Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Front Bioeng Biotechnol ; 12: 1397465, 2024.
Article in English | MEDLINE | ID: mdl-38812919

ABSTRACT

Protein crystallization as opposed to well-established chromatography processes has the benefits to reduce production costs while reaching a comparable high purity. However, monitoring crystallization processes remains a challenge as the produced crystals may interfere with analytical measurements. Especially for capturing proteins from complex feedstock containing various impurities, establishing reliable process analytical technology (PAT) to monitor protein crystallization processes can be complicated. In heterogeneous mixtures, important product characteristics can be found by multivariate analysis and chemometrics, thus contributing to the development of a thorough process understanding. In this project, an analytical set-up is established combining offline analytics, on-line ultraviolet visible light (UV/Vis) spectroscopy, and in-line Raman spectroscopy to monitor a stirred-batch crystallization process with multiple phases and species being present. As an example process, the enzyme Lactobacillus kefir alcohol dehydrogenase (LkADH) was crystallized from clarified Escherichia coli (E. coli) lysate on a 300 mL scale in five distinct experiments, with the experimental conditions changing in terms of the initial lysate solution preparation method and precipitant concentration. Since UV/Vis spectroscopy is sensitive to particles, a cross-flow filtration (cross-flow filtration)-based bypass enabled the on-line analysis of the liquid phase providing information on the lysate composition regarding the nucleic acid to protein ratio. A principal component analysis (PCA) of in situ Raman spectra supported the identification of spectra and wavenumber ranges associated with productspecific information and revealed that the experiments followed a comparable, spectral trend when crystals were present. Based on preprocessed Raman spectra, a partial least squares (PLS) regression model was optimized to monitor the target molecule concentration in real-time. The off-line sample analysis provided information on the crystal number and crystal geometry by automated image analysis as well as the concentration of LkADH and host cell proteins (HCPs) In spite of a complex lysate suspension containing scattering crystals and various impurities, it was possible to monitor the target molecule concentration in a heterogeneous, multi-phase process using spectroscopic methods. With the presented analytical set-up of off-line, particle-sensitive on-line, and in-line analyzers, a crystallization capture process can be characterized better in terms of the geometry, yield, and purity of the crystals.

2.
Anal Bioanal Chem ; 414(21): 6379-6391, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35661232

ABSTRACT

Since preparative chromatography is a sustainability challenge due to large amounts of consumables used in downstream processing of biomolecules, protein crystallization offers a promising alternative as a purification method. While the limited crystallizability of proteins often restricts a broad application of crystallization as a purification method, advances in molecular biology, as well as computational methods are pushing the applicability towards integration in biotechnological downstream processes. However, in industrial and academic settings, monitoring protein crystallization processes non-invasively by microscopic photography and automated image evaluation remains a challenging problem. Recently, the identification of single crystal objects using deep learning has been the subject of increased attention for various model systems. However, the advancement of crystal detection using deep learning for biotechnological applications is limited: robust models obtained through supervised machine learning tasks require large-scale and high-quality data sets usually obtained in large projects through extensive manual labeling, an approach that is highly error-prone for dense systems of transparent crystals. For the first time, recent trends involving the use of synthetic data sets for supervised learning are transferred, thus generating photorealistic images of virtual protein crystals in suspension (PCS) through the use of ray tracing algorithms, accompanied by specialized data augmentations modelling experimental noise. Further, it is demonstrated that state-of-the-art models trained with the large-scale synthetic PCS data set outperform similar fine-tuned models based on the average precision metric on a validation data set, followed by experimental validation using high-resolution photomicrographs from stirred tank protein crystallization processes.


