Skip to the main content

Original scientific paper

https://doi.org/10.5599/admet.2023

Food and bile micelle binding of quaternary ammonium compounds

Takeru Sumiji
Kiyohiko Sugano


Full text: english pdf 461 Kb

page 409-417

downloads: 46

cite

Download JATS file


Abstract

Background and Purpose
Physiologically-based biopharmaceutics modeling (PBBM) has been widely used to predict the oral absorption of drugs. However, the prediction of food effects on oral drug absorption is still challenging, especially for negative food effects. Marked negative food effects have been reported in most cases of quaternary ammonium compounds (QAC). However, the mechanism has remained unclear. The purpose of the present study was to investigate the bile micelle and food binding of QACs as a mechanism of the negative food effect.
Experimental Approach
Trospium (TRS), propantheline (PPT), and ambenonium (AMB) were selected as model QAC drugs. The oral absorption of these QACs has been reported to be reduced by 77% (TRS), > 66% (PPT), and 79% (AMB), when taken with food. The fasted and fed state simulated intestinal fluids (FaSSIF and FeSSIF, containing 3 and 15 mM taurocholic acid, respectively) with or without FDA breakfast homogenate (BFH) were used as the simulated intestinal fluid. The unbound fraction (fu) of the QACs in these media was measured by dynamic dialysis.
Key Results
The fu ratios (FeSSIF/ FaSSIF) were 0.67 (TRS), 0.47 (PPT), and 0.76 (AMB). When BFH was added to FeSSIF, it was reduced to 0.39 (TRS), 0.28 (PPT), and 0.59 (AMB).
Conclusion
These results suggested that bile micelle and food binding play an important role in the negative food effect on the oral absorption of QACs.

Keywords

negative food effect; unbound fraction; simulated intestinal fluid; dynamic dialysis; intestinal membrane permeation

Hrčak ID:

308785

URI

https://hrcak.srce.hr/308785

Publication date:

21.9.2023.

Visits: 154 *




Introduction

Physiologically-based biopharmaceutics modeling (PBBM) has been widely used to predict the oral absorption of drugs [1]. Food effect prediction is one of the main areas where PBBM is anticipated to be useful [2-8]. However, accurate food effect prediction is still challenging [9-11]. In the fed state, the bile micelle concentration is significantly increased [12]. In addition, various food components and their digestants exist in the gastrointestinal (GI) tract in the fed state. Therefore, the interactions between a drug molecule and bile micelles/ food components can affect oral drug absorption.

A negative food effect is often observed for high solubility/ low permeability drugs (biopharmaceutical classification system (BCS) class III) [13] such as hydrophilic tertiary amines and quaternary ammonium compounds (QAC) [14]. A marked negative food effect has been observed especially in QACs [3]. In most BCS III drugs, the rate and extent of fraction dose absorbed (Fa) is limited by epithelial membrane permeation (Fa rate-limiting step (FaRLS): PL-E) [2,15]. Recently, bile micelle binding was reported to be able to quantitatively elucidate the negative food effect for hydrophilic tertiary amines [3]. On the other hand, for QACs, bile micelle binding alone was not sufficient to elucidate the marked negative food effect [3]. In addition to bile micelles, food components coexist in the fed-state intestine. However, the food binding of QACs has not been investigated.

In PBBM [1,2], the permeation flux (J) (dimension: amount/ length2/ time) is nominally expressed by the total (= unbound + bound) drug concentration dissolved in the GI fluid (Cdissolv) (dimension: amount/length3) and the effective (apparent) permeation coefficient (Peff) (dimension: length/time) as:

(1)
ADMET-11-2023-e001.jpg

In other words, Peff is defined based on the total drug concentration but not the unbound drug concentration (like plasma clearance is based on the plasma concentration in pharmacokinetics). This definition of Peff has been employed in PBBM (including commercial software). In the case of PL-E, the effect of the unstirred water layer (UWL) adjacent to the membrane is negligible. Considering that only unbound molecules can permeate the epithelial membrane (the free fraction theory),

(2)
ADMET-11-2023-e002.jpg

where fu is the unbound (free) fraction and Pep is the epithelial membrane permeation coefficient of unbound molecules (not affected by bile micelle binding). Bile micelle binding reduces the concentration of unbound drug molecules (= fu Cdissolv) at the surface of the epithelial membrane, leading to a lower flux. This is expressed as a decrease in fu and Peff in PBBM. The reduction of Peff by bile micelle binding has been reported in the literature [16-18]. The fed/fasted Fa ratio can be approximated as the ratio of the fu values in the fasted and fed states (fu, fed / fu, fasted) for Fa < 0.7 cases [2].

