DEVELOPMENT OF EX VIVO PATIENT-DERIVED MODELS TO UNCOVER THE TUMOR-IMMUNE MICROENVIRONMENT IN RENAL CELL CARCINOMA

IF 2.4 3区 医学 Q3 ONCOLOGY
Soki Kashima, Ruchi Gupta, Vivien Moritz, Katherine Sadak, Julia Walker, Adebowale Adeniran, Peter Humphrey, Daniel Dinulescu, Doug Palmer, Scott Hammond, Marcus Bosenberg, Michael Hurwitz, Patrick Kenney, David A. Braun
{"title":"DEVELOPMENT OF EX VIVO PATIENT-DERIVED MODELS TO UNCOVER THE TUMOR-IMMUNE MICROENVIRONMENT IN RENAL CELL CARCINOMA","authors":"Soki Kashima,&nbsp;Ruchi Gupta,&nbsp;Vivien Moritz,&nbsp;Katherine Sadak,&nbsp;Julia Walker,&nbsp;Adebowale Adeniran,&nbsp;Peter Humphrey,&nbsp;Daniel Dinulescu,&nbsp;Doug Palmer,&nbsp;Scott Hammond,&nbsp;Marcus Bosenberg,&nbsp;Michael Hurwitz,&nbsp;Patrick Kenney,&nbsp;David A. Braun","doi":"10.1016/j.urolonc.2024.12.057","DOIUrl":null,"url":null,"abstract":"<div><h3>Introduction</h3><div>With the rise of immune checkpoint inhibitors (ICIs) as the primary treatment option for metastatic renal cell carcinoma (RCC), investigating the role of T cells within the tumor microenvironment (TME) is a critical component of understanding both treatment response and resistance. Prior efforts, including single-cell transcriptomic approaches, have provided an important landscape of T cell transcriptional phenotypes. However, these immuno-profiling efforts require validation through functional interrogation of the TME to facilitate the development of novel immunomodulatory therapies. Thus, we established a patient-derived tumor model (PDTM) system to directly assess the effect of inhibitory immune interactions on T cell function and anti-tumor activity in the RCC TME. In this initial proof-of-concept study, we evaluated T cell activation in the RCC TME using the PDTM system.</div></div><div><h3>Methods</h3><div>Fresh tumor samples were obtained from surgical resections of RCC at Yale-New Haven Hospital. The tumor was minced to ∼1-3 mm³ pieces and suspended in an air-liquid interface system, consisting of tumor fragments embedded in a collagen matrix on an insert with a semi-permeable membrane, exposed to culture media. The tumor fragment and matrix suspension were carefully pipetted onto the Millicell insert, which served as the top layer. The PDTM setup includes an inner dish containing the bottom gel layer and the tissue-containing top layer. To complete the assembly, a low-dose T cell stimulant, 1.5 ml of DMEM media with or without 500 nM of anti-PD-1 monoclonal antibody (aPD1mAb) was added to the outer dish surrounding the insert.</div></div><div><h3>Results</h3><div>We successfully optimized PDTM experimental workflows for culture, dissociation, and analysis using immunohistochemistry (IHC), flow cytometry (FCM), and enzyme-linked immunosorbent assays (ELISA). Hematoxylin and eosin (H&amp;E) staining and IHC showed that the TME cellular architecture and immune cell composition was broadly preserved during the three-day experimental period. Using FCM to analyze the dissociated tumor samples, we identified well-preserved CA9+ tumor cells, CD4+ and CD8+ T cell populations, CD4+CD25+ regulatory T cells, CD56+ natural killer cells, CD20+ B cells, and CD14+CD11b+ myeloid subsets including monocytes and CD163-/+macrophages. Among the T cells, we detected PD1+, LAG3+, TIM3+, and TIGIT+ cells. We tested the effect of the anti-PD-1 antibody on PDTM, and importantly, found that treatment of the PDTM with aPD1mAb resulted in more activated CD8 T cells and higher IFN-γ production than the control samples.</div></div><div><h3>Conclusions</h3><div>Through optimization of assays evaluating T cell cytokine production, we were able to assess multiple axes of T cell function in the RCC TME. This study revealed that our PDTM system preserves the RCC TME for functional interrogation. Furthermore, our system for assessing T cell phenotype and cytokine production successfully demonstrated the activity of PD-1 blockade <em>ex vivo</em>. Taken together, this novel <em>ex vivo</em> PDTM system has extensive applications in the study of RCC, including assessing the impact of ICIs and new combination therapies on T cell function.</div></div>","PeriodicalId":23408,"journal":{"name":"Urologic Oncology-seminars and Original Investigations","volume":"43 3","pages":"Pages 22-23"},"PeriodicalIF":2.4000,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Urologic Oncology-seminars and Original Investigations","FirstCategoryId":"3","ListUrlMain":"https://www.sciencedirect.com/science/article/pii/S1078143924008378","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Introduction

With the rise of immune checkpoint inhibitors (ICIs) as the primary treatment option for metastatic renal cell carcinoma (RCC), investigating the role of T cells within the tumor microenvironment (TME) is a critical component of understanding both treatment response and resistance. Prior efforts, including single-cell transcriptomic approaches, have provided an important landscape of T cell transcriptional phenotypes. However, these immuno-profiling efforts require validation through functional interrogation of the TME to facilitate the development of novel immunomodulatory therapies. Thus, we established a patient-derived tumor model (PDTM) system to directly assess the effect of inhibitory immune interactions on T cell function and anti-tumor activity in the RCC TME. In this initial proof-of-concept study, we evaluated T cell activation in the RCC TME using the PDTM system.

Methods

Fresh tumor samples were obtained from surgical resections of RCC at Yale-New Haven Hospital. The tumor was minced to ∼1-3 mm³ pieces and suspended in an air-liquid interface system, consisting of tumor fragments embedded in a collagen matrix on an insert with a semi-permeable membrane, exposed to culture media. The tumor fragment and matrix suspension were carefully pipetted onto the Millicell insert, which served as the top layer. The PDTM setup includes an inner dish containing the bottom gel layer and the tissue-containing top layer. To complete the assembly, a low-dose T cell stimulant, 1.5 ml of DMEM media with or without 500 nM of anti-PD-1 monoclonal antibody (aPD1mAb) was added to the outer dish surrounding the insert.

Results

We successfully optimized PDTM experimental workflows for culture, dissociation, and analysis using immunohistochemistry (IHC), flow cytometry (FCM), and enzyme-linked immunosorbent assays (ELISA). Hematoxylin and eosin (H&E) staining and IHC showed that the TME cellular architecture and immune cell composition was broadly preserved during the three-day experimental period. Using FCM to analyze the dissociated tumor samples, we identified well-preserved CA9+ tumor cells, CD4+ and CD8+ T cell populations, CD4+CD25+ regulatory T cells, CD56+ natural killer cells, CD20+ B cells, and CD14+CD11b+ myeloid subsets including monocytes and CD163-/+macrophages. Among the T cells, we detected PD1+, LAG3+, TIM3+, and TIGIT+ cells. We tested the effect of the anti-PD-1 antibody on PDTM, and importantly, found that treatment of the PDTM with aPD1mAb resulted in more activated CD8 T cells and higher IFN-γ production than the control samples.

Conclusions

Through optimization of assays evaluating T cell cytokine production, we were able to assess multiple axes of T cell function in the RCC TME. This study revealed that our PDTM system preserves the RCC TME for functional interrogation. Furthermore, our system for assessing T cell phenotype and cytokine production successfully demonstrated the activity of PD-1 blockade ex vivo. Taken together, this novel ex vivo PDTM system has extensive applications in the study of RCC, including assessing the impact of ICIs and new combination therapies on T cell function.
建立离体模型揭示肾细胞癌的肿瘤免疫微环境
随着免疫检查点抑制剂(ICIs)作为转移性肾细胞癌(RCC)的主要治疗选择的兴起,研究T细胞在肿瘤微环境(TME)中的作用是了解治疗反应和耐药性的关键组成部分。先前的努力,包括单细胞转录组学方法,已经提供了T细胞转录表型的重要景观。然而,这些免疫分析工作需要通过对TME的功能询问来验证,以促进新型免疫调节疗法的发展。因此,我们建立了患者源性肿瘤模型(PDTM)系统,直接评估抑制免疫相互作用对RCC TME中T细胞功能和抗肿瘤活性的影响。在这项初步的概念验证研究中,我们使用PDTM系统评估了RCC TME中的T细胞活化。方法采用耶鲁-纽黑文医院肾细胞癌手术切除的新鲜肿瘤标本。将肿瘤切碎至~ 1-3 mm³,悬浮在气液界面系统中,该系统由嵌入胶原基质的肿瘤碎片组成,插入物带有半透膜,暴露于培养基中。将肿瘤碎片和基质悬浮液小心地移到Millicell插入物上,作为顶层。PDTM装置包括包含底部凝胶层和含有组织的顶层的内盘。为了完成组装,将低剂量T细胞刺激剂,1.5 ml含或不含500 nM抗pd -1单克隆抗体(aPD1mAb)的DMEM培养基加入到插入物周围的外皿中。结果通过免疫组织化学(IHC)、流式细胞术(FCM)和酶联免疫吸附试验(ELISA),我们成功地优化了PDTM的培养、分离和分析实验流程。苏木精和伊红(H&;E)染色和免疫组化显示,在3天的实验期间,TME的细胞结构和免疫细胞组成得到了广泛的保存。利用流式细胞仪分析分离的肿瘤样本,我们鉴定了保存完好的CA9+肿瘤细胞、CD4+和CD8+ T细胞群、CD4+CD25+调节性T细胞、CD56+自然杀伤细胞、CD20+ B细胞和CD14+CD11b+骨髓亚群,包括单核细胞和CD163-/+巨噬细胞。在T细胞中,我们检测到PD1+、LAG3+、TIM3+和TIGIT+细胞。我们测试了抗pd -1抗体对PDTM的作用,重要的是,发现用aPD1mAb处理PDTM导致比对照样品更多的活化CD8 T细胞和更高的IFN-γ产生。结论通过优化评估T细胞细胞因子产生的方法,我们能够评估RCC TME中T细胞功能的多个轴。本研究表明PDTM系统保留了RCC TME的功能询问。此外,我们用于评估T细胞表型和细胞因子产生的系统成功地证明了PD-1在体外阻断的活性。总之,这种新的离体PDTM系统在RCC的研究中有广泛的应用,包括评估ICIs和新的联合疗法对T细胞功能的影响。
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
CiteScore
4.80
自引率
3.70%
发文量
297
审稿时长
7.6 weeks
期刊介绍: Urologic Oncology: Seminars and Original Investigations is the official journal of the Society of Urologic Oncology. The journal publishes practical, timely, and relevant clinical and basic science research articles which address any aspect of urologic oncology. Each issue comprises original research, news and topics, survey articles providing short commentaries on other important articles in the urologic oncology literature, and reviews including an in-depth Seminar examining a specific clinical dilemma. The journal periodically publishes supplement issues devoted to areas of current interest to the urologic oncology community. Articles published are of interest to researchers and the clinicians involved in the practice of urologic oncology including urologists, oncologists, and radiologists.
×
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:481959085
Book学术官方微信