首页 > 最新文献

Journal of Chemical Information and Modeling 最新文献

英文 中文
Predicting Collision Cross-Section Values for Small Molecules through Chemical Class-Based Multimodal Graph Attention Network. 通过基于化学类别的多模态图注意网络预测小分子的碰撞截面值
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-07-03 DOI: 10.1021/acs.jcim.3c01934
Cheng Wang, Chuang Yuan, Yahui Wang, Yuying Shi, Tao Zhang, Gary J Patti

Libraries of collision cross-section (CCS) values have the potential to facilitate compound identification in metabolomics. Although computational methods provide an opportunity to increase library size rapidly, accurate prediction of CCS values remains challenging due to the structural diversity of small molecules. Here, we developed a machine learning (ML) model that integrates graph attention networks and multimodal molecular representations to predict CCS values on the basis of chemical class. Our approach, referred to as MGAT-CCS, had superior performance in comparison to other ML models in CCS prediction. MGAT-CCS achieved a median relative error of 0.47%/1.14% (positive/negative mode) and 1.40%/1.63% (positive/negative mode) for lipids and metabolites, respectively. When MGAT-CCS was applied to real-world metabolomics data, it reduced the number of false metabolite candidates by roughly 25% across multiple sample types ranging from plasma and urine to cells. To facilitate its application, we developed a user-friendly stand-alone web server for MGAT-CCS that is freely available at https://mgat-ccs-web.onrender.com. This work represents a step forward in predicting CCS values and can potentially facilitate the identification of small molecules when using ion mobility spectrometry coupled with mass spectrometry.

碰撞截面(CCS)值库有可能促进代谢组学中的化合物鉴定。虽然计算方法为快速增加库规模提供了机会,但由于小分子结构的多样性,准确预测 CCS 值仍具有挑战性。在此,我们开发了一种机器学习(ML)模型,该模型整合了图注意网络和多模态分子表征,可根据化学类别预测 CCS 值。我们的方法被称为 MGAT-CCS,在 CCS 预测方面与其他 ML 模型相比具有更优越的性能。对于脂类和代谢物,MGAT-CCS 的中位相对误差分别为 0.47%/1.14%(正/负模式)和 1.40%/1.63%(正/负模式)。当将 MGAT-CCS 应用于真实世界的代谢组学数据时,它在从血浆、尿液到细胞的多种样本类型中减少了大约 25% 的错误候选代谢物数量。为了方便应用,我们为 MGAT-CCS 开发了一个用户友好的独立网络服务器,可在 https://mgat-ccs-web.onrender.com 免费获取。这项工作标志着我们在预测 CCS 值方面又向前迈进了一步,并有可能在使用离子迁移谱与质谱联用技术时促进小分子的鉴定。
{"title":"Predicting Collision Cross-Section Values for Small Molecules through Chemical Class-Based Multimodal Graph Attention Network.","authors":"Cheng Wang, Chuang Yuan, Yahui Wang, Yuying Shi, Tao Zhang, Gary J Patti","doi":"10.1021/acs.jcim.3c01934","DOIUrl":"https://doi.org/10.1021/acs.jcim.3c01934","url":null,"abstract":"<p><p>Libraries of collision cross-section (CCS) values have the potential to facilitate compound identification in metabolomics. Although computational methods provide an opportunity to increase library size rapidly, accurate prediction of CCS values remains challenging due to the structural diversity of small molecules. Here, we developed a machine learning (ML) model that integrates graph attention networks and multimodal molecular representations to predict CCS values on the basis of chemical class. Our approach, referred to as MGAT-CCS, had superior performance in comparison to other ML models in CCS prediction. MGAT-CCS achieved a median relative error of 0.47%/1.14% (positive/negative mode) and 1.40%/1.63% (positive/negative mode) for lipids and metabolites, respectively. When MGAT-CCS was applied to real-world metabolomics data, it reduced the number of false metabolite candidates by roughly 25% across multiple sample types ranging from plasma and urine to cells. To facilitate its application, we developed a user-friendly stand-alone web server for MGAT-CCS that is freely available at https://mgat-ccs-web.onrender.com. This work represents a step forward in predicting CCS values and can potentially facilitate the identification of small molecules when using ion mobility spectrometry coupled with mass spectrometry.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-07-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141489951","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Chemoinformatics Insights on Molecular Jackhammers and Cancer Cells. 化疗信息学对分子锤和癌细胞的启示
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-07-03 DOI: 10.1021/acs.jcim.4c00806
Ciceron Ayala-Orozco, Hamid Teimouri, Angela Medvedeva, Bowen Li, Alex Lathem, Gang Li, Anatoly B Kolomeisky, James M Tour

