Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 14.869
Filtrar
2.
AAPS J ; 26(4): 65, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38844719

RESUMEN

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.


Asunto(s)
Simulación por Computador , Voluntarios Sanos , Modelos Biológicos , Humanos , Estudios Retrospectivos , Farmacocinética , Hepatopatías/metabolismo , Enfermedades Renales , Preparaciones Farmacéuticas/metabolismo , Preparaciones Farmacéuticas/administración & dosificación , Insuficiencia Renal/metabolismo
3.
Bioinformatics ; 40(6)2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38837345

RESUMEN

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.


Asunto(s)
Biología Computacional , Biología Computacional/métodos , Descubrimiento de Drogas/métodos , Algoritmos , Reposicionamiento de Medicamentos/métodos , Preparaciones Farmacéuticas/metabolismo , Preparaciones Farmacéuticas/química , Humanos
4.
Clin Transl Sci ; 17(5): e13824, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38752574

RESUMEN

Accurate prediction of a new compound's pharmacokinetic (PK) profile is pivotal for the success of drug discovery programs. An initial assessment of PK in preclinical species and humans is typically performed through allometric scaling and mathematical modeling. These methods use parameters estimated from in vitro or in vivo experiments, which although helpful for an initial estimation, require extensive animal experiments. Furthermore, mathematical models are limited by the mechanistic underpinning of the drugs' absorption, distribution, metabolism, and elimination (ADME) which are largely unknown in the early stages of drug discovery. In this work, we propose a novel methodology in which concentration versus time profile of small molecules in rats is directly predicted by machine learning (ML) using structure-driven molecular properties as input and thus mitigating the need for animal experimentation. The proposed framework initially predicts ADME properties based on molecular structure and then uses them as input to a ML model to predict the PK profile. For the compounds tested, our results demonstrate that PK profiles can be adequately predicted using the proposed algorithm, especially for compounds with Tanimoto score greater than 0.5, the average mean absolute percentage error between predicted PK profile and observed PK profile data was found to be less than 150%. The suggested framework aims to facilitate PK predictions and thus support molecular screening and design earlier in the drug discovery process.


Asunto(s)
Descubrimiento de Drogas , Aprendizaje Automático , Animales , Ratas , Descubrimiento de Drogas/métodos , Preparaciones Farmacéuticas/metabolismo , Preparaciones Farmacéuticas/química , Humanos , Modelos Biológicos , Algoritmos , Estructura Molecular , Farmacocinética , Bibliotecas de Moléculas Pequeñas/farmacocinética
5.
Chemosphere ; 358: 142209, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38697564

RESUMEN

Elevated usage of pharmaceutical products leads to the accumulation of emerging contaminants in sewage. In the current work, Ganoderma lucidum (GL) was used to remove pharmaceutical compounds (PCs), proposed as a tertiary method in sewage treatment plants (STPs). The PCs consisted of a group of painkillers (ketoprofen, diclofenac, and dexamethasone), psychiatrists (carbamazepine, venlafaxine, and citalopram), beta-blockers (atenolol, metoprolol, and propranolol), and anti-hypertensives (losartan and valsartan). The performance of 800 mL of synthetic water, effluent STP, and hospital wastewater (HWW) was evaluated. Parameters, including treatment time, inoculum volume, and mechanical agitation speed, have been tested. The toxicity of the GL after treatment is being studied based on exposure levels to zebrafish embryos (ZFET) and the morphology of the GL has been observed via Field Emission Scanning Electron Microscopy (FESEM). The findings conclude that GL can reduce PCs from <10% to >90%. Diclofenac and valsartan are the highest (>90%) in the synthetic model, while citalopram and propranolol (>80%) are in the real wastewater. GL effectively removed pollutants in 48 h, 1% of the inoculum volume, and 50 rpm. The ZFET showed GL is non-toxic (LC50 is 209.95 mg/mL). In the morphology observation, pellets GL do not show major differences after treatment, showing potential to be used for a longer treatment time and to be re-useable in the system. GL offers advantages to removing PCs in water due to their non-specific extracellular enzymes that allow for the biodegradation of PCs and indicates a good potential in real-world applications as a favourable alternative treatment.


