Research paper
Utility of 96 well Caco-2 cell system for increased throughput of P-gp screening in drug discovery

https://doi.org/10.1016/j.ejpb.2004.02.014Get rights and content

Abstract

The use of Caco-2 cells for screening of discovery compounds for their permeability characteristics and P-glycoprotein interactions is well established and used routinely in pharmaceutical industries world-wide. The screening model involves growing cells on 12 or 24 well transwell format. In this manuscript, we report the use of Caco-2 cells grown on 96 well transwell plates for screening compounds for their potential to interact with P-gp. Bi-directionality studies were performed with known P-gp substrates such as saquinavir, indinavir, vinblastine, vincristine, verapamil, digoxin and taxol. P-gp inhibition studies were also conducted using radiolabeled digoxin as the probe. The results demonstrated that P-gp substrates had efflux ratios (Pc (B to A)/Pc (A to B)) in the 96 well format that were comparable to the ratios seen in 12 and 24 well format. Inhibition of digoxin efflux transport in presence of the test compounds (P-gp substrates) demonstrated that 96 well cells express adequate amounts of efflux transporters and perform as well as the 12 and 24 well Caco-2 cells. Thus, the 96 well Caco-2 cell set-up presents a higher throughput permeability model capable of identifying compounds that interact with P-gp and has the potential to significantly increase the efficiency of P-gp screening in early drug discovery.

Introduction

For a new chemical entity (NCE) to become a successful drug there are a multitude of desirable characteristics it should possess: potency to a biological target, selectivity, good stability and physico-chemical properties, minimal toxicity and adequate ADME profile. There are diverse screens that are set-up in order to select the compounds with the most favorable ‘developability’ characteristics with regards to ADME. Good permeability through intestinal membranes that can lead to adequate systemic absorption is a property highly desirable in NCEs. Various assay systems are used in early discovery to screen the permeability properties of compounds [1]. Commonly used permeability assay systems include cell-based models such as Caco-2, MDCK, LLC-PK1 cells; tissue-based models such as Ussing chamber, single pass perfusion; whole animal models such as in vivo screens; and even non-empirical in silico models. Combinatorial chemistry and other advances in synthetic chemistry have led to a tremendous inflow of discovery compounds being fed into the screens for permeability assessment. Some key characteristics desirable in a permeability screen are: high efficiency, high accuracy, time, cost and space effectiveness, and capability for high throughput. Caco-2 cells grown on 12 or 24 well transwell plates have been the staple of the pharmaceutical industry for high-throughput permeability of discovery compounds. Since P-gp is known to play a significant role in the pharmacokinetics of compounds, bi-directional studies in Caco-2 cells are routinely done for identifying compounds that interact with P-gp.

Caco-2 cells are a sturdy cell line that have been used to predict permeability and absorption of compounds in human [2], [3], [4]. Traditionally, these cells are grown on 12 and 24 wells and have been extremely successful in screening discovery compounds. However, advances in the field of molecular biology and combinatorial chemistry have shifted the bottle-neck in drug discovery from the compound synthesis step to compound selection step. There is a tremendous challenge to improve the throughput of existing screening techniques to keep up with compound synthesis. Increasingly a lot of companies have incorporated sophisticated levels of automation into these assays to make it amenable to higher throughput. The throughput is normally a staggering 100s and even 1000s of compounds per week through this screen. However, there are a large number of reasons why even further miniaturization (i.e. 96 well Caco-2) would help in such permeability assays. Apart from the obvious increase in throughput it would lead to tremendous cost reduction (by virtue of decreased cost of media, plates and buffer). One other key advantage over 12 or 24 well set-up is also that much less discovery compound would be required for 96 well to perform the same assay. This can be a significant advantage keeping in mind the hectic pace at which diverse chemotypes are synthesized in early stages of discovery.

One typical problem that has been encountered during miniaturization of any assay has been the increased variability and progressive drop in the ‘quality’ of data generated because of increased ‘quantity’. In general, the levels of expression of transporters in cell models are known to be expressed more consistently in higher surface area wells (6 or 12 well) compared to smaller wells. Since the objective of this study was to optimize the use of 96 well Caco-2 cells, it was important to demonstrate the following week over week: (1) P-gp (and other efflux transporters such as MRP, BCRP and LRP) were consistently expressed to equivalent levels and (2) cell monolayer maintained their integrity. Studies done with reference compounds (metoprolol, cimetidine, mannitol, digoxin) demonstrated that the cells were functional and maintained their utility between passage numbers 40 and 80 (data not included in the manuscript).

