摘要 3131:新型 CDH17 靶向抗体-药物共轭物在胃肠道癌症临床前模型中表现出抗肿瘤疗效

IF 12.5 1区 医学 Q1 ONCOLOGY Cancer research Pub Date : 2024-03-22 DOI:10.1158/1538-7445.am2024-3131
Po Yee Wong, Lin He, Kronos Chow, Kwan Wa Wong, Chui Yee O, Dennis Wong, J. M. Luk, Pui-Chi Lo, K. F. Wong
{"title":"摘要 3131:新型 CDH17 靶向抗体-药物共轭物在胃肠道癌症临床前模型中表现出抗肿瘤疗效","authors":"Po Yee Wong, Lin He, Kronos Chow, Kwan Wa Wong, Chui Yee O, Dennis Wong, J. M. Luk, Pui-Chi Lo, K. F. Wong","doi":"10.1158/1538-7445.am2024-3131","DOIUrl":null,"url":null,"abstract":"\n Antibody-drug conjugates (ADC) for oncology indications have been developed, however, only few of them are approved for the treatment of gastrointestinal (GI) cancers, which represent as a huge unmet medical need. As such, we developed a novel ADC of which monomethyl auristatin E (MMAE), an antimitotic agent, was conjugated to a cadherin-17 (CDH17)-targeting monoclonal antibody via protease-cleavable peptide valine-citrulline linker. CDH17 is highly restricted to the intestinal adherent junctions which is not accessible in healthy individuals. In contrast, it is overexpressed and becomes reachable in over half of the gastric and pancreatic cancer patients and in more than 95% of patients with colorectal cancer, making it a perfect target for ADC. Our ADC is designed to deliver MMAE specifically to CDH17-expressing tumors to exert the cytotoxic effect of MMAE on cancer cell, mitigating the undesired off-target cytotoxicity. LC/MS analysis revealed that MMAE was conjugated in a drug-to-antibody ratio of 4 with a yield of higher than 75%. ELISA showed that the binding of naked antibody and ADC to CDH17 was comparable (EC50, 0.17 nM & 0.3 nM, respectively), indicating the conjugation process did not affect antigen binding. This ADC also bound to different CDH17-expressing cell lines, including pancreatic AsPC1 and gastric AGS (EC50, 2.1 nM & 1.53 nM, respectively). By labelling the ADC with pH-sensitive fluorescent indicator, we illustrated that ADC internalization only occurred in CDH17+ but not CDH17- cells, suggesting the high specificity of the ADC. Similarly, the ADC showed selective killing on CDH17+ cells (EC50, 0.02-0.5 nM), and the data together proposed a positive correlation between ADC internalization level and its cytotoxicity (R2 = 0.4307). This supported the specificity and safety of the ADC in killing CDH17+ cancer cells only without affecting normal cells. Furthermore, a preliminary in-vivo study on subcutaneous AsPC1 xenograft model demonstrated that the ADC significantly suppressed tumor growth without causing any changes in body weight. Our ADC would potentially be a “First-in-class” ADC for the treatment of GI cancers with minimal adverse systemic toxicity. Further pharmacokinetics, safety, and other IND-enabling studies are underway.\n Citation Format: Po Yee Wong, Lin He, Kronos Chow, Kwan Wa Wong, Chui Yee O, Dennis Wong, John Moon Luk, Pui-Chi Lo, Kwong Fai Wong. Novel CDH17-targeting antibody-drug conjugate exhibits anti-tumor efficacy in preclinical models of gastrointestinal cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 3131.","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":null,"pages":null},"PeriodicalIF":12.5000,"publicationDate":"2024-03-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Abstract 3131: Novel CDH17-targeting antibody-drug conjugate exhibits anti-tumor efficacy in preclinical models of gastrointestinal cancers\",\"authors\":\"Po Yee Wong, Lin He, Kronos Chow, Kwan Wa Wong, Chui Yee O, Dennis Wong, J. M. Luk, Pui-Chi Lo, K. F. Wong\",\"doi\":\"10.1158/1538-7445.am2024-3131\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"\\n Antibody-drug conjugates (ADC) for oncology indications have been developed, however, only few of them are approved for the treatment of gastrointestinal (GI) cancers, which represent as a huge unmet medical need. As such, we developed a novel ADC of which monomethyl auristatin E (MMAE), an antimitotic agent, was conjugated to a cadherin-17 (CDH17)-targeting monoclonal antibody via protease-cleavable peptide valine-citrulline linker. CDH17 is highly restricted to the intestinal adherent junctions which is not accessible in healthy individuals. In contrast, it is overexpressed and becomes reachable in over half of the gastric and pancreatic cancer patients and in more than 95% of patients with colorectal cancer, making it a perfect target for ADC. Our ADC is designed to deliver MMAE specifically to CDH17-expressing tumors to exert the cytotoxic effect of MMAE on cancer cell, mitigating the undesired off-target cytotoxicity. LC/MS analysis revealed that MMAE was conjugated in a drug-to-antibody ratio of 4 with a yield of higher than 75%. ELISA showed that the binding of naked antibody and ADC to CDH17 was comparable (EC50, 0.17 nM & 0.3 nM, respectively), indicating the conjugation process did not affect antigen binding. This ADC also bound to different CDH17-expressing cell lines, including pancreatic AsPC1 and gastric AGS (EC50, 2.1 nM & 1.53 nM, respectively). By labelling the ADC with pH-sensitive fluorescent indicator, we illustrated that ADC internalization only occurred in CDH17+ but not CDH17- cells, suggesting the high specificity of the ADC. Similarly, the ADC showed selective killing on CDH17+ cells (EC50, 0.02-0.5 nM), and the data together proposed a positive correlation between ADC internalization level and its cytotoxicity (R2 = 0.4307). This supported the specificity and safety of the ADC in killing CDH17+ cancer cells only without affecting normal cells. Furthermore, a preliminary in-vivo study on subcutaneous AsPC1 xenograft model demonstrated that the ADC significantly suppressed tumor growth without causing any changes in body weight. Our ADC would potentially be a “First-in-class” ADC for the treatment of GI cancers with minimal adverse systemic toxicity. Further pharmacokinetics, safety, and other IND-enabling studies are underway.\\n Citation Format: Po Yee Wong, Lin He, Kronos Chow, Kwan Wa Wong, Chui Yee O, Dennis Wong, John Moon Luk, Pui-Chi Lo, Kwong Fai Wong. Novel CDH17-targeting antibody-drug conjugate exhibits anti-tumor efficacy in preclinical models of gastrointestinal cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 3131.\",\"PeriodicalId\":9441,\"journal\":{\"name\":\"Cancer research\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":12.5000,\"publicationDate\":\"2024-03-22\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Cancer research\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1158/1538-7445.am2024-3131\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"ONCOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer research","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1158/1538-7445.am2024-3131","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

用于肿瘤适应症的抗体药物共轭物(ADC)已经开发出来,但其中只有少数获准用于治疗胃肠道(GI)癌症,而胃肠道癌症是尚未得到满足的巨大医疗需求。因此,我们开发了一种新型 ADC,通过可被蛋白酶清除的肽缬氨酸-瓜氨酸连接体,将单甲基金丝桃素 E(MMAE)(一种抗坏死剂)与粘连蛋白-17(CDH17)靶向单克隆抗体连接在一起。CDH17 高度局限于肠道粘附连接处,健康人无法接触到。相比之下,一半以上的胃癌和胰腺癌患者以及 95% 以上的结直肠癌患者体内的 CDH17 都存在过表达和可触及性,这使其成为 ADC 的完美靶点。我们设计的 ADC 可将 MMAE 特异性地递送到表达 CDH17 的肿瘤中,从而发挥 MMAE 对癌细胞的细胞毒性作用,减少不必要的脱靶细胞毒性。LC/MS分析表明,MMAE的药物抗体比为4,产率高于75%。酶联免疫吸附试验(ELISA)显示,裸抗体和 ADC 与 CDH17 的结合率相当(EC50 分别为 0.17 nM 和 0.3 nM),表明共轭过程不影响抗原结合。这种 ADC 还能与不同的 CDH17 表达细胞系结合,包括胰腺 AsPC1 和胃 AGS(EC50 分别为 2.1 nM 和 1.53 nM)。通过用 pH 敏感荧光指示剂标记 ADC,我们发现 ADC 只在 CDH17+ 细胞中发生内化,而不在 CDH17- 细胞中发生内化,这表明 ADC 具有高度特异性。同样,ADC对CDH17+细胞具有选择性杀伤作用(EC50,0.02-0.5 nM),这些数据表明ADC内化水平与其细胞毒性呈正相关(R2 = 0.4307)。这证明了 ADC 在只杀死 CDH17+ 癌细胞而不影响正常细胞方面的特异性和安全性。此外,对皮下 AsPC1 异种移植模型进行的初步体内研究表明,ADC 能显著抑制肿瘤生长,且不会引起体重变化。我们的 ADC 有可能成为治疗消化道癌症的 "同类首创 "ADC,其不良全身毒性极低。目前正在进行进一步的药代动力学、安全性和其他 IND 研究。引用格式:Po Yee Wong, Lin He, Kronos Chow, Kwan Wa Wong, Chui Yee O, Dennis Wong, John Moon Luk, Pui-Chi Lo, Kwong Fai Wong.新型 CDH17 靶向抗体-药物共轭物在胃肠道癌症临床前模型中表现出抗肿瘤疗效 [摘要].In:美国癌症研究协会 2024 年年会论文集;第一部分(常规摘要);2024 年 4 月 5-10 日;加利福尼亚州圣地亚哥。费城(宾夕法尼亚州):AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 3131.
