Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Brief Bioinform ; 25(4)2024 May 23.
Article in English | MEDLINE | ID: mdl-38960407

ABSTRACT

The optimization of therapeutic antibodies through traditional techniques, such as candidate screening via hybridoma or phage display, is resource-intensive and time-consuming. In recent years, computational and artificial intelligence-based methods have been actively developed to accelerate and improve the development of therapeutic antibodies. In this study, we developed an end-to-end sequence-based deep learning model, termed AttABseq, for the predictions of the antigen-antibody binding affinity changes connected with antibody mutations. AttABseq is a highly efficient and generic attention-based model by utilizing diverse antigen-antibody complex sequences as the input to predict the binding affinity changes of residue mutations. The assessment on the three benchmark datasets illustrates that AttABseq is 120% more accurate than other sequence-based models in terms of the Pearson correlation coefficient between the predicted and experimental binding affinity changes. Moreover, AttABseq also either outperforms or competes favorably with the structure-based approaches. Furthermore, AttABseq consistently demonstrates robust predictive capabilities across a diverse array of conditions, underscoring its remarkable capacity for generalization across a wide spectrum of antigen-antibody complexes. It imposes no constraints on the quantity of altered residues, rendering it particularly applicable in scenarios where crystallographic structures remain unavailable. The attention-based interpretability analysis indicates that the causal effects of point mutations on antibody-antigen binding affinity changes can be visualized at the residue level, which might assist automated antibody sequence optimization. We believe that AttABseq provides a fiercely competitive answer to therapeutic antibody optimization.


Subject(s)
Antigen-Antibody Complex , Deep Learning , Antigen-Antibody Complex/chemistry , Antigens/chemistry , Antigens/genetics , Antigens/metabolism , Antigens/immunology , Antibody Affinity , Amino Acid Sequence , Computational Biology/methods , Humans , Mutation , Antibodies/chemistry , Antibodies/immunology , Antibodies/genetics , Antibodies/metabolism
4.
J Chem Inf Model ; 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38920405

ABSTRACT

Artificial intelligence (AI)-aided drug design has demonstrated unprecedented effects on modern drug discovery, but there is still an urgent need for user-friendly interfaces that bridge the gap between these sophisticated tools and scientists, particularly those who are less computer savvy. Herein, we present DrugFlow, an AI-driven one-stop platform that offers a clean, convenient, and cloud-based interface to streamline early drug discovery workflows. By seamlessly integrating a range of innovative AI algorithms, covering molecular docking, quantitative structure-activity relationship modeling, molecular generation, ADMET (absorption, distribution, metabolism, excretion and toxicity) prediction, and virtual screening, DrugFlow can offer effective AI solutions for almost all crucial stages in early drug discovery, including hit identification and hit/lead optimization. We hope that the platform can provide sufficiently valuable guidance to aid real-word drug design and discovery. The platform is available at https://drugflow.com.

5.
Taiwan J Obstet Gynecol ; 63(3): 405-408, 2024 May.
Article in English | MEDLINE | ID: mdl-38802208

ABSTRACT

OBJECTIVE: Impetigo herpetiformis (IH) is a rare form of pustular psoriasis which may result in maternal and fetal morbidity and even mortality. Deficiency of interleukin-36 receptor antagonist (DITRA) is the most frequently identified genetic defect of IH. Currently there are no biologics approved for IH despite the revolutionary role of biologics in the treatment of plaque and pustular psoriasis. Anecdotal reports of biologics use in DITRA patients with IH are also limited. CASE REPORTS: We present herein a case series of 6 Chinese IH patients harboring IL36RN gene c.115+6T>C mutation during 8 pregnancies, treated with various biologics, including adalimumab, etanercept and secukinumab. CONCLUSION: Most pregnancy courses were uneventful, except for one woman who had recurrent episodes of decreased fetal heart rate variability after adalimumab injections, which subsided after switching to etanercept. The treatment effectiveness and safety demonstrated in our cases suggested the role of biologics for the treatment of IH in patients with DITRA.


