Lead optimization

销售线索优化
  • 文章类型: Journal Article
    药物发现和开发是一项费力且昂贵的工作。药物的成功不仅取决于良好的疗效,还取决于可接受的吸收,分布,新陈代谢,消除,和毒性(ADMET)特性。总的来说,多达50%的药物开发失败是由不良的ADMET概况造成的。作为多参数目标,由于巨大的化学空间和有限的人类专业知识,ADMET特性的优化极具挑战性。在这项研究中,一个叫做化学分子优化的免费平台,表示和翻译(ChemMORT)是为优化多个ADMET端点而不损失效力而开发的(https://cadd。nscc-tj.cn/deploy/chemmort/)。ChemMORT包含三个模块:简化分子输入线输入系统(SMILES)编码器,描述符解码器和分子优化器。SMILES编码器可以生成具有512维向量的分子表示,并且描述符解码器能够高精度地将上述表示转换为相应的分子结构。基于可逆分子表示和粒子群优化策略,分子优化器可用于有效优化不需要的ADMET特性,而不会损失生物活性,基本上完成了逆QSAR的设计。提供了聚(ADP-核糖)聚合酶1抑制剂的约束多目标优化,以探索ChemMORT的实用性。
    Drug discovery and development constitute a laborious and costly undertaking. The success of a drug hinges not only good efficacy but also acceptable absorption, distribution, metabolism, elimination, and toxicity (ADMET) properties. Overall, up to 50% of drug development failures have been contributed from undesirable ADMET profiles. As a multiple parameter objective, the optimization of the ADMET properties is extremely challenging owing to the vast chemical space and limited human expert knowledge. In this study, a freely available platform called Chemical Molecular Optimization, Representation and Translation (ChemMORT) is developed for the optimization of multiple ADMET endpoints without the loss of potency (https://cadd.nscc-tj.cn/deploy/chemmort/). ChemMORT contains three modules: Simplified Molecular Input Line Entry System (SMILES) Encoder, Descriptor Decoder and Molecular Optimizer. The SMILES Encoder can generate the molecular representation with a 512-dimensional vector, and the Descriptor Decoder is able to translate the above representation to the corresponding molecular structure with high accuracy. Based on reversible molecular representation and particle swarm optimization strategy, the Molecular Optimizer can be used to effectively optimize undesirable ADMET properties without the loss of bioactivity, which essentially accomplishes the design of inverse QSAR. The constrained multi-objective optimization of the poly (ADP-ribose) polymerase-1 inhibitor is provided as the case to explore the utility of ChemMORT.
    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
    受体-丝氨酸/苏氨酸蛋白激酶1(RIPK1)在炎症和细胞死亡中起关键调节因子的作用,并参与介导多种炎症或退行性疾病。已经开发了许多变构RIPK1抑制剂(RIPK1i),其中一些已经进入临床评估。最近,与RIPK1的变构口袋和ATP结合位点相互作用的选择性RIPK1i已经开始出现。这里,基于对已报道但机械上非典型的RIPK3i的重新发现,我们报告了一系列新的II型RIPK1i的合理开发。我们还描述了一种有效的结构引导引线优化,选择性,和口服生物利用度RIPK1i,62,其在急性或慢性炎性疾病的小鼠模型中表现出非凡的功效。总的来说,图62提供了用于在动物疾病模型中评估RIPK1的有用工具和用于进一步药物开发的有希望的线索。
    Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) functions as a key regulator in inflammation and cell death and is involved in mediating a variety of inflammatory or degenerative diseases. A number of allosteric RIPK1 inhibitors (RIPK1i) have been developed, and some of them have already advanced into clinical evaluation. Recently, selective RIPK1i that interact with both the allosteric pocket and the ATP-binding site of RIPK1 have started to emerge. Here, we report the rational development of a new series of type-II RIPK1i based on the rediscovery of a reported but mechanistically atypical RIPK3i. We also describe the structure-guided lead optimization of a potent, selective, and orally bioavailable RIPK1i, 62, which exhibits extraordinary efficacies in mouse models of acute or chronic inflammatory diseases. Collectively, 62 provides a useful tool for evaluating RIPK1 in animal disease models and a promising lead for further drug development.
    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
    常染色体显性多囊肾病(ADPKD)是慢性肾病最常见的单基因病因,也是终末期肾病的第四大病因,占需要肾脏替代疗法的流行病例的50%以上。迫切需要改善ADPKD的治疗。对ADPKD病理生理学的最新见解表明,囊肿细胞经历代谢变化,从而上调有氧糖酵解,而不是线粒体呼吸来产生能量,表面上助长其扩散的过程。目前的工作利用这种代谢破坏作为选择性靶向囊肿细胞凋亡的方法。这种小分子治疗策略利用11β-二氯,一种再利用的DNA损伤抗肿瘤剂,通过加剧线粒体氧化应激诱导细胞凋亡。这里,我们证明11β-二氯可有效延缓囊肿生长及其相关的炎症和纤维化事件,从而在围产期和成年ADPKD小鼠模型中保留肾功能。在这两种模型中,Pkd1纯合失活的囊肿细胞在用11β-二氯处理后显示出增强的氧化应激并发生凋亡。抗氧化剂维生素E的共同给药否定了体内11β-二氯的治疗益处,支持氧化应激是作用机制的关键组成部分的结论。作为临床前开发的入门,我们还合成并测试了一种不能直接烷基化DNA的11β-二氯衍生物,同时保留促氧化剂的特点。尽管如此,该衍生物在体内仍保持出色的抗囊性能,并成为开发的主要候选者。
    Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenic cause of chronic kidney disease and the fourth leading cause of end-stage kidney disease, accounting for over 50% of prevalent cases requiring renal replacement therapy. There is a pressing need for improved therapy for ADPKD. Recent insights into the pathophysiology of ADPKD revealed that cyst cells undergo metabolic changes that up-regulate aerobic glycolysis in lieu of mitochondrial respiration for energy production, a process that ostensibly fuels their increased proliferation. The present work leverages this metabolic disruption as a way to selectively target cyst cells for apoptosis. This small-molecule therapeutic strategy utilizes 11beta-dichloro, a repurposed DNA-damaging anti-tumor agent that induces apoptosis by exacerbating mitochondrial oxidative stress. Here, we demonstrate that 11beta-dichloro is effective in delaying cyst growth and its associated inflammatory and fibrotic events, thus preserving kidney function in perinatal and adult mouse models of ADPKD. In both models, the cyst cells with homozygous inactivation of Pkd1 show enhanced oxidative stress following treatment with 11beta-dichloro and undergo apoptosis. Co-administration of the antioxidant vitamin E negated the therapeutic benefit of 11beta-dichloro in vivo, supporting the conclusion that oxidative stress is a key component of the mechanism of action. As a preclinical development primer, we also synthesized and tested an 11beta-dichloro derivative that cannot directly alkylate DNA, while retaining pro-oxidant features. This derivative nonetheless maintains excellent anti-cystic properties in vivo and emerges as the lead candidate for development.
    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
    在过去30年中,农药使用的全球成本效益分析的特点是1990年至2007年期间显着增加,随后下降。这一观察可以归因于几个因素,包括,但不限于,害虫抗性,在行动模式或化学类别方面缺乏新颖性,和监管行动。由于当前和预计的全球人口增长,很明显,对食物的需求,因此,使用杀虫剂来提高产量将会增加。应对这些挑战和需求,同时通过日益严格的监管环境推广新的作物保护剂,需要开发和整合创新的基础设施,新的和可持续的分子的成本和时间有效的发现和发展。在过去的二十年中,人工智能(AI)和化学信息学的重大进展提高了研究科学家在发现生物活性分子方面的决策能力。AI和化学信息学驱动的分子发现提供了将实验从温室转移到虚拟环境的机会,在虚拟环境中可以快速研究数千到数十亿的分子,为铅的产生提供无偏见的假设,优化,以及化合物合成和测试的有效建议。迄今为止,与药物发现相比,公开可用的农业化学研究文献在很大程度上说明了这一点。在这次审查中,我们提供了作物保护发现管道的概述,以及传统的,化学信息学,和人工智能技术可以帮助解决农业化学发现的需求和挑战,以快速开发新型和更可持续的产品。
    The global cost-benefit analysis of pesticide use during the last 30 years has been characterized by a significant increase during the period from 1990 to 2007 followed by a decline. This observation can be attributed to several factors including, but not limited to, pest resistance, lack of novelty with respect to modes of action or classes of chemistry, and regulatory action. Due to current and projected increases of the global population, it is evident that the demand for food, and consequently, the usage of pesticides to improve yields will increase. Addressing these challenges and needs while promoting new crop protection agents through an increasingly stringent regulatory landscape requires the development and integration of infrastructures for innovative, cost- and time-effective discovery and development of novel and sustainable molecules. Significant advances in artificial intelligence (AI) and cheminformatics over the last two decades have improved the decision-making power of research scientists in the discovery of bioactive molecules. AI- and cheminformatics-driven molecule discovery offers the opportunity of moving experiments from the greenhouse to a virtual environment where thousands to billions of molecules can be investigated at a rapid pace, providing unbiased hypothesis for lead generation, optimization, and effective suggestions for compound synthesis and testing. To date, this is illustrated to a far lesser extent in the publicly available agrochemical research literature compared to drug discovery. In this review, we provide an overview of the crop protection discovery pipeline and how traditional, cheminformatics, and AI technologies can help to address the needs and challenges of agrochemical discovery towards rapidly developing novel and more sustainable products.
    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
    时间分割交叉验证被广泛认为是验证用于药物化学项目的预测模型的黄金标准。不幸的是,这种类型的数据在大型药物研究组织之外并不广泛可用。在这里,我们介绍了SIMPD(模拟药物化学项目数据)算法,将公共数据集拆分为训练集和测试集,以模仿在现实世界药物化学项目数据集中观察到的差异。SIMPD使用多目标遗传算法,其目标来自对诺华生物医学研究所内130多个铅优化项目中早期和晚期化合物之间差异的广泛分析。将SIMPD应用于现实世界数据集产生的训练/测试分割比随机或邻居分割等其他标准方法更准确地反映了时间分割所观察到的属性和机器学习性能的差异。我们将SIMPD算法应用于从ChEMBL提取的生物活性数据,并创建了99个公共数据集,可用于验证用于药物化学项目设置的机器学习模型。SIMPD代码和模拟数据集可在github.com/rinikerlab/molecular_time_series上获得开源/开放数据许可。
    Time-split cross-validation is broadly recognized as the gold standard for validating predictive models intended for use in medicinal chemistry projects. Unfortunately this type of data is not broadly available outside of large pharmaceutical research organizations. Here we introduce the SIMPD (simulated medicinal chemistry project data) algorithm to split public data sets into training and test sets that mimic the differences observed in real-world medicinal chemistry project data sets. SIMPD uses a multi-objective genetic algorithm with objectives derived from an extensive analysis of the differences between early and late compounds in more than 130 lead-optimization projects run within the Novartis Institutes for BioMedical Research. Applying SIMPD to the real-world data sets produced training/test splits which more accurately reflect the differences in properties and machine-learning performance observed for temporal splits than other standard approaches like random or neighbor splits. We applied the SIMPD algorithm to bioactivity data extracted from ChEMBL and created 99 public data sets which can be used for validating machine-learning models intended for use in the setting of a medicinal chemistry project. The SIMPD code and simulated data sets are available under open-source/open-data licenses at github.com/rinikerlab/molecular_time_series.
    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
    COVID-19大流行刺激了学术界和工业界的合作药物发现工作,目的是开发针对SARS-CoV-2的疗法和疫苗。在过去的三年中,已经批准并部署了几种新疗法。然而,由于病毒变异的迅速出现,其临床应用已显示出局限性。因此,具有高效和安全性的下一代SARS-CoV-2治疗剂的开发仍然是全球健康的高度优先事项。人们对“回归自然”改善人类健康的方法的认识日益提高,这促使人们对天然产品重新产生了兴趣,尤其是膳食多酚,作为治疗SARS-CoV-2患者的额外治疗策略,由于其良好的安全性,非凡的营养价值,促进健康的好处(包括潜在的抗病毒特性),负担能力,和可用性。在这里,我们描述了膳食多酚姜黄素的生物学特性和多效性分子机制,白藜芦醇,和棉酚作为在体外和体内研究中观察到的SARS-CoV-2及其变体的抑制剂。基于膳食多酚的优点和缺点,并获得最大的好处,几种策略,如纳米技术(例如,具有抗菌-抗病毒能力的姜黄素纳米纤维膜),铅优化(例如,姜黄素的甲基化类似物),联合疗法(例如,植物提取物和微量营养素的特定组合),和广谱活动(例如,棉酚广泛抑制冠状病毒)也被强调为促进抗SARS-CoV-2药物开发的积极因素,以支持有效的长期大流行管理和控制。
    The COVID-19 pandemic has stimulated collaborative drug discovery efforts in academia and the industry with the aim of developing therapies and vaccines that target SARS-CoV-2. Several novel therapies have been approved and deployed in the last three years. However, their clinical application has revealed limitations due to the rapid emergence of viral variants. Therefore, the development of next-generation SARS-CoV-2 therapeutic agents with a high potency and safety profile remains a high priority for global health. Increasing awareness of the \"back to nature\" approach for improving human health has prompted renewed interest in natural products, especially dietary polyphenols, as an additional therapeutic strategy to treat SARS-CoV-2 patients, owing to its good safety profile, exceptional nutritional value, health-promoting benefits (including potential antiviral properties), affordability, and availability. Herein, we describe the biological properties and pleiotropic molecular mechanisms of dietary polyphenols curcumin, resveratrol, and gossypol as inhibitors against SARS-CoV-2 and its variants as observed in in vitro and in vivo studies. Based on the advantages and disadvantages of dietary polyphenols and to obtain maximal benefits, several strategies such as nanotechnology (e.g., curcumin-incorporated nanofibrous membranes with antibacterial-antiviral ability), lead optimization (e.g., a methylated analog of curcumin), combination therapies (e.g., a specific combination of plant extracts and micronutrients), and broad-spectrum activities (e.g., gossypol broadly inhibits coronaviruses) have also been emphasized as positive factors in the facilitation of anti-SARS-CoV-2 drug development to support effective long-term pandemic management and control.
    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
    描述小分子CSF1R抑制剂在临床开发中对集落刺激因子-1受体(CSF1R)的基于结构的抑制作用,用于靶标识别和癌症治疗中的潜在前导优化,因为CSF1R是一种新型的预测生物标志物用于癌症的免疫治疗。
    化合物在分子操作环境中通过诱导拟合对接方案进行了计算机建模(MOE,妈妈.v.2015).CSF1R激酶的三维(3D)X射线结晶结构(蛋白质数据库,ID4R7H)从结构生物信息学研究实验室(RSCB)蛋白质数据库获得。依地替尼的3D构象,DCC-3014,ARRY-382,BLZ-945,Chiauranib,多替尼,和索拉非尼从PubChem数据库获得。这些结构在Amber10:EHT分子力场中建模,快速制备应用用于校正和优化缺失残基的结构,H计数,终端封顶,和候补。结合位点定义在CSF1R激酶的共结晶配体附近。化合物通过三角形匹配器放置方法进行对接,并通过伦敦dG评分函数进行排名。通过诱导拟合方法进一步细化了对接姿势。在DiscoveryStudioVisualizerv17.2中,使用具有最低结合得分(ΔG)的姿势对配体相互作用谱进行建模。共结晶的配体在其apo构象中对接,并计算均方根偏差以验证对接协议。
    所有7种CSF1R抑制剂均与Met637残基相互作用,表现出选择性,依替尼除外。通过与Asp-Phe-Gly(DFG)基序的Asp797相互作用和/或阻碍Glu633和Lys616之间形成的保守盐桥,从而稳定激活环,或与近膜结构域中的色氨酸残基(Trp550)相互作用。DCC-3014、ARRY-382、BLZ-945和索拉非尼以最低结合能与CSF1R激酶结合。
    嘧啶是与CSF1R残基相互作用的有效抑制剂。DCC-3014和ARRY-382表现出优异的药物潜力,表现出良好的结构稳定性和亲和力。
    UNASSIGNED: Delineate structure-based inhibition of colony-stimulating factor-1 receptor (CSF1R) by small molecule CSF1R inhibitors in clinical development for target identification and potential lead optimization in cancer therapeutics since CSF1R is a novel predictive biomarker for immunotherapy in cancer.
    UNASSIGNED: Compounds were in silico modelled by induced fit docking protocol in a molecular operating environment (MOE, MOE.v.2015). The 3-dimensional (3D) X-ray crystallized structure of CSF1R kinase (Protein Databank, ID 4R7H) was obtained from Research Collaboratory for Structural Bioinformatics (RSCB) Protein Databank. The 3D conformers of edicotinib, DCC-3014, ARRY-382, BLZ-945, chiauranib, dovitinib, and sorafenib were obtained from PubChem Database. These structures were modelled in Amber10:EHT molecular force field, and quick prep application was used to correct and optimize the structures for missing residues, H-counts, termini capping, and alternates. The binding site was defined within the vicinity of the co-crystallized ligand of CSF1R kinase. The compounds were docked by the triangular matcher placement method and ranked by the London dG scoring function. The docked poses were further refined by the induced fit method. The pose with the lowest binding score (ΔG) was used to model the ligand interaction profile in Discovery Studio Visualizer v17.2. The co-crystallized ligand was docked in its apo conformation, and root-mean-square deviation was computed to validate the docking protocol.
    UNASSIGNED: All 7 CSF1R inhibitors interact with residue Met637 exhibiting selectivity except for edicotinib. The inhibitors maintain CSF1R in an auto-inhibitory conformation by interacting with Asp797 of the Asp-Phe-Gly (DFG) motif and/or hindering the conserved salt bridge formed between Glu633 and Lys616 thus stabilizing the activation loop, or interacting with tryptophan residue (Trp550) in the juxtamembrane domain. DCC-3014, ARRY-382, BLZ-945, and sorafenib bind with the lowest binding energy with CSF1R kinase.
