MEK

MEK
  • 文章类型: Journal Article
    这项研究调查了FGFR抑制剂futibatinib(FTB)和MEK抑制剂binimetinib(BTB)用于KRASmtNSCLC治疗的组合。开发并验证了一种用于测量大鼠血浆中FTB和BTB浓度的分析方法,遵守USFDA指南。对45μL血浆样品进行液-液萃取,达到6.5-min的运行时间。线性校准曲线范围为2至100ng/mL。日内和日间准确度介于92.06%和100.08%之间。高浓度样品后的四次空白注射解决了明显的残留。不同浓度的萃取回收率平均为92.06%~102.37%。在空白血浆中未检测到显著的内源性干扰。两种药物的LLOQ均为2.0ng/mL。选择性,矩阵效应,稳定性,和稀释完整性满足验收标准。该方法评估了5mg/kg联合治疗中的FTB和BTB相互作用潜力。这些发现为未来的临床试验提供了必要的药代动力学见解。
    This study investigates the combination of FGFR inhibitor futibatinib (FTB) and MEK inhibitor binimetinib (BTB) for KRASmt NSCLC therapy. An analytical method was developed and validated for measuring FTB and BTB concentrations in rat plasma, adhering to USFDA guidelines. Using liquid-liquid extraction on 45-μL plasma samples, a 6.5-min run time was achieved. The linear calibration curve ranged from 2 to 100 ng/mL. Intra-day and inter-day accuracy ranged between 92.06% and 100.08%. Four blank injections post high-concentration samples resolved significant carryover. Extraction recoveries averaged 92.06% to 102.37% across concentrations. No significant endogenous interference was detected in blank plasma. The LLOQ for both drugs was 2.0 ng/mL. Selectivity, matrix effects, stability, and dilution integrity met the acceptance criteria. The method assessed FTB and BTB interaction potential in combination therapy at 5 mg/kg. The findings provide essential pharmacokinetics insights for future clinical trials.
    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
    在复发性神经母细胞瘤中观察到RAS/MAPK途径中的活化突变。临床前研究表明,这些肿瘤对RAS/MAPK通路抑制剂的敏感性增加,如MEK抑制剂。MEK抑制剂不诱导持久的反应作为单一药物,这表明需要确定靶向药物的协同组合以提供治疗益处。我们之前显示了MEK抑制剂之间的临床前治疗协同作用,曲美替尼,和一种对IGF1R特异的单克隆抗体,加尼单抗在RAS突变横纹肌肉瘤中的应用。神经母细胞瘤细胞,像横纹肌肉瘤细胞,对RAS/MAPK和IGF1R/AKT/mTOR途径的抑制敏感。我们假设曲美替尼和加尼单抗的组合对RAS突变的神经母细胞瘤有效。在这项研究中,曲美替尼和加尼单抗协同抑制细胞培养物中神经母细胞瘤细胞增殖并诱导细胞凋亡。我们还观察到在用曲美替尼和加尼单抗治疗的异位和原位异种移植模型中肿瘤起始延迟和生存期延长。然而,在接受曲美替尼和加尼单抗联合治疗的动物中观察到原发性和转移性肿瘤的生长.因此,在复发性或难治性RAS突变神经母细胞瘤患者中检测该组合之前,需要更多的临床前工作.
    Activating mutations in the RAS/MAPK pathway are observed in relapsed neuroblastoma. Preclinical studies indicate that these tumors have an increased sensitivity to inhibitors of the RAS/MAPK pathway, such as MEK inhibitors. MEK inhibitors do not induce durable responses as single agents, indicating a need to identify synergistic combinations of targeted agents to provide therapeutic benefit. We previously showed preclinical therapeutic synergy between a MEK inhibitor, trametinib, and a monoclonal antibody specific for IGF1R, ganitumab in RAS-mutated rhabdomyosarcoma. Neuroblastoma cells, like rhabdomyosarcoma cells, are sensitive to the inhibition of the RAS/MAPK and IGF1R/AKT/mTOR pathways. We hypothesized that the combination of trametinib and ganitumab would be effective in RAS-mutated neuroblastoma. In this study, trametinib and ganitumab synergistically suppressed neuroblastoma cell proliferation and induced apoptosis in cell culture. We also observed a delay in tumor initiation and prolongation of survival in heterotopic and orthotopic xenograft models treated with trametinib and ganitumab. However, the growth of both primary and metastatic tumors was observed in animals receiving the combination of trametinib and ganitumab. Therefore, more preclinical work is necessary before testing this combination in patients with relapsed or refractory RAS-mutated neuroblastoma.
    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:KRAS突变经常发生在癌症中,尤其是胰腺导管腺癌,结直肠癌,和非小细胞肺癌。尽管最近批准了KRASG12C抑制剂,对于所有KRAS突变癌症,目前尚未建立有效的精准治疗.KRAS突变癌症的许多治疗方法,包括表观基因组靶向药物,目前正在调查中。小泛素样修饰蛋白(SUMO)是通过称为SUMO化和去SUMO化的过程与细胞中的其他蛋白质共价连接并分离的小蛋白质家族。我们评估了SUMO化抑制在KRAS突变的癌细胞中是否有效。
    方法:在多种人和小鼠KRAS突变的癌细胞系中评估了第一类SUMO激活酶E抑制剂TAK-981(subasumstat)的功效。使用TaqMan阵列的基因表达测定用于鉴定TAK-981功效的生物标志物。使用免疫印迹分析研究了SUMO化抑制的生物学作用和随后的调节机制,免疫荧光测定,和老鼠模型。
    结果:我们发现TAK-981下调了目前无法用药的MYC的表达,并有效抑制了不同组织类型中表达MYC的KRAS突变癌症的生长。此外,通过MYC过表达使TAK-981抗性细胞对SUMO化抑制敏感。TAK-981通过改变SUMO化和泛素化之间的平衡并促进泛素-蛋白酶体系统中的关键因素MYC和Fbxw7的结合来诱导MYC的蛋白酶体降解。在使用小鼠来源的CMT167细胞系的免疫活性和免疫缺陷小鼠模型中,TAK-981单一疗法的功效是显著的但适度的。由于MAPK抑制KRAS下游途径在KRAS突变癌症中至关重要,我们预计SUMO化和MEK的共同抑制可能是一个很好的选择。令人惊讶的是,TAK-981和曲美替尼联合治疗可显著诱导多种细胞系和基因工程小鼠来源的类器官细胞凋亡.此外,在使用不同组织类型的细胞系的小鼠模型中,联合治疗导致肿瘤长期消退.最后,我们发现联合治疗可补充抑制Rad51和BRCA1并累积DNA损伤.
    结论:我们发现在KRAS突变的癌细胞中,MYC通过SUMO化抑制发生下调。我们的发现表明,SUMO化和MEK的双重抑制可能是通过增强DNA损伤积累来治疗表达MYC的KRAS突变癌症的有希望的治疗方法。
    BACKGROUND: KRAS mutations frequently occur in cancers, particularly pancreatic ductal adenocarcinoma, colorectal cancer, and non-small cell lung cancer. Although KRASG12C inhibitors have recently been approved, effective precision therapies have not yet been established for all KRAS-mutant cancers. Many treatments for KRAS-mutant cancers, including epigenome-targeted drugs, are currently under investigation. Small ubiquitin-like modifier (SUMO) proteins are a family of small proteins covalently attached to and detached from other proteins in cells via the processes called SUMOylation and de-SUMOylation. We assessed whether SUMOylation inhibition was effective in KRAS-mutant cancer cells.
    METHODS: The efficacy of the first-in-class SUMO-activating enzyme E inhibitor TAK-981 (subasumstat) was assessed in multiple human and mouse KRAS-mutated cancer cell lines. A gene expression assay using a TaqMan array was used to identify biomarkers of TAK-981 efficacy. The biological roles of SUMOylation inhibition and subsequent regulatory mechanisms were investigated using immunoblot analysis, immunofluorescence assays, and mouse models.
    RESULTS: We discovered that TAK-981 downregulated the expression of the currently undruggable MYC and effectively suppressed the growth of MYC-expressing KRAS-mutant cancers across different tissue types. Moreover, TAK-981-resistant cells were sensitized to SUMOylation inhibition via MYC-overexpression. TAK-981 induced proteasomal degradation of MYC by altering the balance between SUMOylation and ubiquitination and promoting the binding of MYC and Fbxw7, a key factor in the ubiquitin-proteasome system. The efficacy of TAK-981 monotherapy in immunocompetent and immunodeficient mouse models using a mouse-derived CMT167 cell line was significant but modest. Since MAPK inhibition of the KRAS downstream pathway is crucial in KRAS-mutant cancer, we expected that co-inhibition of SUMOylation and MEK might be a good option. Surprisingly, combination treatment with TAK-981 and trametinib dramatically induced apoptosis in multiple cell lines and gene-engineered mouse-derived organoids. Moreover, combination therapy resulted in long-term tumor regression in mouse models using cell lines of different tissue types. Finally, we revealed that combination therapy complementally inhibited Rad51 and BRCA1 and accumulated DNA damage.
    CONCLUSIONS: We found that MYC downregulation occurred via SUMOylation inhibition in KRAS-mutant cancer cells. Our findings indicate that dual inhibition of SUMOylation and MEK may be a promising treatment for MYC-expressing KRAS-mutant cancers by enhancing DNA damage accumulation.
    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
    胰腺癌(PC)显示出很高的致死率,在晚期患者的情况下,只能通过手术和化疗或姑息治疗的组合来面对。此外,PC肿瘤富含对现有化学治疗剂具有抗性的癌症干细胞(CSC)亚群,这引起了对新药鉴定的重要需求。为了填补这个空白,我们已经测试了微生物提取物的抗肿瘤活性,这种化学多样性提供了广泛的潜在新生物活性化合物。来自真菌Onychocolasp.的提取物。CF-107644通过高通量筛选进行测定,然后进行生物测定指导的分级分离,并鉴定和分离了六种具有抗肿瘤活性的二苯甲酮衍生物:甲骨癌A-F(#1-6)。用光谱法建立了化合物的结构,包括ESI-TOFMS,1D和2DNMR分析和X射线衍射。化合物#1-4显著抑制胰腺肿瘤细胞系的生长,低微摩尔中位数有效剂量(ED50s)。由于其促凋亡作用,化合物#1(甲骨颈酮A)被优先用于进一步分析,在3D球体和胰腺CSC上进一步验证。蛋白质表达测定表明,该作用在机械上与MEK癌基因信号通路的抑制有关。还通过减少由CSC产生的胰腺异种移植小鼠模型中的肿瘤生长,在体内证明了甲乔酮A的功效。总之,数据支持甲乔酮A是一种有前途的新型小分子,可用于开发PC的新疗法。
    Pancreatic cancer (PC) shows a high fatality rate that can only be faced with a combination of surgery and chemotherapy or palliative treatment in the case of advanced patients. Besides, PC tumors are enriched with subpopulations of cancer stem cells (CSCs) that are resistant to the existing chemotherapeutic agents, which raises an important need for the identification of new drugs. To fill this gap, we have tested the anti-tumoral activity of microbial extracts, which chemical diversity offers a broad spectrum of potential new bioactive compounds. Extracts derived from the fungus Onychocola sp. CF-107644 were assayed via high throughput screening followed by bioassay-guided fractionation and resulted in the identification and isolation of six benzophenone derivatives with antitumoral activity: onychocolones A-F (#1-6). The structures of the compounds were established by spectroscopic methods, including ESI-TOF MS, 1D and 2D NMR analyses and X-ray diffraction. Compounds #1-4 significantly inhibited the growth of the pancreas tumoral cell lines, with low-micromolar Median Effective Doses (ED50s). Compound #1 (onychocolone A) was prioritized for further profiling due to its pro-apoptotic effect, which was further validated on 3D spheroids and pancreatic CSCs. Protein expression assays showed that the effect was mechanistically linked to the inhibition of MEK onco-signaling pathway. The efficacy of onychocolone A was also demonstrated in vivo by the reduction of tumor growth in a pancreatic xenograft mouse model generated by CSCs. Altogether, the data support that onychocolone A is a promising new small molecule for hit-to-lead development of a new treatment for PC.
    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:有证据表明,RAF/MEK/ERK网络的较低活性与COVID-19轻度和中度病程的阳性结果相关。这种级联反应在COVID-19脓毒症中的作用仍未确定。因此,我们检验了以下假设:在COVID-19诱导的脓毒症中,RAF/MEK/ERK网络的活性与对30天生存率的影响相关.
    方法:我们使用了来自多中心CovidDataNet的81名前瞻性招募患者的生物材料。NRW研究队列(德国临床试验注册:DRKS00026184)与他们收集的病史,生命体征,实验室参数,微生物学发现和患者预后。通过使用邻近连接测定评估ERK磷酸化来测量ERK活性。
    结果:在校正的Cox回归模型中,诊断为COVID-19诱导的脓毒症后4天ERK活性增加与生存机会增加三倍以上相关。ERK活性独立于其他混杂因素,如Charlson合并症指数或SOFA评分(HR0.28,95%CI0.10-0.84,p=0.02)。
    结论:在COVID-19脓毒症过程中,RAF/MEK/ERK网络的高活性是一个保护因素,可能表明免疫系统恢复。需要进一步的研究来证实这些结果。
    OBJECTIVE: There is evidence that lower activity of the RAF/MEK/ERK network is associated with positive outcomes in mild and moderate courses of COVID-19. The effect of this cascade in COVID-19 sepsis is still undetermined. Therefore, we tested the hypothesis that activity of the RAF/MEK/ERK network in COVID-19-induced sepsis is associated with an impact on 30-day survival.
    METHODS: We used biomaterial from 81 prospectively recruited patients from the multicentric CovidDataNet.NRW-study cohort (German clinical trial registry: DRKS00026184) with their collected medical history, vital signs, laboratory parameters, microbiological findings and patient outcome. ERK activity was measured by evaluating ERK phosphorylation using a Proximity Ligation Assay.
    RESULTS: An increased ERK activity at 4 days after diagnosis of COVID-19-induced sepsis was associated with a more than threefold increased chance of survival in an adjusted Cox regression model. ERK activity was independent of other confounders such as Charlson Comorbidity Index or SOFA score (HR 0.28, 95% CI 0.10-0.84, p = 0.02).
    CONCLUSIONS: High activity of the RAF/MEK/ERK network during the course of COVID-19 sepsis is a protective factor and may indicate recovery of the immune system. Further studies are needed to confirm these results.
    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:对于局部晚期或潜在可切除的转移性黑色素瘤,新辅助治疗有望改善可操作性和临床结果,而不仅仅是前期手术和辅助治疗。
    方法:47例连续患者接受新辅助辅助BRAF抑制剂(BRAFi)/MEK抑制剂(MEKi)和手术治疗。
    结果:12例(26%)患者达到病理完全缓解,10例(21%)患者达到接近完全缓解。在整个群体中,中位无复发生存期为19.4个月,中位无远处转移生存期(mDMFS)为21.9个月.在病理完全缓解(pCR)/接近pCR的患者中,中位无复发生存期(RFS)和无远处转移生存期(DMFS)明显长于病理轻微缓解的患者,RFS的风险比(HR)=0.37(p=.005),DMFS的HR=0.33(p=.002)。中位随访52.5个月后,自BRAFi/MEKi治疗开始以来的中位无进展生存期为25.1个月.自开始新辅助治疗以来的中位治疗失败时间为22.2个月,pCR/近pCR患者明显更长(HR=0.45;p=0.022)。新辅助治疗未导致任何新的特定手术并发症。48个月后,在pCR/near-pCR和病理部分反应/病理无反应的患者中,RFS和总生存率分别为36.3%和64.8%或20%和37.4%,分别。
    结论:作者证实,BRAFi/MEKi联合应用是III/IV期黑色素瘤围手术期治疗的一种有效且安全的方案。对新辅助治疗的主要病理反应是这些患者中包括DMFS在内的复发的替代标志物。
    结论:我们的研究对诊断为边缘可切除的III或IV期黑色素瘤患者的新辅助-辅助系统治疗进行了大量综合分析。新辅助治疗有效地减少了疾病的体积,这有助于随后的手术切除。中位随访52.5个月后,自治疗开始以来的中位无进展生存期为25.1个月.12例患者有完全的病理反应,10例患者有接近完全的病理反应,他们的中位无复发生存期和无远处转移生存期明显长于病理部分反应或无反应的患者。对新辅助治疗的完全/接近完全的病理反应是无复发的替代标记,包括无远处转移,这些患者的生存。
    BACKGROUND: Neoadjuvant-adjuvant therapy for locally advanced or potentially resectable metastatic melanoma was expected to improve operability and clinical outcomes over upfront surgery and adjuvant treatment only.
