首页 > 最新文献

Experimental and Molecular Therapeutics最新文献

英文 中文
Abstract 1238: Treating estrogen receptor (ER)-negative and triple-negative breast cancer by targeting STAT3 signaling with putative STAT3 inhibitors 1238:利用STAT3抑制剂靶向STAT3信号通路治疗雌激素受体(ER)阴性和三阴性乳腺癌
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-1238
Hyejin Kim, Doerte R. Fricke, Jimin Xu, Haiying Chen, Jia Zhou, Q. Shen
{"title":"Abstract 1238: Treating estrogen receptor (ER)-negative and triple-negative breast cancer by targeting STAT3 signaling with putative STAT3 inhibitors","authors":"Hyejin Kim, Doerte R. Fricke, Jimin Xu, Haiying Chen, Jia Zhou, Q. Shen","doi":"10.1158/1538-7445.AM2021-1238","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1238","url":null,"abstract":"","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"75857930","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 1130: First-in-class KAT6A/KAT6B inhibitor CTx-648 (PF-9363) demonstrates potent anti-tumor activity in ER+ breast cancer with KAT6A dysregulation 摘要:一流的KAT6A/KAT6B抑制剂CTx-648 (PF-9363)在KAT6A失调的ER+乳腺癌中显示出强大的抗肿瘤活性
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-1130
Shikhar Sharma, Jay Chung, S. Uryu, Amanda M. Rickard, N. Nady, Showkhin Khan, Zhenxiong Wang, Yong Zhang, Haikuo Zhang, P. Kung, E. Greenwald, K. Maegley, P. Bingham, Hieu Lam, Y. E. Bozikis, Hendrik Falk, E. Allan, V. Avery, M. Butler, M. Camerino, Catalina Carrasco-Pozo, S. Charman, Melissa J. Davis, M. Dawson, Dawson Sarah-Jane, M. de Silva, M. Dennis, O. Dolezal, Rachel Lagiakos, G. Lindeman, Laura MacPherson, S. Nuttall, T. Peat, B. Ren, Alexandra E Stupple, Elliot E. Surgenor, Chin Wee Tan, T. Thomas, J. Visvader, A. Voss, F. Vaillant, K. White, J. Whittle, Yuqing Yang, Soroor Hediyeh-zadeh, P. Stupple, I. Street, B. Monahan, T. Paul
KAT6A is a lysine histone acetyltransferase (HAT) of the MYST family of HATs. KAT6A, and its paralog KAT6B, have been shown to acetylate histone H3K23Ac and regulate diverse biological processes, including transcription, cell-cycle progression, stem cell maintenance and development. Molecular dysregulation of KAT6A has been observed in several cancers, including amplifications in breast, lung, ovarian cancer along with oncogenic fusions in AML. In breast cancer, KAT6A is amplified as part of the 8p11 amplicon in 10-15% of the patient population, which correlates with a worse clinical outcome in the estrogen receptor+ (ER+) subtype. Here we present identification of a first-in-class potent KAT6A/KAT6B tool inhibitor CTx-648 (PF-9363), that possesses high selectivity versus other MYST family members (KAT7, KAT5, KAT8) and other KATs, demonstrating anti-tumor activity in breast cancer. Using genetic and pharmacological approaches, we have demonstrated several ER+ breast cancer cell lines including KAT6A amplified and over-expressing models, are dependent on KAT6A enzymatic function. Epigenomic profiling studies using bulk and nascent RNA-seq combined with ATAC-seq revealed CTx-648 leads to downregulation of a specific set of genes involved in ESR1 pathway, cell cycle and stem cell pathways. In vivo target validation studies showed strong anti-tumor activity of CTx-648 in several ER+ breast cancer cell line and patient-derived xenograft models, including models harboring endocrine therapy resistance ESR1 mutations, highlighting promise for this novel therapy in ER+ breast cancer population. Based on the strength of the pre-clinical data, a selective KAT6 inhibitor (PF-07248144) is now commencing a Phase 1 clinical study in Advanced or Metastatic Solid Tumors. Citation Format: Shikhar Sharma, Jay Chung, Sean Uryu, Amanda Rickard, Natalie Nady, Showkhin Khan, Zhenxiong Wang, Yong Zhang, Haikuo Zhang, Pei-Pei Kung, Eric Greenwald, Karen Maegley, Patrick Bingham, Hieu Lam, Ylva E. Bozikis, Hendrik Falk, Elizabeth Allan, Vicky M. Avery, Miriam S. Butler, Michelle A. Camerino, Catalina Carrasco-Pozo, Susan A. Charman, Melissa J. Davis, Mark A. Dawson, Dawson Sarah-Jane, Melanie de Silva, Matthew L. Dennis, Olan Dolezal, Rachel Lagiakos, Geoffrey J. Lindeman, Laura MacPherson, Stewart Nuttall, Thomas S. Peat, Bin Ren, Alexandra E. Stupple, Elliot Surgenor, Chin Wee Tan, Tim Thomas, Jane E. Visvader, Anne K. Voss, Francois Vaillant, Karen L. White, James Whittle, Yuqing Yang, Soroor Hediyeh-Zadeh, Paul A. Stupple, Ian P. Street, Brendon J. Monahan, Thomas Paul. First-in-class KAT6A/KAT6B inhibitor CTx-648 (PF-9363) demonstrates potent anti-tumor activity in ER+ breast cancer with KAT6A dysregulation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1130.
