We exposed a microphysiological system (MPS) with intestine and liver organoids to acetaminophen (APAP). This article describes the methods for organoid production and APAP pharmacokinetic and toxicological property assessments in the MPS. It also describes the tissue functionality analyses necessary to validate the results.
The recently introduced microphysiological systems (MPS) cultivating human organoids are expected to perform better than animals in the preclinical tests phase of drug developing process because they are genetically human and recapitulate the interplay among tissues. In this study, the human intestinal barrier (emulated by a co-culture of Caco-2 and HT-29 cells) and the liver equivalent (emulated by spheroids made of differentiated HepaRG cells and human hepatic stellate cells) were integrated into a two-organ chip (2-OC) microfluidic device to assess some acetaminophen (APAP) pharmacokinetic (PK) and toxicological properties. The MPS had three assemblies: Intestine only 2-OC, Liver only 2-OC, and Intestine/Liver 2-OC with the same media perfusing both organoids. For PK assessments, we dosed the APAP in the media at preset timepoints after administering it either over the intestinal barrier (emulating the oral route) or in the media (emulating the intravenous route), at 12 µM and 2 µM respectively. The media samples were analyzed by reversed-phase high-pressure liquid chromatography (HPLC). Organoids were analyzed for gene expression, for TEER values, for protein expression and activity, and then collected, fixed, and submitted to a set of morphological evaluations. The MTT technique performed well in assessing the organoid viability, but the high content analyses (HCA) were able to detect very early toxic events in response to APAP treatment. We verified that the media flow does not significantly affect the APAP absorption whereas it significantly improves the liver equivalent functionality. The APAP human intestinal absorption and hepatic metabolism could be emulated in the MPS. The association between MPS data and in silico modeling has great potential to improve the predictability of the in vitro methods and provide better accuracy than animal models in pharmacokinetic and toxicological studies.
Due to genomic and proteomic differences, animal models have limited predictive value for several human outcomes. Moreover, they are time-consuming, expensive and ethically questionable1. MPS is a relatively new technology that aims at improving the predictive power and reduce the costs and time spent with pre-clinical tests. They are microfluidic devices cultivating organoids (artificial mimetics functional units of organs) under media flow that promotes organoid-organoid communication. Organoids made of human cells increase translational relevance2,3,4. MPS is expected to perform better than the animal tests because they are genetically human and recapitulate the interplay among tissues. When fully functional, the MPS will provide more meaningful results, at higher speed and lower costs and risks4. Many groups are developing MPS for several purposes, especially disease models to tests drug’s efficacy.
Exposure level is one of the most critical parameters for evaluating drug efficacy and toxicity5,6,7,8,9,10,11,12. MPS allows organoid integration that emulates systemic exposure and is expected to perform better than the traditional 2D human tissue culture. This technology can significantly improve the prediction of compound intestinal absorption and liver metabolism4.
An MPS integrating human equivalent model of intestine and liver is a good starting point, considering the central role of these two organs in drug bioavailability and systemic exposure13,14,15. APAP is an attractive drug for studying an MPS without a kidney equivalent because its metabolization is done mainly by the liver16,17.
The 2-OC is a two-chamber microfluidic device suitable for the culture of two different human equivalent tissues/organoids interconnected by microchannels16. In order to emulate an in vitro human oral/intravenous administration of a drug and assess the effects of the cross-talk between the intestine and liver equivalents on APAP pharmacokinetics, besides the organoids functionality and viability, three different MPS assemblies were performed: (1) an “Intestine 2-OC MPS” comprised of an intestine equivalent based in a culture insert containing a Caco-2 + HT-29 cells coculture, integrated into the 2-OC device; (2) a “Liver 2-OC MPS” comprised of liver spheroids made of HepaRG + HHSteC (Human Hepatic Stellate Cells) integrated in the 2-OC device; and (3) an “Intestine/Liver 2-OC MPS” comprised of the intestine equivalent in one device compartment communicating with the liver equivalent in the other by the media flow through the microfluidic channels.
All assays were performed under static (no flow) and dynamic (with flow) conditions due to the impact of the mechanical stimuli (compression, stretching, and shear) on the cell viability and functionalities18,19,20. The present article describes the protocol for APAP oral/intravenous administration emulation and the respective absorption/metabolism and toxicological analyses in the 2-OC MPS containing human intestine and liver equivalent models.
1. Production of tissue equivalents for cultivation in the 2-OC
2. Integration of intestine and liver equivalents in a 2-OC MPS
3. Acetaminophen (APAP) preparation
4. Test substance administration and media sampling
5. Instrumentation and chromatographic conditions
6. Tissue equivalents viability/functionality
To perform the PK APAP tests in the 2-OC MPS, the first step is to manufacture the human intestine and liver equivalents (organoids). They are integrated into the 2-OC microfluidic device (Figure 1A) 24 h before starting of the PK APAP assay. The next day, the medium is changed, and the model is exposed to APAP. Figure 1 illustrates the intestine and liver equivalents placed inside the 2-OC device (Figure 1B) and the APAP PK experiment time course (Figure 1C). We performed a MTT assay, TEER measurements, HCA, real-time PCR, western blotting, histology, and confocal fluorescence microscopy in 2D culture and 3D organoids to check the tissue’s viability and detect possible APAP toxic effects. In the confocal fluorescence microscopy images, the intestine equivalent samples stained with DAPI and Phalloidin (for nuclei and actin respectively) were presented as a contiguous barrier for the non-treated (Figure 2A) and the 12 µM APAP treated samples (Figure 2B). As seen in Figure 2C, the MTT assay showed relative cell viability levels above 70%, indicating the absence of relevant cytotoxic effects in response to APAP exposure at 12 µM concentration33,34,35,36,37. The positive control (100 mM) induced significant cell death (survival below 5%). The Caco-2/HT-29 viability and proper differentiation as well as the intestine equivalent barrier integrity were verified by TEER evolution during the differentiation period (Figure 2D). APAP did not cause any alteration in the TEER values as shown in Figure 2E. The expression of the active sodium-coupled glucose transporters SLC5A1, multidrug resistance transporter MDR1 and sodium-potassium ATPase were analyzed, to verify the APAP treatment impact over cell barriers formation and basal functionality. As demonstrated in Figure 2F–H, both non-treated and APAP treated intestine equivalents have shown similar expression of SLC5A1 and NaKATPase. The oral administration of 12 µM APAP induced marked an increase in the MDR1 mRNA levels in intestines equivalents after 24 h (Figure 2H). We also performed HCA of cell phenotypic changes by a fluorophore dye mixture for nuclei and mitochondrial mass content. Positive controls were 100 mM APAP and 1% NaOH.
