phenotypic drug discovery

表型药物发现
  • 文章类型: Journal Article
    我国资源丰富、治疗复杂疾病疗效确切的中药具有巨大的发展潜力。然而,复杂的成分,药效学物质和作用机制不明确,缺乏合理的临床安全性和有效性评价方法限制了以中药为基础的创新药物的研发。人工智能和仿生学等前沿学科的进展,特别是细胞绘画和器官芯片的出现,有助于根据模型特征的变化来识别和表征中药中的有效成分,从而为中药的开发和应用提供更准确的指导。表型药物发现在中药创新药物研发中的应用日益受到重视。近年来,表型药物发现技术不断进步,提高了新药的早期发现率和药物研发的成功率。因此,表型药物发现逐渐成为新药研究的关键工具。本文论述了中药在创新药物发现和开发中的巨大潜力,由细胞绘画等尖端技术支持,深度学习,和器官芯片,推动中医药进入新的发展阶段。
    Traditional Chinese medicine with rich resources in China and definite therapeutic effects on complex diseases demonstrates great development potential. However, the complex composition, the unclear pharmacodynamic substances and mechanisms of action, and the lack of reasonable methods for evaluating clinical safety and efficacy have limited the research and development of innovative drugs based on traditional Chinese medicine. The progress in cutting-edge disciplines such as artificial intelligence and biomimetics, especially the emergence of cell painting and organ-on-a-chip, helps to identify and characterize the active ingredients in traditional Chinese medicine based on the changes in model characteristics, thus providing more accurate guidance for the development and application of traditional Chinese medicine. The application of phenotypic drug discovery in the research and development of innovative drugs based on traditional Chinese medicine is gaining increasing attention. In recent years, the technology for phenotypic drug discovery keeps advancing, which improves the early discovery rate of new drugs and the success rate of drug research and development. Accordingly, phenotypic drug discovery gradually becomes a key tool for the research on new drugs. This paper discusses the enormous potential of traditional Chinese medicine in the discovery and development of innovative drugs and illustrates how the application of phenotypic drug discovery, supported by cutting-edge technologies such as cell painting, deep learning, and organ-on-a-chip, propels traditional Chinese medicine into a new stage of development.
    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
    表型药物发现(PDD),这涉及直接利用生物系统来发现有效的药物,近年来经历了一次复苏。过去几年人工智能(AI)的快速发展为在微流体平台上增强表型药物筛选提供了许多机会。利用其预测能力,数据分析,高效的数据处理,等。微流控与AI相结合有望彻底改变表型药物发现的格局。通过将先进的微流体平台与AI算法集成,研究人员可以快速筛选大型化合物库,确定新的候选药物,并以前所未有的速度和效率阐明复杂的生物途径。这篇综述概述了基于AI的微流体及其在药物发现中的应用的最新进展和挑战。我们讨论了微流控系统的协同组合,用于高通量筛选和AI驱动的表型表征分析,药物-靶标相互作用,和预测建模。此外,我们强调了AI驱动的微流体实现自动化药物筛选系统的潜力。总的来说,AI驱动的微流体代表了一种有希望的方法,通过实现快速,成本效益高,和治疗相关化合物的准确鉴定。
    Phenotypic drug discovery (PDD), which involves harnessing biological systems directly to uncover effective drugs, has undergone a resurgence in recent years. The rapid advancement of artificial intelligence (AI) over the past few years presents numerous opportunities for augmenting phenotypic drug screening on microfluidic platforms, leveraging its predictive capabilities, data analysis, efficient data processing, etc. Microfluidics coupled with AI is poised to revolutionize the landscape of phenotypic drug discovery. By integrating advanced microfluidic platforms with AI algorithms, researchers can rapidly screen large libraries of compounds, identify novel drug candidates, and elucidate complex biological pathways with unprecedented speed and efficiency. This review provides an overview of recent advances and challenges in AI-based microfluidics and their applications in drug discovery. We discuss the synergistic combination of microfluidic systems for high-throughput screening and AI-driven analysis for phenotype characterization, drug-target interactions, and predictive modeling. In addition, we highlight the potential of AI-powered microfluidics to achieve an automated drug screening system. Overall, AI-powered microfluidics represents a promising approach to shaping the future of phenotypic drug discovery by enabling rapid, cost-effective, and accurate identification of therapeutically relevant compounds.
