首页 > 最新文献

Journal of Chemical Information and Modeling 最新文献

英文 中文
Stereoisomers Are Not Machine Learning's Best Friends. 立体异构体不是机器学习的好朋友
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-07-01 DOI: 10.1021/acs.jcim.4c00318
Gökhan Tahıl, Fabien Delorme, Daniel Le Berre, Éric Monflier, Adlane Sayede, Sébastien Tilloy

This study addresses the challenge of accurately identifying stereoisomers in cheminformatics, which originates from our objective to apply machine learning to predict the association constant between cyclodextrin and a guest. Identifying stereoisomers is indeed crucial for machine learning applications. Current tools offer various molecular descriptors, including their textual representation as Isomeric SMILES that can distinguish stereoisomers. However, such representation is text-based and does not have a fixed size, so a conversion is needed to make it usable to machine learning approaches. Word embedding techniques can be used to solve this problem. Mol2vec, a word embedding approach for molecules, offers such a conversion. Unfortunately, it cannot distinguish between stereoisomers due to its inability to capture the spatial configuration of molecular structures. This study proposes several approaches that use word embedding techniques to handle molecular discrimination using stereochemical information on molecules or considering Isomeric SMILES notation as a text in Natural Language Processing. Our aim is to generate a distinct vector for each unique molecule, correctly identifying stereoisomer information in cheminformatics. The proposed approaches are then compared to our original machine learning task: predicting the association constant between cyclodextrin and a guest molecule.

本研究解决了在化学信息学中准确识别立体异构体的难题,这源于我们应用机器学习预测环糊精与客体之间关联常数的目标。识别立体异构体对于机器学习应用确实至关重要。目前的工具提供了各种分子描述符,包括可以区分立体异构体的异构体 SMILES 文本表述。然而,这种表示法是基于文本的,没有固定的大小,因此需要进行转换,使其适用于机器学习方法。词嵌入技术可用于解决这一问题。分子词嵌入方法 Mol2vec 就提供了这种转换。遗憾的是,由于无法捕捉分子结构的空间构型,它无法区分立体异构体。本研究提出了几种使用单词嵌入技术的方法,利用分子的立体化学信息或将异构体 SMILES 符号视为自然语言处理中的文本来处理分子判别问题。我们的目标是为每个独特的分子生成一个不同的向量,从而正确识别化学信息学中的立体异构体信息。然后将所提出的方法与我们最初的机器学习任务进行比较:预测环糊精与客体分子之间的关联常数。
{"title":"Stereoisomers Are Not Machine Learning's Best Friends.","authors":"Gökhan Tahıl, Fabien Delorme, Daniel Le Berre, Éric Monflier, Adlane Sayede, Sébastien Tilloy","doi":"10.1021/acs.jcim.4c00318","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00318","url":null,"abstract":"<p><p>This study addresses the challenge of accurately identifying stereoisomers in cheminformatics, which originates from our objective to apply machine learning to predict the association constant between cyclodextrin and a guest. Identifying stereoisomers is indeed crucial for machine learning applications. Current tools offer various molecular descriptors, including their textual representation as Isomeric SMILES that can distinguish stereoisomers. However, such representation is text-based and does not have a fixed size, so a conversion is needed to make it usable to machine learning approaches. Word embedding techniques can be used to solve this problem. Mol2vec, a word embedding approach for molecules, offers such a conversion. Unfortunately, it cannot distinguish between stereoisomers due to its inability to capture the spatial configuration of molecular structures. This study proposes several approaches that use word embedding techniques to handle molecular discrimination using stereochemical information on molecules or considering Isomeric SMILES notation as a text in Natural Language Processing. Our aim is to generate a distinct vector for each unique molecule, correctly identifying stereoisomer information in cheminformatics. The proposed approaches are then compared to our original machine learning task: predicting the association constant between cyclodextrin and a guest molecule.</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":"141464276","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
Scaffold-Hopped Compound Identification by Ligand-Based Approaches with a Prospective Affinity Test. 通过基于配体的方法和前瞻性亲和力测试进行支架跳转化合物鉴定。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-07-01 DOI: 10.1021/acs.jcim.4c00342
Itsuki Maeda, Shunsuke Tamura, Yoshihiro Ogura, Takayuki Serizawa, Takashi Shimada, Ryo Kunimoto, Tomoyuki Miyao

Scaffold-hopped (SH) compounds are bioactive compounds structurally different from known active compounds. Identifying SH compounds in the ligand-based approaches has been a central issue in medicinal chemistry, and various molecular representations of scaffold hopping have been proposed. However, appropriate representations for SH compound identification remain unclear. Herein, the ability of SH compound identification among several representations was fairly evaluated based on retrospective validation and prospective demonstration. In the retrospective validation, the combinations of two screening algorithms and four two- and three-dimensional molecular representations were compared using controlled data sets for the early identification of SH compounds. We found that the combination of the support vector machine and extended connectivity fingerprint with bond diameter 4 (SVM-ECFP4) and SVM and the rapid overlay of chemical structures (SVM-ROCS) showed a relatively high performance. The compounds that were highly ranked by SVM-ROCS did not share substructures with the active training compounds, while those ranked by SVM-ECFP4 were mostly recombinant. In the prospective demonstration, 93 SH compounds were prepared by screening the Namiki database using SVM-ROCS, targeting ABL1 inhibitors. The primary screening using surface plasmon resonance suggested five active compounds; however, in the competitive binding assays with adenosine triphosphate, no hits were found.

