted: 05/25/2009] [Indexed: 10/20/2022]
|
78
|
Sugano K. Calculation of fraction of dose absorbed: comparison between analytical solution based on one compartment steady state concentration approximation and dynamic seven compartment model. CHEM-BIO INFORMATICS JOURNAL 2009. [DOI: 10.1273/cbij.9.75] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Kiyohiko Sugano
- Global Research & Development, Sandwich Laboratories, Research Formulation, Pfizer Inc
| |
Collapse
|