甘pican-1靶向抗体-药物共轭物可抑制甘pican-1阳性胶质母细胞瘤的生长

IF 4.8 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology
Shun Uchida , Satoshi Serada , Yuji Suzuki , Eiji Funajima , Kei Kitakami , Kazumasa Dobashi , Satomi Tamatani , Yuichi Sato , Takaaki Beppu , Kuniaki Ogasawara , Testuji Naka
{"title":"甘pican-1靶向抗体-药物共轭物可抑制甘pican-1阳性胶质母细胞瘤的生长","authors":"Shun Uchida ,&nbsp;Satoshi Serada ,&nbsp;Yuji Suzuki ,&nbsp;Eiji Funajima ,&nbsp;Kei Kitakami ,&nbsp;Kazumasa Dobashi ,&nbsp;Satomi Tamatani ,&nbsp;Yuichi Sato ,&nbsp;Takaaki Beppu ,&nbsp;Kuniaki Ogasawara ,&nbsp;Testuji Naka","doi":"10.1016/j.neo.2024.100982","DOIUrl":null,"url":null,"abstract":"<div><p>Glioblastoma is the deadliest form of brain tumor. The presence of the blood–brain barrier (BBB) significantly hinders chemotherapy, necessitating the development of innovative treatment options for this tumor. This report presents the <em>in vitro</em> and <em>in vivo</em> efficacy of an antibody–drug conjugate (ADC) that targets glypican-1 (GPC1) in glioblastoma. The GPC1-ADC was created by conjugating a humanized anti-GPC1 antibody (clone T2) with monomethyl auristatin E (MMAE) <em>via</em> maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl linkers. Immunohistochemical staining analysis of a glioblastoma tissue microarray revealed that GPC1 expression was elevated in more than half of the cases. GPC1-ADC, when bound to GPC1, was efficiently and rapidly internalized in glioblastoma cell lines. It inhibited the growth of GPC1-positive glioma cell lines by inducing cell cycle arrest in the G2/M phase and triggering apoptosis <em>in vitro</em>. We established a heterotopic xenograft model by subcutaneously implanting KALS-1 and administered GPC1-ADC intravenously. GPC1-ADC significantly inhibited tumor growth and increased the number of mitotic cells. We also established an orthotopic xenograft model by intracranially implanting luciferase-transfected KS-1-Luc#19. After injecting Evans blue and resecting brain tissues, dye leakage was observed in the implantation area, confirming BBB disruption. We administered GPC1-ADC intravenously and measured the luciferase activity using an <em>in vivo</em> imaging system. GPC1-ADC significantly inhibited tumor growth and extended survival. In conclusion, GPC1-ADC demonstrated potent intracranial activity against GPC1-positive glioblastoma in an orthotopic xenograft model. These results indicate that GPC1-ADC could represent a groundbreaking new therapy for treating glioblastoma beyond the BBB.</p></div>","PeriodicalId":18917,"journal":{"name":"Neoplasia","volume":null,"pages":null},"PeriodicalIF":4.8000,"publicationDate":"2024-02-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1476558624000198/pdfft?md5=30ea273849b18d645bc33f865a23dfba&pid=1-s2.0-S1476558624000198-main.pdf","citationCount":"0","resultStr":"{\"title\":\"Glypican-1-targeted antibody–drug conjugate inhibits the growth of glypican-1-positive glioblastoma\",\"authors\":\"Shun Uchida ,&nbsp;Satoshi Serada ,&nbsp;Yuji Suzuki ,&nbsp;Eiji Funajima ,&nbsp;Kei Kitakami ,&nbsp;Kazumasa Dobashi ,&nbsp;Satomi Tamatani ,&nbsp;Yuichi Sato ,&nbsp;Takaaki Beppu ,&nbsp;Kuniaki Ogasawara ,&nbsp;Testuji Naka\",\"doi\":\"10.1016/j.neo.2024.100982\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<div><p>Glioblastoma is the deadliest form of brain tumor. The presence of the blood–brain barrier (BBB) significantly hinders chemotherapy, necessitating the development of innovative treatment options for this tumor. This report presents the <em>in vitro</em> and <em>in vivo</em> efficacy of an antibody–drug conjugate (ADC) that targets glypican-1 (GPC1) in glioblastoma. The GPC1-ADC was created by conjugating a humanized anti-GPC1 antibody (clone T2) with monomethyl auristatin E (MMAE) <em>via</em> maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl linkers. Immunohistochemical staining analysis of a glioblastoma tissue microarray revealed that GPC1 expression was elevated in more than half of the cases. GPC1-ADC, when bound to GPC1, was efficiently and rapidly internalized in glioblastoma cell lines. It inhibited the growth of GPC1-positive glioma cell lines by inducing cell cycle arrest in the G2/M phase and triggering apoptosis <em>in vitro</em>. We established a heterotopic xenograft model by subcutaneously implanting KALS-1 and administered GPC1-ADC intravenously. GPC1-ADC significantly inhibited tumor growth and increased the number of mitotic cells. We also established an orthotopic xenograft model by intracranially implanting luciferase-transfected KS-1-Luc#19. After injecting Evans blue and resecting brain tissues, dye leakage was observed in the implantation area, confirming BBB disruption. We administered GPC1-ADC intravenously and measured the luciferase activity using an <em>in vivo</em> imaging system. GPC1-ADC significantly inhibited tumor growth and extended survival. In conclusion, GPC1-ADC demonstrated potent intracranial activity against GPC1-positive glioblastoma in an orthotopic xenograft model. These results indicate that GPC1-ADC could represent a groundbreaking new therapy for treating glioblastoma beyond the BBB.</p></div>\",\"PeriodicalId\":18917,\"journal\":{\"name\":\"Neoplasia\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":4.8000,\"publicationDate\":\"2024-02-27\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.sciencedirect.com/science/article/pii/S1476558624000198/pdfft?md5=30ea273849b18d645bc33f865a23dfba&pid=1-s2.0-S1476558624000198-main.pdf\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Neoplasia\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://www.sciencedirect.com/science/article/pii/S1476558624000198\",\"RegionNum\":2,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"Biochemistry, Genetics and Molecular Biology\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Neoplasia","FirstCategoryId":"3","ListUrlMain":"https://www.sciencedirect.com/science/article/pii/S1476558624000198","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"Biochemistry, Genetics and Molecular Biology","Score":null,"Total":0}
引用次数: 0

