c-Met

C - MET
  • 文章类型: Journal Article
    Vater壶腹腺癌(AAC)是一种罕见的恶性肿瘤,具有由各种组织学亚型引起的异质性肿瘤。需要新的治疗策略。本研究检查了表皮生长因子受体(EGFR),人表皮生长因子受体2(HER2),和c-Met在AAC中的表达,考虑到他们作为药物目标的潜力。在87例接受根治性切除术的患者中,EGFR过度表达占87.4%,HER2为11.5%,和c-Met在50%。EGFR过表达在胰胆管亚型中更为常见(p=0.018),并且与较高的组织学分级有关(p=0.008)。HER2与临床病理特征无关,而c-Met在淋巴结阴性组中更为常见(p=0.004),并且通常与EGFR共表达(p=0.049)。EGFR阳性患者的无病(HR=2.89;95%CI,1.35-6.20;p=0.061)和总生存率(HR=6.89;95%CI,2.94-16.2;p=0.026)均低于EGFR阴性患者。HER2阳性AAC显示出生存期缩短的趋势,虽然没有统计学意义,c-Met对生存结局无影响.在全身性疾病的背景下,生存结果没有根据EGFR而变化,HER2和c-Met表达,但HER2阳性组的无进展生存期有下降趋势(HR=1.90;95%CI,0.56-6.41;p=0.166).这项研究强调了EGFR的潜力,HER2和c-Met作为AAC个性化治疗的目标,需要进一步研究以评估靶向治疗。
    Adenocarcinoma of the ampulla of Vater (AAC) is a rare malignancy with heterogeneous tumors arising from various histologic subtypes, necessitating new therapeutic strategies. This study examines epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), and c-Met expression in AAC, given their potential as druggable targets. Among 87 patients who underwent curative resection, EGFR overexpression was found in 87.4%, HER2 in 11.5%, and c-Met in 50%. EGFR overexpression was more common in the pancreatobiliary subtype (p = 0.018) and associated with a higher histologic grade (p = 0.008). HER2 did not correlate with clinicopathological features, while c-Met was more common in node-negative groups (p = 0.004) and often co-expressed with EGFR (p = 0.049). EGFR-positive patients had worse disease-free (HR = 2.89; 95% CI, 1.35-6.20; p = 0.061) and overall survival (HR = 6.89; 95% CI, 2.94-16.2; p = 0.026) than EGFR-negative patients. HER2-positive AAC showed a trend towards shorter survival, although not statistically significant, and c-Met had no impact on survival outcomes. In the context of systemic disease, survival outcomes did not vary according to EGFR, HER2, and c-Met expression, but the HER2-positive group showed a trend towards inferior progression-free survival (HR = 1.90; 95% CI, 0.56-6.41; p = 0.166). This study underscores the potential of EGFR, HER2, and c-Met as targets for personalized therapy in AAC, warranting further research to evaluate targeted treatments.
    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
    癌症的发展和生长从根本上取决于代谢中的促肿瘤变化。癌细胞通常从氧化磷酸化转变为主要能量来源,并更严重地依赖糖酵解。受体酪氨酸激酶(RTKs)是一种类型的受体,其与向促肿瘤代谢的这种转变有关。RTK是癌症生长和转移的重要驱动因素。一个这样的RTK家族是MET家族,由MET和RON(MST1R)组成。在多种肿瘤类型中,MET或RON的过表达与较差的癌症患者预后相关。MET和RON信号通过增加MYC转录因子活性上调关键糖酵解酶的表达来促进糖酵解增加。此外,MET和RON信号通过上调SREBP2诱导的胆固醇生物合成酶的表达,促进糖酵解下游胆固醇生物合成的增加。这些新陈代谢的变化,由RTK活动驱动,通过药理学抑制或饮食修饰,为限制肿瘤生长和转移提供潜在的靶标。本文综述了由RTK的MET家族驱动的促肿瘤代谢变化。在这样做的时候,我们将就导致患者预后恶化的代谢途径提供我们独特的观点,并为未来的研究提供建议。
    The development and growth of cancer is fundamentally dependent on pro-tumor changes in metabolism. Cancer cells generally shift away from oxidative phosphorylation as the primary source of energy and rely more heavily on glycolysis. Receptor tyrosine kinases (RTKs) are a type of receptor that is implicated in this shift to pro-tumor metabolism. RTKs are important drivers of cancer growth and metastasis. One such family of RTKs is the MET family, which consists of MET and RON (MST1R). The overexpression of either MET or RON has been associated with worse cancer patient prognosis in a variety of tumor types. Both MET and RON signaling promote increased glycolysis by upregulating the expression of key glycolytic enzymes via increased MYC transcription factor activity. Additionally, both MET and RON signaling promote increased cholesterol biosynthesis downstream of glycolysis by upregulating the expression of SREBP2-induced cholesterol biosynthesis enzymes via CTTNB1. These changes in metabolism, driven by RTK activity, provide potential targets in limiting tumor growth and metastasis via pharmacological inhibition or modifications in diet. This review summarizes pro-tumor changes in metabolism driven by the MET family of RTKs. In doing so, we will offer our unique perspective on metabolic pathways that drive worse patient prognosis and provide suggestions for future study.
