targeted protein degradation

靶向蛋白质降解
  • 文章类型: Journal Article
    乳腺癌(BC)占全球女性癌症患者癌症病例的30%,这表明迫切需要开发靶向BCs的选择性疗法。最近,蛋白水解靶向嵌合体(PROTAC)已成为靶向乳腺癌的有希望的策略。PROTAC是由靶蛋白配体组成的嵌合分子,E3连接酶配体,和共轭接头,使其能够通过招募紧密靠近的E3连接酶来促进所需靶蛋白的降解。由于引起BC的蛋白质的催化行为和直接降解,PROTAC可以实现低剂量的高药效,引起了人们对其治疗潜力的极大关注。这篇综述提供了当前开发的针对BCs的PROTACs的案例,具体取决于BCs的类型,限制,以及PROTAC靶向BCs的观点。
    Breast cancer (BC) accounts for 30% of cancer cases among women cancer patient globally, indicating the urgent need for the development of selective therapies targeting BCs. Recently, proteolysis-targeting chimera (PROTAC) has been emerged as promising strategy to target breast cancer. PROTAC is a chimeric molecule consisting with target protein ligand, E3 ligase ligand, and conjugating linkers, enabling it to facilitate the degradation of desired target proteins via recruiting E3 ligase in close proximity. Due to the catalytic behavior and direct degradation of BC-causing proteins, PROTAC could achieve high drug efficacy with low doses, resulting in a great attention for its potential as therapeutics. This review provides cases of the current developed PROTACs targeting BCs depending on the type of BCs, limitation, and perspective of PROTAC in targeting BCs.
    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
    靶向蛋白质降解(TPD)允许细胞维持功能性蛋白质组并快速适应变化的条件。重新利用TPD去活化特定蛋白质的方法在治疗和研究应用中已显示出显著的潜力。这些方法中的大多数是基于蛋白水解靶向嵌合体(PROTACs),它将蛋白质靶标连接到E3泛素连接酶,导致目标蛋白的泛素降解。在这项研究中,我们介绍了一种基于纳米抗体缀合的含泛素调节X结构域(PUX)衔接蛋白的非泛素依赖性TPD方法。我们表明,基于PUX的NAnobody降解物(P-NAD)可以通过CDC48展开酶的拟南芥和人类直系同源物展开目标蛋白,而无需泛素化或启动基序。我们证明了P-NAD质粒可以转染到人细胞系中,其中产生的P-NAD使用内源性CDC48机制用于143kDa多域蛋白的不依赖泛素的TPD。因此,P-NAD为不依赖泛素的治疗性TPD方法铺平了道路。此外,模块化P-NAD设计与体外和细胞检测相结合,为阐明植物和动物中基于CDC48的TPD的功能方面提供了一个通用平台.
    Targeted protein degradation (TPD) allows cells to maintain a functional proteome and to rapidly adapt to changing conditions. Methods that repurpose TPD for the deactivation of specific proteins have demonstrated significant potential in therapeutic and research applications. Most of these methods are based on proteolysis targeting chimaeras (PROTACs) which link the protein target to an E3 ubiquitin ligase, resulting in the ubiquitin-based degradation of the target protein. In this study, we introduce a method for ubiquitin-independent TPD based on nanobody-conjugated plant ubiquitin regulatory X domain-containing (PUX) adaptor proteins. We show that the PUX-based NAnobody Degraders (P-NADs) can unfold a target protein through the Arabidopsis and human orthologues of the CDC48 unfoldase without the need for ubiquitination or initiating motifs. We demonstrate that P-NAD plasmids can be transfected into a human cell line, where the produced P-NADs use the endogenous CDC48 machinery for ubiquitin-independent TPD of a 143 kDa multidomain protein. Thus, P-NADs pave the road for ubiquitin-independent therapeutic TPD approaches. In addition, the modular P-NAD design combined with in vitro and cellular assays provide a versatile platform for elucidating functional aspects of CDC48-based TPD in plants and animals.