KAT6A是lysine histone acetyltransferase (HAT)的MYST家族成员。KAT6A及其类似物KAT6B已被证明可以使组蛋白H3K23Ac乙酰化,并调节多种生物过程,包括转录、细胞周期进程、干细胞维持和发育。已经在几种癌症中观察到KAT6A的分子失调,包括乳腺癌、肺癌、卵巢癌的扩增以及AML的致癌融合。在乳腺癌中,10-15%的患者群体中,KAT6A作为8p11扩增子的一部分被扩增,这与雌激素受体+ (ER+)亚型较差的临床结果相关。在这里,我们鉴定了一种一流的强效KAT6A/KAT6B工具抑制剂CTx-648 (sf -9363),它对其他MYST家族成员(KAT7, KAT5, KAT8)和其他KATs具有高选择性,显示出抗乳腺癌的活性。利用遗传和药理学方法,我们已经证明了几种ER+乳腺癌细胞系,包括KAT6A扩增和过表达模型,依赖于KAT6A酶的功能。使用大体积RNA-seq和新生RNA-seq结合ATAC-seq进行的表观基因组分析研究显示,CTx-648可导致ESR1通路、细胞周期和干细胞通路中一组特定基因的下调。体内靶标验证研究显示,CTx-648在多种ER+乳腺癌细胞系和患者来源的异种移植模型(包括内分泌治疗耐药ESR1突变模型)中具有较强的抗肿瘤活性,突出了这种新疗法在ER+乳腺癌人群中的应用前景。基于临床前数据的强度,一种选择性KAT6抑制剂(PF-07248144)目前正在进行晚期或转移性实体瘤的1期临床研究。引文格式:Shikhar Sharma、Jay Chung、Sean Uryu、Amanda Rickard、Natalie Nady、Showkhin Khan、王振雄、张勇、张海国、龚佩佩、Eric Greenwald、Karen Maegley、Patrick Bingham、林晓秀、Ylva E. Bozikis、Hendrik Falk、Elizabeth Allan、Vicky M. Avery、Miriam S. Butler、Michelle A. Camerino、Catalina Carrasco-Pozo、Susan A. Charman、Melissa J. Davis、Mark A. Dawson、Dawson Sarah-Jane、Melanie de Silva、Matthew L. Dennis、Olan Dolezal、Rachel Lagiakos、Geoffrey J. Lindeman、Laura MacPherson, Stewart Nuttall, Thomas S. Peat, Bin Ren, Alexandra E. Stupple, Elliot Surgenor, Chin Wee Tan, Tim Thomas, Jane E. Visvader, Anne K. Voss, Francois Vaillant, Karen L. White, James Whittle, Yuqing Yang, Soroor Hediyeh-Zadeh, Paul A. Stupple, Ian P. Street, Brendon J. Monahan, Thomas Paul。一流的KAT6A/KAT6B抑制剂CTx-648 (PF-9363)在KAT6A失调的ER+乳腺癌中显示出强大的抗肿瘤活性[摘要]。见:美国癌症研究协会2021年年会论文集;2021年4月10日至15日和5月17日至21日。费城(PA): AACR;癌症杂志,2021;81(13 -增刊):1130。
{"title":"Abstract 1130: First-in-class KAT6A/KAT6B inhibitor CTx-648 (PF-9363) demonstrates potent anti-tumor activity in ER+ breast cancer with KAT6A dysregulation","authors":"Shikhar Sharma, Jay Chung, S. Uryu, Amanda M. Rickard, N. Nady, Showkhin Khan, Zhenxiong Wang, Yong Zhang, Haikuo Zhang, P. Kung, E. Greenwald, K. Maegley, P. Bingham, Hieu Lam, Y. E. Bozikis, Hendrik Falk, E. Allan, V. Avery, M. Butler, M. Camerino, Catalina Carrasco-Pozo, S. Charman, Melissa J. Davis, M. Dawson, Dawson Sarah-Jane, M. de Silva, M. Dennis, O. Dolezal, Rachel Lagiakos, G. Lindeman, Laura MacPherson, S. Nuttall, T. Peat, B. Ren, Alexandra E Stupple, Elliot E. Surgenor, Chin Wee Tan, T. Thomas, J. Visvader, A. Voss, F. Vaillant, K. White, J. Whittle, Yuqing Yang, Soroor Hediyeh-zadeh, P. Stupple, I. Street, B. Monahan, T. Paul","doi":"10.1158/1538-7445.AM2021-1130","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1130","url":null,"abstract":"KAT6A is a lysine histone acetyltransferase (HAT) of the MYST family of HATs. KAT6A, and its paralog KAT6B, have been shown to acetylate histone H3K23Ac and regulate diverse biological processes, including transcription, cell-cycle progression, stem cell maintenance and development. Molecular dysregulation of KAT6A has been observed in several cancers, including amplifications in breast, lung, ovarian cancer along with oncogenic fusions in AML. In breast cancer, KAT6A is amplified as part of the 8p11 amplicon in 10-15% of the patient population, which correlates with a worse clinical outcome in the estrogen receptor+ (ER+) subtype. Here we present identification of a first-in-class potent KAT6A/KAT6B tool inhibitor CTx-648 (PF-9363), that possesses high selectivity versus other MYST family members (KAT7, KAT5, KAT8) and other KATs, demonstrating anti-tumor activity in breast cancer. Using genetic and pharmacological approaches, we have demonstrated several ER+ breast cancer cell lines including KAT6A amplified and over-expressing models, are dependent on KAT6A enzymatic function. Epigenomic profiling studies using bulk and nascent RNA-seq combined with ATAC-seq revealed CTx-648 leads to downregulation of a specific set of genes involved in ESR1 pathway, cell cycle and stem cell pathways. In vivo target validation studies showed strong anti-tumor activity of CTx-648 in several ER+ breast cancer cell line and patient-derived xenograft models, including models harboring endocrine therapy resistance ESR1 mutations, highlighting promise for this novel therapy in ER+ breast cancer population. Based on the strength of the pre-clinical data, a selective KAT6 inhibitor (PF-07248144) is now commencing a Phase 1 clinical study in Advanced or Metastatic Solid Tumors. Citation Format: Shikhar Sharma, Jay Chung, Sean Uryu, Amanda Rickard, Natalie Nady, Showkhin Khan, Zhenxiong Wang, Yong Zhang, Haikuo Zhang, Pei-Pei Kung, Eric Greenwald, Karen Maegley, Patrick Bingham, Hieu Lam, Ylva E. Bozikis, Hendrik Falk, Elizabeth Allan, Vicky M. Avery, Miriam S. Butler, Michelle A. Camerino, Catalina Carrasco-Pozo, Susan A. Charman, Melissa J. Davis, Mark A. Dawson, Dawson Sarah-Jane, Melanie de Silva, Matthew L. Dennis, Olan Dolezal, Rachel Lagiakos, Geoffrey J. Lindeman, Laura MacPherson, Stewart Nuttall, Thomas S. Peat, Bin Ren, Alexandra E. Stupple, Elliot Surgenor, Chin Wee Tan, Tim Thomas, Jane E. Visvader, Anne K. Voss, Francois Vaillant, Karen L. White, James Whittle, Yuqing Yang, Soroor Hediyeh-Zadeh, Paul A. Stupple, Ian P. Street, Brendon J. Monahan, Thomas Paul. First-in-class KAT6A/KAT6B inhibitor CTx-648 (PF-9363) demonstrates potent anti-tumor activity in ER+ breast cancer with KAT6A dysregulation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1130.","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"74438784","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: 7
Abstract 1427: Sensitizing HL60 acute myeloid leukemia cells to decitabine with pterostilbene 摘要1427:紫檀芪使HL60急性髓性白血病细胞对地西他滨敏感