The purpose of the present study was to investigate the bile micelle and food binding of QACs as a mechanism of the negative food effect. Trospium (TRS), propantheline (PPT), and ambenonium (AMB) were selected as model drugs (Figure 1).

The solubility values of these drugs are high (> 100 mg mL-1) [3]. QACs are permanently positively charged, showing low organic solvent partition [19] and low artificial membrane permeation [20,21]. The Caco-2 permeability was reported to be 0.20 and 0.15×10-6 cm s-1 for TRS and AMB, respectively [3]. In fasted-state humans, the Fa values were reported to be about 13 % (TRS) and 6 % (PPT) [22-24]. Therefore, these QACs are classified as high solubility/ low permeability drugs (BCS III), and their oral absorption is limited by epithelial membrane permeation (FaRLS: PL-E). Following postprandial administration, the oral absorption of these QAC drugs is markedly reduced by 77 % (TRS), > 66 % (PPT), and 79 % (AMB), compared to those in the fasted state [25-27]. In this study, the unbound fractions (fu) of the QACs were measured by dynamic dialysis in the simulated intestinal fluids consisting of bile micelles and food homogenates [28-30]. In a bile micelle solution, bile acids exist as an equilibrium between the monomer and micellar states. When using equilibrium dialysis, bile micelle monomers can diffuse through the dialysis membrane pore and form micelles on the opposite side after equilibrium. Therefore, instead of equilibrium dialysis, dynamic dialysis has been used to determine the micelle binding of drugs.

Experimental

Material

Trospium chloride and caffeine were purchased from Tokyo Chemical Industry Co., Ltd (Tokyo, Japan). Sodium chloride (NaCl), sodium dihydrogen phosphate dihydrate (NaH2PO4 2H2O), 8N NaOH, taurocholic acid (TC), and oleic acid (OA) were purchased from FUJIFILM Wako Pure Chemical Corporation (Osaka, Japan). Propantheline bromide was purchased from Funakoshi Co., Ltd (Tokyo, Japan). Ambenonium dichloride was purchased from Toronto Research Chemicals (Ontario, Canada). Egg yolk lecithin (EL) was purchased from NOF Corporation (Tokyo, Japan). Glyceryl mono-oleate (GM) was purchased from Nippon Surfactant Industries Co., Ltd (Tokyo, Japan). A cellulose dialysis membrane was purchased from As-One Corporation (Osaka, Japan).

The Food and Drug Adminstration (FDA) breakfast ingredients were purchased from the local market (bacon (Itoham Foods Inc, Japan), toast (Choujuku bread, Pasco Shikishima Corporation, Japan), egg (Odama mix, I.T.S Farm Co., Ltd, Japan), hash browns (Hoshino potato, Heinz Japan Ltd, Japan), whole milk (Oishii Megumilk Snow Brand Milk, MEGMILK SNOW BRAND Co., Ltd, Japan), and butter (Hokkaido-butter, MEGMILK SNOW BRAND Co., Ltd, Japan)).

Methods

Processing of FDA Breakfast

The FDA breakfast consisted of one strip of bacon, half a slice of toast, one fried egg, 55 g of hash browns, 30 g of butter, and 100 mL of whole milk. After cooking, the FDA breakfast was homogenized for 15 seconds by a food processor. Before use, the FDA breakfast homogenate (BFH) was divided into small aliquots and stored in a freezer (-30 °C).

Measurement of the unbound fraction by dynamic dialysis

Dynamic dialysis was performed by using a side-by-side chamber (SANPLATEC Co., Ltd (Osaka, Japan)) with a cellulose dialysis membrane (molecular weight cut-off: 3500). The membrane area was 2.0 cm2. The fluid volume was 1.5 mL on both the donor and acceptor sides. The fasted and fed state simulated intestinal fluids (FaSSIF and FeSSIF, respectively) with or without BFH were used as test media (Table 1) [31]. A test medium containing 0.5 mM of each QAC was added to the donor side and blank FaSSIF was added to the acceptor side. After incubation at 37 °C for 1 h, the concentration of the QAC in the acceptor side was determined by HPLC (Shimazu Prominence LC-20 series, column: Zorbax Eclipse Plus (C18 2.1×50 mm, 3.5 μm) (Agilent Technologies), flow rate: 0.6 mL min-1, mobile phase: 0.1 % trifluoroacetic acid acetonitrile/ /water (25 % (TRS), 35 % (PPT), 20 % (AMB), and 10 % (caffeine)), detection: UV (260 nm (TRS), 280 nm (PPT), 265 nm (AMB), and 275 nm (caffeine)), column temperature: 40 °C, and injection volume of 10 μL).

