temsirolimus

坦西罗莫司
  • 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
    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)

  • 文章类型: Journal Article
    背景:确定有效的免疫抑制策略对于解决器官移植后的免疫排斥反应至关重要。本研究探讨了替西罗莫司潜在的免疫抑制作用和机制。雷帕霉素衍生物,在器官移植中。
    方法:使用宫颈套管技术与BALB/c供体和C57BL/6受体建立小鼠心脏同种异体移植模型。小鼠胃内给药替西罗莫司并评价移植物存活。组织学染色用于评估病理变化。BrdU测定用于测量脾T细胞增殖。流式细胞术用于定量调节性T细胞(Tregs),CD4+T细胞,和CD8+T细胞。ELISA和qPCR检测Foxp3、IL-4、IFN-γ,和TGF-β表达。
    结果:替西罗莫司在20mg/kg/天时显示出有效的免疫抑制作用,显著抑制T细胞增殖(84.6%,P<0.0001)和延长移植物存活(中位数49天vs.控制8.5天,P<0.0001)。然而,停药后中位生存期降至34.5天.坦西罗莫司也减少脾CD4+和CD8+T细胞(2.85%和2.92%,P<0.001)和抗体水平(IgM,IgG1,IgG2)增加11.85-29.09%(P<0.0001)和增加的Tregs,Foxp3、IL-4(P<0.01)、和TGF-β(P<0.05),同时降低IFN-γ(P<0.001)。
    结论:替西罗莫司表现出有效的免疫抑制作用,成为缓解器官移植排斥反应的有力候选者。
    Identifying effective immunosuppressive strategies is critical for addressing immunological rejection following organ transplantation. This study explores the potential immunosuppressive effects and mechanisms of temsirolimus, a rapamycin derivative, in organ transplantation.
    A mouse cardiac allograft model was established using a cervical cannula technique with BALB/c donors and C57BL/6 recipients. Mice were administered temsirolimus intragastrically and graft survival was evaluated. Histological staining was used to assess pathological changes. The BrdU assay was used to measure splenic T cell proliferation. Flow cytometry was used to quantify regulatory T cells (Tregs), CD4+ T cells, and CD8+ T cells. ELISA and qPCR assays were used to determine Foxp3, IL-4, IFN-γ, and TGF-β expression.
    Temsirolimus displayed potent immunosuppressive effects at 20 mg/kg/day, significantly inhibiting T cell proliferation (84.6%, P < 0.0001) and prolonging graft survival (median 49 days vs. 8.5 days in controls, P < 0.0001). However, median survival decreased to 34.5 days upon withdrawal. Temsirolimus also reduced splenic CD4+ and CD8+ T cells (2.85% and 2.92%, P < 0.001) and antibody levels (IgM, IgG1, IgG2) by 11.85-29.09% (P < 0.0001) and increased Tregs, Foxp3, IL-4 (P < 0.01), and TGF-β (P < 0.05), while decreasing IFN-γ (P < 0.001).
    Temsirolimus exhibited potent immunosuppressive effects, emerging as a strong candidate to mitigate organ transplant rejection.