支架跳跃(SH)化合物是在结构上不同于已知活性化合物的生物活性化合物。在基于配体的方法中识别 SH 化合物一直是药物化学领域的核心问题,人们提出了各种支架跳跃的分子表示方法。然而,用于 SH 化合物鉴定的适当表征仍不明确。在此,我们通过回顾性验证和前瞻性论证,对几种表征之间的SH化合物鉴定能力进行了公正的评估。在回顾性验证中,使用对照数据集比较了两种筛选算法和四种二维和三维分子表征的组合,以早期识别 SH 化合物。我们发现,支持向量机和具有键直径 4 的扩展连通性指纹(SVM-ECFP4)以及 SVM 和化学结构快速叠加(SVM-ROCS)的组合显示出相对较高的性能。SVM-ROCS 排名靠前的化合物与活性训练化合物不共享子结构,而 SVM-ECFP4 排名靠前的化合物大多是重组化合物。在前瞻性演示中,通过使用 SVM-ROCS 对并木数据库进行筛选,制备了 93 个 SH 化合物,目标是 ABL1 抑制剂。利用表面等离子共振进行的初筛提出了五个活性化合物;但是,在与三磷酸腺苷的竞争性结合试验中,没有发现任何命中化合物。
{"title":"Scaffold-Hopped Compound Identification by Ligand-Based Approaches with a Prospective Affinity Test.","authors":"Itsuki Maeda, Shunsuke Tamura, Yoshihiro Ogura, Takayuki Serizawa, Takashi Shimada, Ryo Kunimoto, Tomoyuki Miyao","doi":"10.1021/acs.jcim.4c00342","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00342","url":null,"abstract":"<p><p>Scaffold-hopped (SH) compounds are bioactive compounds structurally different from known active compounds. Identifying SH compounds in the ligand-based approaches has been a central issue in medicinal chemistry, and various molecular representations of scaffold hopping have been proposed. However, appropriate representations for SH compound identification remain unclear. Herein, the ability of SH compound identification among several representations was fairly evaluated based on retrospective validation and prospective demonstration. In the retrospective validation, the combinations of two screening algorithms and four two- and three-dimensional molecular representations were compared using controlled data sets for the early identification of SH compounds. We found that the combination of the support vector machine and extended connectivity fingerprint with bond diameter 4 (SVM-ECFP4) and SVM and the rapid overlay of chemical structures (SVM-ROCS) showed a relatively high performance. The compounds that were highly ranked by SVM-ROCS did not share substructures with the active training compounds, while those ranked by SVM-ECFP4 were mostly recombinant. In the prospective demonstration, 93 SH compounds were prepared by screening the Namiki database using SVM-ROCS, targeting ABL1 inhibitors. The primary screening using surface plasmon resonance suggested five active compounds; however, in the competitive binding assays with adenosine triphosphate, no hits were found.</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":"141475407","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
CapsEnhancer: An Effective Computational Framework for Identifying Enhancers Based on Chaos Game Representation and Capsule Network. CapsEnhancer:基于混沌博弈表征和胶囊网络识别增强子的有效计算框架。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-06-30 DOI: 10.1021/acs.jcim.4c00546
Lantian Yao, Peilin Xie, Jiahui Guan, Chia-Ru Chung, Yixian Huang, Yuxuan Pang, Huacong Wu, Ying-Chih Chiang, Tzong-Yi Lee

Enhancers are a class of noncoding DNA, serving as crucial regulatory elements in governing gene expression by binding to transcription factors. The identification of enhancers holds paramount importance in the field of biology. However, traditional experimental methods for enhancer identification demand substantial human and material resources. Consequently, there is a growing interest in employing computational methods for enhancer prediction. In this study, we propose a two-stage framework based on deep learning, termed CapsEnhancer, for the identification of enhancers and their strengths. CapsEnhancer utilizes chaos game representation to encode DNA sequences into unique images and employs a capsule network to extract local and global features from sequence "images". Experimental results demonstrate that CapsEnhancer achieves state-of-the-art performance in both stages. In the first and second stages, the accuracy surpasses the previous best methods by 8 and 3.5%, reaching accuracies of 94.5 and 95%, respectively. Notably, this study represents the pioneering application of computer vision methods to enhancer identification tasks. Our work not only contributes novel insights to enhancer identification but also provides a fresh perspective for other biological sequence analysis tasks.