Permeated% was calculated as the ratio of the concentration in the acceptor chamber at 1 h and the theoretical equilibrium concentration (0.25 mM) (× 100 to convert to %). The unbound fraction (fu) was calculated as the ratio of permeated% in each medium and blank FaSSIF.

Results

Figure 2 shows the fu values of each QAC. Permeated% at 1h, fu, and the fed/fasted fu ratio are summarized inTable 2. The permeated% and the fu value of QACs were decreased by bile micelles and BFH, and more significantly decreased when bile micelles and BFH coexisted (decrease by 44 to 74 %). Caffeine was used as a reference un-ionized drug. Permeated% was slightly decreased (by 16 %) in the FeSSIF+BFH condition.

Discussion

In this study, the bile micelles decreased the fu values of the QACs in good agreement with the previous study using the Caco-2 membrane [3]. The addition of BFH to FeSSIF resulted in a more significant decrease in the fu values, comparable to the clinically observed negative food effects on the oral absorption of trospium and propantheline (approximately 70 % reduction). This result suggests that the reduction of fu in the fed state is likely to be at least one of the main reasons for the negative food effect on the oral absorption of QACs.

The addition of BFH increases the viscosity of the simulated intestinal fluid. The fluid viscosity could affect the results of dynamic dialysis. To investigate this point, caffeine was used as a control compound. Caffeine is a hydrophilic, non-ionized drug that is unlikely to bind to bile micelles or food components. Therefore, permeated% of caffeine would be mainly affected by fluid viscosity. The permeated% of caffeine in FeSSIF + BFH was reduced markedly less than that of QACs. Therefore, the permeated% of the QACs was mainly affected by the food/bile micelle binding, rather than an increase in fluid viscosity.

BFH contains various components, such as lipids and organic acids. QAC is a cationic compound with lipophilic hydrocarbon moieties (Figure 1). Therefore, both electrostatic and hydrophobic interactions may be formed between a QAC and the components in the FeSSIF+BFH medium. We also investigated the effect of crude bile (ox-bile) on the fu value, however, little or no difference was observed between ox-bile and TC (data not shown). Further investigation is required to clarify molecular-level interactions between bile micelles/food and QAC.

Previously, the negative food effect on the oral absorption of trospium has been extensively investigated [25]. The disintegration time and dissolution rate of trospium formulations were reduced due to the increase in fluid viscosity caused by food. It was suggested that this was one of the reasons for the negative food effect [32]. Because trospium is mainly absorbed from the upper small intestine [33], a slowed disintegration time would reduce its absorption [34]. However, a marked negative food effect was also observed when trospium was administered as an oral solution [25]. In addition, if an increase in fluid viscosity were the mechanism for the marked negative food effect, it would have been observed in a wide range of drug products. However, this is not the case. Therefore, an increase in fluid viscosity may not be the main reason for the marked negative food effect on the oral absorption of trospium. In another report, the reduction of ion-pair transport of trospium with a bile acid anion was also suggested as the mechanism of the negative food effect [35]. In that report, it was suggested that bile acids increased the permeation of trospium by ion-pair transport in the fasted state. In the presence of dietary lipids in the fed state, the bile micelle concentration available for ion-pair transport was suggested to be reduced because bile acids are consumed for lipid solubilization. However, an increase in trospium permeability by bile micelles was not statistically significant in Caco-2 [35] and was slightly observed only after 60 min incubation in the rat small intestine [35]. Similarly, in our previous study using Caco-2 cells, bile micelles did not increase the apparent permeability of trospium [3]. Taken together, the results of the present study suggested that the reduction of the unbound fraction by food/bile micelle binding is more likely to be the reason for the negative food effect observed for trospium. This mechanism can also explain the reason that trospium is absorbed from two intestinal absorption windows [25]. A small portion of trospium is first absorbed before binding to food/bile micelles. Food/bile micelle binding would then occur and reduce the membrane flux of trospium. Over time, the food components are digested and absorbed along the GI tract. In addition, bile acids are re-absorbed in the lower part of the small intestine by the site-specific bile acid transporter. Therefore, trospium is released as it transits along the GI tract, resulting in bimodal plasma-concentration time profiles [16,36].

