Androgen receptor

雄激素受体
  • 文章类型: Journal Article
    雄激素受体(AR)是前列腺癌(PCa)的关键驱动因素,因此,当前的主流治疗针对该分子。然而,这些疗法通常会产生耐药性,因此,必须评估其他目标以改善患者预后.因此,寻求间接靶向AR的替代方法。AR与其他信号通路的串扰,包括几个蛋白激酶信号级联,已被确定为对抗治疗耐药性的潜在途径。蛋白激酶的酪蛋白激酶1(CK1)家族磷酸化多种底物,允许它们调节从细胞周期到DNA损伤修复的各种途径。以及它在PCa中下调的几种信号通路中的作用,突变数据表明它有可能促进前列腺癌的发生。CK1α是一种预测通过磷酸化调节AR活性的同种型,并与几种其他癌症类型的进展有关。在这次审查中,我们探讨CK1的正常生物学功能是如何在癌症中下调的,对信号通路的影响以及这如何促进前列腺肿瘤发生,特别关注CK1α同种型作为PCa的新治疗靶标。
    The androgen receptor (AR) is a key driver of prostate cancer (PCa) and, as such, current mainstay treatments target this molecule. However, resistance commonly arises to these therapies and, therefore, additional targets must be evaluated to improve patient outcomes. Consequently, alternative approaches for indirectly targeting the AR are sought. AR crosstalk with other signalling pathways, including several protein kinase signalling cascades, has been identified as a potential route to combat therapy resistance. The casein kinase 1 (CK1) family of protein kinases phosphorylate a multitude of substrates, allowing them to regulate a diverse range of pathways from the cell cycle to DNA damage repair. As well as its role in several signalling pathways that are de-regulated in PCa, mutational data suggest its potential to promote prostate carcinogenesis. CK1α is one isoform predicted to regulate AR activity via phosphorylation and has been implicated in the progression of several other cancer types. In this review, we explore how the normal biological function of CK1 is de-regulated in cancer, the impact on signalling pathways and how this contributes towards prostate tumourigenesis, with a particular focus on the CK1α isoform as a novel therapeutic target for PCa.
    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
    Latrophilins(LPHN),一组已知与蜘蛛毒latrotoxin(LTX)结合的G蛋白偶联受体,在肿瘤性疾病中仍然基本上没有特征。在本研究中,我们旨在确定LPHNs在前列腺癌进展中的作用.我们评估了LPHN的行动,包括LPHN1,LPHN2和LPHN3,通过其配体(例如,α-LTX,FLRT3)治疗或shRNA感染,以及手术标本。在雄激素受体(AR)阳性的LNCaP/C4-2/22Rv1细胞中,二氢睾酮显著增加LPHN的表达水平,而染色质免疫沉淀分析显示内源性ARs的结合,每个LPHN的启动子区包括AR-V7。用α-LTX或FLRT3处理导致诱导细胞活力和AR阳性和AR阴性细胞系的迁移。α-LTX和FLRT3还增强Bcl-2和JAK2和STAT3的磷酸化形式的表达。同时,每个LPHN的敲除对配体处理介导的所有LPHN都显示出相反的作用。在根治性前列腺切除术标本中的免疫组织化学进一步显示每个LPHN在前列腺癌中的表达显著升高,与邻近的正常前列腺相比,在单变量和多变量设置中,这与术后生化复发的风险显著升高相关.这些发现表明,LPHNs作为ARs的下游效应子,促进雄激素敏感性的生长,抗阉割,甚至AR阴性前列腺癌。
    Latrophilins (LPHNs), a group of the G-protein-coupled receptor to which a spider venom latrotoxin (LTX) is known to bind, remain largely uncharacterized in neoplastic diseases. In the present study, we aimed to determine the role of LPHNs in the progression of prostate cancer. We assessed the actions of LPHNs, including LPHN1, LPHN2, and LPHN3, in human prostate cancer lines via their ligand (e.g., α-LTX, FLRT3) treatment or shRNA infection, as well as in surgical specimens. In androgen receptor (AR)-positive LNCaP/C4-2/22Rv1 cells, dihydrotestosterone considerably increased the expression levels of LPHNs, while chromatin immunoprecipitation assay revealed the binding of endogenous ARs, including AR-V7, to the promoter region of each LPHN. Treatment with α-LTX or FLRT3 resulted in induction in the cell viability and migration of both AR-positive and AR-negative lines. α-LTX and FLRT3 also enhanced the expression of Bcl-2 and phosphorylated forms of JAK2 and STAT3. Meanwhile, the knockdown of each LPHN showed opposite effects on all of those mediated by ligand treatment. Immunohistochemistry in radical prostatectomy specimens further showed the significantly elevated expression of each LPHN in prostate cancer, compared with adjacent normal-appearing prostate, which was associated with a significantly higher risk of postoperative biochemical recurrence in both univariate and multivariable settings. These findings indicate that LPHNs function as downstream effectors of ARs and promote the growth of androgen-sensitive, castration-resistant, or even AR-negative prostate cancer.
