Structure-based drug discovery

基于结构的药物发现
  • 文章类型: Journal Article
    蛋白质晶体学在药物发现中成功应用的关键先决条件是为新药靶蛋白建立强大的结晶系统,以足够快的速度递送晶体结构,最迟,在随后的领导过程中。产生具有高结晶可能性的良好折叠蛋白的第一个关键步骤是鉴定靶蛋白的合适截短变体。在一些情况下,仅最佳长度变体不足以支持结晶,并且需要引入另外的表面突变以获得合适的晶体。在这一贡献中,提出了四个案例研究,其中合理设计的表面修饰是建立靶蛋白结晶条件的关键(蛋白激酶Aurora-C,IRAK4和BUB1,以及KRAS-SOS1复合物)。描述了导致衍射良好的晶体的设计过程,并分析了晶体堆积,以回顾性地了解特定表面突变如何促进成功结晶。所提出的设计方法通常用于我们的团队,以支持建立强大的结晶系统,从而使结构指导的抑制剂优化成为制药研究中的命中铅和铅优化项目。
    A key prerequisite for the successful application of protein crystallography in drug discovery is to establish a robust crystallization system for a new drug-target protein fast enough to deliver crystal structures when the first inhibitors have been identified in the hit-finding campaign or, at the latest, in the subsequent hit-to-lead process. The first crucial step towards generating well folded proteins with a high likelihood of crystallizing is the identification of suitable truncation variants of the target protein. In some cases an optimal length variant alone is not sufficient to support crystallization and additional surface mutations need to be introduced to obtain suitable crystals. In this contribution, four case studies are presented in which rationally designed surface modifications were key to establishing crystallization conditions for the target proteins (the protein kinases Aurora-C, IRAK4 and BUB1, and the KRAS-SOS1 complex). The design process which led to well diffracting crystals is described and the crystal packing is analysed to understand retrospectively how the specific surface mutations promoted successful crystallization. The presented design approaches are routinely used in our team to support the establishment of robust crystallization systems which enable structure-guided inhibitor optimization for hit-to-lead and lead-optimization projects in pharmaceutical research.
    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
    在寻找新的机会来开发布氏锥虫磷酸二酯酶B1(TbrPDEB1)抑制剂,这些抑制剂对脱靶人PDE4(hPDE4)具有选择性,使用各种生化方法研究了碎片生长运动的不同阶段,结构,热力学,和动力学结合测定。确定了结合动力学的显着差异,并用计算方法探索了这种动力学选择性,包括分子动力学和相互作用指纹分析。这些研究表明,GlnQ.50和抑制剂之间的关键氢键暴露于TbrPDEB1中的水通道,导致快速解结合。这种水通道不存在于hPDE4中,导致具有较长停留时间的抑制剂。计算机辅助药物设计方案被应用于最近公开的具有不同支架的TbrPDEB1抑制剂,并且我们的结果证实通过破坏水通道来屏蔽该关键氢键代表了开发更具选择性的TbrPDEB1抑制剂的可行设计策略。我们的工作展示了如何使用计算协议来理解溶剂动力学对抑制剂结合的贡献,我们的结果可应用于相关寄生虫中发现的同源PDEs的选择性抑制剂的设计。
    In search of new opportunities to develop Trypanosoma brucei phosphodiesterase B1 (TbrPDEB1) inhibitors that have selectivity over the off-target human PDE4 (hPDE4), different stages of a fragment-growing campaign were studied using a variety of biochemical, structural, thermodynamic, and kinetic binding assays. Remarkable differences in binding kinetics were identified and this kinetic selectivity was explored with computational methods, including molecular dynamics and interaction fingerprint analyses. These studies indicate that a key hydrogen bond between GlnQ.50 and the inhibitors is exposed to a water channel in TbrPDEB1, leading to fast unbinding. This water channel is not present in hPDE4, leading to inhibitors with a longer residence time. The computer-aided drug design protocols were applied to a recently disclosed TbrPDEB1 inhibitor with a different scaffold and our results confirm that shielding this key hydrogen bond through disruption of the water channel represents a viable design strategy to develop more selective inhibitors of TbrPDEB1. Our work shows how computational protocols can be used to understand the contribution of solvent dynamics to inhibitor binding, and our results can be applied in the design of selective inhibitors for homologous PDEs found in related parasites.
