Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Christer Tannergren is active.

Publication


Featured researches published by Christer Tannergren.


European Journal of Pharmaceutical Sciences | 2014

PBPK models for the prediction of in vivo performance of oral dosage forms

Edmund S. Kostewicz; Leon Aarons; Martin Bergstrand; Michael B. Bolger; Aleksandra Galetin; Oliver J. D. Hatley; Masoud Jamei; Richard Lloyd; Xavier Pepin; Amin Rostami-Hodjegan; Erik Sjögren; Christer Tannergren; David B. Turner; Christian Wagner; Werner Weitschies; Jennifer B. Dressman

Drug absorption from the gastrointestinal (GI) tract is a highly complex process dependent upon numerous factors including the physicochemical properties of the drug, characteristics of the formulation and interplay with the underlying physiological properties of the GI tract. The ability to accurately predict oral drug absorption during drug product development is becoming more relevant given the current challenges facing the pharmaceutical industry. Physiologically-based pharmacokinetic (PBPK) modeling provides an approach that enables the plasma concentration-time profiles to be predicted from preclinical in vitro and in vivo data and can thus provide a valuable resource to support decisions at various stages of the drug development process. Whilst there have been quite a few successes with PBPK models identifying key issues in the development of new drugs in vivo, there are still many aspects that need to be addressed in order to maximize the utility of the PBPK models to predict drug absorption, including improving our understanding of conditions in the lower small intestine and colon, taking the influence of disease on GI physiology into account and further exploring the reasons behind population variability. Importantly, there is also a need to create more appropriate in vitro models for testing dosage form performance and to streamline data input from these into the PBPK models. As part of the Oral Biopharmaceutical Tools (OrBiTo) project, this review provides a summary of the current status of PBPK models available. The current challenges in PBPK set-ups for oral drug absorption including the composition of GI luminal contents, transit and hydrodynamics, permeability and intestinal wall metabolism are discussed in detail. Further, the challenges regarding the appropriate integration of results from in vitro models, such as consideration of appropriate integration/estimation of solubility and the complexity of the in vitro release and precipitation data, are also highlighted as important steps to advancing the application of PBPK models in drug development. It is expected that the innovative integration of in vitro data from more appropriate in vitro models and the enhancement of the GI physiology component of PBPK models, arising from the OrBiTo project, will lead to a significant enhancement in the ability of PBPK models to successfully predict oral drug absorption and advance their role in preclinical and clinical development, as well as for regulatory applications.


European Journal of Pharmaceutical Sciences | 2014

In vivo methods for drug absorption - comparative physiologies, model selection, correlations with in vitro methods (IVIVC), and applications for formulation/API/excipient characterization including food effects.

Erik Sjögren; Bertil Abrahamsson; Patrick Augustijns; Dieter Becker; Michael B. Bolger; Marcus E. Brewster; Joachim Brouwers; Talia Flanagan; Matthew D. Harwood; Christian Heinen; René Holm; Hans-Paul Juretschke; Marlies Kubbinga; Anders Lindahl; Viera Lukacova; Uwe Münster; Sibylle Neuhoff; Mai Anh Nguyen; Achiel Van Peer; Christos Reppas; Amin Rostami Hodjegan; Christer Tannergren; Werner Weitschies; Clive G. Wilson; Patricia Zane; Hans Lennernäs; Peter Langguth

This review summarizes the current knowledge on anatomy and physiology of the human gastrointestinal tract in comparison with that of common laboratory animals (dog, pig, rat and mouse) with emphasis on in vivo methods for testing and prediction of oral dosage form performance. A wide range of factors and methods are considered in addition, such as imaging methods, perfusion models, models for predicting segmental/regional absorption, in vitro in vivo correlations as well as models to investigate the effects of excipients and the role of food on drug absorption. One goal of the authors was to clearly identify the gaps in todays knowledge in order to stimulate further work on refining the existing in vivo models and demonstrate their usefulness in drug formulation and product performance testing.


