Dual-target inhibitor

  • 文章类型: Journal Article
    双特异性酪氨酸磷酸化调节激酶2(DYRK2)和组蛋白脱乙酰酶8(HDAC8)已显示与几种癌症的发展有关。这里,我们通过联合虚拟筛选方案鉴定了双靶点DYRK2/HDAC8抑制剂(DYC-1).DYC-1表现出对DYRK2(IC50=5.27±0.13nM)和HDAC8(IC50=8.06±0.47nM)的纳摩尔抑制活性。分子动力学模拟显示DYC-1与DYRK2和HDAC8具有正的结合稳定性。重要的是,细胞毒性实验表明DYC-1对人肝癌具有优越的抗增殖活性,尤其是SK-HEP-1细胞,对正常肝细胞无明显抑制作用。此外,DYC-1对SK-HEP-1异种移植瘤的生长表现出较强的抑制作用,且无明显副作用。这些数据表明DYC-1是用于治疗肝细胞癌的高效低毒性抗肿瘤剂。
    Dual-specificity tyrosine phosphorylation-regulated kinase 2 (DYRK2) and histone deacetylase 8 (HDAC8) have been shown to be associated with the development of several cancers. Here, we identified a dual-target DYRK2/HDAC8 inhibitor (DYC-1) through a combined virtual screening protocol. DYC-1 exhibited nanomolar inhibitory activity against both DYRK2 (IC50 = 5.27 ± 0.13 nM) and HDAC8 (IC50 = 8.06 ± 0.47 nM). Molecular dynamics simulations showed that DYC-1 had positive binding stability with DYRK2 and HDAC8. Importantly, the cytotoxicity assay indicated that DYC-1 exhibited superior antiproliferative activity against human liver cancer, especially SK-HEP-1 cells, and had no significant inhibition on normal liver cells. Moreover, DYC-1 showed a strong inhibitory effect on the growth of SK-HEP-1 xenograft tumors with no significant side effects. These data suggest that DYC-1 is a high-efficacy and low-toxic antitumor agent for the treatment of hepatocellular carcinoma.
    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
    本研究旨在开发第一个同时靶向盘状结构域受体1(DDR1)和表皮生长因子受体(EGFR)的双靶点小分子抑制剂。在非小细胞肺癌(NSCLC)中起着至关重要的相互依赖的作用,表现出协同抑制作用。发现了一系列针对DDR1和EGFR的创新双靶点抑制剂。这些化合物是使用基于先导化合物BZF02的结构优化策略设计和合成的,采用4,6-嘧啶二胺作为核心支架,随后对其生物活动进行了调查。在这些化合物中,选择D06并显示针对DDRl和EGFR的微摩尔酶效力。随后,观察到化合物D06抑制NSCLC细胞增殖和侵袭。证明可接受的药代动力学性能,化合物D06在NSCLCPC-9/GR异种移植模型中表现出其抗肿瘤活性,没有明显的毒性或明显的体重减轻。这些集体结果展示了一种有效的双靶向抑制剂的成功合成,提示联合靶向DDR1和EGFR治疗DDR1/EGFR阳性NSCLC的潜在疗效。
    This study aimed to develop the first dual-target small molecule inhibitor concurrently targeting Discoidin domain receptor 1 (DDR1) and Epidermal growth factor receptor (EGFR), which play a crucial interdependent roles in non-small cell lung cancer (NSCLC), demonstrating a synergistic inhibitory effect. A series of innovative dual-target inhibitors for DDR1 and EGFR were discovered. These compounds were designed and synthesized using structural optimization strategies based on the lead compound BZF02, employing 4,6-pyrimidine diamine as the core scaffold, followed by an investigation of their biological activities. Among these compounds, D06 was selected and showed micromolar enzymatic potencies against DDR1 and EGFR. Subsequently, compound D06 was observed to inhibit NSCLC cell proliferation and invasion. Demonstrating acceptable pharmacokinetic performance, compound D06 exhibited its anti-tumor activity in NSCLC PC-9/GR xenograft models without apparent toxicity or significant weight loss. These collective results showcase the successful synthesis of a potent dual-targeted inhibitor, suggesting the potential therapeutic efficacy of co-targeting DDR1 and EGFR for DDR1/EGFR-positive NSCLC.