One of the most challenging tasks in modern medicine is to find novel efficient cancer therapeutic methods with minimal side effects. The recent discovery of several classes of organic molecules known as "molecular jackhammers" is a promising development in this direction. It is known that these molecules can directly target and eliminate cancer cells with no impact on healthy tissues. However, the underlying microscopic picture remains poorly understood. We present a study that utilizes theoretical analysis together with experimental measurements to clarify the microscopic aspects of jackhammers' anticancer activities. Our physical-chemical approach combines statistical analysis with chemoinformatics methods to design and optimize molecular jackhammers. By correlating specific physical-chemical properties of these molecules with their abilities to kill cancer cells, several important structural features are identified and discussed. Although our theoretical analysis enhances understanding of the molecular interactions of jackhammers, it also highlights the need for further research to comprehensively elucidate their mechanisms and to develop a robust physical-chemical framework for the rational design of targeted anticancer drugs.

现代医学中最具挑战性的任务之一是寻找副作用最小的新型高效癌症治疗方法。最近发现的几类被称为 "分子锤 "的有机分子是这一方向上一个很有希望的发展。众所周知,这些分子可以直接靶向消除癌细胞,而对健康组织没有影响。然而,人们对其背后的微观图景仍然知之甚少。我们的研究利用理论分析和实验测量来阐明千层锤抗癌活性的微观方面。我们的物理化学方法将统计分析与化学信息学方法相结合,以设计和优化分子千层塔。通过将这些分子的特定物理化学特性与其杀死癌细胞的能力相关联,我们发现并讨论了几个重要的结构特征。虽然我们的理论分析加深了人们对 "千斤顶 "分子相互作用的理解,但它也强调了进一步研究的必要性,以全面阐明它们的作用机制,并为靶向抗癌药物的合理设计开发一个强大的物理化学框架。
{"title":"Chemoinformatics Insights on Molecular Jackhammers and Cancer Cells.","authors":"Ciceron Ayala-Orozco, Hamid Teimouri, Angela Medvedeva, Bowen Li, Alex Lathem, Gang Li, Anatoly B Kolomeisky, James M Tour","doi":"10.1021/acs.jcim.4c00806","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00806","url":null,"abstract":"<p><p>One of the most challenging tasks in modern medicine is to find novel efficient cancer therapeutic methods with minimal side effects. The recent discovery of several classes of organic molecules known as \"molecular jackhammers\" is a promising development in this direction. It is known that these molecules can directly target and eliminate cancer cells with no impact on healthy tissues. However, the underlying microscopic picture remains poorly understood. We present a study that utilizes theoretical analysis together with experimental measurements to clarify the microscopic aspects of jackhammers' anticancer activities. Our physical-chemical approach combines statistical analysis with chemoinformatics methods to design and optimize molecular jackhammers. By correlating specific physical-chemical properties of these molecules with their abilities to kill cancer cells, several important structural features are identified and discussed. Although our theoretical analysis enhances understanding of the molecular interactions of jackhammers, it also highlights the need for further research to comprehensively elucidate their mechanisms and to develop a robust physical-chemical framework for the rational design of targeted anticancer drugs.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-07-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141489948","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Docking Score ML: Target-Specific Machine Learning Models Improving Docking-Based Virtual Screening in 155 Targets. Docking Score ML:目标特异性机器学习模型,改进基于对接的 155 个目标的虚拟筛选。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-07-03 DOI: 10.1021/acs.jcim.4c00072
Haihan Liu, Baichun Hu, Peiying Chen, Xiao Wang, Hanxun Wang, Shizun Wang, Jian Wang, Bin Lin, Maosheng Cheng

In drug discovery, molecular docking methods face challenges in accurately predicting energy. Scoring functions used in molecular docking often fail to simulate complex protein-ligand interactions fully and accurately leading to biases and inaccuracies in virtual screening and target predictions. We introduce the "Docking Score ML", developed from an analysis of over 200,000 docked complexes from 155 known targets for cancer treatments. The scoring functions used are founded on bioactivity data sourced from ChEMBL and have been fine-tuned using both supervised machine learning and deep learning techniques. We validated our approach extensively using multiple data sets such as validation of selectivity mechanism, the DUDE, DUD-AD, and LIT-PCBA data sets, and performed a multitarget analysis on drugs like sunitinib. To enhance prediction accuracy, feature fusion techniques were explored. By merging the capabilities of the Graph Convolutional Network (GCN) with multiple docking functions, our results indicated a clear superiority of our methodologies over conventional approaches. These advantages demonstrate that Docking Score ML is an efficient and accurate tool for virtual screening and reverse docking.