Together with the previous studies [2,3], this study suggested that the fu values in the fasted and fed states are required for PBBM to predict the negative food effect. The FeSSIF+BFH medium can be a useful simulated medium to measure the fu value in vitro. It should be noted that even for high permeability/low solubility drugs (BCS II), the Peff values differ between fasted and fed conditions. BCS II roughly corresponds to solubility- UWL permeation limited absorption (FaRLS: SL-U)). Bile micelle binding reduces the effective diffusion coefficient of a drug in the UWL adjacent to the intestinal wall [2,8]. Therefore, the fu values in the fasted and fed states are required for the quantitative food effects prediction for SL-U (in this case, an increase in Cdissolv by bile micelles is slightly canceled out by a reduction in Peff, resulting in a positive food effect [2]). Furthermore, the fu value is critical to elucidate the solubility-permeability trade-off for low solubility/low permeability drugs (BCS IV, roughly corresponds to solubility- epithelial membrane permeation limited absorption (FaRLS: SL-E)) [2]. A simple empirical extrapolation from apparent Caco-2 permeability data (Papp) like log Peff = A log Papp + B should not be used for the food effect prediction by PBBM because the effect of food/ bile micelle binding on Peff cannot be considered in this method [37].

For PBBM to be used to waive a clinical food effect study, accurate food effect prediction is required. For diagnosing the bioequivalence between the fasted and fed states, the prediction error must be less than 20 %. To achieve this goal, further investigation on food and bile micelle binding is required. Food digestion would change the chemical composition of the digest. The effect of food digestion on the fu value would be of great interest and should be investigated in the future.

The fu measurement of high solubility/low permeability drugs has been challenging. The Caco-2 assay has previously been used for the fu measurement [3]. However, Caco-2 cells may be damaged by food components. In the case of transcellular permeation, parallel artificial membrane permeation assay (PAMPA) may be used to assess the food and bile micelle binding of a drug [38,39]. However, because QACs are permanently charged, they are absorbed mainly via the paracellular route [40]. In such cases, dynamic dialysis would be a good tool to assess the food and bile micelle binding of a drug.

Conclusions

In conclusion, the results of this study suggested that the reduction of fu in the fed state is likely to be the reason for the negative food effect on the oral absorption of QACs. The FeSSIF+BFH medium can be useful for assessing the food and bile micelle binding of a drug. Dynamic dialysis would be a good tool to measure a fu value in a medium containing bile micelles.

Notes

[1] Conflicts of interest Conflict of interest: The Author(s) declare(s) that they have no conflicts of interest to disclose.

[2] Author Contributions: Material preparation, data collection and analysis were performed by Takeru Sumiji. Kiyohiko Sugano supervised all phases of the study, including the manuscript writing. All authors read and approved the final manuscript.

[3] Data availability: The datasets generated during and/or analyzed during the current study are available from the corresponding author on request.

References

[1] 

Sugano K.. Biopharmaceutics modeling and simulations: theory, practice, methods, and applications. John Wiley & Sons, Hoboken, New Jersey, USA, 2012. https://doi.org/10.1002/9781118354339.fmatter. https://doi.org/10.1002/9781118354339.fmatter

[2] 

Sugano K.; Kataoka M.; Mathews C.C.; Yamashita S.. Prediction of food effect by bile micelles on oral drug absorption considering free fraction in intestinal fluid. European Journal of Pharmaceutical Sciences 40 (2010) 118-124. https://doi.org/10.1016/j.ejps.2010.03.011. https://doi.org/10.1016/j.ejps.2010.03.011

[3] 

Akiyama Y.; Ito S.; Fujita T.; Sugano K.. Prediction of negative food effect induced by bile micelle binding on oral absorption of hydrophilic cationic drugs. European Journal of Pharmaceutical Sciences 155 (2020) 105543. https://doi.org/10.1016/j.ejps.2020.105543. https://doi.org/10.1016/j.ejps.2020.105543

[4] 