    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
    肝细胞生长因子(HGF)在多个系统中表达,介导多种生物学活性,如有丝分裂,运动性,和形态发生。越来越多的研究揭示了HGF在从产前到成年期的卵巢和睾丸生理中的表达模式和功能。HGF通过调节卵巢中卵泡膜细胞和颗粒细胞的功能来调节卵泡生成和类固醇生成。它还介导体细胞增殖和类固醇生成,从而影响男性的精子发生。除了对生殖系统的生理影响外,近年来,HGF在治疗男性和女性不育症的临床前研究中显示出优势。特别是在妇女与卵巢早衰。本文旨在总结HGF在生殖系统中的多效性功能,为其临床应用提供前景。
    Hepatocyte growth factor (HGF) is expressed in multiple systems and mediates a variety of biological activities, such as mitosis, motility, and morphogenesis. A growing number of studies have revealed the expression patterns and functions of HGF in ovarian and testicular physiology from the prenatal to the adult stage. HGF regulates folliculogenesis and steroidogenesis by modulating the functions of theca cells and granulosa cells in the ovary. It also mediates somatic cell proliferation and steroidogenesis, thereby affecting spermatogenesis in males. In addition to its physiological effects on the reproductive system, HGF has shown advantages in preclinical studies over recent years for the treatment of male and female infertility, particularly in women with premature ovarian insufficiency. This review aims to summarize the pleiotropic functions of HGF in the reproductive system and to provide prospects for its clinical application.
    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
    作为空气交换的场所,肺组织长期暴露于大量的外来病原体。因此,肺已经发展出精致而复杂的免疫系统。除了它们的物理和化学屏障作用,肺上皮细胞可通过表达Toll样受体(TLRs)和其他模式识别受体,随着细胞因子的分泌。新的证据表明,肺上皮细胞可以产生和分泌免疫球蛋白(Ig),包括IgM,IgA,或IgG,从而执行抗体功能。此外,已经发现恶性转化的肺上皮细胞产生高水平的Ig,主要是IgG,不能实现抗体的作用,而是进行促进肿瘤的活动。结构分析表明,由于独特的糖基化修饰,肺癌细胞产生的IgG的生物学活性与正常肺上皮细胞产生的Ig的生物学活性不同。具体来说,唾液酸化IgG(SIA-IgG),以在IgyCH1的Asn162位点的非传统N-糖基化修饰为特征,在肿瘤干细胞中高度表达。已经证明SIA-IgG依赖于这种独特的唾液酸化修饰来促进肿瘤发生。转移,和免疫逃避。目前的研究结果证明,肺上皮细胞产生的Ig具有多方面的生物学活性,包括生理条件下的免疫防御功能,同时在恶性转化过程中获得促进肿瘤的活性。这些见解作为新的生物标志物和靶标,具有诊断和治疗肺癌的潜力。
    As the locus for air exchange, lung tissue is perpetually exposed to a significant quantity of foreign pathogens. Consequently, lung has developed a refined and intricate immune system. Beyond their physical and chemical barrier roles, lung epithelial cells can contribute to immune defence through the expression of Toll-like receptors (TLRs) and other pattern recognition receptors, along with the secretion of cytokines. Emerging evidence demonstrates that lung epithelial cells can generate and secrete immunoglobulins (Igs), including IgM, IgA, or IgG, thus performing antibody function. Moreover, malignantly transformed lung epithelial cells have been discovered to produce high levels of Ig, predominantly IgG, which do not fulfill the role of antibodies, but instead carries out tumour-promoting activity. Structural analysis has indicated that the biological activity of IgG produced by lung cancer cells differs from that of Igs produced by normal lung epithelial cells due to the unique glycosylation modification. Specifically, the sialylated IgG (SIA-IgG), characterised by a non-traditional N-glycosylation modification at the Asn162 site of Igγ CH1, is highly expressed in tumour stem cells. It has been demonstrated that SIA-IgG relies on this unique sialylation modification to promote tumorigenesis, metastasis, and immune evasion. Current results have proven that the Ig produced by lung epithelial cells has multifaceted biological activities, including immune defence functions under physiological conditions, while acquiring tumour-promoting activity during malignant transformation. These insights possess potential for the diagnosis and treatment of lung cancer as novel biomarkers and targets.