在药物发现过程中,分子对接方法在准确预测能量方面面临挑战。分子对接中使用的评分函数往往不能全面准确地模拟复杂的蛋白质配体相互作用,从而导致虚拟筛选和靶点预测的偏差和不准确。我们介绍的 "Docking Score ML "是通过分析 155 个已知癌症治疗靶点的 20 多万个对接复合物而开发的。所使用的评分函数基于来自 ChEMBL 的生物活性数据,并利用监督机器学习和深度学习技术进行了微调。我们利用选择性机制验证、DUDE、DUD-AD 和 LIT-PCBA 数据集等多个数据集广泛验证了我们的方法,并对舒尼替尼等药物进行了多靶点分析。为了提高预测准确性,研究人员探索了特征融合技术。通过将图形卷积网络(GCN)的功能与多种对接函数相结合,我们的结果表明我们的方法明显优于传统方法。这些优势表明,Docking Score ML 是一种高效、准确的虚拟筛选和反向对接工具。
{"title":"Docking Score ML: Target-Specific Machine Learning Models Improving Docking-Based Virtual Screening in 155 Targets.","authors":"Haihan Liu, Baichun Hu, Peiying Chen, Xiao Wang, Hanxun Wang, Shizun Wang, Jian Wang, Bin Lin, Maosheng Cheng","doi":"10.1021/acs.jcim.4c00072","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00072","url":null,"abstract":"<p><p>In drug discovery, molecular docking methods face challenges in accurately predicting energy. Scoring functions used in molecular docking often fail to simulate complex protein-ligand interactions fully and accurately leading to biases and inaccuracies in virtual screening and target predictions. We introduce the \"Docking Score ML\", developed from an analysis of over 200,000 docked complexes from 155 known targets for cancer treatments. The scoring functions used are founded on bioactivity data sourced from ChEMBL and have been fine-tuned using both supervised machine learning and deep learning techniques. We validated our approach extensively using multiple data sets such as validation of selectivity mechanism, the DUDE, DUD-AD, and LIT-PCBA data sets, and performed a multitarget analysis on drugs like sunitinib. To enhance prediction accuracy, feature fusion techniques were explored. By merging the capabilities of the Graph Convolutional Network (GCN) with multiple docking functions, our results indicated a clear superiority of our methodologies over conventional approaches. These advantages demonstrate that Docking Score ML is an efficient and accurate tool for virtual screening and reverse docking.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-07-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141489950","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Accelerating Polymer Discovery with Uncertainty-Guided PGCNN: Explainable AI for Predicting Properties and Mechanistic Insights. 利用不确定性引导的 PGCNN 加速聚合物发现:可解释的人工智能,用于预测特性和机理洞察。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-07-02 DOI: 10.1021/acs.jcim.4c00555
Shuyu Wang, Hongxing Yue, Xiaoming Yuan

Deep learning holds great potential for expediting the discovery of new polymers from the vast chemical space. However, accurately predicting polymer properties for practical applications based on their monomer composition has long been a challenge. The main obstacles include insufficient data, ineffective representation encoding, and lack of explainability. To address these issues, we propose an interpretable model called the Polymer Graph Convolutional Neural Network (PGCNN) that can accurately predict various polymer properties. This model is trained using the RadonPy data set and validated using experimental data samples. By integrating evidential deep learning with the model, we can quantify the uncertainty of predictions and enable sample-efficient training through uncertainty-guided active learning. Additionally, we demonstrate that the global attention of the graph embedding can aid in discovering underlying physical principles by identifying important functional groups within polymers and associating them with specific material attributes. Lastly, we explore the high-throughput screening capability of our model by rapidly identifying thousands of promising candidates with low and high thermal conductivity from a pool of one million hypothetical polymers. In summary, our research not only advances our mechanistic understanding of polymers using explainable AI but also paves the way for data-driven trustworthy discovery of polymer materials.

