MET oncogene

  • 文章类型: Journal Article
    MET受体是“侵入性生长”的主要驱动因素之一,在胚胎发育和组织修复过程中必不可少的多方面生物学反应,被癌细胞篡夺以诱导和维持恶性表型。MET是癌症中激活的最重要的癌基因之一,自癌症靶向治疗的最初时代以来,人们一直在探索其抑制作用。已经开发了不同的方法来阻碍MET信号传导和/或减少作为转化标志的MET(过)表达。考虑到癌症免疫疗法获得的极大兴趣,这篇综述评估了基于免疫功能开发的治疗方法中靶向MET的机会,在MET损伤对诱导有效反应至关重要的情况下(即,当MET是恶性肿瘤的驱动因素时),或者当阻断MET代表一种增强治疗的方法(即,当MET是肿瘤适应性的佐剂时)。
    The MET receptor is one of the main drivers of \'invasive growth\', a multifaceted biological response essential during embryonic development and tissue repair that is usurped by cancer cells to induce and sustain the malignant phenotype. MET stands out as one of the most important oncogenes activated in cancer and its inhibition has been explored since the initial era of cancer-targeted therapy. Different approaches have been developed to hamper MET signaling and/or reduce MET (over)expression as a hallmark of transformation. Considering the great interest gained by cancer immunotherapy, this review evaluates the opportunity of targeting MET within therapeutic approaches based on the exploitation of immune functions, either in those cases where MET impairment is crucial to induce an effective response (i.e., when MET is the driver of the malignancy), or when blocking MET represents a way for potentiating the treatment (i.e., when MET is an adjuvant of tumor fitness).
    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:癌症中MET受体的异常激活是通过遗传改变或,更频繁地,通过转录上调。一部分MET扩增或突变的肿瘤对MET靶向剂敏感,但是他们的反应通常是短暂的,作为次级阻力最终发生。由于在没有遗传改变的情况下,MET通常不是肿瘤驱动因素,过表达MET的肿瘤对MET靶向治疗没有/反应差。因此,绝大多数显示MET激活的肿瘤仍代表未满足的医疗需求.
    方法:在这里,我们提出了一种基于表达嵌合抗原受体(CAR)的T淋巴细胞的免疫治疗策略,该T淋巴细胞靶向不同组织型的MET过表达肿瘤。我们设计了两种不同的MET-CAR构建体,并针对不同的MET过表达模型测试了MET-CAR-T细胞的细胞毒活性,包括肿瘤细胞系,原发性癌细胞,类器官,和免疫缺陷小鼠的异种移植物。
    结果:我们证明了MET-CAR-T对表达MET的细胞具有特异性细胞毒活性。细胞杀伤与细胞表面表达的MET水平成正比。虽然CAR-T细胞毒性在生理水平上与携带MET的细胞相比是最小的,基本上保留正常细胞,与MET过度表达的肿瘤相比,活性是强劲的,显着控制肿瘤细胞在体外和体内的生长。值得注意的是,MET-CAR-T细胞还能够在下游信号转导中携带次级突变的MET扩增的癌细胞中制动对MET靶向剂的获得性抗性。
    结论:我们在临床前水平设定并验证了一种MET-CAR免疫治疗策略,该策略可能有益于不适合使用抑制分子进行MET靶向治疗的癌症,包括那些表现出主要或次要阻力的。
    BACKGROUND: Aberrant activation of the MET receptor in cancer is sustained by genetic alterations or, more frequently, by transcriptional upregulations. A fraction of MET-amplified or mutated tumors are sensible to MET targeting agents, but their responsiveness is typically short-lasting, as secondary resistance eventually occurs. Since in the absence of genetic alterations MET is usually not a tumor driver, MET overexpressing tumors are not/poorly responsive to MET targeted therapies. Consequently, the vast majority of tumors exhibiting MET activation still represent an unmet medical need.
    METHODS: Here we propose an immunotherapy strategy based on T lymphocytes expressing a Chimeric Antigen Receptor (CAR) targeting MET overexpressing tumors of different histotypes. We engineered two different MET-CAR constructs and tested MET-CAR-T cell cytotoxic activity against different MET overexpressing models, including tumor cell lines, primary cancer cells, organoids, and xenografts in immune-deficient mice.
