首页 > 最新文献

Journal of Computer-Aided Molecular Design最新文献

英文 中文
Correlation of protein binding pocket properties with hits' chemistries used in generation of ultra-large virtual libraries. 用于生成超大型虚拟库的蛋白质结合袋特性与命中化学成分的相关性。
IF 3.5 3区 生物学 Q1 Chemistry Pub Date : 2024-05-16 DOI: 10.1007/s10822-024-00562-4
Robert X Song, Marc C Nicklaus, Nadya I Tarasova

Although the size of virtual libraries of synthesizable compounds is growing rapidly, we are still enumerating only tiny fractions of the drug-like chemical universe. Our capability to mine these newly generated libraries also lags their growth. That is why fragment-based approaches that utilize on-demand virtual combinatorial libraries are gaining popularity in drug discovery. These à la carte libraries utilize synthetic blocks found to be effective binders in parts of target protein pockets and a variety of reliable chemistries to connect them. There is, however, no data on the potential impact of the chemistries used for making on-demand libraries on the hit rates during virtual screening. There are also no rules to guide in the selection of these synthetic methods for production of custom libraries. We have used the SAVI (Synthetically Accessible Virtual Inventory) library, constructed using 53 reliable reaction types (transforms), to evaluate the impact of these chemistries on docking hit rates for 40 well-characterized protein pockets. The data shows that the virtual hit rates differ significantly for different chemistries with cross coupling reactions such as Sonogashira, Suzuki-Miyaura, Hiyama and Liebeskind-Srogl coupling producing the highest hit rates. Virtual hit rates appear to depend not only on the property of the formed chemical bond but also on the diversity of available building blocks and the scope of the reaction. The data identifies reactions that deserve wider use through increasing the number of corresponding building blocks and suggests the reactions that are more effective for pockets with certain physical and hydrogen bond-forming properties.

尽管可合成化合物虚拟库的规模正在迅速增长,但我们仍然只列举了类药物化学宇宙中的极小部分。我们挖掘这些新生成化合物库的能力也落后于它们的增长。这就是为什么利用按需虚拟组合库的基于片段的方法在药物发现领域越来越受欢迎的原因。这些 "点菜式 "文库利用在目标蛋白质口袋部分有效结合的合成块,并利用各种可靠的化学方法将它们连接起来。然而,目前还没有数据表明,用于制作按需文库的化学物质对虚拟筛选过程中的命中率有潜在影响。在选择这些合成方法来生产定制文库时,也没有任何指导规则。我们使用 53 种可靠的反应类型(转换)构建的 SAVI(可合成虚拟库存)库,评估了这些化学方法对 40 个特征明确的蛋白质口袋的对接命中率的影响。数据显示,不同化学反应的虚拟命中率差别很大,交叉偶联反应(如 Sonogashira、Suzuki-Miyaura、Hiyama 和 Liebeskind-Srogl 偶联)的命中率最高。虚拟命中率似乎不仅取决于所形成化学键的性质,还取决于可用构件的多样性和反应的范围。这些数据确定了值得通过增加相应构筑模块的数量来更广泛使用的反应,并提出了对具有某些物理和氢键形成特性的口袋更有效的反应。
{"title":"Correlation of protein binding pocket properties with hits' chemistries used in generation of ultra-large virtual libraries.","authors":"Robert X Song, Marc C Nicklaus, Nadya I Tarasova","doi":"10.1007/s10822-024-00562-4","DOIUrl":"10.1007/s10822-024-00562-4","url":null,"abstract":"<p><p>Although the size of virtual libraries of synthesizable compounds is growing rapidly, we are still enumerating only tiny fractions of the drug-like chemical universe. Our capability to mine these newly generated libraries also lags their growth. That is why fragment-based approaches that utilize on-demand virtual combinatorial libraries are gaining popularity in drug discovery. These à la carte libraries utilize synthetic blocks found to be effective binders in parts of target protein pockets and a variety of reliable chemistries to connect them. There is, however, no data on the potential impact of the chemistries used for making on-demand libraries on the hit rates during virtual screening. There are also no rules to guide in the selection of these synthetic methods for production of custom libraries. We have used the SAVI (Synthetically Accessible Virtual Inventory) library, constructed using 53 reliable reaction types (transforms), to evaluate the impact of these chemistries on docking hit rates for 40 well-characterized protein pockets. The data shows that the virtual hit rates differ significantly for different chemistries with cross coupling reactions such as Sonogashira, Suzuki-Miyaura, Hiyama and Liebeskind-Srogl coupling producing the highest hit rates. Virtual hit rates appear to depend not only on the property of the formed chemical bond but also on the diversity of available building blocks and the scope of the reaction. The data identifies reactions that deserve wider use through increasing the number of corresponding building blocks and suggests the reactions that are more effective for pockets with certain physical and hydrogen bond-forming properties.</p>","PeriodicalId":621,"journal":{"name":"Journal of Computer-Aided Molecular Design","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-05-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11098933/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140943273","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Reactivities of acrylamide warheads toward cysteine targets: a QM/ML approach to covalent inhibitor design 丙烯酰胺弹头对半胱氨酸目标的反应活性:共价抑制剂设计的 QM/ML 方法
IF 3.5 3区 生物学 Q1 Chemistry Pub Date : 2024-05-01 DOI: 10.1007/s10822-024-00560-6
Aaron D. Danilack, Callum J. Dickson, Cihan Soylu, Mike Fortunato, Stephane Rodde, Hagen Munkler, Viktor Hornak, Jose S. Duca

Covalent inhibition offers many advantages over non-covalent inhibition, but covalent warhead reactivity must be carefully balanced to maintain potency while avoiding unwanted side effects. While warhead reactivities are commonly measured with assays, a computational model to predict warhead reactivities could be useful for several aspects of the covalent inhibitor design process. Studies have shown correlations between covalent warhead reactivities and quantum mechanic (QM) properties that describe important aspects of the covalent reaction mechanism. However, the models from these studies are often linear regression equations and can have limitations associated with their usage. Applications of machine learning (ML) models to predict covalent warhead reactivities with QM descriptors are not extensively seen in the literature. This study uses QM descriptors, calculated at different levels of theory, to train ML models to predict reactivities of covalent acrylamide warheads. The QM/ML models are compared with linear regression models built upon the same QM descriptors and with ML models trained on structure-based features like Morgan fingerprints and RDKit descriptors. Experiments show that the QM/ML models outperform the linear regression models and the structure-based ML models, and literature test sets demonstrate the power of the QM/ML models to predict reactivities of unseen acrylamide warhead scaffolds. Ultimately, these QM/ML models are effective, computationally feasible tools that can expedite the design of new covalent inhibitors.

与非共价抑制相比,共价抑制具有许多优势,但必须仔细平衡共价弹头反应性,以保持效力,同时避免不必要的副作用。虽然弹头反应性通常通过化验来测量,但预测弹头反应性的计算模型对共价抑制剂设计过程的多个方面都很有用。研究表明,共价弹头反应活性与描述共价反应机理重要方面的量子力学(QM)特性之间存在相关性。然而,这些研究中的模型通常是线性回归方程,在使用时可能会受到限制。使用 QM 描述子预测共价弹头反应性的机器学习(ML)模型的应用在文献中并不多见。本研究使用按不同理论水平计算的 QM 描述符来训练 ML 模型,以预测共价丙烯酰胺弹头的反应性。QM/ML模型与基于相同QM描述符建立的线性回归模型以及基于摩根指纹和RDKit描述符等基于结构特征训练的ML模型进行了比较。实验表明,QM/ML 模型优于线性回归模型和基于结构的 ML 模型,文献测试集证明了 QM/ML 模型预测未见丙烯酰胺弹头支架反应性的能力。最终,这些 QM/ML 模型是有效的、计算上可行的工具,可以加快新共价抑制剂的设计。
{"title":"Reactivities of acrylamide warheads toward cysteine targets: a QM/ML approach to covalent inhibitor design","authors":"Aaron D. Danilack, Callum J. Dickson, Cihan Soylu, Mike Fortunato, Stephane Rodde, Hagen Munkler, Viktor Hornak, Jose S. Duca","doi":"10.1007/s10822-024-00560-6","DOIUrl":"https://doi.org/10.1007/s10822-024-00560-6","url":null,"abstract":"<p>Covalent inhibition offers many advantages over non-covalent inhibition, but covalent warhead reactivity must be carefully balanced to maintain potency while avoiding unwanted side effects. While warhead reactivities are commonly measured with assays, a computational model to predict warhead reactivities could be useful for several aspects of the covalent inhibitor design process. Studies have shown correlations between covalent warhead reactivities and quantum mechanic (QM) properties that describe important aspects of the covalent reaction mechanism. However, the models from these studies are often linear regression equations and can have limitations associated with their usage. Applications of machine learning (ML) models to predict covalent warhead reactivities with QM descriptors are not extensively seen in the literature. This study uses QM descriptors, calculated at different levels of theory, to train ML models to predict reactivities of covalent acrylamide warheads. The QM/ML models are compared with linear regression models built upon the same QM descriptors and with ML models trained on structure-based features like Morgan fingerprints and RDKit descriptors. Experiments show that the QM/ML models outperform the linear regression models and the structure-based ML models, and literature test sets demonstrate the power of the QM/ML models to predict reactivities of unseen acrylamide warhead scaffolds. Ultimately, these QM/ML models are effective, computationally feasible tools that can expedite the design of new covalent inhibitors.</p>","PeriodicalId":621,"journal":{"name":"Journal of Computer-Aided Molecular Design","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140836933","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
De novo drug design as GPT language modeling: large chemistry models with supervised and reinforcement learning 作为 GPT 语言建模的新药设计:采用监督和强化学习的大型化学模型
IF 3.5 3区 生物学 Q1 Chemistry Pub Date : 2024-04-22 DOI: 10.1007/s10822-024-00559-z
Gavin Ye
{"title":"De novo drug design as GPT language modeling: large chemistry models with supervised and reinforcement learning","authors":"Gavin Ye","doi":"10.1007/s10822-024-00559-z","DOIUrl":"https://doi.org/10.1007/s10822-024-00559-z","url":null,"abstract":"","PeriodicalId":621,"journal":{"name":"Journal of Computer-Aided Molecular Design","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-04-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140673020","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
From UK-2A to florylpicoxamid: Active learning to identify a mimic of a macrocyclic natural product 从UK-2A到氟啶虫酰胺:主动学习识别大环天然产物的模拟物
IF 3.5 3区 生物学 Q1 Chemistry Pub Date : 2024-04-17 DOI: 10.1007/s10822-024-00555-3
Ann E. Cleves, Ajay N. Jain, David A. Demeter, Zachary A. Buchan, Jeremy Wilmot, Erin N. Hancock

Scaffold replacement as part of an optimization process that requires maintenance of potency, desirable biodistribution, metabolic stability, and considerations of synthesis at very large scale is a complex challenge. Here, we consider a set of over 1000 time-stamped compounds, beginning with a macrocyclic natural-product lead and ending with a broad-spectrum crop anti-fungal. We demonstrate the application of the QuanSA 3D-QSAR method employing an active learning procedure that combines two types of molecular selection. The first identifies compounds predicted to be most active of those most likely to be well-covered by the model. The second identifies compounds predicted to be most informative based on exhibiting low predicted activity but showing high 3D similarity to a highly active nearest-neighbor training molecule. Beginning with just 100 compounds, using a deterministic and automatic procedure, five rounds of 20-compound selection and model refinement identifies the binding metabolic form of florylpicoxamid. We show how iterative refinement broadens the domain of applicability of the successive models while also enhancing predictive accuracy. We also demonstrate how a simple method requiring very sparse data can be used to generate relevant ideas for synthetic candidates.

作为优化过程的一部分,支架置换要求维持药效、理想的生物分布、代谢稳定性,并考虑大规模合成,这是一项复杂的挑战。在这里,我们考虑了一组超过 1000 个有时间戳的化合物,从一个大环天然产物先导化合物开始,到一个广谱作物抗真菌药物。我们展示了 QuanSA 3D-QSAR 方法的应用,该方法采用了一种结合两种分子选择类型的主动学习程序。第一种是在最有可能被模型很好覆盖的化合物中识别出最有活性的化合物。第二种方法是根据预测活性较低,但与高活性近邻训练分子的三维相似性较高的情况,确定预测信息量最大的化合物。从仅有的 100 个化合物开始,使用确定性的自动程序,经过五轮 20 个化合物的筛选和模型完善,确定了氟啶虫酰胺的结合代谢形式。我们展示了迭代改进如何拓宽连续模型的适用范围,同时提高预测准确性。我们还展示了如何利用一种需要非常稀少数据的简单方法来产生合成候选化合物的相关想法。
{"title":"From UK-2A to florylpicoxamid: Active learning to identify a mimic of a macrocyclic natural product","authors":"Ann E. Cleves, Ajay N. Jain, David A. Demeter, Zachary A. Buchan, Jeremy Wilmot, Erin N. Hancock","doi":"10.1007/s10822-024-00555-3","DOIUrl":"https://doi.org/10.1007/s10822-024-00555-3","url":null,"abstract":"<p>Scaffold replacement as part of an optimization process that requires maintenance of potency, desirable biodistribution, metabolic stability, and considerations of synthesis at very large scale is a complex challenge. Here, we consider a set of over 1000 time-stamped compounds, beginning with a macrocyclic natural-product lead and ending with a broad-spectrum crop anti-fungal. We demonstrate the application of the QuanSA 3D-QSAR method employing an active learning procedure that combines two types of molecular selection. The first identifies compounds predicted to be most active of those most likely to be well-covered by the model. The second identifies compounds predicted to be most <i>informative</i> based on exhibiting low predicted activity but showing high 3D similarity to a highly active nearest-neighbor training molecule. Beginning with just 100 compounds, using a deterministic and automatic procedure, five rounds of 20-compound selection and model refinement identifies the binding metabolic form of florylpicoxamid. We show how iterative refinement broadens the domain of applicability of the successive models while also enhancing predictive accuracy. We also demonstrate how a simple method requiring very sparse data can be used to generate relevant ideas for synthetic candidates.</p>","PeriodicalId":621,"journal":{"name":"Journal of Computer-Aided Molecular Design","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-04-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140614475","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
On the relevance of query definition in the performance of 3D ligand-based virtual screening 基于三维配体的虚拟筛选性能中查询定义的相关性
IF 3.5 3区 生物学 Q1 Chemistry Pub Date : 2024-04-04 DOI: 10.1007/s10822-024-00561-5
Javier Vázquez, Ricardo García, Paula Llinares, F. Javier Luque, Enric Herrero

Ligand-based virtual screening (LBVS) methods are widely used to explore the vast chemical space in the search of novel compounds resorting to a variety of properties encoded in 1D, 2D or 3D descriptors. The success of 3D-LBVS is affected by the overlay of molecular pairs, thus making selection of the template compound, search of accessible conformational space and choice of the query conformation to be potential factors that modulate the successful retrieval of actives. This study examines the impact of adopting different choices for the query conformation of the template, paying also attention to the influence exerted by the structural similarity between templates and actives. The analysis is performed using PharmScreen, a 3D LBVS tool that relies on similarity measurements of the hydrophobic/philic pattern of molecules, and Phase Shape, which is based on the alignment of atom triplets followed by refinement of the volume overlap. The study is performed for the original DUD-E+ database and a Morgan Fingerprint filtered version (denoted DUD-E+-Diverse; available in https://github.com/Pharmacelera/Query-models-to-3DLBVS), which was prepared to minimize the 2D resemblance between template and actives. Although in most cases the query conformation exhibits a mild influence on the overall performance, a critical analysis is made to disclose factors, such as the content of structural features between template and actives and the induction of conformational strain in the template, that underlie the drastic impact of the query definition in the recovery of actives for certain targets. The findings of this research also provide valuable guidance for assisting the selection of the query definition in 3D LBVS campaigns.

Graphical Abstract

基于配体的虚拟筛选(LBVS)方法被广泛用于探索广阔的化学空间,利用一维、二维或三维描述符编码的各种特性寻找新型化合物。3D-LBVS 的成功与否受分子对叠加的影响,因此模板化合物的选择、可访问构象空间的搜索和查询构象的选择是影响成功检索活性物质的潜在因素。本研究探讨了模板查询构象的不同选择所产生的影响,同时还关注了模板与活性物质之间结构相似性所产生的影响。分析使用 PharmScreen 和 Phase Shape 进行,前者是一种三维 LBVS 工具,依赖于分子疏水/亲水模式的相似性测量;后者则基于原子三元组的排列,然后对体积重叠进行细化。该研究针对原始 DUD-E+ 数据库和摩根指纹过滤版本(DUD-E+-Diverse,可在 https://github.com/Pharmacelera/Query-models-to-3DLBVS 上查阅)进行,后者是为了尽量减少模板和活性物质之间的二维相似性而准备的。虽然在大多数情况下,查询构象对总体性能的影响较小,但通过批判性分析揭示了一些因素,如模板与活性物之间的结构特征内容以及模板中构象应变的诱导,这些因素是查询定义对某些靶标的活性物恢复产生巨大影响的原因。这项研究的结果还为在三维 LBVS 活动中协助选择查询定义提供了有价值的指导。
{"title":"On the relevance of query definition in the performance of 3D ligand-based virtual screening","authors":"Javier Vázquez, Ricardo García, Paula Llinares, F. Javier Luque, Enric Herrero","doi":"10.1007/s10822-024-00561-5","DOIUrl":"https://doi.org/10.1007/s10822-024-00561-5","url":null,"abstract":"<p>Ligand-based virtual screening (LBVS) methods are widely used to explore the vast chemical space in the search of novel compounds resorting to a variety of properties encoded in 1D, 2D or 3D descriptors. The success of 3D-LBVS is affected by the overlay of molecular pairs, thus making selection of the template compound, search of accessible conformational space and choice of the query conformation to be potential factors that modulate the successful retrieval of actives. This study examines the impact of adopting different choices for the query conformation of the template, paying also attention to the influence exerted by the structural similarity between templates and actives. The analysis is performed using PharmScreen, a 3D LBVS tool that relies on similarity measurements of the hydrophobic/philic pattern of molecules, and Phase Shape, which is based on the alignment of atom triplets followed by refinement of the volume overlap. The study is performed for the original DUD-E<sup>+</sup> database and a Morgan Fingerprint filtered version (denoted DUD-E<sup>+</sup>-Diverse; available in https://github.com/Pharmacelera/Query-models-to-3DLBVS), which was prepared to minimize the 2D resemblance between template and actives. Although in most cases the query conformation exhibits a mild influence on the overall performance, a critical analysis is made to disclose factors, such as the content of structural features between template and actives and the induction of conformational strain in the template, that underlie the drastic impact of the query definition in the recovery of actives for certain targets. The findings of this research also provide valuable guidance for assisting the selection of the query definition in 3D LBVS campaigns.</p><h3 data-test=\"abstract-sub-heading\">Graphical Abstract</h3>","PeriodicalId":621,"journal":{"name":"Journal of Computer-Aided Molecular Design","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-04-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140568082","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Computational peptide discovery with a genetic programming approach 利用遗传编程方法计算肽的发现
IF 3.5 3区 生物学 Q1 Chemistry Pub Date : 2024-04-03 DOI: 10.1007/s10822-024-00558-0
Nicolas Scalzitti, Iliya Miralavy, David E. Korenchan, Christian T. Farrar, Assaf A. Gilad, Wolfgang Banzhaf

The development of peptides for therapeutic targets or biomarkers for disease diagnosis is a challenging task in protein engineering. Current approaches are tedious, often time-consuming and require complex laboratory data due to the vast search spaces that need to be considered. In silico methods can accelerate research and substantially reduce costs. Evolutionary algorithms are a promising approach for exploring large search spaces and can facilitate the discovery of new peptides. This study presents the development and use of a new variant of the genetic-programming-based POET algorithm, called POET(_{Regex}), where individuals are represented by a list of regular expressions. This algorithm was trained on a small curated dataset and employed to generate new peptides improving the sensitivity of peptides in magnetic resonance imaging with chemical exchange saturation transfer (CEST). The resulting model achieves a performance gain of 20% over the initial POET models and is able to predict a candidate peptide with a 58% performance increase compared to the gold-standard peptide. By combining the power of genetic programming with the flexibility of regular expressions, new peptide targets were identified that improve the sensitivity of detection by CEST. This approach provides a promising research direction for the efficient identification of peptides with therapeutic or diagnostic potential.

开发用于治疗靶点或疾病诊断的生物标志物的多肽是蛋白质工程中一项具有挑战性的任务。由于需要考虑巨大的搜索空间,目前的方法繁琐、耗时且需要复杂的实验室数据。硅学方法可以加快研究速度并大幅降低成本。进化算法是一种探索大型搜索空间的有前途的方法,可促进新肽的发现。本研究介绍了基于遗传编程的 POET 算法的新变体 POET(_{Regex})的开发和使用,其中个体由正则表达式列表表示。该算法是在一个小型策划数据集上训练的,并用于生成新的肽,以提高肽在化学交换饱和转移(CEST)磁共振成像中的灵敏度。与最初的 POET 模型相比,生成的模型性能提高了 20%,与黄金标准肽相比,预测候选肽的性能提高了 58%。通过将遗传编程的强大功能与正则表达式的灵活性相结合,确定了新的多肽靶标,提高了 CEST 检测的灵敏度。这种方法为高效鉴定具有治疗或诊断潜力的多肽提供了一个前景广阔的研究方向。
{"title":"Computational peptide discovery with a genetic programming approach","authors":"Nicolas Scalzitti, Iliya Miralavy, David E. Korenchan, Christian T. Farrar, Assaf A. Gilad, Wolfgang Banzhaf","doi":"10.1007/s10822-024-00558-0","DOIUrl":"https://doi.org/10.1007/s10822-024-00558-0","url":null,"abstract":"<p>The development of peptides for therapeutic targets or biomarkers for disease diagnosis is a challenging task in protein engineering. Current approaches are tedious, often time-consuming and require complex laboratory data due to the vast search spaces that need to be considered. <i>In silico</i> methods can accelerate research and substantially reduce costs. Evolutionary algorithms are a promising approach for exploring large search spaces and can facilitate the discovery of new peptides. This study presents the development and use of a new variant of the genetic-programming-based POET algorithm, called POET<span>(_{Regex})</span>, where individuals are represented by a list of regular expressions. This algorithm was trained on a small curated dataset and employed to generate new peptides improving the sensitivity of peptides in magnetic resonance imaging with chemical exchange saturation transfer (CEST). The resulting model achieves a performance gain of 20% over the initial POET models and is able to predict a candidate peptide with a 58% performance increase compared to the gold-standard peptide. By combining the power of genetic programming with the flexibility of regular expressions, new peptide targets were identified that improve the sensitivity of detection by CEST. This approach provides a promising research direction for the efficient identification of peptides with therapeutic or diagnostic potential.</p>","PeriodicalId":621,"journal":{"name":"Journal of Computer-Aided Molecular Design","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-04-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140568079","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identifying and characterising promising small molecule inhibitors of kinesin spindle protein using ligand-based virtual screening, molecular docking, molecular dynamics and MM‑GBSA calculations. 利用基于配体的虚拟筛选、分子对接、分子动力学和 MM-GBSA 计算,确定并表征有前途的驱动蛋白纺锤体小分子抑制剂。
IF 3.5 3区 生物学 Q1 Chemistry Pub Date : 2024-04-01 DOI: 10.1007/s10822-024-00553-5
Samia A Elseginy

The kinesin spindle protein (Eg5) is a mitotic protein that plays an essential role in the formation of the bipolar spindles during the mitotic phase. Eg5 protein controls the segregation of the chromosomes in mitosis which renders it a vital target for cancer treatment. In this study our approach to identifying novel scaffold for Eg5 inhibitors is based on targeting the novel allosteric pocket (α4/α6/L11). Extensive computational techniques were applied using ligand-based virtual screening and molecular docking by two approaches, MOE and AutoDock, to screen a library of commercial compounds. We identified compound 8-(3-(1H-imidazol-1-ylpropylamino)-3-methyl-7-((naphthalen-3-yl)methyl)-1H-purine-2, 6 (3H,7H)-dione (compound 5) as a novel scaffold for Eg5 inhibitors. This compound inhibited cancer cell Eg5 ATPase at 2.37 ± 0.15 µM. The molecular dynamics simulations revealed that the identified compound formed stable interactions in the allosteric pocket (α4/α6/L11) of the receptor, indicating its potential as a novel Eg5 inhibitor.

驱动蛋白纺锤体蛋白(Eg5)是一种有丝分裂蛋白,在有丝分裂阶段双极纺锤体的形成过程中起着至关重要的作用。Eg5 蛋白控制着有丝分裂过程中染色体的分离,因此成为癌症治疗的一个重要靶点。在这项研究中,我们以新型异构口袋(α4/α6/L11)为目标,为 Eg5 抑制剂寻找新型支架。我们采用配体虚拟筛选和分子对接(MOE 和 AutoDock)两种方法,对商业化合物库进行了广泛的计算技术筛选。我们发现化合物 8-(3-(1H-咪唑-1-基丙基氨基)-3-甲基-7-((萘-3-基)甲基)-1H-嘌呤-2, 6 (3H, 7H)-二酮(化合物 5)是 Eg5 抑制剂的新型支架。该化合物对癌细胞 Eg5 ATPase 的抑制作用为 2.37 ± 0.15 µM。分子动力学模拟显示,所发现的化合物在受体的异构口袋(α4/α6/L11)中形成了稳定的相互作用,表明其具有作为新型 Eg5 抑制剂的潜力。
{"title":"Identifying and characterising promising small molecule inhibitors of kinesin spindle protein using ligand-based virtual screening, molecular docking, molecular dynamics and MM‑GBSA calculations.","authors":"Samia A Elseginy","doi":"10.1007/s10822-024-00553-5","DOIUrl":"10.1007/s10822-024-00553-5","url":null,"abstract":"<p><p>The kinesin spindle protein (Eg5) is a mitotic protein that plays an essential role in the formation of the bipolar spindles during the mitotic phase. Eg5 protein controls the segregation of the chromosomes in mitosis which renders it a vital target for cancer treatment. In this study our approach to identifying novel scaffold for Eg5 inhibitors is based on targeting the novel allosteric pocket (α4/α6/L11). Extensive computational techniques were applied using ligand-based virtual screening and molecular docking by two approaches, MOE and AutoDock, to screen a library of commercial compounds. We identified compound 8-(3-(1H-imidazol-1-ylpropylamino)-3-methyl-7-((naphthalen-3-yl)methyl)-1H-purine-2, 6 (3H,7H)-dione (compound 5) as a novel scaffold for Eg5 inhibitors. This compound inhibited cancer cell Eg5 ATPase at 2.37 ± 0.15 µM. The molecular dynamics simulations revealed that the identified compound formed stable interactions in the allosteric pocket (α4/α6/L11) of the receptor, indicating its potential as a novel Eg5 inhibitor.</p>","PeriodicalId":621,"journal":{"name":"Journal of Computer-Aided Molecular Design","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10982093/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140331462","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Benchmarking ANI potentials as a rescoring function and screening FDA drugs for SARS-CoV-2 Mpro. 将 ANI 电位作为重构函数的基准,并筛选用于 SARS-CoV-2 Mpro 的 FDA 药物。
IF 3.5 3区 生物学 Q1 Chemistry Pub Date : 2024-03-27 DOI: 10.1007/s10822-024-00554-4
Irem N Zengin, M Serdar Koca, Omer Tayfuroglu, Muslum Yildiz, Abdulkadir Kocak

Here, we introduce the use of ANI-ML potentials as a rescoring function in the host-guest interaction in molecular docking. Our results show that the "docking power" of ANI potentials can compete with the current scoring functions at the same level of computational cost. Benchmarking studies on CASF-2016 dataset showed that ANI is ranked in the top 5 scoring functions among the other 34 tested. In particular, the ANI predicted interaction energies when used in conjunction with GOLD-PLP scoring function can boost the top ranked solution to be the closest to the x-ray structure. Rapid and accurate calculation of interaction energies between ligand and protein also enables screening of millions of drug candidates/docking poses. Using a unique protocol in which docking by GOLD-PLP, rescoring by ANI-ML potentials and extensive MD simulations along with end state free energy methods are combined, we have screened FDA approved drugs against the SARS-CoV-2 main protease (Mpro). The top six drug molecules suggested by the consensus of these free energy methods have already been in clinical trials or proposed as potential drug molecules in previous theoretical and experimental studies, approving the validity and the power of accuracy in our screening method.

在此,我们介绍在分子对接中使用 ANI-ML 电位作为主客体相互作用的重新评分函数。我们的研究结果表明,在计算成本相同的情况下,ANI 电位的 "对接能力 "可以与当前的评分函数相媲美。在 CASF-2016 数据集上进行的基准研究表明,在其他 34 种测试的评分函数中,ANI 位列前 5。特别是,ANI预测的相互作用能与GOLD-PLP评分函数结合使用时,能使排名第一的解决方案与X射线结构最接近。快速准确地计算配体与蛋白质之间的相互作用能还能筛选数百万个候选药物/对接方案。我们采用了一种独特的方案,将 GOLD-PLP 的对接、ANI-ML 电位的重构、大量 MD 模拟以及终态自由能方法结合在一起,针对 SARS-CoV-2 主要蛋白酶(Mpro)筛选出了经 FDA 批准的药物。这些自由能方法一致推荐的前六种药物分子已经进入临床试验阶段,或在以前的理论和实验研究中被推荐为潜在的药物分子,这证明了我们筛选方法的有效性和准确性。
{"title":"Benchmarking ANI potentials as a rescoring function and screening FDA drugs for SARS-CoV-2 M<sup>pro</sup>.","authors":"Irem N Zengin, M Serdar Koca, Omer Tayfuroglu, Muslum Yildiz, Abdulkadir Kocak","doi":"10.1007/s10822-024-00554-4","DOIUrl":"10.1007/s10822-024-00554-4","url":null,"abstract":"<p><p>Here, we introduce the use of ANI-ML potentials as a rescoring function in the host-guest interaction in molecular docking. Our results show that the \"docking power\" of ANI potentials can compete with the current scoring functions at the same level of computational cost. Benchmarking studies on CASF-2016 dataset showed that ANI is ranked in the top 5 scoring functions among the other 34 tested. In particular, the ANI predicted interaction energies when used in conjunction with GOLD-PLP scoring function can boost the top ranked solution to be the closest to the x-ray structure. Rapid and accurate calculation of interaction energies between ligand and protein also enables screening of millions of drug candidates/docking poses. Using a unique protocol in which docking by GOLD-PLP, rescoring by ANI-ML potentials and extensive MD simulations along with end state free energy methods are combined, we have screened FDA approved drugs against the SARS-CoV-2 main protease (M<sup>pro</sup>). The top six drug molecules suggested by the consensus of these free energy methods have already been in clinical trials or proposed as potential drug molecules in previous theoretical and experimental studies, approving the validity and the power of accuracy in our screening method.</p>","PeriodicalId":621,"journal":{"name":"Journal of Computer-Aided Molecular Design","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-03-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10965596/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140292429","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The AI-driven Drug Design (AIDD) platform: an interactive multi-parameter optimization system integrating molecular evolution with physiologically based pharmacokinetic simulations. 人工智能驱动的药物设计(AIDD)平台:一个交互式多参数优化系统,将分子进化与基于生理学的药代动力学模拟融为一体。
IF 3.5 3区 生物学 Q1 Chemistry Pub Date : 2024-03-19 DOI: 10.1007/s10822-024-00552-6
Jeremy Jones, Robert D Clark, Michael S Lawless, David W Miller, Marvin Waldman

Computer-aided drug design has advanced rapidly in recent years, and multiple instances of in silico designed molecules advancing to the clinic have demonstrated the contribution of this field to medicine. Properly designed and implemented platforms can drastically reduce drug development timelines and costs. While such efforts were initially focused primarily on target affinity/activity, it is now appreciated that other parameters are equally important in the successful development of a drug and its progression to the clinic, including pharmacokinetic properties as well as absorption, distribution, metabolic, excretion and toxicological (ADMET) properties. In the last decade, several programs have been developed that incorporate these properties into the drug design and optimization process and to varying degrees, allowing for multi-parameter optimization. Here, we introduce the Artificial Intelligence-driven Drug Design (AIDD) platform, which automates the drug design process by integrating high-throughput physiologically-based pharmacokinetic simulations (powered by GastroPlus) and ADMET predictions (powered by ADMET Predictor) with an advanced evolutionary algorithm that is quite different than current generative models. AIDD uses these and other estimates in iteratively performing multi-objective optimizations to produce novel molecules that are active and lead-like. Here we describe the AIDD workflow and details of the methodologies involved therein. We use a dataset of triazolopyrimidine inhibitors of the dihydroorotate dehydrogenase from Plasmodium falciparum to illustrate how AIDD generates novel sets of molecules.

近年来,计算机辅助药物设计发展迅猛,硅学设计分子进入临床的多个实例证明了这一领域对医学的贡献。设计和实施得当的平台可以大大缩短药物开发的时间和成本。虽然这些工作最初主要集中在靶点亲和力/活性上,但现在人们认识到,其他参数对药物的成功开发和进入临床同样重要,包括药代动力学特性以及吸收、分布、代谢、排泄和毒理学(ADMET)特性。在过去的十年中,已经有多个程序将这些特性纳入了药物设计和优化过程,并在不同程度上实现了多参数优化。在此,我们介绍人工智能驱动药物设计(AIDD)平台,该平台通过将基于生理的高通量药代动力学模拟(由 GastroPlus 提供支持)和 ADMET 预测(由 ADMET Predictor 提供支持)与先进的进化算法相结合,实现了药物设计过程的自动化。AIDD 在迭代执行多目标优化时使用这些和其他估计值,以产生具有活性和先导性的新分子。在此,我们将介绍 AIDD 的工作流程以及相关方法的细节。我们使用恶性疟原虫二氢烟酸脱氢酶的三唑并嘧啶抑制剂数据集来说明 AIDD 如何生成新分子集。
{"title":"The AI-driven Drug Design (AIDD) platform: an interactive multi-parameter optimization system integrating molecular evolution with physiologically based pharmacokinetic simulations.","authors":"Jeremy Jones, Robert D Clark, Michael S Lawless, David W Miller, Marvin Waldman","doi":"10.1007/s10822-024-00552-6","DOIUrl":"10.1007/s10822-024-00552-6","url":null,"abstract":"<p><p>Computer-aided drug design has advanced rapidly in recent years, and multiple instances of in silico designed molecules advancing to the clinic have demonstrated the contribution of this field to medicine. Properly designed and implemented platforms can drastically reduce drug development timelines and costs. While such efforts were initially focused primarily on target affinity/activity, it is now appreciated that other parameters are equally important in the successful development of a drug and its progression to the clinic, including pharmacokinetic properties as well as absorption, distribution, metabolic, excretion and toxicological (ADMET) properties. In the last decade, several programs have been developed that incorporate these properties into the drug design and optimization process and to varying degrees, allowing for multi-parameter optimization. Here, we introduce the Artificial Intelligence-driven Drug Design (AIDD) platform, which automates the drug design process by integrating high-throughput physiologically-based pharmacokinetic simulations (powered by GastroPlus) and ADMET predictions (powered by ADMET Predictor) with an advanced evolutionary algorithm that is quite different than current generative models. AIDD uses these and other estimates in iteratively performing multi-objective optimizations to produce novel molecules that are active and lead-like. Here we describe the AIDD workflow and details of the methodologies involved therein. We use a dataset of triazolopyrimidine inhibitors of the dihydroorotate dehydrogenase from Plasmodium falciparum to illustrate how AIDD generates novel sets of molecules.</p>","PeriodicalId":621,"journal":{"name":"Journal of Computer-Aided Molecular Design","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-03-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140157358","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
SpaceGrow: efficient shape-based virtual screening of billion-sized combinatorial fragment spaces. SpaceGrow:基于形状的亿万级组合片段空间高效虚拟筛选。
IF 3.5 3区 生物学 Q1 Chemistry Pub Date : 2024-03-17 DOI: 10.1007/s10822-024-00551-7
Sophia M N Hönig, Florian Flachsenberg, Christiane Ehrt, Alexander Neumann, Robert Schmidt, Christian Lemmen, Matthias Rarey

The growing size of make-on-demand chemical libraries is posing new challenges to cheminformatics. These ultra-large chemical libraries became too large for exhaustive enumeration. Using a combinatorial approach instead, the resource requirement scales approximately with the number of synthons instead of the number of molecules. This gives access to billions or trillions of compounds as so-called chemical spaces with moderate hardware and in a reasonable time frame. While extremely performant ligand-based 2D methods exist in this context, 3D methods still largely rely on exhaustive enumeration and therefore fail to apply. Here, we present SpaceGrow: a novel shape-based 3D approach for ligand-based virtual screening of billions of compounds within hours on a single CPU. Compared to a conventional superposition tool, SpaceGrow shows comparable pose reproduction capacity based on RMSD and superior ranking performance while being orders of magnitude faster. Result assessment of two differently sized subsets of the eXplore space reveals a higher probability of finding superior results in larger spaces highlighting the potential of searching in ultra-large spaces. Furthermore, the application of SpaceGrow in a drug discovery workflow was investigated in four examples involving G protein-coupled receptors (GPCRs) with the aim to identify compounds with similar binding capabilities and molecular novelty.

按需制造化学库的规模不断扩大,给化学信息学带来了新的挑战。这些超大型化学库变得过于庞大,无法进行详尽的枚举。如果采用组合方法,资源需求将与合成子的数量而不是分子的数量成近似比例。这样,就能以适度的硬件和合理的时间范围访问数十亿或数万亿化合物的所谓化学空间。在这种情况下,虽然基于配体的二维方法性能极佳,但三维方法在很大程度上仍依赖于穷举法,因此并不适用。在此,我们介绍 SpaceGrow:一种基于形状的新型三维方法,可在数小时内利用单个 CPU 对数十亿化合物进行基于配体的虚拟筛选。与传统的叠加工具相比,SpaceGrow 基于 RMSD 显示出相当的姿态再现能力和卓越的排序性能,同时速度快了几个数量级。对 eXplore 空间的两个不同大小的子集进行的结果评估显示,在较大的空间中找到卓越结果的概率更高,这突出表明了在超大空间中搜索的潜力。此外,在涉及 G 蛋白偶联受体(GPCR)的四个实例中,研究了 SpaceGrow 在药物发现工作流程中的应用,目的是找出具有相似结合能力和分子新颖性的化合物。
{"title":"SpaceGrow: efficient shape-based virtual screening of billion-sized combinatorial fragment spaces.","authors":"Sophia M N Hönig, Florian Flachsenberg, Christiane Ehrt, Alexander Neumann, Robert Schmidt, Christian Lemmen, Matthias Rarey","doi":"10.1007/s10822-024-00551-7","DOIUrl":"10.1007/s10822-024-00551-7","url":null,"abstract":"<p><p>The growing size of make-on-demand chemical libraries is posing new challenges to cheminformatics. These ultra-large chemical libraries became too large for exhaustive enumeration. Using a combinatorial approach instead, the resource requirement scales approximately with the number of synthons instead of the number of molecules. This gives access to billions or trillions of compounds as so-called chemical spaces with moderate hardware and in a reasonable time frame. While extremely performant ligand-based 2D methods exist in this context, 3D methods still largely rely on exhaustive enumeration and therefore fail to apply. Here, we present SpaceGrow: a novel shape-based 3D approach for ligand-based virtual screening of billions of compounds within hours on a single CPU. Compared to a conventional superposition tool, SpaceGrow shows comparable pose reproduction capacity based on RMSD and superior ranking performance while being orders of magnitude faster. Result assessment of two differently sized subsets of the eXplore space reveals a higher probability of finding superior results in larger spaces highlighting the potential of searching in ultra-large spaces. Furthermore, the application of SpaceGrow in a drug discovery workflow was investigated in four examples involving G protein-coupled receptors (GPCRs) with the aim to identify compounds with similar binding capabilities and molecular novelty.</p>","PeriodicalId":621,"journal":{"name":"Journal of Computer-Aided Molecular Design","volume":null,"pages":null},"PeriodicalIF":3.5,"publicationDate":"2024-03-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10944417/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140139700","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Journal of Computer-Aided Molecular Design
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
×
提示
确定
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