Subject(s)
Machine Learning , Neural Networks, Computer , Algorithms , Crystallization , Image Processing, Computer-Assisted/methods , Proteins
4.
SLAS Discov ; 26(8): 961-973, 2021 09.
Article in English | MEDLINE | ID: mdl-34308708

ABSTRACT

Acoustic droplet ejection (ADE)-open port interface (OPI)-mass spectrometry (MS) has recently been introduced as a versatile analytical method that combines fast and contactless acoustic sampling with sensitive and accurate electrospray ionization (ESI)-MS-based analyte detection. The potential of the technology to provide label-free measurements in subsecond analytical cycle times makes it an attractive option for high-throughput screening (HTS). Here, we report the first implementation of ADE-OPI-MS in a fully automated HTS environment, based on the example of a biochemical assay aiming at the identification of small-molecule inhibitors of the cyclic guanosine monophosphate-adenosine monophosphate (GMP-AMP) synthase (cGAS). First, we describe the optimization of the method to enable sensitive and accurate determination of enzyme activity and inhibition in miniaturized 1536-well microtiter plate format. Then we show both results from a validation single-concentration screen using a test set of 5500 compounds, and the subsequent concentration-response testing of selected hits in direct comparison with a previously established matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) readout. Finally, we present the development of an in-line OPI cleaning procedure aiming to match the instrument robustness required for large-scale HTS campaigns. Overall, this work points to critical method development parameters and provides guidance for the establishment of integrated ADE-OPI-MS as HTS-compatible technology for early drug discovery.


Subject(s)
Automation, Laboratory , Drug Discovery/methods , High-Throughput Screening Assays/methods , Mass Spectrometry/methods , Drug Discovery/standards , High-Throughput Screening Assays/standards , Humans , Mass Spectrometry/standards , Spectrometry, Mass, Electrospray Ionization/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
5.
Rapid Commun Mass Spectrom ; 35(12): e9096, 2021 Jun 30.
Article in English | MEDLINE | ID: mdl-33837598

ABSTRACT

RATIONALE: The low speed and low flexibility of most liquid chromatography/tandem mass spectrometry (LC/MS/MS) approaches in early drug discovery delay sample analysis from routine in vivo studies within the same day. A high-throughput platform for the rapid quantification of drug compounds in various in vivo assays was developed and established in routine bioanalysis. METHODS: Automated selection of an efficient and adequate LC method was realized by autonomous sample qualification for ultrafast batch gradients (9 s/sample) or for fast linear gradients (45 s/sample) if samples required chromatography. The hardware and software components of our Rapid and Integrated Analysis System (RIAS) were streamlined for increased analytical throughput via state-of-the-art automation while maintaining high analytical quality. RESULTS: Online decision-making was based on a quick assay suitability test (AST), based on a small and dedicated sample set evaluated by two different strategies. 84% of the acquired data points were within ±30% accuracy and 93% of the deviations between the lower limit of quantitation (LLOQ) values were ≤2-fold compared with standard LC/MS/MS systems. Speed, flexibility and overall automation significantly improved. CONCLUSIONS: The developed platform provided an analysis time of only 10 min (batch-mode) and 47 min (gradient-mode) per standard pharmacokinetic (PK) study (62 injections). Automation, data evaluation and results handling were optimized to pave the way for machine learning based on decision-making regarding the evaluation strategy of the AST.


Subject(s)
Chromatography, High Pressure Liquid/methods , Drug Discovery/methods , Machine Learning , Tandem Mass Spectrometry/methods , Automation , High-Throughput Screening Assays/methods , Limit of Detection , Pharmaceutical Preparations/analysis
6.
SLAS Discov ; 26(1): 44-57, 2021 01.
Article in English | MEDLINE | ID: mdl-33073664