Riedmaier A.E.. Predicting Food Effects: Are We There Yet. The AAPS Journal 24 (2022) 25. https://doi.org/10.1208/s12248-021-00674-x. https://doi.org/10.1208/s12248-021-00674-x

[5] 

Kesisoglou F.. Can PBPK Modeling Streamline Food Effect Assessments. The Journal of Clinical Pharmacology 60(Suppl 1) (2020) S98-S104. https://doi.org/10.1002/jcph.1678. https://doi.org/10.1002/jcph.1678

[6] 

Pepin X.J.H.; Huckle J.E.; Alluri R.V.; Basu S.; Dodd S.; Parrott N.; Riedmaier A.E.. Understanding Mechanisms of Food Effect and Developing Reliable PBPK Models Using a Middle-out Approach. The AAPS Journal 23 (2021) 12. https://doi.org/10.1208/s12248-020-00548-8. https://doi.org/10.1208/s12248-020-00548-8

[7] 

Riedmaier A.E.; 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 C.C.; 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. The AAPS Journal 22 (2020) 123. https://doi.org/10.1208/s12248-020-00508-2. https://doi.org/10.1208/s12248-020-00508-2

[8] 

Ramasubramanian H.; Castleberry S.. Biopharmaceutical Modeling of Food Effect–Exploring the Role of Dietary Fat. Molecular Pharmaceutics 20 (2023) 2726-2737. https://doi.org/10.1021/acs.molpharmaceut.3c00170. https://doi.org/10.1021/acs.molpharmaceut.3c00170

[9] 

Cheng L.; Wong H.. Food Effects on Oral Drug Absorption: Application of Physiologically-Based Pharmacokinetic Modeling as a Predictive Tool. Pharmaceutics 12 (2020) 672. https://doi.org/10.3390/pharmaceutics12070672. https://doi.org/10.3390/pharmaceutics12070672

[10] 

Li M.; Zhao P.; Pan Y.; Wagner C.. Predictive Performance of Physiologically Based Pharmacokinetic Models for the Effect of Food on Oral Drug Absorption: Current Status. CPT: Pharmacometrics & Systems Pharmacology 7 (2018) 82-89. https://doi.org/10.1002/psp4.12260. https://doi.org/10.1002/psp4.12260

[11] 

Vinarov Z.; Abrahamsson B.; Artursson P.; Batchelor H.; Berben P.; Bernkop-Schnürch A.; Butler J.; Ceulemans J.; Davies N.; Dupont D.; Flaten G.E.; Fotaki N.; Griffin B.T.; Jannin V.; Keemink J.; Kesisoglou F.; Koziolek M.; Kuentz M.; Mackie A.; Meléndez-Martínez A.J.; McAllister M.; Müllertz A.; O'Driscoll C.M.; Parrott N.; Paszkowska J.; Pavek P.; Porter C.J.H.; Reppas C.; Stillhart C.; Sugano K.; Toader E.; Valentová K.; Vertzoni M.; De Wildt S.N.; Wilson C.G.; Augustijns P.. Current challenges and future perspectives in oral absorption research: An opinion of the UNGAP network. Advanced Drug Delivery Reviews 171 (2021) 289-331. https://doi.org/10.1016/j.addr.2021.02.001. https://doi.org/10.1016/j.addr.2021.02.001

[12] 

Kambayashi Y. Shirasaka. Food effects on gastrointestinal physiology and drug absorption. Drug Metabolism and Pharmacokinetics 48 (2023) 100488. https://doi.org/10.1016/j.dmpk.2022.100488. https://doi.org/10.1016/j.dmpk.2022.100488

[13] 

Yu L.X.; Amidon G.L.; Polli J.E.; Zhao H.; Mehta M.U.; Conner D.P.; Shah V.P.; Lesko L.J.; Chen M.L.; Lee V.H.; Hussain A.S.. Biopharmaceutics classification system: the scientific basis for biowaiver extensions. Pharmaceutical Research 19 (2002) 921-925. https://doi.org/10.1023/a:1016473601633. https://doi.org/10.1023/a:1016473601633

[14] 

Singh B.N.. A quantitative approach to probe the dependence and correlation of food-effect with aqueous solubility, dose/solubility ratio, and partition coefficient (Log P) for orally active drugs administered as immediate-release formulations. Drug Development Research 65 (2005) 55-75. https://doi.org/10.1002/ddr.20008. https://doi.org/10.1002/ddr.20008

[15] 

