1. Introduction
In the small molecule chemical space within Lipinski’s rules of five (Ro5) [
1], compounds exhibit a high range of solubility/permeability, which can be correlated reasonably well to the fraction absorbed (f
a). For cellular permeability (P
app), this correlation represents a sigmoidal curve, where highly absorbed compounds mostly demonstrate high permeability [
2]. However, for compounds with a P
app < 10 × 10
−6 cm/s, the accuracy to predict f
a decreases, and for permeabilities < 1 × 10
−6 cm/s, traditional cellular P
app becomes merely a qualitative marker (low permeability = likely poorly absorbed). Unfortunately, the majority of more complex small molecule drugs beyond the rule of five (bRo5), such as proteolysis-targeting chimeras (PROTACs), fall within this range [
3]. Besides a lack in predictability, traditional cellular permeability assays are often not able to generate a permeability value, because of low detection sensitivity due to the very low permeability, as well as nonspecific binding, of the compound to the incubation setup.
Novel in silico-based and partitioning-based approaches, such as AB-MPS-score [
4], Lipophilic Permeability Efficiency (LPE) [
5], ChameLogD, Experimental Polar Surface Area (EPSA) [
6], Chamelogk [
7], and the EPSA-to-TPSA Ratio (ETR) [
8], provide high-throughput support for bRo5 projects, especially at early stages [
9].
These strategies either address multiple physicochemical parameters in parallel or consider the conformational change of a compound to reduce polarity during membrane passage, often coined “chameleonicity”. Such polarity-reducing effects can lead to a better permeation and absorption of bulky molecules [
8].
Although these models provide a physicochemical representation of permeability for early compound ranking and absorption prediction, they overlook the complexities of biological behavior caused by para- and transcellular transport, compartmental binding, or active transport, and may need a cellular system to better predict permeability and absorption. As the standard cellular transwell
® approach is not suitable for compounds with very low permeability and unfavorable physicochemical properties, previous studies aimed to modify the setup to enable better compound recovery and sensitivity. These approaches included the use of bovine serum albumin (BSA) in the transport medium [
10] or the prolongation of the incubation time within the assay. To mirror the physiological lag time of slowly permeating compounds, pre-incubation steps can assure the measurement of permeability at steady state. These improvements have been shown to better characterize the transport of bRo5 compounds [
11].
The goal of the present study was to identify the most suitable conditions for a simple cell-based assay that provides superior performance with respect to the characterization of permeability, the modeling of gut absorption, and to improve overall data quality and reproducibility.
Therefore, we investigated the relevance of a pre-incubation step, optimized LC-MS/MS analytics, and the addition of BSA for the measurement of permeability of complex chemical entities. To verify the suitability of the setup, we analyzed 61 known reference compounds with available human fa. Lastly, we measured the permeability and efflux of 741 internal compounds, many of which violated two or more Lipinski rules (bRo5) and compared the results to rodent absorption.
2. Materials and Methods
2.1. Materials
Unless stated otherwise, all compounds including inhibitors were obtained internally from AbbVie chemical substance management system (Ludwigshafen, Germany). Assay-ready Caco-2 cells were purchased from acCELLerate (Hamburg, Germany). Buffers and cell culture reagents, e.g., Hank’s Balanced Salt Solution (HBSS), Dulbecco’s Phosphate Buffered Saline (DPBS), trypsin/EDTA, Dulbecco’s Modified Eagle Medium (DMEM), sodium pyruvate, L-glutamine, non-essential amino acids (NEA), fetal bovine serum (FBS), and bovine serum albumin (BSA) were all purchased from Thermo Fisher Scientific (St. Leon-Rot, Germany). All plasticware was internally available in AbbVie.
2.2. Cell Culture and Plate Preparation
Frozen assay-ready Caco-2 cells [
12] in cryovials were taken from liquid nitrogen storage and thawed in a water bath at 37 °C for 2 min. The cells were transferred into a 50 mL Falcon tube and 8 mL of prewarmed cell culture medium (DMEM supplemented with 10% FBS, 1% NEA, 1% L-glutamine, and 1% sodium pyruvate) was added. The cell suspension was centrifugated at 80×
g for 4 min and the supernatant cautiously aspirated. The cell pellet was resuspended in 10 mL of fresh cell culture medium and mixed gently. After equilibration for 30 min at room temperature, the cells were seeded into transwell plates.