    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
    理由:由于癌基因表达产物通常表现出上调或异常激活的活性,开发一种调节异常蛋白质水平的技术代表了治疗肿瘤和蛋白质异常相关疾病的可行方法。方法:我们首先筛选出具有高靶向降解效率的eMIATAC组分,探索eMIATAC诱导靶蛋白降解的机制,并通过蛋白质印迹和流式细胞术验证了目标蛋白的降解效率。接下来,我们将eMIATAC与一些可控元件重组以验证靶蛋白的可调节降解性能。随后,我们构建了能够靶向降解AKT1的eMIATAC,并在体内外验证了其对GBM细胞发育的影响。最后,我们将eMIATAC与CAR序列连接,构建具有低BATF蛋白水平的CAR-T细胞,并验证其抗肿瘤功效的变化.结果:我们开发了一种基于内体-微自噬-溶酶体途径的系统,用于降解内源性蛋白:内体-微自噬流变性嵌合体(eMIATAC),依赖于Vps4A而不是溶酶体相关膜蛋白2A(LAMP2A)与伴侣Hsc70和目标蛋白(POI)结合。然后,复合体被晚期内体运送到溶酶体,其中降解发生类似于微自噬。eMIATAC证明了准确性,效率,可逆性,以及降解靶蛋白EGFP的可控性。此外,eMIATAC在体内和体外靶向内源性蛋白质时在敲低POI方面表现出优异的性能。结论:eMIATACs不仅可以直接敲除异常蛋白用于胶质瘤治疗,还可以通过敲除T细胞耗竭相关蛋白增强CAR-T细胞治疗肿瘤的疗效。新开发的eMIATAC系统有望成为蛋白质击倒策略的新工具。通过实现对内源性蛋白质水平的直接控制,eMIATAC有可能彻底改变癌症和遗传疾病的治疗方法。
    Rationale: Since oncogene expression products often exhibit upregulation or abnormally activated activity, developing a technique to regulate abnormal protein levels represent a viable approach for treating tumors and protein abnormality-related diseases. Methods: We first screened out eMIATAC components with high targeted degradation efficiency and explored the mechanism by which eMIATAC induced target protein degradation, and verified the degradation efficiency of the target protein by protein imprinting and flow cytometry. Next, we recombined eMIATAC with some controllable elements to verify the regulatable degradation performance of the target protein. Subsequently, we constructed eMIATAC that can express targeted degradation of AKT1 and verified its effect on GBM cell development in vitro and in vivo. Finally, we concatenated eMIATAC with CAR sequences to construct CAR-T cells with low BATF protein levels and verified the changes in their anti-tumor efficacy. Results: we developed a system based on the endosome-microautophagy-lysosome pathway for degrading endogenous proteins: endosome-MicroAutophagy TArgeting Chimera (eMIATAC), dependent on Vps4A instead of lysosomal-associated membrane protein 2A (LAMP2A) to bind to the chaperone Hsc70 and the protein of interest (POI). The complex was then transported to the lysosome by late endosomes, where degradation occurred similarly to microautophagy. The eMIATACs demonstrated accuracy, efficiency, reversibility, and controllability in degrading the target protein EGFP. Moreover, eMIATAC exhibited excellent performance in knocking down POI when targeting endogenous proteins in vivo and in vitro. Conclusions: The eMIATACs could not only directly knock down abnormal proteins for glioma treatment but also enhance the therapeutic effect of CAR-T cell therapy for tumors by knocking down T cell exhaustion-related proteins. The newly developed eMIATAC system holds promise as a novel tool for protein knockdown strategies. By enabling direct control over endogenous protein levels, eMIATAC has the potential to revolutionize treatment for cancer and genetic diseases.