深度学习在加速从广阔的化学空间中发现新聚合物方面具有巨大潜力。然而,根据单体成分准确预测实际应用中的聚合物特性一直是个难题。主要障碍包括数据不足、表征编码无效以及缺乏可解释性。为了解决这些问题,我们提出了一种可解释的模型,即聚合物图卷积神经网络(PGCNN),它可以准确预测各种聚合物特性。该模型使用 RadonPy 数据集进行训练,并使用实验数据样本进行验证。通过将证据深度学习与该模型相结合,我们可以量化预测的不确定性,并通过不确定性引导的主动学习实现样本高效训练。此外,我们还证明了图嵌入的全局注意力可以通过识别聚合物中的重要官能团并将它们与特定材料属性联系起来,帮助发现潜在的物理原理。最后,我们从一百万种假设聚合物中快速识别出数千种具有低导热性和高导热性的候选聚合物,从而探索了我们模型的高通量筛选能力。总之,我们的研究不仅利用可解释的人工智能推进了我们对聚合物的机理理解,还为数据驱动的聚合物材料可信发现铺平了道路。
{"title":"Accelerating Polymer Discovery with Uncertainty-Guided PGCNN: Explainable AI for Predicting Properties and Mechanistic Insights.","authors":"Shuyu Wang, Hongxing Yue, Xiaoming Yuan","doi":"10.1021/acs.jcim.4c00555","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00555","url":null,"abstract":"<p><p>Deep learning holds great potential for expediting the discovery of new polymers from the vast chemical space. However, accurately predicting polymer properties for practical applications based on their monomer composition has long been a challenge. The main obstacles include insufficient data, ineffective representation encoding, and lack of explainability. To address these issues, we propose an interpretable model called the Polymer Graph Convolutional Neural Network (PGCNN) that can accurately predict various polymer properties. This model is trained using the RadonPy data set and validated using experimental data samples. By integrating evidential deep learning with the model, we can quantify the uncertainty of predictions and enable sample-efficient training through uncertainty-guided active learning. Additionally, we demonstrate that the global attention of the graph embedding can aid in discovering underlying physical principles by identifying important functional groups within polymers and associating them with specific material attributes. Lastly, we explore the high-throughput screening capability of our model by rapidly identifying thousands of promising candidates with low and high thermal conductivity from a pool of one million hypothetical polymers. In summary, our research not only advances our mechanistic understanding of polymers using explainable AI but also paves the way for data-driven trustworthy discovery of polymer materials.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141475401","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CSM Software: Continuous Symmetry and Chirality Measures for Quantitative Structural Analysis. CSM 软件:用于定量结构分析的连续对称性和手性度量。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-07-02 DOI: 10.1021/acs.jcim.4c00609
Inbal Tuvi-Arad, Yaffa Shalit, Gil Alon

We present a comprehensive and updated Python-based open software to calculate continuous symmetry measures (CSMs) and their related continuous chirality measure (CCM) of molecules across chemistry. These descriptors are used to quantify distortion levels of molecular structures on a continuous scale and were proven insightful in numerous studies. The input information includes the coordinates of the molecular geometry and a desired cyclic symmetry point group (i.e., Cs, Ci, Cn, or Sn). The results include the coordinates of the nearest symmetric structure that belong to the desired symmetry point group, the permutation that defines the symmetry operation, the direction of the symmetry element in space, and a number, between zero and 100, representing the level of symmetry or chirality. Rather than treating symmetry as a binary property by which a structure is either symmetric or asymmetric, the CSM approach quantifies the level of gray between black and white and allows one to follow the course of change. The software can be downloaded from https://github.com/continuous-symmetry-measure/csm or used online at https://csm.ouproj.org.il.