Subject(s)
Adalimumab , Antibodies, Monoclonal, Humanized , Etanercept , Pregnancy Complications , Psoriasis , Humans , Female , Pregnancy , Adult , Antibodies, Monoclonal, Humanized/therapeutic use , Etanercept/therapeutic use , Adalimumab/therapeutic use , Pregnancy Complications/drug therapy , Psoriasis/drug therapy , Psoriasis/genetics , Antibodies, Monoclonal/therapeutic use , Interleukins/genetics , Biological Products/therapeutic use , China , Mutation , East Asian People
6.
Acc Chem Res ; 57(10): 1500-1509, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38577892

ABSTRACT

Molecular docking, also termed ligand docking (LD), is a pivotal element of structure-based virtual screening (SBVS) used to predict the binding conformations and affinities of protein-ligand complexes. Traditional LD methodologies rely on a search and scoring framework, utilizing heuristic algorithms to explore binding conformations and scoring functions to evaluate binding strengths. However, to meet the efficiency demands of SBVS, these algorithms and functions are often simplified, prioritizing speed over accuracy.The emergence of deep learning (DL) has exerted a profound impact on diverse fields, ranging from natural language processing to computer vision and drug discovery. DeepMind's AlphaFold2 has impressively exhibited its ability to accurately predict protein structures solely from amino acid sequences, highlighting the remarkable potential of DL in conformation prediction. This groundbreaking advancement circumvents the traditional search-scoring frameworks in LD, enhancing both accuracy and processing speed and thereby catalyzing a broader adoption of DL algorithms in binding pose prediction. Nevertheless, a consensus on certain aspects remains elusive.In this Account, we delineate the current status of employing DL to augment LD within the VS paradigm, highlighting our contributions to this domain. Furthermore, we discuss the challenges and future prospects, drawing insights from our scholarly investigations. Initially, we present an overview of VS and LD, followed by an introduction to DL paradigms, which deviate significantly from traditional search-scoring frameworks. Subsequently, we delve into the challenges associated with the development of DL-based LD (DLLD), encompassing evaluation metrics, application scenarios, and physical plausibility of the predicted conformations. In the evaluation of LD algorithms, it is essential to recognize the multifaceted nature of the metrics. While the accuracy of binding pose prediction, often measured by the success rate, is a pivotal aspect, the scoring/screening power and computational speed of these algorithms are equally important given the pivotal role of LD tools in VS. Regarding application scenarios, early methods focused on blind docking, where the binding site is unknown. However, recent studies suggest a shift toward identifying binding sites rather than solely predicting binding poses within these models. In contrast, LD with a known pocket in VS has been shown to be more practical. Physical plausibility poses another significant challenge. Although DLLD models often achieve higher success rates compared to traditional methods, they may generate poses with implausible local structures, such as incorrect bond angles or lengths, which are disadvantageous for postprocessing tasks like visualization. Finally, we discuss the future perspectives for DLLD, emphasizing the need to improve generalization ability, strike a balance between speed and accuracy, account for protein conformation flexibility, and enhance physical plausibility. Additionally, we delve into the comparison between generative and regression algorithms in this context, exploring their respective strengths and potential.


Subject(s)
Deep Learning , Molecular Docking Simulation , Ligands , Proteins/chemistry , Proteins/metabolism , Algorithms , Drug Discovery
7.
J Cheminform ; 16(1): 38, 2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38556873

ABSTRACT

Accurate prediction of the enzyme comission (EC) numbers for chemical reactions is essential for the understanding and manipulation of enzyme functions, biocatalytic processes and biosynthetic planning. A number of machine leanring (ML)-based models have been developed to classify enzymatic reactions, showing great advantages over costly and long-winded experimental verifications. However, the prediction accuracy for most available models trained on the records of chemical reactions without specifying the enzymatic catalysts is rather limited. In this study, we introduced BEC-Pred, a BERT-based multiclassification model, for predicting EC numbers associated with reactions. Leveraging transfer learning, our approach achieves precise forecasting across a wide variety of Enzyme Commission (EC) numbers solely through analysis of the SMILES sequences of substrates and products. BEC-Pred model outperformed other sequence and graph-based ML methods, attaining a higher accuracy of 91.6%, surpassing them by 5.5%, and exhibiting superior F1 scores with improvements of 6.6% and 6.0%, respectively. The enhanced performance highlights the potential of BEC-Pred to serve as a reliable foundational tool to accelerate the cutting-edge research in synthetic biology and drug metabolism. Moreover, we discussed a few examples on how BEC-Pred could accurately predict the enzymatic classification for the Novozym 435-induced hydrolysis and lipase efficient catalytic synthesis. We anticipate that BEC-Pred will have a positive impact on the progression of enzymatic research.