    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
    在过去的二十年中,靶向蛋白质降解技术作为消除原本难以治疗的致病蛋白质的革命性策略已经获得了巨大的动力。在开发的各种方法中,利用身体的先天蛋白质稳态机制来实现这一目的,溶酶体靶向嵌合体(LYTAC)通过将靶蛋白与溶酶体运输受体偶联来利用溶酶体降解途径,代表了最新的创新。这些嵌合体独特地用于降解膜结合和细胞外的蛋白质,约占所有蛋白质组的40%。最近开发了几种新的LYTAC配方,为治疗降解剂的设计和开发提供有价值的见解。本文综述了LYTAC技术的最新进展,它的实际应用,以及决定目标降解效率的因素。还讨论了该技术的潜力和新兴趋势。LYTAC技术为靶向蛋白质降解提供了一个有希望的途径,有可能彻底改变众多疾病的治疗前景。
    Targeted protein degradation technology has gained substantial momentum over the past two decades as a revolutionary strategy for eliminating pathogenic proteins that are otherwise refractory to treatment. Among the various approaches developed to harness the body\'s innate protein homeostasis mechanisms for this purpose, lysosome targeting chimeras (LYTACs) that exploit the lysosomal degradation pathway by coupling the target proteins with lysosome-trafficking receptors represent the latest innovation. These chimeras are uniquely tailored to degrade proteins that are membrane-bound and extracellular, encompassing approximately 40% of all proteome. Several novel LYTAC formulas have been developed recently, providing valuable insights for the design and development of therapeutic degraders. This review delineates the recent progresses of LYTAC technology, its practical applications, and the factors that dictate target degradation efficiency. The potential and emerging trends of this technology are discussed as well. LYTAC technology offers a promising avenue for targeted protein degradation, potentially revolutionizing the therapeutic landscape for numerous diseases.
    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
    靶向蛋白质降解是一种新兴且快速发展的治疗策略。特别是,与小分子相比,基于生物制品的靶向降解模式(bioPROTACs)相对缺乏探索。这里,我们研究了目标亲和力,细胞定位,和bioPROTACs的效价影响含有蛋白酪氨酸磷酸酶2(SHP2)的致癌磷酸酶src-同源性2的靶向降解的功效。我们确定通过bioprotacs二价募集SHP2作为一种广泛适用的策略来提高效力。此外,我们证明,SHP2靶向的生物PROTACs可以有效地对抗癌症中存在的功能获得SHP2突变体,否则用小分子构建体选择性靶向是具有挑战性的。总的来说,这项研究证明了生物蛋白质用于具有挑战性的目标的实用性,并进一步阐述了治疗性生物方案的设计原则。
    Targeted protein degradation is an emergent and rapidly evolving therapeutic strategy. In particular, biologics-based targeted degradation modalities (bioPROTACs) are relatively under explored compared to small molecules. Here, we investigate how target affinity, cellular localization, and valency of bioPROTACs impact efficacy of targeted degradation of the oncogenic phosphatase src-homology 2 containing protein tyrosine phosphatase-2 (SHP2). We identify bivalent recruitment of SHP2 by bioPROTACs as a broadly applicable strategy to improve potency. Moreover, we demonstrate that SHP2-targeted bioPROTACs can effectively counteract gain-of-function SHP2 mutants present in cancer, which are otherwise challenging to selectively target with small molecule constructs. Overall, this study demonstrates the utility of bioPROTACs for challenging targets, and further explicates design principles for therapeutic bioPROTACs.
    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
    Vepdegestrant(ARV-471)是一种口服PROTACER降解剂,可结合E3泛素连接酶和ER,直接触发ER的泛素化及其随后的蛋白酶体降解。在一项首次进入人类的I/II期研究中,在ER+/HER2-晚期乳腺癌患者中,vepdegestrant单药治疗的临床活性耐受性良好。全球,随机III期VERITAC-2研究比较了在使用CDK4/6抑制剂加内分泌治疗后,Vepdegestrant与氟维司群在ER+/HER2-晚期乳腺癌成人患者中的疗效和安全性.将在意向治疗人群和ESR1突变阳性亚群中评估盲化独立中央审查(主要终点)的无进展生存率。次要终点包括总生存期,肿瘤反应,安全,药代动力学,患者报告的结果,和循环肿瘤DNA生物标志物。临床试验注册:NCT05654623(ClinicalTrials.gov)。