Asunto(s)
Reishi , Aguas Residuales , Contaminantes Químicos del Agua , Pez Cebra , Aguas Residuales/química , Contaminantes Químicos del Agua/toxicidad , Animales , Reishi/metabolismo , Eliminación de Residuos Líquidos/métodos , Preparaciones Farmacéuticas/análisis , Preparaciones Farmacéuticas/metabolismo , Malasia , Aguas del Alcantarillado/química , Aguas del Alcantarillado/microbiología , Biodegradación Ambiental , Diclofenaco/toxicidad
6.
Expert Opin Drug Deliv ; 21(4): 553-572, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38720439

RESUMEN

INTRODUCTION: Intranasal administration is an effective drug delivery routes in modern pharmaceutics. However, unlike other in vivo biological barriers, the nasal mucosal barrier is characterized by high turnover and selective permeability, hindering the diffusion of both particulate drug delivery systems and drug molecules. The in vivo fate of administrated nanomedicines is often significantly affected by nano-biointeractions. AREAS COVERED: The biological barriers that nanomedicines encounter when administered intranasally are introduced, with a discussion on the factors influencing the interaction between nanomedicines and the mucus layer/mucosal barriers. General design strategies for nanomedicines administered via the nasal route are further proposed. Furthermore, the most common methods to investigate the characteristics and the interactions of nanomedicines when in presence of the mucus layer/mucosal barrier are briefly summarized. EXPERT OPINION: Detailed investigation of nanomedicine-mucus/mucosal interactions and exploration of their mechanisms provide solutions for designing better intranasal nanomedicines. Designing and applying nanomedicines with mucus interaction properties or non-mucosal interactions should be customized according to the therapeutic need, considering the target of the drug, i.e. brain, lung or nose. Then how to improve the precise targeting efficiency of nanomedicines becomes a difficult task for further research.


Asunto(s)
Administración Intranasal , Sistemas de Liberación de Medicamentos , Moco , Nanomedicina , Mucosa Nasal , Mucosa Nasal/metabolismo , Humanos , Animales , Moco/metabolismo , Permeabilidad , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/metabolismo , Diseño de Fármacos , Nanopartículas
8.
Int J Mol Sci ; 25(9)2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38731891

RESUMEN

The past five decades have witnessed remarkable advancements in the field of inhaled medicines targeting the lungs for respiratory disease treatment. As a non-invasive drug delivery route, inhalation therapy offers numerous benefits to respiratory patients, including rapid and targeted exposure at specific sites, quick onset of action, bypassing first-pass metabolism, and beyond. Understanding the characteristics of pulmonary drug transporters and metabolizing enzymes is crucial for comprehending efficient drug exposure and clearance processes within the lungs. These processes are intricately linked to both local and systemic pharmacokinetics and pharmacodynamics of drugs. This review aims to provide a comprehensive overview of the literature on lung transporters and metabolizing enzymes while exploring their roles in exogenous and endogenous substance disposition. Additionally, we identify and discuss the principal challenges in this area of research, providing a foundation for future investigations aimed at optimizing inhaled drug administration. Moving forward, it is imperative that future research endeavors to focus on refining and validating in vitro and ex vivo models to more accurately mimic the human respiratory system. Such advancements will enhance our understanding of drug processing in different pathological states and facilitate the discovery of novel approaches for investigating lung-specific drug transporters and metabolizing enzymes. This deeper insight will be crucial in developing more effective and targeted therapies for respiratory diseases, ultimately leading to improved patient outcomes.