ABSTRACT

Demonstration of in vitro target engagement for small-molecule ligands by measuring binding to a molecular target is an established approach in early drug discovery and a pivotal step in high-throughput screening (HTS)-based compound triaging. We describe the setup, evaluation, and application of a ligand binding assay platform combining automated affinity selection (AS)-based sample preparation and label-free matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) analysis. The platform enables mass spectrometry (MS)-based HTS for small-molecule target interactions from single-compound incubation mixtures and is embedded into a regular assay automation environment. Efficient separation of target-ligand complexes is achieved by in-plate size exclusion chromatography (SEC), and small-molecule ligands are subsequently identified by MALDI-TOF analysis. In contrast to alternative HTS-capable binding assay formats, MALDI-TOF AS-MS is capable of identifying orthosteric and allosteric ligands, as shown for the model system protein tyrosine phosphatase 1B (PTP1B), irrespective of protein function. Furthermore, determining relative binding affinities (RBAs) enabled ligand ranking in accordance with functional inhibition and reference data for PTP1B and a number of diverse protein targets. Finally, we present a validation screen of more than 23,000 compounds within 24 h, demonstrating the general applicability of the platform for the HTS-compatible assessment of protein-ligand interactions.


Subject(s)
Drug Discovery/methods , High-Throughput Screening Assays/methods , Proteins/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Automation, Laboratory , Humans , Ligands
7.
Anal Chem ; 92(18): 12242-12249, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32786476

ABSTRACT

We present an acoustic ejection mass spectrometry (AEMS) setup for contactless electrospray ionization mass spectrometry (ESI-MS)-based sample injection at a sampling rate faster than current ESI and matrix-assisted laser desorption ionization (MALDI) techniques. For the direct transfer of samples out of 384-well plates into a modified ESI source, an open port interface (OPI) was combined with a modified acoustic droplet ejection (ADE) system. AEMS has the potential to eliminate bottlenecks known from classical MS approaches, such as speed, reproducibility, carryover, ion suppression, as well as sample preparation and consumption. This setup provided a drastically reduced transfer distance between OPI and ESI electrode for optimum throughput performance and broadens the scope of applications for this emerging technique. To simulate label-free applications of drug metabolism and pharmacokinetics (DMPK) analysis and high-throughput screening (HTS) campaigns, two stress tests were performed regarding ion suppression and system endurance in combination with minor sample preparation. The maximum sampling rate was 6 Hz for dextromethorphan and d3-dextrorphan (each 100 nM) for 1152 injections in 63 s at full width at half-maximum (FWHM) of 105 ms and a relative standard deviation (%RSD) of 7.7/7.5% without internal standard correction. Enzyme assay buffer and crude dog plasma caused signal suppression of 51/73% at %RSD of 5.7/6.7% (n = 120). An HTS endurance buffer was used for >25 000 injections with minor OPI pollution and constant signals (%RSD = 8.5%, FWHM of 177 ms ± 8.5%, n = 10 557). The optimized hardware and method setup resulted in high-throughput performance and enables further implementation in a fully automated platform for ESI-MS-based high-throughput screening.


Subject(s)
Acoustics , Cytochrome P-450 Enzyme System/blood , Dextromethorphan/analysis , Dextrorphan/analysis , High-Throughput Screening Assays , Animals , Cytochrome P-450 Enzyme System/metabolism , Dogs , Electrodes , Female , High-Throughput Screening Assays/instrumentation , Male , Particle Size , Spectrometry, Mass, Electrospray Ionization/instrumentation , Time Factors
8.
Sci Rep ; 10(1): 13022, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32747751

ABSTRACT

In order to overcome the challenges associated with a limited number of airway epithelial cells that can be obtained from clinical sampling and their restrained capacity to divide ex vivo, miniaturization of respiratory drug discovery assays is of pivotal importance. Thus, a 96-well microplate system was developed where primary human small airway epithelial (hSAE) cells were cultured at an air-liquid interface (ALI). After four weeks of ALI culture, a pseudostratified epithelium containing basal, club, goblet and ciliated cells was produced. The 96-well ALI cultures displayed a cellular composition, ciliary beating frequency, and intercellular tight junctions similar to 24-well conditions. A novel custom-made device for 96-parallelized transepithelial electric resistance (TEER) measurements, together with dextran permeability measurements, confirmed that the 96-well culture developed a tight barrier function during ALI differentiation. 96-well hSAE cultures were responsive to transforming growth factor ß1 (TGF-ß1) and tumor necrosis factor α (TNF-α) in a concentration dependent manner. Thus, the miniaturized cellular model system enables the recapitulation of a physiologically responsive, differentiated small airway epithelium, and a robotic integration provides a medium throughput approach towards pharmaceutical drug discovery, for instance, in respect of fibrotic distal airway/lung diseases.


