DOT1L Is a Novel Cancer Stem Cell Target for Triple-Negative Breast Cancer

Author:

Kurani Hetakshi123,Razavipour Seyedeh Fatemeh3ORCID,Harikumar Kuzhuvelil B.14ORCID,Dunworth Matthew5ORCID,Ewald Andrew J.56ORCID,Nasir Apsra3,Pearson Gray3,Van Booven Derek7,Zhou Zhiqun1ORCID,Azzam Diana8ORCID,Wahlestedt Claes9ORCID,Slingerland Joyce3ORCID

Affiliation:

1. 1Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida.

2. 2Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida.

3. 3Breast Cancer Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, District of Columbia.

4. 4Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala, India.

5. 5Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland.

6. 6Cancer Invasion and Metastasis Program, Sidney Kimmel Comprehensive Cancer Center, and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.

7. 7John P. Hussman Institute of Human Genomics, University of Miami Miller School of Medicine, Miami, Florida.

8. 8Department of Environmental Health Sciences, Florida International University, Miami, Florida.

9. 9Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, Florida.

Abstract

Abstract Purpose: Although chemotherapies kill most cancer cells, stem cell–enriched survivors seed metastasis, particularly in triple-negative breast cancers (TNBC). TNBCs arise from and are enriched for tumor stem cells. Here, we tested if inhibition of DOT1L, an epigenetic regulator of normal tissue stem/progenitor populations, would target TNBC stem cells. Experimental Design: Effects of DOT1L inhibition by EPZ-5676 on stem cell properties were tested in three TNBC lines and four patient-derived xenograft (PDX) models and in isolated cancer stem cell (CSC)-enriched ALDH1+ and ALDH1− populations. RNA sequencing compared DOT1L regulated pathways in ALDH1+ and ALDH1− cells. To test if EPZ-5676 decreases CSC in vivo, limiting dilution assays of EPZ-5676/vehicle pretreated ALDH1+ and ALDH1− cells were performed. Tumor latency, growth, and metastasis were evaluated. Antitumor activity was also tested in TNBC PDX and PDX-derived organoids. Results: ALDH1+ TNBC cells exhibit higher DOT1L and H3K79me2 than ALDH1−. DOT1L maintains MYC expression and self-renewal in ALDH1+ cells. Global profiling revealed that DOT1L governs oxidative phosphorylation, cMyc targets, DNA damage response, and WNT activation in ALDH1+ but not in ALDH1− cells. EPZ-5676 reduced tumorspheres and ALDH1+ cells in vitro and decreased tumor-initiating stem cells and metastasis in xenografts generated from ALDH1+ but not ALDH1− populations in vivo. EPZ-5676 significantly reduced growth in vivo of one of two TNBC PDX tested and decreased clonogenic 3D growth of two other PDX-derived organoid cultures. Conclusions: DOT1L emerges as a key CSC regulator in TNBC. Present data support further clinical investigation of DOT1L inhibitors to target stem cell–enriched TNBC.

Funder

Breast Cancer Research Foundation

DOD | US Army | Medical Command | Congressionally Directed Medical Research Programs

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