首页 > 最新文献

Resistance Mechanisms最新文献

英文 中文
Abstract A47: Long-term treatment of bortezomib reduced resistance to doxorubicin by reducing CerS6/GCS and elevating CerS2/GBA expressions 摘要A47:长期治疗硼替佐米通过降低CerS6/GCS和升高CerS2/GBA表达来降低对阿霉素的耐药性
Pub Date : 2018-08-01 DOI: 10.1158/1557-3125.ADVBC17-A47
Kim Shin, Kim Dongeun, Park Inkeun, Park Woo-Jae
Multiple drug resistance (MDR) is main cause of chemotherapy failure in breast cancer. Herein, we tried to find the mechanism of MDR. Since overexpression of glucosylceramide synthase (GCS) plays an important role in development of MDR, we analyzed expression levels of many molecules from TCGA data to find a connection between GCS and other molecules. Interestingly, correlations between GCS and ceramide synthase 6 (CerS6), and between glucocerebosidase (GBA) and CerS2, 4, 5 were analyzed. To further analyze whether these correlations are true, we treated C16~C24-ceramide to MD-MBA-361 cells. C24-ceramide treatment for 48 and 72 hrs increased GBA expression, which increased susceptibility of apoptosis upon doxorubicin treatment. Moreover, C16-ceramide treatment for 16 weeks increased GCS expression, which also induced MDR1 protein expression. Recently we found that bortezomib, an FDA-approved proteasome inhibitor, increased CerS2 and decreased CerS6 expressions in a dose-dependent manner. To further understand whether bortezomib affects MDR, bortezomib was treated with doxorubicin for 6-8 months and it prevented doxorubicin-induced MDR development by reducing CerS6 and elevating CerS2. In conclusion, downregulation of CerS6 and upregulation of CerS2 is a good strategy to prevent development of MDR, and bortezomib can be used for the prevention of acquired MDR. Citation Format: Kim Shin, Kim Dongeun, Park Inkeun, Park Woo-Jae. Long-term treatment of bortezomib reduced resistance to doxorubicin by reducing CerS6/GCS and elevating CerS2/GBA expressions [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Cancer Research; 2017 Oct 7-10; Hollywood, CA. Philadelphia (PA): AACR; Mol Cancer Res 2018;16(8_Suppl):Abstract nr A47.
多重耐药(MDR)是乳腺癌化疗失败的主要原因。在此,我们试图找到耐多药的机制。由于葡萄糖神经酰胺合成酶(glucosylceramide synthase, GCS)的过表达在MDR的发展中起着重要作用,我们从TCGA数据中分析了许多分子的表达水平,以寻找GCS与其他分子之间的联系。有趣的是,研究人员分析了GCS与神经酰胺合成酶6 (CerS6)、葡萄糖糖苷酶(GBA)与CerS2、4,5之间的相关性。为了进一步分析这些相关性是否成立,我们将C16~ c24 -神经酰胺作用于MD-MBA-361细胞。c24 -神经酰胺处理48和72小时后,GBA表达增加,增加了阿霉素处理后细胞凋亡的易感性。此外,c16 -神经酰胺处理16周后,GCS表达增加,也诱导了MDR1蛋白的表达。最近,我们发现fda批准的蛋白酶体抑制剂硼替佐米(bortezomib)以剂量依赖的方式增加CerS2并降低CerS6的表达。为了进一步了解硼替佐米是否影响MDR,我们将硼替佐米与阿霉素联合治疗6-8个月,通过降低CerS6和提高CerS2来预防阿霉素诱导的MDR的发生。综上所述,下调CerS6和上调CerS2是预防MDR发展的良好策略,硼替佐米可用于预防获得性MDR。引文格式:金信,金东根,朴仁根,朴宇宰。长期治疗硼替佐米通过降低CerS6/GCS和升高CerS2/GBA表达来降低对阿霉素的耐药[摘要]。摘自:AACR特别会议论文集:乳腺癌研究进展;2017年10月7-10日;费城(PA): AACR;中华肿瘤杂志,2018;16(8):1 - 7。
{"title":"Abstract A47: Long-term treatment of bortezomib reduced resistance to doxorubicin by reducing CerS6/GCS and elevating CerS2/GBA expressions","authors":"Kim Shin, Kim Dongeun, Park Inkeun, Park Woo-Jae","doi":"10.1158/1557-3125.ADVBC17-A47","DOIUrl":"https://doi.org/10.1158/1557-3125.ADVBC17-A47","url":null,"abstract":"Multiple drug resistance (MDR) is main cause of chemotherapy failure in breast cancer. Herein, we tried to find the mechanism of MDR. Since overexpression of glucosylceramide synthase (GCS) plays an important role in development of MDR, we analyzed expression levels of many molecules from TCGA data to find a connection between GCS and other molecules. Interestingly, correlations between GCS and ceramide synthase 6 (CerS6), and between glucocerebosidase (GBA) and CerS2, 4, 5 were analyzed. To further analyze whether these correlations are true, we treated C16~C24-ceramide to MD-MBA-361 cells. C24-ceramide treatment for 48 and 72 hrs increased GBA expression, which increased susceptibility of apoptosis upon doxorubicin treatment. Moreover, C16-ceramide treatment for 16 weeks increased GCS expression, which also induced MDR1 protein expression. Recently we found that bortezomib, an FDA-approved proteasome inhibitor, increased CerS2 and decreased CerS6 expressions in a dose-dependent manner. To further understand whether bortezomib affects MDR, bortezomib was treated with doxorubicin for 6-8 months and it prevented doxorubicin-induced MDR development by reducing CerS6 and elevating CerS2. In conclusion, downregulation of CerS6 and upregulation of CerS2 is a good strategy to prevent development of MDR, and bortezomib can be used for the prevention of acquired MDR. Citation Format: Kim Shin, Kim Dongeun, Park Inkeun, Park Woo-Jae. Long-term treatment of bortezomib reduced resistance to doxorubicin by reducing CerS6/GCS and elevating CerS2/GBA expressions [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Cancer Research; 2017 Oct 7-10; Hollywood, CA. Philadelphia (PA): AACR; Mol Cancer Res 2018;16(8_Suppl):Abstract nr A47.","PeriodicalId":20897,"journal":{"name":"Resistance Mechanisms","volume":"5 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2018-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"81774490","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract B36: Progesterone receptor signaling in estrogen receptor-positive breast cancer B36:雌激素受体阳性乳腺癌的孕激素受体信号
Pub Date : 2018-08-01 DOI: 10.1158/1557-3125.ADVBC17-B36
Amy E. Young, Jane Guan, A. Daemen, L. Friedman, Kui Lin
Approximately 65% of breast cancers are estrogen receptor alpha (ERα)-positive and depend on ERα signaling for growth. Clinical data indicate that ERα-positive breast cancers have a better prognosis when another hormone receptor, the progesterone receptor (PR), is coexpressed. Recent studies show that in the presence of its ligand progesterone, PR redirects ERα chromatin binding and transcriptional activity, thereby impacting prognosis and therapeutic response. To further examine PR function, we knocked out the PGR gene in the ERα-positive breast cancer cell lines MCF-7 and T47D using CRISPR-mediated gene editing. Knockout clones from each model were analyzed for ER- and PR-mediated transcriptional activity and gene expression profile, as well as response to selective estrogen receptor degraders (SERDs). The T47D cell line expresses high levels of PR in the presence or absence of estrogen, and stimulation with progesterone induces robust PR phosphorylation and modulation of PR target gene expression. Furthermore, progesterone attenuates estradiol-induced cell proliferation in a dose-dependent manner. Knockout of PR in this model abrogates the ability of progesterone to induce gene expression changes or attenuate estradiol-induced cell proliferation. In contrast, the MCF-7 cell line expresses low basal levels of PR, and is refractory to stimulation with progesterone. Interestingly, in estrogenic growth media, knockout of PR in this model results in basal gene expression changes that resemble a gene signature associated with tamoxifen resistance. Consistent with these findings, knockout of PR reduces the response to SERDs in a 5-day cell proliferation assay. ChIP-seq and RNA-seq analyses are ongoing to examine how loss of PR impacts ERα chromatin binding and transcriptional output. Citation Format: Amy Young, Jane Guan, Anneleen Daemen, Lori Friedman, Kui Lin. Progesterone receptor signaling in estrogen receptor-positive breast cancer [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Cancer Research; 2017 Oct 7-10; Hollywood, CA. Philadelphia (PA): AACR; Mol Cancer Res 2018;16(8_Suppl):Abstract nr B36.
