CRPC

CRPC
  • 文章类型: Journal Article
    背景:在美国,2022年共有268490名男性被发现患有前列腺癌,因此使其成为男性中最常见的癌症,占男性人群所有癌症的27%。在所有男性癌症中,是第五大死因,有34,500人死亡,死亡率为11%。2019年,病例总数为94,748例,是男性中的主要癌症,占所有男性癌症的11%。在死亡率方面,它排名第七,13,217例死亡,死亡率为1.6%。然而,转移性去势敏感性前列腺癌(mCSPC)的新治疗方案已经出现.多西他赛已被证明对mCSPC和去势抵抗前列腺癌(CRPC)均有效。多西他赛尚未在日本获得批准,也没有在大规模研究中得到验证。此外,多西他赛治疗后可以使用几种药物,但尚不清楚哪一种最有效。我们使用了一个大型的日本健康保险数据库,以确定在接受多西他赛的患者中,哪种药物作为下一线治疗最有效。
    方法:我们使用了来自医疗机构的数据,使用了诊断程序组合(DPC),它提供了对医学分类的全面评估。医疗数据视觉数据库覆盖了日本约23%的DPC医院。这项研究分析了2938例接受多西他赛的mCSPC患者,其次是CRPC,2008年4月至2021年12月。该研究集中在三种药物上:恩扎鲁他胺,醋酸阿比特龙,和卡巴他赛.由于患者数量少,其他药物被排除在外。分析了以下数据:年龄,CRPC诊断日期,骨转移的存在,药物类型,和预后。
    结果:本研究纳入了1997例接受多西他赛预先治疗mCSPC后的CRPC患者(恩扎鲁他胺[ENZ]组,n=998;醋酸阿比特龙[ABI]组,n=617;和卡巴他赛[CBZ]组,n=382)。恩杂鲁胺组自药物开始的总生存期(OS)时间为456天,明显长于卡巴他赛组(p=0.017,HR0.94)(ENZ:ABIp=0.54,HR0.94;ABI:CBZp=0.14,HR0.75)。在接受恩杂鲁胺作为二线药物的组中,还比较了三线药物的OS,使用醋酸阿比特龙作为三线药物的组(ENZ-ABI组),和使用醋酸阿比特龙作为二线药物的组。在ENZ-ABI组和ABI-ENZ组之间比较三线药物开始时的OS,接受恩扎鲁他胺作为三线药物,但没有显着差异(269vs.281天,p=0.85;HR1.03)。
    结论:ENZ显示在停止多西他赛后相对于卡巴他赛延长OS。与ABI和CBZ相比,ENZ与更长的药物使用时间相关。
    BACKGROUND: In the United States, a total of 268,490 men were found to have prostate cancer in 2022, thus making it the most common cancer in men, accounting for 27% of all cancers in the male population. Among all cancers in men, it was the fifth leading cause of death, with 34,500 deaths and a mortality rate of 11%. In 2019, the total number of cases was 94,748, making it the leading cancer in males, accounting for 11% of all male cancers. In terms of mortality, it ranked seventh, with 13,217 deaths and a mortality rate of 1.6%. However, new treatment options for metastatic castration-sensitive prostate cancer (mCSPC) have emerged. Docetaxel has been shown to be effective for both mCSPC and castration-resistant prostate cancer (CRPC). Upfront docetaxel has not been approved in Japan, nor has it been validated in large-scale studies. Furthermore, several agents can be used after docetaxel treatment, but it is unclear which is the most effective. We used a large Japanese health insurance database to determine which agent would be the most effective as a next-line therapy in patients who had received docetaxel.
    METHODS: We used data from medical institutions using the Diagnosis Procedure Combination (DPC), which provides a comprehensive evaluation of medical classifications. The Medical Data Vision database covers approximately 23% of DPC hospitals in Japan. This study analyzed 2938 patients with mCSPC who received docetaxel, followed by CRPC, between April 2008 and December 2021. The study focused on three agents: enzalutamide, abiraterone acetate, and cabazitaxel. Other agents were excluded due to the small number of patients. The following data were analyzed: age, date of CRPC diagnosis, presence of bone metastasis, drug type, and prognosis.