European Journal of Pharmaceutical Sciences | 2013

Comprehensive study on regional human intestinal permeability and prediction of fraction absorbed of drugs using the Ussing chamber technique

Åsa Sjöberg; Mareike Lutz; Christer Tannergren; Caroline Wingolf; Anders Borde; Anna-Lena Ungell

The purpose of this study was to evaluate the use of human intestinal tissue in Ussing chamber to predict oral and colonic drug absorption and intestinal metabolism. Data on viability, correlation between apparent permeability coefficients (P(app)) and fraction absorbed (f(a)) after oral and colonic administration, regional permeability, active uptake and efflux of drugs as well as intestinal metabolism were compiled from experiments using 159 human donors. Permeability coefficients for up to 28 drugs were determined using one or several of four intestinal regions: duodenum, jejunum, ileum and colon and 10 drugs were studied bidirectionally. Viability was monitored simultaneously with transport experiments by recording potential difference (PD), short-circuit current (SCC) and the resistance (TER). Intestinal metabolism was studied using testosterone and midazolam as probe substrates. There was a steep sigmoidal correlation between P(app) in the Ussing chamber, using jejunal segments, and oral f(a) in humans, for a set of 25 drugs (R(2): 0.85, p<0.01). A clear sigmoidal relationship was also obtained between P(app) in colonic segments and f(a) after colonic administration in humans for a set of 10 drugs (R(2): 0.93, p<0.05). Regional permeability data showed a tendency for highly permeable compounds to have higher or similar P(app) in colon as in the small intestinal segments, while the colonic regions showed a lower P(app) for more polar compounds as well as for d-glucose and l-leucine. Bidirectional transport (mucosa to serosa and serosa to mucosa direction) in jejunum showed well functioning efflux- and uptake asymmetry. Intestinal metabolic extraction during transport across jejunum segments was found for both testosterone and midazolam. In conclusion, viable excised human intestine mounted in the Ussing chamber, is a powerful technique for predicting regional fraction absorbed (f(a)), transporter-mediated uptake or efflux as well as intestinal metabolism of drug candidates in man. Furthermore, a sigmoidal relationship of P(app) vs. f(a) was obtained when permeability data from the present study were merged with data from 2 other independent laboratories (R(2): 0.83, p<0.01). The correlation curve reported can be used by any laboratory for predictions of human permeability and f(a)(.) In addition, for the first time a correlation curve between colonic P(app) and human colonic f(a) is reported, which demonstrates the usefulness of this methodology in early assessment of the colonic absorption potential of extended release formulation candidates.


European Journal of Pharmaceutical Sciences | 2013

In silico predictions of gastrointestinal drug absorption in pharmaceutical product development : Application of the mechanistic absorption model GI-Sim

Erik Sjögren; Jan Westergren; Iain Grant; Gunilla Hanisch; Lennart Lindfors; Hans Lennernäs; Bertil Abrahamsson; Christer Tannergren

Oral drug delivery is the predominant administration route for a major part of the pharmaceutical products used worldwide. Further understanding and improvement of gastrointestinal drug absorption predictions is currently a highly prioritized area of research within the pharmaceutical industry. The fraction absorbed (fabs) of an oral dose after administration of a solid dosage form is a key parameter in the estimation of the in vivo performance of an orally administrated drug formulation. This study discloses an evaluation of the predictive performance of the mechanistic physiologically based absorption model GI-Sim. GI-Sim deploys a compartmental gastrointestinal absorption and transit model as well as algorithms describing permeability, dissolution rate, salt effects, partitioning into micelles, particle and micelle drifting in the aqueous boundary layer, particle growth and amorphous or crystalline precipitation. Twelve APIs with reported or expected absorption limitations in humans, due to permeability, dissolution and/or solubility, were investigated. Predictions of the intestinal absorption for different doses and formulations were performed based on physicochemical and biopharmaceutical properties, such as solubility in buffer and simulated intestinal fluid, molecular weight, pK(a), diffusivity and molecule density, measured or estimated human effective permeability and particle size distribution. The performance of GI-Sim was evaluated by comparing predicted plasma concentration-time profiles along with oral pharmacokinetic parameters originating from clinical studies in healthy individuals. The capability of GI-Sim to correctly predict impact of dose and particle size as well as the in vivo performance of nanoformulations was also investigated. The overall predictive performance of GI-Sim was good as >95% of the predicted pharmacokinetic parameters (C(max) and AUC) were within a 2-fold deviation from the clinical observations and the predicted plasma AUC was within one standard deviation of the observed mean plasma AUC in 74% of the simulations. GI-Sim was also able to correctly capture the trends in dose- and particle size dependent absorption for the study drugs with solubility and dissolution limited absorption, respectively. In addition, GI-Sim was also shown to be able to predict the increase in absorption and plasma exposure achieved with nanoformulations. Based on the results, the performance of GI-Sim was shown to be suitable for early risk assessment as well as to guide decision making in pharmaceutical formulation development.