The cells were seeded into 0.4 µm Millicell® 96-well transwell plates (Merck Millipore, Darmstadt, Germany), comprising the insert plate and the feeder plate. The insert plate was seeded with 100 µL/well cell suspension containing 40,000 cells. The feeder tray was filled with 28 mL of medium. Unless stated otherwise, Caco-2 cell monolayers were grown for 7–8 days at 37 °C with 5% CO2. Medium was changed on the basolateral side 4–5 days after seeding and apically one day before the experiments to prevent cell starvation. The plate inserts were then transferred into 96-well receiver plates (Merck Millipore, Darmstadt, Germany) filled with 200 µL/well fresh culture medium.
2.3. Bidirectional Transport Studies
If not described differently, compounds were dissolved in dimethyl sulfoxide (DMSO) to 10 mM stock solutions and further diluted in HBSS at pH 7.4 containing the monolayer-integrity marker lucifer yellow (80 µM final conc.) to the required concentrations. Depending on the assay modifications, 1% (w/v) BSA was added to the HBSS buffer. Final DMSO concentrations were max 0.2% (v/v).
Before the assay, culture medium was removed, and the cells were rinsed with HBSS (pH 7.4) once. Compound solutions (1 µM or 3 µM) were added to donor compartments, i.e., apical side of A-to-B transport direction and basolateral side of B-to-A direction. The receiver compartments were filled with corresponding receiver buffer (HBSS pH 7.4 with or without 1% BSA). Compound permeability was evaluated after 60 min incubation at 37 °C, when samples were collected from both apical and basolateral sides of both transwell plates, and then mixed with a quench solution (30% acetonitrile in water or 100% ethanol, containing 25 nM carbutamide as internal LC-MS/MS process control) before measurement using LC-MS/MS. When conducting a pre-incubation step, compound solutions were added to donor compartments and the receiver compartments were filled with corresponding receiver buffer (HBSS pH 7.4 with or without 1% BSA). Unless stated otherwise, the pre-incubation solution was removed after 60–90 min. The cells were then rinsed with HBSS with 1% BSA, and new compound solution (donor compartments) and receiver buffer (receiver compartments) were added for the main incubation (60 min) with subsequent sampling for permeability assessment. To determine compound apparent permeability (P
app), the following equation was used:
where ΔQ is the amount of compound permeated through the monolayer as determined by the response (=peak area) of compound in the receiver well at the end of the experiment, Δt is the incubation time in s, A is the filter surface area (0.11 cm
2), C
1 is the response in the donor well at the end of the experiment, and C
0 is the initial nominal compound concentration. Permeation velocity is expressed as P
app with the unit of 10
−6 cm/s.
Efflux ratio (ER) was calculated as follows:
where P
app,AB and P
app,
BA refer to the mean permeability of two replicates in the direction of apical to basolateral (A-to-B) or basolateral to apical (B-to-A), respectively.
Recovery (%) was determined using the following equation:
C
Acceptor and C
Donor are the responses of compound determined in the acceptor and donor well at the end of the incubation time, respectively. C
0 is the initial nominal compound concentration.
All studies were performed at least in technical duplicates. Mean values were used for final data analysis.
2.4. Analytical Method
All measurements were performed using an Acquity Iclass UPLC (Waters, Milford, MA, USA) system coupled with a tandem mass spectrometer Sciex 6500 (Sciex, Framingham, MA, USA) operating dependent on the tested compounds in positive or negative mode. Chromatographic separation of a 1–7.5 µL injected sample was achieved with a BEH C18 column (2.1 mm × 30 mm, 1.7 μm) kept at 60 °C. The total run time was a linear 1.1 min gradient starting for most tested compounds with 95% water (mobile phase A) and 5% acetonitrile (mobile phase B), both acidified with 0.1% formic acid and peaking with 95% mobile phase B. Optimized mass transitions were used to detect incubated substances. Acquisition and analysis of the data were performed using Analyst 1.7.2 and Discovery Quant 3.0.1 (Sciex). Variability within and across assays was assessed using carbutamide as an analytical process control substance. Results were reported as peak area (response). For compound optimization, a 40 nM test solution was injected and optimized in terms of chromatographic separation, ionization, and fragmentation. A signal-to-noise ratio of at least 3 compared to blank injection was considered as appropriate. The analytical threshold for detection was considered sufficient when a minimum peak area in the donor compartment to enable a Papp of 10−6 cm/s (or 10−7 cm/s) was theoretically achievable. This translated to a peak area threshold of at least 300,000 or 3,000,000, respectively, for the 40 nM test solution, assuming comparable matrix conditions and extraction efficiencies in the donor and acceptor.
