Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-1363
Stefanie D. Krens, W. V. Boxtel, M. Uijen, F. Jansman, I. Desar, S. Mulder, C. V. Herpen, N. P. Erp
Background Cabozantinib is approved for treatment of renal cell cancer (RCC) in a starting dose of 60 mg. However, in the registration studies dose reductions were needed in 46-62% of patients due to toxicity. Improved clinical efficacy of cabozantinib was observed in RCC patients with an average exposure of > 750 ug/L. Data about cabozantinib pharmacokinetics in patients routinely treated with this drug is scarce, while treatment is challenging due to toxicity. Therefore, we explored the cabozantinib exposure in patients with two different tumour types in our clinic. Methods Clinical data and cabozantinib trough concentrations (Cmin) were collected from all patients treated with cabozantinib with at least one measured Cmin at steady state in our clinic between Jan 2018 - Aug 2020. We included salivary gland cancer (SGC) patients treated in a phase II study and RCC patients from our outpatient clinic. Geometric mean (GM) Cmin at the start dose, at 40 mg and at best tolerated dose (BTD) were compared between the two tumour types. Results In total 47 patients were included. All patients with SGC (n=22) started with 60 mg, while 13 of 25 patients with RCC started with 40 mg. GM Cmin level at the start dose was 1350 µg/L (95% CI 1182-1781) vs. 689 µg/L (95% CI 576-823) (P 750 ug/L. Conclusion Unexpectedly, cabozantinib levels were significantly higher in patients with SGC compared to those with RCC at the dose of 40 mg. However, cabozantinib levels at BTD were comparable between patients with SGC and RCC. At BTD, the majority of patients did not reach the target of >750 µg/L. For most patients with RCC, the cabozantinib level at BTD corresponded to a dose of 40 mg. Therefore, 40 mg instead of 60 mg as a starting dose followed by dose-adjustment based on exposure and tolerability may be preferred in patients with RCC. Although cabozantinib is not registered for SGC, an even lower starting dose of 20 mg/day may be required in these patients based on cabozantinib levels at BTD. Future studies should focus on identifying an optimal and tolerable exposure-response target value for cabozantinib and elucidate factors that contribute to the differences in exposure, in order to individualise and improve treatment. Citation Format: Stefanie D. Krens, Wim van Boxtel, Maike J. Uijen, Frank G. Jansman, Ingrid M. Desar, Sasja F. Mulder, Carla M. van Herpen, Nielka P. van Erp. Exposure-toxicity analysis of cabozantinib in patients with salivary gland cancer and renal cell 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 1363.
Cabozantinib被批准用于治疗肾细胞癌(RCC),起始剂量为60mg。然而,在登记研究中,由于毒性,46-62%的患者需要减少剂量。在平均暴露量> 750 ug/L的RCC患者中,卡博桑替尼的临床疗效得到改善。常规使用卡博赞替尼治疗的患者的药代动力学数据很少,而由于其毒性,治疗具有挑战性。因此,我们在我们的临床中探讨了两种不同肿瘤类型患者的卡博赞替尼暴露。方法收集2018年1月至2020年8月在我院接受卡博赞替尼治疗且至少有一项Cmin处于稳态的患者的临床数据和卡博赞替尼谷浓度(Cmin)。我们纳入了在II期研究中治疗的唾液腺癌(SGC)患者和门诊诊所的RCC患者。比较两种肿瘤类型在起始剂量、40mg和最佳耐受剂量(BTD)时的几何平均(GM) Cmin。结果共纳入47例患者。所有SGC患者(n=22)起始剂量为60mg,而25例RCC患者中有13例起始剂量为40mg。起始剂量时GM Cmin水平分别为1350µg/L (95% CI 1182 ~ 1781)和689µg/L (95% CI 576 ~ 823) (P 750 ug/L)。出乎意料的是,在40mg剂量下,SGC患者的卡博赞替尼水平明显高于RCC患者。然而,在SGC和RCC患者中,BTD的卡博赞替尼水平是相当的。在BTD时,大多数患者未达到>750µg/L的目标。对于大多数RCC患者,BTD时的卡博赞替尼水平相当于40mg的剂量。因此,RCC患者首选40mg而不是60mg作为起始剂量,然后根据暴露和耐受性进行剂量调整。虽然卡博赞替尼没有注册用于SGC,但基于BTD时卡博赞替尼的水平,这些患者可能需要更低的起始剂量20mg /天。未来的研究应侧重于确定卡博赞替尼的最佳和可耐受的暴露-反应目标值,并阐明导致暴露差异的因素,以便个性化和改善治疗。引用格式:Stefanie D. Krens, Wim van Boxtel, Maike J. Uijen, Frank G. Jansman, Ingrid M. Desar, Sasja F. Mulder, Carla M. van Herpen, Nielka P. van Erp。卡博赞替尼在涎腺癌和肾细胞癌患者中的暴露毒性分析[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):1363。
{"title":"Abstract 1363: Exposure-toxicity analysis of cabozantinib in patients with salivary gland cancer and renal cell cancer","authors":"Stefanie D. Krens, W. V. Boxtel, M. Uijen, F. Jansman, I. Desar, S. Mulder, C. V. Herpen, N. P. Erp","doi":"10.1158/1538-7445.AM2021-1363","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1363","url":null,"abstract":"Background Cabozantinib is approved for treatment of renal cell cancer (RCC) in a starting dose of 60 mg. However, in the registration studies dose reductions were needed in 46-62% of patients due to toxicity. Improved clinical efficacy of cabozantinib was observed in RCC patients with an average exposure of > 750 ug/L. Data about cabozantinib pharmacokinetics in patients routinely treated with this drug is scarce, while treatment is challenging due to toxicity. Therefore, we explored the cabozantinib exposure in patients with two different tumour types in our clinic. Methods Clinical data and cabozantinib trough concentrations (Cmin) were collected from all patients treated with cabozantinib with at least one measured Cmin at steady state in our clinic between Jan 2018 - Aug 2020. We included salivary gland cancer (SGC) patients treated in a phase II study and RCC patients from our outpatient clinic. Geometric mean (GM) Cmin at the start dose, at 40 mg and at best tolerated dose (BTD) were compared between the two tumour types. Results In total 47 patients were included. All patients with SGC (n=22) started