European Journal of Pharmaceutical Sciences | 2017

IMI – Oral biopharmaceutics tools project – Evaluation of bottom-up PBPK prediction success part 3: Identifying gaps in system parameters by analysing In Silico performance across different compound classes

Adam S. Darwich; Alison Margolskee; Xavier Pepin; Leon Aarons; Aleksandra Galetin; Amin Rostami-Hodjegan; Sara Carlert; Maria Hammarberg; Constanze Hilgendorf; Pernilla Johansson; Eva Karlsson; Dónal Murphy; Christer Tannergren; Helena Thörn; Mohammed Yasin; Florent Mazuir; Olivier Nicolas; Sergej Ramusovic; Christine Xu; Shriram M. Pathak; Timo Korjamo; Johanna Laru; Jussi Malkki; Sari Pappinen; Johanna Tuunainen; Jennifer B. Dressman; Simone Hansmann; Edmund S. Kostewicz; Handan He; Tycho Heimbach

&NA; Three Physiologically Based Pharmacokinetic software packages (GI‐Sim, Simcyp® Simulator, and GastroPlus™) were evaluated as part of the Innovative Medicine Initiative Oral Biopharmaceutics Tools project (OrBiTo) during a blinded “bottom‐up” anticipation of human pharmacokinetics. After data analysis of the predicted vs. measured pharmacokinetics parameters, it was found that oral bioavailability (Foral) was underpredicted for compounds with low permeability, suggesting improper estimates of intestinal surface area, colonic absorption and/or lack of intestinal transporter information. Foral was also underpredicted for acidic compounds, suggesting overestimation of impact of ionisation on permeation, lack of information on intestinal transporters, or underestimation of solubilisation of weak acids due to less than optimal intestinal model pH settings or underestimation of bile micelle contribution. Foral was overpredicted for weak bases, suggesting inadequate models for precipitation or lack of in vitro precipitation information to build informed models. Relative bioavailability was underpredicted for both high logP compounds as well as poorly water‐soluble compounds, suggesting inadequate models for solubility/dissolution, underperforming bile enhancement models and/or lack of biorelevant solubility measurements. These results indicate areas for improvement in model software, modelling approaches, and generation of applicable input data. However, caution is required when interpreting the impact of drug‐specific properties in this exercise, as the availability of input parameters was heterogeneous and highly variable, and the modellers generally used the data “as is” in this blinded bottom‐up prediction approach. Graphical Abstract Figure. No caption available.


European Journal of Pharmaceutical Sciences | 2017

IMI - Oral biopharmaceutics tools project - Evaluation of bottom-up PBPK prediction success part 2 : An introduction to the simulation exercise and overview of results

Alison Margolskee; Adam S. Darwich; Xavier Pepin; Leon Aarons; Aleksandra Galetin; Amin Rostami-Hodjegan; Sara Carlert; Maria Hammarberg; Constanze Hilgendorf; Pernilla Johansson; Eva Karlsson; Dónal Murphy; Christer Tannergren; Helena Thörn; Mohammed Yasin; Florent Mazuir; Olivier Nicolas; Sergej Ramusovic; Christine Xu; Shriram M. Pathak; Timo Korjamo; Johanna Laru; Jussi Malkki; Sari Pappinen; Johanna Tuunainen; Jennifer B. Dressman; Simone Hansmann; Edmund S. Kostewicz; Handan He; Tycho Heimbach