    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
    帕司替尼是一种口服药物,可以抑制多种激酶,包括JAK2,FLT3,IRAK和STAT3。它最近已被批准用于治疗血小板减少症和骨髓纤维化患者。目前,研究正在探索pacritinib在治疗其他类型癌症如白血病中的潜在用途。乳腺癌和前列腺癌。我们的研究旨在探讨单独使用帕克替尼及其联合标准治疗对肾细胞癌(RCC)的影响。我们表明,帕克替尼剂量依赖性地降低了RCC细胞的活力,在纳摩尔或低微摩尔浓度范围内具有IC50。帕瑞替尼抑制细胞增殖,减少集落形成,和增加细胞凋亡。有趣的是,帕克替尼与替西罗莫司和舒尼替尼合用时表现出协同作用,但是当与阿霉素联合使用时具有拮抗作用,在一组RCC细胞系中。我们还证实了pacritinib与替西罗莫司和舒尼替尼的组合在RCC小鼠模型中产生了协同作用。在整个治疗期间完全抑制肿瘤生长。机制研究表明,JAK2的抑制,而不是IRAK,是pacritinib抗RCC活性的主要因素。我们的研究首次证明了pacritinib有望作为RCC的治疗选择,并强调了靶向JAK2/STAT信号通路在RCC中的治疗潜力。
    Pacritinib is an oral medication that inhibits several kinases including JAK2, FLT3, IRAK and STAT3. It has been recently approved to treat patients with thrombocytopenia and myelofibrosis. Studies are currently exploring the potential use of pacritinib in treating other types of cancer such as leukaemia, breast cancer and prostate cancer. Our study aimed to investigate the effects of pacritinib alone and its combination with standard of care in renal cell carcinoma (RCC). We showed that pacritinib dose-dependently decreased viability of RCC cells, with IC50 at nanomolar or low micromolar concentration rage. Pacritinib inhibited cell proliferation, decreased colony formation, and increased apoptosis. Interestingly, pacritinib exhibited synergistic effects when combined with temsirolimus and sunitinib, but antagonistic effects when combined with doxorubicin, in a panel of RCC cell lines. We also confirmed that the combination of pacritinib with temsirolimus and sunitinib resulted in synergistic effects in RCC mouse models, with complete inhibition of tumour growth throughout the treatment period. Mechanistic studies indicated that the inhibition of JAK2, but not IRAK, was the main contributor to the anti-RCC activity of pacritinib. Our study is the first to demonstrate that pacritinib shows promise as a treatment option for RCC and underscores the therapeutic potential of targeting the JAK2/STAT signalling pathway in RCC.
    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
    替西罗莫司是第一代mTOR抑制剂,通常用于临床治疗与肺损伤相关的癌症。然而,这种不利影响的潜在机制仍然难以捉摸。内皮屏障功能障碍在中性粒细胞浸润到肺泡中起关键作用,最终导致肺损伤。本研究表明,替西罗莫司诱导内皮细胞粘附分子的异常表达,导致中性粒细胞浸润增强和随后的肺损伤。小鼠模型的结果表明,替西罗莫司破坏了毛细血管-肺泡屏障功能,并促进了嗜中性粒细胞跨肺泡腔内内皮的迁移。与我们的体内观察一致,替西罗莫司损害了人肺内皮细胞单层内的细胞间屏障功能,导致中性粒细胞浸润增加。此外,在体外和体内实验中,我们证明了替西罗莫司诱导的中性粒细胞经内皮迁移是由血小板内皮细胞粘附分子-1(PECAM-1)介导的.总的来说,这些发现强调了替西罗莫司在体外和体内通过PECAM-1依赖性途径诱导内皮屏障功能障碍,最终导致中性粒细胞浸润和随后的肺损伤。
    Temsirolimus is a first-generation mTOR inhibitor commonly used in the clinical treatment of cancers that is associated with lung injury. However, the mechanism underlying this adverse effect remains elusive. Endothelial barrier dysfunction plays a pivotal role in the infiltration of neutrophils into the pulmonary alveoli, which eventually induces lung injury. The present study demonstrates that temsirolimus induces the aberrant expression of adhesion molecules in endothelial cells, leading to enhanced neutrophil infiltration and subsequent lung injury. Results of a mouse model revealed that temsirolimus disrupted capillary-alveolar barrier function and facilitated neutrophil transmigration across the endothelium within the alveolar space. Consistent with our in vivo observations, temsirolimus impaired intercellular barrier function within monolayers of human lung endothelial cells, resulting in increased neutrophil infiltration. Furthermore, we demonstrated that temsirolimus-induced neutrophil transendothelial migration was mediated by platelet endothelial cell adhesion molecule-1 (PECAM-1) in both in vitro and in vivo experiments. Collectively, these findings highlight that temsirolimus induces endothelial barrier dysfunction via PECAM-1-dependent pathway both in vitro and in vivo, ultimately leading to neutrophil infiltration and subsequent pulmonary injury.