Asunto(s)
Pulmón , Proteínas de Transporte de Membrana , Humanos , Administración por Inhalación , Pulmón/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Animales , Preparaciones Farmacéuticas/metabolismo , Preparaciones Farmacéuticas/administración & dosificación , Transporte Biológico
9.
Expert Opin Drug Metab Toxicol ; 20(5): 319-332, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38785066

RESUMEN

INTRODUCTION: Medications are frequently prescribed for patients with irritable bowel syndrome (IBS) or disorders of gut brain interaction. The level of drug metabolism and modifications in drug targets determine medication efficacy to modify motor or sensory function as well as patient response outcomes. AREAS COVERED: The literature search included PubMed searches with the terms: pharmacokinetics, pharmacogenomics, epigenetics, clinical trials, irritable bowel syndrome, disorders of gut brain interaction, and genome-wide association studies. The main topics covered in relation to irritable bowel syndrome were precision medicine, pharmacogenomics related to drug metabolism, pharmacogenomics related to mechanistic targets, and epigenetics. EXPERT OPINION: Pharmacogenomics impacting drug metabolism [CYP 2D6 (cytochrome P450 2D6) or 2C19 (cytochrome P450 2C19)] is the most practical approach to precision medicine in the treatment of IBS. Although there are proof of concept studies that have documented the importance of genetic modification of transmitters or receptors in altering responses to medications in IBS, these principles have rarely been applied in patient response outcomes. Genome-wide association (GWAS) studies have now documented the association of symptoms with genetic variation but not the evaluation of treatment responses. Considerably more research, particularly focused on patient response outcomes and epigenetics, is essential to impact this field in clinical medicine.


Asunto(s)
Estudio de Asociación del Genoma Completo , Síndrome del Colon Irritable , Farmacogenética , Medicina de Precisión , Humanos , Síndrome del Colon Irritable/tratamiento farmacológico , Síndrome del Colon Irritable/genética , Medicina de Precisión/métodos , Citocromo P-450 CYP2C19/genética , Citocromo P-450 CYP2D6/genética , Citocromo P-450 CYP2D6/metabolismo , Fármacos Gastrointestinales/farmacología , Fármacos Gastrointestinales/farmacocinética , Fármacos Gastrointestinales/administración & dosificación , Preparaciones Farmacéuticas/metabolismo , Preparaciones Farmacéuticas/administración & dosificación , Epigénesis Genética , Animales
10.
Eur J Pharm Sci ; 198: 106799, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38754592

RESUMEN

The clearance concept has been used in pharmacokinetics for over 50 years. However, there is still much debate regarding mathematical clearance models. A recent article discussed that there is a critical error in a basic assumption that leads to the mechanistic hepatic clearance models (Benet, L.Z., Sodhi, J.K., 2024. Are all measures of liver Kpuu a function of FH, as determined following oral dosing, or have we made a critical error in defining hepatic drug clearance? European Journal of Pharmaceutical Sciences 196, 106,753. https://doi.org/10.1016/j.ejps.2024.106753). This commentary discusses this point based on the extended clearance model (ECM), which is increasingly used in modern drug discovery and development. Confusion about clearance can be avoided by using clearly defined drug concentrations based on hierarchical body structures.


Asunto(s)
Hígado , Modelos Biológicos , Humanos , Hígado/metabolismo , Administración Oral , Preparaciones Farmacéuticas/metabolismo , Preparaciones Farmacéuticas/administración & dosificación , Tasa de Depuración Metabólica , Farmacocinética , Animales
11.
Bioinformatics ; 40(5)2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38730554