Additionally, we analyzed whether 12 µM APAP could induce cytotoxicity to the 2D Caco-2/HT-29 co-culture. The intestinal cells images acquired with fluorescence microscopy shown in Figure 2I corroborates the MTT data, which have demonstrated that 12 µM APAP did not cause significant cytotoxicity in the Caco-2/HT-29 intestinal equivalents.
The assessment of hepatic spheroids basal viability and the cytotoxicity in response to 2 µM APAP were done by MTT assay and morphological analyses by confocal fluorescence microscopy, H&E histology, and HCA assays. As shown in Figure 3A, the MTT assay was unable to identify any relevant cytotoxicity in response to the 2 μm APA treatment in samples taken from the Liver 2-OC assembly under both static and dynamic conditions. The cell viability decreased but remained over 80% for both 12 h and 24 h time-points33,34,35,36,37. The positive control treatments (100 mM APAP and 1% NaOH) induced significant tissue damages (viability below 10%). Microscopic confocal images indicate the absence of a necrotic center in the liver spheroids in both basal or APAP treatment conditions, and no evidence of significant death rates (Figure 3B-C). However, when we analyzed multiple cellular phenotypic changes following the vehicle or 2 µM APAP administration in the 2D HepaRG/HHSteC coculture through HCA assay, using 100 mM APAP (C+) as a positive control, contradictorily to the results of the MTT assay, the hepatic cells demonstrated early cytotoxic responses to 2 µM APAP treatment (Figure 3D). After 24 h, there was a decrease in the number of cells, in the nuclear area and an increase in mitochondrial mass. Additionally, a fluorophore dye cocktail containing Hoechst 33342 and MitoTracker Deep Red was used to stain the 3D hepatic spheroids (Figure 3J). Fiji software was used to evaluate 3D spherical architecture homogeneity among several spheroids (Figure 3E–I). The graphic shown in Figure 3E shows the similarity among liver spheroids total area. The aspect ratio (Figure 3F) around 1 means an absence of bias during the confection process of the spheroids. The evaluation also indicated that the majority of the spheroids were roughly rounded (Figure 3G). The evaluation of the morphology perimeter and cell distribution was done by circularity (Figure 3I) and by solidity calculation (Figure 3H), respectively. We concluded that the methodology to confectioning the liver spheroids had generated organoids with a smooth perimeter, compatible with spherical growth, no biases, or necrosis during the process.
As demonstrated in Figure 4A-B, the liver spheroids showed a relative basal high level of albumin and GST mRNA expression respectively, indicating proper basal functionality. Nevertheless, the 2 µM APAP treatment for 24 h induced a decrease in the albumin and the GST mRNA expression levels, suggesting impairment of liver spheroids functionally at 24 h time point of APAP treatment.
The detection of the CYP3A4 and UGT1A1 mRNA expression levels demonstrates the liver equivalents metabolic capacity. The CYP3A4 mRNA basal level (Figure 4C) was consistent with previous reports6. The APAP treatment induced a trend for a decrease in both CYP3A4 mRNA and UGT1A1 expression in response to APAP treatment (Figure 4C-D) once again corroborating the hypothesis of impairment of liver spheroids functionally at 24 h time point of APAP treatment.
Additionally, experiments of western blotting and in vitro enzymatic activity were performed in order to analyze the albumin protein expression, as well as, the CYP 3A4 activity in liver equivalents at basal and at APAP treatment conditions. We found that 2 µM APAP treatment performed at the Liver 2-OC MPS, induced a reduction in the liver equivalent total albumin expression at 12 h and 24 h time-points at both static (Figure 4E) and dynamic (Figure 4F) conditions. On the other hand, liver equivalents samples from dynamic conditions demonstrated a trend to present higher levels of protein expression of albumin when compared to the static conditions (Figure 4G). CYP 3A4 in vitro assay performed at liver equivalents from Liver 2-OC MPS shown that 2 µM APAP treatment for 12 h or 24 h was capable to induce a robust and significant impairment of CYP 3A4 activity at both static and dynamic conditions (Figure 4H-I). More interesting, the presence of media flow (dynamic) has induced a significant improvement of liver equivalents CYP 3A4 activity levels when compared to conditions in which the liver equivalents were kept in absence of circulating medium (static conditions) (Figure 4J).