大约65%的乳腺癌是雌激素受体α (ERα)阳性,并依赖于ERα信号的生长。临床资料表明,当另一种激素受体孕激素受体(PR)共表达时,er α阳性乳腺癌预后较好。最近的研究表明,在其配体黄体酮的存在下,PR可重定向ERα染色质结合和转录活性,从而影响预后和治疗反应。为了进一步研究PR的功能,我们利用crispr介导的基因编辑技术敲除er α阳性乳腺癌细胞系MCF-7和T47D中的PGR基因。分析每个模型的敲除克隆的ER和pr介导的转录活性和基因表达谱,以及对选择性雌激素受体降解物(serd)的反应。T47D细胞系在雌激素存在或不存在的情况下均表达高水平的PR,黄体酮刺激可诱导PR磷酸化并调节PR靶基因的表达。此外,黄体酮以剂量依赖的方式减弱雌二醇诱导的细胞增殖。在该模型中,敲除PR可消除黄体酮诱导基因表达改变或减弱雌二醇诱导的细胞增殖的能力。相比之下,MCF-7细胞系表达低基础水平的PR,并且对黄体酮刺激不耐受。有趣的是,在雌激素生长培养基中,在该模型中敲除PR导致基础基因表达变化,类似于与他莫昔芬耐药相关的基因特征。与这些发现一致,在为期5天的细胞增殖试验中,敲除PR降低了对SERDs的反应。ChIP-seq和RNA-seq分析正在研究PR缺失如何影响ERα染色质结合和转录输出。引文格式:Amy Young, Jane Guan, Anneleen Daemen, Lori Friedman, Kui Lin。雌激素受体阳性乳腺癌的孕激素受体信号传导[摘要]。摘自:AACR特别会议论文集:乳腺癌研究进展;2017年10月7-10日;费城(PA): AACR;癌症学报,2018;16(8 -增刊):摘要nr B36。
{"title":"Abstract B36: Progesterone receptor signaling in estrogen receptor-positive breast cancer","authors":"Amy E. Young, Jane Guan, A. Daemen, L. Friedman, Kui Lin","doi":"10.1158/1557-3125.ADVBC17-B36","DOIUrl":"https://doi.org/10.1158/1557-3125.ADVBC17-B36","url":null,"abstract":"Approximately 65% of breast cancers are estrogen receptor alpha (ERα)-positive and depend on ERα signaling for growth. Clinical data indicate that ERα-positive breast cancers have a better prognosis when another hormone receptor, the progesterone receptor (PR), is coexpressed. Recent studies show that in the presence of its ligand progesterone, PR redirects ERα chromatin binding and transcriptional activity, thereby impacting prognosis and therapeutic response. To further examine PR function, we knocked out the PGR gene in the ERα-positive breast cancer cell lines MCF-7 and T47D using CRISPR-mediated gene editing. Knockout clones from each model were analyzed for ER- and PR-mediated transcriptional activity and gene expression profile, as well as response to selective estrogen receptor degraders (SERDs). The T47D cell line expresses high levels of PR in the presence or absence of estrogen, and stimulation with progesterone induces robust PR phosphorylation and modulation of PR target gene expression. Furthermore, progesterone attenuates estradiol-induced cell proliferation in a dose-dependent manner. Knockout of PR in this model abrogates the ability of progesterone to induce gene expression changes or attenuate estradiol-induced cell proliferation. In contrast, the MCF-7 cell line expresses low basal levels of PR, and is refractory to stimulation with progesterone. Interestingly, in estrogenic growth media, knockout of PR in this model results in basal gene expression changes that resemble a gene signature associated with tamoxifen resistance. Consistent with these findings, knockout of PR reduces the response to SERDs in a 5-day cell proliferation assay. ChIP-seq and RNA-seq analyses are ongoing to examine how loss of PR impacts ERα chromatin binding and transcriptional output. Citation Format: Amy Young, Jane Guan, Anneleen Daemen, Lori Friedman, Kui Lin. Progesterone receptor signaling in estrogen receptor-positive breast cancer [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Cancer Research; 2017 Oct 7-10; Hollywood, CA. Philadelphia (PA): AACR; Mol Cancer Res 2018;16(8_Suppl):Abstract nr B36.","PeriodicalId":20897,"journal":{"name":"Resistance Mechanisms","volume":"15 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2018-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"72733826","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract A45: Kinome rewiring reveals AURKA is a molecular barrier to the efficacy of PI3K/mTOR-pathway inhibitors in breast cancer 摘要:Kinome rewiring揭示AURKA是PI3K/ mtor通路抑制剂在乳腺癌中的作用的分子屏障
Pub Date : 2018-08-01 DOI: 10.1158/1557-3125.ADVBC17-A45
Hayley J. Donnella, James T. Webber, K. Shokat, A. Goga, J. Gordan, Sourav Bandyopadhyay
Dysregulation of the PI3K-AKT-mTOR signaling network is a prominent feature of breast cancers. However, clinical responses to drugs targeting this pathway have been modest. We hypothesized that dynamic changes in signaling, including adaptation and feedback, limit drug efficacy. Using a quantitative proteomics approach, we mapped dynamic changes in the kinome in response to various agents and identified signaling changes that correlate with drug sensitivity. Measurement of dynamics across a panel of breast cancer cell lines identified that maintenance of CDK4 and AURKA activity was associated with drug resistance. We tested whether incomplete inhibition of CDK4 or AURKA was a source of therapy failure and found that inhibition of either was sufficient to sensitize most breast cancer cells to PI3K, AKT, and mTOR inhibitors. In particular, drug combinations including the AURKA inhibitor MLN8237 were highly synergistic and induced apoptosis through enhanced suppression of mTOR signaling to S6 and 4E-BP1 leading to tumor regression in vivo. This approach identifies survival factors whose presence limits the efficacy of target therapy and suggests that Aurora kinase coinhibition could unlock the full potential of PI3K-AKT-mTOR pathway inhibitors in breast cancer. Citation Format: Hayley Donnella, James Webber, Kevan Shokat, Andrei Goga, John Gordan, Sourav Bandyopadhyay. Kinome rewiring reveals AURKA is a molecular barrier to the efficacy of PI3K/mTOR-pathway inhibitors in breast cancer [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Cancer Research; 2017 Oct 7-10; Hollywood, CA. Philadelphia (PA): AACR; Mol Cancer Res 2018;16(8_Suppl):Abstract nr A45.