RESUMEN

MOTIVATION: Enhanced by contemporary computational advances, the prediction of drug-target interactions (DTIs) has become crucial in developing de novo and effective drugs. Existing deep learning approaches to DTI prediction are frequently beleaguered by a tendency to overfit specific molecular representations, which significantly impedes their predictive reliability and utility in novel drug discovery contexts. Furthermore, existing DTI networks often disregard the molecular size variance between macro molecules (targets) and micro molecules (drugs) by treating them at an equivalent scale that undermines the accurate elucidation of their interaction. RESULTS: We propose a novel DTI network with a differential-scale scheme to model the binding site for enhancing DTI prediction, which is named as BindingSiteDTI. It explicitly extracts multiscale substructures from targets with different scales of molecular size and fixed-scale substructures from drugs, facilitating the identification of structurally similar substructural tokens, and models the concealed relationships at the substructural level to construct interaction feature. Experiments conducted on popular benchmarks, including DUD-E, human, and BindingDB, shown that BindingSiteDTI contains significant improvements compared with recent DTI prediction methods. AVAILABILITY AND IMPLEMENTATION: The source code of BindingSiteDTI can be accessed at https://github.com/MagicPF/BindingSiteDTI.


Asunto(s)
Descubrimiento de Drogas , Sitios de Unión , Humanos , Descubrimiento de Drogas/métodos , Preparaciones Farmacéuticas/química , Preparaciones Farmacéuticas/metabolismo , Biología Computacional/métodos , Aprendizaje Profundo
12.
Clin Pharmacokinet ; 63(5): 561-588, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38748090

RESUMEN

Human milk is a remarkable biofluid that provides essential nutrients and immune protection to newborns. Breastfeeding women consuming medications could pass the drug through their milk to neonates. Drugs can be transferred to human milk by passive diffusion or active transport. The physicochemical properties of the drug largely impact the extent of drug transfer into human milk. A comprehensive understanding of the physiology of human milk formation, composition of milk, mechanisms of drug transfer, and factors influencing drug transfer into human milk is critical for appropriate selection and use of medications in lactating women. Quantification of drugs in the milk is essential for assessing the safety of pharmacotherapy during lactation. This can be achieved by developing specific, sensitive, and reproducible analytical methods using techniques such as liquid chromatography coupled with mass spectrometry. The present review briefly discusses the physiology of human milk formation, composition of human milk, mechanisms of drug transfer into human milk, and factors influencing transfer of drugs from blood to milk. We further expand upon and critically evaluate the existing analytical approaches/assays used for the quantification of drugs in human milk.


Asunto(s)
Leche Humana , Humanos , Leche Humana/química , Leche Humana/metabolismo , Preparaciones Farmacéuticas/metabolismo , Femenino , Lactancia/metabolismo , Lactancia Materna , Recién Nacido , Cromatografía Liquida/métodos , Espectrometría de Masas/métodos
13.
J Chem Inf Model ; 64(10): 4348-4358, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38709146

RESUMEN

Developing new pharmaceuticals is a costly and time-consuming endeavor fraught with significant safety risks. A critical aspect of drug research and disease therapy is discerning the existence of interactions between drugs and proteins. The evolution of deep learning (DL) in computer science has been remarkably aided in this regard in recent years. Yet, two challenges remain: (i) balancing the extraction of profound, local cohesive characteristics while warding off gradient disappearance and (ii) globally representing and understanding the interactions between the drug and target local attributes, which is vital for delivering molecular level insights indispensable to drug development. In response to these challenges, we propose a DL network structure, MolLoG, primarily comprising two modules: local feature encoders (LFE) and global interactive learning (GIL). Within the LFE module, graph convolution networks and leap blocks capture the local features of drug and protein molecules, respectively. The GIL module enables the efficient amalgamation of feature information, facilitating the global learning of feature structural semantics and procuring multihead attention weights for abstract features stemming from two modalities, providing biologically pertinent explanations for black-box results. Finally, predictive outcomes are achieved by decoding the unified representation via a multilayer perceptron. Our experimental analysis reveals that MolLoG outperforms several cutting-edge baselines across four data sets, delivering superior overall performance and providing satisfactory results when elucidating various facets of drug-target interaction predictions.


