Preclinical evaluation of an 18F-labeled Nε-acryloyllysine piperazide for covalent targeting of transglutaminase 2

IF 4.4 Q1 CHEMISTRY, INORGANIC & NUCLEAR EJNMMI Radiopharmacy and Chemistry Pub Date : 2024-01-02 DOI:10.1186/s41181-023-00231-1
Robert Wodtke, Markus Laube, Sandra Hauser, Sebastian Meister, Friedrich-Alexander Ludwig, Steffen Fischer, Klaus Kopka, Jens Pietzsch, Reik Löser
{"title":"Preclinical evaluation of an 18F-labeled Nε-acryloyllysine piperazide for covalent targeting of transglutaminase 2","authors":"Robert Wodtke,&nbsp;Markus Laube,&nbsp;Sandra Hauser,&nbsp;Sebastian Meister,&nbsp;Friedrich-Alexander Ludwig,&nbsp;Steffen Fischer,&nbsp;Klaus Kopka,&nbsp;Jens Pietzsch,&nbsp;Reik Löser","doi":"10.1186/s41181-023-00231-1","DOIUrl":null,"url":null,"abstract":"<div><h3>Background</h3><p>Transglutaminase 2 (TGase 2) is a multifunctional protein and has a prominent role in various (patho)physiological processes. In particular, its transamidase activity, which is rather latent under physiological conditions, gains importance in malignant cells. Thus, there is a great need of theranostic probes for targeting tumor-associated TGase 2, and targeted covalent inhibitors appear to be particularly attractive as vector molecules. Such an inhibitor, equipped with a radionuclide suitable for noninvasive imaging, would be supportive for answering the general question on the possibility for functional characterization of tumor-associated TGase 2. For this purpose, the recently developed <sup>18</sup>F-labeled <i>N</i><sup>ε</sup>-acryloyllysine piperazide <b>[</b><sup><b>18</b></sup><b>F]7b</b>, which is a potent and selective irreversible inhibitor of TGase 2, was subject to a detailed radiopharmacological characterization herein.</p><h3>Results</h3><p>An alternative radiosynthesis of <b>[</b><sup><b>18</b></sup><b>F]7b</b> is presented, which demands less than 300 µg of the respective trimethylammonio precursor per synthesis and provides <b>[</b><sup><b>18</b></sup><b>F]7b</b> in good radiochemical yields (17 ± 7%) and high (radio)chemical purities (≥ 99%). Ex vivo biodistribution studies in healthy mice at 5 and 60 min <i>p.i.</i> revealed no permanent enrichment of <sup>18</sup>F-activity in tissues with the exception of the bone tissue. In vivo pretreatment with ketoconazole and in vitro murine liver microsome studies complemented by mass spectrometric analysis demonstrated that bone uptake originates from metabolically released [<sup>18</sup>F]fluoride. Further metabolic transformations of <b>[</b><sup><b>18</b></sup><b>F]7b</b> include mono-hydroxylation and glucuronidation. Based on blood sampling data and liver microsome experiments, pharmacokinetic parameters such as plasma and intrinsic clearance were derived, which substantiated the apparently rapid distribution of <b>[</b><sup><b>18</b></sup><b>F]7b</b> in and elimination from the organisms. A TGase 2-mediated uptake of <b>[</b><sup><b>18</b></sup><b>F]7b</b> in different tumor cell lines could not be proven. Moreover, evaluation of <b>[</b><sup><b>18</b></sup><b>F]7b</b> in melanoma tumor xenograft models based on A375-hS100A4 (TGase 2 +) and MeWo (TGase 2 −) cells by ex vivo biodistribution and PET imaging studies were not indicative for a specific targeting.</p><h3>Conclusion</h3><p><b>[</b><sup><b>18</b></sup><b>F]7b</b> is a valuable radiometric tool to study TGase 2 in vitro under various conditions. However, its suitability for targeting tumor-associated TGase 2 is strongly limited due its unfavorable pharmacokinetic properties as demonstrated in rodents. Consequently, from a radiochemical perspective <b>[</b><sup><b>18</b></sup><b>F]7b</b> requires appropriate structural modifications to overcome these limitations.</p></div>","PeriodicalId":534,"journal":{"name":"EJNMMI Radiopharmacy and Chemistry","volume":null,"pages":null},"PeriodicalIF":4.4000,"publicationDate":"2024-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ejnmmipharmchem.springeropen.com/counter/pdf/10.1186/s41181-023-00231-1","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"EJNMMI Radiopharmacy and Chemistry","FirstCategoryId":"1085","ListUrlMain":"https://link.springer.com/article/10.1186/s41181-023-00231-1","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CHEMISTRY, INORGANIC & NUCLEAR","Score":null,"Total":0}
引用次数: 0

Abstract

Background

Transglutaminase 2 (TGase 2) is a multifunctional protein and has a prominent role in various (patho)physiological processes. In particular, its transamidase activity, which is rather latent under physiological conditions, gains importance in malignant cells. Thus, there is a great need of theranostic probes for targeting tumor-associated TGase 2, and targeted covalent inhibitors appear to be particularly attractive as vector molecules. Such an inhibitor, equipped with a radionuclide suitable for noninvasive imaging, would be supportive for answering the general question on the possibility for functional characterization of tumor-associated TGase 2. For this purpose, the recently developed 18F-labeled Nε-acryloyllysine piperazide [18F]7b, which is a potent and selective irreversible inhibitor of TGase 2, was subject to a detailed radiopharmacological characterization herein.

Results

An alternative radiosynthesis of [18F]7b is presented, which demands less than 300 µg of the respective trimethylammonio precursor per synthesis and provides [18F]7b in good radiochemical yields (17 ± 7%) and high (radio)chemical purities (≥ 99%). Ex vivo biodistribution studies in healthy mice at 5 and 60 min p.i. revealed no permanent enrichment of 18F-activity in tissues with the exception of the bone tissue. In vivo pretreatment with ketoconazole and in vitro murine liver microsome studies complemented by mass spectrometric analysis demonstrated that bone uptake originates from metabolically released [18F]fluoride. Further metabolic transformations of [18F]7b include mono-hydroxylation and glucuronidation. Based on blood sampling data and liver microsome experiments, pharmacokinetic parameters such as plasma and intrinsic clearance were derived, which substantiated the apparently rapid distribution of [18F]7b in and elimination from the organisms. A TGase 2-mediated uptake of [18F]7b in different tumor cell lines could not be proven. Moreover, evaluation of [18F]7b in melanoma tumor xenograft models based on A375-hS100A4 (TGase 2 +) and MeWo (TGase 2 −) cells by ex vivo biodistribution and PET imaging studies were not indicative for a specific targeting.

Conclusion

[18F]7b is a valuable radiometric tool to study TGase 2 in vitro under various conditions. However, its suitability for targeting tumor-associated TGase 2 is strongly limited due its unfavorable pharmacokinetic properties as demonstrated in rodents. Consequently, from a radiochemical perspective [18F]7b requires appropriate structural modifications to overcome these limitations.

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
用于共价靶向转谷氨酰胺酶 2 的 18F 标记 Nε-acryloyllysine piperazide 的临床前评估。
背景:转谷氨酰胺酶 2(TGase 2)是一种多功能蛋白质,在各种(病理)生理过程中发挥着重要作用。特别是,它在生理条件下潜伏的转酰胺酶活性在恶性细胞中变得更加重要。因此,亟需针对肿瘤相关 TGase 2 的治疗探针,而靶向共价抑制剂作为载体分子似乎特别有吸引力。这种抑制剂配有适合无创成像的放射性核素,将有助于回答关于肿瘤相关 TGase 2 功能特征描述可能性的一般性问题。为此,最近开发的 18F 标记 Nε-acryloyllysine piperazide [18F]7b 是 TGase 2 的强效选择性不可逆抑制剂,本文对其进行了详细的放射药理学表征:结果:本文介绍了[18F]7b 的另一种放射合成方法,每次合成只需不到 300 µg 的相应三甲氨基前体,而且[18F]7b 的放射化学收率高(17 ± 7%),(放射)化学纯度高(≥ 99%)。在健康小鼠体内进行的体内生物分布研究(5 分钟和 60 分钟 p.i.)显示,除骨组织外,18F 活性在其他组织中没有永久性富集。体内酮康唑预处理和体外小鼠肝微粒体研究以及质谱分析表明,骨吸收源于代谢释放的[18F]氟化物。18F]7b 的进一步代谢转化包括单羟基化和葡萄糖醛酸化。根据血液采样数据和肝脏微粒体实验,得出了血浆和内在清除率等药代动力学参数,证实了[18F]7b在生物体内的分布和消除速度明显较快。在不同的肿瘤细胞系中,TGase 2 介导的[18F]7b 吸收尚未得到证实。此外,通过体内外生物分布和 PET 成像研究,对基于 A375-hS100A4(TGase 2 +)和 MeWo(TGase 2 -)细胞的黑色素瘤异种移植模型中的 [18F]7b 进行了评估,结果并不表明其具有特定的靶向性:结论:[18F]7b 是在各种条件下研究体外 TGase 2 的重要放射性测量工具。结论:[18F]7b 是研究各种条件下体外 TGase 2 的重要放射性计量工具,但由于其在啮齿类动物中表现出的不利药代动力学特性,它在靶向肿瘤相关 TGase 2 方面的适用性受到很大限制。因此,从放射化学的角度来看,[18F]7b 需要进行适当的结构改造才能克服这些限制。
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 去求助
来源期刊
CiteScore
7.20
自引率
8.70%
发文量
30
审稿时长
5 weeks
期刊最新文献
The development of 177Lu-DOTA-CC-PSMA following a unified "Click Chemistry" protocol of synthesizing metal nuclide-conjugated radiopharmaceuticals. In vitro and in vivo analyses of eFAP: a novel FAP-targeting small molecule for radionuclide theranostics and other oncological interventions. Generic semi-automated radiofluorination strategy for single domain antibodies: [18F]FB-labelled single domain antibodies for PET imaging of fibroblast activation protein-α or folate receptor-α overexpression in cancer. Process validation and preclinical development of a new PET cerebral blood flow tracer [11C]MMP for initial clinical trials. Preclinical evaluation of CXCR4 peptides for targeted radionuclide therapy in glioblastoma
×
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