During compound optimization, a series of alternative chromatographic separations were tested and used if binding (fronting) or retention (tailing) was reduced or signal-to-noise for a 40 nM test solution increased, compared to the standard method described above. The adaptions contained but were not restricted to different column chemistry (Acquity Xbridge C8 (Waters, 2.1 mm × 30 mm, 3.5 µm)) or the replacement of mobile phase A with 25 mM ammonium bicarbonate supplemented with 25 mM ammonium hydroxide. For tacrolimus, chromatography was a linear 1.1 min gradient starting with 50% 10 mM ammonium formate (mobile phase A) and 50% acetonitrile (mobile phase B), both acidified with 0.1% formic acid and peaking with 98% mobile phase B.
2.5. Detailed Workflow of the Final Assay Setup
Three Caco-2-based transport study designs serve as the basis for comparing in vitro systems in this study (
Table 1). Our goal was to develop an assay with optimized throughput and robust turnaround time. As a first step to achieve this goal, we utilized assay-ready cells for a shortened cultivation period of 7 days. The comparison with the commonly used 21-day cultivation time showed highly comparable outcomes in terms of permeability and efflux (
Figure S2). By modifications and comparisons with compounds of known absorption, the study parameters described in 2.3 have been identified as a superior design (
Figure S1).
The standard method is designed as a high-throughput assay with a short incubation time and without the use of BSA. It can accommodate up to 3 compounds cassetted at an individual concentration of 1 µM, allowing for a capacity of 132 compounds/assay along with four reference standards.
The BSA-modified standard method aims to characterize compounds that are not accessible with the standard method. In addition to adding 1% BSA to the assay buffer to solubilize compounds and to prevent non-specific binding, higher concentrations of 3 µM are used in a non-cassetted approach. This assay provides a lower throughput (44 compounds/assay).
Lastly, we introduced an equilibrated method that includes a pre-incubation step to saturate the system and achieve better compound recovery. This method mimics steady-state permeation, which is particularly important for bRo5 compounds with high molar mass, such as PROTACs. To further optimize this assay, the analytical method was improved to enhance sensitivity, as previously described (2.4). Like the BSA-modified standard method, this approach covers 44 compounds/assay. The workflow of the equilibrated method is depicted in
Figure 1.
2.6. Evaluation of Pre-Incubation Modification for the Equilibrated Method
We investigated the pre-incubation process, focusing on the use of BSA as a supplement in the buffer. A set of 21 representative compounds (Ro5 and bRo5;
Table 2) was tested in either setup and permeabilities were compared to a full 24 h pre-incubation setup as a control, accounting for total equilibration of the system, as described by Cui et al. [
11]. Based on these results, the final setting of the equilibrated method was established without the use of BSA in the pre-incubation but with BSA in the main incubation (
Figure 2).
2.7. In Vivo Absorption
To compare the in vitro results with in vivo absorption, we utilized either known literature human fraction absorbed (f
a) or approximated internal rodent fafg, assuming the fraction escaping gut metabolism (f
g) to be close to 1. The reference compounds included in this study are listed in
Table 3.
The fafg values were obtained from internal rodent (mouse and rat) in vivo intravenous (IV) and oral (PO) pharmacokinetic studies. Specifically, the rodent IV blood or plasma clearance (Cl
B or Cl
P) was used to calculate the hepatic bioavailability (f
h) using the extraction ratio, assuming a blood to plasma ratio of 1:
Hepatic blood flow (Q
h) has been estimated, based on internal and external sources, to be 5.2 L/h/kg for mice and 3.8 L/h/kg for rats [
13,
14,
15,
16]. The area under the concentration vs. time curves (0 to t) were dose-normalized and used to calculate the total oral bioavailability, F
PO.
where AUC is the area under the blood- or plasma-level curve and D the respective IV and PO doses.
Approximated intestinal absorption expressed as fafg was then calculated as follows:
Table 3.
Reference compounds and their observed human f
a values. Human f
a values taken from [
17]. Values were rounded to two significant figures.
Table 3.
Reference compounds and their observed human f
a values. Human f
a values taken from [
17]. Values were rounded to two significant figures.