    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
    胃癌和胃食管连接部癌是全球肿瘤相关死亡的主要原因。尽管免疫治疗和分子靶向治疗的进展扩大了治疗选择,对于不可切除或转移性胃癌患者,他们的预后没有显著改变。少数患者,特别是那些PD-L1阳性的,HER-2阳性,或MSI高肿瘤,可能从晚期阶段的免疫检查点抑制剂和/或HER-2定向疗法中受益更多。然而,对于那些缺乏特定靶标和独特分子特征的人,常规化疗仍然是唯一推荐的有效和持久的治疗方案.在这次审查中,我们总结了各种信号通路的作用,并进一步研究了可用的靶标。然后,晚期胃癌II/III期临床试验的当前结果,随着现有生物标志物的优势和局限性,具体讨论。最后,当遇到重大挑战时,我们将提供我们对精准治疗模式的见解。
    Gastric cancer and gastroesophageal junction cancer represent the leading cause of tumor-related death worldwide. Although advances in immunotherapy and molecular targeted therapy have expanded treatment options, they have not significantly altered the prognosis for patients with unresectable or metastatic gastric cancer. A minority of patients, particularly those with PD-L1-positive, HER-2-positive, or MSI-high tumors, may benefit more from immune checkpoint inhibitors and/or HER-2-directed therapies in advanced stages. However, for those lacking specific targets and unique molecular features, conventional chemotherapy remains the only recommended effective and durable regimen. In this review, we summarize the roles of various signaling pathways and further investigate the available targets. Then, the current results of phase II/III clinical trials in advanced gastric cancer, along with the superiorities and limitations of the existing biomarkers, are specifically discussed. Finally, we will offer our insights in precision treatment pattern when encountering the substantial challenges.
    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
    分子工程能够创造具有新功能的适体,但前提是对其结构和识别机制的深刻理解。由于c-MET/HGF信号通路在肿瘤发展和侵袭中的关键作用,细胞间充质上皮转化因子(c-MET)引起了广泛关注。这项研究报道了构建与c-MET和其他特定蛋白质结合的新型嵌合适体的策略。c-MET被鉴定为DNA适体的分子靶标,HF3-58,通过细胞-SELEX选择。系统研究了HF3-58与c-MET的结合结构和机理,露出脚手架,认可,和冗余区域。通过分子工程设计,冗余区域被其他具有茎环结构的适体取代,产生新的嵌合适体,具有双特异性结合c-MET和特异性蛋白。嵌合双特异性适体HF-3b显示出介导T细胞与肿瘤细胞粘附的能力,提示在肿瘤免疫治疗中的前瞻性用途。这些发现表明适体HF3-58可以作为分子工程平台用于开发靶向c-MET的多种多功能配体。此外,全面了解适体的结合机制将为功能性适体的设计提供指导,显著扩大其潜在的应用。
    Molecular engineering enables the creation of aptamers with novel functions, but the prerequisite is a deep understanding of their structure and recognition mechanism. The cellular-mesenchymal epithelial transition factor (c-MET) is garnering significant attention due to the critical role of the c-MET/HGF signaling pathway in tumor development and invasion. This study reports a strategy for constructing novel chimeric aptamers that bind to both c-MET and other specific proteins. c-MET was identified to be the molecular target of a DNA aptamer, HF3-58, selected through cell-SELEX. The binding structure and mechanism of HF3-58 with c-MET were systematically studied, revealing the scaffold, recognition, and redundancy regions. Through molecular engineering design, the redundancy region was replaced with other aptamers possessing stem-loop structures, yielding novel chimeric aptamers with bispecificity for binding to c-MET and specific proteins. A chimeric bispecific aptamer HF-3b showed the ability to mediate the adhesion of T-cells to tumor cells, suggesting the prospective utility in tumor immunotherapy. These findings suggest that aptamer HF3-58 can serve as a molecular engineering platform for the development of diverse multifunctional ligands targeting c-MET. Moreover, comprehensive understanding of the binding mechanisms of aptamers will provide guidance for the design of functional aptamers, significantly expanding their potential applications.