    UNASSIGNED: Pyrimidines are potent inhibitors that interact with CSF1R residues. DCC-3014 and ARRY-382 exhibit exceptional pharmaceutical potential exhibiting great structural stability and affinity.
    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
    结合和调节蛋白质功能的小分子的鉴定和优化是药物开发早期阶段的关键步骤。几十年来,这个过程从使用计算模型中受益匪浅,这些计算模型可以提供对分子结合亲和力和优化的见解。在过去的几年里,各种类型的深度学习模型在改进和增强传统计算方法的性能方面显示出巨大的潜力。在这一章中,我们概述了基于深度学习的最新发展及其在药物发现中的应用。根据每种方法旨在解决的任务,我们将这些方法分为四个子类别。对于每个子类别,我们提供了该方法的一般框架,并讨论了各个方法。
    Identification and optimization of small molecules that bind to and modulate protein function is a crucial step in the early stages of drug development. For decades, this process has benefitted greatly from the use of computational models that can provide insights into molecular binding affinity and optimization. Over the past several years, various types of deep learning models have shown great potential in improving and enhancing the performance of traditional computational methods. In this chapter, we provide an overview of recent deep learning-based developments with applications in drug discovery. We classify these methods into four subcategories dependent on the task each method is aiming to solve. For each subcategory, we provide the general framework of the approach and discuss individual methods.
    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
    探索无毒和具有成本效益的膳食成分,如表没食子儿茶素3-没食子酸酯和杨梅素,健康改善和疾病治疗最近引起了大量的研究关注。最近的COVID-19大流行为研究和鉴定能够治疗病毒感染的饮食成分提供了独特的机会,以及收集应对突发公共卫生事件带来的重大挑战所需的证据。由于饮食成分具有良好的安全性,因此具有很大的潜力,可以作为进一步开发用于治疗和预防SARS-CoV-2感染的药物的起点。广谱抗病毒活性,和多器官保护能力。这里,我们回顾了当前的知识特征-化学成分,生物活性特性,和假定的作用机制-具有靶向SARS-CoV-2及其变体潜力的天然生物活性膳食类黄酮。值得注意的是,我们提出了有希望的策略(联合治疗,引线优化,和药物递送),以克服天然膳食类黄酮的固有缺陷,如有限的生物利用度和差的稳定性。
    The exploration of non-toxic and cost-effective dietary components, such as epigallocatechin 3-gallate and myricetin, for health improvement and disease treatment has recently attracted substantial research attention. The recent COVID-19 pandemic has provided a unique opportunity for the investigation and identification of dietary components capable of treating viral infections, as well as gathering the evidence needed to address the major challenges presented by public health emergencies. Dietary components hold great potential as a starting point for further drug development for the treatment and prevention of SARS-CoV-2 infection owing to their good safety, broad-spectrum antiviral activities, and multi-organ protective capacity. Here, we review current knowledge of the characteristics-chemical composition, bioactive properties, and putative mechanisms of action-of natural bioactive dietary flavonoids with the potential for targeting SARS-CoV-2 and its variants. Notably, we present promising strategies (combination therapy, lead optimization, and drug delivery) to overcome the inherent deficiencies of natural dietary flavonoids, such as limited bioavailability and poor stability.
    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
    可解释的机器学习越来越多地用于药物发现,以帮助合理化化合物的性质预测。特征归因技术是确定哪些分子子结构负责预测的性质变化的流行选择。然而,到目前为止,已经建立的分子特征归因方法对于流行的深度学习算法,如图神经网络(GNN),特别是与更简单的建模替代方案相比,例如随机森林和原子掩蔽。为了缓解这个问题,提出了对GNN回归目标的修改,以专门说明分子对之间的共同核心结构。所提出的方法在最近提出的可解释性基准上显示出更高的准确性。这种方法有可能有助于药物发现管道中的模型可解释性,特别是在研究特定化学系列的铅优化工作中。
    Explainable machine learning is increasingly used in drug discovery to help rationalize compound property predictions. Feature attribution techniques are popular choices to identify which molecular substructures are responsible for a predicted property change. However, established molecular feature attribution methods have so far displayed low performance for popular deep learning algorithms such as graph neural networks (GNNs), especially when compared with simpler modeling alternatives such as random forests coupled with atom masking. To mitigate this problem, a modification of the regression objective for GNNs is proposed to specifically account for common core structures between pairs of molecules. The presented approach shows higher accuracy on a recently-proposed explainability benchmark. This methodology has the potential to assist with model explainability in drug discovery pipelines, particularly in lead optimization efforts where specific chemical series are investigated.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号