本文章由计算机程序翻译,如有差异,请以英文原文为准。
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Abstract 3131: Novel CDH17-targeting antibody-drug conjugate exhibits anti-tumor efficacy in preclinical models of gastrointestinal cancers
Antibody-drug conjugates (ADC) for oncology indications have been developed, however, only few of them are approved for the treatment of gastrointestinal (GI) cancers, which represent as a huge unmet medical need. As such, we developed a novel ADC of which monomethyl auristatin E (MMAE), an antimitotic agent, was conjugated to a cadherin-17 (CDH17)-targeting monoclonal antibody via protease-cleavable peptide valine-citrulline linker. CDH17 is highly restricted to the intestinal adherent junctions which is not accessible in healthy individuals. In contrast, it is overexpressed and becomes reachable in over half of the gastric and pancreatic cancer patients and in more than 95% of patients with colorectal cancer, making it a perfect target for ADC. Our ADC is designed to deliver MMAE specifically to CDH17-expressing tumors to exert the cytotoxic effect of MMAE on cancer cell, mitigating the undesired off-target cytotoxicity. LC/MS analysis revealed that MMAE was conjugated in a drug-to-antibody ratio of 4 with a yield of higher than 75%. ELISA showed that the binding of naked antibody and ADC to CDH17 was comparable (EC50, 0.17 nM & 0.3 nM, respectively), indicating the conjugation process did not affect antigen binding. This ADC also bound to different CDH17-expressing cell lines, including pancreatic AsPC1 and gastric AGS (EC50, 2.1 nM & 1.53 nM, respectively). By labelling the ADC with pH-sensitive fluorescent indicator, we illustrated that ADC internalization only occurred in CDH17+ but not CDH17- cells, suggesting the high specificity of the ADC. Similarly, the ADC showed selective killing on CDH17+ cells (EC50, 0.02-0.5 nM), and the data together proposed a positive correlation between ADC internalization level and its cytotoxicity (R2 = 0.4307). This supported the specificity and safety of the ADC in killing CDH17+ cancer cells only without affecting normal cells. Furthermore, a preliminary in-vivo study on subcutaneous AsPC1 xenograft model demonstrated that the ADC significantly suppressed tumor growth without causing any changes in body weight. Our ADC would potentially be a “First-in-class” ADC for the treatment of GI cancers with minimal adverse systemic toxicity. Further pharmacokinetics, safety, and other IND-enabling studies are underway. Citation Format: Po Yee Wong, Lin He, Kronos Chow, Kwan Wa Wong, Chui Yee O, Dennis Wong, John Moon Luk, Pui-Chi Lo, Kwong Fai Wong. Novel CDH17-targeting antibody-drug conjugate exhibits anti-tumor efficacy in preclinical models of gastrointestinal cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 3131.
求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Cancer research
Cancer research 医学-肿瘤学
CiteScore
16.10
自引率
0.90%
发文量
7677
审稿时长
2.5 months
期刊介绍: Cancer Research, published by the American Association for Cancer Research (AACR), is a journal that focuses on impactful original studies, reviews, and opinion pieces relevant to the broad cancer research community. Manuscripts that present conceptual or technological advances leading to insights into cancer biology are particularly sought after. The journal also places emphasis on convergence science, which involves bridging multiple distinct areas of cancer research. With primary subsections including Cancer Biology, Cancer Immunology, Cancer Metabolism and Molecular Mechanisms, Translational Cancer Biology, Cancer Landscapes, and Convergence Science, Cancer Research has a comprehensive scope. It is published twice a month and has one volume per year, with a print ISSN of 0008-5472 and an online ISSN of 1538-7445. Cancer Research is abstracted and/or indexed in various databases and platforms, including BIOSIS Previews (R) Database, MEDLINE, Current Contents/Life Sciences, Current Contents/Clinical Medicine, Science Citation Index, Scopus, and Web of Science.
期刊最新文献
AKT and the Hallmarks of Cancer SOS1 Inhibition Enhances the Efficacy of KRASG12C Inhibitors and Delays Resistance in Lung Adenocarcinoma Targeted Degradation of SOS1 Exhibits Potent Anticancer Activity and Overcomes Resistance in KRAS-Mutant Tumors and BCR-ABL-Positive Leukemia Distant Metastases of Breast Cancer Resemble Primary Tumors in Cancer Cell Composition but Differ in Immune Cell Phenotypes Ephrin A1 Stimulates CCL2 Secretion to Facilitate Pre-metastatic Niche Formation and Promote Gastric Cancer Liver Metastasis
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
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
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
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