Her2 internalization

  • 文章类型: Journal Article
    在过去的十年中,HER2靶向疗法在乳腺癌治疗方面取得了进步。临床上,通过免疫组织化学或通过荧光原位杂交的基因调控评估肿瘤细胞表面的HER2水平来确定HER2治疗的合格性.HER2治疗在HER2水平升高的患者中并不总是有效,这质疑HER2的量是否足以预测患者的预后。此外,HER2靶向抗体-药物偶联物(ADC)Enhertu®最近被批准用于转移性低HER2癌症,证实HER2治疗对低HER2水平患者的益处。为了评估HER2水平与治疗效果之间的相关性,我们使用流式细胞术定量了8种细胞系的HER2,同时测定了2种HER2靶向ADC的毒性.高HER2细胞系和低HER2细胞系均对ADC具有显著的毒性反应。我们量化了HER2内在化,发现HER2水平和内在化百分比之间没有相关性。我们发现了一个有用的指标,表明运输到溶酶体的HER2受体的最小数量足以提供有效的治疗。我们的结果表明,目前确定HER2治疗资格的标准可能会限制患者获得有效治疗。总之,HER2水平并不完全足以确定对ADC治疗的反应。
    HER2-targeting therapies have advanced breast cancer treatment over the past decade. Clinically, eligibility for HER2 therapies is determined by assessing HER2 levels on tumor cell surfaces through immunohistochemistry or by gene regulation through fluorescence in situ hybridization. HER2 therapies are not always effective in patients with elevated levels of HER2, questioning whether the amount of HER2 is sufficiently predictive of patient outcomes. Additionally, the HER2-targeting antibody-drug conjugate (ADC) Enhertu® was recently approved for metastasized HER2-low cancers, confirming the benefits of HER2 treatment for patients with low HER2 levels. To evaluate the correlation between HER2 levels and treatment efficacy, we quantified HER2 on eight cell lines using flow cytometry while simultaneously determining the toxicity of two HER2-targeting ADCs. Both HER2-high cell lines and HER2-low cell lines had significant toxicity responses to ADCs. We quantified HER2 internalization and found no correlation between HER2 levels and the percentage of internalization. We found a useful metric suggesting that a minimum number of HER2 receptors trafficked to lysosomes is sufficient to provide effective treatment. Our results indicate that the current standards of determining eligibility for HER2 therapy could limit patients\' access to effective treatment. In conclusion, HER2 levels are not wholly adequate to determine the response to ADC treatment.
    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
    The overexpression of tyrosine kinase HER2 in numerous cancers, connected with fierce signaling and uncontrolled proliferation, makes it a suitable target for immunotherapy. The acquisition of resistance to currently used compounds and the multiplicity of signaling pathways involved prompted research into the discovery of novel binders as well as treatment options with multiple targeting and multispecific agents. Here we constructed an anti-HER2 tetravalent and biparatopic symmetrical IgG-like molecule by combining the Fab of pertuzumab with a HER2-specific Fcab (Fc fragment with antigen binding), which recognizes an epitope overlapping with trastuzumab. In the strongly HER2-positive cell line SK-BR-3, the molecule induced a rapid and efficient reduction in surface HER2 levels. A potent anti-proliferative effect, specific for the HER2-positive cell line, was observed in vitro, following the induction of apoptosis, and this could not be achieved with treatment with the mixture of pertuzumab and the parental Fcab. The inhibitory cytotoxic effect of our antibody as a single agent makes it a promising contribution to the armory of anti-cancer molecules directed against HER2-addicted cells.
    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
    单克隆抗体可以通过融合方法或其CDR环的修饰获得第二抗原的接合特性。而且还通过修改它们的恒定域,例如在mAb2形式中,其中CH3结构域中的一组突变的氨基酸残基实现与第二抗原的高亲和力特异性相互作用。我们测试了通过在具有曲妥珠单抗可变结构域和VEGF结合CH3结构域的模型支架中用一对抗原结合CH3结构域替换FabCH1/CL结构域对引入第二抗原的多个结合位点的可能性。此类双特异性分子以“Fab样”形式和全长抗体形式产生。使用质谱法,新的构建体具有预期的分子组成。它们在标准实验室条件下表现得很高,用蛋白A和凝胶过滤纯化为单体,具有高热稳定性。保留了它们与两种靶抗原的高亲和力结合。最后,由于与内源性分泌的细胞因子的交联水平提高,Her2/VEGF结合结构域交换的双特异性抗体能够介导对Her2过表达的细胞系SK-BR-3增强的表面Her2内化作用。最后,具有以交换的抗原结合CH3结构域为特征的Fab的双特异性抗体在抗原结合位点的定位和效价方面提供了替代解决方案。
    Monoclonal antibodies can acquire the property of engagement of a second antigen via fusion methods or modification of their CDR loops, but also by modification of their constant domains, such as in the mAb2 format where a set of mutated amino acid residues in the CH3 domains enables a high-affinity specific interaction with the second antigen. We tested the possibility of introducing multiple binding sites for the second antigen by replacing the Fab CH1/CL domain pair with a pair of antigen-binding CH3 domains in a model scaffold with trastuzumab variable domains and VEGF-binding CH3 domains. Such bispecific molecules were produced in a \"Fab-like\" format and in a full-length antibody format. Novel constructs were of expected molecular composition using mass spectrometry. They were expressed at a high level in standard laboratory conditions, purified as monomers with Protein A and gel filtration and were of high thermostability. Their high-affinity binding to both target antigens was retained. Finally, the Her2/VEGF binding domain-exchanged bispecific antibody was able to mediate a potentiated surface Her2-internalization effect on the Her2-overexpressing cell line SK-BR-3 due to improved level of cross-linking with the endogenously secreted cytokine. To conclude, bispecific antibodies with Fabs featuring exchanged antigen-binding CH3 domains offer an alternative solution in positioning and valency of antigen binding sites.
    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
    背景:人表皮生长因子受体2(HER2)在具有高转移率的人乳腺癌亚组中被扩增并成为临床靶标。涉及抗体曲妥珠单抗和曲妥珠单抗-emtansine(T-DM1)的靶向治疗极大地改善了HER2阳性(HER2+)乳腺癌患者的预后。然而,对这些靶向治疗的耐药性会发展并限制其疗效。这里,我们在HER2+乳腺癌模型中测试内吞衔接蛋白内皮素A2(EndoII)的参与,以及他们对曲妥珠单抗和T-DM1治疗的反应。
    方法:用免疫组织化学方法分析EndoⅡ在人乳腺肿瘤和淋巴结转移中的表达。在HER2+癌细胞系(SK-BR-3和HCC1954)中实现了EndoII的稳定沉默,以测试EndoII对HER2水平的影响,定位和信令,细胞运动和肿瘤转移。使用实时细胞运动性和细胞毒性测定测试EndoII沉默对HER2+癌细胞对曲妥珠单抗或T-DM1治疗的应答的影响。
    结果:在HER2阳性肿瘤中检测到高EndoII蛋白表达,并且在mRNA水平上与淋巴结阳性HER2+乳腺癌的总体生存率较差有关.EndoII在HER2+细胞系中的稳定沉默导致细胞表面HER2水平升高,表皮生长因子诱导的HER2内化受损,和下游效应子激酶Akt和Erk的信号传导减少。EndoII沉默还导致HER2+癌细胞在体外的迁移和侵袭减少,和小鼠尾静脉注射后肺接种受损。此外,EndoII沉默也损害了响应曲妥珠单抗的HER2内化,并导致用T-DM1处理的HER2+癌细胞的细胞毒性反应降低。
    结论:我们的研究提供了EndoII在HER2+癌细胞运动性和HER2运输中的功能的新证据,这与曲妥珠单抗或T-DM1的有效治疗有关。因此,EndoII的差异表达可能与对基于曲妥珠单抗的HER2+癌症治疗的敏感性或耐药性有关.
    BACKGROUND: Human epidermal growth factor receptor-2 (HER2) is amplified and a clinical target in a subset of human breast cancers with high rates of metastasis. Targeted therapies involving the antibody trastuzumab and trastuzumab-emtansine (T-DM1) have greatly improved outcomes for HER2-positive (HER2+) breast cancer patients. However, resistance to these targeted therapies can develop and limit their efficacy. Here, we test the involvement of the endocytic adaptor protein endophilin A2 (Endo II) in HER2+ breast cancer models, and their responses to treatments with trastuzumab and T-DM1.
    METHODS: Endo II expression in human breast tumors and lymph node metastases were analyzed by immunohistochemistry. Stable silencing of Endo II was achieved in HER2+ cancer cell lines (SK-BR-3 and HCC1954) to test Endo II effects on HER2 levels, localization and signaling, cell motility and tumor metastasis. The effects of Endo II silencing on the responses of HER2+ cancer cells to trastuzumab or T-DM1 treatments were tested using real-time cell motility and cytotoxicity assays.
    RESULTS: High Endo II protein expression was detected in HER2-positive tumors, and was linked to worse overall survival in node-positive HER2+ breast cancers at the mRNA level. Stable silencing of Endo II in HER2+ cell lines led to elevated levels of HER2 on the cell surface, impaired epidermal growth factor-induced HER2 internalization, and reduced signaling to downstream effector kinases Akt and Erk. Endo II silencing also led to decreased migration and invasion of HER2+ cancer cells in vitro, and impaired lung seeding following tail vein injection in mice. In addition, Endo II silencing also impaired HER2 internalization in response to Trastuzumab, and led to reduced cytotoxicity response in HER2+ cancer cells treated with T-DM1.
    CONCLUSIONS: Our study provides novel evidence of Endo II function in HER2+ cancer cell motility and trafficking of HER2 that relates to effective treatments with trastuzumab or T-DM1. Thus, differential expression of Endo II may relate to sensitivity or resistance to trastuzumab-based therapies for HER2+ cancers.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号