    RESULTS: This study included 1997 patients with CRPC after upfront docetaxel therapy for mCSPC (enzalutamide [ENZ] group, n = 998; abiraterone acetate [ABI] group, n = 617; and cabazitaxel [CBZ] group, n = 382). The overall survival (OS) time from drug initiation was 456 days in the enzalutamide group, which was significantly longer than that in the cabazitaxel group (p = 0.017, HR 0.94) (ENZ: ABI p = 0.54, HR 0.94; ABI: CBZ p = 0.14, HR 0.75). OS was also compared for the third-line drug in the group that received enzalutamide as the second-line drug, the group that used abiraterone acetate as the third-line drug (ENZ-ABI group), and the group that used abiraterone acetate as the second-line drug. OS from the start of the third-line drug was compared between the ENZ-ABI group and the ABI-ENZ group, which received enzalutamide as the third-line drug, but showed no significant difference (269 vs. 281 days, p = 0.85; HR 1.03).
    CONCLUSIONS: ENZ was shown to prolong OS relative to cabazitaxel after the cessation of docetaxel. ENZ was associated with a longer duration of drug use than ABI and CBZ.
    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
    前列腺癌(PCa)是男性泌尿生殖系统最常诊断的恶性肿瘤。传统上,局限性PCa的治疗基于手术或放疗,而激素疗法用于晚期.然而,放射性标记的实施彻底改变了前列腺癌的面貌。具体来说,已经在PCa治疗时代的不同方面研究了前列腺特异性膜抗原(PSMA)。
    对PSMA放射性标记对前列腺癌治疗的影响进行了文献综述。PSMA示踪剂最初用作成像技术。之后,PSMA标记有呈现细胞毒性能力的同位素,例如Luttium-117和actinium-225,而关于使用放射性配体免疫疗法的报道也存在。同时,正在进行的试验检查了新型放射性核素的开发以及PSMA靶向配体的演变。
    目前,前列腺癌的PSMA放射性配体治疗被批准在疾病的转移阶段。同时,关于其在较不晚期阶段的可能作用,存在各种试验。然而,在这些实现之前,应该解决大量的参数,如PSMA剂量,剂量测定问题及其安全性。未来必须进行精心设计的研究,并选择适当的患者,以进一步探索PSMA放射性配体疗法的观点。
    UNASSIGNED: The prostate cancer (PCa) consists the most frequently diagnosed malignancy of urogenital system in males. Traditionally, treatment of localized PCa was based on surgery or radiotherapy while hormonotherapy was used in more advanced stages. However, the implementation of radiolabels has revolutionized the landscape of prostate cancer. Specifically, prostate-specific membrane antigen (PSMA) has been investigated in different aspects of PCa therapeutic era.
    UNASSIGNED: A literature review is presented about the implications of PSMA radiolabels on prostate cancer treatment. PSMA tracers were initially used as an imaging technique. Afterwards, PSMA labeled with isotopes presenting cytotoxic abilities, such as lutetium-117 and actinium-225, while reports exist about the use of radioligand immunotherapy. Meanwhile, ongoing trials examine the development of novel radionuclides as well as the evolution of the PSMA-targeted ligands.
    UNASSIGNED: Currently, PSMA radioligand treatment of prostate cancer is approved in the metastatic stage of the disease. Meanwhile, a variety of trials exist about its possible role in less advanced stages. However, plenty of parameters should be addressed before these implementations, such as PSMA dosage, dosimetry issues, and its safety profile. A future well-designed study with proper patient selection is mandatory to further explore PSMA radioligand theranostics perspectives.
