NRF2 translation block by inhibition of cap-dependent initiation sensitizes lymphoma cells to ferroptosis and CAR-T immunotherapy

Paola Manara, Austin D Newsam, Venu VG Saralamma, Marco V Russo, Alicia Bilbao Martinez, Nikolai Fattakhov, Tyler A Cunningham, Abdessamad Y Alaoui, Dhanvantri Chahar, Alexandra M Carbone, Olivia B Lightfuss, Alexa M Barroso, Kyle S Hoffman, Francesco Maura, Daniel Bilbao, Jonathan H Schatz
{"title":"NRF2 translation block by inhibition of cap-dependent initiation sensitizes lymphoma cells to ferroptosis and CAR-T immunotherapy","authors":"Paola Manara, Austin D Newsam, Venu VG Saralamma, Marco V Russo, Alicia Bilbao Martinez, Nikolai Fattakhov, Tyler A Cunningham, Abdessamad Y Alaoui, Dhanvantri Chahar, Alexandra M Carbone, Olivia B Lightfuss, Alexa M Barroso, Kyle S Hoffman, Francesco Maura, Daniel Bilbao, Jonathan H Schatz","doi":"10.1101/2024.09.09.612133","DOIUrl":null,"url":null,"abstract":"Cancers coopt stress-response pathways to drive oncogenesis, dodge immune surveillance, and resist cytotoxic therapies. Several of these provide protection from ferroptosis, iron-mediated oxidative cell death. Here, we found dramatic sensitization to ferroptosis upon disruption of cap-dependent translation in diffuse large B-cell lymphoma (DLBCL). Specifically, rocaglate inhibitors of the eIF4A1 RNA helicase synergized with pharmacologic ferroptosis inducers, driven by a collapse of glutathione production that protects polyunsaturated fatty acids from ferroptotic oxidation. These effects occur despite initial up-regulation of specific protective factors. We find lost translation of NRF2, oncogenic master regulator of antioxidant gene-expression, is a key consequence of eIF4A1 inhibition. In vivo, combination of the clinical rocaglate zotatifin with a pharmacologically optimized ferroptosis inducer eradicated DLBCL patient derived xenografts. Moreover, we found zotatifin pre-exposure sensitized DLBCL to CD19-directed chimeric antigen receptor (CAR-19) T cells. Translational disruption therefore provides new opportunities to leverage therapeutic impacts of ferroptosis inducers including cytotoxic immunotherapies.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"bioRxiv - Cancer Biology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1101/2024.09.09.612133","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Cancers coopt stress-response pathways to drive oncogenesis, dodge immune surveillance, and resist cytotoxic therapies. Several of these provide protection from ferroptosis, iron-mediated oxidative cell death. Here, we found dramatic sensitization to ferroptosis upon disruption of cap-dependent translation in diffuse large B-cell lymphoma (DLBCL). Specifically, rocaglate inhibitors of the eIF4A1 RNA helicase synergized with pharmacologic ferroptosis inducers, driven by a collapse of glutathione production that protects polyunsaturated fatty acids from ferroptotic oxidation. These effects occur despite initial up-regulation of specific protective factors. We find lost translation of NRF2, oncogenic master regulator of antioxidant gene-expression, is a key consequence of eIF4A1 inhibition. In vivo, combination of the clinical rocaglate zotatifin with a pharmacologically optimized ferroptosis inducer eradicated DLBCL patient derived xenografts. Moreover, we found zotatifin pre-exposure sensitized DLBCL to CD19-directed chimeric antigen receptor (CAR-19) T cells. Translational disruption therefore provides new opportunities to leverage therapeutic impacts of ferroptosis inducers including cytotoxic immunotherapies.
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
通过抑制帽子依赖性起始阻断 NRF2 翻译,使淋巴瘤细胞对铁变态反应和 CAR-T 免疫疗法敏感
癌症利用应激反应途径驱动肿瘤发生,躲避免疫监视,抵制细胞毒疗法。其中有几种途径能保护细胞免受铁中毒(铁介导的氧化性细胞死亡)的伤害。在这里,我们发现在弥漫大 B 细胞淋巴瘤(DLBCL)中,当帽子依赖性翻译被破坏时,细胞会对铁氧化反应产生极大的敏感性。具体来说,eIF4A1 RNA 螺旋酶的洛卡格雷特抑制剂与药物性铁氧化诱导剂协同作用,谷胱甘肽的产生会崩溃,而谷胱甘肽能保护多不饱和脂肪酸不被铁氧化。尽管最初上调了特定的保护因子,但这些影响还是发生了。我们发现,抗氧化基因表达的致癌主调节因子 NRF2 的翻译丢失是 eIF4A1 抑制的一个关键后果。在体内,将临床罗卡酯佐他替芬与药理上优化的铁变态反应诱导剂结合使用,可根除DLBCL患者衍生的异种移植物。此外,我们还发现佐他替芬的预暴露可使DLBCL对CD19导向的嵌合抗原受体(CAR-19)T细胞敏感。因此,转化干扰为利用铁变态反应诱导剂(包括细胞毒性免疫疗法)的治疗效果提供了新的机会。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
自引率
0.00%
发文量
0
期刊最新文献
Craters on the melanoma surface facilitate tumor-immune interactions and demonstrate pathologic response to checkpoint blockade in humans DNFE: Directed-network flow entropy for detecting the tipping points during biological processes Transcriptional program-based deciphering of the MET exon 14 skipping regulation network Mutant p53 Misfolding and Aggregation Precedes Transformation into High-Grade Serous Ovarian Carcinoma Integrative multiomic approaches reveal ZMAT3 and p21 as conserved hubs in the p53 tumor suppression network
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1