    RESULTS: We proved that MET-CAR-T exerted a specific cytotoxic activity against MET expressing cells. Cell killing was proportional to the level of MET expressed on the cell surface. While CAR-T cytotoxicity was minimal versus cells carrying MET at physiological levels, essentially sparing normal cells, the activity versus MET overexpressing tumors was robust, significantly controlling tumor cell growth in vitro and in vivo. Notably, MET-CAR-T cells were also able to brake acquired resistance to MET targeting agents in MET amplified cancer cells carrying secondary mutations in downstream signal transducers.
    CONCLUSIONS: We set and validated at the pre-clinical level a MET-CAR immunotherapy strategy potentially beneficial for cancers not eligible for MET targeted therapy with inhibitory molecules, including those exhibiting primary or secondary resistance.
    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:肝细胞生长因子(HGF)受体MET是原发性肿瘤阶段或对肿瘤靶向酪氨酸激酶抑制剂(TKIs)治疗的获得性耐药的非小细胞肺癌细胞(NSCLC)亚群中的致癌驱动因子。
    UNASSIGNED:本文总结了通过扩增和突变(包括外显子14畸变)导致MET过表达和激活的机制。此外,讨论了将MET检测和分类为肿瘤驱动因素的方法以及用于患者治疗的选择性TKI。
    未经证实:NSCLC中激酶的激活突变和重排是成功治疗干预的目标。然而,MET激活涉及许多复杂的改变,包括基因扩增,通过METex14外显子跳跃和大量基因突变来防止降解。高水平的MET表达是肿瘤对TKI的反应的前提,并且在缺乏MET扩增的标准化和一致测定的情况下,在原发性病变和对突变的EGFR定向治疗表现出抗性的肿瘤中,难以确认MET依赖性肿瘤进展。
    UNASSIGNED: The hepatocyte growth factor (HGF) receptor MET is an oncogenic driver in a subpopulation of Non-small Lung Cancer Cells (NSCLC) at the primary tumor stage or in acquired resistance to treatment with tumor-targeting tyrosine kinase inhibitors (TKIs).
    UNASSIGNED: This article summarizes the mechanisms leading to overexpression and activation of MET by amplification and mutations including exon 14 aberrations. Furthermore, the methods to detect and categorize MET as a tumor driver and the selective TKIs for patient treatment are discussed.
    UNASSIGNED: Activating mutations and rearrangements of kinases in NSCLC are the target of successful therapeutic intervention. However, MET activation involves a number of complex alterations including gene amplification, prevention of degradation by METex14 exon skipping and a host of gene mutations. A high-level MET expression is the precondition for tumor responses to TKIs and the confirmation of MET-dependent tumor progression is difficult in primary lesions and in tumors exhibiting resistance to mutated EGFR-directed therapy in absence of standardized and concordant assays of MET amplification.
    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:由MET癌基因编码的酪氨酸激酶受体是癌症的主要参与者。当MET负责转化表型(MET成瘾癌症)的发作和进展时,其致癌激活的有效阻断导致有效的肿瘤生长抑制。
    方法:在这里,我们描述了一种分子工程化的MET抗体(hOA-DN30),并在体外和体内验证了其在MET成瘾癌症模型中的药理活性。还评估了非人灵长类动物中的药代动力学和安全性。
    结果:hOA-DN30通过剂量和时间依赖性地从细胞表面去除受体(脱落)而有效地损害MET活化和细胞内信号级联。体外,该抗体通过阻断细胞增殖并同时诱导多种MET成瘾的人类肿瘤细胞系中的细胞死亡来抑制细胞生长。在小鼠异种移植物中,hOA-DN30诱导了令人印象深刻的肿瘤量减少,有一个广阔的治疗窗口。此外,该抗体对MET成瘾的胃肿瘤产生的患者源性异种移植物显示出很高的治疗效果,导致肿瘤完全消退和治疗停止后的长期效果。最后,hOA-DN30在食蟹猴中显示出非常有利的药代动力学特征和基本的耐受性。
    结论:hOA-DN30同时擦除细胞表面MET并释放“诱饵”受体胞外区的独特能力导致了最重要的MET阻断作用。其在大量临床前模型中的显著疗效,以及其在非人灵长类动物中的药理特征和安全性,强烈设想这种新型单臂MET治疗性抗体在MET成瘾癌症治疗中的成功临床应用。
    BACKGROUND: The tyrosine kinase receptor encoded by the MET oncogene is a major player in cancer. When MET is responsible for the onset and progression of the transformed phenotype (MET-addicted cancers), an efficient block of its oncogenic activation results in potent tumor growth inhibition.