    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
    实体肿瘤以及白血病和淋巴瘤显示出显著的核结构变化,包括核大小和形状,核仁的数量和大小,和染色质纹理。这些改变已用于癌症诊断,可能与癌细胞的功能特性改变有关。核基质(NM)代表核的结构组成,由核层和孔复合物组成,内部的核糖核酸蛋白质网络,和残留的核仁.在核微环境中,NM与多蛋白复合物有关,如基础转录因子,信号蛋白,组蛋白修饰因子,和染色质重塑机制直接或间接通过支架蛋白。因此,NM组成的改变可能导致DNA拓扑结构的改变和各种基因相互作用的变化,然后可以参与癌症过程的级联。使用雄激素敏感的前列腺癌细胞系,LNCaP,和它的雄激素非依赖性衍生物,LN96,NM蛋白的常规2D蛋白质组学分析显示,在NM蛋白中检测到富含嘌呤的元件结合蛋白α(PURα),并在细胞系之间差异表达。在这篇文章中,我们将回顾该分子在前列腺癌中的潜在作用。
    Solid tumors as well as leukemias and lymphomas show striking changes in nuclear structure including nuclear size and shape, the number and size of nucleoli, and chromatin texture. These alterations have been used in cancer diagnosis and might be related to the altered functional properties of cancer cells. The nuclear matrix (NM) represents the structural composition of the nucleus and consists of nuclear lamins and pore complexes, an internal ribonucleic protein network, and residual nucleoli. In the nuclear microenvironment, the NM is associated with multi-protein complexes, such as basal transcription factors, signaling proteins, histone-modifying factors, and chromatin remodeling machinery directly or indirectly through scaffolding proteins. Therefore, alterations in the composition of NM could result in altered DNA topology and changes in the interaction of various genes, which could then participate in a cascade of the cancer process. Using an androgen-sensitive prostate cancer cell line, LNCaP, and its androgen-independent derivative, LN96, conventional 2D-proteomic analysis of the NM proteins revealed that purine-rich element binding protein alpha (PURα) was detected in the NM proteins and differentially expressed between the cell lines. In this article, we will review the potential role of the molecule in prostate cancer.
    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
    在这项工作中,一系列姜黄素衍生物(1a-1h,2a-2g,和3a-3c)被合成用于抑制去势抵抗的前列腺癌细胞。所有合成的化合物通过1HNMR表征,13CNMR,HRMS,和熔点。体外细胞毒性研究表明,化合物1a,1e,1f,1h,2g,3a,和3c显示与ASC-J9相比对22Rvl和C4-2细胞相似或增强的细胞毒性,其他合成的化合物显示与ASC-J9相比对22Rvl和C4-2细胞降低的细胞毒性。进行分子对接模拟以研究合成的化合物与雄激素受体的结合亲和力和可能的结合模式。结果表明,与ASC-J9相比,所有合成的化合物都表现出更高的cdocker相互作用能。化合物1h,2g,和3c不仅显示出对22Rv1和C4-2细胞的强细胞毒性,而且显示与雄激素受体的高结合亲和力。在雄激素受体抑制研究中,与ASC-J9相比,化合物1f和2g对C4-2细胞显示出相似的雄激素受体抑制作用,与ASC-J9、1f和2g相比,化合物3c显示出显著增强的AR抑制作用。化合物1a,1e,1f,1h,2g,在这项工作中制备的3a和3c具有用于去势抵抗前列腺癌治疗的巨大潜力。
