Small molecule inhibitors

小分子抑制剂
  • 文章类型: Journal Article
    蛋白酪氨酸磷酸酶非受体2型(PTPN2)是一种去磷酸化具有酪氨酸残基的蛋白质的酶,从而在体内调节相关的信号通路。取决于上下文,PTPN2充当肿瘤抑制因子或肿瘤启动子。在某些癌症中,比如结直肠,肺癌,PTPN2缺陷可能损害蛋白酪氨酸激酶途径,通常在癌细胞中过度激活,并抑制肿瘤的发展和进展。然而,PTPN2还可以通过调节免疫细胞和细胞因子来抑制肿瘤免疫。结构,功能,本文综述了PTPN2在各种肿瘤细胞中的底物。综述了PTPN2小分子抑制剂和降解剂的研究现状。它还强调了开发PTPN2抑制剂作为抗癌药物的潜在机遇和挑战。
    Protein tyrosine phosphatase nonreceptor type 2 (PTPN2) is an enzyme that dephosphorylates proteins with tyrosine residues, thereby modulating relevant signaling pathways in vivo. PTPN2 acts as tumor suppressor or tumor promoter depending on the context. In some cancers, such as colorectal, and lung cancer, PTPN2 defects could impair the protein tyrosine kinase pathway, which is often over-activated in cancer cells, and inhibit tumor development and progression. However, PTPN2 can also suppress tumor immunity by regulating immune cells and cytokines. The structure, functions, and substrates of PTPN2 in various tumor cells were reviewed in this paper. And we summarized the research status of small molecule inhibitors and degraders of PTPN2. It also highlights the potential opportunities and challenges for developing PTPN2 inhibitors as anticancer drugs.
    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
    而受体酪氨酸激酶样孤儿受体1(ROR1)通常在正常组织中低水平表达或不存在,它的表达在各种恶性肿瘤和病症中显著升高,包括慢性淋巴细胞白血病(CLL),乳腺癌,卵巢癌,黑色素瘤,和肺腺癌。这一显著特征将ROR1定位为肿瘤特异性治疗的有吸引力的靶标。目前,几种针对ROR1的靶向药物正在进行临床开发,包括单克隆抗体,抗体-药物偶联物(ADC),和嵌合抗原受体T细胞疗法(CAR-T)。此外,有四种小分子抑制剂被设计为与ROR1结合,为开发靶向ROR1的PROTAC降解剂提供了有希望的途径。这篇综述提供了对ROR1结构和功能特征的最新见解,胚胎发育的影响,细胞存活信号通路,和进化目标策略,所有这些都有可能推进恶性肿瘤的治疗。
    While receptor tyrosine kinase-like orphan receptor 1 (ROR1) is typically expressed at low levels or absent in normal tissues, its expression is notably elevated in various malignant tumors and conditions, including chronic lymphocytic leukemia (CLL), breast cancer, ovarian cancer, melanoma, and lung adenocarcinoma. This distinctive feature positions ROR1 as an attractive target for tumor-specific treatments. Currently, several targeted drugs directed at ROR1 are undergoing clinical development, including monoclonal antibodies, antibody-drug conjugates (ADCs), and chimeric antigen receptor T-cell therapy (CAR-T). Additionally, there are four small molecule inhibitors designed to bind to ROR1, presenting promising avenues for the development of PROTAC degraders targeting ROR1. This review offers updated insights into ROR1\'s structural and functional characteristics, embryonic development implications, cell survival signaling pathways, and evolutionary targeting strategies, all of which have the potential to advance the treatment of malignant tumors.
    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
    动脉粥样硬化(AS)是各种心血管疾病背后的主要病理和全球死亡和残疾的主要原因。最近的研究表明,AS是一种由多种因素引起的慢性血管炎症性疾病。在这种情况下,NLRP3炎性体,作为免疫系统的信号传感器,在AS的发生和进展中起着至关重要的作用。NLRP3炎性体参与内皮损伤,泡沫细胞的形成,AS中的焦亡。因此,靶向NLRP3炎性体为AS提供了一种新的治疗策略.这篇综述强调了对AS发病机制和靶向NLRP3炎性体的药物治疗的最新见解,专注于小分子抑制剂的最佳靶标。这些见解对于合理的药物设计和新的靶向NLRP3炎性体抑制剂治疗AS的药理学评估是有价值的。
    Atherosclerosis (AS) is the primary pathology behind various cardiovascular diseases and the leading cause of death and disability globally. Recent evidence suggests that AS is a chronic vascular inflammatory disease caused by multiple factors. In this context, the NLRP3 inflammasome, acting as a signal transducer of the immune system, plays a critical role in the onset and progression of AS. The NLRP3 inflammasome is involved in endothelial injury, foam cell formation, and pyroptosis in AS. Therefore, targeting the NLRP3 inflammasome offers a new treatment strategy for AS. This review highlights the latest insights into AS pathogenesis and the pharmacological therapies targeting the NLRP3 inflammasome, focusing on optimal targets for small molecule inhibitors. These insights are valuable for rational drug design and the pharmacological assessment of new targeted NLRP3 inflammasome inhibitors in treating AS.