To find the most sensitive analytical condition regarding HPLC analysis, several parameters were investigated including the composition of the mobile phase, the type, and concentration of additives. It was found that acetonitrile gave better chromatogram resolution and appropriate retention time than methanol. Fast and reproducible separation of APAP was obtained using a C18 reversed-phase column. The APAP retention time (Rt) value was 9.27 ± 0.19 minutes. Selectivity for APAP is indicated by the shape and symmetrical resolution of the peak, as well as by the lack of interfering peaks from the DMEM and Williams cell culture media.
APAP standard concentrations in DMEM S and Williams E S cell culture media diluted with ammonium acetate buffer (1:1, v/v) ranging from 0.25 to 100.00 µM were used to build the calibration curves. The linearity of the method was determined at nine concentration levels. The data are shown in Table 2 and Table 3. The relationship between APAP concentration and the peak areas was described by the linear regression equations: y = 16106*x + 3579.8 (R2=1, in DMEM medium) and y = 16397*x + 2475.1 (R2=1, in Williams medium), in which “x” is APAP nominal concentration in µM and “y” is the chromatogram peak area of APAP in AU. At the upper limit of quantification (i.e., 100.00 µM), the percentage deviation and the inter-run variability values were less than 2.50%. The accuracy and the precision for nine concentration levels, excluding the 0.50 µM (LLOQ), were within an acceptable range with DEV and C.V. values less than 7.00% (Table 2 and Table 3).
The analytical method inter- and intra-run accuracy and precision, at four tested concentrations, fell within the generally accepted criteria for bioanalytical assays. The reproducibility of the method was evaluated by analyzing replicates of APAP quality control samples of 0.50 (LLOQ), 4.50, 45.00 and 90.00 µM. The intra-run and inter-run average results are reported in Table 4. The accuracy and precision of the assay are demonstrated by DEV values ≤ 15.00% and by C.V. values ≤ 7.00%, respectively.
LOD was determined as the sample whose signal-to-noise ratio (S/N) was just greater than 3 and corresponded to a 0.25 µM APAP. On the other hand, the LLOQ, estimated with 0.50 µM APAP samples, displayed S/N ratio equal to 10. Furthermore, we found accuracy values (DEV%) ranging within ≤ 19.00% of the nominal concentration values. The intra- and inter-run variabilities were demonstrated by C.V. ≤ 18.77%, as shown in Table 2, Table 3 and Table 4)29,30.
The APAP PK analyses were performed in three different 2-OC MPS assemblies: 1) Intestine 2-OC, containing the intestine equivalent only; 2) Liver 2-OC, containing the liver spheroids only and 3) Intestine/Liver 2-OC with both intestinal barrier and liver spheroids.
For absorption studies, the oral route was mimicked by the administration of 12 µM APAP on the intestine equivalent apical side. APAP concentrations were measured by HPLC/UV, in the medium samples, collected from apical and basolateral intestinal equivalents sides, in both static and dynamic conditions. The APAP kinetics in the medium collected from the apical and basolateral sides demonstrated that the intestine model was able to absorb the APAP. There was a progressive APAP concentration decrease in the apical side (Figure 5A) concomitantly to APAP concentration increase at the intestinal basolateral side (Figure 5B). The maximum concentrations (Cmax) in the medium was around 2 µM for both static and dynamic conditions, after 12 h of the administration (Tmax).
For metabolism studies, the intravenous administration was mimicked by the application of 2 µM APAP in the medium of the liver compartment. The APAP concentration kinetics in the media under both static and dynamic conditions indicated that only at the dynamic conditions the decreases in the APAP concentration could be detected, reaching 0.87 µM APAP 12 h after 2 µM APAP administration (T1/2 = 12 h). The liver equivalents showed minimal metabolic efficiency under static conditions (Figure 5C). The APAP concentration reached 1.7 µM 12 h after APAP administration. The integrated, systemic like APAP absorption and metabolism evaluation was performed in the Intestine/Liver 2-OC model. The APAP was administered over the apical side of the intestine equivalent, emulating the oral route. Medium samples were collected from both Intestinal sides and also from the liver compartment. Figure 5D shows the progressive decay of the APAP concentration at the apical side in both static and dynamic conditions.
Figure 5E shows distinguishable absorption and metabolism phases. The flow also impacted in the intestinal absorption. The APAP Cmax in the medium changed from 2 µM on the “Intestine 2-OC” (Figure 5B) assembly to 1.7 µM for the dynamic “Intestine/Liver 2-OC” (Figure 5E). Figure 5F shows a direct comparison between the concentration–time profile of APAP in our dynamic “Intestine/Liver 2-OC” microphysiological system (red curve and y axis) and a representative profile obtained in humans after a single oral dose of 1000 mg (black curve and y axis).
Figure 1: Schematic step compilation for PK studies in the 2-OC MPS. A) Schematic drawing of the 2-OC MPS, showing the intestinal and hepatic human tissue equivalents in bottom-up view. B) 2-OC MPS photograph with the intestinal and liver equivalents integrated into the device in bottom-up view, with representative optical microscopy images. C) Timeline of tissue equivalents preparation, APAP treatment, and culture medium collections phases for organoid manufacturing, and pharmacokinetic and toxicological assessments. This figure has been modified from Marin et al. Acetaminophen absorption and metabolism in an intestine/liver microphysiological system. Chem Biol Interact. 299, 59-76 (2019). Please click here to view a larger version of this figure.