Sugano K.; Terada K.. Rate- and Extent-Limiting Factors of Oral Drug Absorption: Theory and Applications. Journal of Pharmaceutical Sciences 104 (2015) 2777-2788. https://doi.org/10.1002/jps.24391. https://doi.org/10.1002/jps.24391

[16] 

Lennernäs H.; Regårdh C.G.. Evidence for an interaction between the beta-blocker pafenolol and bile salts in the intestinal lumen of the rat leading to dose-dependent oral absorption and double peaks in the plasma concentration-time profile. Pharmaceutical Research 10 (1993) 879-883. https://doi.org/10.1023/a:1018965328626. https://doi.org/10.1023/a:1018965328626

[17] 

Yamaguchi T.; Ikeda C.; Sekine Y.. Intestinal absorption of a beta-adrenergic blocking agents nadolol. II. Mechanism of the inhibitory effect on the intestinal absorption of nadolol by sodium cholate in rats. Chemical and Pharmaceutical Bulletin (Tokyo) 34 (1986) 3836-3843. https://doi.org/10.1248/cpb.34.3836. https://doi.org/10.1248/cpb.34.3836

[18] 

Ingels F.; Beck B.; Oth M.; Augustijns P.. Effect of simulated intestinal fluid on drug permeability estimation across Caco-2 monolayers. International Journal of Pharmaceutics 274 (2004) 221-232. https://doi.org/10.1016/j.ijpharm.2004.01.014. https://doi.org/10.1016/j.ijpharm.2004.01.014

[19] 

Bouchard G.; Carrupt P.A.; Testa B.; Gobry V.; Girault H.H.. The apparent lipophilicity of quaternary ammonium ions is influenced by galvani potential difference, not ion-pairing: a cyclic voltammetry study. Pharmaceutical Research 18 (2001) 702-708. https://doi.org/10.1023/A:1011001914685. https://doi.org/10.1023/A:1011001914685

[20] 

Sugano K.; Nabuchi Y.; Machida M.; Asoh Y.. Permeation characteristics of a hydrophilic basic compound across a bio-mimetic artificial membrane. International Journal of Pharmaceutics 275 (2004) 271-278. https://doi.org/10.1016/j.ijpharm.2004.02.010. https://doi.org/10.1016/j.ijpharm.2004.02.010

[21] 

Fischer H.; Kansy M.; Avdeef A.; Senner F.. Permeation of permanently positive charged molecules through artificial membranes--influence of physico-chemical properties. European Journal of Pharmaceutical Sciences 31 (2007) 32-42. https://doi.org/10.1016/j.ejps.2007.02.001. https://doi.org/10.1016/j.ejps.2007.02.001

[22] 

Langguth P.; Kubis A.; Krumbiegel G.; Lang W.; Merkle H.P.; Wächter W.; Spahn-Langguth H.; Weyhenmeyer R.. Intestinal absorption of the quaternary trospium chloride: permeability-lowering factors and bioavailabilities for oral dosage forms. European Journal of Pharmaceutics and Biopharmaceutics 43 (1997) 265-272. https://doi.org/10.1016/S0939-6411(97)00050-7. https://doi.org/10.1016/S0939-6411(97)00050-7

[23] 

Doroshyenko O.; Jetter A.; Odenthal K.P.; Fuhr U.. Clinical pharmacokinetics of trospium chloride. Clinical Pharmacokinetics 44 (2005) 701-720. https://doi.org/10.2165/00003088-200544070-00003. https://doi.org/10.2165/00003088-200544070-00003

[24] 

Vose C.W.; Ford G.C.; Grigson S.J.; Haskins N.J.; Prout M.; Stevens P.M.; Rose D.A.; Palmer R.F.; Rudel H.. Pharmacokinetics of propantheline bromide in normal man. British Journal of Clinical Pharmacology 7 (1979) 89-93. https://doi.org/10.1111/j.1365-2125.1979.tb00902.x. https://doi.org/10.1111/j.1365-2125.1979.tb00902.x

[25] 

Tadken T.; Weiss M.; Modess C.; Wegner D.; Roustom T.; Neumeister C.; Schwantes U.; Schulz H.U.; Weitschies W.; Siegmund W.. Trospium chloride is absorbed from two intestinal "absorption windows" with different permeability in healthy subjects. International Journal of Pharmaceutics 515 (2016) 367-373. https://doi.org/10.1016/j.ijpharm.2016.10.030. https://doi.org/10.1016/j.ijpharm.2016.10.030

