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: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: 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: Journal Article
    未经证实:在>90%的胰腺导管腺癌(PDAC)中,丝裂原激活蛋白激酶激酶(MEK)被突变的KRAS激活。MEK和黏着斑激酶(FAK)在PDAC中经常被共同激活,为联合使用曲美替尼提供了理论基础。口服变构MEK1/2抑制剂,GSK2256098,一种口服FAK抑制剂。
    UNASSIGNED:一线姑息性化疗后疾病进展的晚期PDAC患者接受GSK2256098250mg每日2次和曲美替尼0.5mg每日1次口服治疗。主要终点是临床获益(CB;完全缓解,部分响应,或疾病稳定≥24周)。计划使用2期minimax设计(P0=0.15,P1=0.40;α=0.05,功率0.86)招募24名患者。如果2/12或更少的患者在阶段1结束时达到CB,则该组合将被认为是无活性的,并且如果在阶段2结束时>7/24的反应可评估患者达到CB,则该组合将被认为是活性的。收集系列血液样品用于循环肿瘤DNA(ctDNA)突变谱。
    UNASSIGNED:16例患者入组,11例患者可评估反应。在这11人中,有10人患有进行性疾病作为最佳肿瘤反应,其中1人病情稳定4个月。未观察到治疗相关的≥3级不良事件(AE)。中位无进展生存期(PFS)为1.6(95%CI:1.5-1.8)个月,中位总生存期(OS)为3.6(95%CI:2.7-未达到)个月。一名无法评估反应的患者实现了5个月的临床稳定性,CA19-9和ctDNA水平降低,在临床进展时在ctDNA中检测到MAP2K1治疗抗性突变。
    UNASSIGNED:GSK2256098和曲美替尼的组合耐受性良好,但在未选择的晚期PDAC中无活性。
    UNASSIGNED: Mitogen-activated protein kinase kinase (MEK) is activated by mutated KRAS in >90% of pancreatic ductal adenocarcinoma (PDAC). MEK and focal adhesion kinase (FAK) are frequently co-activated in PDAC providing a rationale for combining trametinib, an oral allosteric MEK1/2 inhibitor, with GSK2256098, an oral FAK inhibitor.
    UNASSIGNED: Advanced PDAC patients whose disease progressed after first line palliative chemotherapy were treated with GSK2256098 250 mg twice daily and trametinib 0.5 mg once daily orally. The primary endpoint was clinical benefit (CB; complete response, partial response, or stable disease ≥24 weeks). Twenty-four patients were planned to enroll using a 2-stage minimax design (P0=0.15, P1=0.40; alpha =0.05, power 0.86). The combination would be considered inactive if 2/12 or fewer patients achieved CB at the end of stage 1, and would be considered active if >7/24 response-evaluable patients achieved CB by the end of stage 2. Serial blood samples were collected for circulating tumor DNA (ctDNA) mutation profiling.
    UNASSIGNED: Sixteen patients were enrolled and 11 were response evaluable. Of those 11, 10 had progressive disease as best tumor response and one had stable disease for 4 months. No treatment related grade ≥3 adverse events (AEs) were observed. The median progression free survival (PFS) was 1.6 (95% CI: 1.5-1.8) months and the median overall survival (OS) was 3.6 (95% CI: 2.7-not reached) months. One response-inevaluable patient achieved clinical stability for 5 months with reduction in CA19-9 and ctDNA levels with a MAP2K1 treatment resistance mutation detected in ctDNA at clinical progression.
    UNASSIGNED: The combination of GSK2256098 and trametinib was well tolerated but was not active in unselected advanced PDAC.
    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/目标组合BRAF(vemurafenib,Dabrafenib,或恩科非尼)加MEK(曲美替尼,考比替尼,或比米替尼)抑制剂疗法现已广泛用于转移性黑色素瘤的治疗。然而,这些药物的颅内反应数据有限.我们旨在评估BRAF+MEK抑制剂对BRAF突变黑色素瘤脑转移(BM)患者的颅内疗效,并确定这些新药的失败模式,以告知局部颅内治疗的最佳整合。材料和方法我们回顾性回顾了2006年至2020年在我们机构开始BRAF加MEK抑制剂时发生脑转移的BRAF突变黑色素瘤患者的图表,其中至少有一次未经治疗的脑转移。我们收集了每个患者和每个病变的人口统计数据,治疗方式,和结果。局部累积发生率(LF),远端颅内(DF),和颅外衰竭(EF)通过竞争性风险分析计算,将死亡作为竞争性风险,并在最后一次脑部MRI随访时进行审查.LF基于每个病变计算,而DF和EF基于每个患者计算。DF被定义为任何新的颅内病变。使用Kaplan-Meier分析总生存期(OS)。Logistic回归用于确定LF的预测因子。结果我们确定了10例患者的63例未经治疗的脑转移。中位年龄为50.5岁。每位患者的五个最大未治疗的脑转移的直径的中值总和为20mm(四分位距15-39mm),并且所有可测量的病变的中值直径为4mm。中位随访时间为9.0个月(1.4个月-46.2个月)。中位OS为13.6个月。LF的一年累积发病率,DF,EF为17.1%,88.6和71.4%,分别。到LF的中位时间,DF,从BRAF加MEK抑制剂开始的EF为9.0个月,4.7个月,7.0个月,分别。在单变量分析中,BM的较大大小与LF相关(每1毫米直径增加1.13,95%置信区间1.019至1.308,p<0.02)。两名(20%)患者最终接受了立体定向放射外科治疗,和2(20%)接受全脑放疗颅内进展。结论尽管BRAF突变型黑色素瘤伴BM患者对BRAF+MEK抑制剂有良好的局部控制,死亡和远端颅内外进展的竞争风险很高.具有较大脑转移的患者可从局部治疗中获益。
    Purpose/Objectives Combination BRAF (vemurafenib, dabrafenib, or encorafenib) plus MEK (trametinib, cobimetinib, or binimetinib) inhibitor therapy is now widely used in the treatment of metastatic melanoma. However, data for intracranial response to these drugs are limited. We aimed to evaluate the intracranial efficacy of BRAF plus MEK inhibitors in patients with BRAF-mutant melanoma with brain metastases (BM) and to determine patterns of failure of these new agents to inform optimal integration of local intracranial therapy. Materials and methods We retrospectively reviewed charts of patients with BRAF-mutant melanoma with metastasis to the brain with at least one untreated brain metastasis at the time of initiation of BRAF plus MEK inhibitors at our institution from 2006 to 2020. We collected per-patient and per-lesion data on demographics, treatment modality, and outcomes. The cumulative incidence of local (LF), distant intracranial (DF), and extracranial failure (EF) were calculated with competing risk analysis with death as a competing risk and censored at the last brain MRI follow-up. LF was calculated on a per-lesion basis while DF and EF were calculated on a per-patient basis. DF was defined as any new intracranial lesions. Overall survival (OS) was analyzed using Kaplan-Meier. Logistic regression was used to identify predictors for LF. Results We identified 10 patients with 63 untreated brain metastases. The median age was 50.5 years. The median sum of the diameters of the five largest untreated brain metastases per patient was 20 mm (interquartile range 15-39 mm) and the median diameter for all measurable lesions was 4 mm. Median follow-up time was 9.0 months (range 1.4 months-46.2 months). Median OS was 13.6 months. The one-year cumulative incidence of LF, DF, and EF was 17.1%, 88.6, and 71.4%, respectively. The median time to LF, DF, and EF from the start of BRAF plus MEK inhibitors was 9.0 months, 4.7 months, and 7.0 months, respectively. The larger size of the BM was associated with LF on univariate analysis (odds ratio 1.13 per 1 mm increase in diameter, 95% confidence interval 1.019 to 1.308, p<0.02). Two (20%) patients eventually received stereotactic radiosurgery, and 2 (20%) received whole-brain radiotherapy for intracranial progression. Conclusion Although patients with BRAF-mutant melanoma with BM had fair local control on BRAF plus MEK inhibitors, the competing risk of death and distant intracranial and extracranial progression was high. Patients with larger brain metastases may benefit from local therapy.
    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
    未经批准:在携带致癌ALK或ROS1重排的NSCLC患者中,与细胞毒性化疗相比,酪氨酸激酶抑制剂产生了高反应率和无进展生存期的改善;然而,后天的抵抗力最终发展。在临床前模型中,ALK和MEK共抑制能够克服ALK抑制剂抗性。
    UNASSIGNED:开始了ALK/ROS1抑制剂ceritinib和MEK抑制剂trametinib在伴有ALK或ROS1融合的难治性NSCLC患者中的1期研究。使用三加三剂量递增方案。研究了两种剂量水平。主要终点是确定组合的安全性和耐受性。
    未经批准:9名患者(n=8ALK+,n=1ROS1+)参加研究,并完成至少一个治疗周期。最常见的不良事件(所有等级)是腹泻(n=9;100%),皮疹(n=8;89%),腹痛(n=5;56%),天冬氨酸转氨酶/丙氨酸转氨酶水平升高(n=4;44%)。总有效率为22%,而疾病控制率为56%。中位反应持续时间为7.85个月。中位无进展生存期为3.0个月(95%置信区间:1.5-7.0个月)。中位总生存期为8.9个月(95%置信区间:2.0-未达到)。
    UNASSIGNED:本试验的数据表明,色瑞替尼和曲美替尼的组合没有意外毒性,可以确定耐受剂量。在先前的ALK/ROS1抑制剂治疗进展后,一部分患者似乎从该治疗中获得了临床益处。ClinicalTrials.gov标识符:NCT03087448。
    UNASSIGNED: In patients with NSCLC harboring oncogenic ALK or ROS1 rearrangements, tyrosine kinase inhibitors have yielded high response rates and improvements in progression-free survival compared with cytotoxic chemotherapy; however, acquired resistance eventually develops. In preclinical models, ALK and MEK coinhibition was able to overcome ALK inhibitor resistance.
    UNASSIGNED: A phase 1 study of the ALK/ROS1 inhibitor ceritinib and the MEK inhibitor trametinib in patients with refractory NSCLC harboring ALK or ROS1 fusions was initiated. A three plus three dose-escalation scheme was used. Two dose levels were investigated. The primary end point was to determine the safety and tolerability of the combination.
    UNASSIGNED: Nine patients (n = 8 ALK+, n = 1 ROS1+) were enrolled in the study and completed at least one cycle of therapy. The most common adverse events (all grades) were diarrhea (n = 9; 100%), rash (n = 8; 89%), abdominal pain (n = 5; 56%), and elevated aspartate transaminase/alanine transaminase level (n = 4; 44%). The overall response rate was 22%, whereas disease control rate was 56%. Median duration of response was 7.85 months. The median progression-free survival was 3.0 months (95% confidence interval: 1.5-7.0 mo). The median overall survival was 8.9 months (95% confidence interval: 2.0-not reached).
    UNASSIGNED: Data from this trial indicate that the combination of ceritinib and trametinib had no unexpected toxicities and that a tolerable dose could be identified. A subset of patients seemed to obtain clinical benefit from this treatment after progression on prior ALK/ROS1 inhibitor treatment.ClinicalTrials.gov Identifier: NCT03087448.
    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
    放射病是一组临床定义的发育综合征,由RAS/丝裂原活化蛋白(MAPK)级联的种系变体引起。原型RASopathy是Noonan综合征,与心脏皮肤综合征等相关疾病有表型重叠,科斯特洛综合征,努南综合征伴多个腹水,和其他人。在这份最新的审查中,我们总结了有关这些疾病中未满足的治疗需求的最新知识,以及通过RAS/MAPK相关癌症疗法的见解获得的新治疗方法。特别是通过在有严重疾病表现的患者中抑制MEK1/2和mTOR。我们探索了将目前正在开发的更大分子库整合到未来护理计划中的可能性。最后,我们描述了医学和伦理方面的挑战以及该领域未来临床试验的机遇.
    The RASopathies are a group of clinically defined developmental syndromes caused by germline variants of the RAS/mitogen-activated protein (MAPK) cascade. The prototypic RASopathy is Noonan syndrome, which has phenotypic overlap with related disorders such as cardiofaciocutaneous syndrome, Costello syndrome, Noonan syndrome with multiple lentigines, and others. In this state-of-the-art review, we summarize current knowledge on unmet therapeutic needs in these diseases and novel treatment approaches informed by insights from RAS/MAPK-associated cancer therapies, in particular through inhibition of MEK1/2 and mTOR in patients with severe disease manifestations. We explore the possibilities of integrating a larger arsenal of molecules currently under development into future care plans. Lastly, we describe both medical and ethical challenges and opportunities for future clinical trials in the field.
    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
    葡萄膜黑色素瘤(UM)与转移环境中的不良结局有关,并且在几乎所有情况下都存在激活突变,导致MAPK信号传导上调。司米替尼的疗效,MEK1/2的口服变构抑制剂在75mgBID的连续给药方案中受到限制.临床前研究表明,间歇性MEK抑制减少代偿途径活化并促进T细胞活化。我们假设间歇性给药司美替尼可以降低毒性,允许增加剂量的给药,并实现更完整的通路抑制,从而导致提高的抗肿瘤活性。
    我们在转移性UM患者中使用间歇给药方案进行了司美替尼的Ib期试验。主要目的是评估最大耐受剂量(MTD)并评估安全性和耐受性。次要目标包括评估总体反应率(RR),无进展生存期(PFS)和总生存期(OS)。在基线时收集肿瘤活检,在第3天(治疗),并在第11-14天(停止治疗)之间对9名患者进行药效学(PD)评估。
    29名患者入选,并在4个剂量水平(DL;DL1:100mgBID;DL2:125mgBID;DL3:150mgBID;DL4:175mgBID)接受至少一剂司美替尼。所有患者均出现治疗相关不良事件(TRAE),5/29(17%)发展为3级或更高的TRAE。观察到五种剂量限制性毒性(DLT):在DL2中为2/20,在DL3中为2/5,在DL4中为1/1。估计的MTD为150毫克BID(DL3),估计毒性概率为29%(90%概率区间为16%-44%)。未观察到反应;11/29患者达到稳定疾病(SD)的最佳反应。中位PFS和OS分别为1.8个月(95%CI1.7,4.5)和7.1个月(95%CI5.3,11.5)。PD分析证实在第3天所有样品中至少部分途径抑制,其中7个病例在第11-14天之间重新激活。
    我们确定了150mgBID作为间歇性司米替尼的MTD,表示比连续剂量MTD(75mgBID)增加100%。然而,使用该给药方案未观察到显著的临床疗效.
    UNASSIGNED: Uveal melanoma (UM) is associated with poor outcomes in the metastatic setting and harbors activating mutations resulting in upregulation of MAPK signaling in almost all cases. The efficacy of selumetinib, an oral allosteric inhibitor of MEK1/2, was limited when administered at a continual dosing schedule of 75 mg BID. Preclinical studies demonstrate that intermittent MEK inhibition reduces compensatory pathway activation and promotes T cell activation. We hypothesized that intermittent dosing of selumetinib would reduce toxicity, allow for the administration of increased doses, and achieve more complete pathway inhibition, thus resulting in improved antitumor activity.
    UNASSIGNED: We conducted a phase Ib trial of selumetinib using an intermittent dosing schedule in patients with metastatic UM. The primary objective was to estimate the maximum tolerated dose (MTD) and assess safety and tolerability. Secondary objectives included assessment of the overall response rate (RR), progression-free survival (PFS) and overall survival (OS). Tumor biopsies were collected at baseline, on day 3 (on treatment), and between days 11-14 (off treatment) from 9 patients for pharmacodynamic (PD) assessments.