增强子是一类非编码 DNA,通过与转录因子结合成为调控基因表达的关键调控元件。增强子的鉴定在生物学领域具有极其重要的意义。然而,传统的增强子鉴定实验方法需要大量的人力和物力。因此,人们对采用计算方法预测增强子越来越感兴趣。在本研究中,我们提出了一个基于深度学习的两阶段框架,称为 CapsEnhancer,用于识别增强子及其强度。CapsEnhancer 利用混沌博弈表示法将 DNA 序列编码成独特的图像,并利用胶囊网络从序列 "图像 "中提取局部和全局特征。实验结果表明,CapsEnhancer 在两个阶段都取得了最先进的性能。在第一和第二阶段,准确率分别比之前的最佳方法高出 8% 和 3.5%,达到 94.5% 和 95%。值得注意的是,这项研究开创性地将计算机视觉方法应用于增强器识别任务。我们的工作不仅为增强子识别提供了新的见解,也为其他生物序列分析任务提供了新的视角。
{"title":"CapsEnhancer: An Effective Computational Framework for Identifying Enhancers Based on Chaos Game Representation and Capsule Network.","authors":"Lantian Yao, Peilin Xie, Jiahui Guan, Chia-Ru Chung, Yixian Huang, Yuxuan Pang, Huacong Wu, Ying-Chih Chiang, Tzong-Yi Lee","doi":"10.1021/acs.jcim.4c00546","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00546","url":null,"abstract":"<p><p>Enhancers are a class of noncoding DNA, serving as crucial regulatory elements in governing gene expression by binding to transcription factors. The identification of enhancers holds paramount importance in the field of biology. However, traditional experimental methods for enhancer identification demand substantial human and material resources. Consequently, there is a growing interest in employing computational methods for enhancer prediction. In this study, we propose a two-stage framework based on deep learning, termed CapsEnhancer, for the identification of enhancers and their strengths. CapsEnhancer utilizes chaos game representation to encode DNA sequences into unique images and employs a capsule network to extract local and global features from sequence \"images\". Experimental results demonstrate that CapsEnhancer achieves state-of-the-art performance in both stages. In the first and second stages, the accuracy surpasses the previous best methods by 8 and 3.5%, reaching accuracies of 94.5 and 95%, respectively. Notably, this study represents the pioneering application of computer vision methods to enhancer identification tasks. Our work not only contributes novel insights to enhancer identification but also provides a fresh perspective for other biological sequence analysis tasks.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141464273","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
Challenging Complexity with Simplicity: Rethinking the Role of Single-Step Models in Computer-Aided Synthesis Planning 以简单挑战复杂:重新思考单步模型在计算机辅助合成规划中的作用
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-06-28 DOI: 10.1021/acs.jcim.4c00432
Junren Li, Kangjie Lin, Jianfeng Pei, Luhua Lai
Computer-assisted synthesis planning has become increasingly important in drug discovery. While deep-learning models have shown remarkable progress in achieving high accuracies for single-step retrosynthetic predictions, their performances in retrosynthetic route planning need to be checked. This study compares the intricate single-step models with a straightforward template enumeration approach for retrosynthetic route planning on a real-world drug molecule data set. Despite the superior single-step accuracy of advanced models, the template enumeration method with a heuristic-based retrosynthesis knowledge score was found to surpass them in efficiency in searching the reaction space, achieving a higher or comparable solve rate within the same time frame. This counterintuitive result underscores the importance of efficiency and retrosynthesis knowledge in retrosynthesis route planning and suggests that future research should incorporate a simple template enumeration as a benchmark. It also suggests that this simple yet effective strategy should be considered alongside more complex models to better cater to the practical needs of computer-assisted synthesis planning in drug discovery.
计算机辅助合成规划在药物发现中变得越来越重要。虽然深度学习模型在实现单步逆合成预测的高准确度方面取得了显著进展,但它们在逆合成路线规划方面的表现仍有待检验。本研究在真实世界的药物分子数据集上,比较了复杂的单步模型和直接的模板枚举法在逆向合成路线规划中的表现。尽管高级模型的单步精确度较高,但基于启发式逆合成知识评分的模板枚举法在搜索反应空间方面的效率却超过了高级模型,在相同的时间框架内实现了更高或相当的求解率。这一违反直觉的结果强调了效率和逆合成知识在逆合成路线规划中的重要性,并建议未来的研究应以简单的模板枚举法为基准。它还表明,这种简单而有效的策略应与更复杂的模型一起考虑,以更好地满足药物发现中计算机辅助合成规划的实际需要。
{"title":"Challenging Complexity with Simplicity: Rethinking the Role of Single-Step Models in Computer-Aided Synthesis Planning","authors":"Junren Li, Kangjie Lin, Jianfeng Pei, Luhua Lai","doi":"10.1021/acs.jcim.4c00432","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00432","url":null,"abstract":"Computer-assisted synthesis planning has become increasingly important in drug discovery. While deep-learning models have shown remarkable progress in achieving high accuracies for single-step retrosynthetic predictions, their performances in retrosynthetic route planning need to be checked. This study compares the intricate single-step models with a straightforward template enumeration approach for retrosynthetic route planning on a real-world drug molecule data set. Despite the superior single-step accuracy of advanced models, the template enumeration method with a heuristic-based retrosynthesis knowledge score was found to surpass them in efficiency in searching the reaction space, achieving a higher or comparable solve rate within the same time frame. This counterintuitive result underscores the importance of efficiency and retrosynthesis knowledge in retrosynthesis route planning and suggests that future research should incorporate a simple template enumeration as a benchmark. It also suggests that this simple yet effective strategy should be considered alongside more complex models to better cater to the practical needs of computer-assisted synthesis planning in drug discovery.","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-06-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141463620","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
Exploring Structural Requirements for Sigma-1 Receptor Linear Ligands: Experimental and Computational Approaches 探索 Sigma-1 受体线性配体的结构要求:实验和计算方法
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-06-28 DOI: 10.1021/acs.jcim.4c00500
Lisa Lombardo, Salvatore Mirabile, Rosaria Gitto, Giuseppe Cosentino, Stefano Alcaro, Maria Dichiara, Agostino Marrazzo, Emanuele Amata, Francesco Ortuso, Laura De Luca
Sigma-1 receptor (S1R) is involved in a large array of biological functions due to its ability to interact with various proteins and ion channels. Crystal structures of human S1R revealed the trimeric organization for which each protomer comprises the ligand binding pocket. This study applied a multistep computational procedure to develop a pharmacophore model obtained from molecular dynamics simulations of available cocrystal structures of well-known S1R ligands. Apart from the well-established positive ionizable and hydrophobic features, the obtained model included an additional specific hydrophobic feature and different excluded volumes, thus increasing the selectivity of the model as well as a more detailed determination of the distance between two essential features. The obtained pharmacophore model passed the validation test by receiver operating characteristic (ROC) curve analysis of active and inactive S1R ligands. Finally, the pharmacophoric performance was experimentally investigated through the synthesis and binding assay of new 4-phenylpiperazine-based compounds. The most active new ligand 2-(3-methyl-1-piperidyl)-1-(4-phenylpiperazin-1-yl)ethanone (3) showed an S1R affinity close to the reference compound haloperidol (Ki values of 4.8 and 2.6 nM, respectively). The proposed pharmacophore model can represent a useful tool to design and discover new potent S1R ligands.
Sigma-1 受体(S1R)能够与各种蛋白质和离子通道相互作用,因此参与了大量的生物功能。人类 S1R 的晶体结构揭示了三聚体组织,其中每个原体都是配体结合口袋。本研究采用了一个多步骤计算程序,通过对现有的著名 S1R 配体共晶体结构进行分子动力学模拟,建立了一个药理模型。除了公认的正电离和疏水特征外,所获得的模型还包括一个额外的特定疏水特征和不同的排除体积,从而提高了模型的选择性,并更详细地确定了两个基本特征之间的距离。通过对活性和非活性 S1R 配体进行接收器操作特征曲线(ROC)分析,所获得的药效模型通过了验证测试。最后,通过合成和结合测定新的 4-苯基哌嗪基化合物,对其药效性能进行了实验研究。活性最强的新配体 2-(3-甲基-1-哌啶基)-1-(4-苯基哌嗪-1-基)乙酮(3)的 S1R 亲和力接近参考化合物氟哌啶醇(Ki 值分别为 4.8 和 2.6 nM)。所提出的药理模型是设计和发现新的强效 S1R 配体的有用工具。