    METHODS: Forty-seven consecutive patients were treated with neoadjuvant-adjuvant BRAF inhibitors (BRAFi)/MEK inhibitors (MEKi) and surgery.
    RESULTS: Twelve (26%) patients achieved a pathological complete response and 10 (21%) patients achieved a near-complete response. In the whole group, median recurrence-free survival was 19.4 months and median distant metastasis-free survival (mDMFS) was 21.9 months. In patients with a pathological complete response (pCR)/near-pCR median recurrence-free survival (RFS) and distant metastasis-free survival (DMFS) were significantly longer than in patients with minor pathological response with hazard ratio (HR) = 0.37 (p = .005) for RFS and HR = 0.33 (p = .002) for DMFS. After median follow-up of 52.5 months, median progression-free survival since BRAFi/MEKi therapy initiation was 25.1 months. The median time-to-treatment-failure since initiation of neoadjuvant therapy was 22.2 months and was significantly longer in patients with pCR/near-pCR (HR = 0.45; p = .022). Neoadjuvant therapy did not result in any new specific complications of surgery. After 48 months, RFS and overall survival were 36.3% and 64.8% or 20% and 37.4% in patients with pCR/near-pCR and pathological partial response/pathological nonresponse, respectively.
    CONCLUSIONS: The authors confirmed that BRAFi/MEKi combination is an effective and safe regimen in the perioperative treatment of stage III/IV melanoma. Major pathological response to neoadjuvant treatment is a surrogate marker of recurrence including DMFS in these patients.
    CONCLUSIONS: Our study presents a large comprehensive analysis of neoadjuvant-adjuvant systemic therapy in patients diagnosed with marginally resectable stage III or IV melanoma. Neoadjuvant therapy effectively reduced the volume of the disease, which facilitated subsequent surgical resection. After median follow-up of 52.5 months, median progression-free survival since therapy initiation was 25.1 months. Twelve patients had complete pathological response and 10 patients had a near-complete pathological response-and together they had median recurrence-free survival and distant metastasis-free survival significantly longer than in patients with pathological partial response or nonresponse. Complete/near-complete pathological response to neoadjuvant treatment is a surrogate marker of recurrence-free, including distant metastasis-free, survival in these patients.
    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
    历史上,尽管KRAS是实体肿瘤中最常见的致癌蛋白之一,但仍被认为是“不可用药”。主要是由于突变同工型中缺乏药理学上的“可药物”口袋。然而,能够靶向突变KRAS同工型特别是KRASG12C突变癌症的药物设计的开创性发展,已经为包含大量靶向不同信号通路的抑制剂的联合疗法的出现打开了大门。SHP2信号通路,主要以激活细胞内信号传导途径(如KRAS)而闻名,已成为此类联合疗法的潜在靶标,因为它已成为连接KRAS和免疫信号传导途径的信号蛋白,并为理解RAS/ERK/MAPK信号传导级联的重叠区域提供了联系。因此,具有有效的杀肿瘤活性以及在免疫调节中的作用的SHP2抑制剂引起了研究人员的浓厚兴趣,以探索其作为KRAS突变实体瘤的联合疗法的潜力。然而,这些联合疗法的兴奋需要克服耐药性和毒性增强带来的挑战。在这次审查中,我们将讨论KRAS和SHP2信号通路及其在免疫调节和肿瘤微环境调节中的作用,并分析针对这些信号通路的不同联合疗法的积极作用和缺点,以及它们目前和未来治疗实体肿瘤的潜力.
    Historically, KRAS has been considered \'undruggable\' inspite of being one of the most frequently altered oncogenic proteins in solid tumors, primarily due to the paucity of pharmacologically \'druggable\' pockets within the mutant isoforms. However, pioneering developments in drug design capable of targeting the mutant KRAS isoforms especially KRASG12C-mutant cancers, have opened the doors for emergence of combination therapies comprising of a plethora of inhibitors targeting different signaling pathways. SHP2 signaling pathway, primarily known for activation of intracellular signaling pathways such as KRAS has come up as a potential target for such combination therapies as it emerged to be the signaling protein connecting KRAS and the immune signaling pathways and providing the link for understanding the overlapping regions of RAS/ERK/MAPK signaling cascade. Thus, SHP2 inhibitors having potent tumoricidal activity as well as role in immunomodulation have generated keen interest in researchers to explore its potential as combination therapy in KRAS mutant solid tumors. However, the excitement with these combination therapies need to overcome challenges thrown up by drug resistance and enhanced toxicity. In this review, we will discuss KRAS and SHP2 signaling pathways and their roles in immunomodulation and regulation of tumor microenvironment and also analyze the positive effects and drawbacks of the different combination therapies targeted at these signaling pathways along with their present and future potential to treat solid tumors.
    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: Published Erratum
    [这更正了文章DOI:10.3389/fonc.202.782877。].
    [This corrects the article DOI: 10.3389/fonc.2022.782877.].
    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
    发作间尖峰是脑电图放电,发生在或靠近产生癫痫发作的脑区。虽然它们在癫痫发作中的作用还没有得到很好的理解,尖峰对认知和行为有深远的影响,取决于它们发生的地点和时间。我们先前证明,人类新皮质的尖峰区域在浅表皮质I-III层中显示出持续的MAPK激活,并且与更深皮质区域的微病变相关,其特征是神经元核蛋白染色减少和小胶质细胞浸润增加。基于这些发现,我们选择调查微病变内的其他神经元群体,特别是抑制性中间神经元。此外,我们假设加标足以在大鼠皮质内诱导类似的细胞结构变化,并且抑制MAPK信号传导,使用MAP2K抑制剂,不仅会抑制尖峰形成,还会减少这些细胞结构变化并改善行为结果。为了测试这些假设,我们分析了16例需要皮质切除的难治性癫痫患者的组织样本.我们还利用了破伤风毒素诱导的发作间尖峰动物模型,旨在在雄性Sprague-Dawley大鼠中产生没有癫痫发作的尖峰。大鼠装有硬膜外电极,为了在研究期间记录脑电图,并实施自动算法来量化尖峰。六个月后,处死动物以评估慢性加标对皮质细胞结构的影响.这里,我们表明,微病变可能会促进兴奋性,这是由于抑制性神经元的显着减少,这可能是促进浅层发作间尖峰的原因。同样,我们发现在大鼠模型中诱发癫痫尖峰产生类似的变化,包括减少的神经元核蛋白,钙结合蛋白和小白蛋白阳性神经元和增加的小胶质细胞,表明尖峰足以诱导人类的这些细胞结构变化。最后,我们认为MAPK信号传导是产生这些病理变化的驱动力。使用CI-1040抑制MAP2K,无论是急性还是在尖峰出现后,导致更少的间期尖峰,减少小胶质细胞活化和减少抑制性神经元损失。处理过的动物的高振幅明显较少,短期尖峰,这与巴恩斯迷宫上改进的空间记忆性能相关。一起,我们的结果为癫痫皮质潜在的细胞结构发病机制提供了证据,可以通过早期和延迟的MAP2K抑制来改善。这些发现强调了CI-1040作为药物治疗的潜在作用,可以预防癫痫患者和非癫痫性尖峰相关神经行为障碍患者的癫痫活动发展并减少认知障碍。
    Interictal spikes are electroencephalographic discharges that occur at or near brain regions that produce epileptic seizures. While their role in generating seizures is not well understood, spikes have profound effects on cognition and behaviour, depending on where and when they occur. We previously demonstrated that spiking areas of human neocortex show sustained MAPK activation in superficial cortical Layers I-III and are associated with microlesions in deeper cortical areas characterized by reduced neuronal nuclear protein staining and increased microglial infiltration. Based on these findings, we chose to investigate additional neuronal populations within microlesions, specifically inhibitory interneurons. Additionally, we hypothesized that spiking would be sufficient to induce similar cytoarchitectonic changes within the rat