    METHODS: Here we describe a molecular engineered MET antibody (hOA-DN30) and validate its pharmacological activity in MET-addicted cancer models in vitro and in vivo. Pharmacokinetics and safety profile in non-human primates have also been assessed.
    RESULTS: hOA-DN30 efficiently impaired MET activation and the intracellular signalling cascade by dose and time dependent removal of the receptor from the cell surface (shedding). In vitro, the antibody suppressed cell growth by blocking cell proliferation and by concomitantly inducing cell death in multiple MET-addicted human tumor cell lines. In mice xenografts, hOA-DN30 induced an impressive reduction of tumor masses, with a wide therapeutic window. Moreover, the antibody showed high therapeutic efficacy against patient-derived xenografts generated from MET-addicted gastric tumors, leading to complete tumor regression and long-lasting effects after treatment discontinuation. Finally, hOA-DN30 showed a highly favorable pharmacokinetic profile and substantial tolerability in Cynomolgus monkeys.
    CONCLUSIONS: hOA-DN30 unique ability to simultaneously erase cell surface MET and release the \'decoy\' receptor extracellular region results in a paramount MET blocking action. Its remarkable efficacy in a large number of pre-clinical models, as well as its pharmacological features and safety profile in non-human primates, strongly envisage a successful clinical application of this novel single-arm MET therapeutic antibody for the therapy of MET-addicted cancers.
    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
    胰腺导管腺癌是一种侵袭性肿瘤,其特征是存在丰富的基质区室,对恶性表型有重要贡献。胰腺星状细胞是存在于基质中的特殊成纤维细胞,是细胞外基质蛋白的主要来源。促炎细胞因子,和生长因子,包括肝细胞生长因子(HGF)。开发人类胰腺星状细胞和癌细胞的共培养系统,我们证明,HGF/MET轴抑制后,成纤维细胞活化降低。为了揭示肿瘤中MET激活所协调的维持基质调节的信号通路,我们分析了用HGF刺激和用HGF/MET抑制剂处理的胰腺癌细胞中的基因表达谱.转录组分析表明,在所有由HGF调节的基因中,在使用抑制剂治疗后,125个基因的子集恢复至基础水平.通过独创性通路分析检查这些基因,生腱蛋白C成为一个有希望的候选者,将MET信号传导和肿瘤微环境联系起来。在体外和体内的RNA和蛋白质水平上验证了胰腺癌细胞中MET依赖性生腱蛋白C的调节。总之,这项工作确定生腱蛋白C是由MET激活调节的基因,提示在胰腺肿瘤进展过程中发生的MET介导的肿瘤-基质相互作用中的作用。
    Pancreatic ductal adenocarcinoma is an aggressive tumor characterized by the presence of an abundant stromal compartment contributing significantly to the malignant phenotype. Pancreatic stellate cells are peculiar fibroblasts present in the stroma and represent the predominant source of extracellular matrix proteins, pro-inflammatory cytokines, and growth factors, including hepatocyte growth factor (HGF). Exploiting a co-culture system of human pancreatic stellate cells and cancer cells, we demonstrated that fibroblast activation was reduced upon HGF/MET axis inhibition. To unveil the signaling pathways sustaining stroma modulation orchestrated by MET activation in the tumor, we analyzed the gene expression profile in pancreatic cancer cells stimulated with HGF and treated with HGF/MET inhibitors. Transcriptome analysis showed that, among all the genes modulated by HGF, a subset of 125 genes was restored to the basal level following treatment with the inhibitors. By examining these genes via ingenuity pathway analysis, tenascin C emerged as a promising candidate linking MET signaling and tumor microenvironment. MET-dependent tenascin C modulation in pancreatic cancer cells was validated at RNA and protein levels both in vitro and in vivo. In conclusion, this work identifies tenascin C as a gene modulated by MET activation, suggesting a role in MET-mediated tumor-stroma interplay occurring during pancreatic tumor progression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    The MET oncogene encodes a tyrosine kinase receptor involved in the control of a complex network of biological responses that include protection from apoptosis and stimulation of cell growth during embryogenesis, tissue regeneration, and cancer progression. We