&NA; Orally administered drugs are subject to a number of barriers impacting bioavailability (Foral), causing challenges during drug and formulation development. Physiologically‐based pharmacokinetic (PBPK) modelling can help during drug and formulation development by providing quantitative predictions through a systems approach. The performance of three available PBPK software packages (GI‐Sim, Simcyp®, and GastroPlus™) were evaluated by comparing simulated and observed pharmacokinetic (PK) parameters. Since the availability of input parameters was heterogeneous and highly variable, caution is required when interpreting the results of this exercise. Additionally, this prospective simulation exercise may not be representative of prospective modelling in industry, as API information was limited to sparse details. 43 active pharmaceutical ingredients (APIs) from the OrBiTo database were selected for the exercise. Over 4000 simulation output files were generated, representing over 2550 study arm‐institution‐software combinations and approximately 600 human clinical study arms simulated with overlap. 84% of the simulated study arms represented administration of immediate release formulations, 11% prolonged or delayed release, and 5% intravenous (i.v.). Higher percentages of i.v. predicted area under the curve (AUC) were within two‐fold of observed (52.9%) compared to per oral (p.o.) (37.2%), however, Foral and relative AUC (Frel) between p.o. formulations and solutions were generally well predicted (64.7% and 75.0%). Predictive performance declined progressing from i.v. to solution and immediate release tablet, indicating the compounding error with each layer of complexity. Overall performance was comparable to previous large‐scale evaluations. A general overprediction of AUC was observed with average fold error (AFE) of 1.56 over all simulations. AFE ranged from 0.0361 to 64.0 across the 43 APIs, with 25 showing overpredictions. Discrepancies between software packages were observed for a few APIs, the largest being 606, 171, and 81.7‐fold differences in AFE between SimCYP and GI‐Sim, however average performance was relatively consistent across the three software platforms. Graphical abstract Figure. No caption available.


European Journal of Pharmaceutical Sciences | 2014

Evaluation of an in vitro faecal degradation method for early assessment of the impact of colonic degradation on colonic absorption in humans.

Christer Tannergren; Anders Borde; Cecilia Boreström; Bertil Abrahamsson; Anders Lindahl

The objective of this study was to develop and evaluate an in vitro method to investigate bacterial-mediated luminal degradation of drugs in colon in humans. This would be a valuable tool for the assessment of drug candidates during early drug development, especially for compounds intended to be developed as oral extended release formulations. Freshly prepared faecal homogenate from healthy human volunteers (n=3-18), dog (n=6) and rat (colon and caecal content, n=3) was homogenised with 3.8 parts (w/w) physiological saline under anaerobical conditions. Four model compounds (almokalant, budesonide, ximelagatran and metoprolol) were then incubated (n=3-18) separately in the human faecal homogenate for up to 120min at 37°C. In addition, ximelagatran was also incubated in the faecal or colonic content from dog and rat. The mean (±SD) in vitro half-life for almokalant, budesonide and ximelagatran was 39±1, 68±21 and 26±12min, respectively, in the human faecal homogenate. Metoprolol was found to be stable in the in vitro model. The in vitro degradation data was then compared to literature data on fraction absorbed after direct colon administration in humans. The percentage of drug remaining after 60min of in vitro incubation correlated (R(2)=0.90) with the fraction absorbed from colon in humans. The mean in vitro half-life of ximelagatran was similar in human faeces (26±12min) and rat colon content (34±31min), but significantly (p<0.05) longer in rat caecum content (50±11min) and dog faeces (126±17min). The in vitro method is in vivo relevant both qualitatively as all the model drugs that undergoes colonic degradation in vivo was rapidly degraded in the faecal homogenates as well as quantitatively since a correlation was established between percentage degraded in vitro at 60min and fraction absorbed in the colon for the model drugs, which have no other absorption limiting properties. Also, the method is easy to use from a technical point of view, which suggests that the method is suitable for use in early assessment of colonic absorption of extended release formulation candidates. Further improvement of the confidence in the use of the method would either require an extension of the correlation, which most likely will require more human regional absorption studies, or by including colonic degradation rate as an input in a physiological mechanistic absorption model and evaluate if the prediction of the plasma exposure after colonic administration of the present model drugs is improved.


European Journal of Pharmaceutical Sciences | 2017

IMI – oral biopharmaceutics tools project – evaluation of bottom-up PBPK prediction success part 1: Characterisation of the OrBiTo database of compounds

Alison Margolskee; Adam S. Darwich; Xavier Pepin; Shriram M. Pathak; Michael B. Bolger; Leon Aarons; Amin Rostami-Hodjegan; Jonas Angstenberger; Franziska Graf; Loic Laplanche; Thomas J. J. Müller; Sara Carlert; Pankaj Daga; Dónal Murphy; Christer Tannergren; Mohammed Yasin; Susanne Greschat-Schade; Wolfgang Mück; Uwe Muenster; Dorina van der Mey; Kerstin J. Frank; Richard Lloyd; Lieve Adriaenssen; Jan Bevernage; Loeckie de Zwart; Dominique Swerts; Christophe Tistaert; An Van den Bergh; Achiel Van Peer; Stefania Beato

