Abstract P5-04-19: Sulindac sulfide as a non-immune suppressive gamma secretase modifier to target triple negative breast cancer

Fokhrul M. Hossain, Deniz A. Ucar, Samarpan Majumder, M. Matossian, G. Monticone, Keli Xu, Yong Ran, L. Minter, Y. Xi, M. Burow, T. Golde, B. Osborne, L. Miele
{"title":"Abstract P5-04-19: Sulindac sulfide as a non-immune suppressive gamma secretase modifier to target triple negative breast cancer","authors":"Fokhrul M. Hossain, Deniz A. Ucar, Samarpan Majumder, M. Matossian, G. Monticone, Keli Xu, Yong Ran, L. Minter, Y. Xi, M. Burow, T. Golde, B. Osborne, L. Miele","doi":"10.1158/1538-7445.sabcs19-p5-04-19","DOIUrl":null,"url":null,"abstract":"Triple negative breast cancer (TNBC) is defined as pathologically negative for estrogen receptor (ER-), progesterone receptor (PR-), and human epidermal growth factor receptor 2 amplification (HER2-). TNBCs are a heterogeneous group of clinically aggressive cancers with high risk of recurrence and metastasis, and current treatment options remain limited. Immunotherapy with checkpoint inhibitors shows promise. However, recent data show that crosstalk between cancer stem cells (CSC) and the immune microenvironment leads to immunotherapy resistance, while myeloid-derived suppressor cells (MDSC) promote CSC survival via Notch signaling. Strong evidence supports the involvement of Notch, a prominent CSC pathway, in TNBC progression. Expression of Notch1 and its ligand Jagged1 correlate with poor prognosis. Notch inhibitors, Including Gamma Secretase Inhibitors (GSIs) are quite effective in preclinical models of TNBC. However the success of GSIs in clinical trials has been limited by their intestinal toxicity and potential for adverse immunological effects. Our overarching goal is to replace GSIs with agents that lack their systemic toxicity and adverse immunological effects. We identified Sulindac Sulfide (SS), the active metabolite of FDA-approved NSAID Sulindac, as a potential candidate to replace GSIs. We confirmed that SS has Gamma Secretase Modifier (GSM) activity, in addition to cyclo-oxygenase (COX) inhibition. SS inhibits Notch1 cleavage in TNBC cells, but not in murine T-cells. SS significantly inhibited mammospheres growth in all human and murine TNBC models we tested: 1) human MDA-MB-231 cells; 2) murine TNBC model C0321, from targeted conditional knockout of Lunatic Fringe (LFng-/-); and 3) Two TNBC patient-derived xenograft models, 2K1 and 4IC. In C0321 tumors in mice, we found that SS had remarkable single-agent anti-tumor activity and virtually eliminated Notch1 expression in tumors without intestinal toxicity. SS caused an increase in intra-tumoral CD11c+ dendritic cells and CD8 cells. SS did not affect the numbers of tumor infiltrating macrophages or myeloid-derived suppressor cells (MDSC). However, SS blocked the immunosuppressive function of bone marrow-derived MDSC. RNA-Sequencing of SS-treated tumors revealed significant reduction of CXCL14, EGR1, HOXC6, MAGI2, NCAM1, APOE, CLU (a Wnt target), DTX4 (an E3-ligase positive regulator of Notch activation), and TGFB3 genes and upregulation of CCL17, EPCAM, FABP4, C4A, LTF, ZBTB16, INADL, and FGFR2 genes. Importantly, SS enhanced the antitumor effect of a-PDL1 immunotherapy in our 0321 TNBC mouse model. Our data support further investigation of SS for the treatment of TNBC, with standard of care or with immunotherapy. Repurposing an FDA-approved, safe agent for the treatment of TNBC may be significantly easier and more cost-effective than developing unproven investigational agents. Citation Format: Fokhrul Hossain, Deniz A Ucar, Samarpan Majumder, Margarite Matossian, Giulia Monticone, Keli Xu, Yong Ran, Lisa Minter, Yaguang Xi, Matthew Burow, Todd Golde, Barbara Osborne, Lucio Miele. Sulindac sulfide as a non-immune suppressive gamma secretase modifier to target triple negative breast cancer [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr P5-04-19.","PeriodicalId":20307,"journal":{"name":"Poster Session Abstracts","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2020-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Poster Session Abstracts","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1158/1538-7445.sabcs19-p5-04-19","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

Triple negative breast cancer (TNBC) is defined as pathologically negative for estrogen receptor (ER-), progesterone receptor (PR-), and human epidermal growth factor receptor 2 amplification (HER2-). TNBCs are a heterogeneous group of clinically aggressive cancers with high risk of recurrence and metastasis, and current treatment options remain limited. Immunotherapy with checkpoint inhibitors shows promise. However, recent data show that crosstalk between cancer stem cells (CSC) and the immune microenvironment leads to immunotherapy resistance, while myeloid-derived suppressor cells (MDSC) promote CSC survival via Notch signaling. Strong evidence supports the involvement of Notch, a prominent CSC pathway, in TNBC progression. Expression of Notch1 and its ligand Jagged1 correlate with poor prognosis. Notch inhibitors, Including Gamma Secretase Inhibitors (GSIs) are quite effective in preclinical models of TNBC. However the success of GSIs in clinical trials has been limited by their intestinal toxicity and potential for adverse immunological effects. Our overarching goal is to replace GSIs with agents that lack their systemic toxicity and adverse immunological effects. We identified Sulindac Sulfide (SS), the active metabolite of FDA-approved NSAID Sulindac, as a potential candidate to replace GSIs. We confirmed that SS has Gamma Secretase Modifier (GSM) activity, in addition to cyclo-oxygenase (COX) inhibition. SS inhibits Notch1 cleavage in TNBC cells, but not in murine T-cells. SS significantly inhibited mammospheres growth in all human and murine TNBC models we tested: 1) human MDA-MB-231 cells; 2) murine TNBC model C0321, from targeted conditional knockout of Lunatic Fringe (LFng-/-); and 3) Two TNBC patient-derived xenograft models, 2K1 and 4IC. In C0321 tumors in mice, we found that SS had remarkable single-agent anti-tumor activity and virtually eliminated Notch1 expression in tumors without intestinal toxicity. SS caused an increase in intra-tumoral CD11c+ dendritic cells and CD8 cells. SS did not affect the numbers of tumor infiltrating macrophages or myeloid-derived suppressor cells (MDSC). However, SS blocked the immunosuppressive function of bone marrow-derived MDSC. RNA-Sequencing of SS-treated tumors revealed significant reduction of CXCL14, EGR1, HOXC6, MAGI2, NCAM1, APOE, CLU (a Wnt target), DTX4 (an E3-ligase positive regulator of Notch activation), and TGFB3 genes and upregulation of CCL17, EPCAM, FABP4, C4A, LTF, ZBTB16, INADL, and FGFR2 genes. Importantly, SS enhanced the antitumor effect of a-PDL1 immunotherapy in our 0321 TNBC mouse model. Our data support further investigation of SS for the treatment of TNBC, with standard of care or with immunotherapy. Repurposing an FDA-approved, safe agent for the treatment of TNBC may be significantly easier and more cost-effective than developing unproven investigational agents. Citation Format: Fokhrul Hossain, Deniz A Ucar, Samarpan Majumder, Margarite Matossian, Giulia Monticone, Keli Xu, Yong Ran, Lisa Minter, Yaguang Xi, Matthew Burow, Todd Golde, Barbara Osborne, Lucio Miele. Sulindac sulfide as a non-immune suppressive gamma secretase modifier to target triple negative breast cancer [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr P5-04-19.
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
摘要P5-04-19: Sulindac sulfide作为一种非免疫抑制性γ分泌酶修饰剂靶向三阴性乳腺癌
三阴性乳腺癌(TNBC)被定义为病理阴性雌激素受体(ER-)、孕激素受体(PR-)和人表皮生长因子受体2扩增(HER2-)。tnbc是一种异质性的临床侵袭性癌症,具有较高的复发和转移风险,目前的治疗选择仍然有限。免疫疗法与检查点抑制剂显示出希望。然而,最近的数据显示,癌症干细胞(CSC)和免疫微环境之间的串扰导致免疫治疗抵抗,而髓源性抑制细胞(MDSC)通过Notch信号通路促进CSC存活。强有力的证据支持Notch(一种突出的CSC通路)参与TNBC的进展。Notch1及其配体Jagged1的表达与预后不良相关。Notch抑制剂,包括γ分泌酶抑制剂(GSIs)在临床前TNBC模型中非常有效。然而,由于其肠道毒性和潜在的不良免疫效应,gsi在临床试验中的成功受到限制。我们的首要目标是用缺乏全身毒性和不良免疫效应的药物取代gsi。我们发现Sulindac Sulfide (SS)是fda批准的非甾体抗炎药Sulindac的活性代谢物,是替代GSIs的潜在候选药物。我们证实SS除了抑制环加氧酶(COX)外,还具有γ分泌酶修饰剂(GSM)活性。SS抑制TNBC细胞的Notch1切割,但对小鼠t细胞没有抑制作用。在我们测试的所有人类和小鼠TNBC模型中,SS显著抑制乳腺球体的生长:1)人MDA-MB-231细胞;2)小鼠TNBC模型C0321,来自有针对性的条件敲除Lunatic Fringe (lfg -/-);3)两种TNBC患者来源的异种移植模型,2K1和4IC。在小鼠C0321肿瘤中,我们发现SS具有显著的单药抗肿瘤活性,几乎消除了肿瘤中Notch1的表达,而没有肠道毒性。SS引起肿瘤内CD11c+树突状细胞和CD8细胞的增加。SS不影响肿瘤浸润性巨噬细胞或髓源性抑制细胞(MDSC)的数量。然而,SS阻断了骨髓源性MDSC的免疫抑制功能。ss治疗肿瘤的rna测序显示,CXCL14、EGR1、HOXC6、MAGI2、NCAM1、APOE、CLU (Wnt靶点)、DTX4 (Notch激活的e3连接酶阳性调节因子)和TGFB3基因显著降低,CCL17、EPCAM、FABP4、C4A、LTF、ZBTB16、INADL和FGFR2基因上调。重要的是,在我们的0321 TNBC小鼠模型中,SS增强了a-PDL1免疫治疗的抗肿瘤作用。我们的数据支持进一步研究SS治疗TNBC,标准护理或免疫治疗。重新利用fda批准的安全药物治疗TNBC可能比开发未经证实的研究性药物更容易和更具成本效益。硫化苏林达作为非免疫抑制性γ分泌酶修饰剂靶向三阴性乳腺癌[摘要]。摘自:2019年圣安东尼奥乳腺癌研讨会论文集;2019年12月10日至14日;费城(PA): AACR;中国癌症杂志,2020;31(增刊):P5-04-19。
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
Abstract PS6-01: How low is low risk: MINDACT updated outcome and treatment benefit in patients considered clinical low risk and stratified by genomic signature, age and nodal status Abstract PS14-01: Radium-223 in women with HR-positive bone-metastatic breast cancer receiving endocrine therapy: International phase 2, randomized, double-blind, placebo-controlled trial Abstract PS6-25: Cts5 tested in a Brazilian population: A tool that can predict global survival in early breast cancer ER+/HER2-, as well as the response to extended endocrine therapy Abstract PS1-32: A multicentre prospective feasibility study of carbon dye tattooing of biopsied axillary node and surgical localisation in breast cancer patients Abstract PS12-09: Pre-operative pembrolizumab (Pembro) with radiation therapy (RT) in patients with operable triple-negative breast cancer (TNBC)
×
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