关键词: FRET HER2 breast tumor fluorescence lifetime imaging metabolism target engagement targeted drug delivery tumor microenvironment

来  源:   DOI:10.1101/2024.01.12.575453   PDF(Pubmed)

Abstract:
UNASSIGNED: Trastuzumab (TZM) is a monoclonal antibody that targets the human epidermal growth factor receptor (HER2) and is clinically used for the treatment of HER2-positive breast tumors. However, the tumor microenvironment can limit the access of TZM to the HER2 targets across the whole tumor and thereby compromise TZM\'s therapeutic efficacy. An imaging methodology that can non-invasively quantify the binding of TZM-HER2, which is required for therapeutic action, and distribution within tumors with varying tumor microenvironments is much needed.
UNASSIGNED: We performed near-infrared (NIR) fluorescence lifetime (FLI) Forster Resonance Energy Transfer (FRET) to measure TZM-HER2 binding, using in vitro microscopy and in vivo widefield macroscopy, in HER2 overexpressing breast and ovarian cancer cells and tumor xenografts, respectively. Immunohistochemistry was used to validate in vivo imaging results.
UNASSIGNED: NIR FLI FRET in vitro microscopy data show variations in intracellular distribution of bound TZM in HER2-positive breast AU565 and AU565 tumor-passaged XTM cell lines in comparison to SKOV-3 ovarian cancer cells. Macroscopy FLI (MFLI) FRET in vivo imaging data show that SKOV-3 tumors display reduced TZM binding compared to AU565 and XTM tumors, as validated by ex vivo immunohistochemistry. Moreover, AU565/XTM and SKOV-3 tumor xenografts display different amounts and distributions of TME components, such as collagen and vascularity. Therefore, these results suggest that SKOV-3 tumors are refractory to TZM delivery due to their disrupted vasculature and increased collagen content.
UNASSIGNED: Our study demonstrates that FLI is a powerful analytical tool to monitor the delivery of antibody drug tumor both in cell cultures and in vivo live systems. Especially, MFLI FRET is a unique imaging modality that can directly quantify target engagement with potential to elucidate the role of the TME in drug delivery efficacy in intact live tumor xenografts.
摘要:
曲妥珠单抗(TZM)是一种靶向人表皮生长因子受体(HER2)的单克隆抗体,临床上用于治疗HER2阳性乳腺肿瘤。然而,肿瘤微环境可限制TZM进入整个肿瘤的HER2靶点,从而损害TZM的治疗效果.一种成像方法,可以非侵入性地量化TZM-HER2的结合,这是治疗作用所必需的,并且在具有不同肿瘤微环境的肿瘤内分布是非常需要的。
我们进行了近红外(NIR)荧光寿命(FLI)福斯特共振能量转移(FRET)来测量TZM-HER2结合,使用体外显微镜和体内宽视野显微镜,在过表达HER2的乳腺癌和卵巢癌细胞和肿瘤异种移植物中,分别。免疫组织化学用于验证体内成像结果。
NIRFLIFRET体外显微镜数据显示与SKOV-3卵巢癌细胞相比,HER2阳性乳腺AU565和AU565肿瘤传代的XTM细胞系中结合的TZM的细胞内分布变化。宏观FLI(MFLI)FRET体内成像数据显示,与AU565和XTM肿瘤相比,SKOV-3肿瘤显示TZM结合减少,通过离体免疫组织化学验证。此外,AU565/XTM和SKOV-3肿瘤异种移植物显示不同数量和分布的TME成分,如胶原蛋白和血管。因此,这些结果表明,SKOV-3肿瘤由于其血管破坏和胶原蛋白含量增加而难以接受TZM递送.
我们的研究表明,FLI是一种强大的分析工具,可以在细胞培养和体内活系统中监测抗体药物肿瘤的递送。尤其是,MFLIFRET是一种独特的成像模式,可以直接量化靶标接合,有可能阐明TME在完整的活肿瘤异种移植物中的药物递送功效中的作用。
公众号