Figure 2: Viability and toxicological assessment of the intestine equivalents. A) Representative confocal fluorescence microscopy images of non-treated Caco-2/HT-29 cells stained with cell nuclei and actin filaments fluorescent dye (DAPI and Phalloidin respectively); 63x Magnification, zoom 2.6. B) Representative confocal fluorescence microscopy images of Caco-2/HT-29 cells treated with 12 µM APAP for 24 h, stained with nuclei and actin filaments fluorescent dye (DAPI and Phalloidin respectively); 63x Magnification, zoom 2.6. C) Intestine equivalents viability evaluation by MTT assay in both static and dynamic conditions. The values represented by the bars in the graph are percent calculated relative to vehicle control (time-point named as 0)*P<0.05 0 vs treatment. D) TEER evolution during the 21 days of differentiation. *P<0.05 day 1 vs other days. E) TEER values after APAP administration into the Intestine 2-OC MPS under dynamic conditions. Gene expression in intestines equivalents. Absorption potential of the intestine barrier and possible effects of 12 µM APAP for 24 h under dynamic condition was verified by SLC5A1 (F), Na-K-ATPase (G) and MDR1 (H) expression. Values represent the mean ± SEM of three independent experiments. The result is expressed as a ratio to housekeeping GAPDH. *P<0.05 vehicle vs APAP. I) Operetta image-based HCA performed by the Columbus® 2.4.0. software. Representative images of 2D intestine co-culture in different time points after 12 µM APAP treatment. Negative controls were medium (0 h) or vehicle (0.5% ethanol). Positive controls shown here is the 100 mM APAP. This figure has been modified from Marin et al. Acetaminophen absorption and metabolism in an intestine/liver microphysiological system. Chem Biol Interact. 299, 59-76 (2019). Please click here to view a larger version of this figure.
Figure 3: Viability and toxicological assessment of the liver equivalents. A) Liver equivalents viability evaluation by MTT assay in both static and dynamic conditions. The values represented by the bars in the graph are percent calculated relative to vehicle control (time-point named as 0) *P<0.05 0 vs treatment. B) Representatives confocal images captured from the vehicle and 2 µM APAP 24 h treated liver spheroids from an inner section. C) Representatives H&E (hematoxylin and eosin staining) images captured from the vehicle and 2 µM APAP 24 h treated liver spheroids from an inner section. Scale bar = 50 µm. D) Representative images of 2D liver co-culture in different time points after 2 µM APAP treatment. Samples treated with vehicle and with 2 µM APAP were considered in these analyses. The fluorophore dye mixture includes Hoechst for nuclear staining and Mitotracker Deep Red for mitochondria mass staining. Negative controls were medium (0 h) or vehicle (0.5% ethanol). Positive controls were 100 mM APAP. Measurements of whole spheroids images captured were performed using Fiji software. E) Frequency distributions of the area F) aspect ratio, G) roundness, H) solidity, I) circularity. N = 85. *p < 0,05. J) Representative images of 3D liver spheroids acquired by the Operetta using the LWD 10x objective. This figure has been modified from Marin et al. Acetaminophen absorption and metabolism in an intestine/liver microphysiological system. Chem Biol Interact. 299, 59-76 (2019). Please click here to view a larger version of this figure.
Figure 4: Liver viability/functionality and possible effects of 2 µM APAP under static and dynamic conditions over it were verified by gene and protein expression and by enzymatic activity. A) albumin gene expression. B) GSTA2 gene expression. Liver capability to perform phase I and phase II metabolism and possible effects of 2 µM APAP for 24 h under dynamic condition over it were verified by gene expression of CYP3A4 (C) and by UGT1A1 (D) respectively. E) Total albumin protein expression under static condition. F) Total albumin protein expression under dynamic conditions. condition. G) Comparative graph illustrating the difference in total albumin expression in liver equivalents cultivated and treated under static or dynamic conditions. H) CYP 3A4 in vitro enzymatic activity under static conditions. I) CYP 3A4 in vitro enzymatic activity under dynamic conditions. J) Comparative graph illustrating the difference in CYP 3A4 activity in liver equivalents cultivated and treated under static or dynamic conditions. Values represent the mean ± SEM of three independent experiments. The data of gene expression is expressed as a ratio to housekeeping GAPDH. The data of protein expression is expressed as a ratio to vinculin protein. *P<0.05 vehicle vs APAP treatment. This figure has been modified from Marin et al. Acetaminophen absorption and metabolism in an intestine/liver microphysiological system. Chem Biol Interact. 299, 59-76 (2019). Please click here to view a larger version of this figure.
Figure 5: Analyzes of APAP pharmacokinetics in 2-OC MPS. APAP absorption profile after 12 µM APAP administration at the apical side of the Intestine 2-OC MPS preparation. The intestinal barrier was made of a coculture of Caco-2/HT-29 cell lines (A) Static and dynamic APAP concentrations in the medium from the apical side (representing the human intestinal lumen side). (B) Static and dynamic APAP concentrations in the medium from the basolateral side (representing the human intestinal bloodstream side). *P<0.05 static vs dynamic conditions. C) APAP metabolism profile in the Liver 2-OC MPS by HepaRG/HHSTeC liver spheroids. Comparison of static and dynamic conditions after a 2 µM APAP administration into the medium. *P<0.05 0h vs 6 h, 12 h, and 24 h APAP treatment. APAP absorption and metabolism profile after 12 µM administration on the intestinal barrier apical side of the Intestine/Liver 2-OC MPS preparation. This emulates the oral route. The intestinal barrier was made of Caco-2/HT-29 cell lines and the liver equivalent made of spheroids of HepaRG/HHSTeC cell lines. (D) Intestinal/Liver 2-OC APAP concentrations in the apical side of the intestinal barrier under static and dynamic conditions. (E) Intestinal/Liver 2-OC APAP concentrations in the medium under static and dynamic conditions. *P<0.05 static vs dynamic conditions. (F) Comparison between the concentration–time profile of APAP in our microphysiological system (red curve and y axis) and a representative profile obtained in humans after a single oral dose of 1000 mg (black curve and y axis). Data was extracted from plots using WebPlotDigitizer 4.2 (https://automeris.io/WebPlotDigitizer). This figure has been modified from Marin et al. Acetaminophen absorption and metabolism in an intestine/liver microphysiological system. Chem Biol Interact. 299, 59-76 (2019). Please click here to view a larger version of this figure.