    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
    恶性疟原虫氨酰基tRNA合成酶(PfaaRSs)是有效的抗疟靶标,对于寄生虫生命周期的每个阶段的蛋白质组保真度和总体寄生虫存活至关重要。到目前为止,这些蛋白质中的一些已被单靶向产生抑制剂化合物,其已被抗性的发生所限制,其可通过泛抑制策略来克服。因此,在这里,第一次,我们报告了丝裂霉素(MMC)作为1a类可能的泛抑制剂的鉴定和体外抗疟原虫验证(精氨酰(A)-,半胱氨酰(C),异亮氨酸(I)-,亮氨酰(L),甲硫氨酰基(M),和valyl(V)-)PfaaRSs,假设可能是其先前报道的对核糖体RNA抑制蛋白质翻译和生物合成的活性的基础。我们结合了多种基于计算机结构的发现策略,这些策略首先有助于鉴定每种疟原虫蛋白中化合物优先靶向的功能性和可药用位点:Pf-ARS中的Ins1-Ins2域;Pf-CRS中的反密码子结合域;Pf-IRS和Pf-MRS中的CP1编辑域;Pf-LRS中的C末端域;和Pf-VRS中的CP核心区域。分子动力学研究进一步揭示了MMC变构诱导每种蛋白质整体结构的变化。同样,显著的结构扰动是由该化合物跨越蛋白质的功能结构域引起的。更多,MMC诱导催化核苷酸和氨基酸底物结合的系统改变,最终导致与关键活性位点残基的关键相互作用丧失,并最终降低所有蛋白质的核苷酸结合亲和力。从结合能计算中推导出来。这些完全证实了MMC均匀地破坏了靶蛋白和必需底物的结构。Further,MMC对寄生虫的Dd2和3D7菌株的IC50<5μM,这使其成为疟疾药物开发的良好起点。我们相信,我们的研究结果将对当前寻找高效的多阶段抗疟药物具有重要意义。
    Plasmodium falciparum aminoacyl tRNA synthetases (PfaaRSs) are potent antimalarial targets essential for proteome fidelity and overall parasite survival in every stage of the parasite\'s life cycle. So far, some of these proteins have been singly targeted yielding inhibitor compounds that have been limited by incidences of resistance which can be overcome via pan-inhibition strategies. Hence, herein, for the first time, we report the identification and in vitro antiplasmodial validation of Mitomycin (MMC) as a probable pan-inhibitor of class 1a (arginyl(A)-, cysteinyl(C), isoleucyl(I)-, leucyl(L), methionyl(M), and valyl(V)-) PfaaRSs which hypothetically may underlie its previously reported activity on the ribosomal RNA to inhibit protein translation and biosynthesis. We combined multiple in silico structure-based discovery strategies that first helped identify functional and druggable sites that were preferentially targeted by the compound in each of the plasmodial proteins: Ins1-Ins2 domain in Pf-ARS; anticodon binding domain in Pf-CRS; CP1-editing domain in Pf-IRS and Pf-MRS; C-terminal domain in Pf-LRS; and CP-core region in Pf-VRS. Molecular dynamics studies further revealed that MMC allosterically induced changes in the global structures of each protein. Likewise, prominent structural perturbations were caused by the compound across the functional domains of the proteins. More so, MMC induced systematic alterations in the binding of the catalytic nucleotide and amino acid substrates which culminated in the loss of key interactions with key active site residues and ultimate reduction in the nucleotide-binding affinities across all proteins, as deduced from the binding energy calculations. These altogether confirmed that MMC uniformly disrupted the structure of the target proteins and essential substrates. Further, MMC demonstrated IC50 < 5 μM against the Dd2 and 3D7 strains of parasite making it a good starting point for malarial drug development. We believe that findings from our study will be important in the current search for highly effective multi-stage antimalarial drugs.
