Multidimensional Molecular Profiling of Metastatic Triple-Negative Breast Cancer and Immune Checkpoint Inhibitor Benefit

Author:

Barroso-Sousa Romualdo1ORCID,Forman Juliet234,Collier Katharine5,Weber Zachary T.5ORCID,Jammihal Tejas R.2ORCID,Kao Katrina Z.2,Richardson Edward T.6ORCID,Keenan Tanya2ORCID,Cohen Ofir2,Manos Michael P.2ORCID,Brennick Ryan C.2ORCID,Ott Patrick A.2,Hodi F. Stephen2,Dillon Deborah A.6,Attaya Victoria2,O'Meara Tess7ORCID,Lin Nancy U.28ORCID,Van Allen Eliezer M.2ORCID,Rodig Scott6,Winer Eric P.28ORCID,Mittendorf Elizabeth A.89ORCID,Wu Catherine J.2,Wagle Nikhil28ORCID,Stover Daniel G.5ORCID,Shukla Sachet A.234ORCID,Tolaney Sara M.28

Affiliation:

1. Oncology Center, Hospital Sírio-Libanês, Brasília, Brazil

2. Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA

3. Broad Institute of MIT and Harvard, Cambridge, MA

4. Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA

5. Ohio State University College of Medicine, Columbus, OH

6. Department of Pathology, Brigham and Women's Hospital, Boston, MA

7. Internal Medicine, Brigham and Women's Hospital, Boston, MA

8. Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA

9. Divison of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA

Abstract

PURPOSE In metastatic triple-negative breast cancer (mTNBC), consistent biomarkers of immune checkpoint inhibitor (ICI) therapy benefit remain elusive. We evaluated the immune, genomic, and transcriptomic landscape of mTNBC in patients treated with ICIs. METHODS We identified 29 patients with mTNBC treated with pembrolizumab or atezolizumab, either alone (n = 9) or in combination with chemotherapy (n = 14) or targeted therapy (n = 6), who had tumor tissue and/or blood available before ICI therapy for whole-exome sequencing. RNA sequencing and CIBERSORTx-inferred immune population analyses were performed (n = 20). Immune cell populations and programmed death-ligand 1 expression were assessed using multiplexed immunofluorescence (n = 18). Clonal trajectories were evaluated via serial tumor/circulating tumor DNA whole-exome sequencing (n = 4). Association of biomarkers with progression-free survival and overall survival (OS) was assessed. RESULTS Progression-free survival and OS were longer in patients with high programmed death-ligand 1 expression and tumor mutational burden. Patients with longer survival also had a higher relative inferred fraction of CD8+ T cells, activated CD4+ memory T cells, M1 macrophages, and follicular helper T cells and enrichment of inflammatory gene expression pathways. A mutational signature of defective repair of DNA damage by homologous recombination was enriched in patients with both shorter OS and primary resistance. Exploratory analysis of clonal evolution among four patients treated with programmed cell death protein 1 blockade and a tyrosine kinase inhibitor suggested that clonal stability post-treatment was associated with short time to progression. CONCLUSION This study identified potential biomarkers of response to ICIs among patients with mTNBC: high tumor mutational burden; presence of CD8+, CD4 memory T cells, follicular helper T cells, and M1 macrophages; and inflammatory gene expression pathways. Pretreatment deficiencies in the homologous recombination DNA damage repair pathway and the absence of or minimal clonal evolution post-treatment may be associated with worse outcomes.

Publisher

American Society of Clinical Oncology (ASCO)

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