    VERITAC-2是一项比较vepdegestrant的临床试验,一种降解雌激素受体的新药,对于ER+/HER2-晚期乳腺癌患者的现有治疗称为氟维司群:雌激素受体阳性(ER+)/人类表皮生长因子受体2阴性(HER2-)乳腺癌在雌激素反应中生长,体内的激素,并且具有低水平或没有HER2蛋白。患有ER+/HER2-晚期乳腺癌的人,传播到身体的另一部分,或不能通过手术切除通常用细胞周期蛋白依赖性激酶(CDK)4/6抑制剂和内分泌疗法治疗,但是他们的癌症可能会在这些治疗上变得更糟,需要新的治疗方法。Fulvestrant,一种附着在雌激素受体上的内分泌疗法,降低雌激素对肿瘤的影响,可以减缓或阻止癌症的生长。Vepdegestrant,一种新药正在测试ER+乳腺癌,是一种PROtesolutionTogeting嵌合体(PROTAC)蛋白质降解剂,它附着在雌激素受体上,并使它们被标记为通过细胞的天然蛋白质处置系统去除。通过去除雌激素受体,vepdegestrant可能导致肿瘤停止生长或缩小。本文介绍了VERITAC-2期临床研究,比较了Vepdegestrant与氟维司群在先前接受CDK4/6抑制剂和内分泌疗法治疗的ER/HER2晚期乳腺癌患者中的应用。患者将被随机分配接受vepdegestrant(每天口服一次的药丸)或氟维司群(注射到肌肉中)。这项研究的目的是找出使用vepdegegestrant或fulvestrant的癌症没有恶化的人的寿命。VERITAC-2还将研究人们在研究期间的寿命,人们可能会经历的副作用,以及整个研究过程中人们的整体幸福感。
    Vepdegestrant (ARV-471) is an oral PROTAC ER degrader that binds an E3 ubiquitin ligase and ER to directly trigger ubiquitination of ER and its subsequent proteasomal degradation. In a first-in-human Phase I/II study, vepdegestrant monotherapy was well tolerated with clinical activity in pretreated patients with ER+/HER2- advanced breast cancer. The global, randomized Phase III VERITAC-2 study compares efficacy and safety of vepdegestrant versus fulvestrant in adults with ER+/HER2- advanced breast cancer after treatment with a CDK4/6 inhibitor plus endocrine therapy. Progression-free survival by blinded independent central review (primary end point) will be assessed in the intention-to-treat population and ESR1 mutation-positive subpopulation. Secondary end points include overall survival, tumor response, safety, pharmacokinetics, patient-reported outcomes, and circulating tumor DNA biomarkers.Clinical trial registration: NCT05654623 (ClinicalTrials.gov).
    VERITAC-2 is a clinical trial comparing vepdegestrant, a new drug that degrades estrogen receptors, to an existing treatment called fulvestrant in patients with ER+/HER2- advanced breast cancer: Estrogen receptor-positive (ER+)/human epidermal growth factor receptor 2-negative (HER2-) breast cancer grows in response to estrogen, a hormone in the body, and has low levels or no HER2 protein. People living with ER+/HER2- advanced breast cancer that has grown, spread to another part of the body, or cannot be removed by surgery are often treated with cyclin-dependent kinase (CDK) 4/6 inhibitors and endocrine therapies, but their cancer may get worse on these treatments and new treatments are needed. Fulvestrant, an endocrine therapy that attaches to estrogen receptors, lowers estrogen\'s effect on tumors and can slow or stop cancer growth. Vepdegestrant, a new medicine being tested for ER+ breast cancer, is a PROteolysis TArgeting Chimera (PROTAC) protein degrader that attaches to estrogen receptors and causes them to be tagged for removal by the cell\'s natural protein disposal system. By removing estrogen receptors, vepdegestrant may cause tumors to stop growing or shrink.This paper describes the Phase III VERITAC-2 clinical study comparing vepdegestrant versus fulvestrant in people living with ER+/HER2- advanced breast cancer previously treated with a CDK4/6 inhibitor and endocrine therapy.Patients will be randomly assigned to receive vepdegestrant (a pill taken once daily by mouth) or fulvestrant (a shot given into the muscle). The purpose of the study is to find out how long people live without their cancer getting worse with vepdegestrant or fulvestrant. VERITAC-2 will also look at how long people live during the study, side effects people may experience, and the overall well-being of people throughout the study.
