QEPPI

QEPPI
  • 文章类型: Journal Article
    蛋白质-蛋白质相互作用(PPIs)与各种疾病相关;因此,它们是药物发现的重要目标。然而,PPI靶向药物的物理化学经验特性与常规小分子口服药物不同,遵守“五规则(RO5)”。因此,使用常规方法开发PPI靶向药物,例如分子生成模型,具有挑战性。在这项研究中,我们提出了一种基于深度强化学习的分子生成模型,该模型专门用于生产PPI抑制剂。通过引入可以代表PPI抑制剂性质的评分函数,我们成功地产生了潜在的PPI抑制剂化合物.这些新构建的虚拟化合物具有PPI抑制剂所需的特性,它们与市售的PPI库相似。虚拟化合物可作为虚拟库免费获得。
    Protein-protein interactions (PPIs) are associated with various diseases; hence, they are important targets in drug discovery. However, the physicochemical empirical properties of PPI-targeted drugs are distinct from those of conventional small molecule oral pharmaceuticals, which adhere to the \"rule of five (RO5)\". Therefore, developing PPI-targeted drugs using conventional methods, such as molecular generation models, is challenging. In this study, we propose a molecular generation model based on deep reinforcement learning that is specialized for the production of PPI inhibitors. By introducing a scoring function that can represent the properties of PPI inhibitors, we successfully generated potential PPI inhibitor compounds. These newly constructed virtual compounds possess the desired properties for PPI inhibitors, and they show similarity to commercially available PPI libraries. The virtual compounds are freely available as a virtual library.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    设计/发现药物的过程涉及鉴定和设计具有所需特性并与给定疾病相关靶标良好结合的新分子。有效识别潜在候选药物的主要挑战之一是探索广阔的药物样化学空间,以找到具有所需物理化学性质和生物学特性的新型化学结构。此外,目前可用的分子库的化学空间仅占全部可能的药物样化学空间的一小部分.深层分子生成模型受到了广泛关注,并为分子的设计和发现提供了一种替代方法。为了有效地探索类似药物的空间,我们首先构建了药物样数据集,然后使用条件随机转换方法,以分子接入系统(MACCS)指纹为条件,进行了药物样分子的生成设计,并将其与以前发表的分子生成模型进行了比较.结果表明,深层分子生成模型探索了更广泛的类药物化学空间。生成的药物样分子与已知药物共享化学空间,通过定量估计药物相似度(QED)和定量估计蛋白质-蛋白质相互作用靶向药物相似度(QEPPI)相结合捕获的药物样空间可以覆盖更大的药物样空间。最后,我们展示了该模型在设计MDM2-p53蛋白-蛋白相互作用抑制剂中的潜在应用。我们的结果证明了深层分子生成模型在药物样化学空间和分子设计中的指导探索的潜在应用。
    The process of design/discovery of drugs involves the identification and design of novel molecules that have the desired properties and bind well to a given disease-relevant target. One of the main challenges to effectively identify potential drug candidates is to explore the vast drug-like chemical space to find novel chemical structures with desired physicochemical properties and biological characteristics. Moreover, the chemical space of currently available molecular libraries is only a small fraction of the total possible drug-like chemical space. Deep molecular generative models have received much attention and provide an alternative approach to the design and discovery of molecules. To efficiently explore the drug-like space, we first constructed the drug-like dataset and then performed the generative design of drug-like molecules using a Conditional Randomized Transformer approach with the molecular access system (MACCS) fingerprint as a condition and compared it with previously published molecular generative models. The results show that the deep molecular generative model explores the wider drug-like chemical space. The generated drug-like molecules share the chemical space with known drugs, and the drug-like space captured by the combination of quantitative estimation of drug-likeness (QED) and quantitative estimate of protein-protein interaction targeting drug-likeness (QEPPI) can cover a larger drug-like space. Finally, we show the potential application of the model in design of inhibitors of MDM2-p53 protein-protein interaction. Our results demonstrate the potential application of deep molecular generative models for guided exploration in drug-like chemical space and molecular design.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    我们构建了一个蛋白质-蛋白质相互作用(PPI)靶向的药物相似度数据集,并提出了一个深层的分子生成框架,以从种子化合物的特征中生成新的药物相似度分子。这个框架从已发表的分子生成模型中获得灵感,使用与PPI抑制剂相关的关键特征作为输入,并开发用于PPI抑制剂从头分子设计的深层分子生成模型。第一次,以PPI为目标的化合物的定量估计指数被应用于PPI靶向化合物从头设计的分子生成模型的评估。我们的结果估计产生的分子具有更好的PPI靶向药物相似性和药物相似性。此外,我们的模型还表现出与其他几种最先进的分子生成模型相当的性能。如化学空间分析所证明的,所产生的分子与iPPI-DB抑制剂共享化学空间。探索了PPI抑制剂的肽表征设计和基于配体的PPI抑制剂设计。最后,我们建议,该框架将是PPI靶向治疗的从头设计的重要一步.
    We construct a protein-protein interaction (PPI) targeted drug-likeness dataset and propose a deep molecular generative framework to generate novel drug-likeness molecules from the features of the seed compounds. This framework gains inspiration from published molecular generative models, uses the key features associated with PPI inhibitors as input and develops deep molecular generative models for de novo molecular design of PPI inhibitors. For the first time, quantitative estimation index for compounds targeting PPI was applied to the evaluation of the molecular generation model for de novo design of PPI-targeted compounds. Our results estimated that the generated molecules had better PPI-targeted drug-likeness and drug-likeness. Additionally, our model also exhibits comparable performance to other several state-of-the-art molecule generation models. The generated molecules share chemical space with iPPI-DB inhibitors as demonstrated by chemical space analysis. The peptide characterization-oriented design of PPI inhibitors and the ligand-based design of PPI inhibitors are explored. Finally, we recommend that this framework will be an important step forward for the de novo design of PPI-targeted therapeutics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    药物相似度定量可用于筛选候选药物。定量药物相似度(QED)通常用于评估定量药物功效,但不适用于筛选针对蛋白质-蛋白质相互作用(PPI)的化合物。最近引起了人们的注意。因此,我们开发了以PPI为目标的化合物的定量估计指数(QEPPI),专门用于PPI靶向化合物的早期筛选。QEPPI是针对PPI靶向药物的QED方法的扩展,该方法基于药物/化合物的可用信息对理化性质进行建模,特别是那些报告对PPI采取行动的人。FDA批准的iPPI-DB中的药物和化合物,其中包括PPI抑制剂和稳定剂,使用QEPPI进行评估。结果表明,QEPPI比QED更适合于PPI靶向化合物的早期筛查。QEPPI也被认为是“四规则”(RO4)的扩展概念,PPI抑制剂指数。我们使用F分数和其他指标评估了QEPPI和RO4对PPI目标化合物和FDA批准的药物数据集的歧视性表现。RO4和QEPPI的F评分分别为0.451和0.501。QEPPI显示出更好的性能,并能够量化早期PPI药物发现的药物相似度。因此,它可以用作初始过滤器来有效地筛选PPI靶向化合物。
    Drug-likeness quantification is useful for screening drug candidates. Quantitative estimates of drug-likeness (QED) are commonly used to assess quantitative drug efficacy but are not suitable for screening compounds targeting protein-protein interactions (PPIs), which have recently gained attention. Therefore, we developed a quantitative estimate index for compounds targeting PPIs (QEPPI), specifically for early-stage screening of PPI-targeting compounds. QEPPI is an extension of the QED method for PPI-targeting drugs that models physicochemical properties based on the information available for drugs/compounds, specifically those reported to act on PPIs. FDA-approved drugs and compounds in iPPI-DB, which comprise PPI inhibitors and stabilizers, were evaluated using QEPPI. The results showed that QEPPI is more suitable than QED for early screening of PPI-targeting compounds. QEPPI was also considered an extended concept of the \"Rule-of-Four\" (RO4), a PPI inhibitor index. We evaluated the discriminatory performance of QEPPI and RO4 for datasets of PPI-target compounds and FDA-approved drugs using F-score and other indices. The F-scores of RO4 and QEPPI were 0.451 and 0.501, respectively. QEPPI showed better performance and enabled quantification of drug-likeness for early-stage PPI drug discovery. Hence, it can be used as an initial filter to efficiently screen PPI-targeting compounds.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号