Subject(s)
Bronchioles/cytology , Epithelial Cells/cytology , Miniaturization/instrumentation , Miniaturization/methods , Models, Biological , Air , Automation , Biomarkers/metabolism , Cells, Cultured , Fibrosis , Humans , Respiratory Mucosa/cytology
9.
Sensors (Basel) ; 21(1)2020 Dec 29.
Article in English | MEDLINE | ID: mdl-33383748

ABSTRACT

Vehicle-to-everything (V2X) communication is seen as one of the main enabling technologies for automated vehicles. Collective perception is especially promising, as it allows connected traffic participants to "see through the eyes of others" by sharing sensor-detected objects via V2X communication. Its benefit is typically assessed in terms of the increased object update rate, redundancy, and awareness. To determine the safety improvement thanks to collective perception, the authors introduce new metrics, which quantify the environmental risk awareness of the traffic participants. The performance of the V2X service is then analyzed with the help of the test platform TEPLITS, using real traffic traces from German highways, amounting to over 100 h of total driving time. The results in the considered scenarios clearly show that collective perception not only contributes to the accuracy and integrity of the vehicles' environmental perception, but also that a V2X market penetration of at least 25% is necessary to increase traffic safety from a "risk of serious traffic accidents" to a "residual hypothetical risk of collisions without minor injuries" for traffic participants equipped with non-redundant 360° sensor systems. These results support the ongoing worldwide standardization efforts of the collective perception service.

10.
SLAS Discov ; 25(4): 372-383, 2020 04.
Article in English | MEDLINE | ID: mdl-31583948

ABSTRACT

Comprehensive and unbiased detection methods are a prerequisite for high-throughput screening (HTS) campaigns within drug discovery research. Label-free matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry (MS) has been introduced as an HTS-compatible readout for biochemical test systems to support the drug discovery process. So far, reported HTS applications were based on surface-modified systems or proof-of-concept studies. We present the utilization of a MALDI-TOF-based screening platform to identify inhibitors of human cyclic GMP-AMP synthase (cGAS), a mediator of innate immune response whose aberration has been causally correlated to a number of inflammatory disorders. In this context, the development and validation of a MALDI-TOF-based activity assay is reported to demonstrate fast, robust, and accurate detection of chemical cGAS inhibition by direct quantification of the physiological reaction product cyclic GMP-ATP (cGAMP). Results from a screen of a diverse library of more than 1 million small molecules in 1536-well format against the catalytic cGAS activity are presented with excellent assay performance and data quality. Identified hits were qualified in dose-response experiments and confirmed by RapidFire-MS measurements. Conclusively, the presented data provide the first proof of applicability of direct automated MALDI-TOF MS as a readout strategy for large-scale drug discovery HTS campaigns.


Subject(s)
DNA/genetics , High-Throughput Screening Assays , Nucleotidyltransferases/antagonists & inhibitors , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Cytosol/enzymology , DNA/drug effects , Drug Discovery , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Humans , Nucleotidyltransferases/genetics , Small Molecule Libraries/pharmacology
11.
SLAS Discov ; 24(7): 766-777, 2019 08.
Article in English | MEDLINE | ID: mdl-31059309