    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
    多年来,人二氢乳清酸脱氢酶(hDHODH),它是嘧啶从头生物合成途径的关键参与者,已经成为治疗几种疾病的目标,包括自身免疫性疾病和急性骨髓性白血病,以及宿主靶向抗病毒治疗。对其抑制剂结合行为的分子探索为创新药物设计提供了有希望的候选药物。酶促药效团的详细描述驱动了完善的抑制性支架的装饰,从而获得进一步的体外和体内功效。在目前的工作中,使用X射线晶体学,在以极性基于吡啶的部分为特征的有效抑制剂(化合物18)的结合状态中鉴定出非典型重排。晶体结构表明,在结合化合物18后,参与辅因子结合位点门控机制的蛋白质环的动力学受到三个水分子的存在的调节。从而微调结合袋的极性/疏水性。这些溶剂分子参与氢键网的形成,其中它们中的一个与化合物18的吡啶部分建立直接接触,从而为重新评估hDHODH的抑制铺平了道路。使用综合方法,这种调制的热力学是通过等温滴定量热法与分子建模相结合来描述的。这些结构见解将指导未来的药物设计,以获得更精细的Kd/logD7.4平衡,并在水溶性方面鉴定具有药物样特征的膜可渗透分子。
    Over the years, human dihydroorotate dehydrogenase (hDHODH), which is a key player in the de novo pyrimidine-biosynthesis pathway, has been targeted in the treatment of several conditions, including autoimmune disorders and acute myelogenous leukaemia, as well as in host-targeted antiviral therapy. A molecular exploration of its inhibitor-binding behaviours yielded promising candidates for innovative drug design. A detailed description of the enzymatic pharmacophore drove the decoration of well-established inhibitory scaffolds, thus gaining further in vitro and in vivo efficacy. In the present work, using X-ray crystallography, an atypical rearrangement was identified in the binding pose of a potent inhibitor characterized by a polar pyridine-based moiety (compound 18). The crystal structure shows that upon binding compound 18 the dynamics of a protein loop involved in a gating mechanism at the cofactor-binding site is modulated by the presence of three water molecules, thus fine-tuning the polarity/hydrophobicity of the binding pocket. These solvent molecules are engaged in the formation of a hydrogen-bond mesh in which one of them establishes a direct contact with the pyridine moiety of compound 18, thus paving the way for a reappraisal of the inhibition of hDHODH. Using an integrated approach, the thermodynamics of such a modulation is described by means of isothermal titration calorimetry coupled with molecular modelling. These structural insights will guide future drug design to obtain a finer Kd/logD7.4 balance and identify membrane-permeable molecules with a drug-like profile in terms of water solubility.
    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
    TIGIT主要在T细胞上表达,并且是在肿瘤微环境中与其配体PVR结合的抑制性检查点受体。抗TIGIT单克隆抗体(mAb),例如Ociperlimab和Tiragolumab阻断TIGIT-PVR相互作用,并且正在临床开发中。然而,这些单克隆抗体的分子阻断机制仍然难以捉摸。这里,我们报告了TIGIT与Ociperlimab_Fab和Tiragolumab_Fab复合物的晶体结构,揭示了两种mAb与PVR的空间位阻大结合TIGIT。此外,鉴定了几个关键的表位残基。有趣的是,当将pH从7.4降低至6.0时,Ociperlimab对TIGIT的结合亲和力增加约17倍。我们的结构显示了ASP103HCDR3和HIS76TIGIT之间的强静电相互作用,解释了Ociperlimab的pH响应机制。相比之下,Tiragolumab不显示酸性pH依赖性结合增强。我们的结果提供了有价值的信息,可以帮助提高治疗性抗体对癌症治疗的疗效。
    TIGIT is mainly expressed on T cells and is an inhibitory checkpoint receptor that binds to its ligand PVR in the tumor microenvironment. Anti-TIGIT monoclonal antibodies (mAbs) such as Ociperlimab and Tiragolumab block the TIGIT-PVR interaction and are in clinical development. However, the molecular blockade mechanism of these mAbs remains elusive. Here, we report the crystal structures of TIGIT in complex with Ociperlimab_Fab and Tiragolumab_Fab revealing that both mAbs bind TIGIT with a large steric clash with PVR. Furthermore, several critical epitopic residues are identified. Interestingly, the binding affinity of Ociperlimab toward TIGIT increases approximately 17-fold when lowering the pH from 7.4 to 6.0. Our structure shows a strong electrostatic interaction between ASP103HCDR3 and HIS76TIGIT explaining the pH-responsive mechanism of Ociperlimab. In contrast, Tiragolumab does not show an acidic pH-dependent binding enhancement. Our results provide valuable information that could help to improve the efficacy of therapeutic antibodies for cancer treatment.
    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
    AlphaFold系列的推出将深度学习技术带入了分子结构科学。作为另一个关键问题,基于结构的蛋白质-配体结合亲和力预测迫切需要先进的计算技术。深度学习准备好解码这个问题了吗?在这里,我们回顾了主流的基于结构的,这个问题的深度学习方法,专注于分子表征,学习架构和模型可解释性。已生成模型分类。为了弥补这些模型之间缺乏有效的比较,我们从统一的基础上认识和评价代表,优点和缺点进行了讨论。这篇综述将潜在地有益于基于结构的药物发现和相关领域。
    The launch of AlphaFold series has brought deep-learning techniques into the molecular structural science. As another crucial problem, structure-based prediction of protein-ligand binding affinity urgently calls for advanced computational techniques. Is deep learning ready to decode this problem? Here we review mainstream structure-based, deep-learning approaches for this problem, focusing on molecular representations, learning architectures and model interpretability. A model taxonomy has been generated. To compensate for the lack of valid comparisons among those models, we realized and evaluated representatives from a uniform basis, with the advantages and shortcomings discussed. This review will potentially benefit structure-based drug discovery and related areas.
