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
    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
    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
    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)

  • 文章类型: Journal Article
    胰腺导管腺癌(PDAC)恶性程度极高,进展迅速。PDAC对当前治疗方法的总体响应率仍然不令人满意。因此,确定新的靶标并阐明与PDAC进展相关的潜在机制可能会提供额外的治疗策略.AHNAK2在多种肿瘤中异常表达并发挥促肿瘤发生作用。然而,AHNAK2在PDAC中的生物学作用尚不清楚。
    通过免疫组织化学(IHC)和定量实时聚合酶链反应(qRT-PCR)检测PDAC和配对的非肿瘤组织中AHNAK2的表达。进行慢病毒敲除以研究AHNAK2对胰腺癌细胞生物学功能的影响。采用皮下细胞来源的异种移植(CDX)模型和AHNAK2沉默的KPC自发小鼠模型,观察AHNAK2对肿瘤生长和预后的影响。使用蛋白质印迹评估响应于HGF处理的蛋白水平的c-MET的表达。
    我们的结果表明,AHNAK2在PDAC临床样本中高表达,并与不良预后相关。敲除能显著抑制AHNAK2的增殖,迁移,和胰腺癌细胞的侵袭。AHNAK2敲低或敲除导致具有PDAC的小鼠中的肿瘤生长抑制和延长的存活。此外,AHNAK2和c-MET表达水平在转录后水平显示出显著的正相关。机械上,AHNAK2通过阻止c-MET降解和持续激活HGF/c-MET信号通路促进肿瘤进展。
    总的来说,我们的研究表明,AHNAK2通过调节c-MET信号通路在PDAC进展中发挥重要作用,靶向AHNAK2可能是PDAC的有效治疗策略。
    UNASSIGNED: Pancreatic ductal adenocarcinoma (PDAC) is extremely malignant and rapidly progresses. The overall response rate of PDAC to current treatment methods is still unsatisfactory. Thus, identifying novel targets and clarifying the underlying mechanisms associated with PDAC progression may potentially offer additional treatment strategies. AHNAK2 is aberrantly expressed in a variety of tumors and exerts pro-tumorigenic effects. However, the biological role of AHNAK2 in PDAC remains poorly understood.
    UNASSIGNED: The expression of AHNAK2 in PDAC and paired non-tumor tissues was detected by immunohistochemistry (IHC) and quantitative real-time polymerase chain reaction (qRT-PCR). Lentivirus knockdown was performed to investigate the impact of AHNAK2 on the biological function of pancreatic cancer cells. The subcutaneous cell-derived xenograft (CDX) model and the KPC spontaneous mouse model with AHNAK2 silencing were used to observe the effects of AHNAK2 on tumor growth and prognosis. The expression of c-MET at protein level in response to HGF treatment was assessed using western blot.
    UNASSIGNED: Our results demonstrated that AHNAK2 was highly expressed in PDAC clinical samples and associated with poor prognosis. Knockdown of AHNAK2 significantly inhibited the proliferation, migration, and invasion of pancreatic cancer cells. AHNAK2 knockdown or knockout resulted in tumor growth suppression and prolonged survival in mice with PDAC. In addition, AHNAK2 and c-MET expression levels showed a significant positive correlation at the post-transcriptional level. Mechanistically, AHNAK2 promoted tumor progression by preventing c-MET degradation and persistently activating the HGF/c-MET signaling pathway.
    UNASSIGNED: Overall, our study revealed that AHNAK2 plays an important role in PDAC progression by modulating the c-MET signaling pathway, and targeting AHNAK2 may be an effective therapeutic strategy for 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
    胆囊起源的神经内分泌癌(NEC)特别罕见,仅占原发性胆囊恶性肿瘤的0.38%,标准疗法是有限的。MET基因编码酪氨酸激酶受体,C-Met.MET的致病变异,如MET外显子14跳跃和MET扩增,导致过度的下游信号,促进肿瘤进展。一种MET抑制剂,卡马替尼,阻断c-Met的信号传导,并已被食品和药物管理局批准用于具有MET外显子14跳跃的非小细胞肺癌。据报道,卡马替尼在其他具有MET扩增的癌症中的有效性,但尚未报道具有MET变体的NEC。这里,我们介绍了一个72岁的女性胆囊的NEC与多个肝脏和淋巴结转移,对常规化疗耐药,包括卡铂加依托泊苷作为一线治疗和伊立替康作为二线治疗,但她对卡马替尼有反应.治疗6周后,CT扫描显示部分反应(尺寸减少80%),但13周后,观察到肝转移的再生长。在这里,我们报告了卡马替尼对MET扩增的胆囊源性NEC患者的有意义疗效.
