针对 ESR1 激活突变的疫苗可引起抗肿瘤免疫反应,并抑制耐药 ER+ 乳腺癌的雌激素信号传导。

IF 4.1 4区 医学 Q2 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Human Vaccines & Immunotherapeutics Pub Date : 2024-12-31 Epub Date: 2024-02-08 DOI:10.1080/21645515.2024.2309693
Gabrielle P Dailey, Christopher A Rabiola, Gangjun Lei, Junping Wei, Xiao-Yi Yang, Tao Wang, Cong-Xiao Liu, Melissa Gajda, Amy C Hobeika, Amanda Summers, Robert D Marek, Michael A Morse, Herbert K Lyerly, Erika J Crosby, Zachary C Hartman
{"title":"针对 ESR1 激活突变的疫苗可引起抗肿瘤免疫反应,并抑制耐药 ER+ 乳腺癌的雌激素信号传导。","authors":"Gabrielle P Dailey, Christopher A Rabiola, Gangjun Lei, Junping Wei, Xiao-Yi Yang, Tao Wang, Cong-Xiao Liu, Melissa Gajda, Amy C Hobeika, Amanda Summers, Robert D Marek, Michael A Morse, Herbert K Lyerly, Erika J Crosby, Zachary C Hartman","doi":"10.1080/21645515.2024.2309693","DOIUrl":null,"url":null,"abstract":"<p><p>ER+ breast cancers (BC) are characterized by the elevated expression and signaling of estrogen receptor alpha (<i>ESR1)</i>, which renders them sensitive to anti-endocrine therapy. While these therapies are clinically effective, prolonged treatment inevitably results in therapeutic resistance, which can occur through the emergence of gain-of-function mutations in <i>ESR1</i>. The central importance of <i>ESR1</i> and development of mutated forms of <i>ESR1</i> suggest that vaccines targeting these proteins could potentially be effective in preventing or treating endocrine resistance. To explore the potential of this approach, we developed several recombinant vaccines encoding different mutant forms of <i>ESR1</i> (<i>ESR1</i>mut) and validated their ability to elicit <i>ESR1</i>-specific T cell responses. We then developed novel <i>ESR1</i>mut-expressing murine mammary cancer models to test the anti-tumor potential of <i>ESR1</i>mut vaccines. We found that these vaccines could suppress tumor growth, <i>ESR1</i>mut expression and estrogen signaling in vivo. To illustrate the applicability of these findings, we utilize HPLC to demonstrate the presentation of <i>ESR1</i> and <i>ESR1</i>mut peptides on human ER+ BC cell MHC complexes. We then show the presence of human T cells reactive to <i>ESR1</i>mut epitopes in an ER+ BC patient. These findings support the development of <i>ESR1</i>mut vaccines, which we are testing in a Phase I clinical trial.</p>","PeriodicalId":49067,"journal":{"name":"Human Vaccines & Immunotherapeutics","volume":null,"pages":null},"PeriodicalIF":4.1000,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10857653/pdf/","citationCount":"0","resultStr":"{\"title\":\"Vaccines targeting <i>ESR1</i> activating mutations elicit anti-tumor immune responses and suppress estrogen signaling in therapy resistant ER+ breast cancer.\",\"authors\":\"Gabrielle P Dailey, Christopher A Rabiola, Gangjun Lei, Junping Wei, Xiao-Yi Yang, Tao Wang, Cong-Xiao Liu, Melissa Gajda, Amy C Hobeika, Amanda Summers, Robert D Marek, Michael A Morse, Herbert K Lyerly, Erika J Crosby, Zachary C Hartman\",\"doi\":\"10.1080/21645515.2024.2309693\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>ER+ breast cancers (BC) are characterized by the elevated expression and signaling of estrogen receptor alpha (<i>ESR1)</i>, which renders them sensitive to anti-endocrine therapy. While these therapies are clinically effective, prolonged treatment inevitably results in therapeutic resistance, which can occur through the emergence of gain-of-function mutations in <i>ESR1</i>. The central importance of <i>ESR1</i> and development of mutated forms of <i>ESR1</i> suggest that vaccines targeting these proteins could potentially be effective in preventing or treating endocrine resistance. To explore the potential of this approach, we developed several recombinant vaccines encoding different mutant forms of <i>ESR1</i> (<i>ESR1</i>mut) and validated their ability to elicit <i>ESR1</i>-specific T cell responses. We then developed novel <i>ESR1</i>mut-expressing murine mammary cancer models to test the anti-tumor potential of <i>ESR1</i>mut vaccines. We found that these vaccines could suppress tumor growth, <i>ESR1</i>mut expression and estrogen signaling in vivo. To illustrate the applicability of these findings, we utilize HPLC to demonstrate the presentation of <i>ESR1</i> and <i>ESR1</i>mut peptides on human ER+ BC cell MHC complexes. We then show the presence of human T cells reactive to <i>ESR1</i>mut epitopes in an ER+ BC patient. These findings support the development of <i>ESR1</i>mut vaccines, which we are testing in a Phase I clinical trial.</p>\",\"PeriodicalId\":49067,\"journal\":{\"name\":\"Human Vaccines & Immunotherapeutics\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":4.1000,\"publicationDate\":\"2024-12-31\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10857653/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Human Vaccines & Immunotherapeutics\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1080/21645515.2024.2309693\",\"RegionNum\":4,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2024/2/8 0:00:00\",\"PubModel\":\"Epub\",\"JCR\":\"Q2\",\"JCRName\":\"BIOTECHNOLOGY & APPLIED MICROBIOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Human Vaccines & Immunotherapeutics","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1080/21645515.2024.2309693","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/2/8 0:00:00","PubModel":"Epub","JCR":"Q2","JCRName":"BIOTECHNOLOGY & APPLIED MICROBIOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

ER+乳腺癌(BC)的特点是雌激素受体α(ESR1)的表达和信号传导升高,这使它们对抗内分泌治疗敏感。虽然这些疗法在临床上很有效,但长期治疗不可避免地会产生耐药性,而耐药性的产生可能是由于 ESR1 发生了功能增益突变。ESR1的核心重要性和ESR1突变形式的发展表明,针对这些蛋白的疫苗有可能有效预防或治疗内分泌耐药性。为了探索这种方法的潜力,我们开发了几种编码不同突变形式 ESR1(ESR1mut)的重组疫苗,并验证了它们诱导 ESR1 特异性 T 细胞应答的能力。然后,我们开发了新型 ESR1mut 表达小鼠乳腺癌模型,以测试 ESR1mut 疫苗的抗肿瘤潜力。我们发现这些疫苗可以抑制体内肿瘤生长、ESR1mut 表达和雌激素信号传导。为了说明这些发现的适用性,我们利用高效液相色谱法证明了 ESR1 和 ESR1mut 肽在人 ER+ BC 细胞 MHC 复合物上的呈现。然后,我们在一名ER+ BC患者体内发现了对ESR1mut表位有反应的人类T细胞。这些发现支持了 ESR1mut 疫苗的开发,我们正在对其进行 I 期临床试验。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
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
Vaccines targeting ESR1 activating mutations elicit anti-tumor immune responses and suppress estrogen signaling in therapy resistant ER+ breast cancer.

ER+ breast cancers (BC) are characterized by the elevated expression and signaling of estrogen receptor alpha (ESR1), which renders them sensitive to anti-endocrine therapy. While these therapies are clinically effective, prolonged treatment inevitably results in therapeutic resistance, which can occur through the emergence of gain-of-function mutations in ESR1. The central importance of ESR1 and development of mutated forms of ESR1 suggest that vaccines targeting these proteins could potentially be effective in preventing or treating endocrine resistance. To explore the potential of this approach, we developed several recombinant vaccines encoding different mutant forms of ESR1 (ESR1mut) and validated their ability to elicit ESR1-specific T cell responses. We then developed novel ESR1mut-expressing murine mammary cancer models to test the anti-tumor potential of ESR1mut vaccines. We found that these vaccines could suppress tumor growth, ESR1mut expression and estrogen signaling in vivo. To illustrate the applicability of these findings, we utilize HPLC to demonstrate the presentation of ESR1 and ESR1mut peptides on human ER+ BC cell MHC complexes. We then show the presence of human T cells reactive to ESR1mut epitopes in an ER+ BC patient. These findings support the development of ESR1mut vaccines, which we are testing in a Phase I clinical trial.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Human Vaccines & Immunotherapeutics
Human Vaccines & Immunotherapeutics BIOTECHNOLOGY & APPLIED MICROBIOLOGY-IMMUNOLOGY
CiteScore
7.90
自引率
8.30%
发文量
489
审稿时长
3-6 weeks
期刊介绍: (formerly Human Vaccines; issn 1554-8619) Vaccine research and development is extending its reach beyond the prevention of bacterial or viral diseases. There are experimental vaccines for immunotherapeutic purposes and for applications outside of infectious diseases, in diverse fields such as cancer, autoimmunity, allergy, Alzheimer’s and addiction. Many of these vaccines and immunotherapeutics should become available in the next two decades, with consequent benefit for human health. Continued advancement in this field will benefit from a forum that can (A) help to promote interest by keeping investigators updated, and (B) enable an exchange of ideas regarding the latest progress in the many topics pertaining to vaccines and immunotherapeutics. Human Vaccines & Immunotherapeutics provides such a forum. It is published monthly in a format that is accessible to a wide international audience in the academic, industrial and public sectors.
期刊最新文献
Wilms' tumor 1 -targeting cancer vaccine: Recent advancements and future perspectives. Toll-like receptor agonists as cancer vaccine adjuvants. Hotspot areas of tetanus-unprotected births and its associated factors in Ethiopia: Spatial analysis of EDHS data. How well does vaccine literacy predict intention to vaccinate and vaccination status? A systematic review and meta-analysis. Hypophysitis after COVID-19 vaccination in a patient with Rathke's cleft cyst: A case report.
×
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
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