Identification of potent biparatopic antibodies targeting FGFR2 fusion driven cholangiocarcinoma.

Saireudee Chaturantabut, Sydney Oliver, Dennie T. Frederick, Jiwan Kim, Foxy P Robinson, Alessandro Sinopoli, Tian-Yu Song, Diego J Rodriguez, Liang Chang, Devishi Kesar, Yao He, Meilani Ching, Ruvimbo Dzvurumi, Adel Atari, Yuen-Yi Tseng, Nabeel Bardeesy, William R Sellers
{"title":"Identification of potent biparatopic antibodies targeting FGFR2 fusion driven cholangiocarcinoma.","authors":"Saireudee Chaturantabut, Sydney Oliver, Dennie T. Frederick, Jiwan Kim, Foxy P Robinson, Alessandro Sinopoli, Tian-Yu Song, Diego J Rodriguez, Liang Chang, Devishi Kesar, Yao He, Meilani Ching, Ruvimbo Dzvurumi, Adel Atari, Yuen-Yi Tseng, Nabeel Bardeesy, William R Sellers","doi":"10.1101/2024.09.16.613045","DOIUrl":null,"url":null,"abstract":"Translocations involving FGFR2 gene fusions are common in cholangiocarcinoma and predict response to FGFR kinase inhibitors. However, the rate and durability of response are limited due to the emergence of resistance, typically involving acquired FGFR2 kinase domain mutations, and to sub-optimal dosing, relating to drug adverse effects. Here, we report the development of biparatopic antibodies targeting the FGFR2 extracellular domain (ECD), as candidate therapeutics. Biparatopic antibodies can overcome drawbacks of standard bivalent monoparatopic antibodies, which often show poor inhibitory or even agonist activity against oncogenic receptors. We show that oncogenic transformation by FGFR2 fusions requires an intact ECD. Moreover, by systematically generating biparatopic antibodies that target distinct epitope pairs along the FGFR2 ECD, we identified antibodies that effectively block signaling and malignant growth driven by FGFR2-fusions. Importantly, these antibodies demonstrate efficacy in vivo, synergy with FGFR inhibitors, and activity against FGFR2 fusions harboring kinase domain mutations. Thus, biparatopic antibodies may serve as new treatment options for patients with FGFR2-altered cholangiocarcinoma.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"bioRxiv - Cancer Biology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1101/2024.09.16.613045","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Translocations involving FGFR2 gene fusions are common in cholangiocarcinoma and predict response to FGFR kinase inhibitors. However, the rate and durability of response are limited due to the emergence of resistance, typically involving acquired FGFR2 kinase domain mutations, and to sub-optimal dosing, relating to drug adverse effects. Here, we report the development of biparatopic antibodies targeting the FGFR2 extracellular domain (ECD), as candidate therapeutics. Biparatopic antibodies can overcome drawbacks of standard bivalent monoparatopic antibodies, which often show poor inhibitory or even agonist activity against oncogenic receptors. We show that oncogenic transformation by FGFR2 fusions requires an intact ECD. Moreover, by systematically generating biparatopic antibodies that target distinct epitope pairs along the FGFR2 ECD, we identified antibodies that effectively block signaling and malignant growth driven by FGFR2-fusions. Importantly, these antibodies demonstrate efficacy in vivo, synergy with FGFR inhibitors, and activity against FGFR2 fusions harboring kinase domain mutations. Thus, biparatopic antibodies may serve as new treatment options for patients with FGFR2-altered cholangiocarcinoma.
鉴定针对 FGFR2 融合驱动的胆管癌的强效双抗体。
涉及 FGFR2 基因融合的易位在胆管癌中很常见,可预测对 FGFR 激酶抑制剂的反应。然而,由于耐药性的出现(通常涉及获得性 FGFR2 激酶域突变)以及与药物不良反应有关的次优剂量,反应的速率和持久性受到了限制。在此,我们报告了针对表皮生长因子受体2胞外结构域(ECD)的双位点抗体作为候选疗法的开发情况。双配位抗体可以克服标准双价单配位抗体的缺点,标准双价单配位抗体通常对致癌受体的抑制甚至激动活性较差。我们的研究表明,FGFR2 融合体的致癌转化需要完整的 ECD。此外,通过系统地生成以 FGFR2 ECD 上不同表位对为靶点的双靶抗体,我们发现了能有效阻断 FGFR2 融合体信号传导和恶性生长的抗体。重要的是,这些抗体在体内具有疗效,能与表皮生长因子受体抑制剂产生协同作用,并对激酶域突变的表皮生长因子受体2融合具有活性。因此,双抗体可作为FGFR2改变胆管癌患者的新治疗方案。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
自引率
0.00%
发文量
0
×
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
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信