The use of 96 well Caco-2 cell system that can perform as well as the 12 or 24 well Caco-2 can significantly increase the productivity needed in early drug discovery. Moreover, the fact that the 96 well Caco-2 cells retains adequate as well as consistent expression of efflux transporter (i.e. P-gp) make them an attractive model for P-gp screening.

Section snippets

Materials and methods

Caco-2 cells (passage # 17) were obtained from the American Type Culture Collection (Rockville, MD). Dulbecco's modified Eagle medium, non-essential amino acids and antibiotic–antimiotic were purchased from JHR Biosciences (Lenexa, KS). Fetal bovine serum was obtained from Hyclone Lab. Inc. (Logan, Utah). HTS-Transwell® inserts (surface area: 1, 0.33 and 0.1 cm2 for 12, 24 and 96 well, respectively) with a polycarbonate membrane (0.4 μm pore size) were purchased from Costar (Cambridge, MA).

Bi-directional P-gp substrate assay results

A set of well-characterized and well-studied compounds were selected to test the validity of the P-gp substrate assay in Caco-2 cells grown on 12, 24 and 96 wells. The bi-directional permeability values for these compounds across the Caco-2 monolayers grown on 12 well format is shown in Fig. 1. Mannitol permeability values were very low (<25 nm/s) in both direction suggesting that the cells had intact monolayers and that mannitol was not interacting with any efflux or influx transporter

Discussions

P-gp is one of the most extensively studied ATP-binding cassette transporter which is known to operate as a defense mechanism by expelling toxic xenobiotics out of the system. It is a ubiquitous transporter which is present on the apical surface of the enterocytes, canalicular membrane of hepatocytes, and on the apical surface of kidney, placenta and endothelial cells of brain membrane. Due to its strategic location in some of these key tissues it is widely recognized that P-gp plays a pivotal

Acknowledgements

This work was supported by Bristol-Myers Squibb-Pharmaceutical Research Institute, Princeton. Special thanks to Melissa Yarde for providing excellent technical support with cell culture.

References (20)

There are more references available in the full text version of this article.

Cited by (86)

  • The Caco-2 Model: Modifications and enhancements to improve efficiency and predictive performance

    2022, International Journal of Pharmaceutics
    Citation Excerpt :

    The Caco-2 monolayer allows the study of all major types of absorption pathways like the passive transcellular and paracellular diffusion, carrier mediated active and facilitated transport (influx and efflux). Thus, it provides a high throughput screening platform to assess, differentiate, and classify the permeability of drug-like molecules(Balimane et al., 2004; Biganzoli et al., 1999; Marino et al., 2005), to study the effects of using novel delivery systems such as liposomes(Degim et al., 2004), microspheres(Björk et al., 1995), microemulsions(Ibrahim et al., 2020), nanospheres(Weissenboeck et al., 2004) on the permeability of drugs. Excipients that show a promise in increasing the permeability of drugs are often tested using the Caco-2 model (Chao et al., 1998; Karlsson et al., 1999; Tirumalasetty and Eley, 2005; Ward et al., 2000).

  • Interactions between artemisinin derivatives and P-glycoprotein

    2019, Phytomedicine
    Citation Excerpt :

    Regarding probe substrate bidirectional permeability assays, the most commonly used cell lines also were Caco-2 and MDCK-MDR1 cells (Chang et al., 2006; Fenner et al., 2009; Tang et al., 2002; Zhang and Morris, 2003). Digoxin and rhodamine 123 were the most commonly used probe substrates for P-gp in bidirectional permeability assays (Balimane et al., 2004; Chang et al., 2006; Taub et al., 2005; Volpe, 2011; Zhang and Morris, 2003). As shown in Fig. 4A (control), the P app (BA) of probe substrate is significantly higher than P app (AB) caused by polarized P-gp transport (Callegari et al., 2011; Senarathna et al., 2016; Taub et al., 2005; Zhang et al., 2012).

  • Establishment and characterization of an MDCK cell line stably-transfected with chicken Abcb1 encoding P-glycoprotein

    2016, Research in Veterinary Science
    Citation Excerpt :

    Studies in humans and rodents have demonstrated that P-gp is involved in the drug disposition and drug-drug interaction. Caco-2, a human colorectal adenocarcinoma cell line, has been accredited by the US FDA for in vitro transport studies (Balimane et al., 2004; Madgula et al., 2007). However, it has been observed that the differences in the pharmacokinetics of a compound may be caused by specific functions of P-gp in different species (Takeuchi et al., 2006).

  • Convallatoxin: A new P-glycoprotein substrate

    2015, European Journal of Pharmacology
View all citing articles on Scopus
View full text