Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 15.096
Filter
1.
J Environ Sci (China) ; 147: 688-713, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39003083

ABSTRACT

Innately designed to induce physiological changes, pharmaceuticals are foreknowingly hazardous to the ecosystem. Advanced oxidation processes (AOPs) are recognized as a set of contemporary and highly efficient methods being used as a contrivance for the removal of pharmaceutical residues. Since reactive oxygen species (ROS) are formed in these processes to interact and contribute directly toward the oxidation of target contaminant(s), a profound insight regarding the mechanisms of ROS leading to the degradation of pharmaceuticals is fundamentally significant. The conceptualization of some specific reaction mechanisms allows the design of an effective and safe degradation process that can empirically reduce the environmental impact of the micropollutants. This review mainly deliberates the mechanistic reaction pathways for ROS-mediated degradation of pharmaceuticals often leading to complete mineralization, with a focus on acetaminophen as a drug waste model.


Subject(s)
Acetaminophen , Reactive Oxygen Species , Acetaminophen/chemistry , Reactive Oxygen Species/metabolism , Water Pollutants, Chemical/chemistry , Oxidation-Reduction , Pharmaceutical Preparations/metabolism
2.
Int J Mol Sci ; 25(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39062996

ABSTRACT

Drug-target interactions underlie the actions of chemical substances in medicine. Moreover, drug repurposing can expand use profiles while reducing costs and development time by exploiting potential multi-functional pharmacological properties based upon additional target interactions. Nonetheless, drug repurposing relies on the accurate identification and validation of drug-target interactions (DTIs). In this study, a novel drug-target interaction prediction model was developed. The model, based on an interactive inference network, contains embedding, encoding, interaction, feature extraction, and output layers. In addition, this study used Morgan and PubChem molecular fingerprints as additional information for drug encoding. The interaction layer in our model simulates the drug-target interaction process, which assists in understanding the interaction by representing the interaction space. Our method achieves high levels of predictive performance, as well as interpretability of drug-target interactions. Additionally, we predicted and validated 22 Alzheimer's disease-related targets, suggesting our model is robust and effective and thus may be beneficial for drug repurposing.


Subject(s)
Drug Repositioning , Drug Repositioning/methods , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Algorithms , Pharmaceutical Preparations/metabolism
3.
Eur J Pharm Sci ; 200: 106838, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38960205

ABSTRACT

Physiologically based pharmacokinetic (PBPK) models which can leverage preclinical data to predict the pharmacokinetic properties of drugs rapidly became an essential tool to improve the efficiency and quality of novel drug development. In this review, by searching the Application Review Files in Drugs@FDA, we analyzed the current application of PBPK models in novel drugs approved by the U.S. Food and Drug Administration (FDA) in the past five years. According to the results, 243 novel drugs were approved by the FDA from 2019 to 2023. During this period, 74 Application Review Files of novel drugs approved by the FDA that used PBPK models. PBPK models were used in various areas, including drug-drug interactions (DDI), organ impairment (OI) patients, pediatrics, drug-gene interaction (DGI), disease impact, and food effects. DDI was the most widely used area of PBPK models for novel drugs, accounting for 74.2 % of the total. Software platforms with graphical user interfaces (GUI) have reduced the difficulty of PBPK modeling, and Simcyp was the most popular software platform among applicants, with a usage rate of 80.5 %. Despite its challenges, PBPK has demonstrated its potential in novel drug development, and a growing number of successful cases provide experience learned for researchers in the industry.


Subject(s)
Drug Approval , Drug Interactions , Models, Biological , Pharmacokinetics , United States Food and Drug Administration , Humans , United States , Pharmaceutical Preparations/metabolism , Animals
4.
SLAS Discov ; 29(5): 100170, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38964171

ABSTRACT

The importance of a drug's kinetic profile and interplay of structure-kinetic activity with PK/PD has long been appreciated in drug discovery. However, technical challenges have often limited detailed kinetic characterization of compounds to the latter stages of projects. This review highlights the advances that have been made in recent years in techniques, instrumentation, and data analysis to increase the throughput of detailed kinetic and mechanistic characterization, enabling its application earlier in the drug discovery process.


