Targeted Degradation of XIAP is Sufficient and Specific to Induce Apoptosis in MYCN-overexpressing High-risk Neuroblastoma

Author:

Choo Zhang'E12ORCID,Koh Xiaoying3ORCID,Wong Megan Rui En4ORCID,Ashokan Ruth Minothini12ORCID,Ali Ahamed Nurul Suhana Binte12ORCID,Kang CongBao3ORCID,Kuick Chik Hong5ORCID,Chang Kenneth Tou En456ORCID,Larisch Sarit7ORCID,Loh Amos Hong Pheng4678ORCID,Chen Zhi Xiong1249ORCID

Affiliation:

1. 1Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.

2. 2NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.

3. 3Experimental Drug Development Centre, A*STAR, Singapore.

4. 4VIVA-KKH Paediatric Brain and Solid Tumour Programme, Children's Blood and Cancer Centre, KK Women's and Children's Hospital, Singapore.

5. 5Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore.

6. 6Duke NUS Medical School, Singapore.

7. 7Cell Death and Cancer Research Laboratory, Department of Human Biology and Medical Sciences, University of Haifa, Haifa, Israel.

8. 8Department of Paediatric Surgery, KK Women's and Children's Hospital, Singapore.

9. 9National University Cancer Institute, Singapore, National University Health System, Singapore.

Abstract

Abstract XIAP, the most potent mammalian inhibitor of apoptosis protein (IAP), critically restricts developmental culling of sympathetic neuronal progenitors, and is correspondingly overexpressed in most MYCN-amplified neuroblastoma tumors. Because apoptosis-related protein in the TGFβ signaling pathway (ARTS) is the only XIAP antagonist that directly binds and degrades XIAP, we evaluated the preclinical effectiveness and tolerability of XIAP antagonism as a novel targeting strategy for neuroblastoma. We found that antagonism of XIAP, but not other IAPs, triggered apoptotic death in neuroblastoma cells. XIAP silencing induced apoptosis while overexpression conferred protection from drug-induced apoptosis. From a screen of IAP inhibitors, first-in-class ARTS mimetic A4 was most effective against high-risk and high XIAP-expressing neuroblastoma cells, and least toxic toward normal liver- and bone marrow–derived cells, compared with pan-IAP antagonists. On target engagement assays and nuclear magnetic resonance spectroscopy, A4 was observed to degrade rather than inhibit XIAP, catalyzing rapid degradation of XIAP through the ubiquitin-proteasome pathway. In MYCN-amplified neuroblastoma patient-derived xenografts, A4 significantly prolonged survival as a single agent, and demonstrated synergism with standard-of-care agents to reduce their effective required doses 3- to 6-fold. Engagement and degradation of XIAP by ARTS mimetics is a novel targeting strategy for neuroblastoma that may be especially effective against MYCN-amplified disease with intrinsically high XIAP expression. First-in-class ARTS mimetic A4 demonstrates preclinical efficacy and warrants further development and study. Significance: XIAP degradation is sufficient to kill MYCN-amplified neuroblastoma which overexpresses and relies on XIAP as a brake against cell death, without affecting normal cells.

Funder

VIVA Foundation for Children with Cancer

Children's Cancer Foundation

Agency for Science, Technology and Research

MOH | National Medical Research Council

Publisher

American Association for Cancer Research (AACR)

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