Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where André Mateus is active.

Publication


Featured researches published by André Mateus.


Molecular Pharmaceutics | 2013

Rapid measurement of intracellular unbound drug concentrations.

André Mateus; Pär Matsson; Per Artursson

Intracellular unbound drug concentrations determine affinity to targets in the cell interior. However, due to difficulties in measuring them, they are often overlooked in pharmacology. Here we present a simple experimental technique for the determination of unbound intracellular drug concentrations in cultured cells that is based on parallel measurements of cellular drug binding and steady-state intracellular drug concentrations. Binding in HEK293 cells was highly correlated with binding in liver-derived systems, whereas binding in plasma did not compare well with cellular binding. Compound lipophilicity increased drug binding, while negative charge and aromatic functional groups decreased binding. Intracellular accumulation of unbound drug was consistent with pH-dependent subcellular sequestration, as confirmed by modeling and by inhibition of subcellular pH gradients. The approach developed here can be used to measure intracellular unbound drug concentrations in more complex systems, for example, cell lines with controlled expression of transporters and enzymes or primary cells.


Nature Communications | 2016

CETSA screening identifies known and novel thymidylate synthase inhibitors and slow intracellular activation of 5-fluorouracil

Helena Almqvist; Hanna Axelsson; Rozbeh Jafari; Chen Dan; André Mateus; Martin Haraldsson; Andreas Larsson; Daniel Martinez Molina; Per Artursson; Thomas Lundbäck; Pär Nordlund

Target engagement is a critical factor for therapeutic efficacy. Assessment of compound binding to native target proteins in live cells is therefore highly desirable in all stages of drug discovery. We report here the first compound library screen based on biophysical measurements of intracellular target binding, exemplified by human thymidylate synthase (TS). The screen selected accurately for all the tested known drugs acting on TS. We also identified TS inhibitors with novel chemistry and marketed drugs that were not previously known to target TS, including the DNA methyltransferase inhibitor decitabine. By following the cellular uptake and enzymatic conversion of known drugs we correlated the appearance of active metabolites over time with intracellular target engagement. These data distinguished a much slower activation of 5-fluorouracil when compared with nucleoside-based drugs. The approach establishes efficient means to associate drug uptake and activation with target binding during drug discovery.


Drug Metabolism and Disposition | 2016

Mechanistic modeling of pitavastatin disposition in sandwich-cultured human hepatocytes: a proteomics-informed bottom-up approach

Anna Vildhede; André Mateus; Elin K. Khan; Yurong Lai; Maria Karlgren; Per Artursson; Maria C. Kjellsson

Isolated human hepatocytes are commonly used to predict transporter-mediated clearance in vivo. Such predictions, however, do not provide estimations of transporter contributions and consequently do not allow predictions of the outcome resulting from a change in the activity of a certain transporter, for example, by inhibition or a genetic variant with reduced function. Pitavastatin is a drug that is heavily dependent on hepatic transporters for its elimination, and it is excreted mainly as unchanged drug in the bile. For this reason, pitavastatin was used as a model drug to demonstrate the applicability of a bottom-up approach to predict transporter-mediated disposition in sandwich-cultured human hepatocytes (SCHHs), allowing for the estimation of transporter contributions. Transport experiments in transfected human embryonic kidney cells (HEK293 cell lines) and inverted membrane vesicles overexpressing each of the relevant transport proteins were used to generate parameter estimates for the mechanistic model. By adjusting for differences in transporter abundance between the in vitro systems and individual SCHH batches, the model successfully predicted time profiles of medium and intracellular accumulation. Our predictions of pitavastatin bile accumulation could not be confirmed, however, because of a very low biliary excretion of pitavastatin in relation to the hepatic uptake in our SCHHs. This study is, to our knowledge, the first to successfully simulate transporter-mediated processes in a complex system such as SCHHs at the level of individual transport proteins using a bottom-up approach.