Asunto(s)
Aprendizaje Profundo , Proteínas , Proteínas/metabolismo , Proteínas/química , Preparaciones Farmacéuticas/química , Preparaciones Farmacéuticas/metabolismo , Descubrimiento de Drogas/métodos , Modelos Moleculares
14.
J Chem Inf Model ; 64(10): 4373-4384, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38743013

RESUMEN

Artificial intelligence-based methods for predicting drug-target interactions (DTIs) aim to explore reliable drug candidate targets rapidly and cost-effectively to accelerate the drug development process. However, current methods are often limited by the topological regularities of drug molecules, making them difficult to generalize to a broader chemical space. Additionally, the use of similarity to measure DTI network links often introduces noise, leading to false DTI relationships and affecting the prediction accuracy. To address these issues, this study proposes an Adaptive Iterative Graph Optimization (AIGO)-DTI prediction framework. This framework integrates atomic cluster information and enhances molecular features through the design of functional group prompts and graph encoders, optimizing the construction of DTI association networks. Furthermore, the optimization of graph structure is transformed into a node similarity learning problem, utilizing multihead similarity metric functions to iteratively update the network structure to improve the quality of DTI information. Experimental results demonstrate the outstanding performance of AIGO-DTI on multiple public data sets and label reversal data sets. Case studies, molecular docking, and existing research validate its effectiveness and reliability. Overall, the method proposed in this study can construct comprehensive and reliable DTI association network information, providing new graphing and optimization strategies for DTI prediction, which contribute to efficient drug development and reduce target discovery costs.


Asunto(s)
Algoritmos , Simulación del Acoplamiento Molecular , Inteligencia Artificial , Preparaciones Farmacéuticas/química , Preparaciones Farmacéuticas/metabolismo , Descubrimiento de Drogas/métodos
15.
Clin Transl Sci ; 17(5): e13810, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38716900

RESUMEN

One of the key pharmacokinetic properties of most small molecule drugs is their ability to bind to serum proteins. Unbound or free drug is responsible for pharmacological activity while the balance between free and bound drug can impact drug distribution, elimination, and other safety parameters. In the hepatic impairment (HI) and renal impairment (RI) clinical studies, unbound drug concentration is often assessed; however, the relevance and impact of the protein binding (PB) results is largely limited. We analyzed published clinical safety and pharmacokinetic studies in subjects with HI or RI with PB assessment up to October 2022 and summarized the contribution of PB results on their label dose recommendations. Among drugs with HI publication, 32% (17/53) associated product labels include PB results in HI section. Of these, the majority (9/17, 53%) recommend dose adjustments consistent with observed PB change. Among drugs with RI publication, 27% (12/44) of associated product labels include PB results in RI section with the majority (7/12, 58%) recommending no dose adjustment, consistent with the reported absence of PB change. PB results were found to be consistent with a tailored dose recommendation in 53% and 58% of the approved labels for HI and RI section, respectively. We further discussed the interpretation challenges of PB results, explored treatment decision factors including total drug concentration, exposure-response relationships, and safety considerations in these case examples. Collectively, comprehending the alterations in free drug levels in HI and RI informs treatment decision through a risk-based approach.


Asunto(s)
Etiquetado de Medicamentos , Unión Proteica , Humanos , Insuficiencia Renal/metabolismo , Relación Dosis-Respuesta a Droga , Preparaciones Farmacéuticas/metabolismo , Preparaciones Farmacéuticas/administración & dosificación , Hepatopatías/metabolismo , Hepatopatías/tratamiento farmacológico , Proteínas Sanguíneas/metabolismo , Cálculo de Dosificación de Drogas
16.
Expert Opin Drug Discov ; 19(6): 671-682, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38722032