    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
    目的:在B细胞急性淋巴细胞白血病(B-ALL)中,目前,成人患者的强化化疗在超过50%的病例中未能达到持久的反应,强调该患者人群迫切需要新的治疗方案。本研究旨在确定HZX-02-059,一种靶向磷脂酰肌醇-3-磷酸5-激酶(PIKfyve)和微管蛋白的新型双靶点抑制剂,对B-ALL细胞是致命的,是B-ALL患者的潜在治疗方法。
    方法:细胞增殖,真空化,凋亡,细胞周期,和体内肿瘤生长进行了评估。此外,进行全基因组RNA测序研究以阐明HZX-02-059在B-ALL中抗白血病活性的作用机制。
    结果:发现HZX-02-059抑制细胞增殖,诱导空泡化,促进细胞凋亡,阻断细胞周期,减少体内肿瘤生长。p53途径的下调和磷酸肌醇3-激酶(PI3K)/AKT途径和下游转录因子c-Myc和NF-κB的抑制是这些观察结果的原因。
    结论:总体而言,这些研究结果表明,HZX-02-059是治疗对常规疗法耐药的B-ALL患者的有前景的药物.
    OBJECTIVE: In B-cell acute lymphoblastic leukemia (B-ALL), current intensive chemotherapies for adult patients fail to achieve durable responses in more than 50% of cases, underscoring the urgent need for new therapeutic regimens for this patient population. The present study aimed to determine whether HZX-02-059, a novel dual-target inhibitor targeting both phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) and tubulin, is lethal to B-ALL cells and is a potential therapeutic for B-ALL patients.
    METHODS: Cell proliferation, vacuolization, apoptosis, cell cycle, and in-vivo tumor growth were evaluated. In addition, Genome-wide RNA-sequencing studies were conducted to elucidate the mechanisms of action underlying the anti-leukemia activity of HZX-02-059 in B-ALL.
    RESULTS: HZX-02-059 was found to inhibit cell proliferation, induce vacuolization, promote apoptosis, block the cell cycle, and reduce in-vivo tumor growth. Downregulation of the p53 pathway and suppression of the phosphoinositide 3-kinase (PI3K)/AKT pathway and the downstream transcription factors c-Myc and NF-κB were responsible for these observations.
    CONCLUSIONS: Overall, these findings suggest that HZX-02-059 is a promising agent for the treatment of B-ALL patients resistant to conventional therapies.
    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
    磷酸肌醇3激酶(PI3K)通过肌醇磷脂在3'-OH位置的磷酸化修饰脂质,从而参与信号转导并对细胞生长等各种生理过程产生影响,新陈代谢,和有机体的发展。PI3K激活也驱动癌细胞生长,生存,和新陈代谢,在不同的人类癌症中观察到该途径的遗传失调。因此,该靶标被认为是各种类型癌症的有希望的潜在治疗靶标.目前,几种选择性PI3K抑制剂和一种双靶点PI3K抑制剂已被批准上市。然而,这些抑制剂中的大多数由于担心其不良反应而面临撤销或自愿撤销适应症.本文对其结构和生物学功能进行了全面的综述,和PI3K抑制剂的临床状况,特别强调双靶点PI3K抑制剂的开发策略和构效关系。这些发现为开发极具前景的靶向PI3K的双靶点药物提供了有价值的见解和未来方向。
    Phosphoinositide 3-kinases (PI3Ks) modify lipids by the phosphorylation of inositol phospholipids at the 3\'-OH position, thereby participating in signal transduction and exerting effects on various physiological processes such as cell growth, metabolism, and organism development. PI3K activation also drives cancer cell growth, survival, and metabolism, with genetic dysregulation of this pathway observed in diverse human cancers. Therefore, this target is considered a promising potential therapeutic target for various types of cancer. Currently, several selective PI3K inhibitors and one dual-target PI3K inhibitor have been approved and launched on the market. However, the majority of these inhibitors have faced revocation or voluntary withdrawal of indications due to concerns regarding their adverse effects. This article provides a comprehensive review of the structure and biological functions, and clinical status of PI3K inhibitors, with a specific emphasis on the development strategies and structure-activity relationships of dual-target PI3K inhibitors. The findings offer valuable insights and future directions for the development of highly promising dual-target drugs targeting PI3K.