with 60 mg, while 13 of 25 patients with RCC started with 40 mg. GM Cmin level at the start dose was 1350 µg/L (95% CI 1182-1781) vs. 689 µg/L (95% CI 576-823) (P 750 ug/L. Conclusion Unexpectedly, cabozantinib levels were significantly higher in patients with SGC compared to those with RCC at the dose of 40 mg. However, cabozantinib levels at BTD were comparable between patients with SGC and RCC. At BTD, the majority of patients did not reach the target of >750 µg/L. For most patients with RCC, the cabozantinib level at BTD corresponded to a dose of 40 mg. Therefore, 40 mg instead of 60 mg as a starting dose followed by dose-adjustment based on exposure and tolerability may be preferred in patients with RCC. Although cabozantinib is not registered for SGC, an even lower starting dose of 20 mg/day may be required in these patients based on cabozantinib levels at BTD. Future studies should focus on identifying an optimal and tolerable exposure-response target value for cabozantinib and elucidate factors that contribute to the differences in exposure, in order to individualise and improve treatment. Citation Format: Stefanie D. Krens, Wim van Boxtel, Maike J. Uijen, Frank G. Jansman, Ingrid M. Desar, Sasja F. Mulder, Carla M. van Herpen, Nielka P. van Erp. Exposure-toxicity analysis of cabozantinib in patients with salivary gland cancer and renal cell 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 1363.","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":"85543595","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}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-1085
F. Sinicrope, Lei Sun, Á. Patai, Tara L. Hogenson, M. Fernandez-Zapico, B. Qin
Background Colorectal cancers (CRCs) are intrinsically drug resistant tumors with frequent low or absent tumor cell PD-L1 expression. Previously, we reported that PD-L1 depletion in CRC cells can confer chemoresistance. Given that JNK signaling has been implicated in cell survival and drug resistance in CRC cells, we examined the role of JNK and its regulation in chemoresistance in CRC cells with loss of PD-L1. Materials & MethodsHuman CRC cell lines (RKO, DLD1 and HCT15) were utilized and parental and PD-L1 knockout/knockdown cells were generated. Cells were treated with a JNK1-3 inhibitor (JNK-IN-8) alone or combined with a MEK1/2 inhibitor (cobimetinib), CPT-11 or oxaliplatin. Protein/protein interaction was analyzed by immunoprecipitation (IP); protein/RNA interaction was analyzed by RNA IP assay. RNA level and half life were measured by QRT-PCR. Apoptosis was measured by cleaved caspase-3 and quantified by Annexin V labeling using FACS. Colony formation assay was performed. ResultsPD-L1 depletion in CRC cells was shown to enhance JNK activity that was due to reduced mRNA stability of the upstream CYLD deubiquitinase. Since PD-L1 competes with the ribonuclease EXOSC10 for binding to CYLD mRNA, loss of PD-L1 increases the interaction of EXOSC10 and CYLD mRNA resulting in its ribonuclease-mediated degradation and activation of JNK signaling. JNK activation increased BIM phosphorylation at Thr116 that promotes its sequestration by MCL-1 and BCL-2. Notably, a selective and irreversible JNK1-3 inhibitor (JNK-IN-8) reduces BIMThr116 phosphorylation and can release BIM from its sequestration by MCL-1 and BCL-2 to promote apoptosis. Furthermore, use of JNK-IN-8, an MCL-1 antagonist (AZD5991), or their combination in tumor cells lacking PD-L1 is shown to promote apoptosis and reduce long-term clonogenic survival induced by multiple anti-cancer drugs. As confirmation, knockdown of PD-L1 can confer drug resistance in a human colon cancer-derived organoid model that can be reversed by JNK-IN-8. Conclusion Tumor cells with low or absent PD-L1 expression show increased JNK activity that promotes BIM sequestration by MCL-1/BCL-2 to confer multiple drug resistance that can be reversed by a JNK inhibitor. Citation Format: Frank A. Sinicrope, Lei Sun, Arpad Patai, Tara L. Hogenson, Martin E. Fernandez-Zapico, Bo Qin. PD-L1-mediated resistance to chemotherapy is overcome by an irreversible JNK inhibitor in colorectal cancer cells [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 1085.
结直肠癌(crc)本质上是耐药肿瘤,肿瘤细胞PD-L1表达经常低或缺失。之前,我们报道过CRC细胞中PD-L1的缺失会导致化疗耐药。考虑到JNK信号与CRC细胞的细胞存活和耐药有关,我们研究了JNK在PD-L1缺失的CRC细胞化疗耐药中的作用及其调控。材料与方法利用人CRC细胞系(RKO、DLD1和HCT15),生成亲代细胞和PD-L1敲除/敲低细胞。细胞用JNK1-3抑制剂(JNK-IN-8)单独或联合MEK1/2抑制剂(cobimetinib)、CPT-11或奥沙利铂处理。免疫沉淀法(IP)分析蛋白/蛋白相互作用;RNA IP法分析蛋白/RNA相互作用。采用QRT-PCR检测RNA水平和半衰期。细胞凋亡用cleaved caspase-3检测,Annexin V标记用FACS定量。进行菌落形成试验。结果CRC细胞中spd - l1缺失可增强JNK活性,这是由于上游CYLD去泛素酶mRNA稳定性降低所致。由于PD-L1与核糖核酸酶EXOSC10竞争结合CYLD mRNA,因此PD-L1的缺失增加了EXOSC10与CYLD mRNA的相互作用,导致其核糖核酸酶介导的降解和JNK信号的激活。JNK激活增加了BIM Thr116位点的磷酸化,促进了MCL-1和BCL-2对其的封存。值得注意的是,一种选择性和不可逆的JNK1-3抑制剂(JNK-IN-8)降低了BIMThr116的磷酸化,可以将BIM从MCL-1和BCL-2的隔离中释放出来,促进细胞凋亡。此外,在缺乏PD-L1的肿瘤细胞中使用JNK-IN-8、MCL-1拮抗剂(AZD5991)或它们的联合可促进细胞凋亡,降低多种抗癌药物诱导的长期克隆生存期。作为证实,PD-L1的敲低可以在人类结肠癌衍生的类器官模型中赋予耐药,这可以通过JNK-IN-8逆转。结论PD-L1低表达或缺失的肿瘤细胞显示JNK活性增加,促进MCL-1/BCL-2对BIM的隔离,从而产生多重耐药,而JNK抑制剂可以逆转这种耐药。引文格式:Frank A. Sinicrope,孙雷,Arpad Patai, Tara L. Hogenson, Martin E. Fernandez-Zapico, Bo Qin。结直肠癌细胞中pd - l1介导的化疗耐药被一种不可逆的JNK抑制剂所克服[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1085期。