摘要

胶质母细胞瘤是最致命的脑肿瘤。血脑屏障(BBB)的存在极大地阻碍了化疗,因此有必要针对这种肿瘤开发创新的治疗方案。本报告介绍了一种以glypican-1(GPC1)为靶点的抗体药物共轭物(ADC)在胶质母细胞瘤中的体外和体内疗效。GPC1-ADC 是通过马来酰亚胺-缬氨酸-瓜氨酸-对氨基苄氧羰基连接体将人源化的抗 GPC1 抗体(克隆 T2)与单甲基金丝桃素 E(MMAE)共轭而制成的。对胶质母细胞瘤组织芯片进行的免疫组化染色分析表明,一半以上的病例中 GPC1 表达升高。GPC1-ADC 与 GPC1 结合后,能在胶质母细胞瘤细胞系中高效、快速地内化。它在体外通过诱导细胞周期停滞在 G2/M 期和引发细胞凋亡来抑制 GPC1 阳性胶质瘤细胞株的生长。我们通过皮下注射 KALS-1 并静脉注射 GPC1-ADC 建立了异位异种移植模型。GPC1-ADC 可明显抑制肿瘤生长并增加有丝分裂细胞数量。我们还通过颅内植入荧光素酶转染的KS-1-Luc#19建立了正位异种移植模型。注射伊文思蓝并切除脑组织后,在植入区域观察到染料渗漏,证实了 BBB 破坏。我们静脉注射了 GPC1-ADC,并使用体内成像系统测量了荧光素酶活性。GPC1-ADC 能明显抑制肿瘤生长并延长存活时间。总之,在正位异种移植模型中,GPC1-ADC 对 GPC1 阳性胶质母细胞瘤表现出了强大的颅内活性。这些结果表明,GPC1-ADC 是一种突破性的新疗法,可用于治疗 BBB 外的胶质母细胞瘤。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Glypican-1-targeted antibody–drug conjugate inhibits the growth of glypican-1-positive glioblastoma

Glioblastoma is the deadliest form of brain tumor. The presence of the blood–brain barrier (BBB) significantly hinders chemotherapy, necessitating the development of innovative treatment options for this tumor. This report presents the in vitro and in vivo efficacy of an antibody–drug conjugate (ADC) that targets glypican-1 (GPC1) in glioblastoma. The GPC1-ADC was created by conjugating a humanized anti-GPC1 antibody (clone T2) with monomethyl auristatin E (MMAE) via maleimidocaproyl-valine-citrulline-p-aminobenzyloxycarbonyl linkers. Immunohistochemical staining analysis of a glioblastoma tissue microarray revealed that GPC1 expression was elevated in more than half of the cases. GPC1-ADC, when bound to GPC1, was efficiently and rapidly internalized in glioblastoma cell lines. It inhibited the growth of GPC1-positive glioma cell lines by inducing cell cycle arrest in the G2/M phase and triggering apoptosis in vitro. We established a heterotopic xenograft model by subcutaneously implanting KALS-1 and administered GPC1-ADC intravenously. GPC1-ADC significantly inhibited tumor growth and increased the number of mitotic cells. We also established an orthotopic xenograft model by intracranially implanting luciferase-transfected KS-1-Luc#19. After injecting Evans blue and resecting brain tissues, dye leakage was observed in the implantation area, confirming BBB disruption. We administered GPC1-ADC intravenously and measured the luciferase activity using an in vivo imaging system. GPC1-ADC significantly inhibited tumor growth and extended survival. In conclusion, GPC1-ADC demonstrated potent intracranial activity against GPC1-positive glioblastoma in an orthotopic xenograft model. These results indicate that GPC1-ADC could represent a groundbreaking new therapy for treating glioblastoma beyond the BBB.

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
Neoplasia
Neoplasia 医学-肿瘤学
CiteScore
9.20
自引率
2.10%
发文量
82
审稿时长
26 days
期刊介绍: Neoplasia publishes the results of novel investigations in all areas of oncology research. The title Neoplasia was chosen to convey the journal’s breadth, which encompasses the traditional disciplines of cancer research as well as emerging fields and interdisciplinary investigations. Neoplasia is interested in studies describing new molecular and genetic findings relating to the neoplastic phenotype and in laboratory and clinical studies demonstrating creative applications of advances in the basic sciences to risk assessment, prognostic indications, detection, diagnosis, and treatment. In addition to regular Research Reports, Neoplasia also publishes Reviews and Meeting Reports. Neoplasia is committed to ensuring a thorough, fair, and rapid review and publication schedule to further its mission of serving both the scientific and clinical communities by disseminating important data and ideas in cancer research.
×
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