    UNASSIGNED: 29 patients were enrolled and received at least one dose of selumetinib across 4 dose levels (DL; DL1: 100 mg BID; DL2: 125 mg BID; DL3: 150 mg BID; DL4: 175 mg BID). All patients experienced a treatment-related adverse event (TRAE), with 5/29 (17%) developing a grade 3 or higher TRAE. Five dose limiting toxicities (DLT) were observed: 2/20 in DL2, 2/5 in DL3, 1/1 in DL4. The estimated MTD was 150 mg BID (DL3), with an estimated probability of toxicity of 29% (90% probability interval 16%-44%). No responses were observed; 11/29 patients achieved a best response of stable disease (SD). The median PFS and OS were 1.8 months (95% CI 1.7, 4.5) and 7.1 months (95% CI 5.3, 11.5). PD analysis demonstrated at least partial pathway inhibition in all samples at day 3, with reactivation between days 11-14 in 7 of those cases.
    UNASSIGNED: We identified 150 mg BID as the MTD of intermittent selumetinib, representing a 100% increase over the continuous dose MTD (75 mg BID). However, no significant clinical efficacy was observed using this dosing schedule.
    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
    结直肠癌(CRC)是美国癌症死亡的第二大原因。RAS途径在超过55%的CRC中被激活,并且已经被靶向用于MEK抑制剂的治疗性干预。不幸的是,许多患者有从头抵抗,或者可以对这种新型药物产生抗药性。我们假设这种抗性的大部分可能通过SRC作为一个共同的信号转导节点,并且SRC的抑制可以抑制MEK抑制抵抗机制。共有分子亚型(CMS)4的CRC肿瘤,富含干细胞,难以成功治疗,并已建议通过耐药机制逃避传统的化疗药物。这里,我们评估了同时靶向两个途径,通过克服耐药性产生有效的治疗。我们表明,与单独使用任一种药物治疗相比,曲美替尼(MEKi)与达沙替尼(SRCi)的组合在16个测试的CRC细胞系中的8个中提供了增强的细胞死亡。为了能够选择敏感的单元格,我们同时评估了经过验证的18基因RAS途径激活签名评分以及13基因MEKi抗性签名评分,我们假设这可以预测肿瘤对这种双重靶向治疗的敏感性。我们发现对双重治疗敏感的细胞系主要是CMS4,并且具有高18基因和高13基因评分,表明这些细胞系具有从头MEKi敏感性的潜力,但受到MEKi抗性的快速发展。13基因得分与SRC激活得分高度相关,表明抗性取决于SRC。我们的数据表明,RAS途径激活和MEKi抗性的基因表达特征评分可能有助于确定哪些CRC肿瘤将对MEKi和SRCi的新型药物组合产生反应。
    Colorectal cancer (CRC) is the second leading cause of cancer death in the United States. The RAS pathway is activated in more than 55% of CRC and has been targeted for therapeutic intervention with MEK inhibitors. Unfortunately, many patients have de novo resistance, or can develop resistance to this new class of drugs. We have hypothesized that much of this resistance may pass through SRC as a common signal transduction node, and that inhibition of SRC may suppress MEK inhibition resistance mechanisms. CRC tumors of the Consensus Molecular Subtype (CMS) 4, enriched in stem cells, are difficult to successfully treat and have been suggested to evade traditional chemotherapy agents through resistance mechanisms. Here, we evaluate targeting two pathways simultaneously to produce an effective treatment by overcoming resistance. We show that combining Trametinib (MEKi) with Dasatinib (SRCi) provides enhanced cell death in 8 of the 16 tested CRC cell lines compared to treatment with either agent alone. To be able to select sensitive cells, we simultaneously evaluated a validated 18-gene RAS pathway activation signature score along with a 13-gene MEKi resistance signature score, which we hypothesize predict tumor sensitivity to this dual targeted therapy. We found the cell lines that were sensitive to the dual treatment were predominantly CMS4 and had both a high 18-gene and a high 13-gene score, suggesting these cell lines had potential for de novo MEKi sensitivity but were subject to the rapid development of MEKi resistance. The 13-gene score is highly correlated to a score for SRC activation, suggesting resistance is dependent on SRC. Our data show that gene expression signature scores for RAS pathway activation and for MEKi resistance may be useful in determining which CRC tumors will respond to the novel drug combination of MEKi and SRCi.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号