Ethanol Ablation Therapy Drives Immune-Mediated Antitumor Effects in Murine Breast Cancer Models

CANCERS(2022)

引用 0|浏览9
暂无评分
摘要
Simple Summary Tumor ablation is the process of directly destroying tumor tissue by injecting a cytotoxic substance, in this case, ethanol ethylcellulose. In this report, we characterized the effect of ablation on local and systemic immunologic markers known to impact disease progression in several mouse models. Ablation improved overall survival in poorly invasive breast cancer models and was notable for demonstrating an increase in tumor infiltrating lymphocytes. However, in a metastatic breast cancer model, the response to ablation was more nuanced: the growth of the primary tumor was only modestly slowed compared to controls, and there was a reduction in pro-tumor granulocytic myeloid derived suppressor cells (gMDSCs) with a reduction in metastatic disease. A single ablation reduced circulating granulocytic colony stimulating factor, tumoral gMDSCs, splenic gMDSCs, and pulmonary gMDSCs, as well as the suppressive ability of MDSCs on CD4 and CD8 T cells. The immunomodulation incited by tumor ablation was utilized to recover response to checkpoint inhibitors, resulting in increased overall survival compared to checkpoint inhibitors alone, demonstrating a proof-of-concept for using ethanol ablation as an adjuvant immunomodulatory therapy. Ethanol ablation is a minimally invasive, cost-effective method of destroying tumor tissue through an intratumoral injection of high concentrations of cytotoxic alcohol. Ethyl-cellulose ethanol (ECE) ablation, a modified version of ethanol ablation, contains the phase-changing polysaccharide ethyl-cellulose to reduce ethanol leakage away from the tumor. Ablation produces tissue necrosis and initiates a wound healing process; however, the characteristic of the immunologic events after ECE ablation of tumors has yet to be explored. Models of triple-negative breast cancer (TNBC), which are classically immunosuppressive and difficult to treat clinically, were used to characterize the immunophenotypic changes after ECE ablation. In poorly invasive TNBC rodent models, the injury to the tumor induced by ECE increased tumor infiltrating lymphocytes (TILs) and reduced tumor growth. In a metastatic TNBC model (4T1), TILs did not increase after ECE ablation, though lung metastases were reduced. 4T1 tumors secrete high levels of granulocytic colony stimulating factor (G-CSF), which induces a suppressive milieu of granulocytic myeloid-derived suppressor cells (gMDSCs) aiding in the formation of metastases and suppression of antitumor immunity. We found that a single intratumoral injection of ECE normalized tumor-induced myeloid changes: reducing serum G-CSF and gMDSC populations. ECE also dampened the suppressive strength of gMDSC on CD4 and CD8 cell proliferation, which are crucial for anti-tumor immunity. To demonstrate the utility of these findings, ECE ablation was administered before checkpoint inhibitor (CPI) therapy in the 4T1 model and was found to significantly increase survival compared to a control of saline and CPI. Sixty days after tumor implant no primary tumors or metastatic lung lesions were found in 6/10 mice treated with CPI plus ECE, compared to 1/10 with ECE alone and 0/10 with CPI and saline.
更多
查看译文
关键词
breast cancer, myeloid derived supressor cells, immunomodulation, tumor ablation, low-resource settings, tumor microenvironment
AI 理解论文
溯源树
样例
生成溯源树,研究论文发展脉络
Chat Paper
正在生成论文摘要