HPLC system | Waters Alliance 2695 (Milford, MA, USA), equipped with quaternary pump, sample manager and degasser | ||
Detector | Waters 2996 Uv-Vis set in 210-400 nm range | ||
System control, data acquisition, and processing | Waters Empower 2002 chromatography software | ||
Column | Reversed-phase Luna C18 | ||
(150 x 4.6 mm I.D.; 5mm particle size) | |||
Phenomenex | |||
Guard Column | Reversed-phase Luna C18 (4 x 3 mm) | ||
Phenomenex | |||
Mobile phase | Solvent A- Acetonitrile | ||
Solvent B- 0.10 M ammonium acetate, pH 6.8 | |||
Isocratic conditions | Time | A (%) | B (%) |
(min.) | |||
15 | 5 | 95 | |
Flow | 1.0 mL/min | ||
Injection volume | 25 μL | ||
Temperature | 25 °C | ||
APAP Detection | UV @ 243 and 254 nm | ||
Run time | 15 minutes |
Table 1: Conditions and parameters to be used for HPLC-UV analyses of APAP in culture medium matrices.
Nominal concentration | Calculated APAP concentration (µM) | Average (µM) | S.D.b | C.V. | DEV | |||||
(µM) | (Triplicate of each concentration) | (µM) | (%)c | (%)d | ||||||
Assay number | 1 | 2 | 3 | 4 | 5 | 6 | ||||
0.25 (LOD) | 0.02 | 0.08 | 0.21 | 0.14 | 0.08 | 0.27 | 0.13 | 0.11 | 84.96 | + 46.05 |
0.50 (LLOQ) | 0.31 | 0.36 | 0.29 | 0.47 | 0.42 | 0.65 | 0.41 | 0.05 | 12.72 | + 17.08 |
1.00 | 0.87 | 0.87 | 0.80 | 1.04 | 1.02 | 1.01 | 0.93 | 0.04 | 3.76 | + 6.65 |
2.50 | 2.44 | 2.61 | 2.52 | 2.42 | 2.56 | 2.54 | 2.52 | 0.04 | 1.54 | -0.62 |
5.00 | 5.02 | 4.99 | 5.06 | 5.01 | 5.01 | 5.05 | 5.02 | 0.09 | 1.88 | -0.45 |
10.00 | 10.21 | 10.13 | 9.96 | 10.25 | 10.08 | 10.21 | 10.14 | 0.10 | 0.97 | -1.41 |
25.00 | 25.33 | 25.28 | 25.20 | 25.13 | 25.14 | 24.92 | 25.17 | 0.36 | 1.45 | -0.67 |
50.00 | 50.30 | 50.04 | 50.51 | 49.70 | 49.98 | 49.19 | 49.95 | 0.86 | 1.71 | +0.09 |
100.00 | 99.75 | 99.90 | 99.70 | 100.09 | 99.97 | 100.40 | 99.97 | 0.69 | 0.69 | +0.03 |
R2 | 1.00 | 1.00 | 0.9999 | 1.00 | 1.00 | 0.9999 |
Table 2: Inter-run variation – accuracy, precision, and linearity of standard curve samples prepared in a mixture of DMEM medium with 0.10 M ammonium acetate buffer (1:1, v/v) from six separate assays.a
aA linear curve was fitted to the data for a response (APAP) versus theoretical concentration as described in Experimental. The calculated concentration was derived from reading the response for each standard sample against the calibration curve. Each entry (assay 1-6) corresponds to the average value of triplicate analysis.
bSD= Standard deviation.
cC.V. (coefficient of variation. precision).
dAccuracy (DEV %) = the deviation of the calculated concentration from the nominal value.
This figure has been modified from Marin et al. Acetaminophen absorption and metabolism in an intestine/liver microphysiological system. Chem Biol Interact. 299, 59-76 (2019).