    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:晚期前列腺癌(PCa)中由雄激素受体(AR)信号通路持续激活驱动的内分泌抵抗是致命的。表征异常AR途径激活的潜在机制以寻找潜在的治疗策略尤为重要。RacGTPase激活蛋白1(RACGAP1)是一种特异性的GTPase激活蛋白。作为一种新的肿瘤原癌基因,RACGAP1的过度表达与多种肿瘤的发生有关。
    方法:采用生物信息学方法分析RACGAP1与AR的表达水平以及AR通路激活的关系。进行qRT-PCR和蛋白质印迹测定以评估PCa细胞中AR/AR-V7和RACGAPl的表达。进行免疫沉淀和免疫荧光实验以检测RACGAP1与AR/AR-V7之间的相互作用和共定位。进行了增益和功能丧失分析,以研究RACGAP1在PCa细胞中的生物学作用,使用MTS和集落形成测定。进行体内实验以评估RACGAP1抑制对肿瘤生长的影响。
    结果:RACGAP1是由AR激活的基因,在CRPC和恩杂鲁胺耐药的PCa患者中,该指标明显上调。AR通过结合其启动子区转录激活RACGAP1表达。相互,核RACGAP1与AR和AR-V7的N末端结构域(NTD)结合,阻断了它们与E3泛素连接酶MDM2的相互作用。因此,这阻止了泛素-蛋白酶体依赖性途径中AR/AR-V7的降解。值得注意的是,RACGAP1和AR/AR-V7之间的正反馈回路导致CRPC的内分泌治疗抵抗。恩杂鲁胺和靶向RACGAP1的体内胆固醇缀合的RIG-IsiRNA药物的组合诱导PCa的异种移植肿瘤生长的有效抑制。
    结论:总之,我们的结果表明,RACGAP1和AR/AR-V7之间的相互调节有助于PCa的内分泌抵抗。这些发现突出了RACGAP1抑制和恩杂鲁胺联合治疗晚期PCa的治疗潜力。
    BACKGROUND: Endocrine resistance driven by sustained activation of androgen receptor (AR) signaling pathway in advanced prostate cancer (PCa) is fatal. Characterization of mechanisms underlying aberrant AR pathway activation to search for potential therapeutic strategy is particularly important. Rac GTPase-activating protein 1 (RACGAP1) is one of the specific GTPase-activating proteins. As a novel tumor proto-oncogene, overexpression of RACGAP1 was related to the occurrence of various tumors.
    METHODS: Bioinformatics methods were used to analyze the relationship of expression level between RACGAP1 and AR as well as AR pathway activation. qRT-PCR and western blotting assays were performed to assess the expression of AR/AR-V7 and RACGAP1 in PCa cells. Immunoprecipitation and immunofluorescence experiments were conducted to detect the interaction and co-localization between RACGAP1 and AR/AR-V7. Gain- and loss-of-function analyses were conducted to investigate the biological roles of RACGAP1 in PCa cells, using MTS and colony formation assays. In vivo experiments were conducted to evaluate the effect of RACGAP1 inhibition on the tumor growth.
    RESULTS: RACGAP1 was a gene activated by AR, which was markedly upregulated in PCa patients with CRPC and enzalutamide resistance. AR transcriptionally activated RACGAP1 expression by binding to its promoter region. Reciprocally, nuclear RACGAP1 bound to the N-terminal domain (NTD) of both AR and AR-V7, blocking their interaction with the E3 ubiquitin ligase MDM2. Consequently, this prevented the degradation of AR/AR-V7 in a ubiquitin-proteasome-dependent pathway. Notably, the positive feedback loop between RACGAP1 and AR/AR-V7 contributed to endocrine therapy resistance of CRPC. Combination of enzalutamide and in vivo cholesterol-conjugated RIG-I siRNA drugs targeting RACGAP1 induced potent inhibition of xenograft tumor growth of PCa.
    CONCLUSIONS: In summary, our results reveal that reciprocal regulation between RACGAP1 and AR/AR-V7 contributes to the endocrine resistance in PCa. These findings highlight the therapeutic potential of combined RACGAP1 inhibition and enzalutamide in treatment of advanced PCa.
    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
    尽管恩杂鲁胺在去势抵抗前列腺癌(CRPC)中的初始疗效,不可避免的阻力仍然是一个重大挑战。这里,恩杂鲁胺和铜离子载体(elesclomol/双硫仑)报道了CRPC细胞中铜依赖性细胞死亡(角化)的协同诱导。机械上,恩杂鲁胺治疗增加CRPC细胞的线粒体依赖性,使它们容易发生角化,正如用铜螯合剂四硫钼酸盐的特异性逆转所证明的那样。这种易感性的特征是角化的标志,包括脂化蛋白聚集和铁硫簇蛋白不稳定。有趣的是,线粒体基质还原酶,FDX1,特别是与elesclomol敏感性相关,这表明两个铜离子载体之间存在潜在的机械差异。值得注意的是,这种协同效应超出了体外模型,证明在22Rv1异种移植物中的功效,小鼠Ptenp53敲除类器官。重要的是,恩扎鲁他胺显著增强恩扎鲁他胺抗性细胞中铜离子载体介导的细胞毒性。总的来说,这些发现表明恩杂鲁胺和铜离子载体协同诱导细胞凋亡,为CRPC提供了一个有希望的治疗途径,可能包括恩杂鲁胺耐药病例。
    Despite the initial efficacy of enzalutamide in castration-resistant prostate cancer (CRPC), inevitable resistance remains a significant challenge. Here, the synergistic induction of copper-dependent cell death (cuproptosis) in CRPC cells is reported by enzalutamide and copper ionophores (elesclomol/disulfiram). Mechanistically, enzalutamide treatment increases mitochondrial dependence in CRPC cells, rendering them susceptible to cuproptosis, as evidenced by specific reversal with the copper chelator tetrathiomolybdate. This susceptibility is characterized by hallmarks of cuproptosis, including lipoylated protein aggregation and iron-sulfur cluster protein instability. Interestingly, the mitochondrial matrix reductase, FDX1, specifically correlates with elesclomol sensitivity, suggesting a potential mechanistic divergence between the two copper ionophores. Notably, this synergistic effect extends beyond in vitro models, demonstrating efficacy in 22Rv1 xenografts, mouse Pten p53 knockout organoids. Importantly, enzalutamide significantly enhances copper ionophore-mediated cytotoxicity in enzalutamide-resistant cells. Collectively, these findings indicate that enzalutamide and copper ionophores synergistically induce cuproptosis, offering a promising therapeutic avenue for CRPC, potentially including enzalutamide-resistant cases.
    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
    全球范围内前列腺癌发病率和死亡率的增加显著影响男性患者的寿命,强调了解其致病机制和调节肿瘤进展的相关分子变化以有效预防和治疗的紧迫性。RNA修饰,一个重要的转录后调控过程,深刻影响肿瘤细胞的生长和代谢,塑造细胞命运.已知有超过170种RNA修饰方法,专注于N6-甲基腺苷的突出研究,N7-甲基鸟苷,N1-甲基腺苷,5-甲基胞苷,假尿苷,和N4-乙酰胞苷修饰。这些改变在转录后错综复杂地调节编码和非编码RNA,影响RNA和蛋白质表达水平的稳定性。本文深入探讨了前列腺癌肿瘤细胞中与各种RNA修饰相关的最新进展和挑战,肿瘤微环境,和核心信号分子雄激素受体。旨在为分子诊断提供新的研究目标和途径,治疗策略,并改善前列腺癌的预后。
    The increasing incidence and mortality of prostate cancer worldwide significantly impact the life span of male patients, emphasizing the urgency of understanding its pathogenic mechanism and associated molecular changes that regulate tumor progression for effective prevention and treatment. RNA modification, an important post-transcriptional regulatory process, profoundly influences tumor cell growth and metabolism, shaping cell fate. Over 170 RNA modification methods are known, with prominent research focusing on N6-methyladenosine, N7-methylguanosine, N1-methyladenosine, 5-methylcytidine, pseudouridine, and N4-acetylcytidine modifications. These alterations intricately regulate coding and non-coding RNA post-transcriptionally, affecting the stability of RNA and protein expression levels. This article delves into the latest advancements and challenges associated with various RNA modifications in prostate cancer tumor cells, tumor microenvironment, and core signaling molecule androgen receptors. It aims to provide new research targets and avenues for molecular diagnosis, treatment strategies, and improvement of the prognosis in prostate 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
