Radiofrequency Ablation Remodels the Tumor Microenvironment and Promotes Neutrophil-Mediated Abscopal Immunomodulation in Pancreatic Cancer

Author:

Faraoni Erika Y.1ORCID,O'Brien Baylee J.1ORCID,Strickland Lincoln N.1ORCID,Osborn Baron K.2ORCID,Mota Victoria1ORCID,Chaney Jarod1ORCID,Atkins Constance Lynn1ORCID,Cen Putao3ORCID,Rowe Julie3ORCID,Cardenas Jessica2ORCID,Poulsen Kyle L.14ORCID,Wray Curtis J.2ORCID,Thosani Nirav C.5ORCID,Bailey-Lundberg Jennifer M.145ORCID

Affiliation:

1. 1Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.

2. 2Department of Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.

3. 3Division of Oncology, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.

4. 4Center for Perioperative Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.

5. 5Center for Interventional Gastroenterology at UTHealth (iGUT), McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.

Abstract

Abstract Pancreatic ductal adenocarcinoma (PDAC) presents a 5-year overall survival rate of 11%, despite efforts to improve clinical outcomes in the past two decades. Therapeutic resistance is a hallmark of this disease, due to its dense and suppressive tumor microenvironment (TME). Endoscopic ultrasound-guided radiofrequency ablation (EUS-RFA) is a promising local ablative and potential immunomodulatory therapy for PDAC. In this study, we performed RFA in a preclinical tumor-bearing KrasG12D; Trp53R172H/+; Pdx1:Cre (KPC) syngeneic model, analyzed local and abscopal affects after RFA and compared our findings with resected PDAC specimens. We found that RFA reduced PDAC tumor progression in vivo and promoted strong TME remodeling. In addition, we discovered tumor-infiltrating neutrophils determined abscopal effects. Using imaging mass cytometry, we showed that RFA elevated dendritic cell numbers in RFA-treated tumors and promoted a significant CD4+ and CD8+ T-cell abscopal response. In addition, RFA elevated levels of programmed death-ligand 1 (PD-L1) and checkpoint blockade inhibition targeting PD-L1 sustained tumor growth reduction in the context of RFA. This study indicates RFA treatment, which has been shown to increase tumor antigen shedding, promotes antitumor immunity. This is critical in PDAC where recent clinical immunotherapy trials have not resulted in substantial changes in overall survival.

Funder

National Institute on Alcohol Abuse and Alcoholism

National Institute of Diabetes and Digestive and Kidney Diseases

National Pancreas Foundation

National Cancer Institute

Publisher

American Association for Cancer Research (AACR)

Subject

Cancer Research,Immunology

Cited by 10 articles. 订阅此论文施引文献 订阅此论文施引文献,注册后可以免费订阅5篇论文的施引文献,订阅后可以查看论文全部施引文献

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3