Subject(s)
Drug Discovery , High-Throughput Screening Assays , Drug Discovery/methods , Kinetics , High-Throughput Screening Assays/methods , Humans , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Structure-Activity Relationship
5.
J Chem Inf Model ; 64(14): 5427-5438, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38976447

ABSTRACT

In drug candidate design, clearance is one of the most crucial pharmacokinetic parameters to consider. Recent advancements in machine learning techniques coupled with the growing accumulation of drug data have paved the way for the construction of computational models to predict drug clearance. However, concerns persist regarding the reliability of data collected from public sources, and a majority of current in silico quantitative structure-property relationship models tend to neglect the influence of molecular chirality. In this study, we meticulously examined human liver microsome (HLM) data from public databases and constructed two distinct data sets with varying HLM data quantity and quality. Two baseline models (RF and DNN) and three chirality-focused GNNs (DMPNN, TetraDMPNN, and ChIRo) were proposed, and their performance on HLM data was evaluated and compared with each other. The TetraDMPNN model, which leverages chirality from 2D structure, exhibited the best performance with a test R2 of 0.639 and a test root-mean-squared error of 0.429. The applicability domain of the model was also defined by using a molecular similarity-based method. Our research indicates that graph neural networks capable of capturing molecular chirality have significant potential for practical application and can deliver superior performance.


Subject(s)
Microsomes, Liver , Neural Networks, Computer , Humans , Microsomes, Liver/metabolism , Stereoisomerism , Quantitative Structure-Activity Relationship , Machine Learning , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism
6.
J Chem Inf Model ; 64(14): 5646-5656, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38976879

ABSTRACT

Predicting drug-target interactions (DTIs) is one of the crucial tasks in drug discovery, but traditional wet-lab experiments are costly and time-consuming. Recently, deep learning has emerged as a promising tool for accelerating DTI prediction due to its powerful performance. However, the models trained on limited known DTI data struggle to generalize effectively to novel drug-target pairs. In this work, we propose a strategy to train an ensemble of models by capturing both domain-generic and domain-specific features (E-DIS) to learn diverse domain features and adapt them to out-of-distribution data. Multiple experts were trained on different domains to capture and align domain-specific information from various distributions without accessing any data from unseen domains. E-DIS provides a comprehensive representation of proteins and ligands by capturing diverse features. Experimental results on four benchmark data sets in both in-domain and cross-domain settings demonstrated that E-DIS significantly improved model performance and domain generalization compared to existing methods. Our approach presents a significant advancement in DTI prediction by combining domain-generic and domain-specific features, enhancing the generalization ability of the DTI prediction model.


Subject(s)
Deep Learning , Drug Discovery , Proteins , Drug Discovery/methods , Proteins/chemistry , Proteins/metabolism , Ligands , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Protein Domains
7.
Pharm Res ; 41(7): 1391-1400, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38981900

ABSTRACT

PURPOSE: Evaluation of distribution kinetics is a neglected aspect of pharmacokinetics. This study examines the utility of the model-independent parameter whole body distribution clearance (CLD) in this respect. METHODS: Since mammillary compartmental models are widely used, CLD was calculated in terms of parameters of this model for 15 drugs. The underlying distribution processes were explored by assessment of relationships to pharmacokinetic parameters and covariates. RESULTS: The model-independence of the definition of the parameter CLD allowed a comparison of distributional properties of different drugs and provided physiological insight. Significant changes in CLD were observed as a result of drug-drug interactions, transporter polymorphisms and a diseased state. CONCLUSION: Total distribution clearance CLD is a useful parameter to evaluate distribution kinetics of drugs. Its estimation as an adjunct to the model-independent parameters clearance and steady-state volume of distribution is advocated.