RESUMEN

INTRODUCTION: For rational drug design, it is crucial to understand the receptor-drug binding processes and mechanisms. A new era for the use of computer simulations in predicting drug-receptor interactions at an atomic level has begun with remarkable advances in supercomputing and methodological breakthroughs. AREAS COVERED: End-point free energy calculation methods such as Molecular Mechanics/Poisson Boltzmann Surface Area (MM/PBSA) or Molecular-Mechanics/Generalized Born Surface Area (MM/GBSA), free energy perturbation (FEP), and thermodynamic integration (TI) are commonly used for binding free energy calculations in drug discovery. In addition, kinetic dissociation and association rate constants (koff and kon) play critical roles in the function of drugs. Nowadays, Molecular Dynamics (MD) and enhanced sampling simulations are increasingly being used in drug discovery. Here, the authors provide a review of the computational techniques used in drug binding free energy and kinetics calculations. EXPERT OPINION: The applications of computational methods in drug discovery and design are expanding, thanks to improved predictions of the binding free energy and kinetic rates of drug molecules. Recent microsecond-timescale enhanced sampling simulations have made it possible to accurately capture repetitive ligand binding and dissociation, facilitating more efficient and accurate calculations of ligand binding free energy and kinetics.


Asunto(s)
Diseño de Fármacos , Descubrimiento de Drogas , Simulación de Dinámica Molecular , Termodinámica , Humanos , Simulación por Computador , Descubrimiento de Drogas/métodos , Cinética , Preparaciones Farmacéuticas/química , Preparaciones Farmacéuticas/metabolismo , Unión Proteica
17.
Expert Opin Drug Discov ; 19(6): 683-698, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38727016

RESUMEN

INTRODUCTION: Prediction of pharmacokinetic (PK) properties is crucial for drug discovery and development. Machine-learning (ML) models, which use statistical pattern recognition to learn correlations between input features (such as chemical structures) and target variables (such as PK parameters), are being increasingly used for this purpose. To embed ML models for PK prediction into workflows and to guide future development, a solid understanding of their applicability, advantages, limitations, and synergies with other approaches is necessary. AREAS COVERED: This narrative review discusses the design and application of ML models to predict PK parameters of small molecules, especially in light of established approaches including in vitro-in vivo extrapolation (IVIVE) and physiologically based pharmacokinetic (PBPK) models. The authors illustrate scenarios in which the three approaches are used and emphasize how they enhance and complement each other. In particular, they highlight achievements, the state of the art and potentials of applying machine learning for PK prediction through a comphrehensive literature review. EXPERT OPINION: ML models, when carefully crafted, regularly updated, and appropriately used, empower users to prioritize molecules with favorable PK properties. Informed practitioners can leverage these models to improve the efficiency of drug discovery and development process.


Asunto(s)
Desarrollo de Medicamentos , Descubrimiento de Drogas , Aprendizaje Automático , Modelos Biológicos , Farmacocinética , Humanos , Descubrimiento de Drogas/métodos , Desarrollo de Medicamentos/métodos , Animales , Preparaciones Farmacéuticas/metabolismo , Preparaciones Farmacéuticas/química , Preparaciones Farmacéuticas/administración & dosificación
18.
Expert Opin Drug Metab Toxicol ; 20(5): 377-397, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38706437

RESUMEN

INTRODUCTION: Carboxylesterase 1 (CES1) and carboxylesterase 2 (CES2) are among the most abundant hydrolases in humans, catalyzing the metabolism of numerous clinically important medications, such as methylphenidate and clopidogrel. The large interindividual variability in the expression and activity of CES1 and CES2 affects the pharmacokinetics (PK) and pharmacodynamics (PD) of substrate drugs. AREAS COVERED: This review provides an up-to-date overview of CES expression and activity regulations and examines their impact on the PK and PD of CES substrate drugs. The literature search was conducted on PubMed from inception to January 2024. EXPERT OPINION: Current research revealed modest associations of CES genetic polymorphisms with drug exposure and response. Beyond genomic polymorphisms, transcriptional and posttranslational regulations can also significantly affect CES expression and activity and consequently alter PK and PD. Recent advances in plasma biomarkers of drug-metabolizing enzymes encourage the research of plasma protein and metabolite biomarkers for CES1 and CES2, which could lead to the establishment of precision pharmacotherapy regimens for drugs metabolized by CESs. Moreover, our understanding of tissue-specific expression and substrate selectivity of CES1 and CES2 has shed light on improving the design of CES1- and CES2-activated prodrugs.