    In this work, a series of curcumin derivatives (1a-1h, 2a-2g, and 3a-3c) were synthesized for the suppression of castration-resistant prostate cancer cells. All synthesized compounds were characterized by 1H NMR, 13C NMR, HRMS, and melting point. The in vitro cytotoxicity study shows that compounds 1a, 1e, 1f, 1h, 2g, 3a, and 3c display similar or enhanced cytotoxicity against 22Rv1 and C4-2 cells as compared to ASC-J9, other synthesized compounds display reduced cytotoxicity against 22Rv1 and C4-2 cells as compared to ASC-J9. Molecular docking simulation was performed to study the binding affinity and probable binding modes of the synthesized compounds with androgen receptor. The results show that all synthesized compounds exhibit higher cdocker interaction energies as compared to ASC-J9. Compounds 1h, 2g, and 3c not only show strong cytotoxicity against 22Rv1 and C4-2 cells but also exhibit high binding affinity with androgen receptor. In androgen receptor suppression study, compounds 1f and 2g show similar androgen receptor suppression effect as compared to ASC-J9 on C4-2 cells, compound 3c displays significantly enhanced AR suppression effect as compared to ASC-J9, 1f and 2g. Compounds 1a, 1e, 1f, 1h, 2g, 3a and 3c prepared in this work have significant potential for castration-resistant prostate cancer 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
    作为全氟辛酸(PFOA)的替代品,六氟环氧丙烷二聚酸(HFPO-DA)和三聚酸(HFPO-TA)已在环境介质甚至人类中越来越多地检测到。它们已被证明对模型物种具有生殖毒性,但它们对人类的影响仍不清楚,因为它们的行动方式存在知识差距。在这里,通过结合斑马鱼试验,研究了两种HFPOs和PFOA的(抗)雄激素作用,并探索了潜在的毒理机制。细胞分析和分子对接模拟。幼年斑马鱼在性别分化过程中暴露于化学物质会促进女性化,HFPO-TA在1μg/L的环境浓度下起作用。化学物质抑制人前列腺细胞的增殖和人和斑马鱼雄激素受体(AR)的转录活性,HFPO-TA显示出最强的效力。分子对接显示化学物质以类似于已知AR拮抗剂的构象与AR结合。结合在体内,体外和计算机模拟结果表明,化学物质可能通过拮抗AR介导的途径破坏性别分化,并提供了更多证据表明HFPO-TA不是PFOA的安全替代品。
    As alternatives of perfluorooctanoic acid (PFOA), hexafluoropropylene oxide dimeric acid (HFPO-DA) and trimeric acid (HFPO-TA) have been detected increasingly in environmental media and even humans. They have been shown to exhibit reproductive toxicity to model species, but their effects on human remain unclear due to the knowledge gap in their mode of action. Herein, (anti-)androgenic effects of the two HFPOs and PFOA were investigated and underlying toxicological mechanism was explored by combining zebrafish test, cell assay and molecular docking simulation. Exposure of juvenile zebrafish to the chemicals during sex differentiation promoted feminization, with HFPO-TA acting at an environmental concentration of 1 μg/L. The chemicals inhibited proliferation of human prostate cells and transcriptional activity of human and zebrafish androgen receptors (AR), with HFPO-TA displaying the strongest potency. Molecular docking revealed that the chemicals bind to AR in a conformation similar to a known AR antagonist. Combined in vivo, in vitro and in silico results demonstrated that the chemicals disrupted sex differentiation likely by antagonizing AR-mediated pathways, and provided more evidence that HFPO-TA is not a safe alternative to PFOA.
    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
    卵巢癌(OC)的治疗仍然是妇科肿瘤学的最大挑战之一。OC中经典类固醇受体的存在使激素治疗成为一种有吸引力的选择;然而,OC对激素治疗的反应不大。这里,我们比较了孕酮(PGR)的表达模式,浆液性OC细胞系和非癌卵巢细胞之间的雄激素(AR)和雌激素α(ERα)受体。使用生物信息学方法分析了这些数据与来自患者肿瘤样品的类固醇受体表达谱和生存结果有关。结果表明,ERα,PGR和AR在OC细胞系中共表达,和来自高等级和低等级OC的患者样品共表达至少两种类固醇受体。AR高表达呈负相关,而ERα和PGR表达与患者生存率呈正相关。与非癌症(HOSEpiC)卵巢细胞相比,AR在1型(SKOV-3)和2型(OVCAR-3)OC细胞系中显示出与ERα和PGR相反的表达模式,AR在1型中下调,在2型OC中上调。与其他受体比率相比,低AR/PGR比率和高ESRl/AR比率与OC的有利生存结果相关。尽管由于分析的原发性肿瘤样本数量少,必须谨慎解释结果,然而,他们建议对ERα的评估,应在患者生物材料中通过免疫组织化学进行AR和PGR,以计划未来的临床试验。
    The treatment of ovarian cancer (OC) remains one of the greatest challenges in gynaecological oncology. The presence of classic steroid receptors in OC makes hormone therapy an attractive option; however, the response of OC to hormone therapy is modest. Here, we compared the expression patterns of progesterone (PGR), androgen (AR) and oestrogen alpha (ERα) receptors between serous OC cell lines and non-cancer ovarian cells. These data were analysed in relation to steroid receptor expression profiles from patient tumour samples and survival outcomes using a bioinformatics approach. The results showed that ERα, PGR and AR were co-expressed in OC cell lines, and patient samples from high-grade and low-grade OC co-expressed at least two steroid receptors. High AR expression was negatively correlated, whereas ERα and PGR expression was positively correlated with patient survival. AR showed the opposite expression pattern to that of ERα and PGR in type 1 (SKOV-3) and 2 (OVCAR-3) OC cell lines compared with non-cancer (HOSEpiC) ovarian cells, with AR downregulated in type 1 and upregulated in type 2 OC. A low AR/PGR ratio and a high ESR1/AR ratio were associated with favourable survival outcomes in OC compared with other receptor ratios. Although the results must be interpreted with caution because of the small number of primary tumour samples analysed, they nevertheless suggest that the evaluation of ERα, AR and PGR by immunohistochemistry should be performed in patient biological material to plan future clinical trials.
    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
    背景:内分泌治疗是肿瘤表达雌激素受体α(ERα)的乳腺癌患者最重要的治疗方式。雄激素受体(AR)也在绝大多数(80-90%)ERα阳性肿瘤中表达。AR靶向药物在临床实践中没有使用,但已在多项试验和临床前研究中进行了评估。
    方法:我们进行了一项全基因组研究,以鉴定激素/药物诱导的单核苷酸多态性(SNP)基因型依赖性基因表达,被称为PGx-eQTL,由AR激动剂(双氢睾酮)或部分拮抗剂(恩杂鲁胺)介导,利用先前充分表征的淋巴母细胞细胞系面板。然后使用我们已经发表的三个全基因组关联(GWAS)研究和来自GWAS目录的其他研究来检查鉴定的SNP-基因对与乳腺癌表型的关联。
    结果:我们确定了13个DHT介导的PGx-eQTL基因座和23个Enz介导的PGx-eQTL基因座,它们与ER拮抗剂或芳香化酶抑制剂(AI)治疗后的乳腺癌预后相关,或具有AI的药效学(PD)作用。发现另外30个基因座与癌症风险和性激素结合球蛋白水平有关。顶部基因座涉及IDH2和TMEM9基因,DHT以PGx-eQTLSNP基因型依赖性方式抑制了它们的表达。这两种基因在乳腺癌中都过表达,并与预后较差有关。因此,AR激动剂对这些基因的抑制可能使具有这些SNP的次要等位基因基因型的患者受益.
    结论:我们确定了与风险相关的AR相关PGx-eQTLSNP基因对,可能为乳腺癌个体化治疗提供潜在生物标志物的内分泌治疗的结局和PD效应。
    BACKGROUND: Endocrine therapy is the most important treatment modality of breast cancer patients whose tumors express the estrogen receptor α (ERα). The androgen receptor (AR) is also expressed in the vast majority (80-90%) of ERα-positive tumors. AR-targeting drugs are not used in clinical practice, but have been evaluated in multiple trials and preclinical studies.
    METHODS: We performed a genome-wide study to identify hormone/drug-induced single nucleotide polymorphism (SNP) genotype - dependent gene-expression, known as PGx-eQTL, mediated by either an AR agonist (dihydrotestosterone) or a partial antagonist (enzalutamide), utilizing a previously well characterized lymphoblastic cell line panel. The association of the identified SNPs-gene pairs with breast cancer phenotypes were then examined using three genome-wide association (GWAS) studies that we have published and other studies from the GWAS catalog.
