Reversal of Lactate and PD-1–mediated Macrophage Immunosuppression Controls Growth of PTEN/p53-deficient Prostate Cancer

Author:

Chaudagar Kiranj1ORCID,Hieromnimon Hanna M.1ORCID,Khurana Rimpi2ORCID,Labadie Brian1ORCID,Hirz Taghreed345ORCID,Mei Shenglin36ORCID,Hasan Raisa78ORCID,Shafran Jordan1ORCID,Kelley Anne1ORCID,Apostolov Eva78ORCID,Al-Eryani Ghamdan78ORCID,Harvey Kate7ORCID,Rameshbabu Srikrishnan1ORCID,Loyd Mayme1ORCID,Bynoe Kaela1ORCID,Drovetsky Catherine1ORCID,Solanki Ani9ORCID,Markiewicz Erica10ORCID,Zamora Marta10ORCID,Fan Xiaobing10ORCID,Schürer Stephan211ORCID,Swarbrick Alex78ORCID,Sykes David B.345ORCID,Patnaik Akash1ORCID

Affiliation:

1. 1Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois.

2. 2Department of Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida.

3. 3Center for Regenerative Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts.

4. 4Harvard Stem Cell Institute, Cambridge, Massachusetts.

5. 5Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts.

6. 6Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts.

7. 7Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.

8. 8St Vincent's Clinical School, Faculty of Medicine and Health, UNSW Sydney, Kensington, New South Wales, Australia.

9. 9Animal Resource Center, University of Chicago, Chicago, Illinois.

10. 10Department of Radiology, University of Chicago, Chicago, Illinois.

11. 11Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida.

Abstract

Abstract Purpose: Phosphatase and tensin homolog (PTEN) loss of function occurs in approximately 50% of patients with metastatic castrate-resistant prostate cancer (mCRPC), and is associated with poor prognosis and responsiveness to standard-of-care therapies and immune checkpoint inhibitors. While PTEN loss of function hyperactivates PI3K signaling, combinatorial PI3K/AKT pathway and androgen deprivation therapy (ADT) has demonstrated limited anticancer efficacy in clinical trials. Here, we aimed to elucidate mechanism(s) of resistance to ADT/PI3K-AKT axis blockade, and to develop rational combinatorial strategies to effectively treat this molecular subset of mCRPC. Experimental Design: Prostate-specific PTEN/p53-deficient genetically engineered mice (GEM) with established 150–200 mm3 tumors, as assessed by ultrasound, were treated with either ADT (degarelix), PI3K inhibitor (copanlisib), or anti–PD-1 antibody (aPD-1), as single agents or their combinations, and tumors were monitored by MRI and harvested for immune, transcriptomic, and proteomic profiling, or ex vivo co-culture studies. Single-cell RNA sequencing on human mCRPC samples was performed using 10X Genomics platform. Results: Coclinical trials in PTEN/p53-deficient GEM revealed that recruitment of PD-1–expressing tumor-associated macrophages (TAM) thwarts ADT/PI3Ki combination–induced tumor control. The addition of aPD-1 to ADT/PI3Ki combination led to TAM-dependent approximately 3-fold increase in anticancer responses. Mechanistically, decreased lactate production from PI3Ki-treated tumor cells suppressed histone lactylation within TAM, resulting in their anticancer phagocytic activation, which was augmented by ADT/aPD-1 treatment and abrogated by feedback activation of Wnt/β-catenin pathway. Single-cell RNA-sequencing analysis in mCRPC patient biopsy samples revealed a direct correlation between high glycolytic activity and TAM phagocytosis suppression. Conclusions: Immunometabolic strategies that reverse lactate and PD-1–mediated TAM immunosuppression, in combination with ADT, warrant further investigation in patients with PTEN-deficient mCRPC.

Funder

Prostate Cancer Foundation

National Cancer Institute

National Heart, Lung, and Blood Institute

State of Florida Biomedical Research Program, Bankhead Cley grant

Publisher

American Association for Cancer Research (AACR)

Subject

Cancer Research,Oncology

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