Integrating multi-omics data to optimize immunotherapy in endometrial cancer: a comprehensive study.

IF 2.8 4区 医学 Q3 ENDOCRINOLOGY & METABOLISM
Xin Guan, Rongchuan Cao, Longbi Liu, Lin Ma, Ning Gao, Yanfei Yang, Mingyue Xiao, Rui Du, Yuzhe Su, Zhen Wang, Xiaofeng Liu, Lu Han
{"title":"Integrating multi-omics data to optimize immunotherapy in endometrial cancer: a comprehensive study.","authors":"Xin Guan, Rongchuan Cao, Longbi Liu, Lin Ma, Ning Gao, Yanfei Yang, Mingyue Xiao, Rui Du, Yuzhe Su, Zhen Wang, Xiaofeng Liu, Lu Han","doi":"10.1007/s12672-025-02978-2","DOIUrl":null,"url":null,"abstract":"<p><p>Immunotherapy represents a pivotal therapeutic modality in endometrial cancer (EC). Nevertheless, the efficacy of this treatment is limited to a subset of patients. The present investigation endeavors to amalgamate multi-omics data in order to elucidate the determinants impacting individual immune responsiveness and enhance the optimization of immunotherapy for EC. To differentiate EC patients into non-response (NR) and response (R), multi-omics data from publicly available databases were employed in conjunction with the TIDE computational framework. The validity of these findings was further confirmed through the utilization of the EaSIeR and ImmunoPhenoScore algorithms. The study employed functional enrichment and gene set variant analysis to discern noteworthy disparities in biological pathways across various groups. Moreover, three deconvolution algorithms (ESTIMATE, TIMER, and EPIC) were employed to quantify the tumor microenvironment (TME). Somatic mutation and copy number variant (CNV) analyses were also conducted to identify genomic alterations that impact immunotherapy. Integrated bulk and single-cell RNA sequencing (scRNA-seq) data were employed to identify cell populations linked to efficacy and deduce cell-cell interactions. The immunotherapy response rates were found to be greater in elderly EC patients aged 65 years and above. The NR group of patients displayed notable enrichment in cellular differentiation, angiogenesis, and tumor proliferation characteristics, as evidenced by higher tumor purity and lower expression of immune checkpoints. Conversely, the R group exhibited a stronger correlation with immunity, as indicated by pathway enrichment and composition of TME. Patients in the NR group demonstrated higher frequencies of somatic mutations, with a 2- to 6-fold disparity between the groups in genes such as RPRD1B and CTNNB1. Patients in the R group exhibited elevated mutation scores and higher mutation frequencies at the same mutation loci compared to those in the NR group. Moreover, the incidence of mutations was more prevalent among patients in the R group. In independent cohorts, the Scissor algorithm suggests that macrophages may exert a substantial impact on immunotherapy response in patients with EC. Subsequent analysis unveiled an enrichment of M2-like tumor-associated macrophages (TAMs) within the TME of patients in the NR group. These macrophages facilitate angiogenesis and cell proliferation through intercellular communication with subpopulations such as endothelial and epithelial cells. TME of patients in the R group exhibited an enrichment of M1-like TAMs, which primarily engaged with immune cells via diverse immune-activating factors. Furthermore, immunohistochemistry and flow cytometry demonstrated that responders to immunotherapy had significantly increased infiltration of M1-like TAMs. M1-like TAMs were shown to inhibit proliferation and migration of Ishikawa cells in co-culture assays. This research offers a comprehensive insight into the multi-omics level factors influencing the immunotherapy response of EC patients, emphasizing the influence of genomic variants and TAMs on said response. This contributes to an enhanced comprehension of the biological mechanisms underlying EC immunotherapy response and aids in the optimization of EC immunotherapy.</p>","PeriodicalId":11148,"journal":{"name":"Discover. Oncology","volume":"16 1","pages":"1161"},"PeriodicalIF":2.8000,"publicationDate":"2025-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12181619/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Discover. Oncology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1007/s12672-025-02978-2","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"ENDOCRINOLOGY & METABOLISM","Score":null,"Total":0}
引用次数: 0

Abstract

Immunotherapy represents a pivotal therapeutic modality in endometrial cancer (EC). Nevertheless, the efficacy of this treatment is limited to a subset of patients. The present investigation endeavors to amalgamate multi-omics data in order to elucidate the determinants impacting individual immune responsiveness and enhance the optimization of immunotherapy for EC. To differentiate EC patients into non-response (NR) and response (R), multi-omics data from publicly available databases were employed in conjunction with the TIDE computational framework. The validity of these findings was further confirmed through the utilization of the EaSIeR and ImmunoPhenoScore algorithms. The study employed functional enrichment and gene set variant analysis to discern noteworthy disparities in biological pathways across various groups. Moreover, three deconvolution algorithms (ESTIMATE, TIMER, and EPIC) were employed to quantify the tumor microenvironment (TME). Somatic mutation and copy number variant (CNV) analyses were also conducted to identify genomic alterations that impact immunotherapy. Integrated bulk and single-cell RNA sequencing (scRNA-seq) data were employed to identify cell populations linked to efficacy and deduce cell-cell interactions. The immunotherapy response rates were found to be greater in elderly EC patients aged 65 years and above. The NR group of patients displayed notable enrichment in cellular differentiation, angiogenesis, and tumor proliferation characteristics, as evidenced by higher tumor purity and lower expression of immune checkpoints. Conversely, the R group exhibited a stronger correlation with immunity, as indicated by pathway enrichment and composition of TME. Patients in the NR group demonstrated higher frequencies of somatic mutations, with a 2- to 6-fold disparity between the groups in genes such as RPRD1B and CTNNB1. Patients in the R group exhibited elevated mutation scores and higher mutation frequencies at the same mutation loci compared to those in the NR group. Moreover, the incidence of mutations was more prevalent among patients in the R group. In independent cohorts, the Scissor algorithm suggests that macrophages may exert a substantial impact on immunotherapy response in patients with EC. Subsequent analysis unveiled an enrichment of M2-like tumor-associated macrophages (TAMs) within the TME of patients in the NR group. These macrophages facilitate angiogenesis and cell proliferation through intercellular communication with subpopulations such as endothelial and epithelial cells. TME of patients in the R group exhibited an enrichment of M1-like TAMs, which primarily engaged with immune cells via diverse immune-activating factors. Furthermore, immunohistochemistry and flow cytometry demonstrated that responders to immunotherapy had significantly increased infiltration of M1-like TAMs. M1-like TAMs were shown to inhibit proliferation and migration of Ishikawa cells in co-culture assays. This research offers a comprehensive insight into the multi-omics level factors influencing the immunotherapy response of EC patients, emphasizing the influence of genomic variants and TAMs on said response. This contributes to an enhanced comprehension of the biological mechanisms underlying EC immunotherapy response and aids in the optimization of EC immunotherapy.

整合多组学数据优化子宫内膜癌免疫治疗:一项综合研究。
免疫治疗是子宫内膜癌(EC)的关键治疗方式。然而,这种治疗的疗效仅限于一小部分患者。本研究试图整合多组学数据,以阐明影响个体免疫反应的决定因素,并加强对EC免疫治疗的优化。为了将EC患者区分为无反应(NR)和反应(R),将来自公开可用数据库的多组学数据与TIDE计算框架结合使用。通过使用easy和ImmunoPhenoScore算法进一步证实了这些发现的有效性。该研究采用功能富集和基因集变异分析来辨别不同群体之间生物途径的显著差异。此外,采用三种反卷积算法(ESTIMATE, TIMER和EPIC)来量化肿瘤微环境(TME)。还进行了体细胞突变和拷贝数变异(CNV)分析,以确定影响免疫治疗的基因组改变。整合的整体和单细胞RNA测序(scRNA-seq)数据用于鉴定与疗效相关的细胞群并推断细胞间相互作用。免疫治疗应答率在65岁及以上的老年EC患者中更高。NR组患者在细胞分化、血管生成、肿瘤增殖等方面表现出明显的富集,肿瘤纯度较高,免疫检查点表达较低。相反,R组与免疫表现出更强的相关性,这与途径富集和TME组成有关。NR组的患者表现出更高的体细胞突变频率,在RPRD1B和CTNNB1等基因上,两组之间的差异为2至6倍。与NR组相比,R组患者在相同突变位点上表现出更高的突变评分和更高的突变频率。此外,突变发生率在R组患者中更为普遍。在独立队列中,剪刀算法提示巨噬细胞可能对EC患者的免疫治疗反应产生实质性影响。随后的分析揭示了NR组患者TME内m2样肿瘤相关巨噬细胞(tam)的富集。这些巨噬细胞通过与亚群(如内皮细胞和上皮细胞)的细胞间通讯促进血管生成和细胞增殖。R组患者的TME表现出m1样tam的富集,其主要通过多种免疫激活因子与免疫细胞结合。此外,免疫组织化学和流式细胞术显示免疫治疗应答者明显增加了m1样tam的浸润。在共培养实验中,m1样tam显示出抑制Ishikawa细胞的增殖和迁移。本研究全面了解了影响EC患者免疫治疗反应的多组学水平因素,强调了基因组变异和tam对免疫治疗反应的影响。这有助于增强对EC免疫治疗反应的生物学机制的理解,并有助于优化EC免疫治疗。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Discover. Oncology
Discover. Oncology Medicine-Endocrinology, Diabetes and Metabolism
CiteScore
2.40
自引率
9.10%
发文量
122
审稿时长
5 weeks
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信