{"title":"Exploring Structural Requirements for Sigma-1 Receptor Linear Ligands: Experimental and Computational Approaches","authors":"Lisa Lombardo, Salvatore Mirabile, Rosaria Gitto, Giuseppe Cosentino, Stefano Alcaro, Maria Dichiara, Agostino Marrazzo, Emanuele Amata, Francesco Ortuso, Laura De Luca","doi":"10.1021/acs.jcim.4c00500","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00500","url":null,"abstract":"Sigma-1 receptor (S1R) is involved in a large array of biological functions due to its ability to interact with various proteins and ion channels. Crystal structures of human S1R revealed the trimeric organization for which each protomer comprises the ligand binding pocket. This study applied a multistep computational procedure to develop a pharmacophore model obtained from molecular dynamics simulations of available cocrystal structures of well-known S1R ligands. Apart from the well-established positive ionizable and hydrophobic features, the obtained model included an additional specific hydrophobic feature and different excluded volumes, thus increasing the selectivity of the model as well as a more detailed determination of the distance between two essential features. The obtained pharmacophore model passed the validation test by receiver operating characteristic (ROC) curve analysis of active and inactive S1R ligands. Finally, the pharmacophoric performance was experimentally investigated through the synthesis and binding assay of new 4-phenylpiperazine-based compounds. The most active new ligand 2-(3-methyl-1-piperidyl)-1-(4-phenylpiperazin-1-yl)ethanone (<b>3</b>) showed an S1R affinity close to the reference compound haloperidol (<i>K</i><sub>i</sub> values of 4.8 and 2.6 nM, respectively). The proposed pharmacophore model can represent a useful tool to design and discover new potent S1R ligands.","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-06-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141463670","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 of Vacuum Ultraviolet/Ultraviolet Gas-Phase Absorption Spectra Using Molecular Feature Representations and Machine Learning 利用分子特征表示和机器学习预测真空紫外/紫外气相吸收光谱
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-06-28 DOI: 10.1021/acs.jcim.4c00676
Linh Ho Manh, Victoria C. P. Chen, Jay Rosenberger, Shouyi Wang, Yujing Yang, Kevin A. Schug
Ultraviolet (UV) absorption spectroscopy is a widely used tool for quantitative and qualitative analyses of chemical compounds. In the gas phase, vacuum UV (VUV) and UV absorption spectra are specific and diagnostic for many small molecules. An accurate prediction of VUV/UV absorption spectra can aid the characterization of new or unknown molecules in areas such as fuels, forensics, and pharmaceutical research. An alternative to quantum chemical spectral prediction is the use of artificial intelligence. Here, different molecular feature representation techniques were used and developed to encode chemical structures for testing three machine learning models to predict gas-phase VUV/UV absorption spectra. Structure data files (.sdf) and VUV/UV absorption spectra for 1397 volatile and semivolatile chemical compounds were used to train and test the models. New molecular features (termed ABOCH) were introduced to better capture pi-bonding, aromaticity, and halogenation. The incorporation of these new features benefited spectral prediction and demonstrated superior performance compared to computationally intensive molecular-based deep learning methods. Of the machine learning methods, the use of a Random Forest regressor returned the best accuracy score with the shortest training time. The developed machine learning prediction model also outperformed spectral predictions based on the time-dependent density functional theory.
紫外线(UV)吸收光谱是一种广泛用于定量和定性分析化合物的工具。在气相中,真空紫外(VUV)和紫外吸收光谱对许多小分子具有特异性和诊断性。准确预测真空紫外/紫外吸收光谱有助于鉴定燃料、法医和药物研究等领域的新分子或未知分子。量子化学光谱预测的另一种方法是使用人工智能。在此,我们使用并开发了不同的分子特征表示技术来编码化学结构,以测试预测气相紫外/紫外吸收光谱的三种机器学习模型。1397 种挥发性和半挥发性化合物的结构数据文件(.sdf)和紫外/紫外吸收光谱被用来训练和测试模型。引入了新的分子特征(称为 ABOCH),以更好地捕捉π键、芳香性和卤化。与计算密集型的基于分子的深度学习方法相比,这些新特征的加入有利于光谱预测,并表现出更优越的性能。在机器学习方法中,使用随机森林回归器的准确率最高,训练时间最短。所开发的机器学习预测模型也优于基于时变密度泛函理论的光谱预测。
{"title":"Prediction of Vacuum Ultraviolet/Ultraviolet Gas-Phase Absorption Spectra Using Molecular Feature Representations and Machine Learning","authors":"Linh Ho Manh, Victoria C. P. Chen, Jay Rosenberger, Shouyi Wang, Yujing Yang, Kevin A. Schug","doi":"10.1021/acs.jcim.4c00676","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00676","url":null,"abstract":"Ultraviolet (UV) absorption spectroscopy is a widely used tool for quantitative and qualitative analyses of chemical compounds. In the gas phase, vacuum UV (VUV) and UV absorption spectra are specific and diagnostic for many small molecules. An accurate prediction of VUV/UV absorption spectra can aid the characterization of new or unknown molecules in areas such as fuels, forensics, and pharmaceutical research. An alternative to quantum chemical spectral prediction is the use of artificial intelligence. Here, different molecular feature representation techniques were used and developed to encode chemical structures for testing three machine learning models to predict gas-phase VUV/UV absorption spectra. Structure data files (.sdf) and VUV/UV absorption spectra for 1397 volatile and semivolatile chemical compounds were used to train and test the models. New molecular features (termed ABOCH) were introduced to better capture pi-bonding, aromaticity, and halogenation. The incorporation of these new features benefited spectral prediction and demonstrated superior performance compared to computationally intensive molecular-based deep learning methods. Of the machine learning methods, the use of a Random Forest regressor returned the best accuracy score with the shortest training time. The developed machine learning prediction model also outperformed spectral predictions based on the time-dependent density functional theory.","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-06-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141463557","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
Boosting the Accuracy and Chemical Space Coverage of the Detection of Small Colloidal Aggregating Molecules Using the BAD Molecule Filter. 利用 BAD 分子过滤器提高胶体聚集小分子检测的准确性和化学空间覆盖率。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-06-26 DOI: 10.1021/acs.jcim.4c00363
Abdallah Abou Hajal, Richard A Bryce, Boulbaba Ben Amor, Noor Atatreh, Mohammad A Ghattas

The ability to conduct effective high throughput screening (HTS) campaigns in drug discovery is often hampered by the detection of false positives in these assays due to small colloidally aggregating molecules (SCAMs). SCAMs can produce artifactual hits in HTS by nonspecific inhibition of the protein target. In this work, we present a new computational prediction tool for detecting SCAMs based on their 2D chemical structure. The tool, called the boosted aggregation detection (BAD) molecule filter, employs decision tree ensemble methods, namely, the CatBoost classifier and the light gradient-boosting machine, to significantly improve the detection of SCAMs. In developing the filter, we explore models trained on individual data sets, a consensus approach using these models, and, third, a merged data set approach, each tailored for specific drug discovery needs. The individual data set method emerged as most effective, achieving 93% sensitivity and 90% specificity, outperforming existing state-of-the-art models by 20 and 5%, respectively. The consensus models offer broader chemical space coverage, exceeding 90% for all testing sets. This feature is an important aspect particularly for early stage medicinal chemistry projects, and provides information on applicability domain. Meanwhile, the merged data set models demonstrated robust performance, with a notable sensitivity of 79% in the comprehensive 10-fold cross-validation test set. A SHAP analysis of model features indicates the importance of hydrophobicity and molecular complexity as primary factors influencing the aggregation propensity. The BAD molecule filter is readily accessible for the public usage on https://molmodlab-aau.com/Tools.html. This filter provides a new, more robust tool for aggregate prediction in the early stages of drug discovery to optimize hit rates and reduce associated testing and validation overheads.

在药物发现过程中,进行有效的高通量筛选(HTS)活动的能力往往会受到小胶体聚集分子(SCAMs)在这些检测中产生假阳性的影响。SCAM 会对蛋白质靶点产生非特异性抑制,从而在 HTS 中产生假阳性。在这项工作中,我们提出了一种基于二维化学结构检测 SCAM 的新型计算预测工具。该工具被称为 "增强聚集检测(BAD)分子过滤器",它采用了决策树集合方法,即 CatBoost 分类器和光梯度提升机,从而显著提高了 SCAM 的检测率。在开发该过滤器的过程中,我们探索了在单个数据集上训练的模型、使用这些模型的共识方法,以及第三种合并数据集方法,每种方法都是针对特定的药物发现需求量身定制的。单个数据集方法最为有效,灵敏度达到 93%,特异性达到 90%,分别比现有的最先进模型高出 20% 和 5%。共识模型提供了更广泛的化学空间覆盖率,所有测试集的覆盖率都超过了 90%。这一特点对于早期阶段的药物化学项目尤为重要,并提供了适用领域的信息。同时,合并数据集模型表现出了强劲的性能,在综合 10 倍交叉验证测试集中的灵敏度高达 79%。对模型特征的 SHAP 分析表明,疏水性和分子复杂性是影响聚集倾向的主要因素。BAD 分子过滤器可在 https://molmodlab-aau.com/Tools.html 上供公众使用。该过滤器为药物发现早期阶段的聚集预测提供了一种新的、更强大的工具,可优化命中率并减少相关的测试和验证开销。