    前列腺癌(PCa)是美国男性中最常见的癌症,也是癌症死亡的第二大原因。雄激素剥夺疗法(ADT)已被系统地用作PCa患者的一线疗法。尽管最初的反应,大多数接受ADT的患者最终经历了肿瘤进展为去势抵抗前列腺癌(CRPC),进一步导致肿瘤转移到远处器官。因此,确定PCa进展的关键分子机制对于开发转移性PCa的新疗法仍然至关重要.以前,我们发现肿瘤抑制性miR-99b-5p在侵袭性非裔美国人(AA)PCa和欧裔美国人(EA)CRPC中经常下调,导致mTOR的上调,雄激素受体(AR),和HIF-1α信号。鉴于mTOR和HIF-1α信号传导是触发上皮间质转化(EMT)激活的关键上游途径,我们假设miR-99b-5p可能在调节EMT介导的PCa转移中起重要的功能作用.为了检验这个假设,一系列的细胞生物学,生物化学,和体外功能测定(伤口愈合,Transwell迁移,细胞/ECM粘附,和毛细管样管形成测定),以检查miR-99b-5p模拟物在调节EMT介导的PCa转移过程中的作用。我们的研究结果表明,miR-99b-5p同时靶向MTOR和AR信号,导致E-cadherin的上调,蜗牛/N-钙黏着蛋白/波形蛋白的下调,抑制EMT介导的PCa转移。单独的MiR-99b-5p以及与恩杂鲁胺或阿比特龙组合显着抑制EMT介导的AAPCa和EACRPC的转移。
    Prostate cancer (PCa) is the most frequently diagnosed cancer and second leading cause of cancer deaths among American men. Androgen deprivation therapy (ADT) has been systemically applied as a first-line therapy for PCa patients. Despite the initial responses, the majority of patients under ADT eventually experienced tumor progression to castration-resistant prostate cancer (CRPC), further leading to tumor metastasis to distant organs. Therefore, identifying the key molecular mechanisms underlying PCa progression remains crucial for the development of novel therapies for metastatic PCa. Previously, we identified that tumor-suppressive miR-99b-5p is frequently downregulated in aggressive African American (AA) PCa and European American (EA) CRPC, leading to upregulation of mTOR, androgen receptor (AR), and HIF-1α signaling. Given the fact that mTOR and HIF-1α signaling are critical upstream pathways that trigger the activation of epithelial-mesenchymal transition (EMT), we hypothesized that miR-99b-5p may play a critical functional role in regulating EMT-mediated PCa metastasis. To test this hypothesis, a series of cell biology, biochemical, and in vitro functional assays (wound healing, transwell migration, cell/ECM adhesion, and capillary-like tube formation assays) were performed to examine the effects of miR-99b-5p mimic on regulating EMT-mediated PCa metastasis processes. Our results have demonstrated that miR-99b-5p simultaneously targets MTOR and AR signaling, leading to upregulation of E-cadherin, downregulation of Snail/N-cadherin/Vimentin, and suppression of EMT-mediated PCa metastasis. MiR-99b-5p alone and in combination with enzalutamide or abiraterone significantly inhibits the EMT-mediated metastasis of AA PCa and EA CRPC.
    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
    背景:细胞周期蛋白依赖性激酶12(CDK12)缺陷型前列腺癌定义了去势抵抗性前列腺癌(CRPC)的亚型,预后不良。目前的治疗,包括PARP抑制剂,对这种CRPC亚型的治疗效果最小,潜在的机制仍然难以捉摸。
    方法:基于生物信息学分析,我们评估了CDK12缺乏与前列腺癌患者预后和治疗抵抗之间的关系。此外,我们使用CRISPR-Cas9技术和基于质谱的代谢组学分析来揭示CDK12缺陷型CRPC的代谢特征.为了阐明CDK12缺陷介导的CRPC代谢重编程的具体机制,我们利用细胞RNA-seq分析和其他分子生物学技术,包括细胞活性氧探针,线粒体功能测定,ChIP-qPCR和RNA稳定性分析,阐明CDK12在调节线粒体功能中的作用及其对铁凋亡的贡献。最后,通过体外药物敏感性测试和小鼠体内实验,我们确定了电子传递链(ETC)抑制剂IACS-010759对CDK12缺陷型CRPC的治疗作用.