8.
Phys Chem Chem Phys ; 26(13): 10323-10335, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38501198

ABSTRACT

Ribonucleic acid (RNA)-ligand interactions play a pivotal role in a wide spectrum of biological processes, ranging from protein biosynthesis to cellular reproduction. This recognition has prompted the broader acceptance of RNA as a viable candidate for drug targets. Delving into the atomic-scale understanding of RNA-ligand interactions holds paramount importance in unraveling intricate molecular mechanisms and further contributing to RNA-based drug discovery. Computational approaches, particularly molecular docking, offer an efficient way of predicting the interactions between RNA and small molecules. However, the accuracy and reliability of these predictions heavily depend on the performance of scoring functions (SFs). In contrast to the majority of SFs used in RNA-ligand docking, the end-point binding free energy calculation methods, such as molecular mechanics/generalized Born surface area (MM/GBSA) and molecular mechanics/Poisson Boltzmann surface area (MM/PBSA), stand as theoretically more rigorous approaches. Yet, the evaluation of their effectiveness in predicting both binding affinities and binding poses within RNA-ligand systems remains unexplored. This study first reported the performance of MM/PBSA and MM/GBSA with diverse solvation models, interior dielectric constants (εin) and force fields in the context of binding affinity prediction for 29 RNA-ligand complexes. MM/GBSA is based on short (5 ns) molecular dynamics (MD) simulations in an explicit solvent with the YIL force field; the GBGBn2 model with higher interior dielectric constant (εin = 12, 16 or 20) yields the best correlation (Rp = -0.513), which outperforms the best correlation (Rp = -0.317, rDock) offered by various docking programs. Then, the efficacy of MM/GBSA in identifying the near-native binding poses from the decoys was assessed based on 56 RNA-ligand complexes. However, it is evident that MM/GBSA has limitations in accurately predicting binding poses for RNA-ligand systems, particularly compared with notably proficient docking programs like rDock and PLANTS. The best top-1 success rate achieved by MM/GBSA rescoring is 39.3%, which falls below the best results given by docking programs (50%, PLNATS). This study represents the first evaluation of MM/PBSA and MM/GBSA for RNA-ligand systems and is expected to provide valuable insights into their successful application to RNA targets.


Subject(s)
Molecular Dynamics Simulation , RNA , Molecular Docking Simulation , Ligands , Reproducibility of Results , Protein Binding , Thermodynamics , Binding Sites
9.
Ital J Dermatol Venerol ; 159(2): 207-208, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38436614
10.
Exp Dermatol ; 33(3): e15056, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38488485

ABSTRACT

Several studies have suggested that mutation of the interleukin 36 receptor antagonist gene (IL36RN) is related to generalized pustular psoriasis (GPP), and the presence of IL36RN mutation may affect the clinical manifestations and treatment responses. However, genetic testing is not routinely available in clinical practice for the diagnosis of GPP. Previously, GPP patients with acrodermatitis continua of Hallopeau (ACH) were found to have a high percentage of carrying IL36RN mutation. In this study, we reported six patients with pustular psoriasis presenting as diffuse palmoplantar erythema with keratoderma among 60 patients who carried IL36RN mutation. ACH was present in five patients and five patients had acute flare of GPP. This unique presentation may serve as a predictor for IL36RN mutation in patients with pustular psoriasis, similar to ACH.


Subject(s)
Psoriasis , Humans , Psoriasis/genetics , Mutation , Erythema , China , Interleukins/genetics
11.
J Chem Inf Model ; 64(4): 1213-1228, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38302422

ABSTRACT

Deep learning-based de novo molecular design has recently gained significant attention. While numerous DL-based generative models have been successfully developed for designing novel compounds, the majority of the generated molecules lack sufficiently novel scaffolds or high drug-like profiles. The aforementioned issues may not be fully captured by commonly used metrics for the assessment of molecular generative models, such as novelty, diversity, and quantitative estimation of the drug-likeness score. To address these limitations, we proposed a genetic algorithm-guided generative model called GARel (genetic algorithm-based receptor-ligand interaction generator), a novel framework for training a DL-based generative model to produce drug-like molecules with novel scaffolds. To efficiently train the GARel model, we utilized dense net to update the parameters based on molecules with novel scaffolds and drug-like features. To demonstrate the capability of the GARel model, we used it to design inhibitors for three targets: AA2AR, EGFR, and SARS-Cov2. The results indicate that GARel-generated molecules feature more diverse and novel scaffolds and possess more desirable physicochemical properties and favorable docking scores. Compared with other generative models, GARel makes significant progress in balancing novelty and drug-likeness, providing a promising direction for the further development of DL-based de novo design methodology with potential impacts on drug discovery.


Subject(s)
Drug Design , RNA, Viral , Ligands , Algorithms , Drug Discovery
12.
Brief Bioinform ; 25(2)2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38340091

ABSTRACT

Discovering effective anti-tumor drug combinations is crucial for advancing cancer therapy. Taking full account of intricate biological interactions is highly important in accurately predicting drug synergy. However, the extremely limited prior knowledge poses great challenges in developing current computational methods. To address this, we introduce SynergyX, a multi-modality mutual attention network to improve anti-tumor drug synergy prediction. It dynamically captures cross-modal interactions, allowing for the modeling of complex biological networks and drug interactions. A convolution-augmented attention structure is adopted to integrate multi-omic data in this framework effectively. Compared with other state-of-the-art models, SynergyX demonstrates superior predictive accuracy in both the General Test and Blind Test and cross-dataset validation. By exhaustively screening combinations of approved drugs, SynergyX reveals its ability to identify promising drug combination candidates for potential lung cancer treatment. Another notable advantage lies in its multidimensional interpretability. Taking Sorafenib and Vorinostat as an example, SynergyX serves as a powerful tool for uncovering drug-gene interactions and deciphering cell selectivity mechanisms. In summary, SynergyX provides an illuminating and interpretable framework, poised to catalyze the expedition of drug synergy discovery and deepen our comprehension of rational combination therapy.


Subject(s)
Drug Discovery , Lung Neoplasms , Humans , Catalysis , Combined Modality Therapy , Research Design
13.
J Chem Theory Comput ; 20(3): 1465-1478, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38300792

ABSTRACT

Multisite λ-dynamics (MSLD) is a highly efficient binding free energy calculation method that samples multiple ligands in a single round by assigning different λ values to the alchemical part of each ligand. This method holds great promise for lead optimization (LO) in drug discovery. However, the complex data preparation and simulation process limits its widespread application in diverse protein-ligand systems. To address this challenge, we developed a comprehensive, open-source, and automated workflow for MSLD calculations based on the BLaDE dynamics engine. This workflow incorporates the Ligand Internal and Cartesian coordinate reconstruction-based alignment algorithm (LIC-align) and an optimized maximum common substructure (MCS) search algorithm to accurately generate MSLD multiple topologies with ideal perturbation patterns. Furthermore, our workflow is highly modularized, allowing straightforward integration and extension of various simulation techniques, and is highly accessible to nonexperts. This workflow was validated by calculating the relative binding free energies of large-scale congeneric ligands, many of which have large perturbing groups. The agreement between the calculations and experiments was excellent, with an average unsigned error of 1.08 ± 0.47 kcal/mol. More than 57.1% of the ligands had an error of less than 1.0 kcal/mol, and the perturbations of 6 targets were fully connected via the calculations, while those of 2 targets were connected via both calculations and experimental data. The Pearson correlation coefficient reached 0.88, indicating that the MSLD workflow provides accurate predictions that can guide lead optimization in drug discovery. We also examined the impact of single-site versus multisite perturbations, ligand grouping by perturbing group size, and the position of the anchor atom on the MSLD performance. By integrating our proposed LIC-align and optimized MCS search algorithm along with the coping strategies to handle challenging molecular substructures, our workflow can handle many realistic scenarios more reasonably than all previously published methods. Moreover, we observed that our MSLD workflow achieved similar accuracy to free energy perturbation (FEP) while improving computational efficiency by over 1 order of magnitude in speedup. These findings provide valuable insights and strategies for further MSLD development, making MSLD a competitive tool for lead optimization.


Subject(s)
Molecular Dynamics Simulation , Proteins , Thermodynamics , Ligands , Workflow , Proteins/chemistry , Protein Binding
14.
Nat Protoc ; 19(4): 1105-1121, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38263521

ABSTRACT

Lead optimization is a crucial step in the drug discovery process, which aims to design potential drug candidates from biologically active hits. During lead optimization, active hits undergo modifications to improve their absorption, distribution, metabolism, excretion and toxicity (ADMET) profiles. Medicinal chemists face key questions regarding which compound(s) should be synthesized next and how to balance multiple ADMET properties. Reliable transformation rules from multiple experimental analyses are critical to improve this decision-making process. We developed OptADMET ( https://cadd.nscc-tj.cn/deploy/optadmet/ ), an integrated web-based platform that provides chemical transformation rules for 32 ADMET properties and leverages prior experimental data for lead optimization. The multiproperty transformation rule database contains a total of 41,779 validated transformation rules generated from the analysis of 177,191 reliable experimental datasets. Additionally, 146,450 rules were generated by analyzing 239,194 molecular data predictions. OptADMET provides the ADMET profiles of all optimized molecules from the queried molecule and enables the prediction of desirable substructure transformations and subsequent validation of drug candidates. OptADMET is based on matched molecular pairs analysis derived from synthetic chemistry, thus providing improved practicality over other methods. OptADMET is designed for use by both experimental and computational scientists.


Subject(s)
Drug Discovery , Internet , Databases, Factual
15.
Chem Sci ; 15(4): 1449-1471, 2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38274053

ABSTRACT

The expertise accumulated in deep neural network-based structure prediction has been widely transferred to the field of protein-ligand binding pose prediction, thus leading to the emergence of a variety of deep learning-guided docking models for predicting protein-ligand binding poses without relying on heavy sampling. However, their prediction accuracy and applicability are still far from satisfactory, partially due to the lack of protein-ligand binding complex data. To this end, we create a large-scale complex dataset containing ∼9 M protein-ligand docking complexes for pre-training, and propose CarsiDock, the first deep learning-guided docking approach that leverages pre-training of millions of predicted protein-ligand complexes. CarsiDock contains two main stages, i.e., a deep learning model for the prediction of protein-ligand atomic distance matrices, and a translation, rotation and torsion-guided geometry optimization procedure to reconstruct the matrices into a credible binding pose. The pre-training and multiple innovative architectural designs facilitate the dramatically improved docking accuracy of our approach over the baselines in terms of multiple docking scenarios, thereby contributing to its outstanding early recognition performance in several retrospective virtual screening campaigns. Further explorations demonstrate that CarsiDock can not only guarantee the topological reliability of the binding poses but also successfully reproduce the crucial interactions in crystalized structures, highlighting its superior applicability.

16.
Expert Opin Biol Ther ; 24(1-2): 37-50, 2024.
Article in English | MEDLINE | ID: mdl-38247394

ABSTRACT

INTRODUCTION: In 2022, U.S. Food and Drug Administration (FDA) approved the first biologics, intravenous spesolimab, for acute flare of generalized pustular psoriasis (GPP). The drug works by blocking IL-36 signaling, the key pathway of GPP. Among the known mutations causing GPP, IL36RN mutations are most common, and the presence of IL36RN mutations had been found to affect the clinical manifestations and treatment response of GPP. AREAS COVERED: Literature search was conducted in PubMed, Embase and ClinicalTrials.gov for relevant studies discussing biologic treatment for GPP with special emphasis on larger studies, pediatric group, pregnant women, and the influence of IL36RN mutation on the effectiveness of biologics. EXPERT OPINION: The approval of spesolimab for GPP flare treatment marks a new era. However, whether spesolimab will be placed as the treatment of choice remains unknown, considering its higher cost, lack of direct comparison with existing biologics, and uncertain effects on co-existing plaque-type psoriasis. However, the demonstration of numerically better efficacy for patients carrying pathogenic IL36RN mutations suggests the role of pharmacogenetics in the choices of GPP treatment. Future randomized studies are warranted to investigate the effectiveness and safety of biologics for GPP in pediatric and pregnant groups.