{"title":"Boosting the Accuracy and Chemical Space Coverage of the Detection of Small Colloidal Aggregating Molecules Using the BAD Molecule Filter.","authors":"Abdallah Abou Hajal, Richard A Bryce, Boulbaba Ben Amor, Noor Atatreh, Mohammad A Ghattas","doi":"10.1021/acs.jcim.4c00363","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00363","url":null,"abstract":"<p><p>The ability to conduct effective high throughput screening (HTS) campaigns in drug discovery is often hampered by the detection of false positives in these assays due to small colloidally aggregating molecules (SCAMs). SCAMs can produce artifactual hits in HTS by nonspecific inhibition of the protein target. In this work, we present a new computational prediction tool for detecting SCAMs based on their 2D chemical structure. The tool, called the boosted aggregation detection (BAD) molecule filter, employs decision tree ensemble methods, namely, the CatBoost classifier and the light gradient-boosting machine, to significantly improve the detection of SCAMs. In developing the filter, we explore models trained on individual data sets, a consensus approach using these models, and, third, a merged data set approach, each tailored for specific drug discovery needs. The individual data set method emerged as most effective, achieving 93% sensitivity and 90% specificity, outperforming existing state-of-the-art models by 20 and 5%, respectively. The consensus models offer broader chemical space coverage, exceeding 90% for all testing sets. This feature is an important aspect particularly for early stage medicinal chemistry projects, and provides information on applicability domain. Meanwhile, the merged data set models demonstrated robust performance, with a notable sensitivity of 79% in the comprehensive 10-fold cross-validation test set. A SHAP analysis of model features indicates the importance of hydrophobicity and molecular complexity as primary factors influencing the aggregation propensity. The BAD molecule filter is readily accessible for the public usage on https://molmodlab-aau.com/Tools.html. This filter provides a new, more robust tool for aggregate prediction in the early stages of drug discovery to optimize hit rates and reduce associated testing and validation overheads.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141449037","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
TransfIGN: A Structure-Based Deep Learning Method for Modeling the Interaction between HLA-A*02:01 and Antigen Peptides. TransfIGN:一种基于结构的深度学习方法,用于模拟 HLA-A*02:01 与抗原多肽之间的相互作用。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-06-26 DOI: 10.1021/acs.jcim.4c00678
Nanqi Hong, Dejun Jiang, Zhe Wang, Huiyong Sun, Hao Luo, Lingjie Bao, Mingli Song, Yu Kang, Tingjun Hou

The intricate interaction between major histocompatibility complexes (MHCs) and antigen peptides with diverse amino acid sequences plays a pivotal role in immune responses and T cell activity. In recent years, deep learning (DL)-based models have emerged as promising tools for accelerating antigen peptide screening. However, most of these models solely rely on one-dimensional amino acid sequences, overlooking crucial information required for the three-dimensional (3-D) space binding process. In this study, we propose TransfIGN, a structure-based DL model that is inspired by our previously developed framework, Interaction Graph Network (IGN), and incorporates sequence information from transformers to predict the interactions between HLA-A*02:01 and antigen peptides. Our model, trained on a comprehensive data set containing 61,816 sequences with 9051 binding affinity labels and 56,848 eluted ligand labels, achieves an area under the curve (AUC) of 0.893 on the binary data set, better than state-of-the-art sequence-based models trained on larger data sets such as NetMHCpan4.1, ANN, and TransPHLA. Furthermore, when evaluated on the IEDB weekly benchmark data sets, our predictions (AUC = 0.816) are better than those of the recommended methods like the IEDB consensus (AUC = 0.795). Notably, the interaction weight matrices generated by our method highlight the strong interactions at specific positions within peptides, emphasizing the model's ability to provide physical interpretability. This capability to unveil binding mechanisms through intricate structural features holds promise for new immunotherapeutic avenues.

主要组织相容性复合物(MHC)与具有不同氨基酸序列的抗原肽之间错综复杂的相互作用在免疫反应和 T 细胞活性中起着关键作用。近年来,基于深度学习(DL)的模型已成为加速抗原肽筛选的有前途的工具。然而,这些模型大多仅依赖于一维氨基酸序列,忽略了三维(3-D)空间结合过程所需的关键信息。在本研究中,我们提出了基于结构的DL模型TransfIGN,该模型受我们之前开发的框架--相互作用图网络(IGN)的启发,并结合了转化物的序列信息来预测HLA-A*02:01与抗原肽之间的相互作用。我们的模型是在包含 61,816 个序列、9051 个结合亲和力标签和 56,848 个洗脱配体标签的综合数据集上训练出来的,在二进制数据集上的曲线下面积(AUC)达到了 0.893,优于在更大数据集上训练出来的基于序列的先进模型,如 NetMHCpan4.1、ANN 和 TransPHLA。此外,在 IEDB 每周基准数据集上进行评估时,我们的预测结果(AUC = 0.816)优于 IEDB 共识(AUC = 0.795)等推荐方法。值得注意的是,我们的方法生成的相互作用权重矩阵突出了肽段内特定位置的强相互作用,强调了模型提供物理可解释性的能力。这种通过复杂的结构特征揭示结合机制的能力为新的免疫治疗途径带来了希望。
