mTOR inhibitors

mTOR 抑制剂
  • 文章类型: Journal Article
    结节性硬化症(TSC)是一种常染色体显性疾病,其特征是中枢神经系统错构瘤的发展,心,皮肤,肺,肾脏和其他表现,包括癫痫发作,皮质块茎,径向迁移线,自闭症和认知障碍。该疾病与TSC1或TSC2基因的致病变异有关,导致mTOR通路的过度激活,细胞生长和新陈代谢的关键调节剂。因此,mTOR通路的过度激活导致异常的组织增殖和实体瘤的发展。肾脏受累于TSC的特点是囊性病变的发展,肾细胞癌和肾血管平滑肌脂肪瘤,可能会进展并引起疼痛,出血,肾功能丧失.在过去的几年里,TSC的治疗方法发生了明显的转变,特别是在解决肾脏表现。mTOR抑制剂已成为主要的治疗选择,而像肾切除术和栓塞术这样的手术干预措施主要用于对临床治疗无反应的并发症,如严重的肾出血。本文就TSC的主要临床特点进行综述,肾脏受累的潜在机制,肾脏病变治疗的最新进展,和未来的前景。
    Tuberous sclerosis complex (TSC) is an autosomal dominant disease characterized by the development of hamartomas in the central nervous system, heart, skin, lungs, and kidneys and other manifestations including seizures, cortical tubers, radial migration lines, autism and cognitive disability. The disease is associated with pathogenic variants in the TSC1 or TSC2 genes, resulting in the hyperactivation of the mTOR pathway, a key regulator of cell growth and metabolism. Consequently, the hyperactivation of the mTOR pathway leads to abnormal tissue proliferation and the development of solid tumors. Kidney involvement in TSC is characterized by the development of cystic lesions, renal cell carcinoma and renal angiomyolipomas, which may progress and cause pain, bleeding, and loss of kidney function. Over the past years, there has been a notable shift in the therapeutic approach to TSC, particularly in addressing renal manifestations. mTOR inhibitors have emerged as the primary therapeutic option, whereas surgical interventions like nephrectomy and embolization being reserved primarily for complications unresponsive to clinical treatment, such as severe renal hemorrhage. This review focuses on the main clinical characteristics of TSC, the mechanisms underlying kidney involvement, the recent advances in therapy for kidney lesions, and the future perspectives.
    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:针对2019年冠状病毒病(COVID-19)的疫苗接种在淋巴管平滑肌瘤病(LAM)患者中的安全性和有效性尚不清楚。这项研究调查了COVID-19疫苗的犹豫,疫苗的安全性和有效性,LAM患者的COVID-19症状。
    结果:总计,181名LAM患者和143名健康个体回答了问卷。LAM患者的接种率为77.34%,15.7%的接种疫苗的LAM患者出现不良事件。接种疫苗降低了LAM患者发生厌食症的风险[OR:0.17,95%CI:(0.07,0.43)],肌痛[OR:0.34,95%CI:(0.13,0.84)],和不适应[OR:0.34,95%CI:(0.14,0.84)]。在LAM患者中,使用mTOR抑制剂可降低COVID-19期间出现症状的风险,包括疲劳[OR:0.18,95%CI:(0.03,0.95)],厌食症[OR:0.30,95%CI:(0.09,0.96)],和不适应[OR:0.20,95%CI:(0.06,0.67)]。
    结论:LAM人群的疫苗接种率低于一般人群,22.7%(41/181)的LAM患者对COVID-19疫苗有犹豫。然而,LAM队列中COVID-19疫苗接种的安全性与健康人群相当,和COVID-19疫苗接种降低了LAM患者COVID-19症状的发生率。此外,mTOR抑制剂似乎不能确定COVID-19期间LAM患者出现并发症的风险更大。
    BACKGROUND: The safety and efficacy of vaccination against coronavirus disease 2019 (COVID-19) in patients with lymphangioleiomyomatosis (LAM) is still unclear. This study investigates COVID-19 vaccine hesitancy, vaccine safety and efficacy, and COVID-19 symptoms in LAM patients.
    RESULTS: In total, 181 LAM patients and 143 healthy individuals responded to the questionnaire. The vaccination rate of LAM patients was 77.34%, and 15.7% of vaccinated LAM patients experienced adverse events. Vaccination decreased the risk of LAM patients developing anorexia [OR: 0.17, 95% CI: (0.07, 0.43)], myalgia [OR: 0.34, 95% CI: (0.13, 0.84)], and ageusia [OR: 0.34, 95% CI: (0.14, 0.84)]. In LAM patients, a use of mTOR inhibitors reduced the risk of developing symptoms during COVID-19, including fatigue [OR: 0.18, 95% CI: (0.03, 0.95)], anorexia [OR: 0.30, 95% CI: (0.09, 0.96)], and ageusia [OR: 0.20, 95% CI: (0.06, 0.67)].
