Genomic screening reveals ubiquitin-like modifier activating enzyme 1 as a potent and druggable target in c-MYC-high triple negative breast cancer models

Author:

Jacob Sheeba1,Turner Tia H23,Cai Jinyang1,Floros Konstantinos V1ORCID,Yu Ann K1,Coon Colin M1,Khatri Rishabh1,Alzubi Mohammad A24,Jakubik Charles T5,Bouck Ynes M1,Puchalapalli Madhavi2ORCID,Shende Mayuri2,Dozmorov Mikhail G6ORCID,Boikos Sosipatros A7,Hu Bin2,Harrell J Chuck234,Benes Cyril H5ORCID,Koblinski Jennifer E2,Costa Carlotta5,Faber Anthony C1

Affiliation:

1. Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Virginia Commonwealth University , Richmond, VA 23298 , USA

2. Department of Pathology, Virginia Commonwealth University School of Medicine , Richmond, VA 23298 , USA

3. Wright Center for Clinical and Translational Research, Virginia Commonwealth University School of Medicine , Richmond, VA 23298 , USA

4. Integrative Life Sciences Program, Virginia Commonwealth University , Richmond, VA 23298 , USA

5. Center for Cancer Research, Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School , Boston, MA 02129 , USA

6. Department of Biostatistics, Virginia Commonwealth University , Richmond, VA 23298 , USA

7. Hematology, Oncology and Palliative Care, School of Medicine and Massey Cancer Center, Virginia Commonwealth University , Richmond, VA 23298 , USA

Abstract

Abstract Triple negative breast cancer (TNBC) accounts for over 30% of all breast cancer (BC)-related deaths, despite accounting for only 10% to 15% of total BC cases. Targeted therapy development has largely stalled in TNBC, underlined by a lack of traditionally druggable addictions like receptor tyrosine kinases (RTKs). Here, through full genome CRISPR/Cas9 screening of TNBC models, we have uncovered the sensitivity of TNBCs to the depletion of the ubiquitin-like modifier activating enzyme 1 (UBA1). Targeting UBA1 with the first-in-class UBA1 inhibitor TAK-243 induced unresolvable endoplasmic reticulum (ER)-stress and activating transcription factor 4 (ATF4)-mediated upregulation of proapoptotic NOXA, leading to cell death. c-MYC expression correlates with TAK-243 sensitivity and cooperates with TAK-243 to induce a stress response and cell death. Importantly, there was an order of magnitude greater sensitivity of TNBC lines to TAK-243 compared to normal tissue-derived cells. In five patient derived xenograft models (PDXs) of TNBC, TAK-243 therapy led to tumor inhibition or frank tumor regression. Moreover, in an intracardiac metastatic model of TNBC, TAK-243 markedly reduced metastatic burden, indicating UBA1 is a potential new target in TNBC expressing high levels of c-MYC.

Funder

National Institutes of Health

Publisher

Oxford University Press (OUP)

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