    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:本研究的目的是通过虚拟筛选鉴定抗EGFR/c-Met的双靶点抑制剂,动态仿真,和生物活性评价。这一努力旨在克服由L858R/T790M突变体诱导的药物抗性的挑战。
    方法:收集活性结构以构建药物分子组。接下来,将性质过滤应用于化合物库中的药物结构。然后通过虚拟筛选和聚类分析鉴定活性化合物。随后,我们对活性化合物进行了MTT抗肿瘤活性评估和激酶抑制试验,以确定最有希望的候选化合物.此外,对所选化合物进行AO染色和JC-1测定。最终,优选化合物与EGFR和c-Met蛋白进行分子对接和分子动力学模拟,分别。
    结果:T13074的IC50对于EGFRL858R/T790M激酶为2.446μM,对于c-Met激酶为7.401nM,强调其在克服EGFRL858R/T790M抗性方面的潜力。此外,T13074对H1975细胞的IC50为1.93μM。AO染色和JC-1测定的结果表明T13074以浓度依赖性方式诱导肿瘤细胞凋亡。值得注意的是,发现T13074与EGFR蛋白之间的结合能为-90.329±16.680kJ/mol,与c-Met蛋白的结合能为-139.935±17.414kJ/mol。
    结论:T13074在体内和体外均表现出优异的抗肿瘤活性,表明其作为双靶点EGFR/c-Met抑制剂的潜在效用。这表明其在克服由L858R/T790M突变诱导的EGFR抗性中的有希望的作用。
    OBJECTIVE: The objective of this study is to identify dual-target inhibitors against EGFR/c-Met through virtual screening, dynamic simulation, and biological activity evaluation. This endeavor is aimed at overcoming the challenge of drug resistance induced by L858R/T790M mutants.
    METHODS: Active structures were gathered to construct sets of drug molecules. Next, property filtering was applied to the drug structures within the compound library. Active compounds were then identified through virtual screening and cluster analysis. Subsequently, we conducted MTT antitumor activity evaluation and kinase inhibition assays for the active compounds to identify the most promising candidates. Furthermore, AO staining and JC-1 assays were performed on the selected compounds. Ultimately, the preferred compounds underwent molecular docking and molecular dynamics simulation with the EGFR and c-Met proteins, respectively.
    RESULTS: The IC50 of T13074 was determined as 2.446 μM for EGFRL858R/T790M kinase and 7.401 nM for c-Met kinase, underscoring its potential in overcoming EGFRL858R/T790M resistance. Additionally, T13074 exhibited an IC50 of 1.93 μM on the H1975 cell. Results from AO staining and JC-1 assays indicated that T13074 induced tumor cell apoptosis in a concentration-dependent manner. Notably, the binding energy between T13074 and EGFR protein was found to be -90.329 ± 16.680 kJ/mol, while the binding energy with c-Met protein was -139.935 ± 17.414 kJ/mol.
    CONCLUSIONS: T13074 exhibited outstanding antitumor activity both in vivo and in vitro, indicating its potential utility as a dual-target EGFR/c-Met inhibitor. This suggests its promising role in overcoming EGFR resistance induced by the L858R/T790M mutation.
    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:肺纤维化(PF)是几种与高发病率和死亡率相关的间质性肺病(ILD)的病理终末期。然而,目前的治疗只能延缓疾病进展,而不是提供治愈。炎症在PF进展中的作用是公认的,但是对免疫调节的新见解对于开发更有效的疗法至关重要。c-MET信号传导与免疫细胞的迁移能力和效应子功能有关。然而,该信号通路在PF相关肺部疾病中的作用仍未被研究.
