4.7 Article

Peripherally-driven myeloid NFkB and IFN/ISG responses predict malignancy risk, survival, and immunotherapy regime in ovarian cancer

期刊

出版社

BMJ PUBLISHING GROUP
DOI: 10.1136/jitc-2021-003609

关键词

tumor biomarkers; tumor microenvironment; immunological techniques; computational biology; immunotherapy

资金

  1. Research Foundation Flanders (FWO) [G0B4620N, G0B4716N, 30837538]
  2. KU Leuven [C14/19/098, C3/21/037, POR/16/040, C24/15/037, PDMT1/21/032]
  3. Kom op Tegen Kanker (Stand Up To Cancer, the Flemish Cancer Society) [KOTK/2018/11509/1, KOTK/2016/10728/2603, KOTK/2019/11955/1, 11758]
  4. Amgen Chair for Therapeutic Advances in Ovarian Cancer [2017/LUF/00069]
  5. Vriendtjes Tegen Kanker [2016_LUF_00078]
  6. VLIR-UOS (iBOF grant) [iBOF/21/048]
  7. FWO-SB PhD Fellowship [1S06821N]
  8. Linbury Trust [LIN2600]
  9. Belgian Federation against Cancer [2018-127, 2016-133]
  10. Fondation Roi-Baudouin

向作者/读者索取更多资源

The study established a novel biomarker resource for serum screening in patients with ovarian cancer to evaluate peripheral immunodynamics, patient survival trends, malignancy risk, and preclinical chemo-immunotherapy strategies. The in silico data-driven framework predicted that analysis of myeloid NF kappa B signaling and interferon-stimulated genes' responses could capture patients' peripheral immunodynamics effectively. Multiparametric serum profiling of patients with ovarian cancer showed that the sFIS assay can decode peripheral immunology, estimate survival trends, and co-estimate malignancy risk.
Background Tumors can influence peripheral immune macroenvironment, thereby creating opportunities for non-invasive serum/plasma immunobiomarkers for immunostratification and immunotherapy designing. However, current approaches for immunobiomarkers' detection are largely quantitative, which is unreliable for assessing functional peripheral immunodynamics of patients with cancer. Hence, we aimed to design a functional biomarker modality for capturing peripheral immune signaling in patients with cancer for reliable immunostratification. Methods We used a data-driven in silico framework, integrating existing tumor/blood bulk-RNAseq or single-cell (sc)RNAseq datasets of patients with cancer, to inform the design of an innovative serum-screening modality, that is, serum-functional immunodynamic status (sFIS) assay. Next, we pursued proof-of-concept analyses via multiparametric serum profiling of patients with ovarian cancer (OV) with sFIS assay combined with Luminex (cytokines/soluble immune checkpoints), CA125-antigen detection, and whole-blood immune cell counts. Here, sFIS assay's ability to determine survival benefit or malignancy risk was validated in a discovery (n=32) and/or validation (n=699) patient cohorts. Lastly, we used an orthotopic murine metastatic OV model, with anti-OV therapy selection via in silico drug-target screening and murine serum screening via sFIS assay, to assess suitable in vivo immunotherapy options. Results In silico data-driven framework predicted that peripheral immunodynamics of patients with cancer might be best captured via analyzing myeloid nuclear factor kappa-light-chain enhancer of activated B cells (NF kappa B) signaling and interferon-stimulated genes' (ISG) responses. This helped in conceptualization of an 'in sitro' (in vitro+in situ) sFIS assay, where human myeloid cells were exposed to patients' serum in vitro, to assess serum-induced (si)-NF kappa B or interferon (IFN)/ISG responses (as active signaling reporter activity) within them, thereby 'mimicking' patients' in situ immunodynamic status. Multiparametric serum profiling of patients with OV established that sFIS assay can: decode peripheral immunology (by indicating higher enrichment of si-NF kappa B over si-IFN/ISG responses), estimate survival trends (si-NF kappa B or si-IFN/ISG responses associating with negative or positive prognosis, respectively), and coestimate malignancy risk (relative to benign/borderline ovarian lesions). Biologically, we documented dominance of pro-tumorigenic, myeloid si-NF kappa B response(HIGH)si-IFN/ISG response(LOW) inflammation in periphery of patients with OV. Finally, in an orthotopic murine metastatic OV model, sFIS assay predicted the higher capacity of chemo-immunotherapy (paclitaxel-carboplatin plus anti-TNF antibody combination) in achieving a pro-immunogenic peripheral milieu (si-IFN/ISG response(HIGH)si-NF kappa B response(LOW)), which aligned with high antitumor efficacy. Conclusions We established sFIS assay as a novel biomarker resource for serum screening in patients with OV to evaluate peripheral immunodynamics, patient survival trends and malignancy risk, and to design preclinical chemo-immunotherapy strategies.

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