    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
    小分子抑制剂(SMI)越来越多地用于非小细胞肺癌的治疗。为了支持药代动力学研究和临床治疗监测,我们的目标是开发和验证一种超高效液相色谱-质谱(UPLC-MS/MS)测定,用于定量八个SMI:adagrasib,阿列替尼,布加替尼,卡马替尼,克唑替尼,洛拉替尼,selpercatinib,还有Sotorasib.通过尝试不同的柱和洗脱液以优化峰形状来进行UPLC-MS/MS测定的开发。该测定基于欧洲药品管理局的指南进行验证。使用甲酸铵在水和甲醇中的梯度洗脱进行色谱分离。使用具有电喷雾电离的三重四极杆串联质谱仪进行检测。氯拉替尼在10-2500μg/L的范围内进行验证,阿来替尼和克唑替尼为25-6250μg/L,25-10,000μg/L的卡马替尼和selpercatinib,50-12,500μg/L的布格替尼,和100-25,000μg/L的阿达格拉西布和索托拉西布。对于所有化合物,不精确性<8.88%并且不精确性<12.5%。8个化合物中的7个在室温下稳定96小时。Sotorasib在室温下稳定8小时。已开发出一种灵敏可靠的方法,可通过一次测定来量化八个SMI,提高靶向治疗的疗效和安全性。
    Small molecule inhibitors (SMIs) are increasingly being used in the treatment of non-small cell lung cancer. To support pharmacokinetic research and clinical treatment monitoring, our aim was to develop and validate an ultra-performance liquid chromatography-mass spectrometry (UPLC-MS/MS) assay for quantification of eight SMIs: adagrasib, alectinib, brigatinib, capmatinib, crizotinib, lorlatinib, selpercatinib, and sotorasib. Development of the UPLC-MS/MS assay was done by trying different columns and eluents to optimize peak shape. The assay was validated based on guidelines of the European Medicines Agency. Chromatographic separation was performed with a gradient elution using ammonium formate in water and methanol. Detection was performed using a triple quadrupole tandem mass spectrometer with electrospray ionization. Validation was performed in a range of 10-2500 μg/L for lorlatinib, 25-6250 μg/L for alectinib and crizotinib, 25-10,000 μg/L for capmatinib and selpercatinib, 50-12,500 μg/L for brigatinib, and 100-25,000 μg/L for adagrasib and sotorasib. Imprecision was <8.88% and inaccuracy was <12.5% for all compounds. Seven out of eight compounds were stable for 96 h at room temperature. Sotorasib was stable for 8 h at room temperature. A sensitive and reliable method has been developed to quantify eight SMIs with a single assay, enhancing efficacy and safety of targeted 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
    热休克蛋白90α(Hsp90α)是一种大量表达且进化保守的分子伴侣。Hsp90α是诱导型Hsp90亚型,并且其在细胞外的表达和分泌(eHsp90α)可以响应于多种细胞应激而被触发,以保护/激活客户蛋白并促进细胞对应激的调节。因此,癌症通常具有高表达水平的细胞内和细胞外(血浆)Hsp90α,允许他们支持他们的肿瘤发生和进展。事实上,(e)Hsp90α参与调节过程,如细胞信号传导,DNA修复,促进上皮-间质转化(EMT),促进血管生成,免疫反应,和细胞迁移。在几种癌症中,Hsp90α水平与癌症进展和严重程度相关。表明它可能是癌症的有用生物标志物或药物靶标。迄今为止,细胞内Hsp90α靶向治疗的发展包括标准的N端ATP竞争性抑制剂和与Hsp90α的中间或C端结构域结合的变构调节因子。作为Hsp90α抑制结果的目标毒性和给药并发症推动了eHsp90α靶向疗法的发展。实例包括抗Hsp90α单克隆抗体和细胞不可渗透的Hsp90α小分子抑制剂。这篇综述旨在讨论Hsp90α在癌症进展中的许多作用,重点是Hsp90α靶向治疗的当前发展。