    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
    表型药物发现(PDD)涉及筛选化合物对细胞的影响,组织,或整个生物体不一定了解潜在的分子靶标。PDD与基于靶标的策略不同,因为它不需要了解特定的药物靶标或其在疾病中的作用。这种方法可以导致发现具有意想不到的治疗效果或应用的药物,并允许根据其功能效果来鉴定药物,而不是通过预定义的基于目标的方法。最终,疾病的定义主要是基于症状,而不是基于机制,治疗方法也应该如此。近年来,人们对PDD重新产生了兴趣,因为它有可能解决人类疾病的复杂性,包括与构成代谢宿主-微生物相互作用中心中心的多个靶标的多种代谢物的整体图景。尽管PDD提出了诸如命中验证和目标反卷积等挑战,大数据时代取得了重大成就。本文探讨了研究人员测试胸腺肽激素作用的经验,胸腺素α-1,在临床前和临床环境中,并讨论其在精准医学时代的治疗效用如何在PDD框架内适应。
    Phenotypic drug discovery (PDD) involves screening compounds for their effects on cells, tissues, or whole organisms without necessarily understanding the underlying molecular targets. PDD differs from target-based strategies as it does not require knowledge of a specific drug target or its role in the disease. This approach can lead to the discovery of drugs with unexpected therapeutic effects or applications and allows for the identification of drugs based on their functional effects, rather than through a predefined target-based approach. Ultimately, disease definitions are mostly symptom-based rather than mechanism-based, and the therapeutics should be likewise. In recent years, there has been a renewed interest in PDD due to its potential to address the complexity of human diseases, including the holistic picture of multiple metabolites engaging with multiple targets constituting the central hub of the metabolic host-microbe interactions. Although PDD presents challenges such as hit validation and target deconvolution, significant achievements have been reached in the era of big data. This article explores the experiences of researchers testing the effect of a thymic peptide hormone, thymosin alpha-1, in preclinical and clinical settings and discuss how its therapeutic utility in the precision medicine era can be accommodated within the PDD framework.
    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
    表型药物发现(PDD)是一种有效的药物发现方法,通过观察对疾病表型的治疗效果,特别是在复杂的疾病系统中。三阴性乳腺癌(TNBC)由几种复杂的疾病特征组成,包括高度的肿瘤异质性,高侵袭和转移潜能,缺乏有效的治疗靶点。因此,通过PDD识别有效和新颖的药物是TNBC药物开发的当前趋势。在这项研究中,使用4-(苯基磺酰基)吗啉作为药效团合成了23个新的小分子。在这些衍生物中,GL24(4m)在MDA-MB-231细胞中表现出最低的半最大抑制浓度值(0.90μM)。为了研究GL24的肿瘤抑制机制,使用转录组学分析来检测GL24治疗后基因表达的扰动。其次是基因本体论(GO)分析,基因集富集分析(GSEA),和京都基因和基因组百科全书(KEGG)分析,多个ER应激依赖性肿瘤抑制信号被识别,如未折叠蛋白反应(UPR),p53通路,G2/M检查点,和E2F目标。由GL24触发的大多数确定的途径最终导致细胞周期停滞,然后导致细胞凋亡。总之,我们开发了一种新型的4-(苯磺酰基)吗啉衍生物GL24,具有通过ER应激依赖性肿瘤抑制信号抑制TNBC细胞生长的强大潜力。
    Phenotypic drug discovery (PDD) is an effective drug discovery approach by observation of therapeutic effects on disease phenotypes, especially in complex disease systems. Triple-negative breast cancer (TNBC) is composed of several complex disease features, including high tumor heterogeneity, high invasive and metastatic potential, and a lack of effective therapeutic targets. Therefore, identifying effective and novel agents through PDD is a current trend in TNBC drug development. In this study, 23 novel small molecules were synthesized using 4-(phenylsulfonyl)morpholine as a pharmacophore. Among these derivatives, GL24 (4m) exhibited the lowest half-maximal inhibitory concentration value (0.90 µM) in MDA-MB-231 cells. To investigate the tumor-suppressive mechanisms of GL24, transcriptomic analyses were used to detect the perturbation for gene expression upon GL24 treatment. Followed by gene ontology (GO) analysis, gene set enrichment analysis (GSEA), and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, multiple ER stress-dependent tumor suppressive signals were identified, such as unfolded protein response (UPR), p53 pathway, G2/M checkpoint, and E2F targets. Most of the identified pathways triggered by GL24 eventually led to cell-cycle arrest and then to apoptosis. In summary, we developed a novel 4-(phenylsulfonyl)morpholine derivative GL24 with a strong potential for inhibiting TNBC cell growth through ER stress-dependent tumor suppressive signals.