    Neuroendocrine carcinoma (NEC) of the gallbladder origin is particularly rare, accounting for only 0.38% of primary malignancies of the gallbladder, and standard therapies are limited. The MET gene encodes the tyrosine kinase receptor, c-Met. Pathogenic variants of MET, such as MET exon 14 skipping and MET amplification, result in excessive downstream signaling that promotes tumor progression. A MET inhibitor, capmatinib, blocks signaling of c-Met and has been approved by the Food and Drug Administration for non-small cell lung cancer with MET exon 14 skipping. The effectiveness of capmatinib has been reported in other cancers with MET amplification, but NEC with MET variants has not been reported. Here, we present a case of a 72-year-old woman with NEC of the gallbladder with multiple liver and lymph node metastases, who was resistant to conventional chemotherapy including carboplatin plus etoposide as first-line treatment and irinotecan as second-line treatment, but she responded to capmatinib. After 6 weeks of treatment, CT scan showed a partial response (80% reduction in size), but after 13 weeks, regrowth of liver metastasis was observed. Herein, we report a meaningful efficacy of capmatinib to the patient of NEC of the gallbladder origin with MET amplification.
    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
    间变性淋巴瘤激酶(ALK)靶向酪氨酸激酶抑制剂(TKIs)可改善患者生存率;然而,一些患者出现ALK-TKI耐药的机制不明.我们调查了ALK阳性非小细胞肺癌(NSCLC)患者的ErbB家族和c-MET表达,以了解它们在ALK-TKI反应中的作用。
    我们研究了72例晚期ALK阳性非小细胞肺癌患者的EML4-ALK融合变异亚型和c-MET免疫染色,EGFR,组织标本上的HER2和HER3在用ALK-TKI治疗前(初次)和后(二次)。我们研究了它们的表达与生存结果的关系,并评估了ALK和EGFR抑制剂在HER3特异性配体HRG1刺激的ALK阳性NSCLC细胞系中的有效性。
    c-MET的高表达,EGFR,HER2和HER3在4.9%中观察到,18.0%,1.6%,和25.8%的原发性肿瘤,分别,和18.5%,37.0%,10.7%,和35.7%的继发性肿瘤,分别。原发性肿瘤中的HER3过表达显示较差的生存率(P=0.132)。在具有EML4-ALK变体1/2(V1/V2)的亚组中,HER3过表达与原发性和继发性肿瘤的低生存率显著相关(分别为P=0.022和P=0.004)。氯拉替尼和厄洛替尼的联合治疗显著降低了ALK阳性NSCLC细胞中HRG1诱导的RTK信号激活。
    HER3过表达可能作为ALK阳性NSCLC的预后标志物,包括ALK-TKI幼稚和治疗病例,特别是那些与EML4-ALKV1/V2。评估HER3表达对于这些患者的治疗计划和结果预测可能至关重要。
    UNASSIGNED: Anaplastic lymphoma kinase (ALK)-targeted tyrosine kinase inhibitors (TKIs) improve patient survival; however, some patients develop ALK-TKI resistance with unidentified mechanisms. We investigated ErbB family and c-MET expression in patients with ALK-positive non-small cell lung cancer (NSCLC) to understand their roles in the ALK-TKI response.
    UNASSIGNED: We studied 72 patients with advanced ALK-positive NSCLC with EML4-ALK fusion variant subtyping and immunostaining for c-MET, EGFR, HER2, and HER3 on tissue specimens both pre- (primary) and post-treatment (secondary) with ALK-TKI. We investigated the association of their expression with survival outcomes and assessed the effectiveness of combining ALK and EGFR inhibitors in ALK-positive NSCLC cell lines stimulated with the HER3-specific ligand HRG1.