&NA; Predicting oral bioavailability (Foral) is of importance for estimating systemic exposure of orally administered drugs. Physiologically‐based pharmacokinetic (PBPK) modelling and simulation have been applied extensively in biopharmaceutics recently. The Oral Biopharmaceutical Tools (OrBiTo) project (Innovative Medicines Initiative) aims to develop and improve upon biopharmaceutical tools, including PBPK absorption models. A large‐scale evaluation of PBPK models may be considered the first step. Here we characterise the OrBiTo active pharmaceutical ingredient (API) database for use in a large‐scale simulation study. The OrBiTo database comprised 83 APIs and 1475 study arms. The database displayed a median logP of 3.60 (2.40–4.58), human blood‐to‐plasma ratio of 0.62 (0.57–0.71), and fraction unbound in plasma of 0.05 (0.01–0.17). The database mainly consisted of basic compounds (48.19%) and Biopharmaceutics Classification System class II compounds (55.81%). Median human intravenous clearance was 16.9 L/h (interquartile range: 11.6–43.6 L/h; n = 23), volume of distribution was 80.8 L (54.5–239 L; n = 23). The majority of oral formulations were immediate release (IR: 87.6%). Human Foral displayed a median of 0.415 (0.203–0.724; n = 22) for IR formulations. The OrBiTo database was found to be largely representative of previously published datasets. 43 of the APIs were found to satisfy the minimum inclusion criteria for the simulation exercise, and many of these have significant gaps of other key parameters, which could potentially impact the interpretability of the simulation outcome. However, the OrBiTo simulation exercise represents a unique opportunity to perform a large‐scale evaluation of the PBPK approach to predicting oral biopharmaceutics. Graphical abstract Figure. No caption available.


International Journal of Pharmaceutics | 2018

The effects of three absorption-modifying critical excipients on the in vivo intestinal absorption of six model compounds in rats and dogs.

David Dahlgren; Carl Roos; P Johansson; Christer Tannergren; Anders Lundqvist; Peter Langguth; Markus Sjöblom; Erik Sjögren; Hans Lennernäs

Graphical abstract Figure. No caption available. ABSTRACT Pharmaceutical excipients that may affect gastrointestinal (GI) drug absorption are called critical pharmaceutical excipients, or absorption‐modifying excipients (AMEs) if they act by altering the integrity of the intestinal epithelial cell membrane. Some of these excipients increase intestinal permeability, and subsequently the absorption and bioavailability of the drug. This could have implications for both the assessment of bioequivalence and the efficacy of the absorption‐enhancing drug delivery system. The absorption‐enhancing effects of AMEs with different mechanisms (chitosan, sodium caprate, sodium dodecyl sulfate (SDS)) have previously been evaluated in the rat single‐pass intestinal perfusion (SPIP) model. However, it remains unclear whether these SPIP data are predictive in a more in vivo like model. The same excipients were in this study evaluated in rat and dog intraintestinal bolus models. SDS and chitosan did exert an absorption‐enhancing effect in both bolus models, but the effect was substantially lower than those observed in the rat SPIP model. This illustrates the complexity of the AME effects, and indicates that additional GI physiological factors need to be considered in their evaluation. We therefore recommend that AME evaluations obtained in transit‐independent, preclinical permeability models (e.g. Ussing, SPIP) should be verified in animal models better able to predict in vivo relevant GI effects, at multiple excipient concentrations.


Current protocols in pharmacology | 2017

The Ussing Chamber Assay to Study Drug Metabolism and Transport in the Human Intestine

Beatrice Kisser; Eva Mangelsen; Caroline Wingolf; Lars Ivo Partecke; Claus-Dieter Heidecke; Christer Tannergren; Stefan Oswald; Markus Keiser

The Ussing chamber is an old but still powerful technique originally designed to study the vectorial transport of ions through frog skin. This technique is also used to investigate the transport of chemical agents through the intestinal barrier as well as drug metabolism in enterocytes, both of which are key determinants for the bioavailability of orally administered drugs. More contemporary model systems, such as Caco‐2 cell monolayers or stably transfected cells, are more limited in their use compared to the Ussing chamber because of differences in expression rates of transporter proteins and/or metabolizing enzymes. While there are limitations to the Ussing chamber assay, the use of human intestinal tissue remains the best laboratory test for characterizing the transport and metabolism of compounds following oral administration. Detailed in this unit is a step‐by‐step protocol for preparing human intestinal tissue, for designing Ussing chamber experiments, and for analyzing and interpreting the findings.

Collaboration


Dive into the Christer Tannergren's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Leon Aarons

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge