Drug-induced cholestasis (DIC) is recognized as a major safety concern in drug development, as it represents one of the three types of drug-induced liver injury (DILI). Cholestasis is characterized by the disruption of bile flow, leading to intrahepatic accumulation of toxic bile acids. Bile acid regulation is a multifarious process, orchestrated by several hepatic mechanisms, namely sinusoidal uptake and efflux, canalicular secretion and intracellular metabolism.
View Article and Find Full Text PDFMachine learning (ML) models have become key in decision-making for many disciplines, including drug discovery and medicinal chemistry. ML models are generally evaluated prior to their usage in high-stakes decisions, such as compound synthesis or experimental testing. However, no ML model is robust or predictive in all real-world scenarios.
View Article and Find Full Text PDFAfter initial triaging using in vitro absorption, distribution, metabolism, and excretion (ADME) assays, pharmacokinetic (PK) studies are the first application of promising drug candidates in living mammals. Preclinical PK studies characterize the evolution of the compound's concentration over time, typically in rodents' blood or plasma. From this concentration-time (-) profiles, PK parameters such as total exposure or maximum concentration can be subsequently derived.
View Article and Find Full Text PDFHepatic bile acid regulation is a multifaceted process modulated by several hepatic transporters and enzymes. Drug-induced cholestasis (DIC), a main type of drug-induced liver injury (DILI), denotes any drug-mediated condition in which hepatic bile flow is impaired. Our ability in translating preclinical toxicological findings to human DIC risk is currently very limited, mainly due to important interspecies differences.
View Article and Find Full Text PDFMachine learning (ML) systems can model quantitative structure-property relationships (QSPR) using existing experimental data and make property predictions for new molecules. With the advent of modalities such as targeted protein degraders (TPD), the applicability of QSPR models is questioned and ML usage in TPD-centric projects remains limited. Herein, ML models are developed and evaluated for TPDs' property predictions, including passive permeability, metabolic clearance, cytochrome P450 inhibition, plasma protein binding, and lipophilicity.
View Article and Find Full Text PDFMost drugs are mainly metabolized by cytochrome P450 (CYP450), which can lead to drug-drug interactions (DDI). Specifically, time-dependent inhibition (TDI) of CYP3A4 isoenzyme has been associated with clinically relevant DDI. To overcome potential DDI issues, high-throughput assays were established to assess the TDI of CYP3A4 during the discovery and lead optimization phases.
View Article and Find Full Text PDFMedicinal chemistry and drug design efforts can be assisted by machine learning (ML) models that relate the molecular structure to compound properties. Such quantitative structure-property relationship models are generally trained on large data sets that include diverse chemical series (global models). In the pharmaceutical industry, these ML global models are available across discovery projects as an "out-of-the-box" solution to assist in drug design, synthesis prioritization, and experiment selection.
View Article and Find Full Text PDFIt is common practice in drug discovery and development to predict in vivo hepatic clearance from in vitro incubations with liver microsomes or hepatocytes using the well-stirred model (WSM). When applying the WSM to a set of approximately 3000 Novartis research compounds, 73% of neutral and basic compounds (extended clearance classification system [ECCS] class 2) were well-predicted within 3-fold. In contrast, only 44% (ECCS class 1A) or 34% (ECCS class 1B) of acids were predicted within 3-fold.
View Article and Find Full Text PDFExplainable machine learning is increasingly used in drug discovery to help rationalize compound property predictions. Feature attribution techniques are popular choices to identify which molecular substructures are responsible for a predicted property change. However, established molecular feature attribution methods have so far displayed low performance for popular deep learning algorithms such as graph neural networks (GNNs), especially when compared with simpler modeling alternatives such as random forests coupled with atom masking.
View Article and Find Full Text PDFMachine learning (ML) has become an indispensable tool to predict absorption, distribution, metabolism, and excretion (ADME) properties in pharmaceutical research. ML algorithms are trained on molecular structures and corresponding ADME assay data to develop quantitative structure-property relationship (QSPR) models. Traditional QSPR models were trained on compound sets of limited size.
View Article and Find Full Text PDFWe report the main conclusions of the first Chemoinformatics and Artificial Intelligence Colloquium, Mexico City, June 15-17, 2022. Fifteen lectures were presented during a virtual public event with speakers from industry, academia, and non-for-profit organizations. Twelve hundred and ninety students and academics from more than 60 countries.
View Article and Find Full Text PDFIn pharmaceutical research, compounds are optimized for metabolic stability to avoid a too fast elimination of the drug. Intrinsic clearance (CL) measured in liver microsomes or hepatocytes is an important parameter during lead optimization. In this work, machine learning models were developed to relate the compound structure to microsomal metabolic stability and predict CL for new compounds.