    结果:CDK12缺陷型前列腺癌重新编程细胞能量代谢以支持其积极进展。特别是,CDK12缺乏增强了线粒体呼吸链的电子转移和ATP合成,从而在CRPC细胞中产生了铁凋亡潜能。然而,CDK12缺乏症下调ACSL4表达,抵消脂质氧化应激,导致CRPC细胞从铁性凋亡中逃脱。此外,靶向ETC在体外和体内基本上抑制CDK12缺陷的CRPC细胞的增殖,提示CDK12缺陷型前列腺癌治疗的潜在新靶点。
    结论:我们的研究结果表明,CDK12缺乏的CRPC中的能量和脂质代谢共同推动了CRPC的进展,并为CDK12缺乏的前列腺癌患者的不良预后提供了代谢方面的见解。
    结论:CDK12缺乏通过细胞代谢重编程促进去势抵抗性前列腺癌(CRPC)进展。CRPC中的CDK12缺乏导致更活跃的线粒体电子传递链(ETC),确保高效的电池能源供应。CDK12磷酸化RNAPolII以确保ACSL4的转录以调节铁凋亡。线粒体ETC抑制剂对CDK12缺陷的CRPC细胞表现出更好的选择性,为这种亚型的CRPC患者提供了一种有希望的新的治疗方法。
    BACKGROUND: Cyclin-dependent kinase 12 (CDK12)-deficient prostate cancer defines a subtype of castration-resistant prostate cancer (CRPC) with a poor prognosis. Current therapy, including PARP inhibitors, shows minimal treatment efficacy for this subtype of CRPC, and the underlying mechanism remains elusive.
    METHODS: Based on bioinformatics analysis, we evaluated the relationship between CDK12 deficiency and prostate cancer patient\'s prognosis and treatment resistance. Furthermore, we used CRISPR-Cas9 technology and mass spectrometry-based metabolomic profiling to reveal the metabolic characteristics of CDK12-deficient CRPC. To elucidate the specific mechanisms of CDK12 deficiency-mediated CRPC metabolic reprogramming, we utilized cell RNA-seq profiling and other molecular biology techniques, including cellular reactive oxygen species probes, mitochondrial function assays, ChIP-qPCR and RNA stability analyses, to clarify the role of CDK12 in regulating mitochondrial function and its contribution to ferroptosis. Finally, through in vitro drug sensitivity testing and in vivo experiments in mice, we identified the therapeutic effects of the electron transport chain (ETC) inhibitor IACS-010759 on CDK12-deficient CRPC.
    RESULTS: CDK12-deficient prostate cancers reprogramme cellular energy metabolism to support their aggressive progression. In particular, CDK12 deficiency enhanced the mitochondrial respiratory chain for electronic transfer and ATP synthesis to create a ferroptosis potential in CRPC cells. However, CDK12 deficiency downregulated ACSL4 expression, which counteracts the lipid oxidation stress, leading to the escape of CRPC cells from ferroptosis. Furthermore, targeting the ETC substantially inhibited the proliferation of CDK12-deficient CRPC cells in vitro and in vivo, suggesting a potential new target for the therapy of CDK12-deficient prostate cancer.
    CONCLUSIONS: Our findings show that energy and lipid metabolism in CDK12-deficient CRPC work together to drive CRPC progression and provide a metabolic insight into the worse prognosis of CDK12-deficient prostate cancer patients.