Journal of Biomolecular Screening | 2016

Direct Measurement of Intracellular Compound Concentration by RapidFire Mass Spectrometry Offers Insights into Cell Permeability

Laurie J. Gordon; Morven Allen; Per Artursson; Michael M. Hann; Bill Leavens; André Mateus; Simon A. Readshaw; Klara Valko; Gareth Wayne; Andrew West

One of the key challenges facing early stage drug discovery is understanding the commonly observed difference between the activity of compounds in biochemical assays and cellular assays. Traditionally, indirect or estimated cell permeability measurements such as estimations from logP or artificial membrane permeability are used to explain the differences. The missing link is a direct measurement of intracellular compound concentration in whole cells. This can, in some circumstances, be estimated from the cellular activity, but this may also be problematic if cellular activity is weak or absent. Advances in sensitivity and throughput of analytical techniques have enabled us to develop a high-throughput assay for the measurement of intracellular compound concentration for routine use to support lead optimization. The assay uses a RapidFire-MS based readout of compound concentration in HeLa cells following incubation of cells with test compound. The initial assay validation was performed by ultra-high performance liquid chromatography tandem mass spectrometry, and the assay was subsequently transferred to RapidFire tandem mass spectrometry. Further miniaturization and optimization were performed to streamline the process, increase sample throughput, and reduce cycle time. This optimization has delivered a semi-automated platform with the potential of production scale compound profiling up to 100 compounds per day.


Journal of Medicinal Chemistry | 2014

A High-Throughput Cell-Based Method to Predict the Unbound Drug Fraction in the Brain

André Mateus; Par̈ Matsson; Per Artursson

Optimization of drug efficacy in the brain requires understanding of the local exposure to unbound drug at the site of action. This relies on measurements of the unbound drug fraction (fu,brain), which currently requires access to brain tissue. Here, we present a novel methodology using homogenates of cultured cells for rapid estimation of fu,brain. In our setup, drug binding to human embryonic kidney cell (HEK293) homogenate was measured in a small-scale dialysis apparatus. To increase throughput, we combined drugs into cassettes for simultaneous measurement of multiple compounds. Our method estimated fu,brain with an average error of 1.9-fold. We propose that our simple method can be used as an inexpensive, easily available and high-throughput alternative to brain tissues excised from laboratory animals. Thereby, estimates of unbound drug exposure can now be implemented at a much earlier stage of the drug discovery process, when molecular property changes are easier to make.


Scientific Reports | 2017

Intracellular drug bioavailability: a new predictor of system dependent drug disposition

André Mateus; Andrea Treyer; Christine Wegler; Maria Karlgren; Pär Matsson; Per Artursson

Intracellular drug exposure is influenced by cell- and tissue-dependent expression of drug-transporting proteins and metabolizing enzymes. Here, we introduce the concept of intracellular bioavailability (Fic) as the fraction of extracellular drug available to bind intracellular targets, and we assess how Fic is affected by cellular drug disposition processes. We first investigated the impact of two essential drug transporters separately, one influx transporter (OATP1B1; SLCO1B1) and one efflux transporter (P-gp; ABCB1), in cells overexpressing these proteins. We showed that OATP1B1 increased Fic of its substrates, while P-gp decreased Fic. We then investigated the impact of the concerted action of multiple transporters and metabolizing enzymes in freshly-isolated human hepatocytes in culture configurations with different levels of expression and activity of these proteins. We observed that Fic was up to 35-fold lower in the configuration with high expression of drug-eliminating transporters and enzymes. We conclude that Fic provides a measurement of the net impact of all cellular drug disposition processes on intracellular bioavailable drug levels. Importantly, no prior knowledge of the involved drug distribution pathways is required, allowing for high-throughput determination of drug access to intracellular targets in highly defined cell systems (e.g., single-transporter transfectants) or in complex ones (including primary human cells).


Proceedings of the National Academy of Sciences of the United States of America | 2017

Prediction of intracellular exposure bridges the gap between target- and cell-based drug discovery

André Mateus; Laurie J. Gordon; Gareth Wayne; Helena Almqvist; Hanna Axelsson; Brinton Seashore-Ludlow; Andrea Treyer; Pär Matsson; Thomas Lundbäck; Andrew West; Michael M. Hann; Per Artursson