    RESULTS: We identified 13 DHT-mediated PGx-eQTL loci and 23 Enz-mediated PGx-eQTL loci that were associated with breast cancer outcomes post ER antagonist or aromatase inhibitors (AI) treatment, or with pharmacodynamic (PD) effects of AIs. An additional 30 loci were found to be associated with cancer risk and sex-hormone binding globulin levels. The top loci involved the genes IDH2 and TMEM9, the expression of which were suppressed by DHT in a PGx-eQTL SNP genotype-dependent manner. Both of these genes were overexpressed in breast cancer and were associated with a poorer prognosis. Therefore, suppression of these genes by AR agonists may benefit patients with minor allele genotypes for these SNPs.
    CONCLUSIONS: We identified AR-related PGx-eQTL SNP-gene pairs that were associated with risks, outcomes and PD effects of endocrine therapy that may provide potential biomarkers for individualized treatment of breast cancer.
    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
    目的:观察Lathyrol对肾细胞癌(RCC)小鼠雄激素受体(AR)和鞘氨醇激酶2(SPHK2)表达的影响,进一步探讨Lathyrol抑制上皮间质转化(EMT)侵袭和发生的机制。
    方法:构建RCC异种移植小鼠模型,将小鼠随机分为模型组,实验组和阴性对照组。实验组用Lathyrol溶液(20mg/kg)胃内灌胃,模型组灌胃0.9%NaCl(与实验组相同体积),阴性对照组腹腔注射2mg/kg顺铂水溶液。记录小鼠体重和肿瘤体积的变化。Westernblot(WB)用于评估AR的蛋白表达水平,p-AR,CYP17A1,PARP1,E-cadherin,N-钙黏着蛋白,波形蛋白,α-SMA,β-连环蛋白,ZO-1SPHK2,金属基质蛋白酶2(MMP2)的蛋白质表达水平,通过免疫组织化学(IHC)评估肿瘤组织中的MMP9和尿激酶型纤溶酶原激活物(uPA)。免疫荧光(IF)染色后评估肿瘤组织中的AR表达。
    结果:给药14天后,与模型组相比,阴性对照组和实验组的肿瘤体积较低;模型之间的肿瘤体积差异,对照组与实验组比较差异有统计学意义(P<0.05)。三组间体重差异无统计学意义(P>0.05)。在模型组中,AR的蛋白质表达水平,p-AR,CYP17A1、SPHK2和PARP1相对升高,E-cadherin和ZO-1蛋白表达水平相对降低(P<0.05),和N-钙黏着蛋白的表达水平,β-连环蛋白,波形蛋白,α-SMA相对升高(P<0.05)。在阴性对照组和实验组中,AR的蛋白质表达水平,p-AR,CYP17A1、SPHK2和PARP1相对降低(P<0.05),E-cadherin和ZO-1蛋白表达水平相对升高(P<0.05),和N-钙黏着蛋白的表达水平,β-连环蛋白,波形蛋白和α-SMA相对降低(P<0.05)。
    结论:Lathyrol和顺铂抑制肾癌移植瘤的增殖,降低AR的蛋白质表达水平,CYP17A1,SPHK2,PARP1,E-cadherin,ZO-1在肿瘤组织中的表达(P<0.05),并促进N-cadherin的蛋白质表达水平,β-连环蛋白,波形蛋白和α-SMA(P<0.05)。因此,Lathyrol通过影响RCC小鼠AR和SPHK2的表达来降低RCC侵袭和EMT。
    OBJECTIVE: To investigate the effects of Lathyrol on the expression of androgen receptor (AR) and sphingosine kinase 2 (SPHK2) in renal cell carcinoma (RCC) mice and to further explore the mechanism by which Lathyrol inhibits the invasion and incidence of epithelial-mesenchymal transition (EMT).
    METHODS: An RCC xenograft mouse model was constructed, and the mice were randomly divided into a model group, an experiment group and a negative control group. The experiment group was intragastrically gavaged with Lathyrol solution (20 mg/kg), the model group was intragastrically gavaged with 0.9% NaCl (same volume as that used in the experiment group), and the negative control group was injected intraperitoneally with 2 mg/kg cisplatin aqueous solution. Changes in the body weight and tumor volume of the mice were recorded. Western blot (WB) was used to assess the protein expression levels of AR, p-AR, CYP17A1, PARP1, E-cadherin, N-cadherin, vimentin, α-SMA, β-catenin, and ZO-1. Protein expression levels of SPHK2, metal matrix protease 2 (MMP2), MMP9 and urokinase-type plasminogen activator (uPA) in tumor tissues were assessed by immunohistochemistry (IHC). AR expression in tumor tissues was assessed after immunofluorescence (IF) staining.