    方法:为了确定免疫细胞中c-MET在肺纤维化进展中的影响,我们在免疫细胞中使用了c-Met的条件性缺失。为了诱导肺纤维化,对小鼠气管内施用博来霉素(BLM)。在21天的过程中,评估小鼠的体重变化,在不同时间点安乐死后,评估支气管肺泡灌洗液细胞和肺组织的炎症和纤维化。此外,在冷冻活检切片中评估c-MET表达,支气管肺泡灌洗液细胞样本和单细胞RNA测序数据集来自不同间质性肺疾病的人类患者。
    结果:c-MET在肺免疫细胞中被诱导表达,特别是在T细胞中,间质巨噬细胞,和中性粒细胞,在BLM诱导的PF小鼠模型的炎症阶段。免疫细胞中c-Met的缺失与BLM处理的小鼠的早期体重恢复和改善的存活率相关。此外,免疫细胞中c-Met的缺失与免疫细胞群的早期募集有关,通常被发现表达c-MET,导致随后细胞毒性和促炎环境的减弱。因此,较不广泛的炎症反应,可能伴随着组织修复,最终导致纤维化病变程度较低。此外,c-MET表达在纤维化ILD患者的肺T细胞中上调,提示c-MET可能参与纤维化疾病的发展。
    结论:这些结果强调了免疫细胞中c-MET信号传导对其增强的不受控制的募集和对促炎和促纤维化表型的激活的关键贡献,导致肺损伤的加重和随之而来的纤维化的发展。
    BACKGROUND: Pulmonary fibrosis (PF) represents the pathologic end stage of several interstitial lung diseases (ILDs) associated with high morbidity and mortality rates. However, current treatments can only delay disease progression rather than provide a cure. The role of inflammation in PF progression is well-established, but new insights into immune regulation are fundamental for developing more efficient therapies. c-MET signaling has been implicated in the migratory capacity and effector functions of immune cells. Nevertheless, the role of this signaling pathway in the context of PF-associated lung diseases remains unexplored.
    METHODS: To determine the influence of c-MET in immune cells in the progression of pulmonary fibrosis, we used a conditional deletion of c-Met in immune cells. To induce pulmonary fibrosis mice were administered with bleomycin (BLM) intratracheally. Over the course of 21 days, mice were assessed for weight change, and after euthanasia at different timepoints, bronchoalveolar lavage fluid cells and lung tissue were assessed for inflammation and fibrosis. Furthermore, c-MET expression was assessed in cryobiopsy sections, bronchoalveolar lavage fluid cells samples and single cell RNA-sequencing dataset from human patients with distinct interstitial lung diseases.
    RESULTS: c-MET expression was induced in lung immune cells, specifically in T cells, interstitial macrophages, and neutrophils, during the inflammatory phase of BLM-induced PF mouse model. Deletion of c-Met in immune cells correlated with earlier weight recovery and improved survival of BLM-treated mice. Moreover, the deletion of c-Met in immune cells was associated with early recruitment of the immune cell populations, normally found to express c-MET, leading to a subsequent attenuation of the cytotoxic and proinflammatory environment. Consequently, the less extensive inflammatory response, possibly coupled with tissue repair, culminated in less exacerbated fibrotic lesions. Furthermore, c-MET expression was up-regulated in lung T cells from patients with fibrosing ILD, suggesting a potential involvement of c-MET in the development of fibrosing disease.
    CONCLUSIONS: These results highlight the critical contribution of c-MET signaling in immune cells to their enhanced uncontrolled recruitment and activation toward a proinflammatory and profibrotic phenotype, leading to the exacerbation of lung injury and consequent development of fibrosis.
    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
    间充质上皮转化因子(c-Met)是一种与细胞增殖相关的受体酪氨酸激酶,生存,入侵,以及几种癌症的转移,包括结直肠癌(CRC),特别是在异常激活时。我们的研究策略性地设计了源自c-Met和抗体Onartuzumab之间相互作用的肽。通过利用循环策略,我们实现了显著增强的肽稳定性和亲和力。我们的体外评估证实,与线性对应物相比,环状肽HYNIC-cycOn表现出更高的亲和力(KD=83.5nM)和更高的特异性。通过体内实验,[99mTc]Tc-HYNIC-cycOn在非肿瘤细胞中显示出优异的肿瘤靶向能力和最小的吸收,如单光子发射计算机断层扫描所证实。值得注意的是,肿瘤与肌肉和肿瘤与肠道的比例,注射后1小时,分别为4.78±0.86和3.24±0.47。在原位CRC模型中观察到相当的比率,记录分别为4.94±0.32和3.88±0.41。总之,[99mTc]Tc-HYNIC-cycOn显示出作为临床应用的候选物的实质性希望。我们表明[99mTc]Tc-HYNIC-cycOn可以有效地靶向和观察体内表达c-Met的肿瘤,为在CRC中检测c-Met时提高诊断准确性提供了一种有前途的方法。
    The mesenchymal-epithelial transition factor (c-Met) is a receptor tyrosine kinase linked to the proliferation, survival, invasion, and metastasis of several types of cancers, including colorectal cancer (CRC), particularly when aberrantly activated. Our study strategically designs peptides derived from interactions between c-Met and the antibody Onartuzumab. By utilizing a cyclic strategy, we achieved significantly enhanced peptide stability and affinity. Our in vitro assessments confirmed that the cyclic peptide HYNIC-cycOn exhibited a higher affinity (KD = 83.5 nM) and greater specificity compared with its linear counterpart. Through in vivo experiments, [99mTc]Tc-HYNIC-cycOn displayed exceptional tumor-targeting capabilities and minimal absorption in nontumor cells, as confirmed by single-photon emission computed tomography. Notably, the ratios of tumor to muscle and tumor to intestine, 1 h postinjection, were 4.78 ± 0.86 and 3.24 ± 0.47, respectively. Comparable ratios were observed in orthotopic CRC models, recording 4.94 ± 0.32 and 3.88 ± 0.41, respectively. In summary, [99mTc]Tc-HYNIC-cycOn shows substantial promise as a candidate for clinical applications. We show that [99mTc]Tc-HYNIC-cycOn can effectively target and visualize c-Met-expressing tumors in vivo, providing a promising approach for enhancing diagnostic accuracy when detecting c-Met in CRC.