previously developed an antagonist antibody (DN30) inducing the physical removal of the receptor from the cell surface and resulting in suppression of the biological responses to MET. In its bivalent form, the antibody displayed a residual agonist activity, due to dimerization of the lingering receptors, and partial activation of the downstream signaling cascade. The balance between the two opposing activities is variable in different biological systems and is hardly predictable. In this study, we generated and characterized two single-chain antibody fragments derived from DN30, sharing the same variable regions but including linkers different in length and composition. The two engineered molecules bind MET with high affinity but induce different biological responses. One behaves as a MET-antagonist, promoting programmed cell death in MET \"addicted\" cancer cells. The other acts as a hepatocyte growth factor (HGF)-mimetic, protecting normal cells from doxorubicin-induced apoptosis. Thus, by engineering the same receptor antibody, it is possible to generate molecules enhancing or inhibiting apoptosis either to kill cancer cells or to protect healthy tissues from the injuries of chemotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    MET, a master gene sustaining \"invasive growth,\" is a relevant target for cancer precision therapy. In the vast majority of tumors, wild-type MET behaves as a \"stress-response\" gene and relies on the ligand (HGF) to sustain cell \"scattering,\" invasive growth and apoptosis protection (oncogene \"expedience\"). In this context, concomitant targeting of MET and HGF could be crucial to reach effective inhibition. To test this hypothesis, we combined an anti-MET antibody (MvDN30) inducing \"shedding\" (i.e., removal of MET from the cell surface), with a \"decoy\" (i.e., the soluble extracellular domain of the MET receptor) endowed with HGF-sequestering ability. To avoid antibody/decoy interaction-and subsequent neutralization-we identified a single aminoacid in the extracellular domain of MET-lysine 842-that is critical for MvDN30 binding and engineered the corresponding recombinant decoyMET (K842E). DecoyMETK842E retains the ability to bind HGF with high affinity and inhibits HGF-induced MET phosphorylation. In HGF-dependent cellular models, MvDN30 antibody and decoyMETK842E used in combination cooperate in restraining invasive growth, and synergize in blocking cancer cell \"scattering.\" The antibody and the decoy unbridle apoptosis of colon cancer stem cells grown in vitro as spheroids. In a preclinical model, built by orthotopic transplantation of a human pancreatic carcinoma in SCID mice engineered to express human HGF, concomitant treatment with antibody and decoy significantly reduces metastatic spread. The data reported indicate that vertical targeting of the MET/HGF axis results in powerful inhibition of ligand-dependent MET activation, providing proof of concept in favor of combined target therapy of MET \"expedience.\"
    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
    由于对癌症患者预后的潜在影响,导致转移扩散的分子和细胞机制一直是争论和科学研究的话题。除了遗传和环境因素,单细胞力学和与周围环境的物理相互作用在确定侵袭性表型中起着相关作用。重建转移性克隆的物理性质可能有助于澄清疾病进展中仍然存在的问题,并导致新的诊断和治疗方法。从这个角度来看,原发性未知的癌症(CUP)确定了研究转移过程中涉及的物理相互作用和力量的理想模型。我们先前已经证明MET癌基因在CUP中以意想不到的高频率突变。我们在这里分析和讨论如何通过体细胞突变的MET激活可能会影响物理特性,从而导致这种高度恶性综合征,正如CUP定义的那样。