    RESULTS: After 14 days of drug administration, compared with that in the model group, the tumor volumes in the negative control and experiment groups were lower; the difference in tumor volume among the model, control and experiment groups was statistically significant (P < 0.05). The differences in body weight among the three groups were not statistically significant (P > 0.05). In the model group, the protein expression levels of AR, p-AR, CYP17A1, SPHK2, and PARP1 were relatively increased, the protein expression levels of E-cadherin and ZO-1 were relatively reduced (P < 0.05), and the protein expression levels of N-cadherin, β-catenin, vimentin, and α-SMA were relatively increased (P < 0.05). In the negative control and experiment groups, the protein expression levels of AR, p-AR, CYP17A1, SPHK2, and PARP1 were relatively decreased (P < 0.05), the protein expression levels of E-cadherin and ZO-1 were relatively increased (P < 0.05), and the protein expression levels of N-cadherin, β-catenin, vimentin and α-SMA were relatively decreased (P < 0.05).
    CONCLUSIONS: Lathyrol and cisplatin inhibit the proliferation of RCC xenografts, reduce the protein expression levels of AR, CYP17A1, SPHK2, PARP1, E-cadherin, and ZO-1 in tumor tissues (P < 0.05), and promote the protein expression levels of N-cadherin, β-catenin, vimentin and α-SMA (P < 0.05). Therefore, Lathyrol reduces RCC invasion and EMT by affecting the expression of AR and SPHK2 in RCC mice.
    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
    调节雄激素受体(AR)生物学活性的主要机制涉及越来越多的翻译后修饰(PTM)。在这篇综述中,我们总结了影响该主要转录因子的PTM的结构和功能影响的最新知识。接下来,我们讨论了这些不同PTM之间的串扰和AR蛋白中修饰残基簇的存在。最后,我们讨论了这些共价修饰对疾病如脊髓和延髓肌萎缩症(肯尼迪病)和前列腺癌的病因的影响,以及药物干预的前景。
    A major mechanism to modulate the biological activities of the androgen receptor (AR) involves a growing number of post-translational modifications (PTMs). In this review we summarise the current knowledge on the structural and functional impact of PTMs that affect this major transcription factor. Next, we discuss the cross-talk between these different PTMs and the presence of clusters of modified residues in the AR protein. Finally, we discuss the implications of these covalent modifications for the aetiology of diseases such as spinal and bulbar muscular atrophy (Kennedy\'s disease) and prostate cancer, and the perspectives for pharmacological intervention.
    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
    雄激素受体(AR)是治疗前列腺癌的一个有吸引力的靶点,考虑其在局限性和转移性前列腺癌的发展和进展中的作用。前列腺癌的高全球死亡率负担,尽管药物治疗如雄激素剥夺或AR拮抗剂治疗,强调了探索替代战略的必要性。一种策略涉及使用异双功能降解剂,也被称为蛋白水解靶向嵌合体,它们是抑制扩增或突变靶标的新型小分子疗法。这里,该研究报告了一种新型的基于小脑的AR降解剂,UBX-390,并证明其优于已建立的AR降解剂的活性,例如ARV-110或ARCC-4,在短期和长期治疗条件下的前列腺癌细胞中。UBX-390抑制AR的染色质结合和基因表达,并在治疗抗性前列腺癌患者中显示出AR突变体降解的实质性功效。UBX-390是一种优化的AR降解剂,具有治疗去势抵抗性前列腺癌的巨大潜力。
    The androgen receptor (AR) is an attractive target for treating prostate cancer, considering its role in the development and progression of localized and metastatic prostate cancer. The high global mortality burden of prostate cancer, despite medical treatments such as androgen deprivation or AR antagonist therapy, highlights the need to explore alternative strategies. One strategy involves the use of heterobifunctional degraders, also known as proteolysis-targeting chimeras, which are novel small-molecule therapeutics that inhibit amplified or mutated targets. Here, the study reports a novel cereblon-based AR degrader, UBX-390, and demonstrates its superior activity over established AR degraders, such as ARV-110 or ARCC-4, in prostate cancer cells under short- and long-term treatment conditions. UBX-390 suppresses chromatin binding and gene expression of AR and demonstrates substantial efficacy in the degradation of AR mutants in patients with treatment-resistant prostate cancer. UBX-390 is presented as an optimized AR degrader with remarkable potential for treating castration-resistant prostate cancer.
    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