    Heat shock protein 90 alpha (Hsp90α) is an abundantly expressed and evolutionarily conserved molecular chaperone. Hsp90α is the inducible Hsp90 isoform, and its expression and secretion extracellularly (eHsp90α) can be triggered in response to a variety of cellular stresses to protect/activate client proteins and to facilitate cellular adjustment to the stress. As a result, cancers often have high expression levels of intracellular and extracellular (plasma) Hsp90α, allowing them to support their oncogenesis and progression. In fact, (e)Hsp90α has been implicated in regulating processes such as cell signaling transduction, DNA repair, promotion of the Epithelial-to-Mesenchymal Transition (EMT), promotion of angiogenesis, immune response, and cell migration. Hsp90α levels have been correlated with cancer progression and severity in several cancers, indicating that it may be a useful biomarker or drug-target for cancer. To date, the development of intracellular Hsp90α-targeted therapies include standard N-terminal ATP-competitive inhibitors and allosteric regulators that bind to Hsp90α\'s middle or C-terminal domain. On-target toxicities and dosing complications as a result of Hsp90α inhibition has driven the development of eHsp90α-targeted therapies. Examples include anti-Hsp90α monoclonal antibodies and cell-impermeable Hsp90α small molecule inhibitors. This review aims to discuss the many roles Hsp90α plays in cancer progression with a focus on the current development of Hsp90α-targeted therapies.
    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
    尽管在癌症预防方面取得了重大进展,筛选,和治疗,仍然有限的治疗选择是提高癌症治愈率的障碍。近年来,为了开发选择性靶向致癌Wnt/β-catenin信号通路的不同组分的治疗方法,人们进行了许多努力。这些包括小分子抑制剂,抗体,最近,基于基因的方法。尽管其中一些在临床试验中显示出有希望的结果,Wnt/β-catenin通路在癌症治疗的常规临床实践中仍未被靶向。至于大多数抗癌治疗,使用Wnt/β-连环蛋白抑制剂的一个关键限制是它们的治疗指数,即,将有效的抗癌活性与可接受的毒性结合起来的困难。保护健康组织免受Wnt/β-连环蛋白抑制剂的影响是一个主要问题,因为Wnt/β-连环蛋白信号通路在成人组织稳态和再生中的重要作用。在这次审查中,我们提供了关于Wnt/β-catenin途径抑制剂的临床试验的最新摘要,检查它们的抗肿瘤活性和相关的不良事件,并探索正在开发的策略,以改善这种治疗方法的益处/风险状况。
    Despite significant progress in cancer prevention, screening, and treatment, the still limited number of therapeutic options is an obstacle towards increasing the cancer cure rate. In recent years, many efforts were put forth to develop therapeutics that selectively target different components of the oncogenic Wnt/β-catenin signaling pathway. These include small molecule inhibitors, antibodies, and more recently, gene-based approaches. Although some of them showed promising outcomes in clinical trials, the Wnt/β-catenin pathway is still not targeted in routine clinical practice for cancer management. As for most anticancer treatments, a critical limitation to the use of Wnt/β-catenin inhibitors is their therapeutic index, i.e., the difficulty of combining effective anticancer activity with acceptable toxicity. Protecting healthy tissues from the effects of Wnt/β-catenin inhibitors is a major issue due to the vital role of the Wnt/β-catenin signaling pathway in adult tissue homeostasis and regeneration. In this review, we provide an up-to-date summary of clinical trials on Wnt/β-catenin pathway inhibitors, examine their anti-tumor activity and associated adverse events, and explore strategies under development to improve the benefit/risk profile of this therapeutic approach.