    CONCLUSIONS: CDK12 deficiency promotes castration-resistant prostate cancer (CRPC) progression by reprogramming cellular metabolism. CDK12 deficiency in CRPC leads to a more active mitochondrial electron transport chain (ETC), ensuring efficient cell energy supply. CDK12 phosphorylates RNA Pol II to ensure the transcription of ACSL4 to regulate ferroptosis. Mitochondrial ETC inhibitors exhibit better selectivity for CDK12-deficient CRPC cells, offering a promising new therapeutic approach for this subtype of CRPC patients.
    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
    雄激素受体信号传导对于前列腺癌治疗抵抗的发展至关重要。在类固醇生成酶中,3β-羟基类固醇脱氢酶(3βHSD)在性腺外雄激素合成中起关键作用,特别是3βHSD1。与原发性前列腺肿瘤相比,在去势抵抗的前列腺癌肿瘤中观察到3βHSD的表达增加,表明他们参与了阉割抵抗。最近的研究将3βHSD1与对雄激素受体信号传导抑制剂的抗性联系起来。3βHSD1表达的调控涉及多种因素,包括转录因子,微环境影响,和转录后修饰。此外,HSD3B1基因型的临床意义,特别是rs1047303变体已经被广泛研究。HSD3B1基因型对治疗结果的影响因治疗而异,提示HSD3B1基因分型在个性化医疗中的潜力。靶向3βHSD可能是前列腺癌治疗的有希望的策略。总的来说,了解3βHSD的作用及其遗传变异可能有助于前列腺癌新疗法的开发和优化。
    Androgen receptor signaling is crucial for the development of treatment resistance in prostate cancer. Among steroidogenic enzymes, 3β-hydroxysteroid dehydrogenases (3βHSDs) play critical roles in extragonadal androgen synthesis, especially 3βHSD1. Increased expression of 3βHSDs is observed in castration-resistant prostate cancer tumors compared with primary prostate tumors, indicating their involvement in castration resistance. Recent studies link 3βHSD1 to resistance to androgen receptor signaling inhibitors. The regulation of 3βHSD1 expression involves various factors, including transcription factors, microenvironmental influences, and posttranscriptional modifications. Additionally, the clinical significance of HSD3B1 genotypes, particularly the rs1047303 variant, has been extensively studied. The impact of HSD3B1 genotypes on treatment outcomes varies according to the therapy administered, suggesting the potential of HSD3B1 genotyping for personalized medicine. Targeting 3βHSDs may be a promising strategy for prostate cancer management. Overall, understanding the roles of 3βHSDs and their genetic variations may enable the development and optimization of novel treatments for 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
    雄激素剥夺治疗(ADT)后去势抵抗性前列腺癌(CRPC)的出现与恶性肿瘤增加和治疗选择有限有关。本研究旨在探讨免疫细胞浸润和炎症细胞因子与YAP1/AR/PSA轴之间的潜在联系,通过探索它们与自噬的相互作用。我们的研究表明,与雄激素依赖性前列腺癌(ADPC)和良性前列腺增生(BPH)的组织相比,CRPC组织中Yes相关蛋白1(YAP1)的表达水平升高。此外,在CRPC组织中观察到YAP1和PSA表达之间的相关性,提示YAP1可能对CRPC内PSA表达有调节作用。C4-2细胞中YAP1表达增强导致雄激素受体(AR)核易位和细胞内前列腺特异性抗原(PSA)水平上调。相反,抑制YAP1导致PSA表达降低,提示YAP1可能通过促进AR核输入来正向调节去势抵抗性前列腺癌(CRPC)的PSA。自噬活性的调节对YAP1、AR、PSA。此外,免疫和炎症研究的最新进展为靶向前列腺癌(PC)的潜在治疗提供了有希望的途径。
    The emergence of castration-resistant prostate cancer (CRPC) following androgen deprivation therapy (ADT) is associated with increased malignancy and limited treatment options. This study aims to investigate potential connections between immune cell infiltration and inflammatory cytokines with the YAP1/AR/PSA axis by exploring their interactions with autophagy. Our research reveals heightened levels of Yes-associated protein 1 (YAP1) expression in CRPC tissues compared with tissues from androgen-dependent prostate cancer (ADPC) and benign prostate hyperplasia (BPH). Additionally, a correlation was observed between YAP1 and PSA expressions in CRPC tissues, suggesting that YAP1 may exert a regulatory influence on PSA expression within CRPC. Enhanced YAP1 expression in C4-2 cells resulted in the upregulation of androgen receptor (AR) nuclear translocation and intracellular prostate-specific antigen (PSA) levels. Conversely, the suppression of YAP1 led to a decrease in PSA expression, suggesting that YAP1 may positively regulate the PSA in castration-resistant prostate cancer (CRPC) by facilitating AR nuclear import. The modulation of the autophagy activity exerts a significant impact on the expression levels of YAP1, the AR, and the PSA. Moreover, recent advancements in immunity and inflammation studies present promising avenues for potential therapies targeting prostate cancer (PC).