Asunto(s)
Hidrolasas de Éster Carboxílico , Humanos , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , Animales , Polimorfismo Genético , Preparaciones Farmacéuticas/metabolismo , Profármacos/farmacocinética , Biomarcadores/metabolismo , Carboxilesterasa
19.
AAPS J ; 26(3): 59, 2024 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724865

RESUMEN

Drug clearance in obese subjects varies widely among different drugs and across subjects with different severity of obesity. This study investigates correlations between plasma clearance (CLp) and drug- and patient-related characteristics in obese subjects, and evaluates the systematic accuracy of common weight-based dosing methods. A physiologically-based pharmacokinetic (PBPK) modeling approach that uses recent information on obesity-related changes in physiology was used to simulate CLp for a normal-weight subject (body mass index [BMI] = 20) and subjects with various severities of obesity (BMI 25-60) for hypothetical hepatically cleared drugs with a wide range of properties. Influential variables for CLp change were investigated. For each drug and obese subject, the exponent that yields perfect allometric scaling of CLp from normal-weight subjects was assessed. Among all variables, BMI and relative changes in enzyme activity resulting from obesity proved highly correlated with obesity-related CLp changes. Drugs bound to α1-acid glycoprotein (AAG) had lower CLp changes compared to drugs bound to human serum albumin (HSA). Lower extraction ratios (ER) corresponded to higher CLp changes compared to higher ER. The allometric exponent for perfect scaling ranged from -3.84 to 3.34 illustrating that none of the scaling methods performed well in all situations. While all three dosing methods are generally systematically accurate for drugs with unchanged or up to 50% increased enzyme activity in subjects with a BMI below 30 kg/m2, in any of the other cases, information on the different drug properties and severity of obesity is required to select an appropriate dosing method for individuals with obesity.


Asunto(s)
Índice de Masa Corporal , Modelos Biológicos , Obesidad , Humanos , Obesidad/metabolismo , Tasa de Depuración Metabólica/fisiología , Preparaciones Farmacéuticas/metabolismo , Preparaciones Farmacéuticas/administración & dosificación , Hígado/metabolismo , Orosomucoide/metabolismo , Albúmina Sérica Humana/metabolismo , Albúmina Sérica Humana/análisis , Masculino , Adulto
20.
J Bioinform Comput Biol ; 22(2): 2450004, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38812467

RESUMEN

Biomolecular interaction recognition between ligands and proteins is an essential task, which largely enhances the safety and efficacy in drug discovery and development stage. Studying the interaction between proteins and ligands can improve the understanding of disease pathogenesis and lead to more effective drug targets. Additionally, it can aid in determining drug parameters, ensuring proper absorption, distribution, and metabolism within the body. Due to incomplete feature representation or the model's inadequate adaptation to protein-ligand complexes, the existing methodologies suffer from suboptimal predictive accuracy. To address these pitfalls, in this study, we designed a new deep learning method based on transformer and GCN. We first utilized the transformer network to grasp crucial information of the original protein sequences within the smile sequences and connected them to prevent falling into a local optimum. Furthermore, a series of dilation convolutions are performed to obtain the pocket features and smile features, subsequently subjected to graphical convolution to optimize the connections. The combined representations are fed into the proposed model for classification prediction. Experiments conducted on various protein-ligand binding prediction methods prove the effectiveness of our proposed method. It is expected that the PfgPDI can contribute to drug prediction and accelerate the development of new drugs, while also serving as a valuable partner for drug testing and Research and Development engineers.


Asunto(s)
Biología Computacional , Descubrimiento de Drogas , Redes Neurales de la Computación , Proteínas , Proteínas/química , Proteínas/metabolismo , Ligandos , Biología Computacional/métodos , Descubrimiento de Drogas/métodos , Aprendizaje Profundo , Unión Proteica , Preparaciones Farmacéuticas/química , Preparaciones Farmacéuticas/metabolismo , Bases de Datos de Proteínas , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA