首页 > 最新文献

Experimental and Molecular Therapeutics最新文献

英文 中文
Abstract 1015: mTOR inhibitor induces non-apoptotic, autophagy dependent cell death (ADCD) in TP53 mutant HNSCC 摘要:mTOR抑制剂在TP53突变型HNSCC中诱导非凋亡、自噬依赖性细胞死亡(ADCD)
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-1015
Maksudul M. Alam, Janmaris Marin Fermin, P. Spiller, Channing Dorr, Xiaohua Rong, Tara Medlin, Alok R. Khandelwal, C. Nathan
{"title":"Abstract 1015: mTOR inhibitor induces non-apoptotic, autophagy dependent cell death (ADCD) in TP53 mutant HNSCC","authors":"Maksudul M. Alam, Janmaris Marin Fermin, P. Spiller, Channing Dorr, Xiaohua Rong, Tara Medlin, Alok R. Khandelwal, C. Nathan","doi":"10.1158/1538-7445.AM2021-1015","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1015","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":"76966503","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 1050: Efficacy of AMG 176 in combination with gilteritinib in preclinical models of acute myeloid leukemia 摘要:AMG 176联合吉特替尼治疗急性髓系白血病临床前模型的疗效
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-1050
Xiao-yun Chen, S. Caenepeel, Brian Belmontes, P. McElroy, K. Rex, T. Osgood, P. Hughes
{"title":"Abstract 1050: Efficacy of AMG 176 in combination with gilteritinib in preclinical models of acute myeloid leukemia","authors":"Xiao-yun Chen, S. Caenepeel, Brian Belmontes, P. McElroy, K. Rex, T. Osgood, P. Hughes","doi":"10.1158/1538-7445.AM2021-1050","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1050","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":"76967051","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 1224: Discovery of novel BAF inhibitors for the treatment of transcription factor-driven cancers 1224:发现新的BAF抑制剂用于治疗转录因子驱动的癌症
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-1224
Richard C. Centore, L. M. Soares, R. Vaswani, Kana Ichikawa, Zhifang Li, Hong-Yan Fan, J. Setser, D. Lahr, L. Zawadzke, Xueying Chen, Kimberly D. Barnash, Jordana Muwanguzi, N. Anthony, Gabriel J. Sandoval, K. Feldman, Ammar Adam, David Huang, Shawn E R Schiller, Kevin Wilson, J. Voigt, M. Hentemann, Millan David Simon, H. Chan, S. Bellon, C. Decicco, Lang Xu
{"title":"Abstract 1224: Discovery of novel BAF inhibitors for the treatment of transcription factor-driven cancers","authors":"Richard C. Centore, L. M. Soares, R. Vaswani, Kana Ichikawa, Zhifang Li, Hong-Yan Fan, J. Setser, D. Lahr, L. Zawadzke, Xueying Chen, Kimberly D. Barnash, Jordana Muwanguzi, N. Anthony, Gabriel J. Sandoval, K. Feldman, Ammar Adam, David Huang, Shawn E R Schiller, Kevin Wilson, J. Voigt, M. Hentemann, Millan David Simon, H. Chan, S. Bellon, C. Decicco, Lang Xu","doi":"10.1158/1538-7445.AM2021-1224","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1224","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":"78145161","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 LB111: Drug repurposing of niclosamide to regulate Wnt/beta-catenin signaling pathway through upregulating SFRP1 in colorectal cancer 摘要:niclosamide通过上调SFRP1调控结直肠癌Wnt/ β -catenin信号通路
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-LB111
M. Wu, Christy W S Tong, Vivi W Yan, K. To
Aim: This study investigated the inhibition of proliferation and invasiveness of colorectal cancer (CRC) by niclosamide via targeting the secreted Frizzled-related protein 1 (SFRP1) and the Wnt/beta-catenin signaling pathway. Background: CRC is the third most common cancer worldwide. Aberrant activation of various signaling pathways in CRC leads to its poor response to chemotherapy. Hyperactivation of Wnt/beta-catenin pathway is known to drive cell proliferation, invasion, and migration of CRC. Epigenetic silencing of extracellular Wnt inhibitors, such as secreted Frizzled-related proteins (SFRPs), has been shown to stabilize beta-catenin and subsequently activate the Wnt signaling pathway. Niclosamide is a clinically approved drug indicated for the treatment of tapeworm infections. We have recently reported the repurposing of niclosamide to potentiate chemotherapeutic drugs for treating CRC by STAT3 inhibition. Niclosamide was also reported to be a Wnt/beta-catenin inhibitor but the precise mechanism is not clear. Methods: The antiproliferative effect was assessed by MTT assay and colony formation assay. The mRNA level, protein expression, and methylation status of SFRP1, and sets of SFRP1 co-expressed genes in CRC were analyzed by the TCGA data portal. The mRNA expression of SFRP1 and CpG island methylator phenotype (CIMP) marker genes were measured by qRT-PCR. DNA methylation was analyzed by bisulfite genomic sequencing. Inhibition of Wnt/beta-catenin and its downstream targets were examined by Western blot analysis. Cell migration and invasion were investigated by wound healing and transwell chamber migration assay. The intracellular localization of beta-catenin was determined by immunofluorescence assay. Results: Niclosamide inhibited