Subject(s)
Metabolic Clearance Rate , Models, Biological , Pharmacokinetics , Humans , Pharmaceutical Preparations/metabolism , Drug Interactions , Tissue Distribution
8.
Protein Sci ; 33(8): e5116, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38979784

ABSTRACT

Interactions between proteins and small organic compounds play a crucial role in regulating protein functions. These interactions can modulate various aspects of protein behavior, including enzymatic activity, signaling cascades, and structural stability. By binding to specific sites on proteins, small organic compounds can induce conformational changes, alter protein-protein interactions, or directly affect catalytic activity. Therefore, many drugs available on the market today are small molecules (72% of all approved drugs in the last 5 years). Proteins are composed of one or more domains: evolutionary units that convey function or fitness either singly or in concert with others. Understanding which domain(s) of the target protein binds to a drug can lead to additional opportunities for discovering novel targets. The evolutionary classification of protein domains (ECOD) classifies domains into an evolutionary hierarchy that focuses on distant homology. Previously, no structure-based protein domain classification existed that included information about both the interaction between small molecules or drugs and the structural domains of a target protein. This data is especially important for multidomain proteins and large complexes. Here, we present the DrugDomain database that reports the interaction between ECOD of human target proteins and DrugBank molecules and drugs. The pilot version of DrugDomain describes the interaction of 5160 DrugBank molecules associated with 2573 human proteins. It describes domains for all experimentally determined structures of these proteins and incorporates AlphaFold models when such structures are unavailable. The DrugDomain database is available online: http://prodata.swmed.edu/DrugDomain/.


Subject(s)
Protein Domains , Proteins , Proteins/chemistry , Proteins/metabolism , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Databases, Protein , Humans , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Evolution, Molecular , Protein Binding
9.
Chem Biol Drug Des ; 104(1): e14576, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38969623

ABSTRACT

Intestinal absorption of compounds is significant in drug research and development. To evaluate this efficiently, a method combining mathematical modeling and molecular simulation was proposed, from the perspective of molecular structure. Based on the quantitative structure-property relationship study, the model between molecular structure and their apparent permeability coefficients was successfully constructed and verified, predicting intestinal absorption of drugs and interpreting decisive structural factors, such as AlogP98, Hydrogen bond donor and Ellipsoidal volume. The molecules with strong lipophilicity, less hydrogen bond donors and receptors, and small molecular volume are more easily absorbed. Then, the molecular dynamics simulation and molecular docking were utilized to study the mechanism of differences in intestinal absorption of drugs and investigate the role of molecular structure. Results indicated that molecules with strong lipophilicity and small volume interacted with the membrane at a lower energy and were easier to penetrate the membrane. Likewise, they had weaker interaction with P-glycoprotein and were easier to escape from it and harder to export from the body. More in, less out, is the main reason these molecules absorb well.


Subject(s)
Hydrogen Bonding , Intestinal Absorption , Molecular Docking Simulation , Molecular Dynamics Simulation , Quantitative Structure-Activity Relationship , Humans , Molecular Structure , Pharmaceutical Preparations/metabolism , Pharmaceutical Preparations/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , Hydrophobic and Hydrophilic Interactions , Permeability
10.
Sci Total Environ ; 946: 174361, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-38960202

ABSTRACT

Pharmaceuticals are emerging contaminants in the environment and are a ubiquitous presence in rivers downstream of wastewater treatment plant outfalls. Questions remain about the persistence of pharmaceuticals in rivers, and the uptake and bioconcentration of pharmaceuticals by aquatic plants. Our study took place in the Yarrowee/Leigh/Barwon River system in southeastern Australia. We quantified the concentrations of five pharmaceuticals (carbamazepine, primidone, propranolol, tramadol, and venlafaxine) in surface water at five sites along a 144-km stretch of river, downstream of the presumed primary point source (a wastewater treatment plant outfall). We quantified pharmaceuticals in the leaves of two aquatic plant species (Phragmites australis and Vallisneria australis) sampled at each site, and calculated bioconcentration factors. All five pharmaceuticals were detected in surface waters, and the highest detected concentration exceeded 500 ng.L-1 (tramadol). Four of the pharmaceuticals (all except tramadol) were detected and quantified at all sites, including the furthest site from the outfall (144 km). Carbamazepine showed less attenuation with distance from the outfall than the other pharmaceuticals. Carbamazepine and venlafaxine were quantified in the leaves of both aquatic plant species (range: 10-31 ng.g-1), and there was evidence that bioconcentration factors increased with decreasing surface water concentrations. The study demonstrates the potential long-distance persistence of pharmaceuticals in river systems, and the bioconcentration of pharmaceuticals by aquatic plants in natural ecosystems. These phenomena deserve greater attention as aquatic plants are a potential point of transfer of pharmaceuticals from aquatic ecosystems to terrestrial food webs.