    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
    Myb在40年前被鉴定为鸡中急性白血病病毒的转化成分。从那时起,越来越明显的是,MYB活性失调是许多血液癌症的特征。包括急性髓系白血病,它代表了许多人中最“上瘾”的癌蛋白,如果不是全部,这样的疾病。由于MYB的这种肿瘤特异性依赖性,它已成为开发特定抗白血病药物的主要重点。人们非常关注中断MYB与合作因素之间相互作用的方法,特别是EP300/KAT3B和CBP/KAT3A。除了通过筛选小分子鉴定的候选物,新药最令人兴奋的前景似乎是设计直接干扰MYB及其辅因子之间界面的肽模拟物。这样的肽结合了高度的靶特异性和良好的功效,包括对正常造血细胞的最小影响。
    Myb was identified over four decades ago as the transforming component of acute leukemia viruses in chickens. Since then it has become increasingly apparent that dysregulated MYB activity characterizes many blood cancers, including acute myeloid leukemia, and that it represents the most \"addictive\" oncoprotein in many, if not all, such diseases. As a consequence of this tumor-specific dependency for MYB, it has become a major focus of efforts to develop specific antileukemia drugs. Much attention is being given to ways to interrupt the interaction between MYB and cooperating factors, in particular EP300/KAT3B and CBP/KAT3A. Aside from candidates identified through screening of small molecules, the most exciting prospect for novel drugs seems to be the design of peptide mimetics that interfere directly at the interface between MYB and its cofactors. Such peptides combine a high degree of target specificity with good efficacy including minimal effects on normal hematopoietic cells.
    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
    核因子-κB(NF-κB)在先天和适应性免疫系统中起着至关重要的作用,显着影响各种生理过程,如细胞增殖,迁移,分化,生存,和干劲。NF-κB在癌症进展和化疗反应中的功能越来越受到重视。这篇综述强调了NF-B在炎症控制中的作用,生物学机制,以及癌症治疗中的治疗意义。NF-κB有助于改变炎症因子的释放,如TNF-α,IL-6和IL-1β,这是调节致癌作用的关键。具体来说,在包括结肠炎在内的条件下,NF-κB上调可以加剧炎症,可能导致结直肠癌的发展。它的关键作用延伸到调节肿瘤微环境,影响巨噬细胞等成分,成纤维细胞,T细胞,和自然杀伤细胞。这种调节影响肿瘤发生并可以抑制抗肿瘤免疫应答。此外,NF-κB调节细胞死亡机制,特别是通过抑制细胞凋亡和铁凋亡。它在各种癌症中刺激或抑制自噬方面也具有双重作用。除了这些功能,NF-κB在控制癌症干细胞中起作用,促进血管生成,通过EMT诱导增加转移潜能,降低肿瘤细胞对化疗和放疗的敏感性。鉴于其致癌能力,研究集中在可以抑制NF-B的天然产物和小分子化合物上,为癌症治疗提供有希望的途径。
    Nuclear factor-kappaB (NF-ĸB) plays a crucial role in both innate and adaptive immune systems, significantly influencing various physiological processes such as cell proliferation, migration, differentiation, survival, and stemness. The function of NF-ĸB in cancer progression and response to chemotherapy has gained increasing attention. This review highlights the role of NF-ĸB in inflammation control, biological mechanisms, and therapeutic implications in cancer treatment. NF-ĸB is instrumental in altering the release of inflammatory factors such as TNF-α, IL-6, and IL-1β, which are key in the regulation of carcinogenesis. Specifically, in conditions including colitis, NF-ĸB upregulation can intensify inflammation, potentially leading to the development of colorectal cancer. Its pivotal role extends to regulating the tumor microenvironment, impacting components such as macrophages, fibroblasts, T cells, and natural killer cells. This regulation influences tumorigenesis and can dampen anti-tumor immune responses. Additionally, NF-ĸB modulates cell death mechanisms, notably by inhibiting apoptosis and ferroptosis. It also has a dual role in stimulating or suppressing autophagy in various cancers. Beyond these functions, NF-ĸB plays a role in controlling cancer stem cells, fostering angiogenesis, increasing metastatic potential through EMT induction, and reducing tumor cell sensitivity to chemotherapy and radiotherapy. Given its oncogenic capabilities, research has focused on natural products and small molecule compounds that can suppress NF-ĸB, offering promising avenues for cancer therapy.
