Development and validation of multivariate predictors of primary endocrine resistance to tamoxifen and aromatase inhibitors in luminal breast cancer reveal drug-specific differences

Guokun Zhang, Vindi Jurinovic, Stephan Bartels, Matthias Christgen, Henriette Christgen, Leonie Donata Kandt, Mieke Raap, Janin Klein, Anna-Lena Katzke, Winfried Hofmann, Doris Steinemann, Ron Kates, Oleg Gluz, Monika Graeser, Sherko Kuemmel, Ulrike Nitz, Christoph Plass, Ulrich Lehmann, Ulrich Mansmann, Clarissa Gerhauser, Nadia Harbeck, Hans H. Kreipe
{"title":"Development and validation of multivariate predictors of primary endocrine resistance to tamoxifen and aromatase inhibitors in luminal breast cancer reveal drug-specific differences","authors":"Guokun Zhang, Vindi Jurinovic, Stephan Bartels, Matthias Christgen, Henriette Christgen, Leonie Donata Kandt, Mieke Raap, Janin Klein, Anna-Lena Katzke, Winfried Hofmann, Doris Steinemann, Ron Kates, Oleg Gluz, Monika Graeser, Sherko Kuemmel, Ulrike Nitz, Christoph Plass, Ulrich Lehmann, Ulrich Mansmann, Clarissa Gerhauser, Nadia Harbeck, Hans H. Kreipe","doi":"10.1101/2023.11.13.23298466","DOIUrl":null,"url":null,"abstract":"Background : Endocrine therapy is highly effective in blocking the estrogen receptor pathway in HR+/HER2- early breast cancer (EBC). However, up to 40% of patients experience relapse during or after adjuvant endocrine therapy. Here, we investigate molecular mechanisms associated with primary resistance to endocrine therapy and develop predictive models. Patients and Methods : In the WSG-ADAPT trial ( NCT01779206 ), HR+/HER2- EBC patients underwent pre-operative short-term endocrine therapy (pET). Treatment response was determined by immunohistochemical in-situ labeling of cycling cells (G1 to M-phase) with Ki67 before and after pET. We performed targeted next generation sequencing and Infinium MethylationEPIC-based DNA methylation analysis post-pET in a discovery cohort (n=364, responder (R) and non-responder (NR) pairs matched for clinicopathologic features) and a validation cohort (n=270, unmatched). Predictive indices of endocrine resistance under both treatments were constructed using lasso penalized logistic regression. A subset of breast cancers from The Cancer Genome Atlas project (TCGA-BRCA) was used for external validation. Results : TP53 mutations were prominently associated with primary resistance to both tamoxifen (TAM) and aromatase inhibitors (AI), with AI non-responders exhibiting resistance in up to 32% of cases. Additionally, we identified distinct DNA methylation patterns in TAM and AI non-responders, with TAM non-responders showing global DNA methylation loss, associated with KRAS signaling, apical junctions and epithelial-mesenchymal transition (EMT). Conversely, we observed methylation gain in AI non-responders affecting developmental transcription factors, hypoxia and estrogen signaling. TAM or AI resistance was associated with increased methylation-inferred proportions of immune cells and decreased proportions of endothelial cells. Based on these findings and patient age, we developed the Predictive Endocrine ResistanCe Index (PERCI). PERCI stratified NR and R cases in both treatment groups and cohorts with high accuracy (ROC AUC TAM discovery 93.9%, validation 83%; AI discovery 98.6%, validation 76.9%). A simplified PERCI efficiently predicted progression-free survival in the TCGA-BRCA sub-cohort (Kaplan-Meier log-rank p-value = 0.03 between low and high PERCI groups). Conclusions : We identified genomic and epigenomic features associated with primary resistance to TMA and AI. By combining information on genomic alterations, patient age, differential methylation and tumor microenvironment (TME) composition, we developed PERCI TAM and PERCI AI as novel predictors of primary resistance, with potential additional prognostic value. Applying PERCI in a clinical setting may allow patient-specific drug selection to overcome resistance. WSG-ADAPT, NCT01779206 , Registered 2013-01-25, retrospectively registered.","PeriodicalId":478577,"journal":{"name":"medRxiv (Cold Spring Harbor Laboratory)","volume":"71 6","pages":"0"},"PeriodicalIF":0.0000,"publicationDate":"2023-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"medRxiv (Cold Spring Harbor Laboratory)","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1101/2023.11.13.23298466","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