    CONCLUSIONS: Vaccination rates in the LAM population were lower than those in the general population, as 22.7% (41/181) of LAM patients had hesitations regarding the COVID-19 vaccine. However, the safety of COVID-19 vaccination in the LAM cohort was comparable to the healthy population, and COVID-19 vaccination decreased the incidence of COVID-19 symptoms in LAM patients. In addition, mTOR inhibitors seem not to determine a greater risk of complications in patients with LAM during COVID-19.
    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
    哺乳动物雷帕霉素靶标(mTOR)的抑制剂,依维莫司,替西罗莫司和雷帕霉素,具有广泛的临床应用;然而,与其他化学治疗剂不可避免的情况一样,抗性发展制约了它们的有效性。一种推定的耐药机制是促进自噬,这是抑制mTOR信号通路的直接结果。自噬主要被认为是一种细胞保护生存机制。其中细胞质成分被回收以产生能量和代谢中间体。依维莫司和替西罗莫司诱导的自噬似乎在很大程度上发挥了保护作用。而细胞毒性功能似乎在雷帕霉素的情况下占主导地位。在这篇综述中,我们概述了在不同肿瘤模型中mTOR抑制剂诱导的自噬,以确定自噬靶向是否可以作为与mTOR抑制相关的辅助治疗的临床应用。
    The inhibitors of mammalian target of rapapmycin (mTOR), everolimus, temsirolimus and rapamycin, have a wide range of clinical utility; however, as is inevitably the case with other chemotherapeutic agents, resistance development constrains their effectiveness. One putative mechanism of resistance is the promotion of autophagy, which is a direct consequence of the inhibition of the mTOR signaling pathway. Autophagy is primarily considered to be a cytoprotective survival mechanism, whereby cytoplasmic components are recycled to generate energy and metabolic intermediates. The autophagy induced by everolimus and temsirolimus appears to play a largely protective function, whereas a cytotoxic function appears to predominate in the case of rapamycin. In this review we provide an overview of the autophagy induced in response to mTOR inhibitors in different tumor models in an effort to determine whether autophagy targeting could be of clinical utility as adjuvant therapy in association with mTOR inhibition.
    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
    磷脂酰肌醇3-激酶(PI3-K)信号通路是癌症中细胞存活的关键途径,因此代表了新的儿科抗癌药物的有趣靶标。然而,针对这一途径的独特临床毒性(导致高血糖)与化疗相结合的困难,儿童肿瘤中罕见的突变和伴随的突变已导致这些抑制剂在治疗成人和儿童中的临床翻译的主要障碍。PIK3CA中的突变预测成人癌症中对PI3-K抑制剂的反应。儿童和成人发生相同的突变,但是它们在儿科中的频率明显较低。在儿童中,高级别神经胶质瘤,尤其是弥漫性中线胶质瘤(DMG),PIK3CA突变发生率最高。新的突变特异性PI3-K抑制剂降低了目标PI3-Kα野生型活性的毒性。mTOR抑制剂依维莫司被批准用于室管膜下巨细胞星形细胞瘤。在儿科癌症中,mTOR抑制剂主要由学术界评估,没有总体战略,在经验性的,突变无关的临床试验,对单一疗法的反应率非常低。因此,mTOR抑制剂用于儿童癌症的单药或联合治疗的未来试验应得到非常有力的生物学理论基础和临床前数据的支持.糖原合酶激酶-3β抑制剂的进一步临床前评估是必需的。同样,即使有AKT突变(~0.1%),AKT抑制剂在儿科癌症中的作用尚不清楚.患者倡导者强烈敦促分析和保存参与临床试验的每个儿童的数据。首要任务是评估特定的突变,中枢神经系统穿透性PI3-K抑制剂在儿童DMG中的合理生物学组合。组合的选择,应基于基因组景观,例如PTEN丢失和临床前数据支持的抗性机制。然而,鉴于涉及的人群非常罕见,需要创新的监管方法来生成适应症的数据。
    Phosphatidylinositol 3-kinase (PI3-K) signalling pathway is a crucial path in cancer for cell survival and thus represents an intriguing target for new paediatric anti-cancer drugs. However, the unique clinical toxicities of targeting this pathway (resulting in hyperglycaemia) difficulties combining with chemotherapy, rarity of mutations in childhood tumours and concomitant mutations have resulted in major barriers to clinical translation of these inhibitors in treating both adults and children. Mutations in PIK3CA predict response to PI3-K inhibitors in adult cancers. The same mutations occur in children as in adults, but they are significantly less frequent in paediatrics. In children, high-grade gliomas, especially diffuse midline gliomas (DMG), have the highest incidence of PIK3CA mutations. New mutation-specific PI3-K inhibitors reduce toxicity from on-target PI3-Kα wild-type activity. The mTOR inhibitor everolimus is approved for subependymal giant cell astrocytomas. In