ABSTRACT

Microbial-dependent trimethylamine (TMA) generation from dietary precursors such as choline was recently linked to cardiovascular diseases (CVDs) as well as chronic kidney disease (CKD). Inhibition of TMA-generating enzymes in gut bacteria would be an innovative approach to treat these diseases. The potential to accurately quantify secreted TMA levels highlights the capacity of mass spectrometry (MS) for tracking microbial TMA-lyase activity. However, high-throughput screening (HTS) by conventional MS instrumentation is hampered by limited sample throughput. Recent advancement in liquid handling and instrumentation of matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) MS provides an HTS-compatible MS technology. The deciphering of enzymatic reactions using this label-free readout has been successfully applied but has thus far been limited to peptide/protein-centric activity assays. Here, we demonstrate the versatile applicability of MALDI-TOF by tracking a small molecule within a highly complex sample background. The key to success for this concept was chemical derivatization of the target molecule enabling quantitative assessment of microbial TMA formation. Further, its potential was demonstrated in a side-by-side comparison to RapidFire-MS in a primary screen and subsequent dose-response experiments. Overall, the established assay enables the screening for microbial TMA-lyase inhibitors and serves as a proof of concept for the applicability of MALDI-TOF for demanding assay concepts per se.


Subject(s)
Drug Discovery/methods , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Lyases/antagonists & inhibitors , Methylamines/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Humans
12.
Methods Mol Biol ; 1994: 101-115, 2019.
Article in English | MEDLINE | ID: mdl-31124108

ABSTRACT

In drug discovery, there is an increasing demand for more physiological in vitro models that recapitulate the disease situation in patients. Human induced pluripotent stem (hiPS) cell-derived model cells could serve this purpose. To date, several directed differentiation approaches have been described to generate definitive endoderm (DE) from hiPS cells, but protocols suitable for drug development and high-throughput screening (HTS) have not been reported yet. In this work, a large-scale expansion of hiPS cells for high-throughput adaption is presented and an optimized stepwise differentiation of hiPS cells into DE cells is described. The produced DE cells were demonstrated to express classical DE markers on the gene expression and protein level. The here described DE cells are multipotent progenitors and act as starting points for a broad spectrum of endodermal model cells in HTS and other areas of drug discovery.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Endoderm/cytology , Induced Pluripotent Stem Cells/drug effects , Cell Differentiation/genetics , Cell Line , Drug Discovery , Endoderm/metabolism , High-Throughput Screening Assays , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism
13.
SLAS Technol ; 24(4): 386-393, 2019 08.
Article in English | MEDLINE | ID: mdl-30698995

ABSTRACT

Label-free in vitro potency assays are an emerging field in drug discovery to enable more physiological conditions, to improve the readout quality, and to save time. For this approach mass spectrometry (MS) is a powerful technology to directly follow physiological processes. The speed of this methodology, however, was for a long time not compatible with chemiluminescence- or fluorescence-based assays. Recent advances in matrix-assisted laser desorption/ionization (MALDI) instrumentation paved the way for high-throughput MS analysis of label-free assays for large compound libraries, whereas electrospray ionization (ESI)-based mass spectrometers equipped with RapidFire autosamplers were limited to medium throughput. Here we present a technological advancement of the RapidFire device to enable cycle times of 2.5 s per sample. This newly developed BLAZE-mode substantially boosted the ESI-MS analysis speed, providing an alternative technology for label-free high-throughput screening.


Subject(s)
Automation, Laboratory/methods , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Spectrometry, Mass, Electrospray Ionization/methods , Automation, Laboratory/instrumentation , High-Throughput Screening Assays/instrumentation
14.
SLAS Technol ; 24(2): 209-221, 2019 04.
Article in English | MEDLINE | ID: mdl-30074850