HCT116 cell proliferation and colony formation in a concentration-dependent manner. Compared with normal colon tissues, CRC expressed significantly lower level of SFRP1 mRNA. SFRP1 methylation was inversely correlated with its mRNA expression. The DNA-demethylating agent 5-azacytidine was shown to restore expression of SFRP1 and CIMP panel genes in HCT116 cells after 5-day treatment. However, SFRP1 mRNA level was only upregulated by 1-day niclosamide treatment, but not 3-day niclosamide treatment. The upregulation of SFRP1 by niclosamide was found not relevant to DNA demethylation according to bisulfite genomic sequencing data. Immunofluorescence and Western blotting analysis showed that the increased SFRP1 expression was accompanied by reduced expression and nuclear accumulation of beta-catenin. Moreover, a low concentration of niclosamide that has minimal effect on cell proliferation, was shown to significantly suppress migration and invasion of HCT116 cells. Conclusion: Niclosamide is a promising candidate for drug repurposing. It was shown to upregulate SFRP1 and subsequently suppress Wnt/beta-catenin signaling, thereby impairing CRC cell migration and invasion. Upregulation of SFRP1 by niclosamide was not
目的:本研究通过靶向分泌的卷曲相关蛋白1 (frfrp1)和Wnt/ β -catenin信号通路,探讨氯硝胺对结直肠癌(CRC)增殖和侵袭性的抑制作用。背景:结直肠癌是全球第三大常见癌症。CRC中各种信号通路的异常激活导致其对化疗的不良反应。已知Wnt/ β -连环蛋白通路的过度激活可驱动结直肠癌的细胞增殖、侵袭和迁移。细胞外Wnt抑制剂的表观遗传沉默,如分泌的卷曲相关蛋白(SFRPs),已被证明可以稳定β -连环蛋白并随后激活Wnt信号通路。氯硝胺是临床批准的治疗绦虫感染的药物。我们最近报道了通过抑制STAT3来强化化疗药物治疗结直肠癌的研究。据报道,氯硝柳胺也是一种Wnt/ β -连环蛋白抑制剂,但其确切机制尚不清楚。方法:采用MTT法和菌落形成法评价其抗增殖作用。通过TCGA数据门户网站分析CRC中SFRP1和SFRP1共表达基因的mRNA水平、蛋白表达和甲基化状态。采用qRT-PCR检测SFRP1和CpG岛甲基化表型(CIMP)标记基因的mRNA表达量。亚硫酸氢盐基因组测序分析DNA甲基化。Western blot检测Wnt/ β -连环蛋白及其下游靶点的抑制作用。采用伤口愈合和跨室迁移实验研究细胞的迁移和侵袭。免疫荧光法测定β -连环蛋白的细胞内定位。结果:氯硝柳胺抑制HCT116细胞增殖和集落形成呈浓度依赖性。与正常结肠组织相比,结直肠癌中SFRP1 mRNA的表达水平明显降低。SFRP1甲基化与其mRNA表达呈负相关。dna去甲基化剂5-氮杂胞苷在治疗5天后恢复HCT116细胞中SFRP1和CIMP面板基因的表达。然而,SFRP1 mRNA水平仅在氯硝柳胺治疗1天时上调,而在氯硝柳胺治疗3天时没有上调。根据亚硫酸氢盐基因组测序数据,发现氯硝柳胺上调SFRP1与DNA去甲基化无关。免疫荧光和Western blotting分析显示,SFRP1表达升高伴随着β -连环蛋白的表达降低和核积累。此外,低浓度的氯硝柳胺对细胞增殖的影响很小,但却能显著抑制HCT116细胞的迁移和侵袭。结论:氯硝柳胺是一种很有前景的药物再利用药物。研究表明,它上调SFRP1并随后抑制Wnt/ β -连环蛋白信号传导,从而损害CRC细胞的迁移和侵袭。氯硝柳胺对SFRP1的上调不依赖于DNA去甲基化。引文格式:吴明霞、唐永森、严伟、杜建华。niclosamide药物再利用通过上调SFRP1调控结直肠癌Wnt/ β -catenin信号通路[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nr LB111。
{"title":"Abstract LB111: Drug repurposing of niclosamide to regulate Wnt/beta-catenin signaling pathway through upregulating SFRP1 in colorectal cancer","authors":"M. Wu, Christy W S Tong, Vivi W Yan, K. To","doi":"10.1158/1538-7445.AM2021-LB111","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-LB111","url":null,"abstract":"Aim: This study investigated the inhibition of proliferation and invasiveness of colorectal cancer (CRC) by niclosamide via targeting the secreted Frizzled-related protein 1 (SFRP1) and the Wnt/beta-catenin signaling pathway. Background: CRC is the third most common cancer worldwide. Aberrant activation of various signaling pathways in CRC leads to its poor response to chemotherapy. Hyperactivation of Wnt/beta-catenin pathway is known to drive cell proliferation, invasion, and migration of CRC. Epigenetic silencing of extracellular Wnt inhibitors, such as secreted Frizzled-related proteins (SFRPs), has been shown to stabilize beta-catenin and subsequently activate the Wnt signaling pathway. Niclosamide is a clinically approved drug indicated for the treatment of tapeworm infections. We have recently reported the repurposing of niclosamide to potentiate chemotherapeutic drugs for treating CRC by STAT3 inhibition. Niclosamide was also reported to be a Wnt/beta-catenin inhibitor but the precise mechanism is not clear. Methods: The antiproliferative effect was assessed by MTT assay and colony formation assay. The mRNA level, protein expression, and methylation status of SFRP1, and sets of SFRP1 co-expressed genes in CRC were analyzed by the TCGA data portal. The mRNA expression of SFRP1 and CpG island methylator phenotype (CIMP) marker genes were measured by qRT-PCR. DNA methylation was analyzed by bisulfite genomic sequencing. Inhibition of Wnt/beta-catenin and its downstream targets were examined by Western blot analysis. Cell migration and invasion were investigated by wound healing and transwell chamber migration assay. The intracellular localization of beta-catenin was determined by immunofluorescence assay. Results: Niclosamide inhibited HCT116 cell proliferation and colony formation in a concentration-dependent manner. Compared with normal colon tissues, CRC expressed significantly lower level of SFRP1 mRNA. SFRP1 methylation was inversely correlated with its mRNA expression. The DNA-demethylating agent 5-azacytidine was shown to restore expression of SFRP1 and CIMP panel genes in HCT116 cells after 5-day treatment. However, SFRP1 mRNA level was only upregulated by 1-day niclosamide treatment, but not 3-day niclosamide treatment. The upregulation of SFRP1 by niclosamide was found not relevant to DNA demethylation according to bisulfite genomic sequencing data. Immunofluorescence and Western blotting analysis showed that the increased SFRP1 expression was accompanied by reduced expression and nuclear accumulation of beta-catenin. Moreover, a low concentration of niclosamide that has minimal effect on cell proliferation, was shown to significantly suppress migration and invasion of HCT116 cells. Conclusion: Niclosamide is a promising candidate for drug repurposing. It was shown to upregulate SFRP1 and subsequently suppress Wnt/beta-catenin signaling, thereby impairing CRC cell migration and invasion. Upregulation of SFRP1 by niclosamide was not ","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":"78317870","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 979: Bcl-2 inhibitor enhances anti-androgen therapy induced regression of castration sensitive prostate cancer 979: Bcl-2抑制剂增强抗雄激素治疗诱导的去势敏感前列腺癌消退
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-979
Rahim Hirani, Subhiksha Nandakumar, T. Kalidindi, Deborah Fidele, Harisha Rajanala, Ying Z. Mazzu, Yuki Yoshikawa, Lina E. Jehane, G. Lee, E. Stanchina, Adam G. Sowalsky, M. Morris, H. Schöder, N. Pillarsetty, L. Mucci, D. Danila, G. Chakraborty, P. Kantoff
{"title":"Abstract 979: Bcl-2 inhibitor enhances anti-androgen therapy induced regression of castration sensitive prostate cancer","authors":"Rahim Hirani, Subhiksha Nandakumar, T. Kalidindi, Deborah Fidele, Harisha Rajanala, Ying Z. Mazzu, Yuki Yoshikawa, Lina E. Jehane, G. Lee, E. Stanchina, Adam G. Sowalsky, M. Morris, H. Schöder, N. Pillarsetty, L. Mucci, D. Danila, G. Chakraborty, P. Kantoff","doi":"10.1158/1538-7445.AM2021-979","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-979","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":"75151409","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 968: Focal adhesion kinase (FAK) inhibitor APG-2449 sensitizes ovarian tumors to chemotherapy via CD44 downregulation 摘要:局灶黏附激酶(FAK)抑制剂APG-2449通过下调CD44使卵巢肿瘤对化疗增敏
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-968
R. Tao, Douglas D Fang, Yuanbao Li, Kaixiang Zhang, Chunhua Xu, X. Fang, Qixin Wang, Dajun Yang, Y. Zhai
Ovarian cancer is one of the deadliest malignancies in women, and up to 70% of patients with epithelial ovarian cancer have FAK overexpression, amplification, or activation. FAK plays an important role in cellular migration, growth factor signaling, cell cycle progression, cellular survival and chemoresistance. This biomarker is also significantly associated with higher tumor stage, metastasis, and shorter overall survival in patients with ovarian cancer. Inhibition of FAK is therefore emerging as a promising treatment target. APG-2449 is a clinical stage FAK/ALK/ROS1 multi-kinase inhibitor. In this study we investigated antitumor activity of APG-2449 combined with standard-of-care chemotherapeutics in ovarian cancer in the preclinical setting. In a mouse xenograft tumor model derived from ovarian cancer cell line OVCAR-3, which was resistant to platinum-based therapies, APG-2449 combined with paclitaxel, and paclitaxel plus carboplatin, synergistically enhanced antitumor activity, whereas the chemotherapeutics showed no activity. Synergistic antitumor activity was also observed in multiple patient-derived xenograft (PDX) models derived from women with chemoinsensitive ovarian cancer, which also frequently expresses high levels of FAK. By comparing gene expression profiles of PDX tumors obtained from responders and nonresponders to the combined therapy, we identified CD44 (a marker for cancer stem cells) as a potentially predictive biomarker. Western blot analysis confirmed higher protein levels of CD44 in pretreated tumors of responders. Interestingly, downregulation of CD44 levels was observed in combination-treated tumors, suggesting that these combinations reduced cancer stem cell populations in ovarian cancer. Accordingly, in ovarian cancer cells exposed to APG-2449 alone or combined with paclitaxel, numbers of cells positive for CD44 or aldehyde dehydrogenase 1 (ALDH1; another marker for cancer stem cells) decreased in a dose-dependent manner. In summary, our data suggest that FAK inhibition by APG-2449 sensitizes ovarian tumors to chemotherapeutics in preclinical tumor models of ovarian cancer. The synergistic antitumor activity was mediated by downregulation of CD44+ or ALDH1+ cancer stem cell populations. These findings encourage clinical development of APG-2449 in combination with chemotherapies for treatment of ovarian cancer. Citation Format: Ran Tao, Douglas D. Fang, Yuanbao Li, Kaixiang Zhang, Chunhua Xu, Xu Fang, Qixin Wang, Dajun Yang, Yifan Zhai. Focal adhesion kinase (FAK) inhibitor APG-2449 sensitizes ovarian tumors to chemotherapy via CD44 downregulation [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 968.
卵巢癌是女性中最致命的恶性肿瘤之一,高达70%的上皮性卵巢癌患者存在FAK过表达、扩增或激活。FAK在细胞迁移、生长因子信号传导、细胞周期进程、细胞存活和化疗耐药等方面发挥重要作用。该生物标志物还与卵巢癌患者较高的肿瘤分期、转移和较短的总生存期显著相关。因此,抑制FAK正成为一个有希望的治疗靶点。APG-2449是一种临床分期FAK/ALK/ROS1多激酶抑制剂。在这项研究中,我们研究了APG-2449联合标准化疗药物在卵巢癌临床前的抗肿瘤活性。在对铂类药物耐药的卵巢癌细胞系OVCAR-3衍生的小鼠异种移植肿瘤模型中,APG-2449联合紫杉醇以及紫杉醇加卡铂可协同增强抗肿瘤活性,而化疗药物则无活性。在来自化疗不敏感卵巢癌女性的多种患者来源的异种移植(PDX)模型中也观察到协同抗肿瘤活性,该模型也经常表达高水平的FAK。通过比较对联合治疗有反应和无反应的PDX肿瘤的基因表达谱,我们确定了CD44(癌症干细胞的标志物)作为潜在的预测性生物标志物。Western blot分析证实,在应答者的预处理肿瘤中,CD44蛋白水平较高。有趣的是,在联合治疗的肿瘤中观察到CD44水平的下调,这表明这些联合治疗减少了卵巢癌中的癌症干细胞数量。因此,在单独或联合紫杉醇暴露于APG-2449的卵巢癌细胞中,CD44或醛脱氢酶1 (ALDH1)阳性的细胞数量;另一个癌症干细胞的标记物)以剂量依赖的方式减少。总之,我们的数据表明,在卵巢癌临床前肿瘤模型中,APG-2449抑制FAK可使卵巢肿瘤对化疗药物增敏。协同抗肿瘤活性是通过下调CD44+或ALDH1+肿瘤干细胞群介导的。这些发现鼓励APG-2449联合化疗治疗卵巢癌的临床发展。引用格式:陶然,方元保,李元保,张开祥,徐春华,方旭,王启新,杨大军,翟一凡。局灶黏附激酶(FAK)抑制剂APG-2449通过下调CD44使卵巢肿瘤对化疗增敏[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968期。
{"title":"Abstract 968: Focal adhesion kinase (FAK) inhibitor APG-2449 sensitizes ovarian tumors to chemotherapy via CD44 downregulation","authors":"R. Tao, Douglas D Fang, Yuanbao Li, Kaixiang Zhang, Chunhua Xu, X. Fang, Qixin Wang, Dajun Yang, Y. Zhai","doi":"10.1158/1538-7445.AM2021-968","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-968","url":null,"abstract":"Ovarian cancer is one of the deadliest malignancies in women, and up to 70% of patients with epithelial ovarian cancer have FAK overexpression, amplification, or activation. FAK plays an important role in cellular migration, growth factor signaling, cell cycle progression, cellular survival and chemoresistance. This biomarker is also significantly associated with higher tumor stage, metastasis, and shorter overall survival in patients with ovarian cancer. Inhibition of FAK is therefore emerging as a promising treatment target. APG-2449 is a clinical stage FAK/ALK/ROS1 multi-kinase inhibitor. In this study we investigated antitumor activity of APG-2449 combined with standard-of-care chemotherapeutics in ovarian cancer in the preclinical setting. In a mouse xenograft tumor model derived from ovarian cancer cell line OVCAR-3, which was resistant to platinum-based therapies, APG-2449 combined with paclitaxel, and paclitaxel plus carboplatin, synergistically enhanced antitumor activity, whereas the chemotherapeutics showed no activity. Synergistic antitumor activity was also observed in multiple patient-derived xenograft (PDX) models derived from women with chemoinsensitive ovarian cancer, which also frequently expresses high levels of FAK. By comparing gene expression profiles of PDX tumors obtained from responders and nonresponders to the combined therapy, we identified CD44 (a marker for cancer stem cells) as a potentially predictive biomarker. Western blot analysis confirmed higher protein levels of CD44 in pretreated tumors of responders. Interestingly, downregulation of CD44 levels was observed in combination-treated tumors, suggesting that these combinations reduced cancer stem cell populations in ovarian cancer. Accordingly, in ovarian cancer cells exposed to APG-2449 alone or combined with paclitaxel, numbers of cells positive for CD44 or aldehyde dehydrogenase 1 (ALDH1; another marker for cancer stem cells) decreased in a dose-dependent manner. In summary, our data suggest that FAK inhibition by APG-2449 sensitizes ovarian tumors to chemotherapeutics in preclinical tumor models of ovarian cancer. The synergistic antitumor activity was mediated by downregulation of CD44+ or ALDH1+ cancer stem cell populations. These findings encourage clinical development of APG-2449 in combination with chemotherapies for treatment of ovarian cancer. Citation Format: Ran Tao, Douglas D. Fang, Yuanbao Li, Kaixiang Zhang, Chunhua Xu, Xu Fang, Qixin Wang, Dajun Yang, Yifan Zhai. Focal adhesion kinase (FAK) inhibitor APG-2449 sensitizes ovarian tumors to chemotherapy via CD44 downregulation [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 968.","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":"75932333","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 1148: SynOV1.1: A synthetic gene circuit controlled oncolytic adenovirus demonstrating high tumor specificity, potent antitumor efficacy and high synergy with an anti-PD-L1 monoclonal antibody in preclinical hepatocellular carcinoma models 摘要SynOV1.1是一种合成的基因回路控制的溶瘤腺病毒,在临床前肝癌模型中具有高肿瘤特异性、强抗肿瘤功效和与抗pd - l1单克隆抗体的高协同作用
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-1148
Qiang Liu, Yubing Cao, Man Zhang, Shuguang Peng, Yiqi Liu, Huiya Huang, Bin-fan Chen
Hepatocellular carcinoma (HCC) is yet the fourth most common cause of cancer-related death although its treatment has been significantly advanced in recent years. Oncolytic viruses are among a novel class of immunotherapies that have shown promise to HCC patients with convincing abilities to selectively replicate in and kill cancer cells as well as to boost tumor immunogenicity. In order to further enhance the safety and efficacy of oncolytic viruses, SynOV1.1, a recombinant replication-competent serotype 5-based adenovirus, was constructed by deleting E1B and partial E3 genes, and incorporating a synthetic sensory switch circuit. The gene circuit was designed to enhance tumor-specific expression of E1A gene and an immune effector, human granulocyte-macrophage colony-stimulating factor (hGM-CSF) by sensing cancer-specific alpha-fetoprotein (AFP) promoter and two microRNA inputs. The specificity of SynOV1.1 to selectively kill target tumor cells and the ability to specifically express hGM-CSF in the target cells were confirmed in numerous human and mouse HCC cell lines with an IC50 approximately 3 to 35-fold less than that of normal cell lines. In vivo, intratumoral injections of SynOV1.1 in immune-deficient mice bearing human HCC cells resulted in a significant reduction of tumor volume (approximately 80%) as compared with the controls (approximately 30% for sorafenib). To further evaluate the effects of antitumor and immune stimulation, SynOV1.1m (SynOV1.1 murine surrogate) was intratumorally injected to the immune-competent mice bearing mouse HCC cells (ie., mHepa1-6), which showed SynOV1.1m induced a greater antitumor effect as well as a more robust tumor-specific immune response as compared to the controls. In addition, SynOV1.1m could significantly inhibit the growth of non-injected distal tumors after it was injected into a mHepa1-6 tumor on the other side of the mouse body, indicating that a systemic immune response was induced by SynOV1.1m. The combination of SynOV1.1m and an anti-mPD-L1 monoclonal antibody (mAb) was further explored and showed a synergistic anti-tumor effect as compared to SynOV1.1m or anti-mPD-L1 mAb alone. Preclinical safety of SynOV1.1 was examined in various toxicology species including golden hamsters, tumor-bearing mice and cynomolgus monkey, in all of which SynOV1.1 was found safe. Overall, preclinical pharmacology and toxicology data support clinical investigation of SynOV1.1 alone or in combination with atezolizumab in advanced HCC patients with positive serum AFP levels. Citation Format: Qiang Liu, Yubing Cao, Man Zhang, Shuguang Peng, Yiqi Liu, Huiya Huang, Bin Chen. SynOV1.1: A synthetic gene circuit controlled oncolytic adenovirus demonstrating high tumor specificity, potent antitumor efficacy and high synergy with an anti-PD-L1 monoclonal antibody in preclinical hepatocellular carcinoma models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and
肝细胞癌(HCC)仍然是癌症相关死亡的第四大常见原因,尽管近年来其治疗取得了显著进展。溶瘤病毒是一类新的免疫疗法之一,它对HCC患者显示出有希望,具有令人信服的选择性复制和杀死癌细胞以及增强肿瘤免疫原性的能力。为了进一步提高溶瘤病毒的安全性和有效性,我们通过删除E1B和部分E3基因,并加入合成的感觉开关电路,构建了具有复制能力的重组血清5型腺病毒SynOV1.1。该基因回路旨在通过检测癌症特异性甲胎蛋白(AFP)启动子和两种microRNA输入,增强E1A基因和免疫效应物人粒细胞-巨噬细胞集落刺激因子(hGM-CSF)的肿瘤特异性表达。SynOV1.1选择性杀伤靶肿瘤细胞的特异性和在靶细胞中特异性表达hGM-CSF的能力在许多人和小鼠HCC细胞系中得到证实,其IC50约为正常细胞系的3至35倍。在体内,在携带人类HCC细胞的免疫缺陷小鼠瘤内注射SynOV1.1,与对照组(索拉非尼约30%)相比,肿瘤体积显著减少(约80%)。为了进一步评估SynOV1.1m (SynOV1.1小鼠替代物)的抗肿瘤和免疫刺激作用,我们将SynOV1.1m (SynOV1.1小鼠替代物)瘤内注射到携带小鼠HCC细胞的免疫能力小鼠(即肝癌细胞)。与对照组相比,SynOV1.1m诱导了更强的抗肿瘤作用以及更强的肿瘤特异性免疫反应。此外,SynOV1.1m注射到小鼠身体另一侧的mHepa1-6肿瘤后,可以显著抑制非注射性远端肿瘤的生长,表明SynOV1.1m诱导了全身免疫反应。进一步探索SynOV1.1m与抗mpd - l1单克隆抗体(mAb)的联合,与SynOV1.1m或抗mpd - l1单克隆抗体单独相比,显示出协同抗肿瘤作用。SynOV1.1在金仓鼠、荷瘤小鼠和食蟹猴等多种毒理学物种的临床前安全性试验均显示为安全。总体而言,临床前药理学和毒理学数据支持SynOV1.1单独或联合atezolizumab用于血清AFP水平阳性的晚期HCC患者的临床研究。引用格式:刘强,曹玉兵,张满,彭曙光,刘益奇,黄惠亚,陈斌。SynOV1.1:一种合成的基因回路控制的溶瘤腺病毒,在临床前肝癌模型中具有高肿瘤特异性、强抗肿瘤功效和与抗pd - l1单克隆抗体的高协同作用[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):1148。
{"title":"Abstract 1148: SynOV1.1: A synthetic gene circuit controlled oncolytic adenovirus demonstrating high tumor specificity, potent antitumor efficacy and high synergy with an anti-PD-L1 monoclonal antibody in preclinical hepatocellular carcinoma models","authors":"Qiang Liu, Yubing Cao, Man Zhang, Shuguang Peng, Yiqi Liu, Huiya Huang, Bin-fan Chen","doi":"10.1158/1538-7445.AM2021-1148","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1148","url":null,"abstract":"Hepatocellular carcinoma (HCC) is yet the fourth most common cause of cancer-related death although its treatment has been significantly advanced in recent years. Oncolytic viruses are among a novel class of immunotherapies that have shown promise to HCC patients with convincing abilities to selectively replicate in and kill cancer cells as well as to boost tumor immunogenicity. In order to further enhance the safety and efficacy of oncolytic viruses, SynOV1.1, a recombinant replication-competent serotype 5-based adenovirus, was constructed by deleting E1B and partial E3 genes, and incorporating a synthetic sensory switch circuit. The gene circuit was designed to enhance tumor-specific expression of E1A gene and an immune effector, human granulocyte-macrophage colony-stimulating factor (hGM-CSF) by sensing cancer-specific alpha-fetoprotein (AFP) promoter and two microRNA inputs. The specificity of SynOV1.1 to selectively kill target tumor cells and the ability to specifically express hGM-CSF in the target cells were confirmed in numerous human and mouse HCC cell lines with an IC50 approximately 3 to 35-fold less than that of normal cell lines. In vivo, intratumoral injections of SynOV1.1 in immune-deficient mice bearing human HCC cells resulted in a significant reduction of tumor volume (approximately 80%) as compared with the controls (approximately 30% for sorafenib). To further evaluate the effects of antitumor and immune stimulation, SynOV1.1m (SynOV1.1 murine surrogate) was intratumorally injected to the immune-competent mice bearing mouse HCC cells (ie., mHepa1-6), which showed SynOV1.1m induced a greater antitumor effect as well as a more robust tumor-specific immune response as compared to the controls. In addition, SynOV1.1m could significantly inhibit the growth of non-injected distal tumors after it was injected into a mHepa1-6 tumor on the other side of the mouse body, indicating that a systemic immune response was induced by SynOV1.1m. The combination of SynOV1.1m and an anti-mPD-L1 monoclonal antibody (mAb) was further explored and showed a synergistic anti-tumor effect as compared to SynOV1.1m or anti-mPD-L1 mAb alone. Preclinical safety of SynOV1.1 was examined in various toxicology species including golden hamsters, tumor-bearing mice and cynomolgus monkey, in all of which SynOV1.1 was found safe. Overall, preclinical pharmacology and toxicology data support clinical investigation of SynOV1.1 alone or in combination with atezolizumab in advanced HCC patients with positive serum AFP levels. Citation Format: Qiang Liu, Yubing Cao, Man Zhang, Shuguang Peng, Yiqi Liu, Huiya Huang, Bin Chen. SynOV1.1: A synthetic gene circuit controlled oncolytic adenovirus demonstrating high tumor specificity, potent antitumor efficacy and high synergy with an anti-PD-L1 monoclonal antibody in preclinical hepatocellular carcinoma models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and","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":"76298151","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: 2
Abstract 935: Preclinical activity of seribantumab in gastrointestinal cancers withNRG1fusions 摘要:西班妥单抗在nrg1融合胃肠道肿瘤中的临床前活性
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-935
I. Odintsov, A. Lui, P. Bloom, M. Vojnic, S. Leland, M. Ladanyi, R. Somwar
Background. Oncogenic rearrangements of the neuregulin 1 gene (NRG1) consist of a 59 partner fused to a 39 NRG1 sequence that retains the EGF-like domain, and are found in 0.2% of solid tumors including lung, breast and gastrointestinal (GI) cancers. Carcinomas of GI origin, including pancreatic and cholangiocarcinoma, represent around 20% of solid tumors harboring NRG1 fusions and there is no approved therapy for this group of cancers. The chimeric NRG1 oncoproteins bind to HER3/ERBB3 leading to trans-activation of other ERBB family members and trigger a signaling cascade that culminates in oncogenesis. Although targeting HER3 represents a rational therapeutic strategy for cancers harboring NRG1 fusions, this has remained relatively unexplored for NRG1 fusion-positive GI malignancies. In this study we investigated the efficacy of the anti-HER3 monoclonal antibody seribantumab in preclinical models of NRG1-driven GI cancers. Methods. We developed models of isogenic pancreatic cancer cells with NRG1 fusions by lentiviral-mediated cDNA expression of ATP1B1-NRG1 and SLC3A2-NRG1 fusions in immortalized pancreatic ductal cells (H6c7). Seribantumab efficacy was evaluated in isogenic cell lines and in patient-derived xenograft (PDX) models of pancreatic adenocarcinoma (CTG-0943, APP-NRG1 fusion) and intrahepatic cholangiocarcinoma (CH-07-0068, RBPMS-NRG1 fusion). Western blotting analysis was used to evaluate protein phosphorylation. Expression of NRG1 fusions was confirmed by RT-PCR and NGS. Results. Expression of NRG1 fusions in H6c7 cells resulted in enhanced phosphorylation of HER3 and AKT and increased sensitivity to afatinib, as compared to empty vector control cells (H6c7-EV). Treatment of H6c7-SLC3A2-NRG1 cells with seribantumab resulted in a dose-dependent inhibition of HER3 and AKT phosphorylation. Seribantumab treatment of H6c7-ATP1B1-NRG1 and H6c7-SLC3A2-NRG1 cells resulted in dose-dependent inhibition of cell growth with IC50 values of 0.05 and 0.2 µM, respectively. In contrast, growth of H6c7-EV cells was much less sensitive to seribantumab (IC50 > 1µM). Tumor growth inhibition was observed after administration of seribantumab to PDX mouse models of pancreatic adenocarcinoma and intrahepatic cholangiocarcinoma. While seribantumab (5 mg and 10 mg per dose, BIW) was equally effective to the clinical equivalent dose of afatinib (5 mg/kg QD) in the cholangiocarcinoma PDX model, the two doses of seribantumab were more effective than afatinib in the pancreatic cancer PDX model, causing tumor shrinkage of up to 55% (23-77% range). There was no shrinkage of afatinib-treated pancreatic PDX tumors. Our results here suggest that seribantumab is effective at reducing tumor growth in preclinical models of gastrointestinal cancers with NRG1 fusions. These data support the use of seribantumab to treat GI and other cancers with NRG1 fusions in the ongoing phase 2 CRESTONE study (NCT#04383210). Citation Format: Igor Odintsov, Allan J. Lui, Paul R. Bloom, M
背景。神经调节蛋白1基因(NRG1)的致癌重排由一个59的伴侣融合到一个39的NRG1序列中,该序列保留了egf样结构域,并且在0.2%的实体肿瘤中发现,包括肺癌、乳腺癌和胃肠道(GI)癌症。胃肠道起源的癌,包括胰腺和胆管癌,约占含有NRG1融合物的实体肿瘤的20%,目前尚无针对这类癌症的批准治疗方法。嵌合的NRG1癌蛋白与HER3/ERBB3结合,导致其他ERBB家族成员的反式激活,并触发信号级联,最终导致肿瘤发生。尽管靶向HER3是治疗含有NRG1融合的癌症的一种合理的治疗策略,但对于NRG1融合阳性的胃肠道恶性肿瘤,这一策略仍相对未被探索。在这项研究中,我们研究了抗her3单克隆抗体西班妥单抗在nrg1驱动的GI癌症的临床前模型中的疗效。方法。我们通过慢病毒介导的ATP1B1-NRG1和SLC3A2-NRG1融合体在永生化胰腺导管细胞(H6c7)中的cDNA表达,建立了NRG1融合的等基因胰腺癌细胞模型。在等基因细胞系和患者来源的胰腺癌(CTG-0943, APP-NRG1融合)和肝内胆管癌(CH-07-0068, RBPMS-NRG1融合)的异种移植(PDX)模型中评估了serbantumab的疗效。Western blotting分析评价蛋白磷酸化水平。RT-PCR和NGS检测证实NRG1的表达。结果。与空载体对照细胞(H6c7- ev)相比,NRG1融合物在H6c7细胞中的表达导致HER3和AKT磷酸化增强,对阿法替尼的敏感性增加。用西班妥单抗治疗H6c7-SLC3A2-NRG1细胞导致HER3和AKT磷酸化的剂量依赖性抑制。西班妥单抗治疗H6c7-ATP1B1-NRG1和H6c7-SLC3A2-NRG1细胞对细胞生长的抑制呈剂量依赖性,IC50值分别为0.05和0.2µM。相比之下,H6c7-EV细胞的生长对西班妥单抗的敏感性要低得多(IC50 bb0 1µM)。用西班妥单抗治疗胰腺腺癌和肝内胆管癌的PDX小鼠模型后,观察到肿瘤生长抑制。虽然在胆管癌PDX模型中,西班妥单抗(5mg和10mg /剂量,BIW)与临床等效剂量的阿法替尼(5mg /kg QD)同样有效,但在胰腺癌PDX模型中,这两种剂量的西班妥单抗比阿法替尼更有效,导致肿瘤缩小高达55%(23-77%范围)。阿法替尼治疗的胰腺PDX肿瘤没有缩小。我们的研究结果表明,在NRG1融合的胃肠道癌症临床前模型中,西班妥单抗可有效降低肿瘤生长。这些数据支持在正在进行的2期CRESTONE研究(nct# 04383210)中使用seribantumab治疗GI和其他NRG1融合的癌症。引文格式:Igor Odintsov, Allan J. Lui, Paul R. Bloom, Morana Vojnic, Shawn Leland, Marc Ladanyi, Romel Somwar。西班妥单抗在NRG1融合的胃肠道肿瘤中的临床前活性[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):935。
{"title":"Abstract 935: Preclinical activity of seribantumab in gastrointestinal cancers withNRG1fusions","authors":"I. Odintsov, A. Lui, P. Bloom, M. Vojnic, S. Leland, M. Ladanyi, R. Somwar","doi":"10.1158/1538-7445.AM2021-935","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-935","url":null,"abstract":"Background. Oncogenic rearrangements of the neuregulin 1 gene (NRG1) consist of a 59 partner fused to a 39 NRG1 sequence that retains the EGF-like domain, and are found in 0.2% of solid tumors including lung, breast and gastrointestinal (GI) cancers. Carcinomas of GI origin, including pancreatic and cholangiocarcinoma, represent around 20% of solid tumors harboring NRG1 fusions and there is no approved therapy for this group of cancers. The chimeric NRG1 oncoproteins bind to HER3/ERBB3 leading to trans-activation of other ERBB family members and trigger a signaling cascade that culminates in oncogenesis. Although targeting HER3 represents a rational therapeutic strategy for cancers harboring NRG1 fusions, this has remained relatively unexplored for NRG1 fusion-positive GI malignancies. In this study we investigated the efficacy of the anti-HER3 monoclonal antibody seribantumab in preclinical models of NRG1-driven GI cancers. Methods. We developed models of isogenic pancreatic cancer cells with NRG1 fusions by lentiviral-mediated cDNA expression of ATP1B1-NRG1 and SLC3A2-NRG1 fusions in immortalized pancreatic ductal cells (H6c7). Seribantumab efficacy was evaluated in isogenic cell lines and in patient-derived xenograft (PDX) models of pancreatic adenocarcinoma (CTG-0943, APP-NRG1 fusion) and intrahepatic cholangiocarcinoma (CH-07-0068, RBPMS-NRG1 fusion). Western blotting analysis was used to evaluate protein phosphorylation. Expression of NRG1 fusions was confirmed by RT-PCR and NGS. Results. Expression of NRG1 fusions in H6c7 cells resulted in enhanced phosphorylation of HER3 and AKT and increased sensitivity to afatinib, as compared to empty vector control cells (H6c7-EV). Treatment of H6c7-SLC3A2-NRG1 cells with seribantumab resulted in a dose-dependent inhibition of HER3 and AKT phosphorylation. Seribantumab treatment of H6c7-ATP1B1-NRG1 and H6c7-SLC3A2-NRG1 cells resulted in dose-dependent inhibition of cell growth with IC50 values of 0.05 and 0.2 µM, respectively. In contrast, growth of H6c7-EV cells was much less sensitive to seribantumab (IC50 > 1µM). Tumor growth inhibition was observed after administration of seribantumab to PDX mouse models of pancreatic adenocarcinoma and intrahepatic cholangiocarcinoma. While seribantumab (5 mg and 10 mg per dose, BIW) was equally effective to the clinical equivalent dose of afatinib (5 mg/kg QD) in the cholangiocarcinoma PDX model, the two doses of seribantumab were more effective than afatinib in the pancreatic cancer PDX model, causing tumor shrinkage of up to 55% (23-77% range). There was no shrinkage of afatinib-treated pancreatic PDX tumors. Our results here suggest that seribantumab is effective at reducing tumor growth in preclinical models of gastrointestinal cancers with NRG1 fusions. These data support the use of seribantumab to treat GI and other cancers with NRG1 fusions in the ongoing phase 2 CRESTONE study (NCT#04383210). Citation Format: Igor Odintsov, Allan J. Lui, Paul R. Bloom, M","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":"74721840","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 1071: Studying the ability of retinoids to inhibit growth of CRC cells based on their retinoid pathway genotype 摘要/ Abstract摘要:基于类维生素a通路基因型研究类维生素a抑制结直肠癌细胞生长的能力
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-1071
Victoria O. Hunsu, C. Facey, Lynn M. Opdenaker, B. Boman
{"title":"Abstract 1071: Studying the ability of retinoids to inhibit growth of CRC cells based on their retinoid pathway genotype","authors":"Victoria O. Hunsu, C. Facey, Lynn M. Opdenaker, B. Boman","doi":"10.1158/1538-7445.AM2021-1071","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1071","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":"74220007","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 912: CLDN6 and CLDN9 dual targeting antibody drug conjugates for the treatment of ovarian and endometrial cancers 912: CLDN6和CLDN9双靶向抗体药物偶联物治疗卵巢癌和子宫内膜癌
Pub Date : 2021-07-01 DOI: 10.1158/1538-7445.AM2021-912
Du Liang, Hongyan Zhang, L. Jin, Yali Chen, Tingting Wan, L. Xu
{"title":"Abstract 912: CLDN6 and CLDN9 dual targeting antibody drug conjugates for the treatment of ovarian and endometrial cancers","authors":"Du Liang, Hongyan Zhang, L. Jin, Yali Chen, Tingting Wan, L. Xu","doi":"10.1158/1538-7445.AM2021-912","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-912","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":"74220494","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