paediatric cancers, mTOR inhibitors have been predominantly evaluated by academia, without an overall strategy, in empiric, mutation-agnostic clinical trials with very low response rates to monotherapy. Therefore, future trials of single agent or combination strategies of mTOR inhibitors in childhood cancer should be supported by very strong biological rationale and preclinical data. Further preclinical evaluation of glycogen synthase kinase-3 beta inhibitors is required. Similarly, even where there is an AKT mutation (∼0.1 %), the role of AKT inhibitors in paediatric cancers remains unclear. Patient advocates strongly urged analysing and conserving data from every child participating in a clinical trial. A priority is to evaluate mutation-specific, central nervous system-penetrant PI3-K inhibitors in children with DMG in a rational biological combination. The choice of combination, should be based on the genomic landscape e.g. PTEN loss and resistance mechanisms supported by preclinical data. However, in view of the very rare populations involved, innovative regulatory approaches are needed to generate data for an indication.
    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
    PI3K/AKT/mTOR信号通路是乳腺癌中最频繁激活的通路之一,并且在几种生理功能的调节中也起着核心作用。通过开发针对三种激酶的抑制剂(PI3K,AKT,和mTOR)。虽然已经开发了多种化合物,目前,只有三种抑制剂被批准在晚期ER阳性患者中靶向该途径,HER2阴性乳腺癌:依维莫司(mTOR抑制剂),alpelisib(PIK3CA抑制剂),和capivasertib(AKT抑制剂)。像大多数靶向抗癌药物一样,耐药性在临床上是一个主要问题,也是经常限制这些药物整体疗效的一个因素.耐药性可以根据其发展的时间范围分为内在或获得性耐药性。尽管内在抗性在特定治疗之前存在,获得性抵抗是由治疗引起的。大多数ER阳性的患者,HER2阴性晚期乳腺癌可能会在其癌症旅程的某个时候提供PI3K/AKT/mTOR通路的抑制剂。可用的选项取决于批准标准和癌症的突变状态。在这一大群患者中,大多数人可能会在某个时候产生抗药性,这使得这是一个感兴趣的领域,目前尚未满足的需求。在这里,我们回顾了针对PI3K/AKT/mTOR信号通路的药物耐药的常见机制,阐述当前的管理方法,并讨论正在进行的临床试验,试图减轻这一重大问题。我们特别强调需要对AKT1抑制剂耐药性进行更多研究。
    The PI3K/AKT/mTOR signalling pathway is one of the most frequently activated pathways in breast cancer and also plays a central role in the regulation of several physiologic functions. There are major efforts ongoing to exploit precision medicine by developing inhibitors that target the three kinases (PI3K, AKT, and mTOR). Although multiple compounds have been developed, at present, there are just three inhibitors approved to target this pathway in patients with advanced ER-positive, HER2-negative breast cancer: everolimus (mTOR inhibitor), alpelisib (PIK3CA inhibitor), and capivasertib (AKT inhibitor). Like most targeted cancer drugs, resistance poses a major problem in the clinical setting and is a factor that has frequently limited the overall efficacy of these agents. Drug resistance can be categorised into intrinsic or acquired resistance depending on the timeframe it has developed within. Whereas intrinsic resistance exists prior to a specific treatment, acquired resistance is induced by a therapy. The majority of patients with ER-positive, HER2-negative advanced breast cancer will likely be offered an inhibitor of the PI3K/AKT/mTOR pathway at some point in their cancer journey, with the options available depending on the approval criteria in place and the cancer\'s mutation status. Within this large cohort of patients, it is likely that most will develop resistance at some point, which makes this an area of interest and an unmet need at present. Herein, we review the common mechanisms of resistance to agents that target the PI3K/AKT/mTOR signalling pathway, elaborate on current management approaches, and discuss ongoing clinical trials attempting to mitigate this significant issue. We highlight the need for additional studies into AKT1 inhibitor resistance in particular.