Reference Compound | Observed Human fa |
---|
Amiloride | 0.53 |
Amprenavir | 0.76 |
Antipyrine | 0.98 |
Azithromycin | 0.43 |
Biperiden | 1.0 |
Boceprevir | 0.92 |
Bosentan | 0.70 |
Bupropion | 0.88 |
Caffeine | 1.0 |
Cetirizine | 0.73 |
Clozapine | 0.99 |
Corticosterone | 1.0 |
Diclofenac | 0.97 |
Dipyridamole | 0.56 |
Doxepin | 0.76 |
Flecainide | 0.82 |
Furosemide | 0.61 |
Gliquidone | 0.95 |
Hydroxychloroquine | 0.90 |
Imatinib | 0.90 |
Indinavir | 0.63 |
Ketoconazole | 0.76 |
Lamotrigine | 0.98 |
Lenalidomide | 0.90 |
Loperamide | 0.52 |
Loratadine | 0.90 |
Methotrexate | 0.64 |
Metoclopramide | 0.89 |
Mexiletine | 0.99 |
Moclobemide | 0.85 |
Naratriptan | 0.70 |
Nicotine | 1.0 |
Nimodipine | 1.0 |
Nortriptyline | 1.0 |
Ondansetron | 1.0 |
Oseltamivir | 0.75 |
Phenazopyridine | 0.90 |
Phenytoin | 0.93 |
Physostigmine | 0.050 |
Pirenzepine | 0.26 |
Propranolol | 0.97 |
Ranitidine | 0.56 |
Rifabutin | 0.53 |
Riluzole | 0.90 |
Risperidone | 0.85 |
Rizatriptan | 0.90 |
Rosiglitazone | 1.0 |
Rosuvastatin | 0.35 |
Roxithromycin | 0.86 |
Selegiline | 1.0 |
Sulfasalazine | 0.33 |
Sumatriptan | 0.58 |
Telmisartan | 0.82 |
Trazodone | 0.98 |
Trihexyphenidyl | 1.0 |
Vardenafil | 0.90 |
Venlafaxine | 0.96 |
Verapamil | 0.92 |
Vismodegib | 0.69 |
Ziprasidone | 0.63 |
Zolmitriptan | 0.92 |
2.8. Statistical Analysis
Significance was tested based on Welch’s t-test assuming unequal variances, using GraphPad Prism Software (Version 9.5.0). Significance was determined based on p < 0.05 (*), p < 0.01 (**), p < 0.005 (***), and p < 0.001 (****).
4. Discussion
Over the past decades, the chemical space in pharmaceutical development of small molecules has shifted towards larger and more complex chemical entities, often with suboptimal physicochemical properties. This trend is caused by the intention to engage formerly non-druggable targets or to induce target degradation with bispecific molecules (PROTACs). Modern compounds often violate more than one of the Lipinski rules and are therefore referred to as beyond rule of five (bRo5) molecules [
4]. Although the fundamental connection between permeability and absorption remains, it has become more relevant to characterize or rank permeability for compounds with lower absorption. As for efficiency reasons, throughout the pharmaceutical industry in vitro permeability assays have been developed for high-throughput and fast turnaround time, the standard incubation time is often too short to reach compound equilibrium in the cell, which limits the possibility of accurately measuring permeability and efflux. Cui et al. approached this challenge by introducing a 24 h pre-incubation, allowing the measurement of the intrinsic permeability at steady-state conditions. This pre-incubated setup achieved an adequate characterization of efflux and permeability for bRo5 substances, previously insufficiently or not characterized [
11].
Besides this approach, several powerful tools based on physicochemical compound parameters have been established [
4,
5,
6,
8]. However, our goal was to develop a cell-based high-throughput method which provides the determination of active cellular permeability and efflux for compounds in the bRo5 chemical space, with a focus on the possibility to predict the properties or rank-order compounds according to their expected intestinal absorption based on this cellular assay. After the identification of the most appropriate assay parameters (
Figure S1), a Caco-2-based system (parameters described in
Section 2.5) was further optimized to cope with known challenges, including assay validity, recovery, and correlation to human f
a and rodent fafg.
To achieve a high-throughput and a rapid turn-around, we applied assay-ready cell culturing and maintained the cells for 7–8 days, in contrast to a more commonly described 21-day cultivation. The 7- and 21-day culturing revealed no significant difference in functional performance (
Figure S2) and has already been shown to be suitable for advanced applications, like permeation studies with complex biorelevant media [
19]. Furthermore, we intended to reduce the pre-incubation time from 24 h to 1 h for improved handling and to avoid the potential cytotoxic effects of test compounds. This was achieved by conducting the pre-incubation step without, and the main incubation step with, BSA to provide a higher fraction unbound for a faster equilibration of the system. The permeability and the ER in this method were comparable to a 24 h pre-incubation (
Section 3.1;
Figure 2) [
11].
We compared the standard method (no pre-incubation, no BSA), the BSA-modified standard method (no pre-incubation, with BSA), and the equilibrated method (pre-incubation without BSA and main incubation with BSA; the exact method description is given in
Section 2.5,
Table 1) for assay validity and compound recovery with a compound set containing mainly bRo5 compounds. The introduction of BSA helped to reduce the number of invalid and qualified results with a valid result rate of 79% compared to 38% with the standard method. However, the equilibrated method exceeded the other two methods by achieving 94% valid results (
Figure 6).
