Unraveling the Mechanism of Epichaperome Modulation by Zelavespib: Biochemical Insights on Target Occupancy and Extended Residence Time at the Site of Action

Author:

Sharma Sahil1ORCID,Joshi Suhasini1,Kalidindi Teja2ORCID,Digwal Chander S.1ORCID,Panchal Palak1,Lee Sang-Gyu2,Zanzonico Pat2,Pillarsetty Nagavarakishore2ORCID,Chiosis Gabriela13

Affiliation:

1. Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA

2. Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA

3. Breast Cancer Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA

Abstract

Drugs with a long residence time at their target sites are often more efficacious in disease treatment. The mechanism, however, behind prolonged retention at the site of action is often difficult to understand for non-covalent agents. In this context, we focus on epichaperome agents, such as zelavespib and icapamespib, which maintain target binding for days despite rapid plasma clearance, minimal retention in non-diseased tissues, and rapid metabolism. They have shown significant therapeutic value in cancer and neurodegenerative diseases by disassembling epichaperomes, which are assemblies of tightly bound chaperones and other factors that serve as scaffolding platforms to pathologically rewire protein–protein interactions. To investigate their impact on epichaperomes in vivo, we conducted pharmacokinetic and target occupancy measurements for zelavespib and monitored epichaperome assemblies biochemically in a mouse model. Our findings provide evidence of the intricate mechanism through which zelavespib modulates epichaperomes in vivo. Initially, zelavespib becomes trapped when epichaperomes bound, a mechanism that results in epichaperome disassembly, with no change in the expression level of epichaperome constituents. We propose that the initial trapping stage of epichaperomes is a main contributing factor to the extended on-target residence time observed for this agent in clinical settings. Zelavespib’s residence time in tumors seems to be dictated by target disassembly kinetics rather than by frank drug–target unbinding kinetics. The off-rate of zelavespib from epichaperomes is, therefore, much slower than anticipated from the recorded tumor pharmacokinetic profile or as determined in vitro using diluted systems. This research sheds light on the underlying processes that make epichaperome agents effective in the treatment of certain diseases.

Funder

NIH

William H.

Alice Goodwin

Commonwealth Foundation for Cancer Research

Experimental Therapeutics Center of MSKCC

BrightFocus Foundation

Publisher

MDPI AG

Subject

General Biochemistry, Genetics and Molecular Biology,Medicine (miscellaneous)

Reference65 articles.

1. Can the flow of medicines be improved? Fundamental pharmacokinetic and pharmacological principles toward improving Phase II survival;Morgan;Drug Discov. Today,2012

2. Markman, M. (2023, July 07). Drugs Can–t Kill a Tumor They Can–t Reach. Medscape, 6 February 2013. Available online: https://www.medscape.com/viewarticle/778630.

3. Improving target assessment in biomedical research: The GOT-IT recommendations;Emmerich;Nat. Rev. Drug Discov.,2021

4. Turn On, Tune In, Turnover! Target Biology Impacts In Vivo Potency, Efficacy, and Clearance;Gabrielsson;Pharmacol. Rev.,2023

5. In vivo potency revisited—Keep the target in sight;Gabrielsson;Pharmacol. Ther.,2018

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