    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
    暂无摘要。
    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
    卵巢癌是涵盖许多不同亚型的总称。子宫内膜样和透明细胞卵巢癌是子宫内膜异位症相关的卵巢癌(EAOC),通常由卵巢中的异位子宫内膜引起。雷帕霉素的机制靶标(mTOR)是细胞内稳态的关键调节因子,在子宫内膜异位症和子宫内膜异位症相关卵巢癌中失调,从具有癌症样特征的良性疾病中,可能有利于癌症发生,通过一个非典型的阶段,弗兰克·恶性肿瘤。在这次审查中,我们关注子宫内膜异位症和EAOCs中的mTOR失调,研究癌症驱动基因突变及其与mTOR通路的潜在相互作用。此外,我们探索转化的复杂发病机理,考虑到环境,荷尔蒙,和表观遗传因素。然后我们讨论绝经后子宫内膜异位症的发病机制和恶变倾向。最后,我们总结了目前mTOR靶向治疗子宫内膜异位症和EAOCs的研究进展.
    Ovarian cancer is an umbrella term covering a number of distinct subtypes. Endometrioid and clear-cell ovarian carcinoma are endometriosis-associated ovarian cancers (EAOCs) frequently arising from ectopic endometrium in the ovary. The mechanistic target of rapamycin (mTOR) is a crucial regulator of cellular homeostasis and is dysregulated in both endometriosis and endometriosis-associated ovarian cancer, potentially favouring carcinogenesis across a spectrum from benign disease with cancer-like characteristics, through an atypical phase, to frank malignancy. In this review, we focus on mTOR dysregulation in endometriosis and EAOCs, investigating cancer driver gene mutations and their potential interaction with the mTOR pathway. Additionally, we explore the complex pathogenesis of transformation, considering environmental, hormonal, and epigenetic factors. We then discuss postmenopausal endometriosis pathogenesis and propensity for malignant transformation. Finally, we summarize the current advancements in mTOR-targeted therapeutics for endometriosis and EAOCs.
    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
    哺乳动物雷帕霉素靶蛋白(mTOR)是一个关键的调节因子,整合不同的环境信号来控制基本的细胞功能,如蛋白质合成,细胞生长,生存,和凋亡。嵌入在信号通路的复杂网络中,mTOR失调与一系列人类疾病的发生和进展有关,包括代谢紊乱,如糖尿病和心血管疾病,以及各种癌症。mTOR在衰老中也具有显著的作用。鉴于其广泛的生物学影响,mTOR信号传导是解决这些复杂病症的主要治疗靶标。mTOR抑制剂的开发已经证明在许多研究领域是有利的。这篇综述深入研究了mTOR信号的重要性,突出了导致疾病的这个复杂网络的关键组成部分。此外,它阐述了mTOR抑制剂的最新发现及其临床意义。该综述还强调了开发具有双重功能的更有效的下一代mTOR抑制剂以有效靶向mTOR途径的重要性。对mTOR信号传导的全面了解将能够开发用于管理与mTOR失调相关的疾病的有效治疗策略。
    The mammalian target of rapamycin (mTOR) is a pivotal regulator, integrating diverse environmental signals to control fundamental cellular functions, such as protein synthesis, cell growth, survival, and apoptosis. Embedded in a complex network of signaling pathways, mTOR dysregulation is implicated in the onset and progression of a range of human diseases, including metabolic disorders such as diabetes and cardiovascular diseases, as well as various cancers. mTOR also has a notable role in aging. Given its extensive biological impact, mTOR signaling is a prime therapeutic target for addressing these complex conditions. The development of mTOR inhibitors has proven advantageous in numerous research domains. This review delves into the significance of mTOR signaling, highlighting the critical components of this intricate network that contribute to disease. Additionally, it addresses the latest findings on mTOR inhibitors and their clinical implications. The review also emphasizes the importance of developing more effective next-generation mTOR inhibitors with dual functions to efficiently target the mTOR pathways. A comprehensive understanding of mTOR signaling will enable the development of effective therapeutic strategies for managing diseases associated with mTOR dysregulation.
    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
    多发性骨髓瘤(MM),最终分化的浆细胞癌,是第二大最常见的血液恶性肿瘤,并且由于耐药性的不可避免的发展而无法治愈。强烈的蛋白质合成是MM细胞的独特特征,支持克隆免疫球蛋白或游离轻链的大规模生产。哺乳动物雷帕霉素靶蛋白(mTOR)激酶被认为是重要细胞过程的主要调节因子。包括调节代谢和蛋白质合成,可以在两个多蛋白复合物中找到,mTORC1和mTORC2。这些复合物的失调与几种类型的癌症有关,包括MM。由于mTOR已被证明在大部分MM患者中异常激活,并在刺激MM细胞存活和对几种现有疗法的抵抗中发挥作用,了解mTOR复合物的调节和功能对于开发更有效的治疗策略至关重要.这篇综述提供了mTOR通路的一般概述,讨论与mTOR复合物的结构和调控相关的关键发现和最新见解。此外,我们重点介绍了mTOR参与蛋白质合成的机制方面的发现,并深入研究了mTOR介导的在MM中发生的过程.最后,我们总结了针对MM中mTOR复合物的药物的进展和当前挑战。
    Multiple Myeloma (MM), a cancer of terminally differentiated plasma cells, is the second most prevalent hematological malignancy and is incurable due to the inevitable development of drug resistance. Intense protein synthesis is a distinctive trait of MM cells, supporting the massive production of clonal immunoglobulins or free light chains. The mammalian target of rapamycin (mTOR) kinase is appreciated as a master regulator of vital cellular processes, including regulation of metabolism and protein synthesis, and can be found in two multiprotein complexes, mTORC1 and mTORC2. Dysregulation of these complexes is implicated in several types of cancer, including MM. Since mTOR has been shown to be aberrantly activated in a large portion of MM patients and to play a role in stimulating MM cell survival and resistance to several existing therapies, understanding the regulation and functions of the mTOR complexes is vital for the development of more effective therapeutic strategies. This review provides a general overview of the mTOR pathway, discussing key discoveries and recent insights related to the structure and regulation of mTOR complexes. Additionally, we highlight findings on the mechanisms by which mTOR is involved in protein synthesis and delve into mTOR-mediated processes occurring in MM. Finally, we summarize the progress and current challenges of drugs targeting mTOR complexes in MM.
    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
    mTORC1在肾细胞癌(RCC)中异常激活,并被rapalogs靶向。至于其他靶向治疗,rapalogs的临床应用受到耐药性发展的限制。抗性通常由目标突变引起,但mTOR突变在RCC中很少发现。和人类一样,延长RCC肿瘤移植物(TG)的rapalog治疗导致耐药性。出乎意料的是,抗性肿瘤的外植体在小鼠的培养和随后的移植中均变得敏感。值得注意的是,尽管肿瘤细胞中存在持续的mTORC1抑制,但仍出现耐药性。相比之下,mTORC1在肿瘤微环境(TME)中重新激活。为了测试TME的作用,我们设计了具有抗性mTOR突变(S2035T)的免疫受损受体小鼠。有趣的是,在mTORS2035T小鼠中,TG对rapalogs产生抗性。尽管肿瘤细胞中存在mTORC1抑制,但仍存在耐药性,并且可以通过将肿瘤细胞与突变成纤维细胞共培养来诱导。因此,TME中mTORC1的强制激活足以赋予rapalogs抗性。这些研究强调了非肿瘤细胞中mTORC1抑制对rapalog抗肿瘤活性的重要性,并为RCC患者缺乏mTOR抗性突变提供了解释。
    mTORC1 is aberrantly activated in renal cell carcinoma (RCC) and is targeted by rapalogs. As for other targeted therapies, rapalogs clinical utility is limited by the development of resistance. Resistance often results from target mutation, but mTOR mutations are rarely found in RCC. As in humans, prolonged rapalog treatment of RCC tumorgrafts (TGs) led to resistance. Unexpectedly, explants from resistant tumors became sensitive both in culture and in subsequent transplants in mice. Notably, resistance developed despite persistent mTORC1 inhibition in tumor cells. In contrast, mTORC1 became reactivated in the tumor microenvironment (TME). To test the role of the TME, we engineered immunocompromised recipient mice with a resistance mTOR mutation (S2035T). Interestingly, TGs became resistant to rapalogs in mTORS2035T mice. Resistance occurred despite mTORC1 inhibition in tumor cells and could be induced by coculturing tumor cells with mutant fibroblasts. Thus, enforced mTORC1 activation in the TME is sufficient to confer resistance to rapalogs. These studies highlight the importance of mTORC1 inhibition in nontumor cells for rapalog antitumor activity and provide an explanation for the lack of mTOR resistance mutations in RCC patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号