[26] 

Moses D.K.; Charles B.G.; Ravenscroft P.J.; Whyte I.M.. Food reduces the oral bioavailability of propantheline bromide in healthy subjects. British Journal of Clinical Pharmacology 16 (1983) 758-759. https://doi.org/10.1111/j.1365-2125.1983.tb02261.x. https://doi.org/10.1111/j.1365-2125.1983.tb02261.x

[27] 

Ohtsubo K.; Fujii N.; Higuchi S.; Aoyama T.; Goto I.; Tatsuhara T.. Influence of food on serum ambenonium concentration in patients with myasthenia gravis. European Journal of Clinical Pharmacology 42 (1992) 371-374. https://doi.org/10.1007/BF00280120. https://doi.org/10.1007/BF00280120

[28] 

Modi S.; Anderson B.D.. Determination of drug release kinetics from nanoparticles: overcoming pitfalls of the dynamic dialysis method. Molecular Pharmaceutics 10 (2013) 3076-3089. https://doi.org/10.1021/mp400154a. https://doi.org/10.1021/mp400154a

[29] 

Guan P.; Lu Y.; Qi J.; Niu M.; Lian R.; Hu F.; Wu W.. Enhanced oral bioavailability of cyclosporine A by liposomes containing a bile salt. International Journal of Nanomedicine 6 (2011) 965-974. https://doi.org/10.2147/IJN.S19259. https://doi.org/10.2147/IJN.S19259

[30] 

Yano K.; Masaoka Y.; Kataoka M.; Sakuma S.; Yamashita S.. Mechanisms of membrane transport of poorly soluble drugs: role of micelles in oral absorption processes. Journal of Pharmaceutical Sciences 99 (2010) 1336-1345. https://doi.org/10.1002/jps.21919. https://doi.org/10.1002/jps.21919

[31] 

Silva D.A.; Davies N.M.; Bou-Chacra N.; Ferraz H.G.; Löbenberg R.. Update on Gastrointestinal Biorelevant Media and Physiologically Relevant Dissolution Conditions. Dissolution Technology 29 (2022) 62-75. https://doi.org/10.14227/DT290222P62. https://doi.org/10.14227/DT290222P62

[32] 

Radwan A.; Amidon G.L.; Langguth P.. Mechanistic investigation of food effect on disintegration and dissolution of BCS class III compound solid formulations: the importance of viscosity. Biopharmaceutics & Drug Disposition 33 (2012) 403-416. https://doi.org/10.1002/bdd.1798. https://doi.org/10.1002/bdd.1798

[33] 

Schröder S.; Jetter A.; Zaigler M.; Weyhenmeyer R.; Krumbiegel G.; Wächter W.; Fuhr U.. Absorption pattern of trospium chloride along the human gastrointestinal tract assessed using local enteral administration. Internatinal Journal of Clinical Pharmacology and Therapeutics 42 (2004) 543-549. https://doi.org/10.5414/cpp42543. https://doi.org/10.5414/cpp42543

[34] 

Cvijić S.; Langguth P.. Improvement of trospium-specific absorption models for fasted and fed states in humans. Biopharmaceutics & Drug Disposition 35 (2014) 553-558. https://doi.org/10.1002/bdd.1911. https://doi.org/10.1002/bdd.1911

[35] 

Heinen C.A.; Reuss S.; Amidon G.L.; Langguth P.. Ion pairing with bile salts modulates intestinal permeability and contributes to food-drug interaction of BCS class III compound trospium chloride. Molecular Pharmaceutics 10 (2013) 3989-3996. https://doi.org/10.1021/mp400179v. https://doi.org/10.1021/mp400179v

[36] 

Davies N.M.; Takemoto J.K.; Brocks D.R.; Yáñez J.A.. Multiple peaking phenomena in pharmacokinetic disposition. Clinical Pharmacokinetics 49 (2010) 351-377. https://doi.org/10.2165/11319320-000000000-00000. https://doi.org/10.2165/11319320-000000000-00000

[37] 

Sugano K.. Lost in modelling and simulation. ADMET & DMPK 9 (2021) 75-109. https://doi.org/10.5599/admet.923. https://doi.org/10.5599/admet.923

[38] 