{"title":"Abstract 1085: PD-L1-mediated resistance to chemotherapy is overcome by an irreversible JNK inhibitor in colorectal cancer cells","authors":"F. Sinicrope, Lei Sun, Á. Patai, Tara L. Hogenson, M. Fernandez-Zapico, B. Qin","doi":"10.1158/1538-7445.AM2021-1085","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1085","url":null,"abstract":"Background Colorectal cancers (CRCs) are intrinsically drug resistant tumors with frequent low or absent tumor cell PD-L1 expression. Previously, we reported that PD-L1 depletion in CRC cells can confer chemoresistance. Given that JNK signaling has been implicated in cell survival and drug resistance in CRC cells, we examined the role of JNK and its regulation in chemoresistance in CRC cells with loss of PD-L1. Materials & MethodsHuman CRC cell lines (RKO, DLD1 and HCT15) were utilized and parental and PD-L1 knockout/knockdown cells were generated. Cells were treated with a JNK1-3 inhibitor (JNK-IN-8) alone or combined with a MEK1/2 inhibitor (cobimetinib), CPT-11 or oxaliplatin. Protein/protein interaction was analyzed by immunoprecipitation (IP); protein/RNA interaction was analyzed by RNA IP assay. RNA level and half life were measured by QRT-PCR. Apoptosis was measured by cleaved caspase-3 and quantified by Annexin V labeling using FACS. Colony formation assay was performed. ResultsPD-L1 depletion in CRC cells was shown to enhance JNK activity that was due to reduced mRNA stability of the upstream CYLD deubiquitinase. Since PD-L1 competes with the ribonuclease EXOSC10 for binding to CYLD mRNA, loss of PD-L1 increases the interaction of EXOSC10 and CYLD mRNA resulting in its ribonuclease-mediated degradation and activation of JNK signaling. JNK activation increased BIM phosphorylation at Thr116 that promotes its sequestration by MCL-1 and BCL-2. Notably, a selective and irreversible JNK1-3 inhibitor (JNK-IN-8) reduces BIMThr116 phosphorylation and can release BIM from its sequestration by MCL-1 and BCL-2 to promote apoptosis. Furthermore, use of JNK-IN-8, an MCL-1 antagonist (AZD5991), or their combination in tumor cells lacking PD-L1 is shown to promote apoptosis and reduce long-term clonogenic survival induced by multiple anti-cancer drugs. As confirmation, knockdown of PD-L1 can confer drug resistance in a human colon cancer-derived organoid model that can be reversed by JNK-IN-8. Conclusion Tumor cells with low or absent PD-L1 expression show increased JNK activity that promotes BIM sequestration by MCL-1/BCL-2 to confer multiple drug resistance that can be reversed by a JNK inhibitor. Citation Format: Frank A. Sinicrope, Lei Sun, Arpad Patai, Tara L. Hogenson, Martin E. Fernandez-Zapico, Bo Qin. PD-L1-mediated resistance to chemotherapy is overcome by an irreversible JNK inhibitor in colorectal cancer cells [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 1085.","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":"81686042","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}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-1328
S. Budhiraja, S. Baisiwala, E. Perrault, Li Chen, Cheol Park, Chidiebere U. Awah, C. Dmello, A. Zolp, A. Sonabend, Atique U. Ahmed
Glioblastoma (GBM) is the most aggressive and common type of adult malignant brain tumor, with 12,000 new diagnoses each year. Even with the current standard of care—surgical resection, radiation, and temozolomide (TMZ)-based chemotherapy—the median survival is about 20 months. This is partly due to the high rate of resistance to conventional therapy, including TMZ, leading to recurrence rates close to 100%. It remains largely unknown what drives the development of this resistance. Many studies have shown differences between primary and recurrent tumors, but a deeper understanding of resistance mechanisms is needed. CRISPR-Cas9 screening is a powerful tool for systematic and unbiased genetic analysis, which we applied to understand TMZ resistance. We performed a genome-wide CRISPR knockout screen in H4 human GBM cells, encompassing over 17,000 genes. A DMSO-treated population was compared with a TMZ-treated population over 14 days. In this drug sensitivity screen, depletion of guides corresponds to a TMZ-resistance gene, whereas enrichment of guides corresponds to a TMZ-sensitivity gene. Analysis showed that there was significant enrichment in guides for known TMZ-sensitivity genes that have been highly cited—ATG14, MSH6, MLH1, and PMS2—thus validating our screen results. However, more importantly, we were able to identify a list of 200 novel genes implicated in TMZ resistance. Pathway analysis revealed that these genes were enriched in Hippo and Notch signaling, both known to play a role in chemoresistance. From this list of novel genes, we identified 4 previously unstudied genes. These genes showed significant elevations in RNA expression (p Citation Format: Shreya Budhiraja, Shivani Baisiwala, Ella Perrault, Li Chen, Cheol Park, Chidiebere Awah, Crismita Dmello, Andrew Zolp, Adam Sonabend, Atique Ahmed. Using whole-genome CRISPR-Cas9 screening to identify resistance networks in glioblastoma [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 1328.