    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
    在克服他汀类药物在心血管疾病(CVD)中表现不佳的策略中,针对前蛋白转化酶枯草杆菌蛋白酶样Kexin9型(PCSK9)的药物的开发被认为是最有前途的药物之一。然而,迄今为止,只有抗PCSK9生物药物获得批准,和口服可用的用于治疗高胆固醇血症的小分子在市场上仍然缺失。在目前的工作中,我们描述了表型方法在鉴定和优化4-氨基-2-吡啶酮衍生物作为具有抗PCSK9活性的新化学型中的应用。从内部收集的化合物开始,对HepG2细胞进行功能测定,然后进行化学驱动的命中优化活动,导致了有效的抗PCSK9候选物5c。这个化合物,在5μM,完全阻断HepG2细胞分泌PCSK9,LDL受体(LDLR)表达显着增加,并通过减少辛伐他汀对PCSK9表达的诱导与辛伐他汀协同作用。最后,化合物5c也被证明在测试浓度(40mg/kg)下在C57BL/6J小鼠中具有良好的耐受性,没有毒性或行为改变的迹象。
    Among the strategies to overcome the underperformance of statins in cardiovascular diseases (CVDs), the development of drugs targeting the Proprotein Convertase Subtilisin-like Kexin type 9 (PCSK9) is considered one of the most promising. However, only anti-PCSK9 biological drugs have been approved to date, and orally available small-molecules for the treatment of hypercholesterolemic conditions are still missing on the market. In the present work, we describe the application of a phenotypic approach to the identification and optimization of 4-amino-2-pyridone derivatives as a new chemotype with anti-PCSK9 activity. Starting from an in-house collection of compounds, functional assays on HepG2 cells followed by a chemistry-driven hit optimization campaign, led to the potent anti-PCSK9 candidate 5c. This compound, at 5 μM, totally blocked PCSK9 secretion from HepG2 cells, significantly increased LDL receptor (LDLR) expression, and acted cooperatively with simvastatin by reducing its induction of PCSK9 expression. Finally, compound 5c also proved to be well tolerated in C57BL/6J mice at the tested concentration (40 mg/kg) with no sign of toxicity or behavior modifications.
    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: Review
    分枝杆菌种类包括大量的病原生物,如结核分枝杆菌,麻风分枝杆菌,和各种非结核分枝杆菌。分枝杆菌膜蛋白大3(MmpL3)是生长和细胞活力所需的必需的霉菌酸和脂质转运蛋白。在过去的十年里,许多研究已经在蛋白质功能方面描述了MmpL3,本地化,regulation,和底物/抑制剂相互作用。这篇综述总结了该领域的新发现,并试图评估我们对MmpL3作为药物靶标的快速扩展理解的未来研究领域。提供了对抑制剂具有抗性的已知MmpL3突变的图集,将氨基酸取代映射到MmpL3的特定结构域。此外,对不同类型的Mmpl3抑制剂的化学特征进行了比较,以提供对不同MmpL3抑制剂的共同和独特特征的见解。
    Mycobacteria species include a large number of pathogenic organisms such as Mycobacterium tuberculosis, Mycobacterium leprae, and various non-tuberculous mycobacteria. Mycobacterial membrane protein large 3 (MmpL3) is an essential mycolic acid and lipid transporter required for growth and cell viability. In the last decade, numerous studies have characterized MmpL3 with respect to protein function, localization, regulation, and substrate/inhibitor interactions. This review summarizes new findings in the field and seeks to assess future areas of research in our rapidly expanding understanding of MmpL3 as a drug target. An atlas of known MmpL3 mutations that provide resistance to inhibitors is presented, which maps amino acid substitutions to specific structural domains of MmpL3. In addition, chemical features of distinct classes of Mmpl3 inhibitors are compared to provide insights into shared and unique features of varied MmpL3 inhibitors.