    UNASSIGNED: High expression of c-MET, EGFR, HER2, and HER3 was observed in 4.9%, 18.0%, 1.6%, and 25.8% of primary tumors, respectively, and 18.5%, 37.0%, 10.7%, and 35.7% of secondary tumors, respectively. HER3 overexpression in primary tumors showed inferior survival (P=0.132). In the subgroup with EML4-ALK variant 1/2 (V1/V2), HER3 overexpression was significantly associated with inferior survival in both primary and secondary tumors (P=0.022 and P=0.004, respectively). Combination treatment with lorlatinib and erlotinib significantly reduced HRG1-induced activation of RTK signaling in ALK-positive NSCLC cells.
    UNASSIGNED: HER3 overexpression has potential as a prognostic marker in ALK-positive NSCLCs, including ALK-TKI naïve and treated cases, especially those with EML4-ALK V1/V2. Assessing HER3 expression may be crucial for treatment planning and outcome prediction in these 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
    c-Met是一种酪氨酸激酶受体,它的异常激活在肿瘤发生中起着关键作用,入侵,和转移性扩散在许多人类肿瘤中。PHA-665752(PHA)是c-Met的抑制剂,对许多血液恶性肿瘤和实体癌具有抗肿瘤作用。然而,c-Met的激活和表达以及PHA对人口腔鳞状细胞癌(OSCC)细胞的作用和抗肿瘤作用尚不清楚。这里,我们研究了c-Met的激活和表达以及PHA对具有高c-Met磷酸化和表达的高度致瘤性HSC-3人OSCC细胞系生长的影响。值得注意的是,c-Met在HSC-3细胞中在Y1234/1235上高表达并磷酸化,和PHA处理显著抑制这些细胞的生长和诱导凋亡。此外,抑制c-Met磷酸化(活化)的PHA进一步引起Src磷酸化和表达水平降低,蛋白激酶B(PKB),哺乳动物雷帕霉素靶蛋白(mTtor),和HSC-3细胞中的髓样细胞白血病-1(Mcl-1)。此外,显示了PHA在HSC-3细胞中的抗血管生成特性,该药物对缺氧诱导因子-1α(HIF-1α)表达的抑制作用证明,一个关键的肿瘤血管生成转录因子。重要的是,c-Met基因消融导致HSC-3细胞生长减少,Src磷酸化和HIF-1α表达减少。一起,这些结果表明c-Met在HSC-3人类口腔癌细胞中高度激活,PHA表现出强烈的抗生长,促凋亡,以及对这些细胞的抗血管生成作用,它们是通过调节多个靶标的磷酸化和表达来介导的,包括c-Met,Src,PKB,mTOR,Mcl-1和HIF-1α。
    c-Met is a tyrosine-kinase receptor, and its aberrant activation plays critical roles in tumorigenesis, invasion, and metastatic spread in many human tumors. PHA-665752 (PHA) is an inhibitor of c-Met and has antitumor effects on many hematological malignancies and solid cancers. However, the activation and expression of c-Met and its role and the antitumor effect of PHA on human oral squamous cell carcinoma (OSCC) cells remain unclear. Here, we investigated the activation and expression of c-Met and the effects of PHA on the growth of a highly tumorigenic HSC-3 human OSCC cell line with high c-Met phosphorylation and expression. Of note, c-Met was highly expressed and phosphorylated on Y1234/1235 in HSC-3 cells, and PHA treatment significantly suppressed the growth and induced apoptosis of these cells. Moreover, PHA that inhibited the phosphorylation (activation) of c-Met further caused the reduced phosphorylation and expression levels of Src, protein kinase B (PKB), mammalian target of rapamycin (mTtor), and myeloid cell leukemia-1 (Mcl-1) in HSC-3 cells. In addition, the antiangiogenic property of PHA in HSC-3 cells was shown, as evidenced by the drug\'s suppressive effect on the expression of hypoxia-inducible factor-1α (HIF-1α), a critical tumor angiogenic transcription factor. Importantly, genetic ablation of c-Met caused the reduced growth of HSC-3 cells and decreased Src phosphorylation and HIF-1α expression. Together, these results demonstrate that c-Met is highly activated in HSC-3 human oral cancer cells, and PHA exhibits strong antigrowth, proapoptotic, and antiangiogenic effects on these cells, which are mediated through regulation of the phosphorylation and expression of multiple targets, including c-Met, Src, PKB, mTOR, Mcl-1, and HIF-1α.