    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
    嵌合抗原受体(CAR)T细胞在临床上产生了巨大的影响,但是通过CAR的有效信号传导可能不利于治疗的安全性和有效性。使用蛋白质降解来控制CAR信号传导可以解决临床前模型中的这些问题。用于调节CAR稳定性的现有策略依赖于小分子来诱导全身性降解。与小分子调控相反,遗传电路提供了一种更精确的方法来以自主的逐细胞方式控制CAR信号。这里,我们描述了一种可编程的蛋白质降解工具,它采用了生物蛋白质降解的框架,异双功能蛋白,由靶识别结构域与募集内源性泛素蛋白酶体系统的结构域融合组成。我们开发了新颖的生物PROTACs,该生物利用紧凑的四残基degron,并使用纳米抗体或合成亮氨酸拉链作为蛋白质结合剂证明了胞浆和膜蛋白靶标的降解。我们的生物PROTAC表现出CAR的有效降解,并且可以抑制原代人T细胞中的CAR信号传导。我们通过构建遗传电路来降解酪氨酸激酶ZAP70,以响应对特定膜结合抗原的识别,从而证明了我们的bioPROTACs的实用性。该回路可以仅在特定细胞群存在的情况下破坏CART细胞信号传导。这些结果表明,bioPROTACs是扩展CAR-T细胞工程工具箱的强大工具。
    Chimeric antigen receptor (CAR) T cells have made a tremendous impact in the clinic, but potent signaling through the CAR can be detrimental to treatment safety and efficacy. The use of protein degradation to control CAR signaling can address these issues in preclinical models. Existing strategies for regulating CAR stability rely on small molecules to induce systemic degradation. In contrast to small molecule regulation, genetic circuits offer a more precise method to control CAR signaling in an autonomous cell-by-cell fashion. Here, we describe a programmable protein degradation tool that adopts the framework of bioPROTACs, heterobifunctional proteins that are composed of a target recognition domain fused to a domain that recruits the endogenous ubiquitin proteasome system. We develop novel bioPROTACs that utilize a compact four-residue degron and demonstrate degradation of cytosolic and membrane protein targets using either a nanobody or synthetic leucine zipper as a protein binder. Our bioPROTACs exhibit potent degradation of CARs and can inhibit CAR signaling in primary human T cells. We demonstrate the utility of our bioPROTACs by constructing a genetic circuit to degrade the tyrosine kinase ZAP70 in response to recognition of a specific membrane-bound antigen. This circuit can disrupt CAR T cell signaling only in the presence of a specific cell population. These results suggest that bioPROTACs are powerful tools for expanding the CAR T cell engineering toolbox.
    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
    阿尔茨海默病(Alzheimer’sdisease,AD)是最常见的慢性神经退行性疾病之一。过度磷酸化tau在AD的神经元功能障碍和突触损伤中起着不可或缺的作用。蛋白水解靶向嵌合体(PROTACs)是一种新型的嵌合分子,可以通过诱导其聚泛素化来降解靶蛋白。这种方法已经显示出降低tau蛋白水平的希望,是AD的潜在治疗靶点。与传统药物疗法相比,使用PROTACs降低tau水平可能为治疗AD提供更具体和有效的策略,副作用少。在本研究中,我们设计并合成了一系列小分子PROTACs来敲除tau蛋白。其中,化合物C8能够降低HEK293细胞中的总tau水平和磷酸化tau水平,其中野生型全长人tau(称为HEK293-htau)和htau过表达的小鼠稳定表达。Western印迹结果表明,C8通过泛素-蛋白酶体系统以时间依赖性方式降解tau蛋白。在过表达htau的小鼠中,新型物体识别和Morris水迷宫测试的结果表明,C8显着改善了认知功能。一起,我们的研究结果表明,使用小分子PROTACC8降解磷酸化tau可能是AD的一种有前景的治疗策略.
    Alzheimer\'s disease (AD) is one of the most common chronic neurodegenerative diseases. Hyperphosphorylated tau plays an indispensable role in neuronal dysfunction and synaptic damage in AD. Proteolysis-targeting chimeras (PROTACs) are a novel type of chimeric molecule that can degrade target proteins by inducing their polyubiquitination. This approach has shown promise for reducing tau protein levels, which is a potential therapeutic target for AD. Compared with traditional drug therapies, the use of PROTACs to reduce tau levels may offer a more specific and efficient strategy for treating AD, with fewer side effects. In the present study, we designed and synthesized a series of small-molecule PROTACs to knock down tau protein. Of these, compound C8 was able to lower both total and phosphorylated tau levels in HEK293 cells with stable expression of wild-type full-length human tau (termed HEK293-htau) and htau-overexpressed mice. Western blot findings indicated that C8 degraded tau protein through the ubiquitin-proteasome system in a time-dependent manner. In htau-overexpressed mice, the results of both the novel object recognition and Morris water maze tests revealed that C8 markedly improved cognitive function. Together, our findings suggest that the use of the small-molecule PROTAC C8 to degrade phosphorylated tau may be a promising therapeutic strategy for AD.