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • DOI:
    文章类型: Journal Article
    恩扎鲁胺是一种用于治疗前列腺癌(PC)的药物,多西他赛是一种用于化疗治疗多种癌症类型的药物,包括PC。这些药物在治疗去势抵抗性前列腺癌(CRPC)中的有效性较差,因此CRPC在很大程度上仍然无法治愈。然而,脂肪酸结合蛋白5(FABP5)的生物抑制剂,dmrFABP5是FABP5的突变形式,不能与脂肪酸结合,最近已显示能够抑制培养的CRPC细胞的致瘤性和转移。本研究调查了dmrFABP5与恩杂鲁胺或多西他赛联合抑制PC细胞致瘤特性的可能协同作用。包括细胞活力,迁移,软琼脂中的侵袭和集落增殖。当dmrFABP5与恩杂鲁胺组合用于雄激素响应性PC22RV1细胞时,观察到对这些特性的高度显着的协同抑制作用。此外,当dmrFABP5与多西他赛联合时,也观察到高度显著的协同抑制作用,并添加到22RV1细胞和高度恶性的细胞中,雄激素受体(AR)阴性Du145细胞。当添加到FABP5阴性细胞系LNCaP时,DmrFABP5单独不能产生任何抑制作用,尽管恩杂鲁胺作为单一药物使用时可以显着抑制LNCaP细胞。dmrFABP5的这些协同抑制作用是通过中断PC细胞中FABP5相关的信号转导途径而产生的。因此,DMrFABP5似乎不仅是一种潜在的单一治疗剂,但它也可以与现有药物联合使用,以抑制AR阳性和AR阴性的PC。
    Enzalutamide is a drug used to treat prostate cancer (PC) and docetaxel is a drug for chemotherapeutic treatment of diverse cancer types, including PC. The effectiveness of these drugs in treating castration-resistant prostate cancer (CRPC) is poor and therefore CRPC is still largely incurable. However, the bio-inhibitor of fatty acid-binding protein 5 (FABP5), dmrFABP5, which is a mutant form of FABP5 incapable of binding to fatty acids, has been shown recently to be able to suppress the tumorigenicity and metastasis of cultured CRPC cells. The present study investigated the possible synergistic effect of dmrFABP5 combined with either enzalutamide or docetaxel on suppressing the tumorigenic properties of PC cells, including cell viability, migration, invasion and colony proliferation in soft agar. A highly significant synergistic inhibitory effect on these properties was observed when dmrFABP5 was used in combination with enzalutamide on androgen-responsive PC 22RV1 cells. Moreover, a highly significant synergistic inhibitory effect was also observed when dmrFABP5 was combined with docetaxel, and added to 22RV1 cells and to the highly malignant, androgen-receptor (AR)-negative Du145 cells. DmrFABP5 alone failed to produce any suppressive effect when added to the FABP5-negative cell line LNCaP, although enzalutamide could significantly suppress LNCaP cells when used as a single agent. These synergistic inhibitory effects of dmrFABP5 were produced by interrupting the FABP5-related signal transduction pathway in PC cells. Thus, dmrFABP5 appears to be not only a potential single therapeutic agent, but it may also be used in combination with existing drugs to suppress both AR-positive and AR-negative PC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号