Nominal concentration | Calculated APAP concentration (µM) | Average | S.D.b | C.V. | DEV | ||||
(µM) | (Triplicate of each concentration) | (µM) | (µM) | (%)c | (%)d | ||||
Assay number | 1 | 2 | 3 | 4 | 5 | ||||
0.25 (LOD) | 0.34 | 0.12 | 0.30 | 0.16 | 0.00 | 0.18 | 0.08 | 45.46 | +26.03 |
0.50 (LLOQ) | 0.36 | 0.44 | 0.49 | 0.43 | 0.40 | 0.43 | 0.02 | 5.84 | +14.97 |
1.00 | 0.87 | 0.98 | 1.04 | 0.94 | 0.83 | 0.93 | 0.04 | 4.67 | +6.85 |
2.50 | 2.41 | 2.46 | 2.49 | 2.52 | 2.43 | 2.46 | 0.06 | 2.39 | +1.56 |
5.00 | 5.00 | 4.99 | 5.12 | 5.10 | 5.14 | 5.07 | 0.15 | 3.00 | -1.38 |
10.00 | 10.08 | 10.01 | 9.93 | 10.10 | 10.29 | 10.08 | 0.18 | 1.80 | -0.81 |
25.00 | 25.14 | 25.18 | 24.96 | 25.32 | 25.35 | 25.19 | 0.45 | 1.78 | -0.76 |
50.00 | 50.19 | 50.23 | 49.83 | 49.56 | 50.17 | 49.99 | 0.87 | 1.75 | +0.01 |
100.00 | 99.87 | 99.84 | 100.10 | 100.13 | 99.80 | 99.95 | 2.12 | 2.12 | +0.05 |
R2 | 1.00 | 1.00 | 1.00 | 1.00 | 1.00 |
Table 3: Inter-run variation – accuracy, precision, and linearity of standard curve samples prepared in a mixture of Williams medium with 0.10 M ammonium acetate buffer (1:1, v/v) from six separate assays.a
aA linear curve was fitted to the data for a response (APAP) versus theoretical concentration as described in Experimental. The calculated concentration was derived from reading the response for each standard sample against a calibration curve. Each entry (assay 1-5) corresponds to the average value of triplicate analysis.
bSD= Standard deviation.
cC.V. (coefficient of variation. precision).
dAccuracy (DEV %) = the deviation of the calculated concentration from the nominal value.
This figure has been modified from Marin et al. Acetaminophen absorption and metabolism in an intestine/liver microphysiological system. Chem Biol Interact. 299, 59-76 (2019).
Williams medium | Nominal concentration | Measured concentration | S.D. | C.V. | DEV |
(µM) | (µM) | (µM) | (%) | (%) | |
Intra-run (n=3) | 0.50 (LLOQ) | 0.49 | 0.08 | 15.55 | +1.77 |
4.50 | 4.59 | 0.23 | 5.10 | -2.06 | |
45.00 | 41.23 | 0.76 | 1.85 | +8.37 | |
90.00 | 82.29 | 1.75 | 2.13 | +8.57 | |
Inter-run (n=15) | 0.50 (LLOQ) | 0.43 | 0.05 | 10.99 | +14.97 |
4.50 | 4.37 | 0.19 | 4.42 | +2.99 | |
45.00 | 42.35 | 0.82 | 1.93 | +5.88 | |
90.00 | 85.22 | 2.25 | 2.65 | +5.31 | |
DMEM medium | Nominal concentration | Measured concentration | S.D. | C.V. | DEV |
(µM) | (µM) | (µM) | (%) | (%) | |
Intra-run (n=3) | 0.50 (LLOQ) | 0.47 | 0.09 | 18.77 | +6.35 |
4.50 | 4.45 | 0.30 | 6.63 | +1.04 | |
45.00 | 44.24 | 1.59 | 3.58 | +1.69 | |
90.00 | 86.40 | 4.09 | 4.73 | +4.00 | |
Inter-run (n=12) | 0.50 (LLOQ) | 0.46 | 0.08 | 17.81 | +7.75 |
4.50 | 5.16 | 0.27 | 5.31 | -14.68 | |
45.00 | 48.99 | 2.10 | 4.29 | -8.86 | |
90.00 | 96.18 | 4.47 | 4.65 | -6.86 |
Table 4: Intra- and inter-run precision and accuracy for APAP in quality control samples.a
aThe data are shown as averages. SD (standard deviation). C.V. (coefficient of variation. precision) and accuracy (percent deviation. DEV%).
This figure has been modified from Marin et al. Acetaminophen absorption and metabolism in an intestine/liver microphysiological system. Chem Biol Interact. 299, 59-76 (2019).
Primer (5’ → 3’) | |||||
Tissue | Gene | Forward | Reverse | ||
Intestine | SGLT1/SLC5A1 | gAgCCCAgCAACTgTCCCAC | CAggCTCCAACACAgACggT | ||
NA-K-ATPase | ACCgCCCAgAAATCCCAAAAC | CAgCggTCATCCCAgTCC | |||
MDR1 | TggATgTTTCCggTTTggAg | TgTgggCTgCTgATATTTTgg | |||
Liver | Albumin | TgCAAggCTgATAAggAg | TTTAgACAgggTgTTggCTTTACAC | ||
GSTA2 | CTgAggAACAAgATgCCAAgC | AgCAgAgggAAggCTggAAATAAg | |||
CPY3A4 | ggAAggTggACCCAgAAACTgC | TTACggTgCCATCCCTTgAC | |||
UGT1A1 | ATgCAAAgCgCATggAgAC | ggTCCTTgTgAAggCTggAg |
Table 5: Real-time qPCR primers to evaluate gene transcription at mRNA level in the 2-OC cultures for intestine and liver tissues
The accurate and reliable assessment of the pharmacologic properties of investigative new drugs is critical for reducing the risk in the following development steps. The MPS is a relatively new technology, that aims at improving the predictive power and reducing the costs and time spent with preclinical tests. Our group is advancing in the assessment of pharmacokinetic and toxicological properties mostly needed for lead optimization. We worked with the 2-OC microfluidic device, which has two chambers, allowing the integration of two organoids. APAP was chosen because it has plenty of high-quality human data, is mostly metabolized by the liver, and also displays hepatotoxic properties. The study protocol aimed to emulate some steps of the human phase I clinical trial, where a drug is administered orally to volunteers, and periodic blood samples are drawn to assess the drugs’ concentration to know the pharmacokinetic properties and biochemical and clinical parameters are collected to assess the safety and tolerability. Therefore, when the MPS evolves enough to provide reliable predictions, it will significantly reduce the risk of failure in the phase I trial. By adding the disease models in the future, the same assumption possibly will apply to the phases II and III clinical trials.