PI3K-AKT-mTOR信号网络的失调是乳腺癌的一个突出特征。然而,针对这一途径的药物的临床反应一直很温和。我们假设信号的动态变化,包括适应和反馈,限制了药物的疗效。使用定量蛋白质组学方法,我们绘制了响应各种药物的激酶组的动态变化,并确定了与药物敏感性相关的信号变化。对一组乳腺癌细胞系的动态测量发现,CDK4和AURKA活性的维持与耐药性有关。我们测试了CDK4或AURKA的不完全抑制是否是治疗失败的原因,并发现任何一种抑制都足以使大多数乳腺癌细胞对PI3K、AKT和mTOR抑制剂敏感。特别是,包括AURKA抑制剂MLN8237在内的药物组合具有高度协同作用,并通过增强对S6和4E-BP1的mTOR信号的抑制,诱导细胞凋亡,从而导致体内肿瘤消退。该方法确定了存在限制靶向治疗疗效的生存因素,并表明极光激酶共抑制可以释放PI3K-AKT-mTOR途径抑制剂在乳腺癌中的全部潜力。引用格式:Hayley Donnella, James Webber, Kevan Shokat, Andrei Goga, John Gordan, Sourav Bandyopadhyay。Kinome重布线表明AURKA是PI3K/ mtor通路抑制剂在乳腺癌中的作用的分子屏障[摘要]。摘自:AACR特别会议论文集:乳腺癌研究进展;2017年10月7-10日;费城(PA): AACR;中华肿瘤杂志,2018;16(8):1 - 5。
{"title":"Abstract A45: Kinome rewiring reveals AURKA is a molecular barrier to the efficacy of PI3K/mTOR-pathway inhibitors in breast cancer","authors":"Hayley J. Donnella, James T. Webber, K. Shokat, A. Goga, J. Gordan, Sourav Bandyopadhyay","doi":"10.1158/1557-3125.ADVBC17-A45","DOIUrl":"https://doi.org/10.1158/1557-3125.ADVBC17-A45","url":null,"abstract":"Dysregulation of the PI3K-AKT-mTOR signaling network is a prominent feature of breast cancers. However, clinical responses to drugs targeting this pathway have been modest. We hypothesized that dynamic changes in signaling, including adaptation and feedback, limit drug efficacy. Using a quantitative proteomics approach, we mapped dynamic changes in the kinome in response to various agents and identified signaling changes that correlate with drug sensitivity. Measurement of dynamics across a panel of breast cancer cell lines identified that maintenance of CDK4 and AURKA activity was associated with drug resistance. We tested whether incomplete inhibition of CDK4 or AURKA was a source of therapy failure and found that inhibition of either was sufficient to sensitize most breast cancer cells to PI3K, AKT, and mTOR inhibitors. In particular, drug combinations including the AURKA inhibitor MLN8237 were highly synergistic and induced apoptosis through enhanced suppression of mTOR signaling to S6 and 4E-BP1 leading to tumor regression in vivo. This approach identifies survival factors whose presence limits the efficacy of target therapy and suggests that Aurora kinase coinhibition could unlock the full potential of PI3K-AKT-mTOR pathway inhibitors in breast cancer. Citation Format: Hayley Donnella, James Webber, Kevan Shokat, Andrei Goga, John Gordan, Sourav Bandyopadhyay. Kinome rewiring reveals AURKA is a molecular barrier to the efficacy of PI3K/mTOR-pathway inhibitors in breast cancer [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Cancer Research; 2017 Oct 7-10; Hollywood, CA. Philadelphia (PA): AACR; Mol Cancer Res 2018;16(8_Suppl):Abstract nr A45.","PeriodicalId":20897,"journal":{"name":"Resistance Mechanisms","volume":"2 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2018-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"77636685","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Abstract A42: Identifying relationships between high expression levels of the HSPA9 gene, putative HSPA9 alterations, and patient survivability in invasive breast carcinomas 摘要:探讨浸润性乳腺癌中HSPA9基因的高表达水平、HSPA9可能的改变与患者存活率之间的关系
Pub Date : 2018-08-01 DOI: 10.1158/1557-3125.ADVBC17-A42
Kofi K. Khamit-Kush, J. Lillard, V. Bond, Ming-bo Huang, W. Roth
Metastasis is the primary cause for the lethality of breast cancer, and is responsible for approximately 90% of breast cancer-related deaths. Tumor cell resistance to cancer treatment continues to stymie efforts to effectively treat breast cancer, which we know to be primarily mediated by a highly conserved molecular chaperone in the heat shock protein 70 family known as mortalin. Mortalin is encoded by the gene HSPA9B localized on chromosome 5q31.1.1, and is low or undetectable in normal unstressed cells while highly expressed in many carcinomas. Breast cancer cell invasion and metastasis are closely related to adverse clinical outcomes and a worsened prognosis for patients. Previous studies have shown that breast cancer patients with high mortalin expression had decreased DFS and OS rates compared to those with low mortalin expression in early- or late-stage breast cancer. Conversely, low expression of mortalin decreases tumor cell progression and inhibits the epithelial-mesenchymal transition. Gene amplification is one of the major pathways by which proto-oncogenes are activated during tumorigenesis, and missense mutations in the gene of interest could potentially render a nonfunctional mortalin product. We examined three different publicly available datasets from the Genome Data Commons for possible relationships between alterations in the HSPA9 gene and BRCA patient survival rates. On a mutation-specific level, we compared the survival rates of the cases with a high frequency of HSPA9 alterations versus the cases without HSPA9 alterations. Our bioinformatics analysis of the BRCA datasets showed fourteen alterations in the HSPA9 gene that correlated to prognosis and survival rate in patients with breast cancer. Amplifications in the HSPA9 gene lead to lower survivability rates for the patient samples, while missense mutations in HSPA9 led to higher survivability rates. Also of interest, missense mutations were far more numerous than any other HSPA9 alteration type. Different alterations in the HSPA9 gene result in different mortalin protein products, ultimately leading to differences in prognosis for patients with invasive breast carcinomas. Our study supports that much of the clinically observable plasticity and heterogeneity occurs within, rather than across, the major biologic subtypes of breast cancer suggested by the variance in HSPA9 alteration types. Furthermore, the study supports that mortalin expression has a significant effect on breast cancer status and lends credence to mortalin as a survival predictor, particularly in BRCA patients. Note: This abstract was not presented at the conference. Citation Format: Kofi K. Khamit-Kush, James W. Lillard, Jr., Vincent C. Bond, Ming Huang, William Roth. Identifying relationships between high expression levels of the HSPA9 gene, putative HSPA9 alterations, and patient survivability in invasive breast carcinomas [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Can
转移是导致乳腺癌死亡的主要原因,约90%的乳腺癌相关死亡是由转移引起的。肿瘤细胞对癌症治疗的耐药性继续阻碍着有效治疗乳腺癌的努力,我们知道乳腺癌主要是由热休克蛋白70家族中一种高度保守的分子伴侣蛋白介导的,这种分子伴侣蛋白被称为死亡蛋白。Mortalin由位于染色体5q31.1.1上的HSPA9B基因编码,在正常的非应激细胞中含量低或检测不到,而在许多肿瘤中高表达。乳腺癌细胞的侵袭和转移与患者的不良临床结局和预后恶化密切相关。先前的研究表明,在早期或晚期乳腺癌中,与死亡率低的乳腺癌患者相比,死亡率高的乳腺癌患者的DFS和OS率降低。相反,低表达的死亡率降低肿瘤细胞的进展和抑制上皮-间质转化。基因扩增是原癌基因在肿瘤发生过程中被激活的主要途径之一,而相关基因的错义突变可能会导致无功能的致死性蛋白产物。我们检查了来自基因组数据共享的三个不同的公开数据集,以寻找HSPA9基因改变与BRCA患者存活率之间的可能关系。在突变特异性水平上,我们比较了HSPA9突变频率高的病例与没有HSPA9突变的病例的存活率。我们对BRCA数据集的生物信息学分析显示,HSPA9基因的14个改变与乳腺癌患者的预后和生存率相关。HSPA9基因的扩增导致患者样本的存活率较低,而HSPA9基因的错义突变导致存活率较高。同样有趣的是,错义突变比任何其他HSPA9变异类型都要多。HSPA9基因的不同改变导致不同的致死性蛋白产物,最终导致浸润性乳腺癌患者预后的差异。我们的研究支持,临床观察到的可塑性和异质性发生在乳腺癌的主要生物学亚型内,而不是跨越HSPA9变异类型的差异。此外,该研究支持mortalin表达对乳腺癌状态有显著影响,并为mortalin作为生存预测因子提供了依据,特别是在BRCA患者中。注:本摘要未在会议上发表。引文格式:Kofi K. khamitt - kush, James W. Lillard, Jr., Vincent C. Bond, Ming Huang, William Roth。确定HSPA9基因高表达水平、推测的HSPA9改变与浸润性乳腺癌患者存活率之间的关系[摘要]。摘自:AACR特别会议论文集:乳腺癌研究进展;2017年10月7-10日;费城(PA): AACR;中华肿瘤杂志,2018;16(8):1 - 2。
{"title":"Abstract A42: Identifying relationships between high expression levels of the HSPA9 gene, putative HSPA9 alterations, and patient survivability in invasive breast carcinomas","authors":"Kofi K. Khamit-Kush, J. Lillard, V. Bond, Ming-bo Huang, W. Roth","doi":"10.1158/1557-3125.ADVBC17-A42","DOIUrl":"https://doi.org/10.1158/1557-3125.ADVBC17-A42","url":null,"abstract":"Metastasis is the primary cause for the lethality of breast cancer, and is responsible for approximately 90% of breast cancer-related deaths. Tumor cell resistance to cancer treatment continues to stymie efforts to effectively treat breast cancer, which we know to be primarily mediated by a highly conserved molecular chaperone in the heat shock protein 70 family known as mortalin. Mortalin is encoded by the gene HSPA9B localized on chromosome 5q31.1.1, and is low or undetectable in normal unstressed cells while highly expressed in many carcinomas. Breast cancer cell invasion and metastasis are closely related to adverse clinical outcomes and a worsened prognosis for patients. Previous studies have shown that breast cancer patients with high mortalin expression had decreased DFS and OS rates compared to those with low mortalin expression in early- or late-stage breast cancer. Conversely, low expression of mortalin decreases tumor cell progression and inhibits the epithelial-mesenchymal transition. Gene amplification is one of the major pathways by which proto-oncogenes are activated during tumorigenesis, and missense mutations in the gene of interest could potentially render a nonfunctional mortalin product. We examined three different publicly available datasets from the Genome Data Commons for possible relationships between alterations in the HSPA9 gene and BRCA patient survival rates. On a mutation-specific level, we compared the survival rates of the cases with a high frequency of HSPA9 alterations versus the cases without HSPA9 alterations. Our bioinformatics analysis of the BRCA datasets showed fourteen alterations in the HSPA9 gene that correlated to prognosis and survival rate in patients with breast cancer. Amplifications in the HSPA9 gene lead to lower survivability rates for the patient samples, while missense mutations in HSPA9 led to higher survivability rates. Also of interest, missense mutations were far more numerous than any other HSPA9 alteration type. Different alterations in the HSPA9 gene result in different mortalin protein products, ultimately leading to differences in prognosis for patients with invasive breast carcinomas. Our study supports that much of the clinically observable plasticity and heterogeneity occurs within, rather than across, the major biologic subtypes of breast cancer suggested by the variance in HSPA9 alteration types. Furthermore, the study supports that mortalin expression has a significant effect on breast cancer status and lends credence to mortalin as a survival predictor, particularly in BRCA patients. Note: This abstract was not presented at the conference. Citation Format: Kofi K. Khamit-Kush, James W. Lillard, Jr., Vincent C. Bond, Ming Huang, William Roth. Identifying relationships between high expression levels of the HSPA9 gene, putative HSPA9 alterations, and patient survivability in invasive breast carcinomas [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Can","PeriodicalId":20897,"journal":{"name":"Resistance Mechanisms","volume":"13 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2018-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"87995081","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Abstract B34: Novel synergistic combination therapies with BET bromodomain inhibitors in triple-negative breast cancer B34: BET溴结构域抑制剂联合治疗三阴性乳腺癌的新协同疗法
Pub Date : 2018-08-01 DOI: 10.1158/1557-3125.ADVBC17-B34
Samantha M. Bevill, N. Sciaky, B. Golitz, N. Rashid, J. Zawistowski, G. Johnson
Adaptive resistance to targeted cancer therapies is a universal problem in cancer treatment where tumor cells circumvent targeted pathway inhibition to reactivate growth signaling. In our previous work, we observed genome-wide enhancer remodeling following MEK inhibition (MEKi) capable of driving adaptive gene transcription in triple-negative breast cancer (TNBC). Adaptive enhancers were enriched for the BET bromodomain protein BRD4 and cotreatment with MEKi and BET inhibitor (JQ1) could durably suppress TNBC growth in multiple cell lines and preclinical mouse models. There are currently 10 BET inhibitors in clinical trials being tested as single agents across multiple tumor types including TNBC. There is also a growing body of preclinical literature using epigenetic inhibitors to block the adaptive ability of tumor cells in combination with multiple targeted therapies. This led us to screen for inhibitors that synergize with JQ1 to suppress growth of TNBC using a 176-compound library enriched for epigenetic and kinase inhibitors. We performed synergy screens in 6 TNBC cell lines across 6 doses of JQ1 and each library compound. Using the Bliss Independence model to assess synergy, we found that inhibition of MEK, CDK9, Aurora Kinase, CREBBP/P300, and BAZ2A/B was strongly synergistic with JQ1. BRD4, CDK9, and the acetyltransferase CREBBP/P300 are all members of the P-TEFb transcriptional elongation complex. When we performed additional synergy screens against the P300 bromodomain inhibitor CPI-637, we found a significant overlap in synergistic targets with the JQ1 screen including MEK, BET bromodomain proteins, ERK, Aurora Kinase, and CDK9. BAZ2A/B inhibition using the small-molecule inhibitor GSK2801, which targets the bromodomain of BAZ2A/B, synergized significantly stronger with JQ1 across all cell lines compared to CPI637. BAZ2A/B proteins are members of nucleosome remodeling complexes that mediate DNA silencing by aiding in recruitment of histone modifying enzymes. Ongoing studies seek to understand the role of BAZ2A/B and the mechanism of GSK2801 synergy with BET bromodomain inhibition using RNA sequencing and ChIP sequencing experiments. These results define novel targets that synergize with JQ1 to suppress tumor cell growth and illuminate additional mechanisms of transcriptional regulation driven by BET bromodomain proteins in TNBC. Citation Format: Samantha M. Bevill, Noah Sciaky, Brian T. Golitz, Naim U. Rashid, Jon S. Zawistowski, Gary L. Johnson. Novel synergistic combination therapies with BET bromodomain inhibitors in triple-negative breast cancer [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Cancer Research; 2017 Oct 7-10; Hollywood, CA. Philadelphia (PA): AACR; Mol Cancer Res 2018;16(8_Suppl):Abstract nr B34.
靶向癌症治疗的适应性耐药是癌症治疗中的一个普遍问题,肿瘤细胞绕过靶向途径抑制来重新激活生长信号。在我们之前的工作中,我们观察到三阴性乳腺癌(TNBC)中MEK抑制(MEKi)后的全基因组增强子重塑能够驱动适应性基因转录。在多种细胞系和临床前小鼠模型中,与MEKi和BET抑制剂(JQ1)共处理的适应性增强子富集了BET溴结构域蛋白BRD4,可以持久抑制TNBC的生长。目前有10种BET抑制剂正在临床试验中作为单一药物治疗多种肿瘤类型,包括TNBC。也有越来越多的临床前文献使用表观遗传抑制剂来阻断肿瘤细胞的适应能力,并结合多种靶向治疗。这促使我们筛选与JQ1协同抑制TNBC生长的抑制剂,使用一个富含表观遗传和激酶抑制剂的176个化合物文库。我们在6个TNBC细胞系中进行了6个剂量的JQ1和每个文库化合物的协同筛选。利用Bliss独立模型评估协同作用,我们发现MEK、CDK9、极光激酶、CREBBP/P300和BAZ2A/B的抑制与JQ1具有很强的协同作用。BRD4、CDK9和乙酰转移酶CREBBP/P300都是P-TEFb转录延伸复合物的成员。当我们对P300溴结构域抑制剂pci -637进行额外的协同筛选时,我们发现与JQ1筛选的协同靶点有显著的重叠,包括MEK、BET溴结构域蛋白、ERK、极光激酶和CDK9。使用靶向BAZ2A/B溴域的小分子抑制剂GSK2801抑制BAZ2A/B,与CPI637相比,在所有细胞系中与JQ1的协同作用明显更强。BAZ2A/B蛋白是核小体重塑复合体的成员,通过帮助募集组蛋白修饰酶介导DNA沉默。正在进行的研究试图通过RNA测序和ChIP测序实验了解BAZ2A/B的作用以及GSK2801协同BET溴域抑制的机制。这些结果确定了与JQ1协同抑制肿瘤细胞生长的新靶点,并阐明了TNBC中BET溴结构域蛋白驱动的转录调控的其他机制。引文格式:Samantha M. Bevill, Noah Sciaky, Brian T. Golitz, Naim U. Rashid, Jon S. Zawistowski, Gary L. Johnson。BET溴结构域抑制剂治疗三阴性乳腺癌的新型协同联合疗法[摘要]。摘自:AACR特别会议论文集:乳腺癌研究进展;2017年10月7-10日;费城(PA): AACR;癌症学报,2018;16(8 -增刊):摘要nr B34。
{"title":"Abstract B34: Novel synergistic combination therapies with BET bromodomain inhibitors in triple-negative breast cancer","authors":"Samantha M. Bevill, N. Sciaky, B. Golitz, N. Rashid, J. Zawistowski, G. Johnson","doi":"10.1158/1557-3125.ADVBC17-B34","DOIUrl":"https://doi.org/10.1158/1557-3125.ADVBC17-B34","url":null,"abstract":"Adaptive resistance to targeted cancer therapies is a universal problem in cancer treatment where tumor cells circumvent targeted pathway inhibition to reactivate growth signaling. In our previous work, we observed genome-wide enhancer remodeling following MEK inhibition (MEKi) capable of driving adaptive gene transcription in triple-negative breast cancer (TNBC). Adaptive enhancers were enriched for the BET bromodomain protein BRD4 and cotreatment with MEKi and BET inhibitor (JQ1) could durably suppress TNBC growth in multiple cell lines and preclinical mouse models. There are currently 10 BET inhibitors in clinical trials being tested as single agents across multiple tumor types including TNBC. There is also a growing body of preclinical literature using epigenetic inhibitors to block the adaptive ability of tumor cells in combination with multiple targeted therapies. This led us to screen for inhibitors that synergize with JQ1 to suppress growth of TNBC using a 176-compound library enriched for epigenetic and kinase inhibitors. We performed synergy screens in 6 TNBC cell lines across 6 doses of JQ1 and each library compound. Using the Bliss Independence model to assess synergy, we found that inhibition of MEK, CDK9, Aurora Kinase, CREBBP/P300, and BAZ2A/B was strongly synergistic with JQ1. BRD4, CDK9, and the acetyltransferase CREBBP/P300 are all members of the P-TEFb transcriptional elongation complex. When we performed additional synergy screens against the P300 bromodomain inhibitor CPI-637, we found a significant overlap in synergistic targets with the JQ1 screen including MEK, BET bromodomain proteins, ERK, Aurora Kinase, and CDK9. BAZ2A/B inhibition using the small-molecule inhibitor GSK2801, which targets the bromodomain of BAZ2A/B, synergized significantly stronger with JQ1 across all cell lines compared to CPI637. BAZ2A/B proteins are members of nucleosome remodeling complexes that mediate DNA silencing by aiding in recruitment of histone modifying enzymes. Ongoing studies seek to understand the role of BAZ2A/B and the mechanism of GSK2801 synergy with BET bromodomain inhibition using RNA sequencing and ChIP sequencing experiments. These results define novel targets that synergize with JQ1 to suppress tumor cell growth and illuminate additional mechanisms of transcriptional regulation driven by BET bromodomain proteins in TNBC. Citation Format: Samantha M. Bevill, Noah Sciaky, Brian T. Golitz, Naim U. Rashid, Jon S. Zawistowski, Gary L. Johnson. Novel synergistic combination therapies with BET bromodomain inhibitors in triple-negative breast cancer [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Cancer Research; 2017 Oct 7-10; Hollywood, CA. Philadelphia (PA): AACR; Mol Cancer Res 2018;16(8_Suppl):Abstract nr B34.","PeriodicalId":20897,"journal":{"name":"Resistance Mechanisms","volume":"27 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2018-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"78056635","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract PR05: The kinome of chemotherapy-resistant TNBC and identification of targetable kinases PR05:化疗耐药TNBC的激酶及靶向激酶的鉴定
Pub Date : 2018-08-01 DOI: 10.1158/1557-3125.ADVBC17-PR05
C. L. V. D. Borden, Nina M. Koemans, B. Pan, Changjun Wang, D. Wolf, Jean-Philippe Coppé, L. Veer
Triple-negative breast cancer (TNBC) accounts for around 15% of all breast cancer cases, with over 35,000 newly diagnosed women per year in the US. TNBC patients are at highest risk for recurrence, and neoadjuvant standard chemotherapy gives pathologic complete response in about 30% of these patients. Currently, no targeted therapy has been conclusively established to improve the outcome of TNBC patients, though successful phase II studies have been completed (1, 2). Therefore, identification of mechanisms that would be targetable is of great importance to improve the management of TNBC significantly. The aim of this study is to identify a unique set of hyperactivated kinases in chemotherapy-resistant TNBC cell lines that can potentially be targeted to achieve therapeutic response using a newly developed high-throughput kinase activity-mapping (HT-KAM) assay. The HT-KAM assay is our new screening technology to assess the catalytic activity of many kinases in parallel, which relies on collections of peptide probes that are used as combinatorial sensors to measure the phosphor-catalytic activity of kinases in large-scale high-throughput ATP-consumption assays (3). The HT-KAM system provides access to a new, untapped, and large resource of biologically meaningful measurements, both as a means to map the entire cancer kinome, and as a means to convert global phospho-signatures into functional patterns of kinase activity signatures. Kinome maps represent how kinase-signaling networks are rewired by drugs/targeted therapies in the context of different cellular backgrounds and exogenously mutated proteins/pathways, and provide insight into potentially targetable kinases. We previously established PhosphoAtlas, a heavily curated, comprehensive catalog database of 1,733 functionally interconnected proteins comprising the human phospho-reactome, including 4,748 unique edges that connect kinases to a target (776 kinase genes, 1,276 substrate protein genes, 2,492 heptameric peptide sequences [HPS]) (4). Here, we made a selection of 640 peptide sensors, capturing the functionality of >110 kinases over >900 kinase-substrate nodes directly relevant to tumor biology that represent the majority of the curated interconnected proteins, from our PhosphoAtlas (4). Earlier, the HT-KAM assay on 225 peptides was applied to explore mechanisms driving the unresponsiveness of colorectal and melanoma cancers to anti-BRAFV600E therapy in cell culture and patient-derived xenografts (PDX) (3). This successfully produced the predictive oncogenic kinome of melanoma tissues from patients suffering from fatal metastatic disease and, more importantly, identified new kinases/nodes that could be targeted to overcome drug resistance (3). In this study, we characterized the phospho-catalytic signatures of 10 TNBC cell lines (BT-549, HCC1143, HCC1395, HCC1937, HCC38, HCC70, HS578T, MDA-MB-231, MDA-MB-436, MDA-MB-468) across 640 peptides, either untreated or treated for 5 weeks with a ch
三阴性乳腺癌(TNBC)约占所有乳腺癌病例的15%,在美国每年有超过35,000名新诊断的女性。TNBC患者的复发风险最高,新辅助标准化疗在约30%的患者中获得病理完全缓解。目前,虽然已经完成了成功的II期研究,但还没有明确的靶向治疗方法来改善TNBC患者的预后(1,2)。因此,确定可靶向的机制对于显著改善TNBC的管理非常重要。本研究的目的是在化疗耐药的TNBC细胞系中鉴定一组独特的高活化激酶,这些激酶可以使用新开发的高通量激酶活性定位(HT-KAM)测定来潜在地靶向实现治疗反应。HT-KAM测定法是我们新的筛选技术,用于并行评估许多激酶的催化活性,它依赖于肽探针的集合,这些探针被用作组合传感器,用于在大规模高通量atp消耗测定中测量激酶的磷酸化催化活性(3)。HT-KAM系统提供了一种新的、未开发的、具有生物学意义的大量测量资源,既可以作为绘制整个癌症激酶组的手段,也可以作为绘制整个癌症激酶组的手段。作为一种将全局磷酸化信号转化为激酶活性信号的功能模式的手段。Kinome图谱代表了在不同细胞背景和外源突变蛋白/途径的背景下,药物/靶向治疗如何重新连接激酶信号网络,并提供了对潜在靶向激酶的见解。我们之前建立了PhosphoAtlas,这是一个精心策划的综合目录数据库,包含1,733个功能相互连接的蛋白质,包括人类磷酸化反应组,包括4,748个连接激酶和目标的独特边缘(776个激酶基因,1,276个底物蛋白基因,2,492个七聚体肽序列[HPS])(4)。在这里,我们选择了640个肽传感器。从我们的PhosphoAtlas(4)中,捕获了与肿瘤生物学直接相关的>110个激酶和>900个激酶-底物节点的功能,这些节点代表了大多数策划的相互连接蛋白。225个多肽的HT-KAM测定被用于探索导致结直肠癌和黑色素瘤癌症对细胞培养和患者来源的异种移植物(PDX)中的抗brafv600e治疗无反应的机制(3)。这成功地产生了来自致命转移性疾病患者的黑色素瘤组织的预测致癌激酶,更重要的是,确定了新的激酶/节点,可以靶向克服耐药性(3)。我们对10个TNBC细胞系(BT-549, HCC1143, HCC1395, HCC1937, HCC38, HCC70, HS578T, MDA-MB-231, MDA-MB-436, MDA-MB-468)在640个多肽上的磷酸化催化特征进行了表征,这些多肽在IC50浓度下未经治疗或使用化疗药物(卡铂,阿霉素,5- fu)治疗5周。为了验证预期的磷酸化特征,我们还评估了选定的纯化重组激酶的催化活性。基于初步数据,我们期望展示TNBC细胞系的致癌基因组的综合图谱,使我们能够根据其来源和治疗来区分TNBC细胞系。引用:1。Engebraaten O, Vollan HKM, Borresen-Dale A-L。三阴性乳腺癌和对新治疗靶点的需求。中华疾病杂志,2013;18(4):1064- 1074。http://www.sciencedirect.com/science/article/pii/S0002944013004616。2. Rugo HS, Olopade OI, DeMichele A,等。维利帕利-卡铂治疗乳腺癌的适应性随机化。中华医学杂志,2016;37(5):391 - 391。PMID: 27406347。3.Coppe JP, Mori M, Pan B,等。磷酸催化回路的功能检测发现了癌症的新脆弱性。提交。4。陈忠,李建平,李建平,等。人类基因组图谱揭示了癌症中磷酸化失调级联反应的突变景观。巨蟹座杂志2016;76:1733-45。PMID: 26921330。此摘要也以海报B37的形式呈现。引文格式:Carolien L. Van Der Borden, Nina M. Koemans, Bo Pan, Changjun Wang, Denise M. Wolf, Jean-Philippe Coppe, Laura J. Van ' t Veer。化疗耐药TNBC的激酶及靶向激酶的鉴定[摘要]。摘自:AACR特别会议论文集:乳腺癌研究进展;2017年10月7-10日;费城(PA): AACR;中华肿瘤杂志,2018;16(8):摘要nr PR05。
{"title":"Abstract PR05: The kinome of chemotherapy-resistant TNBC and identification of targetable kinases","authors":"C. L. V. D. Borden, Nina M. Koemans, B. Pan, Changjun Wang, D. Wolf, Jean-Philippe Coppé, L. Veer","doi":"10.1158/1557-3125.ADVBC17-PR05","DOIUrl":"https://doi.org/10.1158/1557-3125.ADVBC17-PR05","url":null,"abstract":"Triple-negative breast cancer (TNBC) accounts for around 15% of all breast cancer cases, with over 35,000 newly diagnosed women per year in the US. TNBC patients are at highest risk for recurrence, and neoadjuvant standard chemotherapy gives pathologic complete response in about 30% of these patients. Currently, no targeted therapy has been conclusively established to improve the outcome of TNBC patients, though successful phase II studies have been completed (1, 2). Therefore, identification of mechanisms that would be targetable is of great importance to improve the management of TNBC significantly. The aim of this study is to identify a unique set of hyperactivated kinases in chemotherapy-resistant TNBC cell lines that can potentially be targeted to achieve therapeutic response using a newly developed high-throughput kinase activity-mapping (HT-KAM) assay. The HT-KAM assay is our new screening technology to assess the catalytic activity of many kinases in parallel, which relies on collections of peptide probes that are used as combinatorial sensors to measure the phosphor-catalytic activity of kinases in large-scale high-throughput ATP-consumption assays (3). The HT-KAM system provides access to a new, untapped, and large resource of biologically meaningful measurements, both as a means to map the entire cancer kinome, and as a means to convert global phospho-signatures into functional patterns of kinase activity signatures. Kinome maps represent how kinase-signaling networks are rewired by drugs/targeted therapies in the context of different cellular backgrounds and exogenously mutated proteins/pathways, and provide insight into potentially targetable kinases. We previously established PhosphoAtlas, a heavily curated, comprehensive catalog database of 1,733 functionally interconnected proteins comprising the human phospho-reactome, including 4,748 unique edges that connect kinases to a target (776 kinase genes, 1,276 substrate protein genes, 2,492 heptameric peptide sequences [HPS]) (4). Here, we made a selection of 640 peptide sensors, capturing the functionality of >110 kinases over >900 kinase-substrate nodes directly relevant to tumor biology that represent the majority of the curated interconnected proteins, from our PhosphoAtlas (4). Earlier, the HT-KAM assay on 225 peptides was applied to explore mechanisms driving the unresponsiveness of colorectal and melanoma cancers to anti-BRAFV600E therapy in cell culture and patient-derived xenografts (PDX) (3). This successfully produced the predictive oncogenic kinome of melanoma tissues from patients suffering from fatal metastatic disease and, more importantly, identified new kinases/nodes that could be targeted to overcome drug resistance (3). In this study, we characterized the phospho-catalytic signatures of 10 TNBC cell lines (BT-549, HCC1143, HCC1395, HCC1937, HCC38, HCC70, HS578T, MDA-MB-231, MDA-MB-436, MDA-MB-468) across 640 peptides, either untreated or treated for 5 weeks with a ch","PeriodicalId":20897,"journal":{"name":"Resistance Mechanisms","volume":"45 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2018-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"88299608","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract A50: Modeling and targeting of oncogenic proteotoxic stress in drug-resistant breast cancer A50:耐药乳腺癌中致癌蛋白毒性应激的建模和靶向
Pub Date : 2018-08-01 DOI: 10.1158/1557-3125.ADVBC17-A50
Navneet Singh, V. Modur, Belal Muhammad, K. Komurov
ERBB2/HER2-positive breast cancers are associated with poor prognosis, and resistance to HER2-targeted therapy (trastuzumab and lapatinib) presents a significant clinical hurdle. There are no treatment options for metastatic HER2+ BCs that have progressed on HER2-targeted therapy. At present, most of the research on therapeutic strategies in therapy-refractory Her2+ BCs focuses on resensitizing tumor cells to HER2-targeting agents. We have shown that targeting of nononcogenic vulnerabilities, specifically adaptive proteotoxic stress response, is a promising therapeutic approach in this subset of BCs (Singh N. et al, HER2-mTOR signaling-driven breast cancer cells require ER-associated degradation to survive. Science Signaling 2015). We showed that Her2+ BCs have specific dependence on the endoplasmic reticulum (ER)-associated degradation (ERAD) pathway to suppress the hyperactive Her2/mTOR pathway-driven proteotoxic stress. Here, we explore the kinetics of protein homeostatic changes during the course of Her2-targeted drug therapy, and identify optimal therapeutic windows for the targeting of ERAD in the therapy-refractory Her2+ BCs. Using an integrated systems approach, we have modeled the dynamics of signaling and protein homeostasis changes in Her2+ cells during remission, resistance, and relapse phases of Her2-targeted therapy. We found that the resistance and relapse phases of anti-Her2 drug (lapatinib and trastuzumab) treatment are associated with Her2-independent acquisition of mTOR signaling, as previously reported. However, these signaling pathway changes also resulted in heightened proteotoxic burden on the ER due to increased protein synthesis rates, which was further exacerbated upon the release of cells from anti-Her2 treatment. Accordingly, cells in the relapse phase, and even more so upon further drug withdrawal, showed significantly heightened sensitivity to the inhibition of ERAD. This study reveals the kinetics of protein homeostasis associated with the signaling pathway changes during acquired resistance to anti-Her2 therapy, and suggests optimal therapeutic windows for the targeting of ERAD in therapy-refractory Her2+ BCs. Citation Format: Navneet Singh, Vishnu Modur, Belal Muhammad, Kakajan Komurov. Modeling and targeting of oncogenic proteotoxic stress in drug-resistant breast cancer [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Cancer Research; 2017 Oct 7-10; Hollywood, CA. Philadelphia (PA): AACR; Mol Cancer Res 2018;16(8_Suppl):Abstract nr A50.
ERBB2/ her2阳性乳腺癌与预后不良相关,对her2靶向治疗(曲妥珠单抗和拉帕替尼)的耐药性是一个重大的临床障碍。在HER2靶向治疗中进展的转移性HER2+ bc没有治疗选择。目前,对难治性Her2+ bc治疗策略的研究大多集中在使肿瘤细胞对Her2靶向药物重新敏感。我们已经证明,针对非癌性易感性,特别是适应性蛋白毒性应激反应,是一种很有希望的治疗方法,适用于这类bc (Singh N.等),HER2-mTOR信号驱动的乳腺癌细胞需要er相关降解才能存活。Science Signaling, 2015)。我们发现Her2+ bc特异性依赖内质网(ER)相关降解(ERAD)途径来抑制过度活跃的Her2/mTOR途径驱动的蛋白毒性应激。在这里,我们探讨了Her2靶向药物治疗过程中蛋白质稳态变化的动力学,并确定了ERAD靶向治疗难治性Her2+ bc的最佳治疗窗口。使用集成系统方法,我们模拟了Her2+细胞在Her2靶向治疗的缓解、抵抗和复发阶段的信号和蛋白质稳态变化的动力学。我们发现抗her2药物(拉帕替尼和曲妥珠单抗)治疗的耐药和复发阶段与her2非依赖性mTOR信号的获得相关,如先前报道的那样。然而,由于蛋白质合成速率的增加,这些信号通路的改变也导致内质网蛋白毒性负担的增加,这在抗her2治疗释放细胞后进一步加剧。因此,复发期的细胞,尤其是进一步停药后的细胞,对ERAD抑制的敏感性显著提高。该研究揭示了获得性抗Her2治疗耐药过程中与信号通路变化相关的蛋白稳态动力学,并提出了靶向ERAD治疗难治性Her2+ bc的最佳治疗窗口。引用格式:Navneet Singh, Vishnu Modur, Belal Muhammad, Kakajan Komurov。耐药乳腺癌中致癌蛋白毒性应激的建模与靶向研究[摘要]。摘自:AACR特别会议论文集:乳腺癌研究进展;2017年10月7-10日;费城(PA): AACR;中华肿瘤杂志,2018;16(8):1 - 5。
{"title":"Abstract A50: Modeling and targeting of oncogenic proteotoxic stress in drug-resistant breast cancer","authors":"Navneet Singh, V. Modur, Belal Muhammad, K. Komurov","doi":"10.1158/1557-3125.ADVBC17-A50","DOIUrl":"https://doi.org/10.1158/1557-3125.ADVBC17-A50","url":null,"abstract":"ERBB2/HER2-positive breast cancers are associated with poor prognosis, and resistance to HER2-targeted therapy (trastuzumab and lapatinib) presents a significant clinical hurdle. There are no treatment options for metastatic HER2+ BCs that have progressed on HER2-targeted therapy. At present, most of the research on therapeutic strategies in therapy-refractory Her2+ BCs focuses on resensitizing tumor cells to HER2-targeting agents. We have shown that targeting of nononcogenic vulnerabilities, specifically adaptive proteotoxic stress response, is a promising therapeutic approach in this subset of BCs (Singh N. et al, HER2-mTOR signaling-driven breast cancer cells require ER-associated degradation to survive. Science Signaling 2015). We showed that Her2+ BCs have specific dependence on the endoplasmic reticulum (ER)-associated degradation (ERAD) pathway to suppress the hyperactive Her2/mTOR pathway-driven proteotoxic stress. Here, we explore the kinetics of protein homeostatic changes during the course of Her2-targeted drug therapy, and identify optimal therapeutic windows for the targeting of ERAD in the therapy-refractory Her2+ BCs. Using an integrated systems approach, we have modeled the dynamics of signaling and protein homeostasis changes in Her2+ cells during remission, resistance, and relapse phases of Her2-targeted therapy. We found that the resistance and relapse phases of anti-Her2 drug (lapatinib and trastuzumab) treatment are associated with Her2-independent acquisition of mTOR signaling, as previously reported. However, these signaling pathway changes also resulted in heightened proteotoxic burden on the ER due to increased protein synthesis rates, which was further exacerbated upon the release of cells from anti-Her2 treatment. Accordingly, cells in the relapse phase, and even more so upon further drug withdrawal, showed significantly heightened sensitivity to the inhibition of ERAD. This study reveals the kinetics of protein homeostasis associated with the signaling pathway changes during acquired resistance to anti-Her2 therapy, and suggests optimal therapeutic windows for the targeting of ERAD in therapy-refractory Her2+ BCs. Citation Format: Navneet Singh, Vishnu Modur, Belal Muhammad, Kakajan Komurov. Modeling and targeting of oncogenic proteotoxic stress in drug-resistant breast cancer [abstract]. In: Proceedings of the AACR Special Conference: Advances in Breast Cancer Research; 2017 Oct 7-10; Hollywood, CA. Philadelphia (PA): AACR; Mol Cancer Res 2018;16(8_Suppl):Abstract nr A50.","PeriodicalId":20897,"journal":{"name":"Resistance Mechanisms","volume":"43 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2018-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"88565289","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Resistance Mechanisms
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
×
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
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