cortex and that inhibition of MAPK signalling, using a MAP2K inhibitor, would not only inhibit spike formation but also reduce these cytoarchitectonic changes and improve behavioural outcomes. To test these hypotheses, we analysed tissue samples from 16 patients with intractable epilepsy who required cortical resections. We also utilized a tetanus toxin-induced animal model of interictal spiking, designed to produce spikes without seizures in male Sprague-Dawley rats. Rats were fitted with epidural electrodes, to permit EEG recording for the duration of the study, and automated algorithms were implemented to quantify spikes. After 6 months, animals were sacrificed to assess the effects of chronic spiking on cortical cytoarchitecture. Here, we show that microlesions may promote excitability due to a significant reduction of inhibitory neurons that could be responsible for promoting interictal spikes in superficial layers. Similarly, we found that the induction of epileptic spikes in the rat model produced analogous changes, including reduced neuronal nuclear protein, calbindin and parvalbumin-positive neurons and increased microglia, suggesting that spikes are sufficient for inducing these cytoarchitectonic changes in humans. Finally, we implicated MAPK signalling as a driving force producing these pathological changes. Using CI-1040 to inhibit MAP2K, both acutely and after spikes developed, resulting in fewer interictal spikes, reduced microglial activation and less inhibitory neuron loss. Treated animals had significantly fewer high-amplitude, short-duration spikes, which correlated with improved spatial memory performance on the Barnes maze. Together, our results provide evidence for a cytoarchitectonic pathogenesis underlying epileptic cortex, which can be ameliorated through both early and delayed MAP2K inhibition. These findings highlight the potential role for CI-1040 as a pharmacological treatment that could prevent the development of epileptic activity and reduce cognitive impairment in both patients with epilepsy and those with non-epileptic spike-associated neurobehavioural disorders.
    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
    KRAS是人类胃癌中常见的突变癌基因,常与耐药和不良预后相关。在KRAS突变的癌症中组合MEK-CDK4/6抑制的联合临床试验证明了在患者来源的异种移植物中的治疗功效和在患者中的安全性。这里,目前的研究主要集中在靶向CDK4/6和MEK在体外和体内协同阻断KRAS突变胃癌细胞的增殖,并通过AMPK/mTOR通路诱导自噬。此外,自噬抑制剂联合靶向CDK4/6和MEK治疗对KRAS突变胃癌细胞具有显著的抗肿瘤作用。需要临床试验来确定这一发现背后的机制及其临床效用。总之,我们的结果表明,联合靶向MEK和CDK4/6的自噬抑制剂同时阻断多个代谢过程,可能是胃癌的有效治疗方法.
    KRAS is a commonly mutated oncogene in human gastric cancer and is often associated with drug resistance and poor prognosis. Co-clinical trial of combined MEK-CDK4/6 inhibition in KRAS mutated cancers demonstrated therapeutic efficacy in patient-derived xenografts and safety in patients. Here, present research focuses on targeting CDK4/6 and MEK synergistically block the proliferation of KRAS-mutated gastric cancer cells in vitro and in vivo and induced autophagy through the AMPK/mTOR pathway. Furthermore, autophagy inhibitor combined with targeting CDK4/6 and MEK therapy had significant antitumor effects on KRAS mutant gastric cancer cells. Clinical trials are needed to determine the mechanism behind this finding and its clinical utility. In conclusion, our results demonstrate autophagy inhibitor combined targeting MEK and CDK4/6 that concurrently block multiple metabolic processes may be an effective therapeutic approach for gastric cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号