    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
    非霍奇金淋巴瘤(NHL)的发病率随着时间的推移而上升,B细胞淋巴瘤占大多数淋巴瘤。磷酸肌醇3-激酶(PI3K)/v-akt鼠胸腺瘤病毒癌基因同源物1(Akt)/哺乳动物雷帕霉素靶(mTOR)信号通路在多种细胞过程中发挥关键作用,如细胞增殖和存活。在许多不同类型的B细胞淋巴瘤中证实了其在淋巴发生中的作用。本文主要对PI3K/v-akt/mTOR通路在B细胞NHL中的致癌机制进行综述,重点介绍常见的B细胞淋巴瘤类型[弥漫性大B细胞淋巴瘤(DLBCL)和套细胞淋巴瘤(MCL)]。此外,它总结了有关mTOR抑制剂替西罗莫司和依维莫司在B细胞NHL中的临床应用的文献,已经在一系列B细胞恶性肿瘤患者的临床试验中进行了测试,作为单一疗法或与其他药物或方案组合。
    Non-Hodgkin lymphoma\'s (NHL) incidence is rising over time, and B cell lymphomas comprise the majority of lymphomas. The phosphoinositide 3-kinase (PI3K)/v-akt murine thymoma viral oncogene homologue 1 (Akt)/mammalian target of the rapamycin (mTOR) signaling pathway plays a critical role in a variety of cellular processes, such as cell proliferation and survival. Its role in lymphomagenesis is confirmed in many different types of B cell lymphomas. This review is mainly focused on the PI3K/v-akt/mTOR pathway-related oncogenic mechanisms in B cell NHLs with an emphasis on common B cell lymphoma types [diffuse large B cell lymphoma (DLBCL) and mantle cell lymphoma (MCL)]. Furthermore, it summarizes the literature regarding the clinical applications of the mTOR inhibitors temsirolimus and everolimus in B cell NHLs, which have been tested in a range of clinical trials enrolling patients with B cell malignancies, either as monotherapy or in combination with other agents or regimens.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Clinical Trial, Phase II
    我们评估了贝伐单抗联合替西罗莫司对晚期胰腺外神经内分泌肿瘤患者的疗效和安全性。这个NCI赞助的多中心,开放标签,II期研究(NCT01010126)纳入晚期患者,经常性,或转移性胰腺外神经内分泌肿瘤。所有患者均接受替西罗莫司和贝伐单抗治疗,直至疾病进展或出现不可接受的毒性。替西罗莫司25mg在第1、8、15和22天静脉内施用,贝伐单抗10mg/kg在4周周期的第1和15天静脉内施用。停药替西罗莫司或贝伐单抗不需要停药其他药物。主要终点是客观缓解率和6个月无进展生存率。59名患者参加了这项研究,对54例患者的疗效和不良事件进行了评价.中位无进展生存期为7.1个月,坦西罗莫司治疗的中位持续时间为3.9个月,贝伐单抗治疗的中位持续时间为3.5个月.联合治疗的客观有效率为2%,6个月无进展生存率为48%.最常报告的3-4级不良事件包括疲劳(13%),高血压(13%),出血(13%)。接近54%的患者因不良事件而停止治疗,拒绝进一步治疗,或治疗延误。研究中发生了三例死亡,其中两个是由于治疗相关的肠穿孔。鉴于贝伐单抗和替西罗莫司的联合用药的最小疗效和增加的毒性,我们不建议在晚期胰腺外神经内分泌肿瘤患者中使用该方案.
    We assessed the efficacy and safety of combining bevacizumab with temsirolimus in patients with advanced extra-pancreatic neuroendocrine tumors. This NCI-sponsored multicenter, open-label, phase II study (NCT01010126) enrolled patients with advanced, recurrent, or metastatic extra-pancreatic neuroendocrine tumors. All patients were treated with temsirolimus and bevacizumab until disease progression or unacceptable toxicity. Temsirolimus 25 mg was administered i.v. on days 1, 8, 15, and 22 and bevacizumab 10 mg/kg i.v. on days 1 and 15 of a 4-week cycle. Discontinuation of temsirolimus or bevacizumab did not require discontinuation of the other agent. The primary endpoints were objective response rate and 6-month progression-free survival rate. Fifty-nine patients were enrolled in this study, and 54 were evaluated for efficacy and adverse events. While median progression-free survival was 7.1 months, the median duration of treatment with temsirolimus was 3.9 months and that with bevacizumab was 3.5 months. The objective response rate of combination therapy was 2%, and 6-month progression-free survival was 48%. The most frequently reported grade 3-4 adverse events included fatigue (13%), hypertension (13%), and bleeding (13%). Close to 54% of the patients discontinued treatment due to adverse events, refusal of further treatment, or treatment delays. Three deaths occurred in the study, of which two were due to treatment-related bowel perforations. Given the minimal efficacy and increased toxicity seen with the combination of bevacizumab and temsirolimus, we do not recommend the use of this regimen in patients with advanced extra-pancreatic neuroendocrine tumors.
    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:纤维板层肝细胞癌(FLHCC)是一种罕见的肿瘤,表现在没有慢性肝病的年轻患者中。高达80-100%发展为复发性疾病,需要额外的手术或全身治疗。缺乏系统选择和临床前治疗研究。我们先前描述了患者来源的异种移植物(PDX)的发展,允许临床前研究。在这里,我们开发了FLHCCPDX模型,并利用这些模型来定义肿瘤特征和确定全身药物的疗效.
    方法:两名患者在FLHCC切除时获得原发灶和淋巴结转移瘤组织。筛选肿瘤裂解物的蛋白质上调。从转移性和原发性肿瘤组织产生细胞系。用替西罗莫司处理后评估细胞系的活力,吉西他滨/奥沙利铂,跟随FIRINOX。从转移组织开发两个PDX模型。对于体内研究,用替西罗莫司治疗荷瘤小鼠,FOLFIRINOX,和吉西他滨/奥沙利铂。
    结果:从转移性FLHCC成功生成PDX模型,紧密地概括了原始肿瘤。与原发性肿瘤相比,转移组织中mTOR上调。来自转移组织的细胞系显示出对替西罗莫司的显著敏感性。PDX模型的体内测试表明,对单剂替西罗莫司的显着反应具有最小的毒性。
    结论:此处,我们证明了开发紧密概括FLHCC的PDX模型的可行性。与原发性组织相比,转移组织中mTOR上调。使用替西罗莫司治疗的mTOR抑制的功效表明mTOR途径的上调可能是转移性病变中生长的重要机制和治疗的潜在靶标。
    OBJECTIVE: Fibrolamellar hepatocellular carcinoma (FLHCC) is a rare tumor presenting in younger patients without chronic liver disease. Up to 80-100% develop recurrent disease, necessitating additional surgery or systemic treatment. Systemic options and pre-clinical treatment studies are lacking. We previously described patient-derived xenograft (PDX) development, allowing for pre-clinical studies. Herein, we develop FLHCC PDX models and utilize these to define tumor characteristics and determine the efficacy of systemic agents.
    METHODS: Primary and lymph node metastatic tumor tissues were obtained at the time of FLHCC resection in two patients. Tumor lysates were screened for protein upregulation. Cell lines were generated from metastatic and primary tumor tissue. The viability of the cell lines was assessed after treatment with temsirolimus, gemcitabine/oxaliplatin, and FOLFIRINOX. Two PDX models were developed from metastatic tissue. For in vivo studies, tumor-bearing mice were treated with temsirolimus, FOLFIRINOX, and Gemcitabine/oxaliplatin.
    RESULTS: PDX models were successfully generated from metastatic FLHCC, which closely recapitulated the original tumor. Upregulation of mTOR was seen in metastatic tissue compared to primary tumors. Cell lines from metastatic tissue demonstrated significant sensitivity to temsirolimus. In vivo testing of PDX models demonstrated a significant response to single-agent temsirolimus with minimal toxicity.
    CONCLUSIONS: Herein, we demonstrate the feasibility of developing PDX models that closely recapitulate FLHCC. Upregulation of mTOR was seen in metastatic tissue compared to primary tissue. The efficacy of mTOR inhibition with temsirolimus treatment suggests that the upregulation of the mTOR pathway may be a significant mechanism for growth in metastatic lesions and a potential target for therapeutics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Clinical Trial, Phase I
    背景:临床前模型表明抗血管生成疗法之间的协同作用,哺乳动物雷帕霉素靶蛋白(mTOR),和组蛋白去乙酰化酶抑制剂促进抗癌活性。
    方法:该一期研究纳入了2012年4月至2018年之间的47名患者,并确定了安全性,最大耐受剂量(MTD),和剂量限制性毒性(DLT),当结合贝伐单抗,替西罗莫司,晚期癌症患者的丙戊酸。
    结果:入选患者的中位年龄为56岁。对患者进行严重预处理,中位数为4行先前治疗。45名患者(95.7%)经历了一个或多个治疗相关的不良事件(TRAEs)。3级TRAEs为淋巴细胞减少症(14.9%),血小板减少症(8.5%),和粘膜炎(6.4%)。4级TRAE包括淋巴细胞减少(2.1%)和中枢神经系统脑血管缺血(2.1%)。六名患者在3级感染的10个剂量水平上发展了DLT,皮疹,粘膜炎,肠穿孔,脂肪酶升高,和4级脑血管缺血。MTD为剂量水平9(贝伐单抗5mg/kg第1天和第15天静脉内(IV)加替西罗莫司25mg第1、8、15和22天,每次口服(PO)和丙戊酸5mg/kg第1-7天和第15-21天)。3例患者的客观缓解率(ORR)为7.9%,确认部分缓解(PR)(腮腺各1例,卵巢,和阴道癌)。5例(13.1%)患者病情稳定(SD)≥+6个月。临床获益状态(CBR:PR+SD≥+6个月)为21%。
    结论:贝伐单抗联合治疗,替西罗莫司,丙戊酸是可行的,但是有很多毒性,这将需要对未来的临床发展进行仔细的管理(ClinicalTrials.gov标识符:NCT01552434)。
    BACKGROUND: Preclinical models suggest synergy between anti-angiogenesis therapy, mammalian target of rapamycin (mTOR), and histone deacetylase inhibitors to promote anticancer activity.
    METHODS: This phase I study enrolled 47 patients between April 2012 and 2018 and determined safety, maximum tolerated dose (MTD), and dose-limiting toxicities (DLTs) when combining bevacizumab, temsirolimus, and valproic acid in patients with advanced cancer.
    RESULTS: Median age of enrolled patients was 56 years. Patients were heavily pretreated with a median of 4 lines of prior therapy. Forty-five patients (95.7%) experienced one or more treatment-related adverse events (TRAEs). Grade 3 TRAEs were lymphopenia (14.9%), thrombocytopenia (8.5%), and mucositis (6.4%). Grade 4 TRAEs included lymphopenia (2.1%) and CNS cerebrovascular ischemia (2.1%). Six patients developed DLTs across 10 dose levels with grade 3 infection, rash, mucositis, bowel perforation, elevated lipase, and grade 4 cerebrovascular ischemia. The MTD was dose level 9 (bevacizumab 5 mg/kg days 1 and 15 intravenously (IV) plus temsirolimus 25 mg days 1, 8, 15, and 22 IV and valproic acid 5 mg/kg on days 1-7 and 15-21 per orally (PO)). Objective response rate (ORR) was 7.9% with confirmed partial response (PRs) in 3 patients (one each in parotid gland, ovarian, and vaginal cancers). Stable disease (SD) ≥+6 months was seen in 5 patients (13.1%). Clinical benefit state (CBR: PR + SD ≥+6 months) was 21%.
    CONCLUSIONS: Combination therapy with bevacizumab, temsirolimus, and valproic acid was feasible, but there were numerous toxicities, which will require careful management for future clinical development (ClinicalTrials.gov Identifier: NCT01552434).
    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
    PI3K/AKT/mTOR途径的组成型激活在套细胞淋巴瘤(MCL)的发病机制中起重要作用,导致mTOR抑制剂替西罗莫司批准用于复发或难治性MCL。然而,尽管初始反应率良好,已观察到治疗中的早期复发。因此,了解替西罗莫司耐药的潜在机制并制定克服它的策略是非常必要的。这里,我们建立了一个新的temsirolimus耐药MCL细胞系来评估耐药的分子背景。比较替西罗莫司敏感和抗性细胞系的转录组谱分析和基因集富集分析显示PI3K/AKT/mTor-的显着上调,RAS信号-和依赖于RTK的PDGFR-,FGFR-,抗性细胞中的MET-和ALK-信号通路。此外,MET,被称为重要的原癌基因和耐药介质,是抗性细胞中最上调的基因之一。重要的是,Met蛋白在两者中均过表达,MCL细胞具有获得性和内在的替西罗莫司抗性,但在任何替西罗莫司敏感的药物中都检测不到。mTOR和Met信号传导与替西罗莫司和RTK抑制剂克唑替尼的联合药理学抑制显著恢复了对替西罗莫司的敏感性。此外,这种联合治疗被证明在所研究的所有MCL细胞系中具有协同作用,并且在原代MCL细胞中也具有活性.总之,我们首次表明,MET的过表达在介导MCL中替西罗莫司耐药中起重要作用,替西罗莫司和克唑替尼联合治疗是一种非常有前景的MCL治疗方法,也是克服替西罗莫司耐药的有效策略.
    Constitutive activation of the PI3K/AKT/mTOR-pathway plays an important role in the pathogenesis of mantle cell lymphoma (MCL), leading to approval of the mTOR inhibitor temsirolimus for relapsed or refractory MCL. Yet, despite favorable initial response rates, early relapses under treatment have been observed. Therefore, understanding the underlying mechanisms of temsirolimus resistance and developing strategies to overcome it is highly warranted. Here, we established a new temsirolimus-resistant MCL cell line to evaluate the molecular background of resistance to this drug. Transcriptome profiling and gene set enrichment analysis comparing temsirolimus-sensitive and -resistant cell lines showed significant upregulation of PI3K/AKT/mTor-, RAS signaling- and the RTK-dependent PDGFR-, FGFR-, Met- and ALK-signaling-pathways in the resistant cells. Furthermore, MET, known as important proto-oncogene and mediator of drug resistance, was among the most upregulated genes in the resistant cells. Importantly, Met protein was overexpressed in both, MCL cells with acquired as well as intrinsic temsirolimus resistance, but could not be detected in any of the temsirolimus sensitive ones. Combined pharmacological inhibition of mTOR and Met signaling with temsirolimus and the RTK inhibitor crizotinib significantly restored sensitivity to temsirolimus. Furthermore, this combined treatment proved to be synergistic in all MCL cell lines investigated and was also active in primary MCL cells. In summary, we showed for the first time that overexpression of MET plays an important role for mediating temsirolimus resistance in MCL and combined treatment with temsirolimus and crizotinib is a very promising therapeutic approach for MCL and an effective strategy to overcome temsirolimus resistance.
    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