4.7 Article

Tumor microenvironment modulation enhances immunologic benefit of chemoradiotherapy

期刊

出版社

BMC
DOI: 10.1186/s40425-018-0485-9

关键词

Immunotherapy; Tumor microenvironment; Inducible nitric oxide synthase (iNOS); Cyclophosphamide; L-n6-(1-iminoethyl)-lysine (L-NIL); Chemoradiotherapy; Radiotherapy; Head and neck squamous cell carcinoma; Head and neck cancer; Human papillomavirus (HPV)

资金

  1. National Institute of General Medical Sciences T32 predoctoral training grant of the National Institutes of Health [T32GM088129]
  2. National Institute of Dental & Craniofacial Research F31 NRSA training grant of the National Institutes of Health [F31DE026682]
  3. Dutch Cancer Society [2014-6696]
  4. Swim Across America
  5. German Cancer Aid
  6. Cancer Prevention and Research Institutes of Texas [CPRIT RR160027]
  7. McNair Medical Institute at The Robert and Janice McNair Foundation
  8. Kom op tegen Kanker (Stand Up against Cancer)
  9. FWO (Science Foundation Flanders)
  10. Foundation against Cancer
  11. Caroline Weiss Law Endowment for Academic Excellence
  12. Owens Foundation
  13. Cancer Research Institute
  14. National Institutes of Health [NCI/NIDCR 1U01DE028233-01]

向作者/读者索取更多资源

BackgroundChemoradiotherapy (CRT) remains one of the most common cancer treatment modalities, and recent data suggest that CRT is maximally effective when there is generation of an anti-tumoral immune response. However, CRT has also been shown to promote immunosuppressive mechanisms which must be blocked or reversed to maximize its immune stimulating effects.MethodsTherefore, using a preclinical model of human papillomavirus (HPV)-associated head and neck squamous cell carcinoma (HNSCC), we developed a clinically relevant therapy combining CRT and two existing immunomodulatory drugs: cyclophosphamide (CTX) and the small molecule inducible nitric oxide synthase (iNOS) inhibitor L-n6-(1-iminoethyl)-lysine (L-NIL). In this model, we treated the syngeneic HPV-HNSCC mEER tumor-bearing mice with fractionated (10 fractions of 3Gy) tumor-directed radiation and weekly cisplatin administration. We compared the immune responses induced by CRT and those induced by combinatory treatment (CRT+CTX/L-NIL) with flow cytometry, quantitative multiplex immunofluorescence and by profiling immune-related gene expression changes.ResultsWe show that combination treatment favorably remodels the tumor myeloid immune microenvironment including an increase in anti-tumor immune cell types (inflammatory monocytes and M1-like macrophages) and a decrease in immunosuppressive granulocytic myeloid-derived suppressor cells (MDSCs). Intratumoral T cell infiltration and tumor antigen specificity of T cells were also improved, including a 31.8-fold increase in the CD8(+) T cell/ regulatory T cell ratio and a significant increase in tumor antigen-specific CD8(+) T cells compared to CRT alone. CTX/LNIL immunomodulation was also shown to significantly improve CRT efficacy, leading to rejection of 21% established tumors in a CD8-dependent manner.ConclusionsOverall, these data show that modulation of the tumor immune microenvironment with CTX/L-NIL enhances susceptibility of treatment-refractory tumors to CRT. The combination of tumor immune microenvironment modulation with CRT constitutes a translationally relevant approach to enhance CRT efficacy through enhanced immune activation.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据