structure-based drug design

基于结构的药物设计
  • 文章类型: Journal Article
    前列腺癌(PCa)是男性的主要癌症之一,缺乏合适的生物标志物或其调节剂会导致预后不良。膜蛋白(MPs)在PCa的发生和发展中起着至关重要的作用,并且可以成为有吸引力的治疗靶标。然而,靶向MPs的实验局限性阻碍了有效的生物标志物和抑制剂的发现。为了克服这个障碍,计算方法可以产生结构洞察和筛选大型化合物库,加速线索识别和优化。在这次审查中,我们研究了计算机辅助药物设计(CADD)的当前突破,重点是靶向最相关的膜结合PCa生物标志物的基于结构的方法。
    Prostate cancer (PCa) is one of the leading cancers in men and the lack of suitable biomarkers or their modulators results in poor prognosis. Membrane proteins (MPs) have a crucial role in the development and progression of PCa and can be attractive therapeutic targets. However, experimental limitations in targeting MPs hinder effective biomarker and inhibitor discovery. To overcome this barrier, computational methods can yield structural insights and screen large libraries of compounds, accelerating lead identification and optimization. In this review, we examine current breakthroughs in computer-aided drug design (CADD), with emphasis on structure-based approaches targeting the most relevant membrane-bound PCa biomarkers.
    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
    肠道病毒是一种单链,高度多样化的正义RNA病毒,包括人类肠道病毒A-D和人类鼻病毒A-C。他们负责许多疾病和一些感染可以发展到危及生命的并发症,特别是儿童或免疫功能低下的患者。迄今为止,市场上没有针对肠道病毒的治疗方法,脊髓灰质炎病毒(疫苗)和EV-A71(中国疫苗)除外。在(SARS-Cov2)封锁期间和之后不久,肠道病毒感染有所减少,再次发现肠道病毒爆发,尤其是在年幼的孩子。这种重新出现凸显了开发针对肠道病毒的广谱治疗的必要性。在过去的一年里,我们的研究小组发现了一类新的显示抗EV活性的小分子抑制剂.靶向病毒衣壳中众所周知的疏水口袋,这些化合物显示出对EV-A71的微摩尔活性和高选择性指数(SI)(5h:EC50,MRC-5=0.57μM,CC50,MRC-5>20μM,SI>35;EC50,RD=4.38μM,CC50,RD>40μM,SI>9;6c:EC50,MRC-5=0.29μM,CC50,MRC-5>20μM,SI>69;EC50,RD=1.66μM,CC50,RD>40μM,SI>24;参考:VapendavirEC50,MRC-5=0.36μM,CC50,MRC-5>20μM,EC50,RD=0.53μM,CC50,RD>40μM,SI>63)。分析了这些化合物与肠道病毒衣壳的复合物的结合模式,并显示出一系列保守的相互作用。因此,6c及其衍生物是用于治疗肠道病毒感染的有希望的候选物。
    The enterovirus is a genus of single-stranded, highly diverse positive-sense RNA viruses, including Human Enterovirus A-D and Human Rhinovirus A-C species. They are responsible for numerous diseases and some infections can progress to life-threatening complications, particularly in children or immunocompromised patients. To date, there is no treatment against enteroviruses on the market, except for polioviruses (vaccine) and EV-A71 (vaccine in China). Following a decrease in enterovirus infections during and shortly after the (SARS-Cov2) lockdown, enterovirus outbreaks were once again detected, notably in young children. This reemergence highlights on the need to develop broad-spectrum treatment against enteroviruses. Over the last year, our research team has identified a new class of small-molecule inhibitors showing anti-EV activity. Targeting the well-known hydrophobic pocket in the viral capsid, these compounds show micromolar activity against EV-A71 and a high selectivity index (SI) (5h: EC50, MRC-5 = 0.57 μM, CC50, MRC-5 >20 μM, SI > 35; EC50, RD = 4.38 μM, CC50, RD > 40 μM, SI > 9; 6c: EC50, MRC-5 = 0.29 μM, CC50, MRC-5 >20 μM, SI > 69; EC50, RD = 1.66 μM, CC50, RD > 40 μM, SI > 24; Reference: Vapendavir EC50, MRC-5 = 0.36 μM, CC50, MRC-5 > 20 μM, EC50, RD = 0.53 μM, CC50, RD > 40 μM, SI > 63). The binding mode of these compounds in complex with enterovirus capsids was analyzed and showed a series of conserved interactions. Consequently, 6c and its derivatives are promising candidates for the treatment of enterovirus infections.
    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
    基于结构的药物设计(SBDD)的学科已有几十年的历史,人们很容易认为许多药物靶标的实验结构的增殖可能使计算机辅助药物设计(CADD)变得简单。然而,这远非真实。在这次审查中,我们说明了CADD科学家每天在工作中面临的一些挑战,即使是现在.我们使用Rho相关蛋白激酶(ROCK),公共领域的结构和数据,作为一个例子来说明我们在针对这种蛋白质的项目中遇到的一些挑战。我们希望这将有助于防止对CADD可以实现的不切实际的期望,并教育非CADD科学家关于他们的CADD同事仍然面临的挑战。
    The discipline of structure-based drug design (SBDD) is several decades old and it is tempting to think that the proliferation of experimental structures for many drug targets might make computer-aided drug design (CADD) straightforward. However, this is far from true. In this review, we illustrate some of the challenges that CADD scientists face every day in their work, even now. We use Rho-associated protein kinase (ROCK), and public domain structures and data, as an example to illustrate some of the challenges we have experienced during our project targeting this protein. We hope that this will help to prevent unrealistic expectations of what CADD can accomplish and to educate non-CADD scientists regarding the challenges still facing their CADD colleagues.
    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
    武田G蛋白偶联受体5(TGR5)被一级和二级胆汁酸内源性激活。该受体被认为是解决炎症和代谢紊乱的候选靶标。我们已经通过基于结构的方法来靶向TGR5,使用最近解决的实验结构来寻找配体,以及从分子动力学模拟中获得的结构。通过解决正构和假定的变构位点,我们鉴定了激动剂和正变构调节剂.虽然预测的结合位置与其功效不一致,我们的工作有助于激活我们在体外彻底表征的小分子配体。
    The Takeda G protein-coupled receptor 5 (TGR5) is activated endogenously by primary and secondary bile acids. This receptor is considered a candidate target for addressing inflammatory and metabolic disorders. We have targeted TGR5 with structure-based methods for ligand finding using the recently solved experimental structures, as well as structures obtained from molecular dynamics simulations. Through addressing the orthosteric as well as a putative allosteric site, we identified agonists and positive allosteric modulators. While the predicted binding locations were not in line with their efficacy, our work contributes activating small-molecule ligands that we have thoroughly characterized in vitro.
    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
    FGFRs的异常激活在各种癌症中起关键作用,导致几种FGFR抑制剂在临床上的发展。然而,耐药性的出现,主要是由于FGFRs中的看门人突变,限制了其临床疗效。为了解决未满足的医疗需求,设计并合成了一系列5-氨基-1H-吡唑-4-甲酰胺衍生物作为靶向野生型和看门人突变体的新型pan-FGFR共价抑制剂。代表性化合物10h在生化测定中表现出针对FGFR1、FGFR2、FGFR3和FGFR2V564F看门人突变体的纳摩尔活性(IC50=46、41、99和62nM)。此外,10h还强烈抑制了NCI-H520肺癌细胞的增殖,SNU-16和KATOIII胃癌细胞,IC50值分别为19、59和73nM,分别。进一步的X-射线共晶结构显示10h不可逆地结合FGFR1。该研究为FGFRs介导的抗癌药物开发提供了新的有希望的观点。
    The aberrant activation of FGFRs plays a critical role in various cancers, leading to the development of several FGFR inhibitors in clinic. However, the emergence of drug resistance, primarily due to gatekeeper mutations in FGFRs, has limited their clinical efficacy. To address the unmet medical need, a series of 5-amino-1H-pyrazole-4-carboxamide derivatives were designed and synthesized as novel pan-FGFR covalent inhibitors targeting both wild-type and the gatekeeper mutants. The representative compound 10h demonstrated nanomolar activities against FGFR1, FGFR2, FGFR3 and FGFR2 V564F gatekeeper mutant in biochemical assays (IC50 = 46, 41, 99, and 62 nM). Moreover, 10h also strongly suppressed the proliferation of NCI-H520 lung cancer cells, SNU-16 and KATO III gastric cancer cells with IC50 values of 19, 59, and 73 nM, respectively. Further X-ray co-crystal structure revealed that 10h irreversibly binds to FGFR1. The study provides a new promising point for anticancer drug development medicated by FGFRs.
    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
    作为全球卫生挑战,癌症促使人们不断探索基于新靶点的创新疗法。一个有希望的途径是针对shelterin蛋白复合物,端粒的保护对于防止DNA损伤至关重要。shelterin在调节共济失调-毛细血管扩张突变(ATM)和共济失调-毛细血管扩张和Rad3相关(ATR)激酶中的作用,DNA损伤反应(DDR)的关键参与者,确定其在癌细胞中的意义。破坏了Shelterins的防御机制,尤其是在癌细胞中,使端粒脆弱,可能导致基因组不稳定和阻碍癌细胞存活。在这次审查中,我们概述了最近探索shelterins作为潜在抗癌靶标的方法,突出了开发选择性分子的前景,以利用端粒脆弱性对新的创新癌症治疗。
    As a global health challenge, cancer prompts continuous exploration for innovative therapies that are also based on new targets. One promising avenue is targeting the shelterin protein complex, a safeguard for telomeres crucial in preventing DNA damage. The role of shelterin in modulating ataxia-telangiectasia mutated (ATM) and ataxia-telangiectasia and Rad3-related (ATR) kinases, key players in the DNA damage response (DDR), establishes its significance in cancer cells. Disrupting these defence mechanisms of shelterins, especially in cancer cells, renders telomeres vulnerable, potentially leading to genomic instability and hindering cancer cell survival. In this review, we outline recent approaches exploring shelterins as potential anticancer targets, highlighting the prospect of developing selective molecules to exploit telomere vulnerabilities toward new innovative cancer treatments.
    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
    访问小,刚性,富含sp3的分子是挑战蛋白质靶标的药物发现的主要限制。FK506结合蛋白作为药物靶标或分子胶的推动者具有很高的潜力,但在被接受为配体的化学型中却很挑剔。我们在这里报道了一种高度刚性化的模仿三环支架的对映选择性合成,该支架精确定位了与FKBP相互作用的官能团。这是通过以阳极氧化为特征的14步克级合成实现的,立体定向乙烯基化,和N-酰基亚胺环化。基于结构的优化导致发现了具有皮摩尔生化和亚纳摩尔细胞活性的FKBP抑制剂,其代表了迄今为止已知的最有效的FKBP配体。
    Access to small, rigid, and sp3-rich molecules is a major limitation in the drug discovery for challenging protein targets. FK506-binding proteins hold high potential as drug targets or enablers of molecular glues but are fastidious in the chemotypes accepted as ligands. We here report an enantioselective synthesis of a highly rigidified pipecolate-mimicking tricyclic scaffold that precisely positions functional groups for interacting with FKBPs. This was enabled by a 14-step gram-scale synthesis featuring anodic oxidation, stereospecific vinylation, and N-acyl iminium cyclization. Structure-based optimization resulted in the discovery of FKBP inhibitors with picomolar biochemical and subnanomolar cellular activity that represent the most potent FKBP ligands known to date.
    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
    G蛋白偶联受体(GPCRs)在细胞外信号的转导和许多生物过程的调节中起着关键作用,这使得这些膜蛋白成为药物最重要的靶标之一。GPCRs的分辨原子结构的数量的显著增加开启了通过基于结构的药物设计(SBDD)开发靶向这些受体的药物的可能性。SBDD利用有关受体-配体复合物结构的信息来搜索选择性配体,而无需进行广泛的高通量实验配体筛选,并且可以显着扩展用于配体搜索的化学空间。在这次审查中,我们描述了使用X射线衍射分析和低温电子显微镜破译GPCR结构的过程,这是合理设计针对该受体类别的药物的重要阶段。我们的主要目标是向广泛的专家介绍GPCR靶向剂SBDD中使用的实验方法的现代发展和关键特征。
    G protein-coupled receptors (GPCRs) play a key role in the transduction of extracellular signals to cells and regulation of many biological processes, which makes these membrane proteins one of the most important targets for pharmacological agents. A significant increase in the number of resolved atomic structures of GPCRs has opened the possibility of developing pharmaceuticals targeting these receptors via structure-based drug design (SBDD). SBDD employs information on the structure of receptor-ligand complexes to search for selective ligands without the need for an extensive high-throughput experimental ligand screening and can significantly expand the chemical space for ligand search. In this review, we describe the process of deciphering GPCR structures using X-ray diffraction analysis and cryoelectron microscopy as an important stage in the rational design of drugs targeting this receptor class. Our main goal was to present modern developments and key features of experimental methods used in SBDD of GPCR-targeting agents to a wide range of specialists.