    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
    能够从筛选中快速发现可靠的命中,并迅速将其转化为铅化合物,这可以在功能测定中得到验证,是药物发现的核心。新靶标的快速验证和调节剂的鉴定以推进临床前研究可以显着提高药物开发的成功率。我们的SaXPyTM(“X射线快速定位SAR”)平台,适用于任何X射线晶体学药物靶标,将已建立的蛋白质X射线晶体学和基于片段的药物发现(FBDD)方法与先进的计算和药物化学相结合,以在短时间内提供小分子调节剂或靶向蛋白质降解配体。我们的方法,特别是对于难以捉摸的或“不可用的”目标,允许(i)命中生成;(ii)蛋白质-配体相互作用的作图;(iii)评估靶标的可连接性;(iv)发现新的和潜在的变构结合位点;和(v)命中到前导执行。这些进步为化学可处理性和下游生物学提供了信息,并产生了新的知识产权。我们在这里描述了SaXPy在针对DNA聚合酶eta(Polη或POLH)和嘌呤/嘧啶核酸内切酶1(APE1或APEX1)的DNA损伤应答抑制剂的发现和开发中的应用。值得注意的是,我们的SaXPy平台使我们能够解决这些蛋白质与小分子结合的第一个晶体结构,并发现每个靶标的新结合位点。
    The ability to quickly discover reliable hits from screening and rapidly convert them into lead compounds, which can be verified in functional assays, is central to drug discovery. The expedited validation of novel targets and the identification of modulators to advance to preclinical studies can significantly increase drug development success. Our SaXPyTM (\"SAR by X-ray Poses Quickly\") platform, which is applicable to any X-ray crystallography-enabled drug target, couples the established methods of protein X-ray crystallography and fragment-based drug discovery (FBDD) with advanced computational and medicinal chemistry to deliver small molecule modulators or targeted protein degradation ligands in a short timeframe. Our approach, especially for elusive or \"undruggable\" targets, allows for (i) hit generation; (ii) the mapping of protein-ligand interactions; (iii) the assessment of target ligandability; (iv) the discovery of novel and potential allosteric binding sites; and (v) hit-to-lead execution. These advances inform chemical tractability and downstream biology and generate novel intellectual property. We describe here the application of SaXPy in the discovery and development of DNA damage response inhibitors against DNA polymerase eta (Pol η or POLH) and apurinic/apyrimidinic endonuclease 1 (APE1 or APEX1). Notably, our SaXPy platform allowed us to solve the first crystal structures of these proteins bound to small molecules and to discover novel binding sites for each target.
    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
    利什曼病是由20种利什曼原虫引起的,在流行地区影响数百万人。现有的治疗方法不足以有效控制疾病,引起严重的副作用并最终导致耐药性,迫切需要发现新的治疗分子。基于分子靶标的药物发现,目标是一个确定的分子基因,蛋白质或机制,是新疗法的理论驱动方法。人类从膳食来源获得必需氨基酸如苏氨酸,而利什曼原虫是从头合成的。苏氨酸生物合成途径的酶,因此,包括将L-高丝氨酸转化为正磷酸高丝氨酸的限速高丝氨酸激酶(HSK)是理论驱动疗法的有吸引力的靶标。HSK在人类中的缺失及其在多尼利什曼原虫中的存在增加了利用HSK作为抗利什曼原虫治疗开发的分子靶标的机会。在这项研究中,我们利用基于结构的高通量药物发现(SBDD),随后进行生化验证,并从Maybridge化学文库中鉴定出两种潜在的抑制剂(RH00038和S02587),靶向L.donovaniHSK。这两种抑制剂有效地诱导了利什曼原虫donovani的死亡在两个阶段的阿马斯蒂戈特和前乳突。其中一种对寄生虫具有特异性,是标准治疗分子的两倍。
    Leishmaniasis is caused by ∼20 species of Leishmania that affects millions in endemic areas. Available therapies are not sufficient to effectively control the disease, cause severe side effects and eventually lead to drug resistance, making the discovery of novel therapeutic molecules an immediate need. Molecular target-based drug discovery, where the target is a defined molecular gene, protein or a mechanism, is a rationale driven approach for novel therapeutics. Humans obtain the essential amino acid such as threonine from dietary sources, while Leishmania synthesize it de-novo. Enzymes of the threonine biosynthesis pathway, including the rate limiting Homoserine kinase (HSK) which converts L-homoserine into ortho-phospho homoserine are thus attractive targets for rationale driven therapy. The absence of HSK in humans and its presence in Leishmania donovani enhances the opportunity to exploit HSK as a molecular target for anti-leishmanials therapeutic development. In this study, we utilize structure-based high throughput drug discovery (SBDD), followed by biochemical validation and identified two potential inhibitors (RH00038 and S02587) from Maybridge chemical library that targets L. donovani HSK. These two inhibitors effectively induced the mortality of Leishmania donovani in both amastigote and promastigote stages, with one of them being specific to parasite and twice as effective as the standard therapeutic molecule.