我们介绍了一个基于 Python 的全面、最新的开放式软件,用于计算化学分子的连续对称性度量(CSM)及其相关的连续手性度量(CCM)。这些描述符用于量化分子结构在连续尺度上的畸变程度,并在大量研究中得到了证实。输入信息包括分子几何形状的坐标和所需的循环对称点组(即 Cs、Ci、Cn 或 Sn)。结果包括属于所需对称点组的最近对称结构的坐标、定义对称操作的置换、空间对称元素的方向,以及代表对称性或手性级的 0 至 100 之间的数字。CSM 方法不是将对称性视为结构对称或不对称的二元属性,而是量化黑白之间的灰度,并允许人们跟踪变化过程。该软件可从 https://github.com/continuous-symmetry-measure/csm 下载或在 https://csm.ouproj.org.il 在线使用。
{"title":"CSM Software: Continuous Symmetry and Chirality Measures for Quantitative Structural Analysis.","authors":"Inbal Tuvi-Arad, Yaffa Shalit, Gil Alon","doi":"10.1021/acs.jcim.4c00609","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00609","url":null,"abstract":"<p><p>We present a comprehensive and updated Python-based open software to calculate continuous symmetry measures (CSMs) and their related continuous chirality measure (CCM) of molecules across chemistry. These descriptors are used to quantify distortion levels of molecular structures on a continuous scale and were proven insightful in numerous studies. The input information includes the coordinates of the molecular geometry and a desired cyclic symmetry point group (<i>i.e., C</i><sub>s</sub>, <i>C</i><sub>i</sub>, <i>C</i><sub>n</sub>, or <i>S</i><sub>n</sub>). The results include the coordinates of the nearest symmetric structure that belong to the desired symmetry point group, the permutation that defines the symmetry operation, the direction of the symmetry element in space, and a number, between zero and 100, representing the level of symmetry or chirality. Rather than treating symmetry as a binary property by which a structure is either symmetric or asymmetric, the CSM approach quantifies the level of gray between black and white and allows one to follow the course of change. The software can be downloaded from https://github.com/continuous-symmetry-measure/csm or used online at https://csm.ouproj.org.il.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141489949","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Proximity Graph Networks: Predicting Ligand Affinity with Message Passing Neural Networks 邻近图网络:利用信息传递神经网络预测配体亲和性
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-07-02 DOI: 10.1021/acs.jcim.4c00311
Zachary J. Gale-Day, Laura Shub, Kangway V. Chuang, Michael J. Keiser
Message passing neural networks (MPNNs) on molecular graphs generate continuous and differentiable encodings of small molecules with state-of-the-art performance on protein–ligand complex scoring tasks. Here, we describe the proximity graph network (PGN) package, an open-source toolkit that constructs ligand–receptor graphs based on atom proximity and allows users to rapidly apply and evaluate MPNN architectures for a broad range of tasks. We demonstrate the utility of PGN by introducing benchmarks for affinity and docking score prediction tasks. Graph networks generalize better than fingerprint-based models and perform strongly for the docking score prediction task. Overall, MPNNs with proximity graph data structures augment the prediction of ligand–receptor complex properties when ligand–receptor data are available.
分子图上的消息传递神经网络(MPNN)可生成连续且可微分的小分子编码,在蛋白质配体复合物评分任务中具有最先进的性能。在这里,我们介绍了邻近图网络(PGN)软件包,这是一个开源工具包,可根据原子邻近性构建配体-受体图,并允许用户在广泛的任务中快速应用和评估 MPNN 架构。我们通过引入亲和力和对接得分预测任务的基准,展示了 PGN 的实用性。与基于指纹的模型相比,图网络的泛化效果更好,在对接得分预测任务中表现强劲。总体而言,当配体-受体数据可用时,具有接近图数据结构的 MPNNs 可以增强配体-受体复合物特性的预测。
{"title":"Proximity Graph Networks: Predicting Ligand Affinity with Message Passing Neural Networks","authors":"Zachary J. Gale-Day, Laura Shub, Kangway V. Chuang, Michael J. Keiser","doi":"10.1021/acs.jcim.4c00311","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00311","url":null,"abstract":"Message passing neural networks (MPNNs) on molecular graphs generate continuous and differentiable encodings of small molecules with state-of-the-art performance on protein–ligand complex scoring tasks. Here, we describe the proximity graph network (PGN) package, an open-source toolkit that constructs ligand–receptor graphs based on atom proximity and allows users to rapidly apply and evaluate MPNN architectures for a broad range of tasks. We demonstrate the utility of PGN by introducing benchmarks for affinity and docking score prediction tasks. Graph networks generalize better than fingerprint-based models and perform strongly for the docking score prediction task. Overall, MPNNs with proximity graph data structures augment the prediction of ligand–receptor complex properties when ligand–receptor data are available.","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141489782","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ligand-Based Compound Activity Prediction via Few-Shot Learning. 基于配体的化合物活性预测(Few-Shot Learning)。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-07-01 DOI: 10.1021/acs.jcim.4c00485
Peter Eckmann, Jake Anderson, Rose Yu, Michael K Gilson

Predicting the activities of new compounds against biophysical or phenotypic assays based on the known activities of one or a few existing compounds is a common goal in early stage drug discovery. This problem can be cast as a "few-shot learning" challenge, and prior studies have developed few-shot learning methods to classify compounds as active versus inactive. However, the ability to go beyond classification and rank compounds by expected affinity is more valuable. We describe Few-Shot Compound Activity Prediction (FS-CAP), a novel neural architecture trained on a large bioactivity data set to predict compound activities against an assay outside the training set, based on only the activities of a few known compounds against the same assay. Our model aggregates encodings generated from the known compounds and their activities to capture assay information and uses a separate encoder for the new compound whose activity is to be predicted. The new method provides encouraging results relative to traditional chemical-similarity-based techniques as well as other state-of-the-art few-shot learning methods in tests on a variety of ligand-based drug discovery settings and data sets. The code for FS-CAP is available at https://github.com/Rose-STL-Lab/FS-CAP.