    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
    脂滴(LD),最初被认为仅仅是脂质储存结构,是高度动态的细胞器,具有控制细胞命运和行为的复杂功能。近年来,它们作为广泛的人类疾病的治疗靶标的相关性已经得到了很好的确立。因此,开发研究工具的努力已经加强,包括可以在临床相关的基于细胞的模型中准确跟踪LD水平的测定。我们以前报道过,LD的积累决定了足细胞在代谢和非代谢起源的肾脏疾病中的脂毒性和细胞死亡。我们还表明,这些细胞中的LD积累既可以作为疾病进展的标志物,也可以作为治疗靶标。这里,我们描述了一种强大的表型筛选方法,使用分化的人类足细胞,用于鉴定在细胞应激下从LD积累和脂毒性中拯救足细胞的小分子化合物。主要测定进展包括1)使用溶剂化变色染料改善LD染色,减少背景噪音,并提高检测精度,2)使用共焦成像来减少LD的垂直重叠并实现精确计数,3)结合膜和细胞骨架染色,以改善共聚焦模式下的细胞分割,和4)使用优化的斑点检测算法,其需要每次单独运行的最小配置。该测定是稳健的,并且产生始终>0.5的Z因子。
    Lipid droplets (LDs), initially thought to be mere lipid storage structures, are highly dynamic organelles with complex functions that control cell fate and behavior. In recent years, their relevance as therapeutic targets for a wide array of human diseases has been well established. Consequently, efforts to develop tools to study them have intensified, including assays that can accurately track LD levels in clinically relevant cell-based models. We previously reported that LD accumulation destines podocytes for lipotoxicity and cell death in renal diseases of metabolic and nonmetabolic origin. We also showed that LD accumulation in those cells serves as both a marker for disease progression and as a therapeutic target. Here, we describe a robust phenotypic screening method, using differentiated human podocytes, for identifying small-molecule compounds that rescue podocytes from LD accumulation and lipotoxicity under cellular stress. Major assay advances include 1) the use of a solvatochromic dye to improve LD staining, reduce background noise, and improve detection accuracy, 2) use of confocal imaging to reduce vertical overlap of LDs and enable accurate counting, 3) combining membrane and cytoskeleton stains to improve cell segmentation in confocal mode, and 4) use of an optimized spot detection algorithm that requires minimal configuration per individual run. The assay is robust and yields a Z-factor that is consistently >0.5.
    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
    现代药物发现方法通常使用高含量成像来系统地研究化合物的大型文库对细胞的影响。通过自动筛选数千或数百万张图像来识别特定的药物诱导的细胞表型,例如,细胞形态改变,这些方法可以揭示提供治疗前景的“命中”化合物。在过去的几年里,基于深度学习(DL)[机器学习(ML)技术家族]的人工智能(AI)方法几乎破坏了所有图像分析任务,从图像分类到分割。这些强大的方法还有望通过加速有效药物及其作用模式的识别来影响药物发现。在这次审查中,我们强调ML的应用和改编,特别是用于基于细胞的表型药物发现(PDD)的DL方法。
    Modern drug discovery approaches often use high-content imaging to systematically study the effect on cells of large libraries of chemical compounds. By automatically screening thousands or millions of images to identify specific drug-induced cellular phenotypes, for example, altered cellular morphology, these approaches can reveal \'hit\' compounds offering therapeutic promise. In the past few years, artificial intelligence (AI) methods based on deep learning (DL) [a family of machine learning (ML) techniques] have disrupted virtually all image analysis tasks, from image classification to segmentation. These powerful methods also promise to impact drug discovery by accelerating the identification of effective drugs and their modes of action. In this review, we highlight applications and adaptations of ML, especially DL methods for cell-based phenotypic drug discovery (PDD).