Subject(s)
Environmental Monitoring , Rivers , Water Pollutants, Chemical , Rivers/chemistry , Water Pollutants, Chemical/analysis , Water Pollutants, Chemical/metabolism , Pharmaceutical Preparations/metabolism , Pharmaceutical Preparations/analysis , Australia
11.
J Mol Model ; 30(8): 264, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38995407

ABSTRACT

CONTEXT: Accurately predicting plasma protein binding rate (PPBR) and oral bioavailability (OBA) helps to better reveal the absorption and distribution of drugs in the human body and subsequent drug design. Although machine learning models have achieved good results in prediction accuracy, they often suffer from insufficient accuracy when dealing with data with irregular topological structures. METHODS: In view of this, this study proposes a pharmacokinetic parameter prediction framework based on graph convolutional networks (GCN), which predicts the PPBR and OBA of small molecule drugs. In the framework, GCN is first used to extract spatial feature information on the topological structure of drug molecules, in order to better learn node features and association information between nodes. Then, based on the principle of drug similarity, this study calculates the similarity between small molecule drugs, selects different thresholds to construct datasets, and establishes a prediction model centered on the GCN algorithm. The experimental results show that compared with traditional machine learning prediction models, the prediction model constructed based on the GCN method performs best on PPBR and OBA datasets with an inter-molecular similarity threshold of 0.25, with MAE of 0.155 and 0.167, respectively. In addition, in order to further improve the accuracy of the prediction model, GCN is combined with other algorithms. Compared to using a single GCN method, the distribution of the predicted values obtained by the combined model is highly consistent with the true values. In summary, this work provides a new method for improving the rate of early drug screening in the future.


Subject(s)
Machine Learning , Humans , Algorithms , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Neural Networks, Computer , Biological Availability , Protein Binding , Small Molecule Libraries/pharmacokinetics , Small Molecule Libraries/chemistry , Pharmacokinetics , Blood Proteins/metabolism
12.
Drug Metab Dispos ; 52(8): 919-931, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39013583

ABSTRACT

There is overwhelming preference for application of the unphysiologic, well-stirred model (WSM) over the parallel tube model (PTM) and dispersion model (DM) to predict hepatic drug clearance, CLH , despite that liver blood flow is dispersive and closer to the DM in nature. The reasoning is the ease in computation relating the hepatic intrinsic clearance ( CLint ), hepatic blood flow ( QH ), unbound fraction in blood ( fub ) and the transmembrane clearances ( CLin and CLef ) to CLH for the WSM. However, the WSM, being the least efficient liver model, predicts a lower EH that is associated with the in vitro CLint ( Vmax / Km ), therefore requiring scale-up to predict CLH in vivo. By contrast, the miniPTM, a three-subcompartment tank-in-series model of uniform enzymes, closely mimics the DM and yielded similar patterns for CLint versus EH , substrate concentration [S] , and KL / B , the tissue to outflow blood concentration ratio. We placed these liver models nested within physiologically based pharmacokinetic models to describe the kinetics of the flow-limited, phenolic substrate, harmol, using the WSM (single compartment) and the miniPTM and zonal liver models (ZLMs) of evenly and unevenly distributed glucuronidation and sulfation activities, respectively, to predict CLH For the same, given CLint ( Vmax and Km ), the WSM again furnished the lowest extraction ratio ( EH,WSM = 0.5) compared with the miniPTM and ZLM (>0.68). Values of EH,WSM were elevated to those for EH, PTM and EH, ZLM when the Vmax s for sulfation and glucuronidation were raised 5.7- to 1.15-fold. The miniPTM is easily manageable mathematically and should be the new normal for liver/physiologic modeling. SIGNIFICANCE STATEMENT: Selection of the proper liver clearance model impacts strongly on CLH predictions. The authors recommend use of the tank-in-series miniPTM (3 compartments mini-parallel tube model), which displays similar properties as the dispersion model (DM) in relating CLint and [ S ] to CLH as a stand-in for the DM, which best describes the liver microcirculation. The miniPTM is readily modified to accommodate enzyme and transporter zonation.