    The molecular and cellular mechanisms which drive metastatic spread are the topic of constant debate and scientific research due to the potential implications for cancer patients\' prognosis. In addition to genetics and environmental factors, mechanics of single cells and physical interaction with the surrounding environment play relevant role in defining invasive phenotype. Reconstructing the physical properties of metastatic clones may help to clarify still open issues in disease progression as well as to lead to new diagnostic and therapeutic approaches. In this perspective cancer of unknown primary origin (CUP) identify the ideal model to study physical interactions and forces involved in the metastatic process. We have previously demonstrated that MET oncogene is mutated with unexpected high frequency in CUPs. We here analyze and discuss how the MET activation by somatic mutation may affect physical properties in giving rise to such a highly malignant syndrome, as that defined by CUP.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    The discovery of druggable oncogenic drivers (i.e. EGFR and ALK), along with the introduction of comprehensive tumor genotyping techniques into the daily clinical practice define non-small-cell lung cancer (NSCLC) asa group of heterogeneous diseases, requiring a context-personalized clinico-therapeutical approach. Among the most investigated biomarkers, the MET proto-oncogene has been extensively demonstrated to play a crucial role throughout the lung oncogenesis, unbalancing the proliferation/apoptosis signaling and influencing the epithelial-mesenchymal transition and the invasive phenotype. Nevertheless, although different mechanisms eliciting the aberrant MET-associated oncogenic stimulus have been detected in lung cancer (such as gene amplification, increased gene copy number, mutations and MET/HGF overexpression), to date no clinically impactful results have been achieved with anti-MET tyrosine kinase inhibitors and monoclonal antibodies in the context of an unselected or MET enriched population. Recently, MET exon 14 splicing abnormalities have been identified asa potential oncogenic target in lung cancer, able to drive the activity of MET inhibitors in molecularly selected patients. In this paper, the major advancement and drawbacks of MET history in lung cancer are reviewed, underlying the renewed scientific euphoria related to the recent identification of MET exon 14 splicing variants asan actionable oncogenic target.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    胶质母细胞瘤(GBM)含有已知对电离辐射有抗性的干细胞样细胞(GSC),因此导致治疗失败和快速致死的肿瘤复发。已知GSC辐射抗性依赖于DNA损伤反应的有效激活。但将这种反应与茎状态联系起来的机制仍不清楚。这里,我们发现MET受体激酶,GSC的功能标记,在抗放射性GSC的子集中特异性表达,并且在放疗后重复的人GBM中过表达。我们阐明了MET通过一种新机制促进GSC辐射抗性,依靠AKT活性并导致(I)Aurora激酶A的持续激活,ATM激酶,和DNA修复的下游效应,和(ii)p21的磷酸化和细胞质保留,这与抗凋亡功能有关。我们表明,MET药理学抑制会导致DNA损伤在辐照的GSC中积累,并在体外和GSC异种移植产生的GBM中耗尽。因此,临床前证据表明,MET抑制剂可以使肿瘤放射增敏并转化GSC阳性选择,由放射治疗诱导,根除GSC。
    Glioblastoma (GBM) contains stem-like cells (GSCs) known to be resistant to ionizing radiation and thus responsible for therapeutic failure and rapidly lethal tumor recurrence. It is known that GSC radioresistance relies on efficient activation of the DNA damage response, but the mechanisms linking this response with the stem status are still unclear. Here, we show that the MET receptor kinase, a functional marker of GSCs, is specifically expressed in a subset of radioresistant GSCs and overexpressed in human GBM recurring after radiotherapy. We elucidate that MET promotes GSC radioresistance through a novel mechanism, relying on AKT activity and leading to (i) sustained activation of Aurora kinase A, ATM kinase, and the downstream effectors of DNA repair, and (ii) phosphorylation and cytoplasmic retention of p21, which is associated with anti-apoptotic functions. We show that MET pharmacological inhibition causes DNA damage accumulation in irradiated GSCs and their depletion in vitro and in GBMs generated by GSC xenotransplantation. Preclinical evidence is thus provided that MET inhibitors can radiosensitize tumors and convert GSC-positive selection, induced by radiotherapy, into GSC eradication.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号