5 research outputs found

    Deep learning for in vitro prediction of pharmaceutical formulations

    Full text link
    Current pharmaceutical formulation development still strongly relies on the traditional trial-and-error approach by individual experiences of pharmaceutical scientists, which is laborious, time-consuming and costly. Recently, deep learning has been widely applied in many challenging domains because of its important capability of automatic feature extraction. The aim of this research is to use deep learning to predict pharmaceutical formulations. In this paper, two different types of dosage forms were chosen as model systems. Evaluation criteria suitable for pharmaceutics were applied to assessing the performance of the models. Moreover, an automatic dataset selection algorithm was developed for selecting the representative data as validation and test datasets. Six machine learning methods were compared with deep learning. The result shows the accuracies of both two deep neural networks were above 80% and higher than other machine learning models, which showed good prediction in pharmaceutical formulations. In summary, deep learning with the automatic data splitting algorithm and the evaluation criteria suitable for pharmaceutical formulation data was firstly developed for the prediction of pharmaceutical formulations. The cross-disciplinary integration of pharmaceutics and artificial intelligence may shift the paradigm of pharmaceutical researches from experience-dependent studies to data-driven methodologies

    Organic crystal structure prediction via coupled generative adversarial networks and graph convolutional networks

    No full text
    Organic crystal structures exert a profound impact on the physicochemical properties and biological effects of organic compounds. Quantum mechanics (QM)–based crystal structure predictions (CSPs) have somewhat alleviated the dilemma that experimental crystal structure investigations struggle to conduct complete polymorphism studies, but the high computing cost poses a challenge to its widespread application. The present study aims to construct DeepCSP, a feasible pure machine learning framework for minute-scale rapid organic CSP. Initially, based on 177,746 data entries from the Cambridge Crystal Structure Database, a generative adversarial network was built to conditionally generate trial crystal structures under selected feature constraints for the given molecule. Simultaneously, a graph convolutional attention network was used to predict the density of stable crystal structures for the input molecule. Subsequently, the distances between the predicted density and the definition-based calculated density would be considered to be the crystal structure screening and ranking basis, and finally, the density-based crystal structure ranking would be output. Two such distinct algorithms, performing the generation and ranking functionalities, respectively, collectively constitute the DeepCSP, which has demonstrated compelling performance in marketed drug validations, achieving an accuracy rate exceeding 80% and a hit rate surpassing 85%. Inspiringly, the computing speed of the pure machine learning methodology demonstrates the potential of artificial intelligence in advancing CSP research

    Predicting liposome formulations by the integrated machine learning and molecular modeling approaches

    No full text
    Liposome is one of the most widely used carriers for drug delivery because of the great biocompatibility and biodegradability. Due to the complex formulation components and preparation process, formulation screening mostly relies on trial-and-error process with low efficiency. Here liposome formulation prediction models have been built by machine learning (ML) approaches. The important parameters of liposomes, including size, polydispersity index (PDI), zeta potential and encapsulation, are predicted individually by optimal ML algorithm, while the formulation features are also ranked to provide important guidance for formulation design. The analysis of key parameter reveals that drug molecules with logS [-3, -6], molecular complexity [500, 1000] and XLogP3 (≥2) are priority for preparing liposome with higher encapsulation. In addition, naproxen (NAP) and palmatine HCl (PAL) represented the insoluble and water-soluble molecules are prepared as liposome formulations to validate prediction ability. The consistency between predicted and experimental value verifies the satisfied accuracy of ML models. As the drug properties are critical for liposome particles, the molecular interactions and dynamics of NAP and PAL liposome are further investigated by coarse-grained molecular dynamics simulations. The modeling structure reveals that NAP molecules could distribute into lipid layer, while most PAL molecules aggregate in the inner aqueous phase of liposome. The completely different physical state of NAP and PAL confirms the importance of drug properties for liposome formulations. In summary, the general prediction models are built to predict liposome formulations, and the impacts of key factors are analyzed by combing ML with molecular modeling. The availability and rationality of these intelligent prediction systems have been proved in this study, which could be applied for liposome formulation development in the future
    corecore