ABSTRACT

Label-free, mass spectrometric (MS) deciphering of enzymatic reactions by direct analysis of substrate-to-product conversion provides the next step toward more physiological relevant assays within drug discovery campaigns. Reduced risk of suffering from compound interference combined with diminished necessity for tailored signal mediators emphasizes the valuable role of label-free readouts. However, MS-based detection has not hitherto met high-throughput screening (HTS) requirements because of the lack of HTS-compatible sample introduction. In the present study, we report on a fully automated liquid-handling concept built in-house to concatenate biochemical assays with matrix-assisted laser desorption/ionization time-of-flight closing this technological gap. The integrated reformatting from 384- to 1536-well format enables cycle times of 0.6 s/sample for automated spotting and 0.4 s/sample for MS analysis, matching the requirements of HTS compatibility. In-depth examination of spotting quality, quantification accuracy, and instrument robustness together with the implementation of a protein tyrosine phosphatase 1B (PTP1B) inhibitor screening (4896 compounds) demonstrate the potential of the heavily inquired HTS integration of the label-free MS readout. Overall, the presented data demonstrate that the introduced automation concept makes label-free MS-based readouts accessible for HTS within drug discovery campaigns but also in other research areas requiring ultrafast MS-based detection.


Subject(s)
Drug Discovery/methods , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Drug Discovery/instrumentation , Drug Evaluation, Preclinical/instrumentation , High-Throughput Screening Assays/instrumentation , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/instrumentation
15.
Drug Discov Today ; 24(2): 526-532, 2019 02.
Article in English | MEDLINE | ID: mdl-30359770

ABSTRACT

A fierce dispute has arisen between the supporters of phenotypic and target-focused screening regarding which path grants the higher probability of successful drug development. A chance to reconcile these two approaches lies in successful target deconvolution (TD) after phenotypic screens. But, despite the panoply of available in vitro TD methods, the task of matching a phenotypically active compound with a biomolecular target remains challenging. Consequently, this review details the latest developments of in silico techniques that expedite TD. Ultimately, the deconvoluted target allows us to reap the benefits of the phenotypic and target-focused approaches.


Subject(s)
Drug Discovery , Phenotype , Humans
16.
SLAS Discov ; 23(6): 561-573, 2018 07.
Article in English | MEDLINE | ID: mdl-29466676

ABSTRACT

Label-free, mass spectrometric (MS) detection is an emerging technology in the field of drug discovery. Unbiased deciphering of enzymatic reactions is a proficient advantage over conventional label-based readouts suffering from compound interference and intricate generation of tailored signal mediators. Significant evolvements of matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) MS, as well as associated liquid handling instrumentation, triggered extensive efforts in the drug discovery community to integrate the comprehensive MS readout into the high-throughput screening (HTS) portfolio. Providing speed, sensitivity, and accuracy comparable to those of conventional, label-based readouts, combined with merits of MS-based technologies, such as label-free parallelized measurement of multiple physiological components, emphasizes the advantages of MALDI-TOF for HTS approaches. Here we describe the assay development for the identification of protein tyrosine phosphatase 1B (PTP1B) inhibitors. In the context of this precious drug target, MALDI-TOF was integrated into the HTS environment and cross-compared with the well-established AlphaScreen technology. We demonstrate robust and accurate IC50 determination with high accordance to data generated by AlphaScreen. Additionally, a tailored MALDI-TOF assay was developed to monitor compound-dependent, irreversible modification of the active cysteine of PTP1B. Overall, the presented data proves the promising perspective for the integration of MALDI-TOF into drug discovery campaigns.


Subject(s)
Drug Discovery/methods , Enzyme Inhibitors/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , High-Throughput Screening Assays/methods
17.
Drug Metab Dispos ; 46(2): 166-177, 2018 02.
Article in English | MEDLINE | ID: mdl-29212823

ABSTRACT

Deficient functional expression of drug transporters incapacitates most hepatic cell lines as a reliable tool for evaluating transporter-mediated drug-drug interactions. Recently, genetically modified cells (referred to as upcyte hepatocytes) have emerged as an expandable, noncancerous source of human hepatic cells. Herein, we quantified mRNA and protein levels of key hepatobiliary transporters and we assessed associated uptake activity in short- and long-term cultures of upcyte human hepatocytes (UHH) in comparison to cryopreserved primary human hepatocytes (cPHH). Expression of canalicular efflux pumps, such as MRD1/ABCB1, MATE1/SLC47A1, and MRP2/ABCC2, was relatively well preserved in UHH. By contrast, long-term cultivation of UHH in a two-dimensional sandwich configuration [sandwich-cultured upcyte human hepatocytes (SCUHH)] was required to upregulate organic anion-transporting polypeptide OATP1B1/SLCO1B1, OATP2B1/SLCO2B1, NTCP/SLC10A1, and OCT1/SLC22A1 mRNA expression, which correlated well with respective protein abundances. However, mRNA and protein levels of sinusoidal solute carrier transporters, except for NTCP and OATP2B1, remained low in SCUHH compared to sandwich-cultured cPHH. OCT1- and NTCP-mediated uptake of N-methyl-4-phenylpyridinium acetate and taurocholate was demonstrated in both hepatic models, whereas active uptake of OATP1B1/1B3-selective marker substrates, paralleled by markedly reduced SLCO1B1/1B3 expression, were not detectable in SCUHH. Uptake studies under Na+-depletion and excess of taurocholate confirmed the presence of functional NTCP protein and indicated that NTCP, apart from OATP2B1, contributed substantially to the overall hepatic uptake of rosuvastatin in SCUHH. In conclusion, our data suggest that SCUHH, despite their limitation for evaluating OATP1B1/1B3-mediated transport processes, retain NTCP, OATP2B1, and OCT1 transport activities and thus may be considered as a tool for elucidating compensatory uptake pathways for OATP1B1/1B3 substrates.


Subject(s)
Biological Transport/physiology , Hepatocytes/metabolism , Liver/metabolism , Organic Anion Transporters/metabolism , Adult , Cell Culture Techniques/methods , Drug Interactions/physiology , Female , Humans , Male , Middle Aged , Multidrug Resistance-Associated Protein 2 , RNA, Messenger/metabolism
18.
SLAS Discov ; 22(4): 425-432, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28328321

ABSTRACT

Autotaxin (ATX) is a promising drug target for the treatment of several diseases, such as cancer and fibrosis. ATX hydrolyzes lysophosphatidyl choline (LPC) into bioactive lysophosphatidic acid (LPA). The potency of ATX inhibitors can be readily determined by using fluorescence-based LPC derivatives. While such assays are ultra-high throughput, they are prone to false positives compared to assays based on natural LPC. Here we report the development of ultrafast mass spectrometry-based ATX assays enabling the measurement of data points within 13 s, which is 10 times faster than classic liquid chromatography-mass spectrometry. To this end, we set up a novel in vitro and whole-blood assay. We demonstrate that the potencies determined with these assays are in good agreement with the in vivo efficacy and that the whole-blood assay has the best predictive power. This high-throughput label-free approach paired with the translatable data quality is highly attractive for appropriate guidance of medicinal chemists for constructing strong structure-activity relationships.


Subject(s)
Enzyme Inhibitors/blood , High-Throughput Screening Assays , Lysophosphatidylcholines/blood , Lysophospholipids/blood , Mass Spectrometry/methods , Phosphoric Diester Hydrolases/blood , Animals , Dogs , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Haplorhini , Humans , Hydrolysis , Lysophosphatidylcholines/chemistry , Lysophospholipids/antagonists & inhibitors , Lysophospholipids/chemistry , Rats , Rats, Wistar , Recombinant Proteins/blood
19.
J Pharmacol Exp Ther ; 357(3): 554-61, 2016 06.
Article in English | MEDLINE | ID: mdl-27048659

ABSTRACT

BI 1002494 [(R)-4-{(R)-1-[7-(3,4,5-trimethoxy-phenyl)-[1,6]napthyridin-5-yloxy]-ethyl}pyrrolidin-2-one] is a novel, potent, and selective spleen tyrosine kinase (SYK) inhibitor with sustained plasma exposure after oral administration in rats, which qualifies this molecule as a good in vitro and in vivo tool compound. BI 1002494 exhibits higher potency in inhibiting high-affinity IgE receptor-mediated mast cell and basophil degranulation (IC50 = 115 nM) compared with B-cell receptor-mediated activation of B cells (IC50 = 810 nM). This may be explained by lower kinase potency when the physiologic ligand B-cell linker was used, suggesting that SYK inhibitors may exhibit differential potency depending on the cell type and the respective signal transduction ligand. A 3-fold decrease in potency was observed in rat basophils (IC50 = 323 nM) compared with human basophils, but a similar species potency shift was not observed in B cells. The lower potency in rat basophils was confirmed in both ex vivo inhibition of bronchoconstriction in precision-cut rat lung slices and in reversal of anaphylaxis-driven airway resistance in rats. The different cellular potencies translated into different in vivo efficacy; full efficacy in a rat ovalbumin model (that contains an element of mast cell dependence) was achieved with a trough plasma concentration of 340 nM, whereas full efficacy in a rat collagen-induced arthritis model (that contains an element of B-cell dependence) was achieved with a trough plasma concentration of 1400 nM. Taken together, these data provide a platform from which different estimates of human efficacious exposures can be made according to the relevant cell type for the indication intended to be treated.


Subject(s)
B-Lymphocytes/drug effects , B-Lymphocytes/enzymology , Basophils/drug effects , Basophils/enzymology , Naphthyridines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrrolidines/pharmacology , Pyrrolidinones/pharmacology , Syk Kinase/antagonists & inhibitors , Administration, Oral , Animals , Humans , Male , Mast Cells/drug effects , Mast Cells/enzymology , Naphthyridines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyrrolidines/administration & dosage , Pyrrolidinones/administration & dosage , Rats
20.
Drug Metab Dispos ; 44(3): 435-44, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26712819

ABSTRACT

In vitro models based on primary human hepatocytes (PHH) have been advanced for clearance (CL) prediction of metabolically stable compounds, representing state-of-the-art assay systems for drug discovery and development. Yet, limited cell availability and large interindividual variability of metabolic profiles remain shortcomings of PHH. Upcyte human hepatocytes (UHH) represent a novel hepatic cell system derived from PHH, exhibiting proliferative capacity for approximately 35 population doublings. UHH from three donors were evaluated during culture for up to 18 days, investigating relative mRNA expression and in situ enzyme activity of cytochrome P450s (P450s), UDP-glucuronosyltransferases, and sulfotransferases. Furthermore, UHH were used for predicting hepatic CL of 21 marketed low to intermediate CL drugs. In a typical experiment, expansion from 3.9 × 10(6) up to 8.5 × 10(7) cells was achieved during subculture. When maintained at confluence, transcripts of major P450s were expressed at donor-specific levels with sustained activities for the majority of isoforms, showing generally low CYP1A2 and high CYP2B6 activity levels. For donor 151-03, CL prediction based on depletion experiments resulted in an average fold error of 2.0, and 80% of compounds being predicted within twofold to in vivo CL for a subset of 10 low CL drugs. UHH showed sustained and consistent activity of drug-metabolizing enzymes (DME), resulting in highly reproducible CL prediction performance. In conclusion, UHH show promising potential as alternative to PHH for standardized in vitro applications in discovery research based on their stable, hepatocyte-like DME phenotype and virtually unlimited cell availability.


Subject(s)
Hepatocytes/metabolism , Inactivation, Metabolic/physiology , Liver/physiology , Metabolic Clearance Rate/physiology , Adult , Cells, Cultured , Cytochrome P-450 Enzyme System/metabolism , Female , Glucuronosyltransferase/metabolism , Humans , Kinetics , Liver/metabolism , Male , Middle Aged , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...