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-1427
Cayla Boycott, B. Stefańska
{"title":"Abstract 1427: Sensitizing HL60 acute myeloid leukemia cells to decitabine with pterostilbene","authors":"Cayla Boycott, B. Stefańska","doi":"10.1158/1538-7445.AM2021-1427","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1427","url":null,"abstract":"","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"74644995","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 1445: A nuclear localizing peptide that targets glioblastoma 1445:一种靶向胶质母细胞瘤的核定位肽
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-1445
Calvin D. Lewis, A. Singh, V. Kapoor, D. Hallahan
{"title":"Abstract 1445: A nuclear localizing peptide that targets glioblastoma","authors":"Calvin D. Lewis, A. Singh, V. Kapoor, D. Hallahan","doi":"10.1158/1538-7445.AM2021-1445","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1445","url":null,"abstract":"","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"73016999","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 1280: New generation of STING agonists: Development and characterization of a novel series of systemic immunomodulators with improved potency 1280:新一代STING激动剂:开发和表征一系列新的系统性免疫调节剂,具有更高的效力
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-1280
Maciej K. Rogacki, S. Chmielewski, J. Mazurek, M. Zawadzka, K. Wnuk-Lipinska, K. Kuś, Katarzyna Wójcik-Jaszczyńska, Aleksandra Poczkaj, Łukasz Dudek, Wojciech Schonemann, Urszula Głowniak-Kwitek, M. Leś, Marek Wronowski, T. Mahajan, U. Kulesza, Magdalena Zastawna, David Synak, Karol Zuchowicz, Karolina Gluza, K. Banaszak, Karolina Wiatrowska, Izabela Strojny, M. Gładysz, J. Jablonska, Ewelina Gabor-Worwa, Monika Dobrzańska, Raghuram S. Tangirala, P. Littlewood, K. Brzózka
{"title":"Abstract 1280: New generation of STING agonists: Development and characterization of a novel series of systemic immunomodulators with improved potency","authors":"Maciej K. Rogacki, S. Chmielewski, J. Mazurek, M. Zawadzka, K. Wnuk-Lipinska, K. Kuś, Katarzyna Wójcik-Jaszczyńska, Aleksandra Poczkaj, Łukasz Dudek, Wojciech Schonemann, Urszula Głowniak-Kwitek, M. Leś, Marek Wronowski, T. Mahajan, U. Kulesza, Magdalena Zastawna, David Synak, Karol Zuchowicz, Karolina Gluza, K. Banaszak, Karolina Wiatrowska, Izabela Strojny, M. Gładysz, J. Jablonska, Ewelina Gabor-Worwa, Monika Dobrzańska, Raghuram S. Tangirala, P. Littlewood, K. Brzózka","doi":"10.1158/1538-7445.AM2021-1280","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1280","url":null,"abstract":"","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"73062567","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 1061: Characterization of synergistic selinexor combinations with dexamethasone, pomalidomide, elotuzumab, and daratumumab in primary MM cells 摘要/ Abstract摘要:selinexor与地塞米松、泊马度胺、elotuzumab和daratumumab联合在原发MM细胞中的协同作用
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-1061
P. Sudalagunta, M. Meads, R. Canevarolo, Maria C. Silva, C. Cubitt, Gabriel Deavila, R. Alugubelli, C. Logothetis, Amit Kulkarni, Qi Zhang, O. Hampton, C. Walker, Y. Landesman, K. Shain, A. Silva
Introduction. Multiple myeloma (MM) is an all but incurable plasma cell malignancy without predictive biomarkers for approved therapies. Selinexor (SELI), a nuclear export inhibitor targeting exportin 1 (XPO1), is approved with dexamethasone (DEX) with promising SELI-combination studies ongoing. We investigated SELI combinations ex vivo to identify synergistic combinations and companion biomarkers. Methods. We established a platform to perform parallel RNA/exome sequencing and ex vivo drug sensitivity assessment on CD138+ cells from MM patient bone marrow aspirates. At the time of this analysis, 844 different samples with clinical, WES and RNA sequencing data were treated ex vivo with the following agents: SELI (n=75), DEX (192), pomalidomide (POM, 268), elotuzumab (ELO, 21), daratumumab (DARA, 117), SELI+DEX (22), SELI+POM (20), SELI+ELO (21), SELI+DARA (27). Cells were cultured with autologous macrophages, stroma, collagen matrix and patient-derived serum. Cell death (LD50 and AUC) was assessed through digital image analysis. Sequencing was performed through ORIEN/AVATAR. Links between non-synonymous mutations in coding genes and cell death were calculated using T-tests with multiple test correction. Results. Our analysis identified SELI+DEX (number of samples=60, p<1E-9), SELI+POM (57, p<0.001) and SELI+ELO (55, p<0.01) as the most synergistic combinations (BLISS model). SELI+DARA showed synergy in 23 out of 50 samples tested. Notably, both direct drug toxicity and phagocytosis were observed. RNAseq found gene expression associations with drug resistance/response. In turn, gene set enrichment analysis (GSEA) showed that SELI resistance was associated with expression of cell adhesion, inflammatory cytokines, and EMT pathways, while the MYC targets were associated with SELI sensitivity. SELI+ELO resistance was associated with expression of hedgehog signaling pathway, while expression of ribosomal subunits was associated with sensitivity. SELI+POM resistance was linked with lysosome and cell adhesion molecules, while sensitivity was tied to ribosome, spliceosome and RNA polymerase. GSEA also identified G2M, MTORC1, MYC targets, E2F and glycolysis as biomarkers for the SELI+DARA synergistic subgroup. WES also identified mutations associated with SELI sensitivity. Mutation of BCL7A, a protein involved in chromatin remodeling, was associated with sensitivity, and mutation of CEP290, which encodes a microtubule binding protein, was associated with resistance (p<0.05). Both BCL7A and CEP290 contain predicted nuclear export sequences, suggesting they are XPO1 cargoes. Conclusions. We observed ex vivo synergy between SELI and DEX, POM, ELO and DARA, and identified expression signatures and mutations associated with response to these agents. Ongoing analysis of additional samples is being performed to validate these results. Citation Format: Praneeth Reddy Sudalagunta, Mark B. Meads, Rafael Renatino Canevarolo, Maria Coelho Silva, Christopher Cubitt, Gab
介绍。多发性骨髓瘤(MM)是一种几乎无法治愈的浆细胞恶性肿瘤,没有经批准治疗的预测性生物标志物。Selinexor (SELI)是一种靶向出口蛋白1 (XPO1)的核出口抑制剂,已被批准与地塞米松(DEX)联合进行有前景的Selinexor联合研究。我们在体外研究了SELI组合,以确定协同组合和伴随生物标志物。方法。我们建立了一个平台,对MM患者骨髓抽取的CD138+细胞进行平行RNA/外显子组测序和体外药物敏感性评估。在本分析中,844个具有临床、WES和RNA测序数据的不同样本接受了以下药物的体外治疗:SELI (n=75)、DEX(192)、pomalidomide (POM, 268)、elotuzumab (ELO, 21)、daratumumab (DARA, 117)、SELI+DEX(22)、SELI+POM(20)、SELI+ELO(21)、SELI+DARA(27)。用自体巨噬细胞、基质、胶原基质和患者源性血清培养细胞。通过数字图像分析评估细胞死亡(LD50和AUC)。通过ORIEN/AVATAR进行测序。编码基因的非同义突变与细胞死亡之间的联系使用多重检验校正的t检验计算。结果。我们的分析发现SELI+DEX(样本数=60,p<1E-9), SELI+POM (57, p<0.001)和SELI+ELO (55, p<0.01)是最具协同效应的组合(BLISS模型)。SELI+DARA在50个样本中有23个显示协同作用。值得注意的是,直接药物毒性和吞噬作用均被观察到。RNAseq发现基因表达与耐药/反应相关。反过来,基因集富集分析(GSEA)显示SELI抗性与细胞粘附、炎症细胞因子和EMT途径的表达有关,而MYC靶点与SELI敏感性相关。SELI+ELO耐药与hedgehog信号通路的表达有关,而核糖体亚基的表达与敏感性有关。SELI+POM耐药性与溶酶体和细胞粘附分子有关,敏感性与核糖体、剪接体和RNA聚合酶有关。GSEA还确定了G2M、MTORC1、MYC靶点、E2F和糖酵解作为SELI+DARA协同亚组的生物标志物。WES还发现了与SELI敏感性相关的突变。参与染色质重塑的蛋白BCL7A突变与敏感性相关,编码微管结合蛋白CEP290突变与耐药性相关(p<0.05)。BCL7A和CEP290都含有预测的核出口序列,表明它们是XPO1货物。结论。我们观察了SELI与DEX、POM、ELO和DARA的体外协同作用,并确定了与这些药物反应相关的表达特征和突变。正在对其他样品进行持续分析,以验证这些结果。引文格式:Praneeth Reddy Sudalagunta, Mark B. Meads, Rafael Renatino Canevarolo, Maria Coelho Silva, Christopher Cubitt, Gabriel DeAvila, Raghunandan Reddy Alugubelli, Constantine N. Logothetis, Amit Kulkarni, Qi Zhang, Oliver Hampton, Christopher J. Walker, Yosef Landesman, Kenneth Shain, Ariosto Siqueira Silva。selinexor与地塞米松、泊马度胺、elotuzumab和daratumumab在原发MM细胞中的协同联合特性[摘要]。见:美国癌症研究协会2021年年会论文集;2021年4月10日至15日和5月17日至21日。费城(PA): AACR;癌症杂志,2021;81(13 -增刊):摘要第1061期。
{"title":"Abstract 1061: Characterization of synergistic selinexor combinations with dexamethasone, pomalidomide, elotuzumab, and daratumumab in primary MM cells","authors":"P. Sudalagunta, M. Meads, R. Canevarolo, Maria C. Silva, C. Cubitt, Gabriel Deavila, R. Alugubelli, C. Logothetis, Amit Kulkarni, Qi Zhang, O. Hampton, C. Walker, Y. Landesman, K. Shain, A. Silva","doi":"10.1158/1538-7445.AM2021-1061","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1061","url":null,"abstract":"Introduction. Multiple myeloma (MM) is an all but incurable plasma cell malignancy without predictive biomarkers for approved therapies. Selinexor (SELI), a nuclear export inhibitor targeting exportin 1 (XPO1), is approved with dexamethasone (DEX) with promising SELI-combination studies ongoing. We investigated SELI combinations ex vivo to identify synergistic combinations and companion biomarkers. Methods. We established a platform to perform parallel RNA/exome sequencing and ex vivo drug sensitivity assessment on CD138+ cells from MM patient bone marrow aspirates. At the time of this analysis, 844 different samples with clinical, WES and RNA sequencing data were treated ex vivo with the following agents: SELI (n=75), DEX (192), pomalidomide (POM, 268), elotuzumab (ELO, 21), daratumumab (DARA, 117), SELI+DEX (22), SELI+POM (20), SELI+ELO (21), SELI+DARA (27). Cells were cultured with autologous macrophages, stroma, collagen matrix and patient-derived serum. Cell death (LD50 and AUC) was assessed through digital image analysis. Sequencing was performed through ORIEN/AVATAR. Links between non-synonymous mutations in coding genes and cell death were calculated using T-tests with multiple test correction. Results. Our analysis identified SELI+DEX (number of samples=60, p<1E-9), SELI+POM (57, p<0.001) and SELI+ELO (55, p<0.01) as the most synergistic combinations (BLISS model). SELI+DARA showed synergy in 23 out of 50 samples tested. Notably, both direct drug toxicity and phagocytosis were observed. RNAseq found gene expression associations with drug resistance/response. In turn, gene set enrichment analysis (GSEA) showed that SELI resistance was associated with expression of cell adhesion, inflammatory cytokines, and EMT pathways, while the MYC targets were associated with SELI sensitivity. SELI+ELO resistance was associated with expression of hedgehog signaling pathway, while expression of ribosomal subunits was associated with sensitivity. SELI+POM resistance was linked with lysosome and cell adhesion molecules, while sensitivity was tied to ribosome, spliceosome and RNA polymerase. GSEA also identified G2M, MTORC1, MYC targets, E2F and glycolysis as biomarkers for the SELI+DARA synergistic subgroup. WES also identified mutations associated with SELI sensitivity. Mutation of BCL7A, a protein involved in chromatin remodeling, was associated with sensitivity, and mutation of CEP290, which encodes a microtubule binding protein, was associated with resistance (p<0.05). Both BCL7A and CEP290 contain predicted nuclear export sequences, suggesting they are XPO1 cargoes. Conclusions. We observed ex vivo synergy between SELI and DEX, POM, ELO and DARA, and identified expression signatures and mutations associated with response to these agents. Ongoing analysis of additional samples is being performed to validate these results. Citation Format: Praneeth Reddy Sudalagunta, Mark B. Meads, Rafael Renatino Canevarolo, Maria Coelho Silva, Christopher Cubitt, Gab","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"73201598","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 1323: Novel autophagy inhibitor to treat pancreatic cancer 摘要1323:新型自噬抑制剂治疗胰腺癌
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-1323
Mythili Ramachandran, Zhao Ma, Yuanpei Li
{"title":"Abstract 1323: Novel autophagy inhibitor to treat pancreatic cancer","authors":"Mythili Ramachandran, Zhao Ma, Yuanpei Li","doi":"10.1158/1538-7445.AM2021-1323","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1323","url":null,"abstract":"","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"73210547","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 1188: Reclassification of ER+ (luminal A/luminal B1 minus ER low)-like and ER- like breast tumors based on proteomic/gene and clinical outcome signatures 摘要:基于蛋白质组学/基因和临床结局特征的ER+ (luminal A/luminal B1 - ER low)样和ER-样乳腺肿瘤的重新分类
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-1188
Guisong Wang, Punit Shah, R. Searfoss, Leigh Fantacone-Campbell, J. Hooke, B. Deyarmin, Rebecca N. Zingmark, S. Somiari, Jianfang Liu, L. Kvecher, Bradley J. Mostoller, Lori A. Sturtz, Praven-Kumar Raj-Kumar, E. Granger, L. Vahdat, M. Cutler, C. Bountra, R. Sarangarajan, Hai Hu, M. Kiebish, A. Kovatich, N. Narain, C. Shriver
Introduction: Classification of breast cancer can incorporate immunohistochemical (IHC) detection of ER/PR/HER2/KI67 to stratify the subtypes. High throughput proteomics analysis allows for the expansion of biomarker discovery within the subtypes. We evaluated a cohort of 109 tumors characterized as ER+ (Luminal A and Luminal B1; HER2+ and ER low (1-10%) cases were excluded) compared to ER-/HER2- tumors. Utilizing an integrated bioinformatics approach, we developed a proteomic marker signature to reclassify tumors into ER+(like) and ER-(like) tumors. CPTAC (Proteomic)/TCGA (RNAseq) datasets and larger METBRIC and GSE96058 cohorts were used to validate this marker signature. The selected biomarkers demonstrated significant differences impacting survival outcome. Methods: Clinical IHC subtyping of core biopsies was used to select a cohort of patients with ER+/HER2- and ER-/HER2- primary tumors from flash-frozen surgical samples. The positive/negative status of ER/PR/HER2 was defined using updated ASCO 2020 guidelines. Ki-67 status was determined using the 2011 St. Gallen9s International Expert Consensus recommendations. Proteomic analysis was performed using Thermo Q-Exactive+ LC MS/MS analysis. Differential analysis was applied to select the significantly altered proteins between ER+ and ER- cases, Univariate survival analysis was engaged to filter informative protein/genes using TCGA RNA-Seq data. Nearest centroid analysis was deployed to define the classifier to predict novel molecular subtypes. Results/Conclusions: We selected 34 proteins/genes from 164 significantly differentially expressed proteins for further analysis. The centroid model constructed with the 34 proteins defined 2 groups: ER+(like) and ER-(like). An additional 4 groups were defined across subtypes: luminal tumors classified both by IHC and marker signature (LL), luminal tumors classified by IHC but marker signature more like triple negative (LT), triple negative tumors classified by IHC but marker signature more like luminal (TL), and triple negative classified by both IHC and marker signature (TT). This marker signature segregated close to 5000 tumors across CPTAC, TCGA, METABRIC and GSE96058 cohorts. Survival analysis in these groups of patients revealed differences in radiation, hormone/radiation, hormone therapy, and hormone/radiation/chemotherapy treatments. In summary using proteomics data we identified a 34 gene/protein marker signature, validated in large external cohorts and exhibited impact on survival and response to therapy. Further, this signature was enriched in metabolism and microenvironmental associated factors that could represent novel targets or development combination strategies based on this signature. Citation Format: Guisong Wang, Punit Shah, Rick Searfoss, Leigh Fantacone-Campbell, Jeffrey A. Hooke, Brenda Deyarmin, Rebecca N. Zingmark, Stella Somiari, Jianfang Liu, Leonid Kvecher, Bradley Mostoller, Lori A. Sturtz, Praven-Kumar Raj-Kumar, Elder Grang
乳腺癌的分型可结合免疫组化(IHC)检测ER/PR/HER2/KI67进行亚型分型。高通量蛋白质组学分析允许在亚型中扩展生物标志物的发现。我们评估了109例ER+肿瘤(Luminal a和Luminal B1;与ER-/HER2-肿瘤相比,HER2+和ER低(1-10%)的病例被排除。利用综合生物信息学方法,我们开发了一种蛋白质组学标记标记,将肿瘤重新分类为ER+(样)和ER-(样)肿瘤。使用CPTAC (Proteomic)/TCGA (RNAseq)数据集和更大的METBRIC和GSE96058队列来验证该标记签名。所选择的生物标志物显示出影响生存结果的显着差异。方法:采用核心活检的临床免疫组化分型,从速冻手术样本中选择ER+/HER2-和ER-/HER2-原发肿瘤患者。使用更新的ASCO 2020指南定义ER/PR/HER2的阳性/阴性状态。Ki-67状态是根据2011年圣加仑国际专家共识建议确定的。蛋白质组学分析采用Thermo Q-Exactive+ LC MS/MS分析。采用差异分析选择ER+和ER-病例之间显著改变的蛋白,采用单因素生存分析使用TCGA RNA-Seq数据过滤信息蛋白/基因。最近质心分析被用来定义分类器来预测新的分子亚型。结果/结论:我们从164个显著差异表达蛋白中筛选出34个蛋白/基因进行进一步分析。34种蛋白构建质心模型,分为2组:ER+(like)和ER-(like)。另外,根据亚型划分了4组:经免疫组化和标记标记分类的管腔肿瘤(LL)、经免疫组化分类但标记标记更接近三阴性(LT)的管腔肿瘤、经免疫组化分类但标记标记更接近三阴性(TL)的三阴性肿瘤、经免疫组化和标记标记同时分类的三阴性肿瘤(TT)。该标记在CPTAC、TCGA、METABRIC和GSE96058队列中分离了近5000个肿瘤。这些患者的生存分析揭示了放疗、激素/放疗、激素治疗和激素/放疗/化疗治疗的差异。总之,利用蛋白质组学数据,我们确定了34个基因/蛋白质标记,在大型外部队列中得到验证,并显示出对生存和治疗反应的影响。此外,该特征富含代谢和微环境相关因子,可能代表基于该特征的新靶点或开发组合策略。引用格式:Guisong Wang, Punit Shah, Rick Searfoss, Leigh Fantacone-Campbell, Jeffrey A. Hooke, Brenda Deyarmin, Rebecca N. Zingmark, Stella Somiari,刘建芳,Leonid Kvecher, Bradley Mostoller, Lori A. Sturtz, prven - kumar Raj-Kumar, Elder Granger, Linda Vahdat, Mary L. Cutler, Chas Bountra, Rangaprasad Sarangarajan, Hai Hu, Michael A. Kiebish, Albert J. Kovatich, Niven R. Narain, Craig D. Shriver。基于蛋白质组学/基因和临床结局特征的ER+ (luminal A/luminal B1 - ER low)样和ER-样乳腺肿瘤的重新分类[摘要]。见:美国癌症研究协会2021年年会论文集;2021年4月10日至15日和5月17日至21日。费城(PA): AACR;癌症杂志,2021;81(13 -增刊):1188。
{"title":"Abstract 1188: Reclassification of ER+ (luminal A/luminal B1 minus ER low)-like and ER- like breast tumors based on proteomic/gene and clinical outcome signatures","authors":"Guisong Wang, Punit Shah, R. Searfoss, Leigh Fantacone-Campbell, J. Hooke, B. Deyarmin, Rebecca N. Zingmark, S. Somiari, Jianfang Liu, L. Kvecher, Bradley J. Mostoller, Lori A. Sturtz, Praven-Kumar Raj-Kumar, E. Granger, L. Vahdat, M. Cutler, C. Bountra, R. Sarangarajan, Hai Hu, M. Kiebish, A. Kovatich, N. Narain, C. Shriver","doi":"10.1158/1538-7445.AM2021-1188","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1188","url":null,"abstract":"Introduction: Classification of breast cancer can incorporate immunohistochemical (IHC) detection of ER/PR/HER2/KI67 to stratify the subtypes. High throughput proteomics analysis allows for the expansion of biomarker discovery within the subtypes. We evaluated a cohort of 109 tumors characterized as ER+ (Luminal A and Luminal B1; HER2+ and ER low (1-10%) cases were excluded) compared to ER-/HER2- tumors. Utilizing an integrated bioinformatics approach, we developed a proteomic marker signature to reclassify tumors into ER+(like) and ER-(like) tumors. CPTAC (Proteomic)/TCGA (RNAseq) datasets and larger METBRIC and GSE96058 cohorts were used to validate this marker signature. The selected biomarkers demonstrated significant differences impacting survival outcome. Methods: Clinical IHC subtyping of core biopsies was used to select a cohort of patients with ER+/HER2- and ER-/HER2- primary tumors from flash-frozen surgical samples. The positive/negative status of ER/PR/HER2 was defined using updated ASCO 2020 guidelines. Ki-67 status was determined using the 2011 St. Gallen9s International Expert Consensus recommendations. Proteomic analysis was performed using Thermo Q-Exactive+ LC MS/MS analysis. Differential analysis was applied to select the significantly altered proteins between ER+ and ER- cases, Univariate survival analysis was engaged to filter informative protein/genes using TCGA RNA-Seq data. Nearest centroid analysis was deployed to define the classifier to predict novel molecular subtypes. Results/Conclusions: We selected 34 proteins/genes from 164 significantly differentially expressed proteins for further analysis. The centroid model constructed with the 34 proteins defined 2 groups: ER+(like) and ER-(like). An additional 4 groups were defined across subtypes: luminal tumors classified both by IHC and marker signature (LL), luminal tumors classified by IHC but marker signature more like triple negative (LT), triple negative tumors classified by IHC but marker signature more like luminal (TL), and triple negative classified by both IHC and marker signature (TT). This marker signature segregated close to 5000 tumors across CPTAC, TCGA, METABRIC and GSE96058 cohorts. Survival analysis in these groups of patients revealed differences in radiation, hormone/radiation, hormone therapy, and hormone/radiation/chemotherapy treatments. In summary using proteomics data we identified a 34 gene/protein marker signature, validated in large external cohorts and exhibited impact on survival and response to therapy. Further, this signature was enriched in metabolism and microenvironmental associated factors that could represent novel targets or development combination strategies based on this signature. Citation Format: Guisong Wang, Punit Shah, Rick Searfoss, Leigh Fantacone-Campbell, Jeffrey A. Hooke, Brenda Deyarmin, Rebecca N. Zingmark, Stella Somiari, Jianfang Liu, Leonid Kvecher, Bradley Mostoller, Lori A. Sturtz, Praven-Kumar Raj-Kumar, Elder Grang","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"75413814","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 1380: Synergistic effect of the combination of XPO1 and mTORC1/2 inhibition for the treatment of triple-hit DLBCL 摘要1380:XPO1与mTORC1/2联合抑制治疗三击型DLBCL的协同效应
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-1380
Bing-Xu Hou, Aihua Wang, Bo Shan, J. Mei
Background Double/triple-hit lymphoma is an aggressive form of DLBCL with particularly poor outcome. XPO1 (exportin 1) is a well characterized nuclear export protein which is overexpressed in multiple tumor types. XPO1 exports many tumor-suppressor proteins and thus acts as a protooncogene by removing tumor suppressor protein and growth regulatory factors from the nucleus, where they are active, to the cytoplasm. First-in-class, single agent oral XPO1 inhibitor, Selinexor (ATG-010), was recently approved for the treatment of patients with DLBCL (de novo or transformed from follicular NHL) after at least two prior therapies. mTOR complex-1 (mTORC1) and mTOR complex-2 (mTORC2) are critical mediators of the PI3K-AKT pathway. Signaling by the PI3K/AKT/mTOR pathway is frequently deregulated in NHL. Inhibition of mTOR has shown preclinical and clinical efficacy in treating DLBCL. This study tested the antitumor effects induced by the combination of the XPO1 inhibitor, Selinexor and the dual mTORC1/2 kinase inhibitor, ATG-008 (Onatasertib) on triple-hit DLBCL cells, DoHH2. Methods Firstly, the in vitro 50% inhibition concentration (IC50) of two compounds was determined in DoHH2 cell line using CellTiter-Glo luminescent cell viability assay. The synergy effect of compound ATG-008 in combination with ATG-010 were evaluated by combination index (CI). The synergy effect is calculated by the Chou-Talalay Method [1]. The CI score ranging from 0.1-0.9 suggests very strong to slight synergism. The in vivo combination of the drugs were tested in DoHH2 CDX mouse model. The tumor bearing mouse were treated with vehicle control, Selinexor (5mg/kg, MWF), ATG-008 (10mg/kg, QD) or the combination for 21 days. The tumor size was measured twice a week and tumor growth inhibition (TGI) was evaluated compared with vehicle control group. Results Potent in vitro and in vivo anti-tumor efficacy and synergy has been observed for the combination of the two drugs. The in vitro IC50 for DOHH-2 cell line were 0.85µM and 0.08µM for ATG-008 and Selinexor, respectively. Synergy has been observed for most combo concentrations tested with the strongest synergism observed for ATG-008 (1.7µM) + Selinexor (0.16µM). The combination index was 0.48. In the DoHH2 CDX in vivo study, the mono therapy of ATG-008 showed 41% TGI at day 19 after grouping (p value Conclusions Strong synergism has been observed for the combination of XPO1 and mTORC1/2 inhibition by Selinexor and ATG-008, respectively, suggesting promising therapeutic strategies for double/triple-hit lymphoma patients that warrants further investigation. [1]Chou TC. Cancer Res. 2010 Jan 15;70(2):440-6. Citation Format: Bing Hou, Aihua Wang, Bo Shan, Jay Mei. Synergistic effect of the combination of XPO1 and mTORC1/2 inhibition for the treatment of triple-hit DLBCL [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(1
背景:双/三打淋巴瘤是一种侵袭性DLBCL,预后特别差。XPO1(输出蛋白1)是一种特征明确的核输出蛋白,在多种肿瘤类型中过表达。XPO1输出许多肿瘤抑制蛋白,从而作为原癌基因将肿瘤抑制蛋白和生长调节因子从活跃的细胞核移至细胞质。单药口服XPO1抑制剂Selinexor (ATG-010)最近被批准用于治疗至少两次治疗后的DLBCL(新生或从滤泡性NHL转化)患者。mTOR复合物-1 (mTORC1)和mTOR复合物-2 (mTORC2)是PI3K-AKT通路的关键介质。在NHL中,PI3K/AKT/mTOR信号通路经常失调。抑制mTOR在治疗DLBCL方面已显示出临床前和临床疗效。本研究检测了XPO1抑制剂Selinexor与双mTORC1/2激酶抑制剂ATG-008 (Onatasertib)联合使用对三击中DLBCL细胞DoHH2的抗肿瘤作用。方法首先采用CellTiter-Glo荧光细胞活力法测定两种化合物对doh2细胞株体外50%抑制浓度(IC50);采用联合指数(CI)评价复方ATG-008与ATG-010联合用药的协同效应。协同效应采用Chou-Talalay法计算[1]。CI值在0.1-0.9之间,表明协同作用非常强到轻微。在doh2 CDX小鼠模型上进行了药物的体内联合试验。荷瘤小鼠分别用载药对照、赛力纳索(5mg/kg, MWF)、ATG-008 (10mg/kg, QD)或联合用药治疗21 d。每周一次测量肿瘤大小,并与对照组比较肿瘤生长抑制(TGI)。结果两药合用具有较强的体内外抗肿瘤作用和协同作用。ATG-008和Selinexor对DOHH-2细胞株的体外IC50分别为0.85µM和0.08µM。大多数组合浓度均观察到协同作用,其中ATG-008(1.7µM) + Selinexor(0.16µM)的协同作用最强。组合指数为0.48。在DoHH2 CDX体内研究中,分组后第19天,ATG-008单独治疗的TGI为41% (p值)结论Selinexor和ATG-008联合抑制XPO1和mTORC1/2分别具有较强的协同作用,为双/三发淋巴瘤患者提供了有希望的治疗策略,值得进一步研究。[1]周TC。癌症杂志,2010,31(2):444 - 444。引用格式:侯冰,王爱华,单波,梅杰。XPO1与mTORC1/2联合抑制治疗三击型DLBCL的协同作用[摘要]。见:美国癌症研究协会2021年年会论文集;2021年4月10日至15日和5月17日至21日。费城(PA): AACR;癌症杂志,2021;81(13 -增刊):1380。
{"title":"Abstract 1380: Synergistic effect of the combination of XPO1 and mTORC1/2 inhibition for the treatment of triple-hit DLBCL","authors":"Bing-Xu Hou, Aihua Wang, Bo Shan, J. Mei","doi":"10.1158/1538-7445.AM2021-1380","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1380","url":null,"abstract":"Background Double/triple-hit lymphoma is an aggressive form of DLBCL with particularly poor outcome. XPO1 (exportin 1) is a well characterized nuclear export protein which is overexpressed in multiple tumor types. XPO1 exports many tumor-suppressor proteins and thus acts as a protooncogene by removing tumor suppressor protein and growth regulatory factors from the nucleus, where they are active, to the cytoplasm. First-in-class, single agent oral XPO1 inhibitor, Selinexor (ATG-010), was recently approved for the treatment of patients with DLBCL (de novo or transformed from follicular NHL) after at least two prior therapies. mTOR complex-1 (mTORC1) and mTOR complex-2 (mTORC2) are critical mediators of the PI3K-AKT pathway. Signaling by the PI3K/AKT/mTOR pathway is frequently deregulated in NHL. Inhibition of mTOR has shown preclinical and clinical efficacy in treating DLBCL. This study tested the antitumor effects induced by the combination of the XPO1 inhibitor, Selinexor and the dual mTORC1/2 kinase inhibitor, ATG-008 (Onatasertib) on triple-hit DLBCL cells, DoHH2. Methods Firstly, the in vitro 50% inhibition concentration (IC50) of two compounds was determined in DoHH2 cell line using CellTiter-Glo luminescent cell viability assay. The synergy effect of compound ATG-008 in combination with ATG-010 were evaluated by combination index (CI). The synergy effect is calculated by the Chou-Talalay Method [1]. The CI score ranging from 0.1-0.9 suggests very strong to slight synergism. The in vivo combination of the drugs were tested in DoHH2 CDX mouse model. The tumor bearing mouse were treated with vehicle control, Selinexor (5mg/kg, MWF), ATG-008 (10mg/kg, QD) or the combination for 21 days. The tumor size was measured twice a week and tumor growth inhibition (TGI) was evaluated compared with vehicle control group. Results Potent in vitro and in vivo anti-tumor efficacy and synergy has been observed for the combination of the two drugs. The in vitro IC50 for DOHH-2 cell line were 0.85µM and 0.08µM for ATG-008 and Selinexor, respectively. Synergy has been observed for most combo concentrations tested with the strongest synergism observed for ATG-008 (1.7µM) + Selinexor (0.16µM). The combination index was 0.48. In the DoHH2 CDX in vivo study, the mono therapy of ATG-008 showed 41% TGI at day 19 after grouping (p value Conclusions Strong synergism has been observed for the combination of XPO1 and mTORC1/2 inhibition by Selinexor and ATG-008, respectively, suggesting promising therapeutic strategies for double/triple-hit lymphoma patients that warrants further investigation. [1]Chou TC. Cancer Res. 2010 Jan 15;70(2):440-6. Citation Format: Bing Hou, Aihua Wang, Bo Shan, Jay Mei. Synergistic effect of the combination of XPO1 and mTORC1/2 inhibition for the treatment of triple-hit DLBCL [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(1","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"75494416","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 1208: Identifying a novel glycolytic inhibitor for treatment of aggressive prostate cancer 摘要1208:鉴定一种治疗侵袭性前列腺癌的新型糖酵解抑制剂
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-1208
Tanya Stoyanova
Among men, prostate cancer is the second leading cause of cancer-associated mortality, with advanced disease remaining a major clinical challenge. Chalcones are a major class of widely occurring natural products that are intermediates in plant flavonoid isoflavonoid synthesis. They are characterized by an α,β-unsaturated carbonyl structure with two aromatic rings and commonly act as free-radical scavengers. Herein, we describe a chalcone derivative, SU086, as an anticancer agent for prostate cancer. Proteomic and metabolomic profiling demonstrate that SU086 impairs glycolysis, a critical pathway for cancer growth and survival. SU086 inhibited prostate cancer cell growth, migration, and invasion in vitro. Moreover, SU086 significantly delayed the tumor growth of cell line-derived xenograft models of CRPC as well as patient-derived xenografts (PDXs) in vivo, and the proliferation of primary human prostate cancer patient-derived tissues ex vivo. Furthermore, SU086 strongly synergized with standard of care second-generation anti-androgens, enzalutamide and abiraterone, in inhibiting prostate cancer cell growth in vitro and tumor growth in vivo. Our study identifies SU086 alone or in combination therapy settings as a novel treatment for aggressive prostate cancer. We demonstrate that SU086 represents a highly effective therapeutic strategy for both AR-sensitive and AR-insensitive prostate cancers and may potentially be applicable across multiple cancer types. Citation Format: Tanya Ivanova Stoyanova. Identifying a novel glycolytic inhibitor for treatment of aggressive prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1208.
在男性中,前列腺癌是癌症相关死亡的第二大原因,晚期疾病仍然是一个主要的临床挑战。查尔酮是一类广泛存在的天然产物,是植物类黄酮异黄酮合成的中间体。它们具有α,β-不饱和羰基结构和两个芳香环,通常作为自由基清除剂。在这里,我们描述了一种查尔酮衍生物SU086,作为前列腺癌的抗癌剂。蛋白质组学和代谢组学分析表明,SU086损害糖酵解,这是癌症生长和生存的关键途径。SU086在体外抑制前列腺癌细胞的生长、迁移和侵袭。此外,SU086在体内显著延缓了CRPC细胞系来源的异种移植物模型和患者来源的异种移植物(PDXs)的肿瘤生长,以及原发性人前列腺癌患者来源组织的体外增殖。此外,SU086与标准治疗的第二代抗雄激素恩杂鲁胺和阿比特龙在体外抑制前列腺癌细胞生长和体内抑制肿瘤生长方面具有很强的协同作用。我们的研究确定SU086单独或联合治疗是一种治疗侵袭性前列腺癌的新方法。我们证明SU086对于ar敏感性和ar不敏感性前列腺癌都是一种非常有效的治疗策略,并且可能适用于多种癌症类型。引用格式:Tanya Ivanova Stoyanova。鉴定一种治疗侵袭性前列腺癌的新型糖酵解抑制剂[摘要]。见:美国癌症研究协会2021年年会论文集;2021年4月10日至15日和5月17日至21日。费城(PA): AACR;癌症杂志,2021;81(13 -增刊):摘要第1208期。
{"title":"Abstract 1208: Identifying a novel glycolytic inhibitor for treatment of aggressive prostate cancer","authors":"Tanya Stoyanova","doi":"10.1158/1538-7445.AM2021-1208","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1208","url":null,"abstract":"Among men, prostate cancer is the second leading cause of cancer-associated mortality, with advanced disease remaining a major clinical challenge. Chalcones are a major class of widely occurring natural products that are intermediates in plant flavonoid isoflavonoid synthesis. They are characterized by an α,β-unsaturated carbonyl structure with two aromatic rings and commonly act as free-radical scavengers. Herein, we describe a chalcone derivative, SU086, as an anticancer agent for prostate cancer. Proteomic and metabolomic profiling demonstrate that SU086 impairs glycolysis, a critical pathway for cancer growth and survival. SU086 inhibited prostate cancer cell growth, migration, and invasion in vitro. Moreover, SU086 significantly delayed the tumor growth of cell line-derived xenograft models of CRPC as well as patient-derived xenografts (PDXs) in vivo, and the proliferation of primary human prostate cancer patient-derived tissues ex vivo. Furthermore, SU086 strongly synergized with standard of care second-generation anti-androgens, enzalutamide and abiraterone, in inhibiting prostate cancer cell growth in vitro and tumor growth in vivo. Our study identifies SU086 alone or in combination therapy settings as a novel treatment for aggressive prostate cancer. We demonstrate that SU086 represents a highly effective therapeutic strategy for both AR-sensitive and AR-insensitive prostate cancers and may potentially be applicable across multiple cancer types. Citation Format: Tanya Ivanova Stoyanova. Identifying a novel glycolytic inhibitor for treatment of aggressive prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1208.","PeriodicalId":12258,"journal":{"name":"Experimental and Molecular Therapeutics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"73687379","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
Experimental and Molecular Therapeutics
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