    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
    如今,人们普遍认为水分子在配体与分子靶标结合中起关键作用。在分子识别过程中忽略水分子是基于结构的药物发现活动中多次失败的结果。WaterMap的应用,特别是WaterMap引导的分子对接,可以合理准确和快速地描述配体-蛋白质界面处水分子的位置和能量。
    在这篇评论中,作者简要讨论了水在药物设计和发现中的重要性,并简要概述了用于预测溶剂相关效应的计算方法,以便在其他可用技术和工具的背景下介绍WaterMap。在简要描述WaterMap概念之后,介绍了2013年至2023年之间发布的WaterMap辅助虚拟筛选文献。
    近年来,WaterMap软件已广泛用于支持基于结构的药物设计,尤其是基于结构的虚拟筛选。的确,考虑到结合袋中的水分子,它是重新计算对接结果的有用工具。尽管WaterMap允许考虑结合位点中水分子的动态行为,为了获得最佳准确性,建议将其与FEP的MM-GB/SA等其他技术结合使用。
    Nowadays, it is widely accepted that water molecules play a key role in binding a ligand to a molecular target. Neglecting water molecules in the process of molecular recognition was the result of several failures of the structure-based drug discovery campaigns. The application of WaterMap, in particular WaterMap-guided molecular docking, enables the reasonably accurate and quick description of the location and energetics of water molecules at the ligand-protein interface.
    In this review, the authors shortly discuss the importance of water in drug design and discovery and provide a brief overview of the computational approaches used to predict the solvent-related effects for the purposes of presenting WaterMap in the context of other available techniques and tools. A concise description of WaterMap concept is followed by the presentation of WaterMap-assisted virtual screening literature published between 2013 and 2023.
    In recent years, WaterMap software has been extensively used to support structure-based drug design, in particular structure-based virtual screening. Indeed, it is a useful tool to rescore docking results considering water molecules in the binding pocket. Although WaterMap allows for the consideration of the dynamic behavior of water molecules in the binding site, for best accuracy, its application in conjunction with other techniques such as molecular mechanics-generalized Born surface area of FEP (Free Energy Perturbation) is recommended.
    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
    精确的虚拟筛选工具的设计是药物发现中的一个开放挑战。已经在不同的近似水平上开发了几种基于结构的方法。其中,分子对接是一种高效的成熟技术,但通常精度较低。此外,已知对接性能与目标相关,这使得在接近新的蛋白质靶标时,对接程序和相应的评分功能的选择至关重要。为了比较不同对接协议的性能,我们开发了ChemFlow_py,一个自动化的工具来执行对接和重新评分。使用从DUD-E中提取的四种蛋白质系统,每个目标有100种已知的活性化合物和3000种诱饵,我们比较了几种评分策略的性能,包括共识评分。我们发现,平均对接结果可以通过共识排序来提高,它强调了当给定目标的化学信息很少或没有时,共识评分的相关性。ChemFlow_py是一个免费的工具包,用于优化虚拟高通量筛选(vHTS)的性能。该软件可在https://github.com/IFMlab/ChemFlow_py上公开获得。
    The design of accurate virtual screening tools is an open challenge in drug discovery. Several structure-based methods have been developed at different levels of approximation. Among them, molecular docking is an established technique with high efficiency, but typically low accuracy. Moreover, docking performances are known to be target-dependent, which makes the choice of the docking program and corresponding scoring function critical when approaching a new protein target. To compare the performances of different docking protocols, we developed ChemFlow_py, an automated tool to perform docking and rescoring. Using four protein systems extracted from DUD-E with 100 known active compounds and 3000 decoys per target, we compared the performances of several rescoring strategies including consensus scoring. We found that the average docking results can be improved by consensus ranking, which emphasizes the relevance of consensus scoring when little or no chemical information is available for a given target. ChemFlow_py is a free toolkit to optimize the performances of virtual high-throughput screening (vHTS). The software is publicly available at https://github.com/IFMlab/ChemFlow_py .
    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