All studies were performed in three models: Intestine 2-OC (APAP oral administration), Liver 2-OC (Intravenous administration), and Intestine/Liver 2-OC (oral administration). The first model isolated the absorption, the second the metabolism, and the third integrated both. We first produced the organoids and incorporated into the microfluidic device, second administered the APAP and collected the media samples, and last performed a set of tests to assess the cell viability and functionality and the toxicological impact of APAP exposure. For the pharmacokinetic studies, we developed and validated a chromatographic method for APAP quantification in the medium. The validation complied with the Guideline on Bioanalytical Method Validation regarding specificity, linearity, the limit of quantification (LOQ), the limit of detection (LOD), matrix effect, precision, and accuracy, as outlined by FDA29,30. The specificity was established with blank, pooled, and individual biological samples from two different sources. The method performed acceptably during the analysis, and the data confirmed the ability of a simple isocratic mobile phase to separate and quantify APAP.
The viability/functionally of the intestine and liver organoids was assessed by different techniques. For the intestine equivalent, the confocal fluorescence microscopy (Figure 2A-B), the MTT assay (Figure 2C), gene expression assessment (Figure 2D-F) or HCA experiment, did not detect any toxicity 24 h after APAP exposure. Likewise, MTT assay did not detect toxic insults induced by APAP in liver equivalents (Figure 3A). However, the HCA technique added the possibility of detection of very early toxic events (24 h after APAP exposure) for liver cells, by the observation of cell phenotypic changes, with some mechanistic clues (Figure 3D). On the other hand, the confocal fluorescence microscopy (Figure 3B) and histology (Figure 3C) were showed to be a useful complementary tool in the investigation of the viability status of the human tissue’s equivalents. For the liver cells, the simultaneous use of different techniques was very advantageous. The MTT assay, as mentioned before, was not able to detect the APAP cytotoxicity detected by the HCA 24 h after APAP exposure. The gene expression analyses assessed the functionality of the intestine (Figure 2D-F) and liver organoids (Figure 4A-D) at both basal and 24 h after APAP treatment. Under basal conditions, there were normal levels of intestinal and liver-specific markers, suggesting proper functionality. After 24 h of APAP exposure, there was a downregulation of albumin, GST mRNA levels, and a tendency to reduce the CYP3A4 gene expression in liver equivalent tissues, indicating APAP early cytotoxicity. Corroborating these findings, Western blotting experiments showed that the reduction in gene expression was also accompanied by a robust reduction in liver total albumin protein expression in response to APAP treatment (Figure 4E-F), confirming the toxic insults imposed on liver tissue by exposure to APAP. Accordingly, experiments of in vitro enzymatic activity showed a robust and progressive reduction in CYP 3A4 activity levels induced by APAP treatment, in liver equivalents (Figure 4H-I).
Morphometrical statistics of organoids were performed on Image J (Figure 3E-I). The area reveals how close the 2D size is to all organoids analyzed, which could be used as standardization in this protocol so that an unbiased result can be produced. The ‘shape descriptors’ reveal statistics corresponding precisely to the shape morphology. The Aspect Ratio is an index which uses the major and minor axis, so results around 1 indicate no bias (i.e., preferential growth) during organoid formation. Values of Roundness (4 ×[area]/ (π × [major axis]2)) are very sensitive to a preferential growth, which would be revealed as a major axis. Solidity ([area]/ ([convex area])) is essential in showing gross morphology as it is not affected by irregularities in borders since it uses convex area (=envelope). Distributions centered around 1.0 indicate putative spherical growth. Conversely, Circularity (4× π [area]/[perimeter]2) is very sensitive to a complex perimeter, so “cavities” or “pockets” would impact this index. Thus, circularity around 1 also corroborates putative spherical growth, compatible with proper organoid functionality.
The analytical results showed that the MPS could emulate the APAP absorption and metabolism properties, both isolated or integrated in a curve comparable to that produced in vivo (Figure 5F) without the excretion phase. The APAP absorption was similar after oral administration to both Intestine MPS or Intestine/Liver MPS models, under both static and dynamic conditions (Figure 5A-B, Figure 5D-E). In both, there was an APAP concentration decreases at apical side concomitantly to its increase at the basolateral side, with no significant difference for static and dynamic conditions. In contrast, the APAP hepatic metabolism differed under these conditions. The circulating media in MPS seemed to improve the organoid metabolic capability (Figure 5C and Figure 5E). There was a significant APAP decay underflow not seen without flow. Interestingly, in vitro, CYP3A4 activity experiments corroborate the hypothesis that the presence of flow in the system increases the functionality of human liver equivalents. As shown in the graph in Figure 4J, the activity of CYP 3A4 was significantly higher in liver equivalents maintained under flow at both basal and APAP treatment conditions. Likewise, the liver equivalents kept under flow (dynamic) showed a tendency to increase the protein expression of albumin when compared to those kept without flow (static) both at baseline or at APAP treated conditions (Figure 4G).