    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
    通常在多种肿瘤中上调的78kDa葡萄糖调节蛋白(GRP78)是重要的预后标志物,也是抑制肿瘤发生和治疗抗性的有希望的靶标。虽然GRP78已被公认为具有抗凋亡特性的主要内质网(ER)伴侣和未折叠蛋白反应的主要调节因子,它作为癌蛋白表达调节剂的新作用刚刚出现。MYC在约70%的人类癌症中失调,是最常见的活化癌蛋白。然而,尽管最近取得了进展,MYC的治疗靶向仍然具有挑战性.在这里,我们将GRP78确定为抑制MYC表达的新靶标。使用多种MYC依赖性癌症模型,包括头颈部鳞状细胞癌及其顺铂耐药克隆,乳腺和胰腺腺癌,我们的研究表明,通过siRNA敲低GRP78或通过小分子抑制剂(YUM70或HA15)抑制其活性降低c-MYC表达,导致细胞凋亡和细胞活力的丧失。这在2D细胞培养中观察到,3D球体和异种移植模型。机械上,我们确定c-MYC的抑制处于转录后水平,YUM70和HA15处理有效上调真核翻译抑制剂4E-BP1,其靶向对c-MYC翻译起始至关重要的eIF4E.此外,通过siRNA敲除4E-BP1拯救了YUM70介导的c-MYC抑制。由于YUM70还能够抑制N-MYC表达,这项研究提供了一种通过敲低或抑制GRP78来抑制MYC蛋白表达的新方法。
    The 78-kDa glucose regulated protein (GRP78) commonly upregulated in a wide variety of tumors is an important prognostic marker and a promising target for suppressing tumorigenesis and treatment resistance. While GRP78 is well established as a major endoplasmic reticulum (ER) chaperone with anti-apoptotic properties and a master regulator of the unfolded protein response, its new role as a regulator of oncoprotein expression is just emerging. MYC is dysregulated in about 70 % of human cancers and is the most commonly activated oncoprotein. However, despite recent advances, therapeutic targeting of MYC remains challenging. Here we identify GRP78 as a new target for suppression of MYC expression. Using multiple MYC-dependent cancer models including head and neck squamous cell carcinoma and their cisplatin-resistant clones, breast and pancreatic adenocarcinoma, our studies revealed that GRP78 knockdown by siRNA or inhibition of its activity by small molecule inhibitors (YUM70 or HA15) reduced c-MYC expression, leading to onset of apoptosis and loss of cell viability. This was observed in 2D cell culture, 3D spheroid and in xenograft models. Mechanistically, we determined that the suppression of c-MYC is at the post-transcriptional level and that YUM70 and HA15 treatment potently upregulated the eukaryotic translation inhibitor 4E-BP1, which targets eIF4E critical for c-MYC translation initiation. Furthermore, knock-down of 4E-BP1 via siRNA rescued YUM70-mediated c-MYC suppression. As YUM70 is also capable of suppressing N-MYC expression, this study offers a new approach to suppress MYC protein expression through knockdown or inhibition of GRP78.
    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
    部署DNA损伤反应(DDR)对抗各种形式的DNA损伤,确保基因组稳定性。癌细胞对基因组不稳定性的倾向提供了通过抑制DDR途径选择性杀死癌细胞的治疗机会。DNA依赖性蛋白激酶(DNA-PK),核丝氨酸/苏氨酸激酶,对于DNA双链断裂(DSB)修复中的非同源末端连接(NHEJ)途径至关重要。因此,靶向DNA-PK是一种有前途的癌症治疗策略.本文综述了DNA-PK及其相关大蛋白的结构,以及DNA-PK抑制剂的发展过程,以及其临床应用的最新进展。我们强调我们对基于不同支架的DNA-PK抑制剂的开发过程和结构-活性关系(SARs)的分析。我们希望这篇综述将为未来寻求开发新的DNA-PK抑制剂的研究人员提供实用信息。
    The deployment of DNA damage response (DDR) combats various forms of DNA damage, ensuring genomic stability. Cancer cells\' propensity for genomic instability offers therapeutic opportunities to selectively kill cancer cells by suppressing the DDR pathway. DNA-dependent protein kinase (DNA-PK), a nuclear serine/threonine kinase, is crucial for the non-homologous end joining (NHEJ) pathway in the repair of DNA double-strand breaks (DSBs). Therefore, targeting DNA-PK is a promising cancer treatment strategy. This review elaborates on the structures of DNA-PK and its related large protein, as well as the development process of DNA-PK inhibitors, and recent advancements in their clinical application. We emphasize our analysis of the development process and structure-activity relationships (SARs) of DNA-PK inhibitors based on different scaffolds. We hope this review will provide practical information for researchers seeking to develop novel DNA-PK inhibitors in the future.
    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