For shared valid results, there was a significant improvement in recovery using the equilibrated method, with 72% and 94% of the results with compound recovery of >80% for A-to-B and B-to-A permeability, respectively. In contrast, recoveries for both the standard method and the BSA-modified standard method were >80% for only 50% and 39% for A-to-B and B-to-A permeability, respectively.
Although the use of BSA is known to prevent non-specific binding and to improve compound recovery [
10], the pre-incubation step of the equilibrated method further ameliorated the recovery through improved equilibration during the pre-incubation step and, consequently, steady-state compound transport. Adequate recovery rates (>80%) not only enhance the valid result rate but also boost confidence in cellular transport studies by ensuring the assay’s accuracy in measuring compound translocation across the layer.
The standard method and the equilibrated method were thoroughly compared regarding their output for permeability and efflux as well as their correlation to human f
a. In a set of 61 compounds with known human f
a (
Table 3) [
17], 92% and 97% of predictions with P
app,AB and ER were within three-fold, respectively. P
app,AB to f
a between both methods was comparable, and compounds could be classified with no significant differences between the methods. It is important to highlight that the compounds with available human f
a data in the literature were predominantly well-absorbed Ro5 substances with molecular weights mostly below 500 and less than two Lipinski violations (
Figure 10). The use of BSA in the main incubation of the equilibrated method did not have any significant impact on the permeability measurement. In other cell-based experimental setups, it was shown that the correlation dropped (R
2 < 0.5) only at very low fractions unbound to plasma (f
u,p < 0.1%) (
Figure S3). Despite the fact that the free drug hypothesis suggests stronger effects at lower protein binding, our evidence indicates that plasma proteins, like albumin, facilitate cellular uptake of compounds. This discrepancy arises from the dynamic and reversible nature of protein–compound interactions, as opposed to the static theoretical models [
20]. Consequently, this dynamic interaction explains why the inclusion of BSA in cellular transport assays generally does not affect the outcome of the assay significantly [
21].
Due to limited published data on human absorption of bRo5 substances from the existing literature, we applied the equilibrated method to 741 internal compounds with rodent fafg results. fafg served as a surrogate for in vivo absorption. In this compound set, 51% were bRo5 substances. In this analysis, a significant correlation between P
app and absorption was confirmed (
Figure 13). Interestingly, the median absorption (fafg) of the compounds was lower compared to the published compounds with human f
a values when using the same binning. Because of the differences of the chemical entities, the larger AbbVie set contained a higher number of compounds with lower solubility (91%,
Figure 14). Furthermore, many of these compounds were efflux substrates with ER greater than 2 (80%,
Figure 13). The great number of compounds with efflux liability additionally drove a strong correlation between ER and absorption. Overall, P
app and ER both could be used to rank order compounds when applying individual cut-offs with a high rate of true absorption prediction (64–86%). Based on these results and considering that efflux is an active transport component (often missing in non-cellular models), the cellular equilibrated Caco-2 assay is a powerful supplement to close the gaps around the determination of P
app and ER for bRo5 compounds with suboptimal physicochemical properties. With this supplement we are now able to measure 94% of the current AbbVie chemical space reliably, while previously we could only determine P
app and ER of 38% of the studied compounds (
Figure 6).
However, there are also limitations to this test system, as there is a significant fraction of false positives (predicted high fafg but low measured fafg) and false negatives (low permeability but high fafg) (
Figure 15). As this cellular assay only takes permeability into account, the relevance of solubility and intestinal metabolism (f
g) for oral absorption is missing. A compound with poor solubility or high intestinal metabolism may result in false positive results in our assay. On the other hand, higher absorption—despite low permeability—can be the result of either enabling formulation technologies in the corresponding in vivo studies or of particularities of the cellular system. Although binding effects to BSA are considered to be negligible in our assay systems (
Figure S3), especially for compounds with a very low fraction that is unbound, high binding affinity to BSA, or even non-reversible covalent protein binding, may contribute to false positive results.
Taken together, the equilibrated Caco-2 assay provides a powerful cell-based in vitro tool for the characterization of difficult compounds within the bRo5 space, where simpler cellular systems or non-cellular in vitro assays have failed. Due to the complex nature of absorption, it is important to further consider properties like solubility and intestinal metabolism to improve the predictability of in vitro models for oral absorption. Employing the equilibrated Caco-2 assay in conjunction with in vitro assays evaluating solubility and intestinal metabolism in the context of a PBPK model allows for the best prediction of the outcome of human absorption in phase I clinical trials and beyond.