{"title":"TransfIGN: A Structure-Based Deep Learning Method for Modeling the Interaction between HLA-A*02:01 and Antigen Peptides.","authors":"Nanqi Hong, Dejun Jiang, Zhe Wang, Huiyong Sun, Hao Luo, Lingjie Bao, Mingli Song, Yu Kang, Tingjun Hou","doi":"10.1021/acs.jcim.4c00678","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00678","url":null,"abstract":"<p><p>The intricate interaction between major histocompatibility complexes (MHCs) and antigen peptides with diverse amino acid sequences plays a pivotal role in immune responses and T cell activity. In recent years, deep learning (DL)-based models have emerged as promising tools for accelerating antigen peptide screening. However, most of these models solely rely on one-dimensional amino acid sequences, overlooking crucial information required for the three-dimensional (3-D) space binding process. In this study, we propose TransfIGN, a structure-based DL model that is inspired by our previously developed framework, Interaction Graph Network (IGN), and incorporates sequence information from transformers to predict the interactions between HLA-A*02:01 and antigen peptides. Our model, trained on a comprehensive data set containing 61,816 sequences with 9051 binding affinity labels and 56,848 eluted ligand labels, achieves an area under the curve (AUC) of 0.893 on the binary data set, better than state-of-the-art sequence-based models trained on larger data sets such as NetMHCpan4.1, ANN, and TransPHLA. Furthermore, when evaluated on the IEDB weekly benchmark data sets, our predictions (AUC = 0.816) are better than those of the recommended methods like the IEDB consensus (AUC = 0.795). Notably, the interaction weight matrices generated by our method highlight the strong interactions at specific positions within peptides, emphasizing the model's ability to provide physical interpretability. This capability to unveil binding mechanisms through intricate structural features holds promise for new immunotherapeutic avenues.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141449043","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
Correction to "Sensitivity of the RNA Structure to Ion Conditions as Probed by Molecular Dynamics Simulations of Common Canonical RNA Duplexes". 对 "通过分子动力学模拟探究常见典型 RNA 双链体的 RNA 结构对离子条件的敏感性 "的更正。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-06-26 DOI: 10.1021/acs.jcim.4c01067
Petra Kührová, Vojtěch Mlýnský, Michal Otyepka, Jiří Šponer, Pavel Banáš
{"title":"Correction to \"Sensitivity of the RNA Structure to Ion Conditions as Probed by Molecular Dynamics Simulations of Common Canonical RNA Duplexes\".","authors":"Petra Kührová, Vojtěch Mlýnský, Michal Otyepka, Jiří Šponer, Pavel Banáš","doi":"10.1021/acs.jcim.4c01067","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c01067","url":null,"abstract":"","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141453732","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
DrugFlow: An AI-Driven One-Stop Platform for Innovative Drug Discovery. DrugFlow:人工智能驱动的创新药物发现一站式平台。
IF 5.6 2区 化学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-06-26 DOI: 10.1021/acs.jcim.4c00621
Chao Shen, Jianfei Song, Chang-Yu Hsieh, Dongsheng Cao, Yu Kang, Wenling Ye, Zhenxing Wu, Jike Wang, Odin Zhang, Xujun Zhang, Hao Zeng, Heng Cai, Yu Chen, Linkang Chen, Hao Luo, Xinda Zhao, Tianye Jian, Tong Chen, Dejun Jiang, Mingyang Wang, Qing Ye, Jialu Wu, Hongyan Du, Hui Shi, Yafeng Deng, Tingjun Hou

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.

人工智能(AI)辅助药物设计对现代药物发现产生了前所未有的影响,但目前仍迫切需要用户友好型界面,以弥合这些复杂工具与科学家之间的差距,尤其是那些不太精通计算机的科学家。在此,我们介绍了人工智能驱动的一站式平台DrugFlow,它提供了一个简洁、方便、基于云的界面,可简化早期药物发现工作流程。通过无缝集成一系列创新的人工智能算法,包括分子对接、定量结构-活性关系建模、分子生成、ADMET(吸收、分布、代谢、排泄和毒性)预测和虚拟筛选,DrugFlow 可以为早期药物发现的几乎所有关键阶段提供有效的人工智能解决方案,包括新药发现和新药/先导物优化。我们希望该平台能提供足够有价值的指导,帮助实际药物设计和发现。该平台可在 https://drugflow.com 上获取。
{"title":"DrugFlow: An AI-Driven One-Stop Platform for Innovative Drug Discovery.","authors":"Chao Shen, Jianfei Song, Chang-Yu Hsieh, Dongsheng Cao, Yu Kang, Wenling Ye, Zhenxing Wu, Jike Wang, Odin Zhang, Xujun Zhang, Hao Zeng, Heng Cai, Yu Chen, Linkang Chen, Hao Luo, Xinda Zhao, Tianye Jian, Tong Chen, Dejun Jiang, Mingyang Wang, Qing Ye, Jialu Wu, Hongyan Du, Hui Shi, Yafeng Deng, Tingjun Hou","doi":"10.1021/acs.jcim.4c00621","DOIUrl":"https://doi.org/10.1021/acs.jcim.4c00621","url":null,"abstract":"<p><p>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.</p>","PeriodicalId":44,"journal":{"name":"Journal of Chemical Information and Modeling ","volume":null,"pages":null},"PeriodicalIF":5.6,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141449041","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