    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
    分子胶可以诱导靶蛋白和泛素连接酶之间的接近以诱导靶降解,但是发现它们的策略仍然有限。我们筛选了3,200个生物活性小分子,并确定C646需要neddylation依赖性蛋白质降解来诱导细胞毒性。尽管组蛋白乙酰转移酶p300是C646的典型靶标,但我们提供了大量证据表明C646直接靶向并降解Exportin-1(XPO1)。由C646诱导的多种细胞表型在表达已知XPO1C528S药物抗性等位基因的细胞中被消除。虽然XPO1催化许多货物蛋白的核到细胞质的运输,它也直接结合染色质。我们证明p300和XPO1共同占据数百个染色质基因座。使用C646或已知的XPO1调节剂S109降解XPO1会减少XPO1和p300的染色质占用,从而使XPO1能够直接靶向表型p300抑制。这项工作强调了耐药等位基因的实用性,并进一步验证了XPO1作为染色质状态的可靶向调节因子。
    Molecular glues can induce proximity between a target protein and ubiquitin ligases to induce target degradation, but strategies for their discovery remain limited. We screened 3,200 bioactive small molecules and identified that C646 requires neddylation-dependent protein degradation to induce cytotoxicity. Although the histone acetyltransferase p300 is the canonical target of C646, we provide extensive evidence that C646 directly targets and degrades Exportin-1 (XPO1). Multiple cellular phenotypes induced by C646 were abrogated in cells expressing the known XPO1C528S drug-resistance allele. While XPO1 catalyzes nuclear-to-cytoplasmic transport of many cargo proteins, it also directly binds chromatin. We demonstrate that p300 and XPO1 co-occupy hundreds of chromatin loci. Degrading XPO1 using C646 or the known XPO1 modulator S109 diminishes the chromatin occupancy of both XPO1 and p300, enabling direct targeting of XPO1 to phenocopy p300 inhibition. This work highlights the utility of drug-resistant alleles and further validates XPO1 as a targetable regulator of chromatin state.
    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
    PDIA1在癌症中的过度表达刺激了对有效抑制剂的寻求。然而,现有的抑制剂通常只与一个活性位点结合,限制其功效。在我们的研究中,我们通过将PACMA31(PA)类似物与针对小脑的泊马度胺相结合,开发了PROTAC模拟探针dPA。通过蛋白质谱分析和分析,我们证实了dPA与PDIA1活性位点半胱氨酸的特异性相互作用。我们进一步合成了具有噻吩环和各种接头的PROTAC变体以提高降解效率。值得注意的是,H4,具有PEG接头,与PA类似,诱导显著的PDIA1降解并抑制癌细胞增殖。H4的生物安全性与PA相当,强调其在癌症治疗中进一步发展的潜力。我们的发现强调了通过靶向降解抑制PDIA1的新策略,为癌症治疗提供了有希望的前景。这种方法可以克服常规抑制剂的局限性,提出了推进抗癌干预措施的新途径。
    The overexpression of PDIA1 in cancer has spurred the quest for effective inhibitors. However, existing inhibitors often bind to only one active site, limiting their efficacy. In our study, we developed a PROTAC-mimetic probe dPA by combining PACMA31 (PA) analogs with cereblon-directed pomalidomide. Through protein profiling and analysis, we confirmed dPA\'s specific interaction with PDIA1\'s active site cysteines. We further synthesized PROTAC variants with a thiophene ring and various linkers to enhance degradation efficiency. Notably, H4, featuring a PEG linker, induced significant PDIA1 degradation and inhibited cancer cell proliferation similarly to PA. The biosafety profile of H4 is comparable to that of PA, highlighting its potential for further development in cancer therapy. Our findings highlight a novel strategy for PDIA1 inhibition via targeted degradation, offering promising prospects in cancer therapeutics. This approach may overcome limitations of conventional inhibitors, presenting new avenues for advancing anti-cancer interventions.
    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