胶质母细胞瘤(GBM)是最具侵袭性和最常见的成人恶性脑肿瘤类型,每年有12,000例新诊断。即使采用目前的治疗标准——手术切除、放疗和以替莫唑胺(TMZ)为基础的化疗——中位生存期约为20个月。这部分是由于对包括TMZ在内的传统疗法的高耐药性导致复发率接近100%。是什么导致了这种耐药性的发展,这在很大程度上仍然是未知的。许多研究表明原发性和复发性肿瘤之间存在差异,但需要对耐药机制有更深入的了解。CRISPR-Cas9筛选是系统和公正的遗传分析的有力工具,我们将其用于了解TMZ抗性。我们在H4人类GBM细胞中进行了全基因组CRISPR敲除筛选,其中包含超过17,000个基因。将dmso处理的人群与tmz处理的人群进行14天的比较。在这个药敏筛选中,缺失的导体对应于tmz耐药基因,而富集的导体对应于tmz敏感基因。分析显示,已知tmz敏感基因(atg14、MSH6、MLH1和pms2)在指南中显著富集,从而验证了我们的筛选结果。然而,更重要的是,我们能够鉴定出200个与TMZ抗性有关的新基因。通路分析显示,这些基因在Hippo和Notch信号中富集,这两种信号都在化学耐药中起作用。从这个新基因列表中,我们确定了4个以前未研究过的基因。这些基因的RNA表达显著升高(p引文格式:Shreya Budhiraja, Shivani Baisiwala, Ella Perrault, Li Chen, Cheol Park, Chidiebere Awah, Crismita dello, Andrew Zolp, Adam Sonabend, Atique Ahmed)。利用全基因组CRISPR-Cas9筛选鉴定胶质母细胞瘤耐药网络[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):1328。
{"title":"Abstract 1328: Using whole-genome CRISPR-Cas9 screening to identify resistance networks in glioblastoma","authors":"S. Budhiraja, S. Baisiwala, E. Perrault, Li Chen, Cheol Park, Chidiebere U. Awah, C. Dmello, A. Zolp, A. Sonabend, Atique U. Ahmed","doi":"10.1158/1538-7445.AM2021-1328","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1328","url":null,"abstract":"Glioblastoma (GBM) is the most aggressive and common type of adult malignant brain tumor, with 12,000 new diagnoses each year. Even with the current standard of care—surgical resection, radiation, and temozolomide (TMZ)-based chemotherapy—the median survival is about 20 months. This is partly due to the high rate of resistance to conventional therapy, including TMZ, leading to recurrence rates close to 100%. It remains largely unknown what drives the development of this resistance. Many studies have shown differences between primary and recurrent tumors, but a deeper understanding of resistance mechanisms is needed. CRISPR-Cas9 screening is a powerful tool for systematic and unbiased genetic analysis, which we applied to understand TMZ resistance. We performed a genome-wide CRISPR knockout screen in H4 human GBM cells, encompassing over 17,000 genes. A DMSO-treated population was compared with a TMZ-treated population over 14 days. In this drug sensitivity screen, depletion of guides corresponds to a TMZ-resistance gene, whereas enrichment of guides corresponds to a TMZ-sensitivity gene. Analysis showed that there was significant enrichment in guides for known TMZ-sensitivity genes that have been highly cited—ATG14, MSH6, MLH1, and PMS2—thus validating our screen results. However, more importantly, we were able to identify a list of 200 novel genes implicated in TMZ resistance. Pathway analysis revealed that these genes were enriched in Hippo and Notch signaling, both known to play a role in chemoresistance. From this list of novel genes, we identified 4 previously unstudied genes. These genes showed significant elevations in RNA expression (p Citation Format: Shreya Budhiraja, Shivani Baisiwala, Ella Perrault, Li Chen, Cheol Park, Chidiebere Awah, Crismita Dmello, Andrew Zolp, Adam Sonabend, Atique Ahmed. Using whole-genome CRISPR-Cas9 screening to identify resistance networks in glioblastoma [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 1328.","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":"79511556","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}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-1289
W. Baer-Dubowska, Maria Narożna, V. Krajka-Kuźniak, B. Bednarczyk-Cwynar
Chronic inflammation is a key factor in the etiology of neoplastic diseases, including pancreatic cancer. Nonsteroidal anti-inflammatory drugs (NSAIDs) were approved for the chemoprevention of colon tumors and suggested for pancreas cancers prophylaxis. Anti-inflammatory and anti-tumorigenic activities are also exerted by naturally occurring and synthetic triterpenoids. Coupling of triterpenoid analogues with NSAIDs may enhance this effect and prevent the unfavorable side effects related to NSAIDs long-term use. In this study novel oleanolic acid oxime (OAO) derivatives conjugated with indomethacin (IND) differing in substitution group at position C-17 were evaluated in the context of their possible modulating effect of Nrf2-ARE and NF-κB signaling pathways in pancreatic cancer cells.PSN-1 cells were incubated for 24h with IND and OAO-IND derivatives at the concentrations of 10µM and 20µM selected based on the results of the MTT assay. The activation of Nrf2 and NF-κB was assessed by the evaluation of its translocation into the nucleus and binding to specific DNA sequences by the ELISA assay. Expression of Nrf2, SOD-1, NF-κBp50, NF-κBp65 and COX-2 was evaluated by RT-PCR and Western blot methods.Cell viability was affected mostly by 3-indomethacinoxyiminoolean-12-en-28-oic acid morpholide and 3-indomethacinoxyiminoolean-en-28-oic acid benzyl ester. The level of Nrf2 in the nucleus and binding to ARE sequence was decreased in PSN-1 cells, similarly as was NF-κBp50 and NF-κBp65 binding and nuclear accumulation. As result of diminished activation of these transcription factors decreased expression of SOD-1 and COX-2 i.e. mRNA and protein levels were found. These results indicate that the OAO-IND derivatives, particularly morpholide and benzyl ester conjugates, are more potent suppressors of NF-κB signaling pathway than IND alone. Moreover, conjugation of IND with novel OAO may protect cancer cells against chemoresistance through inhibition of the Nrf2-ARE pathway. Those novel compounds might be considered the potential modulators of hepatocellular carcinoma therapy and chemopreventive agents. Funding: This work was supported by Polish National Science Centre, grant 2016/21/B/NZ7/01758. Citation Format: Wanda Baer-Dubowska, Maria Narozna, Violetta Krajka-Kuźniak, Barbara Bednarczyk-Cwynar. Conjugation of indomethacin with novel oleanolic acid oximes increases its Nrf2 and NF-κB signaling pathways modulating effect in pancreatic cancer cells [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 1289.
慢性炎症是包括胰腺癌在内的肿瘤疾病病因学的关键因素。非甾体抗炎药(NSAIDs)已被批准用于结肠肿瘤的化学预防,并建议用于胰腺癌的预防。天然存在的和合成的三萜也具有抗炎和抗肿瘤活性。三萜类似物与非甾体抗炎药的偶联可以增强这种作用,并防止非甾体抗炎药长期使用的不良副作用。本研究评估了新型齐墩果酸肟(OAO)衍生物与吲哚美辛(IND)在C-17位置不同取代基的结合对胰腺癌细胞中Nrf2-ARE和NF-κB信号通路的可能调节作用。根据MTT检测结果选择10µM和20µM浓度的IND和OAO-IND衍生物孵育PSN-1细胞24h。通过ELISA法观察Nrf2和NF-κB的易位及与特定DNA序列的结合,评估其活化程度。RT-PCR和Western blot检测Nrf2、SOD-1、NF-κBp50、NF-κBp65和COX-2的表达。3-吲哚美辛氧亚胺酸-12-烯-28-酸酯和3-吲哚美辛氧亚胺酸-烯-28-酸苄酯对细胞活力的影响最大。在PSN-1细胞中,Nrf2与ARE序列的结合水平降低,NF-κBp50和NF-κBp65与细胞核的结合水平降低。由于这些转录因子的激活减少,SOD-1和COX-2的表达减少,即mRNA和蛋白水平下降。这些结果表明,OAO-IND衍生物,特别是morpholide和benzyl酯缀合物,比单独的IND更有效地抑制NF-κB信号通路。此外,IND与新型OAO的结合可能通过抑制Nrf2-ARE通路来保护癌细胞免受化疗耐药。这些新化合物可能被认为是肝细胞癌治疗和化学预防药物的潜在调节剂。项目资助:波兰国家科学中心资助项目2016/21/B/NZ7/01758。引文格式:Wanda Baer-Dubowska, Maria Narozna, Violetta Krajka-Kuźniak, Barbara Bednarczyk-Cwynar。吲哚美辛与新型齐墩果酸肟偶联可增强其对胰腺癌细胞中Nrf2和NF-κB信号通路的调节作用[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):1289。
{"title":"Abstract 1289: Conjugation of indomethacin with novel oleanolic acid oximes increases its Nrf2 and NF-κB signaling pathways modulating effect in pancreatic cancer cells","authors":"W. Baer-Dubowska, Maria Narożna, V. Krajka-Kuźniak, B. Bednarczyk-Cwynar","doi":"10.1158/1538-7445.AM2021-1289","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1289","url":null,"abstract":"Chronic inflammation is a key factor in the etiology of neoplastic diseases, including pancreatic cancer. Nonsteroidal anti-inflammatory drugs (NSAIDs) were approved for the chemoprevention of colon tumors and suggested for pancreas cancers prophylaxis. Anti-inflammatory and anti-tumorigenic activities are also exerted by naturally occurring and synthetic triterpenoids. Coupling of triterpenoid analogues with NSAIDs may enhance this effect and prevent the unfavorable side effects related to NSAIDs long-term use. In this study novel oleanolic acid oxime (OAO) derivatives conjugated with indomethacin (IND) differing in substitution group at position C-17 were evaluated in the context of their possible modulating effect of Nrf2-ARE and NF-κB signaling pathways in pancreatic cancer cells.PSN-1 cells were incubated for 24h with IND and OAO-IND derivatives at the concentrations of 10µM and 20µM selected based on the results of the MTT assay. The activation of Nrf2 and NF-κB was assessed by the evaluation of its translocation into the nucleus and binding to specific DNA sequences by the ELISA assay. Expression of Nrf2, SOD-1, NF-κBp50, NF-κBp65 and COX-2 was evaluated by RT-PCR and Western blot methods.Cell viability was affected mostly by 3-indomethacinoxyiminoolean-12-en-28-oic acid morpholide and 3-indomethacinoxyiminoolean-en-28-oic acid benzyl ester. The level of Nrf2 in the nucleus and binding to ARE sequence was decreased in PSN-1 cells, similarly as was NF-κBp50 and NF-κBp65 binding and nuclear accumulation. As result of diminished activation of these transcription factors decreased expression of SOD-1 and COX-2 i.e. mRNA and protein levels were found. These results indicate that the OAO-IND derivatives, particularly morpholide and benzyl ester conjugates, are more potent suppressors of NF-κB signaling pathway than IND alone. Moreover, conjugation of IND with novel OAO may protect cancer cells against chemoresistance through inhibition of the Nrf2-ARE pathway. Those novel compounds might be considered the potential modulators of hepatocellular carcinoma therapy and chemopreventive agents. Funding: This work was supported by Polish National Science Centre, grant 2016/21/B/NZ7/01758. Citation Format: Wanda Baer-Dubowska, Maria Narozna, Violetta Krajka-Kuźniak, Barbara Bednarczyk-Cwynar. Conjugation of indomethacin with novel oleanolic acid oximes increases its Nrf2 and NF-κB signaling pathways modulating effect in pancreatic cancer cells [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 1289.","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":"84525641","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}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-1479
J. Adashek, A. Desai, Arjun K. Menta, J. Roszik, V. Subbiah
{"title":"Abstract 1479: Pan-cancer efficacy of BRAF and/or MEK inhibitors in BRAF V600-mutant multiple non-melanoma cancers: A clinico-genomic study","authors":"J. Adashek, A. Desai, Arjun K. Menta, J. Roszik, V. Subbiah","doi":"10.1158/1538-7445.AM2021-1479","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1479","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":"85252796","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}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-1137
Koichi Ito, V. Thodima, Jack Carter, N. Bhagwat, Monisha Sivakumar, Alexander Grego, J. Rager, M. Terai, Takami Sato, O. Abdel-Wahab, Bruce R Ruggeri, P. Scherle, K. Vaddi
{"title":"Abstract 1137: PRMT5 inhibition regulates alternative splicing and DNA damage repair pathways in SF3B1 R625G expressing uveal melanoma cells","authors":"Koichi Ito, V. Thodima, Jack Carter, N. Bhagwat, Monisha Sivakumar, Alexander Grego, J. Rager, M. Terai, Takami Sato, O. Abdel-Wahab, Bruce R Ruggeri, P. Scherle, K. Vaddi","doi":"10.1158/1538-7445.AM2021-1137","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1137","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":"83836155","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}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-1372
Hoai‐Thu Thai, Nadia Gaudel-Dedieu, M. Cerou, Bernard Sebastien, H. Velde, D. Semiond, C. Veyrat‐Follet
Introduction: Isa is a CD38 monoclonal antibody with multiple modes of action for killing tumor cells. Isa, in combination with pomalidomide and dexamethasone (Pd), is approved for the treatment of adult pts with RRMM who have received ≥2 prior therapies including lenalidomide and a proteasome inhibitor. This study characterized the relationship between serum M-protein kinetics and progression free survival (PFS) in RRMM pts from the Phase 3 ICARIA-MM study, and simulated expected PFS outcomes when switching to a hypothetical monthly Isa dosing regimen after 6 months. Methods: A joint model of serum M-protein dynamics and PFS was developed using data from 256 pts. Pts received Isa intravenously, 10 mg/kg once weekly (QW) for 4 weeks (wks), then every other wk (Q2W) for 28-day cycles plus standard Pd (Isa-Pd) or Pd alone (control). A tumor growth inhibition model described serum M-protein kinetics under treatment effects of Isa-Pd or Pd alone; Isa exposure was predicted using individual PK parameters from the population PK analysis (Fau, PAGE Congress, 2019) and Pd exposure was predicted from K-PD model using dosing history. Trial simulations were performed using individual PK/PD parameters of ICARIA-MM pts. Results: The joint model identified instantaneous changes (slope) in serum M-protein as the best on-treatment predictor for PFS and identified baseline pt characteristics impacting serum M-protein kinetics (serum albumin and serum β2 microglobulin on the baseline serum M-protein levels and the non-IgG type on the serum M‐protein growth rate, the serum M-protein slope) and PFS (presence of plasmacytomas). Non-IgG MM pts have similar behavior on serum M-protein kinetics for the first 60 wks even with higher exposure and similar PFS vs IgG MM pts supporting non-dose adjustment based on IgG status. Clinical trial simulation of the Isa-Pd regimen showed that switching pts on treatment at 6 months to a monthly Isa regimen would shorten the median time to progression (TTP) by 4.1 wks and median PFS by 2.3 wks (14.03 to 13.45 months). Based on TTP criteria, pts with no risk of earlier progression while switching to a monthly Isa regimen (57.7%) tend to have lower baseline tumor burden (lower serum M-protein and lower percent of bone marrow plasma cell) and better prognostic factors (higher glomerular filtration rate, higher albumin, lower β2 microglobulin). At 6 months, 85% of these pts had predicted stable ≥VGPR status. Conclusions: Trial simulations support the approved Isa 10 mg/kg QW/Q2W regimen and show that switching to a monthly Isa regimen after 6 months may reduce clinical benefit in the overall population. However, some pts with good prognosis (low tumor burden, low ISS stage, good renal function) and obtaining stable ≥VGPR status by 6 months may switch to a monthly regimen after 6 months without compromising disease progression risk; this hypothesis will be tested in a prospective clinical trial. Citation Format: Hoai-Thu Thai, Nadia Gaudel
Isa是一种CD38单克隆抗体,具有多种杀伤肿瘤细胞的作用模式。Isa联合泊马度胺和地塞米松(Pd)被批准用于治疗既往接受过2种以上治疗(包括来那度胺和蛋白酶体抑制剂)的成年RRMM患者。本研究描述了来自ICARIA-MM 3期研究的RRMM患者血清m蛋白动力学与无进展生存期(PFS)之间的关系,并模拟了6个月后切换到假设的每月Isa给药方案时的预期PFS结果。方法:利用256例患者的数据建立血清m蛋白动力学和PFS联合模型。患者静脉注射Isa,每周一次(QW), 10mg /kg,持续4周(周),然后每隔一周(Q2W), 28天周期加标准Pd (Isa-Pd)或单独Pd(对照组)。肿瘤生长抑制模型描述了Isa-Pd或单独Pd治疗作用下的血清m蛋白动力学;使用群体PK分析中的个体PK参数预测Isa暴露(Fau, PAGE Congress, 2019),使用K-PD模型预测Pd暴露,使用给药历史。使用ICARIA-MM pts的个体PK/PD参数进行试验模拟。结果:联合模型确定了血清M蛋白的瞬时变化(斜率)是治疗期间PFS的最佳预测指标,并确定了影响血清M蛋白动力学的基线pt特征(血清白蛋白和血清β2微球蛋白对基线血清M蛋白水平的影响,非igg类型对血清M蛋白生长速率、血清M蛋白斜率的影响)和PFS(浆细胞瘤的存在)。非IgG MM患者在前60周的血清m蛋白动力学上具有相似的行为,即使在较高的暴露水平下也是如此,并且类似的PFS与IgG MM患者支持基于IgG状态的非剂量调整。Isa- pd方案的临床试验模拟显示,将6个月治疗的患者转换为每月一次的Isa方案将缩短中位进展时间(TTP) 4.1周,中位PFS缩短2.3周(14.03至13.45个月)。基于TTP标准,转换为每月一次Isa方案时无早期进展风险的患者(57.7%)往往具有较低的基线肿瘤负担(较低的血清m蛋白和较低的骨髓浆细胞百分比)和较好的预后因素(较高的肾小球滤过率,较高的白蛋白,较低的β2微球蛋白)。6个月时,85%的患者预测稳定的≥VGPR状态。结论:试验模拟支持已批准的Isa 10 mg/kg QW/Q2W方案,并显示在6个月后切换到每月Isa方案可能会降低总体人群的临床获益。然而,一些预后良好(肿瘤负荷低、ISS分期低、肾功能良好)且在6个月前获得稳定的≥VGPR状态的患者,在不降低疾病进展风险的情况下,可以在6个月后改用月度方案;这一假设将在前瞻性临床试验中得到验证。引文格式:Hoai-Thu Thai, Nadia Gaudel-Dedieu, Marc Cerou, Bernard Sebastien, Helgi van de Velde, Dorothee Semiond, Christine veyratt - follet。基于模型的方法评价Isatuximab (Isa)治疗复发/难治性多发性骨髓瘤(RRMM)的月给药方案[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1372期。
{"title":"Abstract 1372: Model based approach to evaluate Isatuximab (Isa) monthly dosing regimen in relapsed/refractory multiple myeloma (RRMM)","authors":"Hoai‐Thu Thai, Nadia Gaudel-Dedieu, M. Cerou, Bernard Sebastien, H. Velde, D. Semiond, C. Veyrat‐Follet","doi":"10.1158/1538-7445.AM2021-1372","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1372","url":null,"abstract":"Introduction: Isa is a CD38 monoclonal antibody with multiple modes of action for killing tumor cells. Isa, in combination with pomalidomide and dexamethasone (Pd), is approved for the treatment of adult pts with RRMM who have received ≥2 prior therapies including lenalidomide and a proteasome inhibitor. This study characterized the relationship between serum M-protein kinetics and progression free survival (PFS) in RRMM pts from the Phase 3 ICARIA-MM study, and simulated expected PFS outcomes when switching to a hypothetical monthly Isa dosing regimen after 6 months. Methods: A joint model of serum M-protein dynamics and PFS was developed using data from 256 pts. Pts received Isa intravenously, 10 mg/kg once weekly (QW) for 4 weeks (wks), then every other wk (Q2W) for 28-day cycles plus standard Pd (Isa-Pd) or Pd alone (control). A tumor growth inhibition model described serum M-protein kinetics under treatment effects of Isa-Pd or Pd alone; Isa exposure was predicted using individual PK parameters from the population PK analysis (Fau, PAGE Congress, 2019) and Pd exposure was predicted from K-PD model using dosing history. Trial simulations were performed using individual PK/PD parameters of ICARIA-MM pts. Results: The joint model identified instantaneous changes (slope) in serum M-protein as the best on-treatment predictor for PFS and identified baseline pt characteristics impacting serum M-protein kinetics (serum albumin and serum β2 microglobulin on the baseline serum M-protein levels and the non-IgG type on the serum M‐protein growth rate, the serum M-protein slope) and PFS (presence of plasmacytomas). Non-IgG MM pts have similar behavior on serum M-protein kinetics for the first 60 wks even with higher exposure and similar PFS vs IgG MM pts supporting non-dose adjustment based on IgG status. Clinical trial simulation of the Isa-Pd regimen showed that switching pts on treatment at 6 months to a monthly Isa regimen would shorten the median time to progression (TTP) by 4.1 wks and median PFS by 2.3 wks (14.03 to 13.45 months). Based on TTP criteria, pts with no risk of earlier progression while switching to a monthly Isa regimen (57.7%) tend to have lower baseline tumor burden (lower serum M-protein and lower percent of bone marrow plasma cell) and better prognostic factors (higher glomerular filtration rate, higher albumin, lower β2 microglobulin). At 6 months, 85% of these pts had predicted stable ≥VGPR status. Conclusions: Trial simulations support the approved Isa 10 mg/kg QW/Q2W regimen and show that switching to a monthly Isa regimen after 6 months may reduce clinical benefit in the overall population. However, some pts with good prognosis (low tumor burden, low ISS stage, good renal function) and obtaining stable ≥VGPR status by 6 months may switch to a monthly regimen after 6 months without compromising disease progression risk; this hypothesis will be tested in a prospective clinical trial. Citation Format: Hoai-Thu Thai, Nadia Gaudel","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":"82272146","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}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-950
Juniper A Scribner, Francine Z Chen, Ying Li, M. Chiechi, T. Son, J. Hooley, S. Koenig, P. Moore, E. Bonvini, C. Bohac, D. Loo
Introduction: Squamous cell carcinoma of the head and neck (SCCHN) is the seventh most common cancer worldwide, accounting for ~ 4% of all cancers in the United States. Although PD-1-directed therapy has efficacy in SCCHN, 85-95% of patients progress following initial response. B7-H3 is a member of the B7 family of immunomodulatory molecules, is overexpressed in SCCHN, and correlates with disease severity and poor clinical outcome. Furthermore, consistent with its putative coinhibitory function, B7-H3 expression in SCCHN is inversely correlated with the number of tumor infiltrating CD8+ T-cells. We are developing therapeutics targeting B7-H3, including enoblituzumab, an Fc-engineered anti-B7-H3 monoclonal antibody, and MGC018, a duocarmycin-based B7-H3 ADC, both of which are currently being evaluated in clinical studies. Here the potential of MGC018 was explored in preclinical models as a proof of concept for targeting B7-H3 in SCCHN. Methods: Immunohistochemistry studies were performed to define the expression of B7-H3 in SCCHN tissue microarrays (TMA). Single and repeat-dose in vivo efficacy studies were conducted in CD-1 nude mice with cell line-derived SCCHN human tumor xenografts to explore the relationship between Cmax, exposure and antitumor activity, and to define the minimal efficacious dose in these models. Based on results in these cell-derived xenograft (CDX) studies, in vivo efficacy studies were extended to a panel of SCCHN patient-derived xenograft (PDX) models, which more closely mimic the biological characteristics of the patient tumor and exhibit heterogenous expression of B7-H3. Results: Analysis of B7-H3 expression on a SCCHN TMA confirmed and extended previously reported expression of B7-H3 in SCCHN. Of the SCCHN samples evaluated, 90% (36/40) of the tumor samples were positive for B7-H3: 35% (14/40) had H-scores greater than 200, with the remaining 22 samples equally distributed between the H-score range of 101-200 and 1-100 (~ 28% each). MGC018 demonstrated specific, dose-dependent in vitro cytotoxicity toward SCCHN human tumor cell lines. The in vitro cytotoxicity translated to potent antitumor activity in vivo against SCCHN CDX models, with a single administration of 3 mg/kg resulting in complete responses in 7/7 mice in the FaDu model. In the PDX setting (H-scores 120-283), repeat dose administration every week or two weeks with MGC018 at 3 mg/kg/dose, led to regressions and/or stable disease in 10/18 models, and a delay in tumor growth in 5 additional models. Conclusion: B7-H3 is frequently overexpressed in SCCHN. At clinically relevant dose levels, MGC018 demonstrated potent antitumor activity in vivo toward SCCHN CDX mouse models and the majority of SCCHN PDX mouse models examined. These results support SCCHN as a potential indication that may be responsive to ADC-based treatments directed toward B7-H3. Citation Format: Juniper A. Scribner, Francine Z. Chen, Ying Li, Michael Chiechi, Thomas Son, Jeff Hooley, Scott Koe
头颈部鳞状细胞癌(SCCHN)是全球第七大常见癌症,占美国所有癌症的4%。虽然pd -1定向治疗对SCCHN有效,但85-95%的患者在初始反应后进展。B7- h3是免疫调节分子B7家族的成员,在SCCHN中过表达,与疾病严重程度和不良临床预后相关。此外,与推测的共抑制功能一致,B7-H3在SCCHN中的表达与肿瘤浸润CD8+ t细胞的数量呈负相关。我们正在开发针对B7-H3的治疗药物,包括enoblituzumab(一种fc工程抗B7-H3单克隆抗体)和MGC018(一种基于多卡霉素的B7-H3 ADC),这两种药物目前都在临床研究中进行评估。本研究在临床前模型中探索了MGC018的潜力,作为靶向B7-H3治疗SCCHN的概念证明。方法:采用免疫组化方法确定B7-H3在SCCHN组织微阵列(TMA)中的表达。在CD-1裸鼠身上进行单次和重复剂量的体内药效研究,探讨Cmax、暴露与抗肿瘤活性之间的关系,并确定这些模型的最小有效剂量。基于这些细胞源性异种移植物(CDX)研究的结果,体内疗效研究扩展到一组SCCHN患者源性异种移植物(PDX)模型,这些模型更接近于模拟患者肿瘤的生物学特性,并表现出B7-H3的异质表达。结果:B7-H3在SCCHN TMA上的表达分析证实并扩展了先前报道的B7-H3在SCCHN中的表达。在评估的SCCHN样本中,90%(36/40)的肿瘤样本B7-H3阳性,35%(14/40)的肿瘤样本h评分大于200,其余22个样本平均分布在101-200和1-100之间(各约28%)。MGC018对SCCHN人肿瘤细胞系具有特异性、剂量依赖性的体外细胞毒性。体外细胞毒性转化为体内对SCCHN CDX模型的有效抗肿瘤活性,单次给药3mg /kg, FaDu模型中7/7只小鼠完全缓解。在PDX组(h评分120-283)中,每周或每两周重复给药3mg /kg/剂量的MGC018,在10/18模型中导致疾病消退和/或稳定,在另外5个模型中导致肿瘤生长延迟。结论:B7-H3在SCCHN中高表达。在临床相关剂量水平下,MGC018在体内对SCCHN CDX小鼠模型和大多数SCCHN PDX小鼠模型显示出有效的抗肿瘤活性。这些结果支持SCCHN作为一种潜在的适应症,可能对针对B7-H3的adc治疗有反应。引文格式:Juniper A. Scribner, Francine Z. Chen, Ying Li, Michael Chiechi, Thomas Son, Jeff Hooley, Scott Koenig, Paul A. Moore, Ezio Bonvini, Chet Bohac, Deryk Loo。靶向B7-H3治疗头颈部鳞状细胞癌:抗B7-H3抗体-药物偶联物MGC018的临床前概念验证[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950期。
{"title":"Abstract 950: Targeting B7-H3 in squamous cell carcinoma of the head and neck: Preclinical proof-of-concept with the investigational anti-B7-H3 antibody-drug conjugate, MGC018","authors":"Juniper A Scribner, Francine Z Chen, Ying Li, M. Chiechi, T. Son, J. Hooley, S. Koenig, P. Moore, E. Bonvini, C. Bohac, D. Loo","doi":"10.1158/1538-7445.AM2021-950","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-950","url":null,"abstract":"Introduction: Squamous cell carcinoma of the head and neck (SCCHN) is the seventh most common cancer worldwide, accounting for ~ 4% of all cancers in the United States. Although PD-1-directed therapy has efficacy in SCCHN, 85-95% of patients progress following initial response. B7-H3 is a member of the B7 family of immunomodulatory molecules, is overexpressed in SCCHN, and correlates with disease severity and poor clinical outcome. Furthermore, consistent with its putative coinhibitory function, B7-H3 expression in SCCHN is inversely correlated with the number of tumor infiltrating CD8+ T-cells. We are developing therapeutics targeting B7-H3, including enoblituzumab, an Fc-engineered anti-B7-H3 monoclonal antibody, and MGC018, a duocarmycin-based B7-H3 ADC, both of which are currently being evaluated in clinical studies. Here the potential of MGC018 was explored in preclinical models as a proof of concept for targeting B7-H3 in SCCHN. Methods: Immunohistochemistry studies were performed to define the expression of B7-H3 in SCCHN tissue microarrays (TMA). Single and repeat-dose in vivo efficacy studies were conducted in CD-1 nude mice with cell line-derived SCCHN human tumor xenografts to explore the relationship between Cmax, exposure and antitumor activity, and to define the minimal efficacious dose in these models. Based on results in these cell-derived xenograft (CDX) studies, in vivo efficacy studies were extended to a panel of SCCHN patient-derived xenograft (PDX) models, which more closely mimic the biological characteristics of the patient tumor and exhibit heterogenous expression of B7-H3. Results: Analysis of B7-H3 expression on a SCCHN TMA confirmed and extended previously reported expression of B7-H3 in SCCHN. Of the SCCHN samples evaluated, 90% (36/40) of the tumor samples were positive for B7-H3: 35% (14/40) had H-scores greater than 200, with the remaining 22 samples equally distributed between the H-score range of 101-200 and 1-100 (~ 28% each). MGC018 demonstrated specific, dose-dependent in vitro cytotoxicity toward SCCHN human tumor cell lines. The in vitro cytotoxicity translated to potent antitumor activity in vivo against SCCHN CDX models, with a single administration of 3 mg/kg resulting in complete responses in 7/7 mice in the FaDu model. In the PDX setting (H-scores 120-283), repeat dose administration every week or two weeks with MGC018 at 3 mg/kg/dose, led to regressions and/or stable disease in 10/18 models, and a delay in tumor growth in 5 additional models. Conclusion: B7-H3 is frequently overexpressed in SCCHN. At clinically relevant dose levels, MGC018 demonstrated potent antitumor activity in vivo toward SCCHN CDX mouse models and the majority of SCCHN PDX mouse models examined. These results support SCCHN as a potential indication that may be responsive to ADC-based treatments directed toward B7-H3. Citation Format: Juniper A. Scribner, Francine Z. Chen, Ying Li, Michael Chiechi, Thomas Son, Jeff Hooley, Scott Koe","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":"82300927","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}
Pub Date : 2021-07-01DOI: 10.1158/1538-7445.AM2021-1171
Brenna M. Marks, Mengshi Li, Edwin A. Sagastume, M. Schultz, Frances L. Johnson
{"title":"Abstract 1171: Assessing Melanocortin 1 receptor as a target for metastatic melanoma drug delivery","authors":"Brenna M. Marks, Mengshi Li, Edwin A. Sagastume, M. Schultz, Frances L. Johnson","doi":"10.1158/1538-7445.AM2021-1171","DOIUrl":"https://doi.org/10.1158/1538-7445.AM2021-1171","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":"82463225","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}