根据一种或几种现有化合物的已知活性,预测新化合物在生物物理或表型测定中的活性,是早期药物发现的一个共同目标。这一问题可被视为 "少量学习 "挑战,先前的研究已开发出少量学习方法,可将化合物分为活性和非活性两种。然而,超越分类并根据预期亲和力对化合物进行排序的能力更有价值。我们介绍了 "少量化合物活性预测"(FS-CAP),这是一种在大型生物活性数据集上进行训练的新型神经架构,它可以仅根据少数已知化合物对同一检测方法的活性,预测化合物对训练集之外的检测方法的活性。我们的模型汇总了从已知化合物及其活性生成的编码,以捕捉检测信息,并为要预测其活性的新化合物使用单独的编码器。在对各种配体药物发现设置和数据集的测试中,与传统的基于化学相似性的技术以及其他最先进的少量学习方法相比,新方法取得了令人鼓舞的结果。FS-CAP 的代码见 https://github.com/Rose-STL-Lab/FS-CAP。
{"title":"Ligand-Based Compound Activity Prediction via Few-Shot Learning.","authors":"Peter Eckmann, Jake Anderson, Rose Yu, Michael K Gilson","doi":"10.1021/acs.jcim.4c00485","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00485","url":null,"abstract":"<p><p>Predicting the activities of new compounds against biophysical or phenotypic assays based on the known activities of one or a few existing compounds is a common goal in early stage drug discovery. This problem can be cast as a \"few-shot learning\" challenge, and prior studies have developed few-shot learning methods to classify compounds as active versus inactive. However, the ability to go beyond classification and rank compounds by expected affinity is more valuable. We describe <i>Few-Shot Compound Activity Prediction</i> (FS-CAP), a novel neural architecture trained on a large bioactivity data set to predict compound activities against an assay outside the training set, based on only the activities of a few known compounds against the same assay. Our model aggregates encodings generated from the known compounds and their activities to capture assay information and uses a separate encoder for the new compound whose activity is to be predicted. The new method provides encouraging results relative to traditional chemical-similarity-based techniques as well as other state-of-the-art few-shot learning methods in tests on a variety of ligand-based drug discovery settings and data sets. The code for FS-CAP is available at https://github.com/Rose-STL-Lab/FS-CAP.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141475403","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Revealing Comprehensive Food Functionalities and Mechanisms of Action through Machine Learning. 通过机器学习揭示食品的综合功能和作用机理。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-07-01 DOI: 10.1021/acs.jcim.4c00061
Nanako Inoue, Tomokazu Shibata, Yusuke Tanaka, Hiromu Taguchi, Ryusuke Sawada, Kenshin Goto, Shogo Momokita, Morihiro Aoyagi, Takashi Hirao, Yoshihiro Yamanishi

Foods possess a range of unexplored functionalities; however, fully identifying these functions through empirical means presents significant challenges. In this study, we have proposed an in silico approach to comprehensively predict the functionalities of foods, encompassing even processed foods. This prediction is accomplished through the utilization of machine learning on biomedical big data. Our focus revolves around disease-related protein pathways, wherein we statistically evaluate how the constituent compounds collaboratively regulate these pathways. The proposed method has been employed across 876 foods and 83 diseases, leading to an extensive revelation of both food functionalities and their underlying operational mechanisms. Additionally, this approach identifies food combinations that potentially affect molecular pathways based on interrelationships between food functions within disease-related pathways. Our proposed method holds potential for advancing preventive healthcare.

食品具有一系列尚未开发的功能;然而,通过经验手段全面确定这些功能是一项重大挑战。在这项研究中,我们提出了一种硅学方法来全面预测食品的功能,甚至包括加工食品。这种预测是通过对生物医学大数据的机器学习来实现的。我们的重点是围绕与疾病相关的蛋白质通路,通过统计评估组成化合物如何协同调节这些通路。我们已在 876 种食物和 83 种疾病中采用了所提出的方法,从而广泛揭示了食物的功能及其潜在的运行机制。此外,这种方法还能根据食物功能在疾病相关途径中的相互关系,确定可能影响分子途径的食物组合。我们提出的方法具有推进预防保健的潜力。
{"title":"Revealing Comprehensive Food Functionalities and Mechanisms of Action through Machine Learning.","authors":"Nanako Inoue, Tomokazu Shibata, Yusuke Tanaka, Hiromu Taguchi, Ryusuke Sawada, Kenshin Goto, Shogo Momokita, Morihiro Aoyagi, Takashi Hirao, Yoshihiro Yamanishi","doi":"10.1021/acs.jcim.4c00061","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00061","url":null,"abstract":"<p><p>Foods possess a range of unexplored functionalities; however, fully identifying these functions through empirical means presents significant challenges. In this study, we have proposed an <i>in silico</i> approach to comprehensively predict the functionalities of foods, encompassing even processed foods. This prediction is accomplished through the utilization of machine learning on biomedical big data. Our focus revolves around disease-related protein pathways, wherein we statistically evaluate how the constituent compounds collaboratively regulate these pathways. The proposed method has been employed across 876 foods and 83 diseases, leading to an extensive revelation of both food functionalities and their underlying operational mechanisms. Additionally, this approach identifies food combinations that potentially affect molecular pathways based on interrelationships between food functions within disease-related pathways. Our proposed method holds potential for advancing preventive healthcare.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141475406","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Prediction and Interpretation Microglia Cytotoxicity by Machine Learning. 通过机器学习预测和解释小胶质细胞毒性。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-07-01 DOI: 10.1021/acs.jcim.4c00366
Qing Liu, Dakuo He, Mengmeng Fan, Jinpeng Wang, Zeyu Cui, Hao Wang, Yan Mi, Ning Li, Qingqi Meng, Yue Hou