When compared to the concentration-time profile after oral administration of APAP to humans, our microphysiological system shows much larger t1/2 (half-life time) (Figure 5F). This happens, as mentioned before, because while we have a two-organ system with intestine and liver equivalents that can absorb and metabolize the drug, there is no kidney equivalent to excrete APAP and its metabolites from the plasma compartment38. In addition to that, the microphysiological system shows smaller Cmax (peak plasma concentration) and larger tmax (time to reach Cmax) than what is typically observed for humans (Figure 5F). This is a consequence of the differences in the scale of the in vitro and in vivo experiments. Overall, there are three main differences between the concentration–time profile obtained using the microphysiological system and profiles obtained after oral administration of APAP to humans: larger t1/2, smaller Cmax and larger tmax (Figure 5F). While the larger t1/2 is due to the absence of a kidney equivalent to excrete APAP and its metabolites from the plasma equivalent, smaller Cmax and larger tmax are consequences of the small scale of the experiment when compared to the human body. The best scaling strategies for building and operating microphysiological systems are still an active area of research and it is unlikely that a single approach is optimal for all systems39. Additionally, mathematical modeling or machine learning can be used to apply corrections or learn the mapping from the micro scale to full scale in order to extrapolate in vitro data obtained with the microphysiological systems to the in vivo behavior observed in humans40.
The authors have nothing to disclose.
We thanks to Dr. Christie Guguen-Guillouzo, Dr. Philippe Gripon at Unit 522 INSERM and to Dr. Christian Trepo at Unit 271 INSERM for the use of the Biological Material (Hepa RG cells) and for making then available for us in order to perform the academic research.
1x DPBS | Thermo Fisher Scientific | 14190235 | No calcium, no magnesium |
2-OC | TissUse GmbH | Two-organ chip | |
384-well Spheroid Microplate | Corning | 3830 | Black/Clear, Round Bottom, Ultra-Low Attachment |
4% Paraformaldehyde | Use to fix cell | ||
Acetaminophen | Sigma Aldrich | A7085 | Use to MPS assays |
Acetonitrile | Tedia | Used to perform HPLC | |
Alexa Fluor 647 phalloidin | Thermo Fisher Scientific | confocal experiment | |
Ammonium acetate | Sigma Aldrich | Used to perform HPLC | |
Caco-2 cells | Sigma Aldrich | 86010202 | |
Cacodylate buffer | |||
Cell culture flasks | Sarstedt | ||
Confocal Fluorescence microscope | Leica | DMI6000 | |
Cryostat | Leica | CM1950 | |
DMEM high glucose | Thermo Fisher Scientific | 12800017 | Add supplements: 10% fetal bovine serum, 100 units per mL penicillin, 100 µg/mL streptomycin, and 1% non-essential amino acids |
DMSO | Sigma Aldrich | D4540 | Add 2% to HepaRG media |
Ethanol | Synth | ||
Fetal Bovine Serum | Thermo Fisher Scientific | 12657029 | |
Freezing medium OCT | Tissue-Tek | Tissue-Tek® O.C.T.™ Compound is a formulation of watersoluble glycols and resins, providing a convenient specimen matrix for cryostat sectioning at temperatures of -10°C and below. | |
Hematoxylin & Eosin | |||
HepaRG cells | Biopredic International | HPR101 | Undifferentiated cells |
HHSTeC | ScienCell Research Laboratories | 5300 | Cells and all culture supplements |
Hoechst 33342 | HCA experiments | ||
HT-29 cells | Sigma Aldrich | 85061109 | |
Human Insulin | Invitrogen – Thermo Fisher Scientific | 12585014 | |
Hydrocortisone | Sigma Aldrich | H0888 | |
Isopropanol | Merck | 278475 | |
Karnovsky’s fixative | |||
L-glutamine | Thermo Fisher Scientific | A2916801 | |
Luna C18 guard column SS | Phenomenex | Used to perform HPLC | |
Microscope | Leica | DMi4000 | |
Microtome | Leica | RM2245 | |
Millicell 0.4 µm pore size inserts | Merck | PIHP01250 | |
Millicell ERS-2 meter | Merck | MERS00002 | Used to TEER measurement |
MitoTracker Deep Red | HCA experiments | ||
MTT | Thermo Fisher Scientific | M6494 | |
MX3000P system | Agilent Technologies | ||
Neubauer chamber | Counting cells | ||
Operetta High Content Imaging System | Perkin Elmer | Used to perform HCA | |
P450-Glo CYP3A4 Assay with Luciferin-IPA | Promega | Cat.# V9001 | |
Penicillin/Streptomycin | Thermo Fisher Scientific | 15070063 | Cell culture |
Permount | Thermo Fisher Scientific | Histology | |
Primers | RT-qPCR | ||
PVDF membrane | BioRad | ||
PVDF Syringe filter | 0.22 μm pore size | ||
Reversed-phase Luna C18 column | Phenomenex | Used to perform HPLC | |
Shaker (IKA VXR Basic Vibrax) | IKA Works GmbH & Co | 2819000 | Used for spheroids to improve MTT assay |
Stellate Cell Media (STeC CM) | ScienCell | 5301 | Add STeC CM supplements |
SuperScriptIITM Reverse Transcriptase | Thermo Fisher Scientific | ||
SYBR Green PCR Master Mix | Thermo Fisher Scientific | ||
TRizol TM reagent | Thermo Fisher Scientific | Trizol is a monophasic solution of phenol and guanidine isothiocyanate. | |
Trypsin/EDTA solution | Thermo Fisher Scientific | R001100 | |
Ultra-low-attachment plates | Corning | CLS3471-24EA | 6 wells |
Vectashield plus DAPI mounting media | |||
White Opaque 96-well Microplate | PerkinHelmer | ||
Wide-bore tips | |||
Williams E | Pan Biotech | P04-29510 | Add supplements: 10% fetal bovine serum, 2 mM L-glutamine, 100 units per ml penicillin, 100 µg/mL streptomycin and 5 µg/mL human insulin |