Subject(s)
Liver , Metabolic Clearance Rate , Models, Biological , Liver/metabolism , Humans , Metabolic Clearance Rate/physiology , Animals , Pharmaceutical Preparations/metabolism , Hepatobiliary Elimination/physiology , Pharmacokinetics
13.
J Bioinform Comput Biol ; 22(3): 2450010, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39030668

ABSTRACT

Drugs often target specific metabolic pathways to produce a therapeutic effect. However, these pathways are complex and interconnected, making it challenging to predict a drug's potential effects on an organism's overall metabolism. The mapping of drugs with targeting metabolic pathways in the organisms can provide a more complete understanding of the metabolic effects of a drug and help to identify potential drug-drug interactions. In this study, we proposed a machine learning hybrid model Graph Transformer Integrated Encoder (GTIE-RT) for mapping drugs to target metabolic pathways in human. The proposed model is a composite of a Graph Convolution Network (GCN) and transformer encoder for graph embedding and attention mechanism. The output of the transformer encoder is then fed into the Extremely Randomized Trees Classifier to predict target metabolic pathways. The evaluation of the GTIE-RT on drugs dataset demonstrates excellent performance metrics, including accuracy (>95%), recall (>92%), precision (>93%) and F1-score (>92%). Compared to other variants and machine learning methods, GTIE-RT consistently shows more reliable results.


Subject(s)
Computational Biology , Machine Learning , Metabolic Networks and Pathways , Humans , Computational Biology/methods , Pharmaceutical Preparations/metabolism , Algorithms , Models, Biological , Neural Networks, Computer , Drug Interactions
14.
Database (Oxford) ; 20242024 Jul 24.
Article in English | MEDLINE | ID: mdl-39049520

ABSTRACT

In vitro-in vivo extrapolation is a commonly applied technique for liver clearance prediction. Various in vitro models are available such as hepatocytes, human liver microsomes, or recombinant cytochromes P450. According to the free drug theory, only the unbound fraction (fu) of a chemical can undergo metabolic changes. Therefore, to ensure the reliability of predictions, both specific and nonspecific binding in the model should be accounted. However, the fraction unbound in the experiment is often not reported. The study aimed to provide a detailed repository of the literature data on the compound's fu value in various in vitro systems used for drug metabolism evaluation and corresponding human plasma binding levels. Data on the free fraction in plasma and different in vitro models were supplemented with the following information: the experimental method used for the assessment of the degree of drug binding, protein or cell concentration in the incubation, and other experimental conditions, if different from the standard ones, species, reference to the source publication, and the author's name and date of publication. In total, we collected 129 literature studies on 1425 different compounds. The provided data set can be used as a reference for scientists involved in pharmacokinetic/physiologically based pharmacokinetic modelling as well as researchers interested in Quantitative Structure-Activity Relationship models for the prediction of fraction unbound based on compound structure. Database URL: https://data.mendeley.com/datasets/3bs5526htd/1.


Subject(s)
Hepatocytes , Humans , Hepatocytes/metabolism , Metabolic Clearance Rate , Animals , Models, Biological , Pharmaceutical Preparations/metabolism , Protein Binding , Microsomes, Liver/metabolism
15.
Anal Chem ; 96(29): 12129-12138, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38985547

ABSTRACT

As organoids and organ-on-chip (OoC) systems move toward preclinical and clinical applications, there is an increased need for method validation. Using a liquid chromatography-mass spectrometry (LC-MS)-based approach, we developed a method for measuring small-molecule drugs and metabolites in the cell medium directly sampled from liver organoids/OoC systems. The LC-MS setup was coupled to an automatic filtration and filter flush system with online solid-phase extraction (SPE), allowing for robust and automated sample cleanup/analysis. For the matrix, rich in, e.g., protein, salts, and amino acids, no preinjection sample preparation steps (protein precipitation, SPE, etc.) were necessary. The approach was demonstrated with tolbutamide and its liver metabolite, 4-hydroxytolbutamide (4HT). The method was validated for analysis of cell media of human stem cell-derived liver organoids cultured in static conditions and on a microfluidic platform according to Food and Drug Administration (FDA) guidelines with regards to selectivity, matrix effects, accuracy, precision, etc. The system allows for hundreds of injections without replacing chromatography hardware. In summary, drug/metabolite analysis of organoids/OoCs can be performed robustly with minimal sample preparation.