Kansy M.; Senner F.; Gubernator K.. Physicochemical high throughput screening: parallel artificial membrane permeation assay in the description of passive absorption processes. Journal of Medicinal Chemistry 41 (1998) 1007-1010. https://doi.org/10.1021/jm970530e. https://doi.org/10.1021/jm970530e

[39] 

Avdeef A.; Bendels S.; Di L.; Faller B.; Kansy M.; Sugano K.; Yamauchi Y.. PAMPA--critical factors for better predictions of absorption. Journal of Pharmaceutical Sciences 96 (2007) 2893-2909. https://doi.org/10.1002/jps.21068. https://doi.org/10.1002/jps.21068

[40] 

Sugano K.; Takata N.; Machida M.; Saitoh K.; Terada K.. Prediction of passive intestinal absorption using bio-mimetic artificial membrane permeation assay and the paracellular pathway model. International Journal of Pharmaceutics 24 (2002) 241-251. https://doi.org/10.1016/s0378-5173(02)00240-5. https://doi.org/10.1016/s0378-5173(02)00240-5

Floating objects

Figure 1. Chemical structures of model QAC drugs
ADMET-11-2023-g001.jpg
Figure 2. Effect of bile micelles and FDA breakfast homogenate (BFH) on the fu values of QACs. (A) Trospium, (B) propantheline, and (C) ambenonium.
ADMET-11-2023-g002.jpg
Table 1. Composition of test media*
Test mediumTC, mMEL, mMGM, mMOA, mMBFH, wt.%
FaSSIF30.75000
×0.5 FeSSIF7.51.88000
FeSSIF153.75000
×2FeSSIF307.50000
FeSSIFv2102.050.80
BFH added to blank FaSSIF000050
FeSSIF+BFH153.750050

[i] *The components were dissolved in a phosphate buffer (phosphate 28.6 mM, NaCl 106 mM, pH 6.5; blank FaSSIF); QAC: 0.5 mM

Table 2. Permeated% and fu
DrugTest mediumPermeated% at 1 h*fu*fu ratio*,**
TrospiumBlank FaSSIF21.9 ± 0.1--
FaSSIF21.4 ± 1.00.98 ± 0.06-
×0.5 FeSSIF21.1 ± 0.50.96 ± 0.030.99 ± 0.07
FeSSIF14.4 ± 0.60.66 ± 0.030.67 ± 0.02
×2 FeSSIF13.0 ± 2.70.59 ± 0.150.61 ± 0.14
FeSSIF-V216.8 ± 0.60.77 ± 0.030.79 ± 0.01
BFH12.2 ± 1.00.56 ± 0.060.57 ± 0.03
FeSSIF + BFH8.3 ± 0.30.38 ± 0.020.39 ± 0.02
PropanthelineBlank FaSSIF25.6 ± 0.2--
FaSSIF23.2 ± 0.30.90 ± 0.02-
×0.5 FeSSIF16.8 ± 1.50.66 ± 0.070.73 ± 0.08
FeSSIF10.9 ± 0.20.42 ± 0.010.47 ± 0.02
×2 FeSSIF6.1 ± 0.10.24 ± 0.010.26 ± 0.01
FeSSIF-V29.8 ± 0.30.38 ± 0.010.42 ± 0.01
BFH13.0 ± 1.00.51 ± 0.000.56 ± 0.05
FeSSIF + BFH6.6 ± 0.60.26 ± 0.000.28 ± 0.03
AmbenoniumBlank FaSSIF17.5 ± 1.0--
FaSSIF16.8 ± 0.20.96 ± 0.02-
×0.5 FeSSIF16.3 ± 1.00.93 ± 0.070.97 ± 0.06
FeSSIF12.8 ± 0.40.73 ± 0.030.76 ± 0.02
×2 FeSSIF8.7 ± 0.40.49 ± 0.030.52 ± 0.03
FeSSIF-V212.1 ± 0.60.69 ± 0.040.72 ± 0.03
BFH14.0 ± 0.30.80 ± 0.020.84 ± 0.01
FeSSIF + BFH9.8 ± 0.10.56 ± 0.010.59 ± 0.02
CaffeineBlank FaSSIF40.1 ± 0.7-
BFH36.4 ± 2.80.91 ± 0.09-
FeSSIF + BFH33.9 ± 0.80.84 ± 0.03-

[i] *mean ± S.D., n = 3

[ii] **vs. the fu value in FaSSIF


This display is generated from NISO JATS XML with jats-html.xsl. The XSLT engine is libxslt.