Ameliorating microglia-mediated neuroinflammation is a crucial strategy in developing new drugs for neurodegenerative diseases. Plant compounds are an important screening target for the discovery of drugs for the treatment of neurodegenerative diseases. However, due to the spatial complexity of phytochemicals, it becomes particularly important to evaluate the effectiveness of compounds while avoiding the mixing of cytotoxic substances in the early stages of compound screening. Traditional high-throughput screening methods suffer from high cost and low efficiency. A computational model based on machine learning provides a novel avenue for cytotoxicity determination. In this study, a microglia cytotoxicity classifier was developed using a machine learning approach. First, we proposed a data splitting strategy based on the molecule murcko generic scaffold, under this condition, three machine learning approaches were coupled with three kinds of molecular representation methods to construct microglia cytotoxicity classifier, which were then compared and assessed by the predictive accuracy, balanced accuracy, F1-score, and Matthews Correlation Coefficient. Then, the recursive feature elimination integrated with support vector machine (RFE-SVC) dimension reduction method was introduced to molecular fingerprints with high dimensions to further improve the model performance. Among all the microglial cytotoxicity classifiers, the SVM coupled with ECFP4 fingerprint after feature selection (ECFP4-RFE-SVM) obtained the most accurate classification for the test set (ACC of 0.99, BA of 0.99, F1-score of 0.99, MCC of 0.97). Finally, the Shapley additive explanations (SHAP) method was used in interpreting the microglia cytotoxicity classifier and key substructure smart identified as structural alerts. Experimental results show that ECFP4-RFE-SVM have reliable classification capability for microglia cytotoxicity, and SHAP can not only provide a rational explanation for microglia cytotoxicity predictions, but also offer a guideline for subsequent molecular cytotoxicity modifications.

改善小胶质细胞介导的神经炎症是开发治疗神经退行性疾病新药的重要策略。植物化合物是发现治疗神经退行性疾病药物的重要筛选目标。然而,由于植物化学物质的空间复杂性,在化合物筛选的早期阶段,既要评估化合物的有效性,又要避免混入细胞毒性物质变得尤为重要。传统的高通量筛选方法成本高、效率低。基于机器学习的计算模型为细胞毒性测定提供了一条新途径。本研究采用机器学习方法开发了小胶质细胞毒性分类器。首先,我们提出了一种基于分子murcko通用支架的数据拆分策略,在此条件下,将三种机器学习方法与三种分子表征方法相结合,构建了小胶质细胞毒性分类器,并通过预测准确率、平衡准确率、F1-score和Matthews相关系数对其进行了比较和评估。然后,针对高维度的分子指纹引入了递归特征消除与支持向量机(RFE-SVC)降维方法,进一步提高了模型的性能。在所有小神经胶质细胞毒性分类器中,特征选择后与 ECFP4 指纹相结合的 SVM(ECFP4-RFE-SVM)对测试集的分类准确度最高(ACC 为 0.99,BA 为 0.99,F1-score 为 0.99,MCC 为 0.97)。最后,在解释小胶质细胞毒性分类器时使用了 Shapley 加性解释(SHAP)方法,并将关键子结构智能识别为结构警报。实验结果表明,ECFP4-RFE-SVM 对小胶质细胞毒性具有可靠的分类能力,SHAP 不仅可以为小胶质细胞毒性预测提供合理解释,还可以为后续的分子细胞毒性修饰提供指导。
{"title":"Prediction and Interpretation Microglia Cytotoxicity by Machine Learning.","authors":"Qing Liu, Dakuo He, Mengmeng Fan, Jinpeng Wang, Zeyu Cui, Hao Wang, Yan Mi, Ning Li, Qingqi Meng, Yue Hou","doi":"10.1021/acs.jcim.4c00366","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00366","url":null,"abstract":"<p><p>Ameliorating microglia-mediated neuroinflammation is a crucial strategy in developing new drugs for neurodegenerative diseases. Plant compounds are an important screening target for the discovery of drugs for the treatment of neurodegenerative diseases. However, due to the spatial complexity of phytochemicals, it becomes particularly important to evaluate the effectiveness of compounds while avoiding the mixing of cytotoxic substances in the early stages of compound screening. Traditional high-throughput screening methods suffer from high cost and low efficiency. A computational model based on machine learning provides a novel avenue for cytotoxicity determination. In this study, a microglia cytotoxicity classifier was developed using a machine learning approach. First, we proposed a data splitting strategy based on the molecule murcko generic scaffold, under this condition, three machine learning approaches were coupled with three kinds of molecular representation methods to construct microglia cytotoxicity classifier, which were then compared and assessed by the predictive accuracy, balanced accuracy, F<sub>1</sub>-score, and Matthews Correlation Coefficient. Then, the recursive feature elimination integrated with support vector machine (RFE-SVC) dimension reduction method was introduced to molecular fingerprints with high dimensions to further improve the model performance. Among all the microglial cytotoxicity classifiers, the SVM coupled with ECFP4 fingerprint after feature selection (ECFP4-RFE-SVM) obtained the most accurate classification for the test set (ACC of 0.99, BA of 0.99, F<sub>1</sub>-score of 0.99, MCC of 0.97). Finally, the Shapley additive explanations (SHAP) method was used in interpreting the microglia cytotoxicity classifier and key substructure smart identified as structural alerts. Experimental results show that ECFP4-RFE-SVM have reliable classification capability for microglia cytotoxicity, and SHAP can not only provide a rational explanation for microglia cytotoxicity predictions, but also offer a guideline for subsequent molecular cytotoxicity modifications.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141464275","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
QSARtuna: An Automated QSAR Modeling Platform for Molecular Property Prediction in Drug Design. QSARtuna:用于药物设计中分子性质预测的 QSAR 自动建模平台。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-07-01 DOI: 10.1021/acs.jcim.4c00457
Lewis Mervin, Alexey Voronov, Mikhail Kabeshov, Ola Engkvist