Background : Endocrine therapy is highly effective in blocking the estrogen receptor pathway in HR+/HER2- early breast cancer (EBC). However, up to 40% of patients experience relapse during or after adjuvant endocrine therapy. Here, we investigate molecular mechanisms associated with primary resistance to endocrine therapy and develop predictive models. Patients and Methods : In the WSG-ADAPT trial ( NCT01779206 ), HR+/HER2- EBC patients underwent pre-operative short-term endocrine therapy (pET). Treatment response was determined by immunohistochemical in-situ labeling of cycling cells (G1 to M-phase) with Ki67 before and after pET. We performed targeted next generation sequencing and Infinium MethylationEPIC-based DNA methylation analysis post-pET in a discovery cohort (n=364, responder (R) and non-responder (NR) pairs matched for clinicopathologic features) and a validation cohort (n=270, unmatched). Predictive indices of endocrine resistance under both treatments were constructed using lasso penalized logistic regression. A subset of breast cancers from The Cancer Genome Atlas project (TCGA-BRCA) was used for external validation. Results : TP53 mutations were prominently associated with primary resistance to both tamoxifen (TAM) and aromatase inhibitors (AI), with AI non-responders exhibiting resistance in up to 32% of cases. Additionally, we identified distinct DNA methylation patterns in TAM and AI non-responders, with TAM non-responders showing global DNA methylation loss, associated with KRAS signaling, apical junctions and epithelial-mesenchymal transition (EMT). Conversely, we observed methylation gain in AI non-responders affecting developmental transcription factors, hypoxia and estrogen signaling. TAM or AI resistance was associated with increased methylation-inferred proportions of immune cells and decreased proportions of endothelial cells. Based on these findings and patient age, we developed the Predictive Endocrine ResistanCe Index (PERCI). PERCI stratified NR and R cases in both treatment groups and cohorts with high accuracy (ROC AUC TAM discovery 93.9%, validation 83%; AI discovery 98.6%, validation 76.9%). A simplified PERCI efficiently predicted progression-free survival in the TCGA-BRCA sub-cohort (Kaplan-Meier log-rank p-value = 0.03 between low and high PERCI groups). Conclusions : We identified genomic and epigenomic features associated with primary resistance to TMA and AI. By combining information on genomic alterations, patient age, differential methylation and tumor microenvironment (TME) composition, we developed PERCI TAM and PERCI AI as novel predictors of primary resistance, with potential additional prognostic value. Applying PERCI in a clinical setting may allow patient-specific drug selection to overcome resistance. WSG-ADAPT, NCT01779206 , Registered 2013-01-25, retrospectively registered.
乳腺癌患者对他莫昔芬和芳香酶抑制剂原发性内分泌耐药的多变量预测因子的发展和验证揭示了药物特异性差异
背景:内分泌治疗在阻断HR+/HER2-早期乳腺癌(EBC)雌激素受体通路方面非常有效。然而,高达40%的患者在辅助内分泌治疗期间或之后复发。在这里,我们研究了与内分泌治疗原发性耐药相关的分子机制,并建立了预测模型。患者和方法:在WSG-ADAPT试验(NCT01779206)中,HR+/HER2- EBC患者接受术前短期内分泌治疗(pET)。用Ki67免疫组化原位标记pET前后循环细胞(G1期至m期),观察治疗效果。我们在发现队列(n=364,临床病理特征匹配的应答者(R)和非应答者(NR)对)和验证队列(n=270,未匹配)中进行了靶向下一代测序和基于Infinium methylationepic的DNA甲基化分析。采用套索惩罚logistic回归构建两种处理下的内分泌抗性预测指标。来自癌症基因组图谱计划(TCGA-BRCA)的乳腺癌子集被用于外部验证。结果:TP53突变与他莫昔芬(TAM)和芳香酶抑制剂(AI)的原发性耐药显著相关,在高达32%的病例中,AI无应答者表现出耐药。此外,我们在TAM和AI无应答者中发现了不同的DNA甲基化模式,TAM无应答者显示出全局DNA甲基化丢失,与KRAS信号、根尖连接和上皮-间质转化(EMT)相关。相反,我们观察到AI无应答者的甲基化增加会影响发育转录因子、缺氧和雌激素信号。TAM或AI耐药与甲基化推断的免疫细胞比例增加和内皮细胞比例减少有关。基于这些发现和患者年龄,我们开发了预测内分泌抵抗指数(PERCI)。两个治疗组和队列的分层NR和R病例具有较高的准确度(ROC AUC TAM发现率93.9%,验证率83%;AI发现98.6%,验证76.9%)。简化的PERCI有效地预测了TCGA-BRCA亚队列的无进展生存(低和高PERCI组之间的Kaplan-Meier log-rank p值= 0.03)。结论:我们确定了与TMA和AI原发耐药相关的基因组和表观基因组特征。通过结合基因组改变、患者年龄、差异甲基化和肿瘤微环境(TME)组成的信息,我们开发了PERCI TAM和PERCI AI作为原发性耐药的新预测指标,具有潜在的额外预后价值。在临床环境中应用PERCI可能允许针对患者的药物选择来克服耐药性。WSG-ADAPT, NCT01779206, 2013-01-25注册,追溯注册。
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 求助全文
来源期刊
自引率
0.00%
发文量
0
×
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
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信