    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
    PAX8/PPARγ重排,产生PAX8-PPARγ融合蛋白(PPFP),被认为在甲状腺滤泡性肿瘤的发生中起着至关重要的作用。为了确定PPFP靶向的候选药物,并建立甲状腺肿瘤的早期护理标准,我们进行了基于集合对接的化合物筛选.具体来说,我们研究了为PPFP寻找有希望的配体化合物应采用的口袋结构;PPFPPPARγ侧的配体结合口袋的位置与PPARγ相似;然而,由于环境因素,它们之间的形状略有不同。我们开发了一种选择具有相关口袋和高预测精度的PPFP结构的方法,用于配体结合。该方法用PPARγ进行了验证,其结构和活性值对于许多化合物是已知的。然后,我们对在DrugBank数据库中注册的具有噻唑烷主链的97种药物或药物样化合物的PPFP进行了对接计算,这是与PPARγ良好结合的配体的特征之一。此外,使用分子力学泊松-玻尔兹曼表面积方法更可靠地估计了有希望的配体候选物的结合亲和力。因此,我们提出了具有噻唑烷主链的PPFP的有希望的候选药物。
    The PAX8/PPARγ rearrangement, producing the PAX8-PPARγ fusion protein (PPFP), is thought to play an essential role in the oncogenesis of thyroid follicular tumors. To identify PPFP-targeted drug candidates and establish an early standard of care for thyroid tumors, we performed ensemble-docking-based compound screening. Specifically, we investigated the pocket structure that should be adopted to search for a promising ligand compound for the PPFP; the position of the ligand-binding pocket on the PPARγ side of the PPFP is similar to that of PPARγ; however, the shape is slightly different between them due to environmental factors. We developed a method for selecting a PPFP structure with a relevant pocket and high prediction accuracy for ligand binding. This method was validated using PPARγ, whose structure and activity values are known for many compounds. Then, we performed docking calculations to the PPFP for 97 drug or drug-like compounds registered in the DrugBank database with a thiazolidine backbone, which is one of the characteristics of ligands that bind well to PPARγ. Furthermore, the binding affinities of promising ligand candidates were estimated more reliably using the molecular mechanics Poisson-Boltzmann surface area method. Thus, we propose promising drug candidates for the PPFP with a thiazolidine backbone.
    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
    这篇综述旨在强调ADAR蛋白的结构,这些结构对识别其功能至关重要,并与未来的治疗发展有关。ADAR蛋白可以纠正或多样化遗传信息,强调了它们对蛋白质多样性和神经元网络复杂性的关键贡献。ADAR蛋白在RNA编辑中具有许多独立的功能,并通过A-IRNA编辑的机制不断被揭示。提供了对ADAR家族成员-ADAR1,ADAR2和ADAR3-各自的特征在于提供结构多样性和功能变异性的不同同种型的详细检查。显着影响RNA编辑机制并表现出组织特异性调控模式,突出它们的共同特征,例如双链RNA结合结构域(dsRBD)和催化脱氨酶结构域(CDD)。此外,它探讨了ADAR在免疫中的广泛作用,RNA干扰,和疾病调制,证明了它们在疾病的发展和抑制方面的矛盾性质。通过全面的分析,这篇综述旨在强调在治疗策略中靶向ADAR蛋白的潜力,敦促继续调查其生物学机制和健康影响。
    This review aims to highlight the structures of ADAR proteins that have been crucial in the discernment of their functions and are relevant to future therapeutic development. ADAR proteins can correct or diversify genetic information, underscoring their pivotal contribution to protein diversity and the sophistication of neuronal networks. ADAR proteins have numerous functions in RNA editing independent roles and through the mechanisms of A-I RNA editing that continue to be revealed. Provided is a detailed examination of the ADAR family members-ADAR1, ADAR2, and ADAR3-each characterized by distinct isoforms that offer both structural diversity and functional variability, significantly affecting RNA editing mechanisms and exhibiting tissue-specific regulatory patterns, highlighting their shared features, such as double-stranded RNA binding domains (dsRBD) and a catalytic deaminase domain (CDD). Moreover, it explores ADARs\' extensive roles in immunity, RNA interference, and disease modulation, demonstrating their ambivalent nature in both the advancement and inhibition of diseases. Through this comprehensive analysis, the review seeks to underline the potential of targeting ADAR proteins in therapeutic strategies, urging continued investigation into their biological mechanisms and health implications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号