Machine-learning (ML) and deep-learning (DL) approaches to predict the molecular properties of small molecules are increasingly deployed within the design-make-test-analyze (DMTA) drug design cycle to predict molecular properties of interest. Despite this uptake, there are only a few automated packages to aid their development and deployment that also support uncertainty estimation, model explainability, and other key aspects of model usage. This represents a key unmet need within the field, and the large number of molecular representations and algorithms (and associated parameters) means it is nontrivial to robustly optimize, evaluate, reproduce, and deploy models. Here, we present QSARtuna, a molecule property prediction modeling pipeline, written in Python and utilizing the Optuna, Scikit-learn, RDKit, and ChemProp packages, which enables the efficient and automated comparison between molecular representations and machine learning models. The platform was developed by considering the increasingly important aspect of model uncertainty quantification and explainability by design. We provide details for our framework and provide illustrative examples to demonstrate the capability of the software when applied to simple molecular property, reaction/reactivity prediction, and DNA encoded library enrichment classification. We hope that the release of QSARtuna will further spur innovation in automatic ML modeling and provide a platform for education of best practices in molecular property modeling. The code for the QSARtuna framework is made freely available via GitHub.

在 "设计-制造-测试-分析"(DMTA)药物设计周期中,越来越多地采用机器学习(ML)和深度学习(DL)方法来预测小分子的分子特性。尽管如此,只有少数自动化软件包可以帮助开发和部署这些模型,同时还支持不确定性估计、模型可解释性以及模型使用的其他关键方面。这是该领域尚未满足的一个关键需求,而大量的分子表征和算法(以及相关参数)意味着要稳健地优化、评估、复制和部署模型并非易事。在此,我们介绍 QSARtuna,这是一个用 Python 编写的分子性质预测建模管道,它利用 Optuna、Scikit-learn、RDKit 和 ChemProp 软件包,实现了分子表征与机器学习模型之间的高效自动比较。该平台的开发考虑了日益重要的模型不确定性量化和可解释性设计。我们将详细介绍我们的框架,并举例说明该软件在应用于简单分子特性、反应/活性预测和 DNA 编码文库富集分类时的能力。我们希望 QSARtuna 的发布能进一步推动自动 ML 建模的创新,并为分子性质建模的最佳实践教育提供一个平台。QSARtuna 框架的代码可通过 GitHub 免费获取。
{"title":"QSARtuna: An Automated QSAR Modeling Platform for Molecular Property Prediction in Drug Design.","authors":"Lewis Mervin, Alexey Voronov, Mikhail Kabeshov, Ola Engkvist","doi":"10.1021/acs.jcim.4c00457","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00457","url":null,"abstract":"<p><p>Machine-learning (ML) and deep-learning (DL) approaches to predict the molecular properties of small molecules are increasingly deployed within the design-make-test-analyze (DMTA) drug design cycle to predict molecular properties of interest. Despite this uptake, there are only a few automated packages to aid their development and deployment that also support uncertainty estimation, model explainability, and other key aspects of model usage. This represents a key unmet need within the field, and the large number of molecular representations and algorithms (and associated parameters) means it is nontrivial to robustly optimize, evaluate, reproduce, and deploy models. Here, we present QSARtuna, a molecule property prediction modeling pipeline, written in Python and utilizing the Optuna, Scikit-learn, RDKit, and ChemProp packages, which enables the efficient and automated comparison between molecular representations and machine learning models. The platform was developed by considering the increasingly important aspect of model uncertainty quantification and explainability by design. We provide details for our framework and provide illustrative examples to demonstrate the capability of the software when applied to simple molecular property, reaction/reactivity prediction, and DNA encoded library enrichment classification. We hope that the release of QSARtuna will further spur innovation in automatic ML modeling and provide a platform for education of best practices in molecular property modeling. The code for the QSARtuna framework is made freely available via GitHub.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141475405","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"化学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Journal of Chemical Information and Modeling
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1