    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
    干细胞移植和基因疗法为镰状细胞病(SCD)患者提供了潜在的治疗方法。但是这些选择需要先进的医疗设施,而且价格昂贵。因此,对于大多数患有这种疾病的患者,这些治疗方法将无法使用很多年。现在迫切需要的是一种廉价的口服药物,除了羟基脲,FDA批准的唯一一种抑制镰状血红蛋白聚合的药物。这里,我们报告了我们对ScrippsReFRAME药物再利用文库中的12,657种化合物进行表型筛选的第一阶段结果,该方法使用最近开发的高通量试验来测量镰状性状个体红细胞脱氧至0%氧后的镰状培养时间.ReFRAME文库是一个非常重要的集合,因为这些化合物要么是FDA批准的药物,要么已经在临床试验中进行了测试。从剂量反应测量来看,12,657种化合物中的106种在31nM至10μM的浓度范围内表现出统计学上显著的抗凝固作用。抑制性状细胞的镰状化的化合物对于SCD细胞也是有效的。106种抗镰刀菌化合物中有多达21种成为潜在药物。该估计是基于抑制浓度与人血清中口服药物的游离浓度的比较。此外,每个抑制水平的预期治疗潜力可以通过测量不同严重程度的镰状综合征个体的细胞的镰状培养时间来预测。我们的结果应该激励其他人将这106种化合物中的一种或多种开发成治疗SCD的药物。
    Stem cell transplantation and genetic therapies offer potential cures for patients with sickle cell disease (SCD), but these options require advanced medical facilities and are expensive. Consequently, these treatments will not be available for many years to the majority of patients suffering from this disease. What is urgently needed now is an inexpensive oral drug in addition to hydroxyurea, the only drug approved by the FDA that inhibits sickle-hemoglobin polymerization. Here, we report the results of the first phase of our phenotypic screen of the 12,657 compounds of the Scripps ReFRAME drug repurposing library using a recently developed high-throughput assay to measure sickling times following deoxygenation to 0% oxygen of red cells from sickle trait individuals. The ReFRAME library is a very important collection because the compounds are either FDA-approved drugs or have been tested in clinical trials. From dose-response measurements, 106 of the 12,657 compounds exhibit statistically significant antisickling at concentrations ranging from 31 nM to 10 μM. Compounds that inhibit sickling of trait cells are also effective with SCD cells. As many as 21 of the 106 antisickling compounds emerge as potential drugs. This estimate is based on a comparison of inhibitory concentrations with free concentrations of oral drugs in human serum. Moreover, the expected therapeutic potential for each level of inhibition can be predicted from measurements of sickling times for cells from individuals with sickle syndromes of varying severity. Our results should motivate others to develop one or more of these 106 compounds into drugs for treating SCD.
    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
    天然产物代表了前所未有的抗癌化合物的极好来源。然而,确定作用机制仍然是一项重大挑战。已经考虑了几种技术和方法,但成功有限。在这项工作中,我们探索了活细胞成像和机器学习技术的组合,作为一种有前途的工具,可以在快速和负担得起的测试中描述具有抗增殖活性的天然化合物的作用模式。为了开发模型,我们选择了非小细胞肺癌细胞系SW1573,它暴露于已知的抗有丝分裂药物紫杉醇,秋水仙碱和长春碱。我们方法的新颖性集中在两个具有最高相关性的主要特征上,(A)有意义的表型指标,和(b)将表型参数的时间序列快速傅里叶变换(FFT)成它们相应的幅度和相位。由此产生的算法能够对微管干扰物进行聚类,同时紫杉醇与其他治疗呈负相关。FFT方法能够像肉眼检查一样有效地对样本进行分组。这种方法可以很容易地扩展到对大量数据进行分组,而无需视觉监督。
    Natural products represent an excellent source of unprecedented anticancer compounds. However, the identification of the mechanism of action remains a major challenge. Several techniques and methodologies have been considered, but with limited success. In this work, we explored the combination of live cell imaging and machine learning techniques as a promising tool to depict in a fast and affordable test the mode of action of natural compounds with antiproliferative activity. To develop the model, we selected the non-small cell lung cancer cell line SW1573, which was exposed to the known antimitotic drugs paclitaxel, colchicine and vinblastine. The novelty of our methodology focuses on two main features with the highest relevance, (a) meaningful phenotypic metrics, and (b) fast Fourier transform (FFT) of the time series of the phenotypic parameters into their corresponding amplitudes and phases. The resulting algorithm was able to cluster the microtubule disruptors, and meanwhile showed a negative correlation between paclitaxel and the other treatments. The FFT approach was able to group the samples as efficiently as checking by eye. This methodology could easily scale to group a large amount of data without visual supervision.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号