Subject(s)
Biological Products , Psoriasis , Pregnancy , Humans , Child , Female , Interleukins/therapeutic use , Psoriasis/drug therapy , Psoriasis/genetics , Psoriasis/pathology , Mutation , Acute Disease , Chronic Disease , Biological Therapy , Biological Products/therapeutic use
17.
Research (Wash D C) ; 7: 0292, 2024.
Article in English | MEDLINE | ID: mdl-38213662

ABSTRACT

Deep learning (DL)-driven efficient synthesis planning may profoundly transform the paradigm for designing novel pharmaceuticals and materials. However, the progress of many DL-assisted synthesis planning (DASP) algorithms has suffered from the lack of reliable automated pathway evaluation tools. As a critical metric for evaluating chemical reactions, accurate prediction of reaction yields helps improve the practicality of DASP algorithms in the real-world scenarios. Currently, accurately predicting yields of interesting reactions still faces numerous challenges, mainly including the absence of high-quality generic reaction yield datasets and robust generic yield predictors. To compensate for the limitations of high-throughput yield datasets, we curated a generic reaction yield dataset containing 12 reaction categories and rich reaction condition information. Subsequently, by utilizing 2 pretraining tasks based on chemical reaction masked language modeling and contrastive learning, we proposed a powerful bidirectional encoder representations from transformers (BERT)-based reaction yield predictor named Egret. It achieved comparable or even superior performance to the best previous models on 4 benchmark datasets and established state-of-the-art performance on the newly curated dataset. We found that reaction-condition-based contrastive learning enhances the model's sensitivity to reaction conditions, and Egret is capable of capturing subtle differences between reactions involving identical reactants and products but different reaction conditions. Furthermore, we proposed a new scoring function that incorporated Egret into the evaluation of multistep synthesis routes. Test results showed that yield-incorporated scoring facilitated the prioritization of literature-supported high-yield reaction pathways for target molecules. In addition, through meta-learning strategy, we further improved the reliability of the model's prediction for reaction types with limited data and lower data quality. Our results suggest that Egret holds the potential to become an essential component of the next-generation DASP tools.

19.
Trends Pharmacol Sci ; 45(2): 103-106, 2024 02.
Article in English | MEDLINE | ID: mdl-38160084

ABSTRACT

Ligand docking (LD), a technology for predicting protein-ligand (PL)-binding conformations and strengths, plays key roles in virtual screening (VS). However, the accuracy and speed of current LD methodologies remain suboptimal. Here, we discuss how deep learning (DL) could help to bridge this gap by examining recent advancements and projecting future trends.


Subject(s)
Deep Learning , Proteins , Humans , Ligands , Proteins/metabolism , Protein Binding , Protein Conformation , Molecular Docking Simulation
20.
J Chem Inf Model ; 63(24): 7617-7627, 2023 Dec 25.
Article in English | MEDLINE | ID: mdl-38079566

ABSTRACT

The application of Explainable Artificial Intelligence (XAI) in the field of chemistry has garnered growing interest for its potential to justify the prediction of black-box machine learning models and provide actionable insights. We first survey a range of XAI techniques adapted for chemical applications and categorize them based on the technical details of each methodology. We then present a few case studies to illustrate the practical utility of XAI, such as identifying carcinogenic molecules and guiding molecular optimizations, in order to provide chemists with concrete examples of ways to take full advantage of XAI-augmented machine learning for chemistry. Despite the initial success of XAI in chemistry, we still face the challenges of developing more reliable explanations, assuring robustness against adversarial actions, and customizing the explanation for different applications and needs of the diverse scientific community. Finally, we discuss the emerging role of large language models like GPT in generating natural language explanations and discusses the specific challenges associated with them. We advocate that addressing the aforementioned challenges and actively embracing new techniques may contribute to establishing machine learning as an indispensable technique for chemistry in this digital era.


Subject(s)
Artificial Intelligence , Machine Learning , Language
SELECTION OF CITATIONS
SEARCH DETAIL
...