    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
    由于酪氨酸激酶抑制剂与微管靶向药物在抗肿瘤治疗中的临床成功,本文提出了一种新型的双靶标抑制剂的组合靶向设计,具有芳基甲酰脲偶联喹唑啉主链。设计了一系列目标化合物(10a-10r),合成,和特点。生物学评估表明,10c在体外显着增强了10种肿瘤细胞系,IC50值范围为1.04µM至7.66µM。重要的是,10c(IC50=10.66nM)对EGFR激酶的抑制活性优于参考药物吉非替尼(25.42nM),EGFR磷酸化水平降低,AKT,和ERK.此外,10c显著阻碍微管蛋白聚合,破坏了A549细胞的细胞内微管网络,诱导细胞凋亡,导致S期细胞周期停滞,阻碍细胞迁移。在使用A549荷癌裸鼠模型的抗癌评估测试中,10c显示出与吉非替尼相似的治疗效果,但只需要一半的剂量(15毫克/千克)。这些发现表明化合物10c是抗癌疗法的有希望的双靶标候选物。
    Motivated by the clinical success of combining tyrosine kinase inhibitors with microtubule-targeted drugs in antitumor treatment, this paper presents a novel combi-targeting design for dual-target inhibitors, featuring arylformylurea-coupled quinazoline backbones. A series of target compounds (10a-10r) were designed, synthesized, and characterized. Biological assessments demonstrated that 10c notably potentiated ten tumor cell lines in vitro, with IC50 values ranging from 1.04 µM to 7.66 µM. Importantly, 10c (IC50 = 10.66 nM) exhibited superior inhibitory activity against EGFR kinases compared to the reference drug Gefitinib (25.42 nM) and reduced phosphorylated levels of EGFR, AKT, and ERK. Moreover, 10c significantly impeded tubulin polymerization, disrupted the intracellular microtubule network in A549 cells, induced apoptosis, led to S-phase cell cycle arrest, and hindered cell migration. In anticancer evaluation tests using A549 cancer-bearing nude mice models, 10c showed a therapeutic effect similar to Gefitinib, but required only half the dosage (15 mg/kg). These findings indicate that compound 10c is a promising dual-target candidate for anticancer therapy.
    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
    COX-2抑制剂构成了一类抗炎镇痛药,显示出对抗某些类型癌症的潜力。然而,这些抑制剂与心血管毒性有关。此外,尽管单靶标分子对特定靶标具有特异性,它们经常导致安全性差,由于代偿机制的低疗效和耐药性。同时抑制COX-2和另一个靶标的新一代双靶标药物显示出治疗癌症或减少不良心脏作用的强大潜力。本观点关注COX-2的结构和功能及其作为治疗靶标的作用。它还从药物化学的角度探讨了双目标策略的当前状态和未来可能性。
    Inhibitors of COX-2 constitute a class of anti-inflammatory analgesics, showing potential against certain types of cancer. However, such inhibitors are associated with cardiovascular toxicity. Moreover, although single-target molecules possess specificity for particular targets, they often lead to poor safety, low efficacy and drug resistance due to compensatory mechanisms. A new generation of dual-target drugs that simultaneously inhibit COX-2 and another target is showing strong potential to treat cancer or reduce adverse cardiac effects. The present perspective focuses on the structure and functions of COX-2, and its role as a therapeutic target. It also explores the current state and future possibilities for dual-target strategies from a medicinal chemistry perspective.