    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
    转移性胃腺癌(mGAC)的预后仍然较差。受体酪氨酸激酶(RTK)如表皮生长因子受体(EGFR)及其下游效应物(包括磷脂酰肌醇3-激酶(PIK3CA)的催化亚基α)的基因改变在mGAC中很常见。靶向RTK和磷脂酰肌醇-3-激酶(PI3K)治疗已经证明在其他实体瘤中的临床益处,并且考虑到这些途径中存在复发性改变,是针对mGAC的临床开发的关键潜在靶标。此外,RTK/PI3K联合治疗可以克服使用单一疗法出现的代偿机制,改善患者预后。在这里,我们研究了针对我们独特的人类mGAC衍生的PIK3CA功能获得突变体的RTK/PI3K单一和组合药物反应,人表皮生长因子受体2(HER2)阴性,表达EGFR的循环肿瘤细胞系,UWG02CTC,在二维和三维培养条件下模拟转移的不同阶段。UWG02CTC对PI3Kp110α亚基靶向药物PIK-75(IC50=37.0±11.1nM)或alpelisib(7.05±3.7µM)高度响应。药物敏感性在3D条件下显著增加。与EGFR抑制剂吉非替尼联合治疗克服了PI3K/Akt抑制引起的代偿性MAPK/ERK通路上调,这是强烈的协同作用。在器官型分析中,PIK-75加吉非替尼显着损害了UWG02CTC的侵袭。总之,UWG02CTCs是一种强大的离体mGAC药物反应模型,揭示了EGFR/PI3K靶向药物作为HER2阴性的有希望的联合治疗选择。RAS野生型mGAC患者。
    The prognosis for metastatic gastric adenocarcinoma (mGAC) remains poor. Gene alterations in receptor tyrosine kinases (RTKs) such as epidermal growth factor receptor (EGFR) and their downstream effectors including catalytic subunit alpha of the phosphatidylinositol 3-kinase (PIK3CA) are common in mGAC. Targeted RTK and phosphatidylinositol-3-kinase (PI3K) treatments have demonstrated clinical benefits in other solid tumours and are key potential targets for clinical development against mGAC given the presence of recurrent alterations in these pathways. Furthermore, combination RTK/PI3K treatments may overcome compensatory mechanisms that arise using monotherapies, leading to improved patient outcomes. Herein, we investigated RTK/PI3K single and combination drug responses against our unique human mGAC-derived PIK3CA gain-of-function mutant, human epidermal growth factor receptor 2 (HER2)-negative, EGFR-expressing circulating tumour cell line, UWG02CTC, under two- and three-dimensional culture conditions to model different stages of metastasis. UWG02CTCs were highly responsive to the PI3K p110α-subunit targeted drugs PIK-75 (IC50 = 37.0 ± 11.1 nM) or alpelisib (7.05 ± 3.7 µM). Drug sensitivities were significantly increased in 3D conditions. Compensatory MAPK/ERK pathway upregulation by PI3K/Akt suppression was overcome by combination treatment with the EGFR inhibitor gefitinib, which was strongly synergistic. PIK-75 plus gefitinib significantly impaired UWG02CTC invasion in an organotypic assay. In conclusion, UWG02CTCs are a powerful ex vivo mGAC drug responsiveness model revealing EGFR/PI3K-targeted drugs as a promising combination treatment option for HER2-negative, RAS wild-type mGAC patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号