Significance Exposure at the site of action has been identified as one of the three most important factors for success in drug discovery and the design of chemical probes. Modern drug discovery programs have, to a great extent, shifted to intracellular targets, but methods to determine intracellular drug concentrations have been lacking. Here, we use a methodology for predicting intracellular exposure of small-molecule drugs to understand their potency toward intracellular targets. We show that our approach is generally applicable to multiple targets, cell types, and therapeutic areas. We expect that routine measurements of intracellular drug concentration will contribute to reducing the high attrition observed in drug discovery and the design of both better chemical probes and medicines. Inadequate target exposure is a major cause of high attrition in drug discovery. Here, we show that a label-free method for quantifying the intracellular bioavailability (Fic) of drug molecules predicts drug access to intracellular targets and hence, pharmacological effect. We determined Fic in multiple cellular assays and cell types representing different targets from a number of therapeutic areas, including cancer, inflammation, and dementia. Both cytosolic targets and targets localized in subcellular compartments were investigated. Fic gives insights on membrane-permeable compounds in terms of cellular potency and intracellular target engagement, compared with biochemical potency measurements alone. Knowledge of the amount of drug that is locally available to bind intracellular targets provides a powerful tool for compound selection in early drug discovery.


Journal of Medicinal Chemistry | 2017

Identification of Triazolothiadiazoles as Potent Inhibitors of the dCTP Pyrophosphatase 1

Sabin Llona-Minguez; Andreas Höglund; Elisee Wiita; Ingrid Almlöf; André Mateus; José Manuel Calderón-Montaño; Cindy Cazares-Körner; Evert Homan; Olga Loseva; Pawel Baranczewski; Ann-Sofie Jemth; Maria Häggblad; Ulf Martens; Bo Lundgren; Per Artursson; Thomas Lundbäck; Annika Jenmalm Jensen; Ulrika Warpman Berglund; Martin Scobie; Thomas Helleday

The dCTP pyrophosphatase 1 (dCTPase) is involved in the regulation of the cellular dNTP pool and has been linked to cancer progression. Here we report on the discovery of a series of 3,6-disubstituted triazolothiadiazoles as potent dCTPase inhibitors. Compounds 16 and 18 display good correlation between enzymatic inhibition and target engagement, together with efficacy in a cellular synergy model, deeming them as a promising starting point for hit-to-lead development.


Molecular Pharmaceutics | 2018

Intracellular Drug Bioavailability: Effect of Neutral Lipids and Phospholipids

Andrea Treyer; André Mateus; Jacek R. Wiśniewski; Hinnerk Boriss; Pär Matsson; Per Artursson

Intracellular unbound drug concentrations are the pharmacologically relevant concentrations for targets inside cells. Intracellular drug concentrations are determined by multiple processes, including the extent of drug binding to intracellular structures. The aim of this study was to evaluate the effect of neutral lipid (NL) and phospholipid (PL) levels on intracellular drug disposition. The NL and/or PL content of 3T3-L1 cells were enhanced, resulting in phenotypes (in terms of morphology and proteome) reminiscent of adipocytes (high NL and PL) or mild phospholipidosis (only high PL). Intracellular bioavailability ( Fic) was then determined for 23 drugs in these cellular models and in untreated wild-type cells. A higher PL content led to higher intracellular drug binding and a lower Fic. The induction of NL did not further increase drug binding but led to altered Fic due to increased lysosomal pH. Further, there was a good correlation between binding to beads coated with pure PL and intracellular drug binding. In conclusion, our results suggest that PL content is a major determinant of drug binding in cells and that PL beads may constitute a simple alternative to estimating this parameter. Further, the presence of massive amounts of intracellular NLs did not influence drug binding significantly.


Journal of Medicinal Chemistry | 2017

Piperazin-1-ylpyridazine Derivatives Are a Novel Class of Human dCTP Pyrophosphatase 1 Inhibitors

Sabin Llona-Minguez; Andreas Höglund; Artin Ghassemian; Matthieu Desroses; José Manuel Calderón-Montaño; Estefanía Burgos Morón; Nicholas C. K. Valerie; Elisee Wiita; Ingrid Almlöf; Tobias Koolmeister; André Mateus; Cindy Cazares-Körner; Kumar Sanjiv; Evert Homan; Olga Loseva; Pawel Baranczewski; Masoud Darabi; Amir Mehdizadeh; Shabnam Fayezi; Ann-Sofie Jemth; Ulrika Warpman Berglund; Kristmundur Sigmundsson; Thomas Lundbäck; Annika Jenmalm Jensen; Per Artursson; Martin Scobie; Thomas Helleday

Collaboration


Dive into the André Mateus's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge