MEK Inhibition

  • 文章类型: Journal Article
    背景:小儿胃肠胰腺神经内分泌肿瘤极为罕见,导致大多数儿科治疗建议基于来自成人的数据。曲美替尼是一种靶向MEK1/2的激酶抑制剂,已用于治疗Ras途径中存在突变的癌症。
    方法:我们利用已建立的具有已知NRAS突变的人类小儿胃肠胰腺神经内分泌样肿瘤患者异种移植物(PDX)来研究MEK抑制作用。我们评估了曲美替尼对增殖的影响,运动性,和体内肿瘤生长。我们建立了PDX的腹膜内转移模型,表征了转移性PDX的表型和基因型,研究了MEK抑制作用。
    结果:我们发现在曲美替尼治疗下,ERK1/2磷酸化降低的靶参与。曲美替尼导致体外细胞生长和运动减少,在小鼠侧腹肿瘤模型中,肿瘤生长减少,动物存活率增加。最后,我们证明曲美替尼能够显著减少胃肠胰腺神经内分泌腹膜内肿瘤转移.
    结论:这些研究的结果支持MEK抑制在小儿NRAS突变实体瘤中的进一步研究。
    BACKGROUND: Pediatric gastroenteropancreatic neuroendocrine tumors are exceedingly rare, resulting in most pediatric treatment recommendations being based on data derived from adults. Trametinib is a kinase inhibitor that targets MEK1/2 and has been employed in the treatment of cancers harboring mutations in the Ras pathway.
    METHODS: We utilized an established human pediatric gastroenteropancreatic neuroendocrine-like tumor patient-derived xenograft (PDX) with a known NRAS mutation to study the effects of MEK inhibition. We evaluated the effects of trametinib on proliferation, motility, and tumor growth in vivo. We created an intraperitoneal metastatic model of this PDX, characterized both the phenotype and the genotype of the metastatic PDX and again, investigated the effects of MEK inhibition.
    RESULTS: We found target engagement with decreased ERK1/2 phosphorylation with trametinib treatment. Trametinib led to decreased in vitro cell growth and motility, and decreased tumor growth and increased animal survival in a murine flank tumor model. Finally, we demonstrated that trametinib was able to significantly decrease gastroenteropancreatic neuroendocrine intraperitoneal tumor metastasis.
    CONCLUSIONS: The results of these studies support the further investigation of MEK inhibition in pediatric NRAS mutated solid 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:本报告重点介绍了BRAF突变肿瘤治疗的一些最新治疗进展,讨论了BRAF靶向药物观察到的最常见的不良事件,并提出了管理和减轻治疗相关毒性的策略。
    结果:BRAF和MEK抑制剂代表了治疗BRAF突变的恶性肿瘤的重大进展,不同肿瘤类型的数据证明了双重MAPK抑制的抗肿瘤功效。尽管这些药物具有合理的毒性,可以在整个器官系统中产生不同的副作用。激活BRAF突变的发现以及随后BRAF和MEK抑制剂的开发已经改变了BRAF突变恶性肿瘤的治疗算法。随着这些有针对性的方案应用的增加,识别和及时管理其独特的不良事件至关重要.
    OBJECTIVE: This report highlights several of the recent therapeutic advancements in the treatment of BRAF-mutant tumors, discusses the most common adverse events observed with BRAF-targeted agents, and suggests strategies to manage and mitigate treatment-related toxicities.
    RESULTS: BRAF and MEK inhibitors represent a significant advancement in the treatment of BRAF-mutated malignancies with data across tumor types demonstrating the anti-tumor efficacy of dual MAPK inhibition. Although these agents have a reasonable toxicity profile, variable side effects across organ systems can develop. The discovery of activating BRAF mutations and subsequent development of BRAF and MEK inhibitors has transformed the treatment algorithms of BRAF-mutant malignancies. With increased application of these targeted regimens, identification and prompt management of their unique adverse events are crucial.
    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
    目的:1型神经纤维瘤病(NF-1)是由编码神经纤维蛋白的NF1肿瘤抑制基因突变引起的癌症易感性综合征,它充当Ras信号的负调节剂。我们回顾过去,电流,以及与NF-1相关的肿瘤的治疗策略的未来状态。
    结果:NF-1相关肿瘤的治疗努力集中在抑制Ras输出,导致下游MEK抑制丛状神经纤维瘤和低度胶质瘤的临床成功。然而,MEK抑制和阻断Ras信号的类似分子单一疗法方法并不适用于所有患者,并且对更具侵袭性的癌症(如恶性外周神经鞘瘤和高级别神经胶质瘤)显示有限的疗效。激发新的治疗方法。我们强调目前NF-1相关肿瘤的治疗前景,将治疗广泛分类为过去的Ras途径阻断策略,目前的方法针对平行的致癌和肿瘤抑制途径,以及利用免疫疗法中的生物和技术创新的未来研究途径,药理学,和基因传递。
    OBJECTIVE: Neurofibromatosis type 1 (NF-1) is a cancer predisposition syndrome caused by mutations in the NF1 tumor suppressor gene that encodes the neurofibromin protein, which functions as a negative regulator of Ras signaling. We review the past, current, and future state of therapeutic strategies for tumors associated with NF-1.
    RESULTS: Therapeutic efforts for NF-1-associated tumors have centered around inhibiting Ras output, leading to the clinical success of downstream MEK inhibition for plexiform neurofibromas and low-grade gliomas. However, MEK inhibition and similar molecular monotherapy approaches that block Ras signaling do not work for all patients and show limited efficacy for more aggressive cancers such as malignant peripheral nerve sheath tumors and high-grade gliomas, motivating novel treatment approaches. We highlight the current therapeutic landscape for NF-1-associated tumors, broadly categorizing treatment into past strategies for serial Ras pathway blockade, current approaches targeting parallel oncogenic and tumor suppressor pathways, and future avenues of investigation leveraging biologic and technical innovations in immunotherapy, pharmacology, and gene delivery.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Review
    患有野生型BRCA的癌症,同源重组熟练,或对PARP抑制的从头或获得性耐药代表越来越多的患者可能受益于PARP抑制剂组合策略.我们回顾了血管生成的靶向抑制剂,表观遗传调节因子,PI3K,MAPK,和其他细胞信号通路作为同源重组缺陷的诱导剂,为在以前认为对PARP抑制不敏感的情况下使用PARP抑制剂提供支持。
    Cancers with wild-type BRCA, homologous recombination proficiency, or de novo or acquired resistance to PARP inhibition represent a growing population of patients who may benefit from combinatorial PARP inhibitor strategies. We review targeted inhibitors of angiogenesis, epigenetic regulators, and PI3K, MAPK, and other cellular signaling pathways as inducers of homologous recombination deficiency, providing support for the use of PARP inhibitors in contexts not previously considered susceptible to PARP 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
    目的:胰腺导管腺癌(PDAC)的高度异质性的细胞和分子组成不仅促进了异常侵袭性的肿瘤生物学,而且与目前的一刀切的对抗PDAC的治疗策略的概念相矛盾。因此,我们旨在利用以SMAD4缺乏和活化T细胞核因子高表达(SMAD4-/-/NFATc1High)为特征的临床相关分子PDAC亚组的肿瘤生物学意义和治疗脆弱性.
    方法:分析转录组和临床数据,以确定SMAD4-/-/NFATc1高癌症的预后相关性。通过免疫沉淀研究了体外和体内致癌转录因子复合物的形成,邻近连接测定,并验证了交叉模型和物种。SMAD4状态对治疗靶向经典KRAS信号的影响通过全基因组基因表达分析和遗传扰动实验进行了机械破译和证实。分别。在患者来源的类器官和细胞以及转基因和原位PDAC模型中进行了新的定制治疗选择的验证。
    结果:我们的发现确定了侵袭性和化疗耐药SMAD4-/-/NFATc1High亚组的肿瘤生物学特性。机械上,我们确定SMAD4缺陷是致癌NFATc1/SMAD3/cJUN转录因子复合物形成的分子先决条件,驱动RRM1/2的表达式。RRM1/2补充与代谢的吉西他滨直接竞争DNA链掺入的核苷库。通过丝裂原激活的蛋白激酶信号传导抑制(MEKi)分解NFATc1/SMAD3/cJUN复合物可使RRM1/2表达正常化,并与体内吉西他滨治疗协同作用以降低增殖指数。
    结论:我们的结果表明,以SMAD4缺乏和致癌NFATc1/SMAD3/cJUN复合物形成为特征的PDAC暴露了对MEKi/吉西他滨联合治疗的敏感性。
    The highly heterogeneous cellular and molecular makeup of pancreatic ductal adenocarcinoma (PDAC) not only fosters exceptionally aggressive tumor biology, but contradicts the current concept of one-size-fits-all therapeutic strategies to combat PDAC. Therefore, we aimed to exploit the tumor biological implication and therapeutic vulnerabilities of a clinically relevant molecular PDAC subgroup characterized by SMAD4 deficiency and high expression of the nuclear factor of activated T cells (SMAD4-/-/NFATc1High).
    Transcriptomic and clinical data were analyzed to determine the prognostic relevance of SMAD4-/-/NFATc1High cancers. In vitro and in vivo oncogenic transcription factor complex formation was studied by immunoprecipitation, proximity ligation assays, and validated cross model and species. The impact of SMAD4 status on therapeutically targeting canonical KRAS signaling was mechanistically deciphered and corroborated by genome-wide gene expression analysis and genetic perturbation experiments, respectively. Validation of a novel tailored therapeutic option was conducted in patient-derived organoids and cells and transgenic as well as orthotopic PDAC models.
    Our findings determined the tumor biology of an aggressive and chemotherapy-resistant SMAD4-/-/NFATc1High subgroup. Mechanistically, we identify SMAD4 deficiency as a molecular prerequisite for the formation of an oncogenic NFATc1/SMAD3/cJUN transcription factor complex, which drives the expression of RRM1/2. RRM1/2 replenishes nucleoside pools that directly compete with metabolized gemcitabine for DNA strand incorporation. Disassembly of the NFATc1/SMAD3/cJUN complex by mitogen-activated protein kinase signaling inhibition normalizes RRM1/2 expression and synergizes with gemcitabine treatment in vivo to reduce the proliferative index.
    Our results suggest that PDAC characterized by SMAD4 deficiency and oncogenic NFATc1/SMAD3/cJUN complex formation exposes sensitivity to a mitogen-activated protein kinase signaling inhibition and gemcitabine combination 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: Clinical Trial, Phase I
    BRAF/MEK靶向治疗和免疫检查点抑制已显著改善晚期黑色素瘤患者的疾病控制和存活率。然而,大多数患者都没有从这两种疗法中获得持久的益处.由于耐药性的发展,BRAF靶向治疗通常具有有限的疗效持续时间。临床前数据表明,克服对BRAF/MEK靶向治疗的抗性的一种可能方法可能是添加CSF1R抑制。在这项I/II期研究中,我们评估了抗集落刺激因子-1受体(CSF-1R)单克隆抗体LY3022855与BRAF抑制剂vemurafenib和MEK抑制剂cobimetinib在BRAFV600E/K突变转移性黑色素瘤患者中的安全性和有效性。由于申办方终止了LY3022855的开发计划,该试验提前终止。在2017年8月至2018年5月期间,有5名受试者参加。三名患者经历了被认为可能与LY3022855相关的3级事件。没有与LY3022855相关的4级或5级事件。5例患者中有1例完全缓解(CR),而其他4人患有进行性疾病(PD)。中位无进展生存期为3.9个月(90%CI:1.9-37.2个月)。LY3022855的CSF1R抑制与Vemurafenib和cobimetinib的BRAF/MEK抑制组合在小黑色素瘤群体中难以耐受。在这个小样本患者中观察到一个反应,表明这种组合可能值得进一步探索。
    BRAF/MEK targeted therapies and immune checkpoint inhibition have dramatically improved disease control and survival of patients with advanced melanoma. However, most patients do not have durable benefit from either of these therapies. BRAF targeted therapy often has a limited duration of efficacy due to the development of resistance. Pre-clinical data suggest that one possible way to overcome resistance to BRAF/MEK targeted therapy may be the addition of CSF1R inhibition. In this phase I/II study we evaluated the safety and efficacy of LY3022855, an anti-colony stimulating factor-1 receptor (CSF-1R) monoclonal antibody in combination with the BRAF inhibitor vemurafenib and the MEK inhibitor cobimetinib in patients with BRAF V600E/K mutant metastatic melanoma. The trial was terminated early due to discontinuation of the development program for LY3022855 by the sponsor. Between August 2017 and May 2018 five pts were enrolled. Three patients experienced grade 3 events that were deemed possibly related to LY3022855. There were no grade 4 or grade 5 events related to LY3022855. One of the 5 patients had a complete response (CR), whereas the other 4 had progressive disease (PD). Median progression free survival was 3.9 months (90% CI: 1.9-37.2 mos). CSF1R inhibition with LY3022855 in combination with BRAF/MEK inhibition with vemurafenib and cobimetinib was difficult to tolerate in a small melanoma population. One response was observed in this small sample of patients suggesting this combination might be worthy of further exploration.
    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
    近年来,棕色和白色脂肪组织的热分化和激活被认为是治疗和改善肥胖的主要创新和有前途的策略之一。然而,对这一过程的药理学方法有有限和不足的承诺,这对肥胖治疗提出了更大的挑战。这项研究评估了U0126化合物对脂肪生成过程中热分化激活的影响。结果表明,U0126预处理启动白色和棕色前脂肪细胞,以上调产热和线粒体基因,并增强分化过程中的功能。我们确定U0126介导的产热分化诱导部分通过AMPK激活信号发生。这项研究的结果表明,U0126是一种有前途的替代配体,用于寻求增加生热脂肪细胞形成和改善能量消耗的药理学选择。因此,它可以为发现治疗肥胖及其相关并发症的治疗药物铺平道路。
    In recent years, thermogenic differentiation and activation in brown and white adipose tissues have been regarded as one of the major innovative and promising strategies for the treatment and amelioration of obesity. However, the pharmacological approach towards this process has had limited and insufficient commitments, which presents a greater challenge for obesity treatment. This research evaluates the effects of U0126 compound on the activation of thermogenic differentiation during adipogenesis. The results show that U0126 pretreatment primes both white and brown preadipocytes to upregulate thermogenic and mitochondrial genes as well as enhance functions during the differentiation process. We establish that U0126-mediated thermogenic differentiation induction occurs partially via AMPK activation signaling. The findings of this research suggest U0126 as a promising alternative ligand in pursuit of a pharmacological option to increase thermogenic adipocyte formation and improve energy expenditure. Thus it could pave the way for the discovery of therapeutic drugs for the treatment of obesity and its related complications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    由于与Noonan综合征相关的淋巴发育不良引起的难治性乳糜积液由于蛋白质和免疫球蛋白损失而导致大量发病率和死亡率。很少有病例报道曲美替尼治疗成功,所有常规治疗均失败。
    我们介绍了一个患有Noonan综合征和肥厚型心肌病的女孩,她出现了危及生命的难治性乳糜胸,所有常规治疗方案都失败了。她成功地用丝裂原激活的细胞外信号调节激酶抑制剂曲美替尼治疗。
    对于一部分努南综合征和严重肺淋巴管扩张症患者,使用曲美替尼抑制MEK正在成为一种可能的挽救性治疗选择。需要更多的经验来建立最佳的治疗方案和长期结果。
    UNASSIGNED: Refractory chylous effusions due to lymphatic dysplasia related to Noonan syndrome cause significant morbidity and mortality due to protein and immunoglobulin losses. Very few cases have been published reporting successful treatment of patients with trametinib where all conventional treatments had failed.
    UNASSIGNED: We present a girl with Noonan syndrome and hypertrophic cardiomyopathy who presented with life-threatening refractory chylothorax where all conventional treatment options failed. She was successfully treated with mitogen-activated extracellular signal-regulated kinase inhibitor trametinib.
    UNASSIGNED: MEK inhibition with trametinib is emerging as a possible salvage treatment option for a subset of patients with Noonan syndrome and severe pulmonary lymphangiectasia. More experience is required to establish optimal treatment regimen and long-term outcomes.
    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
    背景:随着BRAF/MEK定向治疗和免疫检查点抑制的发展,转移性黑色素瘤患者的预后得到了显着改善。然而,对治疗的抵抗仍然是一个挑战,特别是BRAF/MEK靶向治疗,其疗效持续时间通常有限。临床前数据表明,在BRAF/MEK靶向治疗中添加CSF1抑制可能会降低耐药性并提高疗效。
    方法:我们进行了I/II期研究,以确定在BRAFV600E/K突变转移性黑色素瘤患者中,MCS110抑制CSF1与DRAF/MEK抑制联合dabrafenib/trametinib的安全性和有效性。由于研究赞助者决定停止MCS110的进一步开发,该试验提前终止。
    结果:在2018年9月至2019年7月期间,有6名患者被纳入研究。患者平均分为女性(50%)和男性(50%),中位年龄为59.5岁。(26-71).五名患者经历了可能与其中一种疗法有关的3级毒性,没有4级或5级事件.根据RECIST1.1,一名患者有部分反应(PR),一名患者疾病稳定(SD),3例患者出现疾病进展(PD)。中位无进展生存期为2.3个月(90%CI:1.3个月未达到)。
    结论:MCS110与dabrafenib和trametinib联合用药在小黑素瘤人群中具有良好的耐受性。在这个小样本患者中观察到一个反应,表明这种组合可能值得进一步探索。
    BACKGROUND: Prognosis for patients with metastatic melanoma has been improved dramatically with the development of BRAF/MEK directed therapy and immune checkpoint inhibition. However, resistance to therapy remains a challenge, particularly with BRAF/MEK targeted therapy which often has a limited duration of efficacy. Pre-clinical data suggest that adding CSF1 inhibition to BRAF/MEK targeted therapy may reduce resistance and increase efficacy.
    METHODS: We performed a phase I/II study to determine the safety and efficacy of CSF1 inhibition with MCS110 in combination with BRAF/MEK inhibition with dabrafenib/trametinib in patients with BRAF V600E/K mutant metastatic melanoma. The trial was terminated early due to a decision by the study sponsor to cease further development of MCS110.
    RESULTS: Between September 2018 to July 2019 six patients were enrolled on the study. Patients were evenly split between female (50%) and male (50%) with a median age of 59.5 yrs. (26-71). Five patients experienced grade 3 toxicities that were possibly related to one of the therapies, there were no grade 4 or grade 5 events. One patient had a partial response (PR) by RECIST 1.1, one patient had stable disease (SD), 3 patients had disease progression (PD). Median progression free survival was 2.3 months (90% CI: 1.3 mos to not reached).
    CONCLUSIONS: MCS110 in combination with dabrafenib and trametinib was reasonably well tolerated in a small melanoma population. One response was observed in this small sample of patients suggesting this combination might be worthy of further exploration.
    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
    RAS-MAPK信号通路是人类癌症中最常见的失调通路之一。针对该途径的小分子抑制剂在患有各种癌症类型的患者中具有临床活性并且可以改善患者结果。然而,这些药物的使用与不良反应有关,这可能导致剂量减少或治疗中断。对目标的更好的分子理解,肿瘤外效应可能改善毒性管理。在本研究中,我们的目的是通过药理学抑制RAS-MAPK信号通路,确定肝脏早期启动的生物学变化.为此,我们使用小鼠和人肝细胞细胞系测试了MEK抑制剂PD0325901的作用。雄性C57BL/6小鼠用载体或PD0325901治疗6天,其次是肝脏和表型表征的转录组分析。药物MEK抑制改变了423个基因的表达,其中78个上调,345个下调。我们确认了Shp,转录抑制因子,和Cyp7a1,将胆固醇转化为胆汁酸的限速酶,作为最高差异表达基因。PD0325901处理也影响了参与胆汁酸调节的其他基因,这与血浆胆汁酸组成和粪便胆汁酸组成和总水平的变化以及早期肝毒性的预测性生物标志物升高有关。总之,短期药物MEK抑制导致胆汁酸代谢的深刻变化,这可能解释了药理学抑制RAS-MAPK通路的一些临床不良反应,包括胃肠道并发症和肝毒性。
    The RAS-MAPK signaling pathway is one of the most frequently dysregulated pathways in human cancer. Small molecule inhibitors directed against this pathway have clinical activity in patients with various cancer types and can improve patient outcomes. However, the use of these drugs is associated with adverse effects, which can result in dose reduction or treatment interruption. A better molecular understanding of on-target, off-tumor effects may improve toxicity management. In the present study, we aimed to identify early initiating biological changes in the liver upon pharmacological inhibition of the RAS-MAPK signaling pathway. To this end, we tested the effect of MEK inhibitor PD0325901 using mice and human hepatocyte cell lines. Male C57BL/6 mice were treated with either vehicle or PD0325901 for six days, followed by transcriptome analysis of the liver and phenotypic characterization. Pharmacological MEK inhibition altered the expression of 423 genes, of which 78 were upregulated and 345 were downregulated. We identified Shp, a transcriptional repressor, and Cyp7a1, the rate-limiting enzyme in converting cholesterol to bile acids, as the top differentially expressed genes. PD0325901 treatment also affected other genes involved in bile acid regulation, which was associated with changes in the composition of plasma bile acids and composition and total levels of fecal bile acids and elevated predictive biomarkers of early liver toxicity. In conclusion, short-term pharmacological MEK inhibition results in profound changes in bile acid metabolism, which may explain some of the clinical adverse effects of pharmacological inhibition of the RAS-MAPK pathway, including gastrointestinal complications and hepatotoxicity.
    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