Subject(s)
Liver , Organoids , Humans , Organoids/metabolism , Organoids/cytology , Chromatography, Liquid/methods , Liver/metabolism , Mass Spectrometry/methods , Tolbutamide/metabolism , Tolbutamide/analysis , Lab-On-A-Chip Devices , Pharmaceutical Preparations/metabolism , Pharmaceutical Preparations/analysis , Solid Phase Extraction , Small Molecule Libraries/analysis , Small Molecule Libraries/metabolism , Small Molecule Libraries/chemistry , Liquid Chromatography-Mass Spectrometry
16.
Clin Pharmacokinet ; 63(6): 735-749, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38867094

ABSTRACT

The renal secretion of many drugs is facilitated by membrane transporters, including organic cation transporter 2, multidrug and toxin extrusion protein 1/2-K and organic anion transporters 1 and 3. Inhibition of these transporters can reduce renal excretion of drugs and thereby pose a safety risk. Assessing the risk of inhibition of these membrane transporters by investigational drugs remains a key focus in the evaluation of drug-drug interactions (DDIs). Current methods to predict DDI risk are based on generating in vitro data followed by a clinical assessment using a recommended exogenous probe substrate for the individual drug transporter. More recently, monitoring plasma-based and urine-based endogenous biomarkers to predict transporter-mediated DDIs in early phase I studies represents a promising approach to facilitate, improve and potentially avoid conventional clinical DDI studies. This perspective reviews the evidence for use of these endogenous biomarkers in the assessment of renal transporter-mediated DDI, evaluates how endogenous biomarkers may help to expand the DDI assessment toolkit and offers some potential knowledge gaps. A conceptual framework for assessment that may complement the current paradigm of predicting the potential for renal transporter-mediated DDIs is outlined.


Subject(s)
Biomarkers , Drug Development , Drug Interactions , Membrane Transport Proteins , Humans , Drug Development/methods , Biomarkers/metabolism , Biomarkers/urine , Membrane Transport Proteins/metabolism , Drug Industry/methods , Kidney/metabolism , Kidney/drug effects , Pharmaceutical Preparations/metabolism , Animals
17.
AAPS J ; 26(4): 65, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844719

ABSTRACT

The recruitment of a parallel, healthy participants (HPs) arm in renal and hepatic impairment (RI and HI) studies is a common strategy to assess differences in pharmacokinetics. Limitations in this approach include the underpowered estimate of exposure differences and the use of the drug in a population for which there is no benefit. Recently, a method was published by Purohit et. al. (2023) that leveraged prior population pharmacokinetic (PopPK) modeling-based simulation to infer the distribution of exposure ratios between the RI/HI arms and HPs. The approach was successful, but it was a single example with a robust model having several iterations of development and fitting to extensive HP data. To test in more studies and models at different stages of development, our catalogue of RI/HI studies was searched, and those with suitable properties and from programs with available models were analyzed with the simulation approach. There were 9 studies included in the analysis. Most studies were associated with models that would have been available at the time (ATT) of the study, and all had a current, final model. For 3 studies, the HP PK was not predicted well by the ATT (2) or final (1) models. In comparison to conventional analysis of variance (ANOVA), the simulation approach provided similar point estimates and confidence intervals of exposure ratios. This PopPK based approach can be considered as a method of choice in situations where the simulation of HP data would not be an extrapolation, and when no other complicating factors are present.


Subject(s)
Computer Simulation , Healthy Volunteers , Models, Biological , Humans , Retrospective Studies , Pharmacokinetics , Liver Diseases/metabolism , Kidney Diseases , Pharmaceutical Preparations/metabolism , Pharmaceutical Preparations/administration & dosage , Renal Insufficiency/metabolism
18.
Microb Biotechnol ; 17(6): e14515, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38925623