    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
    三阴性乳腺癌(TNBC)是最具侵袭性的乳腺肿瘤之一,复发和转移率高,预后差。因此,寻找新的靶向治疗策略和开发相应的药物已迫在眉睫。先前的研究表明,CDK12抑制剂与PARP1抑制剂的组合能够在TNBC细胞中诱导合成致死性。这里,我们报道了通过遗传或药理学方法同时抑制CDK12和PARP1可协同抑制TNBC细胞的增殖.然后,设计并合成了一系列靶向CDK12和PARP1的小分子抑制剂。新的双靶点抑制剂(12e)对CDK12(IC50=285nM)和PARP1(IC50=34nM)显示出有效的抑制活性,以及在TNBC细胞系中良好的抗增殖作用。同时,化合物12e通过抑制DNA损伤修复在细胞和异种移植物中显示出良好的协同抗肿瘤功效,促进细胞周期阻滞和凋亡。一起来看,我们成功合成了第一个有效的CDK12-PARP1双重抑制剂,这有望成为TNBC有吸引力的治疗策略。
    Triple negative breast cancer (TNBC) is one of the most aggressive breast tumors, with a high rate of recurrence and metastasis as well as a poor prognosis. Consequently, it is urgent to find new targeted therapeutic strategies and development of corresponding drugs. Previous studies have shown that CDK12 inhibitors in combination with PARP1 inhibitors is able to induce synthetic lethality in TNBC cells. Here, we reported simultaneously inhibition of CDK12 and PARP1 by genetic or pharmacological approaches synergistically inhibited the proliferation of TNBC cells. Then, a series of small molecule inhibitors targeting both CDK12 and PARP1 were designed and synthesized. The new dual-target inhibitor (12e) showed potent inhibitory activity against CDK12 (IC50 = 285 nM) and PARP1 (IC50 = 34 nM), as well as good anti-proliferative effects in TNBC cell lines. Meanwhile, compound 12e showed favorable synergistic anti-tumor efficacy in cells and xenografts by inhibiting DNA damage repair, promoting cell cycle arrest and apoptosis. Taken together, we successfully synthesized the first effective CDK12-PARP1 dual inhibitor, which is expected to be an attractive therapeutic strategy for TNBC.
    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
    与别嘌醇合用,曲尼司特用作尿酸转运蛋白1(URAT1)抑制剂,用于治疗高尿酸血症,但是很少研究其与URAT1抑制活性有关的构效关系。在本文中,在曲尼司特和特权支架吲哚的基础上,使用支架跳跃策略设计和合成类似物1-30。然后,使用14C-尿酸摄取测定用HEK293-URAT1过表达细胞评估URAT1活性。与曲尼司特相比(10μM时抑制率=44.9%),大多数化合物表现出明显的抑制作用,在URAT1上10μM时范围从40.0%到81.0%。令人惊讶的是,随着在吲哚环的5位引入氰基,化合物26和28-30具有黄嘌呤氧化酶(XO)抑制活性。特别是,化合物29对URAT1(10μM时为48.0%)和XO(IC50=1.01μM)具有效力。分子模拟分析表明化合物29的基本结构与URAT1和XO具有亲和力。此外,化合物29在口服剂量为10mg/kg的含氧酸钾诱导的高尿酸血症大鼠模型中显示出显着的低尿酸作用。总之,曲尼司特类似物29被鉴定为URAT1和XO的有效双靶标抑制剂,和一个有希望的铅化合物进一步调查。
    In combination with allopurinol, tranilast is used as an urate transporter 1 (URAT1) inhibitor for the treatment of hyperuricemia, but its structure-activity relationship concerning URAT1 inhibitory activity is rarely studied. In this paper, analogs 1-30 were designed and synthesized using scaffold hopping strategy on the basis of tranilast and the privileged scaffold indole. Then, URAT1 activity was evaluated using 14C-uric acid uptake assay with HEK293-URAT1 overexpressing cells. Compared with tranilast (inhibitory rate = 44.9% at 10 μM), most compounds displayed apparent inhibitory effects, ranging from 40.0% to 81.0% at 10 μM on URAT1. Surprisingly, along with the bringing in of a cyano group at the 5-position of indole ring, compounds 26 and 28-30 exerted xanthine oxidase (XO) inhibitory activity. In particular, compound 29 presented potency on URAT1 (48.0% at 10 μM) and XO (IC50 = 1.01 μM). Molecular simulation analysis revealed that the basic structure of compound 29 had an affinity with URAT1, and XO. Furthermore, compound 29 demonstrated a significant hypouricemic effect in a potassium oxonate-induced hyperuricemia rat model at an oral dose of 10 mg/kg during in vivo tests. In summary, tranilast analog 29 was identified as a potent dual-target inhibitor of URAT1 and XO, and a promising lead compound for further investigation.