    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:FGF/FGFR信号通路在人类癌症中起关键作用。我们分析了靶向FGF/FGFR途径的抑制剂AZD4547的抗肿瘤作用,上皮性卵巢癌(EOC)和克服AZD4547耐药性的策略。
    方法:评估AZD4547对细胞活力/迁移的影响,并使用EOC细胞和患者来源的异种移植物(PDX)模型在腹膜内异种移植物中进行体内实验。AZD4547与SU11274(一种c-Met特异性抑制剂)的联合作用,FGF19特异性siRNA,或FGFR4抑制剂通过MTT测定进行评估。
    结果:AZD4547显著降低了对药物敏感的EOC细胞的存活和迁移,但没有降低耐药细胞的存活和迁移。AZD4547在药物敏感性A2780和SKOV3ip1细胞的异种移植模型中以及在具有药物敏感性的PDX中显着降低了肿瘤重量,但在具有耐药性A2780-CP20和SKOV3-TR细胞的模型中却没有。此外,c-Met在SKOV3-TR和HeyA8-MDR细胞中高表达,和共同施用SU11274和AZD4547协同诱导细胞死亡。此外,FGF19和FGFR4在A2780-CP20细胞中高表达。将AZD4547与FGF19siRNA或与选择性FGFR4抑制剂组合导致A2780-CP20细胞中的细胞增殖显著降低。
    结论:本研究表明AZD4547在药物敏感细胞和PDX模型中具有显著的抗癌作用,但在耐药EOC细胞中没有。在耐药细胞中,c-Met或FGF19/FGFR4的表达水平可能是AZD4547治疗反应的预测生物标志物,靶向c-Met或FGF19/FGFR4的药物与AZD4547的组合策略可能是EOC的有效治疗策略。
    BACKGROUND: The FGF/FGFR signaling pathway plays a critical role in human cancers. We analyzed the anti-tumor effect of AZD4547, an inhibitor targeting the FGF/FGFR pathway, in epithelial ovarian cancer (EOC) and strategies on overcoming AZD4547 resistance.
    METHODS: The effect of AZD4547 on cell viability/migration was evaluated and in vivo experiments in intraperitoneal xenografts using EOC cells and a patient-derived xenograft (PDX) model were performed. The effect of the combination of AZD4547 with SU11274, a c-Met-specific inhibitor, FGF19-specific siRNA, or an FGFR4 inhibitor was evaluated by MTT assay.
    RESULTS: AZD4547 significantly decreased cell survival and migration in drug-sensitive EOC cells but not drug-resistant cells. AZD4547 significantly decreased tumor weight in xenograft models of drug-sensitive A2780 and SKOV3ip1 cells and in a PDX with drug sensitivity but not in models with drug-resistant A2780-CP20 and SKOV3-TR cells. Furthermore, c-Met expression was high in SKOV3-TR and HeyA8-MDR cells, and co-administration of SU11274 and AZD4547 synergistically induced cell death. In addition, expressions of FGF19 and FGFR4 were high in A2780-CP20 cells. Combining AZD4547 with FGF19 siRNA or with a selective FGFR4 inhibitor led to significantly reduced cell proliferation in A2780-CP20 cells.
    CONCLUSIONS: This study showed that AZD4547 has significant anti-cancer effects in drug-sensitive cells and PDX models but not in drug-resistant EOC cells. In drug-resistant cells, the expression level of c-Met or FGF19/FGFR4 may be a predictive biomarker for AZD4547 treatment response, and a combination strategy of drugs targeting c-Met or FGF19/FGFR4 together with AZD4547 may be an effective therapeutic strategy for EOC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号