ABSTRACT

Pharmaceuticals are of increasing environmental concern as they emerge and accumulate in surface- and groundwater systems around the world, endangering the overall health of aquatic ecosystems. Municipal wastewater discharge is a significant vector for pharmaceuticals and their metabolites to enter surface waters as humans incompletely absorb prescription drugs and excrete up to 50% into wastewater, which are subsequently incompletely removed during wastewater treatment. Microalgae present a promising target for improving wastewater treatment due to their ability to remove some pollutants efficiently. However, their inherent metabolic pathways limit their capacity to degrade more recalcitrant organic compounds such as pharmaceuticals. The human liver employs enzymes to break down and absorb drugs, and these enzymes are extensively researched during drug development, meaning the cytochrome P450 enzymes responsible for metabolizing each approved drug are well studied. Thus, unlocking or increasing cytochrome P450 expression in endogenous wastewater microalgae could be a cost-effective strategy to reduce pharmaceutical loads in effluents. Here, we discuss the challenges and opportunities associated with introducing cytochrome P450 enzymes into microalgae. We anticipate that cytochrome P450-engineered microalgae can serve as a new drug removal method and a sustainable solution that can upgrade wastewater treatment facilities to function as "mega livers".


Subject(s)
Cytochrome P-450 Enzyme System , Microalgae , Wastewater , Water Purification , Microalgae/metabolism , Microalgae/enzymology , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme System/genetics , Wastewater/chemistry , Wastewater/microbiology , Pharmaceutical Preparations/metabolism , Water Purification/methods , Water Pollutants, Chemical/metabolism , Humans , Biodegradation, Environmental
19.
Bioinformatics ; 40(6)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38837345

ABSTRACT

MOTIVATION: Accurately identifying the drug-target interactions (DTIs) is one of the crucial steps in the drug discovery and drug repositioning process. Currently, many computational-based models have already been proposed for DTI prediction and achieved some significant improvement. However, these approaches pay little attention to fuse the multi-view similarity networks related to drugs and targets in an appropriate way. Besides, how to fully incorporate the known interaction relationships to accurately represent drugs and targets is not well investigated. Therefore, there is still a need to improve the accuracy of DTI prediction models. RESULTS: In this study, we propose a novel approach that employs Multi-view similarity network fusion strategy and deep Interactive attention mechanism to predict Drug-Target Interactions (MIDTI). First, MIDTI constructs multi-view similarity networks of drugs and targets with their diverse information and integrates these similarity networks effectively in an unsupervised manner. Then, MIDTI obtains the embeddings of drugs and targets from multi-type networks simultaneously. After that, MIDTI adopts the deep interactive attention mechanism to further learn their discriminative embeddings comprehensively with the known DTI relationships. Finally, we feed the learned representations of drugs and targets to the multilayer perceptron model and predict the underlying interactions. Extensive results indicate that MIDTI significantly outperforms other baseline methods on the DTI prediction task. The results of the ablation experiments also confirm the effectiveness of the attention mechanism in the multi-view similarity network fusion strategy and the deep interactive attention mechanism. AVAILABILITY AND IMPLEMENTATION: https://github.com/XuLew/MIDTI.


Subject(s)
Computational Biology , Computational Biology/methods , Drug Discovery/methods , Algorithms , Drug Repositioning/methods , Pharmaceutical Preparations/metabolism , Pharmaceutical Preparations/chemistry , Humans
20.
Expert Opin Drug Metab Toxicol ; 20(6): 459-471, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38832686

ABSTRACT

INTRODUCTION: Advances in the accessibility of manufacturing technologies and iPSC-based modeling have accelerated the overall progress of organs-on-a-chip. Notably, the progress in multi-organ systems is not progressing with equal speed, indicating that there are still major technological barriers to overcome that may include biological relevance, technological usability as well as overall accessibility. AREAS COVERED: We here review the progress in the field of multi-tissue- and body-on-a-chip pre and post- SARS-CoV-2 pandemic and review five selected studies with increasingly complex multi-organ chips aiming at pharmacological studies. EXPERT OPINION: We discuss future and necessary advances in the field of multi-organ chips including how to overcome challenges regarding cell diversity, improved culture conditions, model translatability as well as sensor integrations to enable microsystems to cover organ-organ interactions in not only toxicokinetic but more importantly pharmacodynamic and -kinetic studies.


Subject(s)
COVID-19 , Lab-On-A-Chip Devices , Pharmacokinetics , Humans , Animals , Pharmaceutical Preparations/metabolism , Pharmaceutical Preparations/administration & dosage , Models, Biological , Microphysiological Systems
SELECTION OF CITATIONS
SEARCH DETAIL