    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
    多靶点药物开发已成为发现治疗阿尔茨海默病(AzD)药物的一个有吸引力的策略。在这项研究中,第一次,利用分类树(CT)的基于规则的机器学习(ML)方法用于新型双靶点乙酰胆碱酯酶(AChE)和β位点淀粉样蛋白前体裂解酶1(BACE1)抑制剂的合理设计.来自具有AChE和BACE1测量值的3524种化合物的更新数据从ChEMBL数据库进行整理。AChE和BACE1的训练/外部验证的最佳全局准确性分别为0.85/0.80和0.83/0.81。然后将规则应用于从原始数据库中筛选双重抑制剂。根据从每个分类树获得的最佳规则,确定了一组潜在的AChE和BACE1抑制剂,并使用Murcko型分解分析提取活性片段。使用一致的QSAR模型和对接验证,基于活性片段和预测的AChE和BACE1抑制活性,在计算机中设计了超过250种新型抑制剂。本研究中应用的基于规则的方法和ML方法可能有助于针对AzD的新型AChE和BACE1双重抑制剂的设计和筛选。
    Multi-target drug development has become an attractive strategy in the discovery of drugs to treat of Alzheimer\'s disease (AzD). In this study, for the first time, a rule-based machine learning (ML) approach with classification trees (CT) was applied for the rational design of novel dual-target acetylcholinesterase (AChE) and β-site amyloid-protein precursor cleaving enzyme 1 (BACE1) inhibitors. Updated data from 3524 compounds with AChE and BACE1 measurements were curated from the ChEMBL database. The best global accuracies of training/external validation for AChE and BACE1 were 0.85/0.80 and 0.83/0.81, respectively. The rules were then applied to screen dual inhibitors from the original databases. Based on the best rules obtained from each classification tree, a set of potential AChE and BACE1 inhibitors were identified, and active fragments were extracted using Murcko-type decomposition analysis. More than 250 novel inhibitors were designed in silico based on active fragments and predicted AChE and BACE1 inhibitory activity using consensus QSAR models and docking validations. The rule-based and ML approach applied in this study may be useful for the in silico design and screening of new AChE and BACE1 dual inhibitors against AzD.
    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
    拓扑异构酶II(TopoII)和组蛋白脱乙酰酶(HDAC)都是癌症的重要治疗靶标。在这项研究中,设计并合成了两个含有嘧啶并[5,4-b]吲哚和吡唑并[3,4-d]嘧啶基序的新型化合物作为TopoII/HDAC双重抑制剂。MTT分析表明,所有化合物对三种癌细胞系(MGC-803,MCF-7和U937)均具有潜在的抗增殖活性,对正常细胞系(3T3)具有较低的细胞毒性。在酶活性抑制实验中,化合物7d和8d对TopoⅡ和HDAC表现出优异的双重抑制活性。裂解反应实验显示7d为TopoII毒物,这与对接结果一致。进一步的实验结果表明,化合物7d和8d可以促进MCF-7细胞的凋亡并显着抑制其迁移。分子对接显示化合物7d和8d在活性位点结合TopoII和HDAC。分子动力学模拟表明7d能稳定地结合TopoⅡ和HDAC。
    Both topoisomerase II (Topo II) and histone deacetylase (HDAC) are important therapeutic targets for cancer. In this study, two series of novel compounds containing pyrimido[5,4-b]indole and pyrazolo[3,4-d]pyrimidine motifs were designed and synthesized as dual Topo II/HDAC inhibitors. MTT assay indicated that all the compounds displayed potential antiproliferative activity against three cancer cell lines (MGC-803, MCF-7 and U937) and low cytotoxicity on normal cell line (3T3). In the enzyme activity inhibition experiments, compounds 7d and 8d exhibited excellent dual inhibitory activities against Topo II and HDAC. Cleavage reaction assay showed that 7d was a Topo II poison, which was consistent with the docking results. Further experimental results revealed that compounds 7d and 8d could promote apoptosis and significantly inhibit the migration in MCF-7 cells. Molecular docking showed that compounds 7d and 8d bind Topo II and HDAC at the active sites. Molecular dynamics simulation showed that 7d can stably bind to Topo II and HDAC.
    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