View Article and Find Full Text PDFGraph neural networks (GNNs) recursively propagate signals along the edges of an input graph, integrate node feature information with graph structure, and learn object representations. Like other deep neural network models, GNNs have notorious black box character. For GNNs, only few approaches are available to rationalize model decisions.
View Article and Find Full Text PDFAssessing whether compounds penetrate the brain can become critical in drug discovery, either to prevent adverse events or to reach the biological target. Generally, pre-clinical in vivo studies measuring the ratio of brain and blood concentrations () are required to estimate the brain penetration potential of a new drug entity. In this work, we developed machine learning models to predict in vivo compound brain penetration (as Log) from chemical structure.
View Article and Find Full Text PDFIn chemoinformatics and medicinal chemistry, machine learning has evolved into an important approach. In recent years, increasing computational resources and new deep learning algorithms have put machine learning onto a new level, addressing previously unmet challenges in pharmaceutical research. In silico approaches for compound activity predictions, de novo design, and reaction modeling have been further advanced by new algorithmic developments and the emergence of big data in the field.
View Article and Find Full Text PDFThe support vector machine (SVM) algorithm is one of the most widely used machine learning (ML) methods for predicting active compounds and molecular properties. In chemoinformatics and drug discovery, SVM has been a state-of-the-art ML approach for more than a decade. A unique attribute of SVM is that it operates in feature spaces of increasing dimensionality.
View Article and Find Full Text PDFAs in other areas, artificial intelligence (AI) is heavily promoted in different scientific fields, including chemistry. Although chemistry traditionally tends to be a conservative field and slower than others to adapt new concepts, AI is increasingly being investigated across chemical disciplines. In medicinal chemistry, supported by computer-aided drug design and cheminformatics, computational methods have long been employed to aid in the search for and optimization of active compounds.
View Article and Find Full Text PDFThe prediction of compound properties from chemical structure is a main task for machine learning (ML) in medicinal chemistry. ML is often applied to large data sets in applications such as compound screening, virtual library enumeration, or generative chemistry. Albeit desirable, a detailed understanding of ML model decisions is typically not required in these cases.
View Article and Find Full Text PDFMachine learning is widely applied in drug discovery research to predict molecular properties and aid in the identification of active compounds. Herein, we introduce a new approach that uses model-internal information from compound activity predictions to uncover relationships between target proteins. On the basis of a large-scale analysis generating and comparing machine learning models for more than 200 proteins, feature importance correlation analysis is shown to detect similar compound binding characteristics.
View Article and Find Full Text PDFMachine learning (ML) enables modeling of quantitative structure-activity relationships (QSAR) and compound potency predictions. Recently, multi-target QSAR models have been gaining increasing attention. Simultaneous compound potency predictions for multiple targets can be carried out using ensembles of independently derived target-based QSAR models or in a more integrated and advanced manner using multi-target deep neural networks (MT-DNNs).
View Article and Find Full Text PDFCarbonic anhydrases (CAs) catalyze the physiological hydration of carbon dioxide and are among the most intensely studied pharmaceutical target enzymes. A hallmark of CA inhibition is the complexation of the catalytic zinc cation in the active site. Human () CA isoforms belonging to different families are implicated in a wide range of diseases and of very high interest for therapeutic intervention.
View Article and Find Full Text PDFFor kinase inhibitors, X-ray crystallography has revealed different types of binding modes. Currently, more than 2000 kinase inhibitors with known binding modes are available, which makes it possible to derive and test machine learning models for the prediction of inhibitors with different binding modes. We have addressed this prediction task to evaluate and compare the information content of distinct molecular representations including protein-ligand interaction fingerprints (IFPs) and compound structure-based structural fingerprints (i.
View Article and Find Full Text PDFDifficulties in interpreting machine learning (ML) models and their predictions limit the practical applicability of and confidence in ML in pharmaceutical research. There is a need for agnostic approaches aiding in the interpretation of ML models regardless of their complexity that is also applicable to deep neural network (DNN) architectures and model ensembles. To these ends, the SHapley Additive exPlanations (SHAP) methodology has recently been introduced.
View Article and Find Full Text PDFIn qualitative or quantitative studies of structure-activity relationships (SARs), machine learning (ML) models are trained to recognize structural patterns that differentiate between active and inactive compounds. Understanding model decisions is challenging but of critical importance to guide compound design. Moreover, the interpretation of ML results provides an additional level of model validation based on expert knowledge.
View Article and Find Full Text PDFNoncovalent inhibitors of protein kinases have different modes of action. They bind to the active or inactive form of kinases, compete with ATP, stabilize inactive kinase conformations, or act through allosteric sites. Accordingly, kinase inhibitors have been classified on the basis of different binding modes.
View Article and Find Full Text PDF