关于炎症靶向纳米配体在大脑和其他靶器官中的安全性、药代动力学和生物分布的大型和小型动物研究

Sydney Risen, Breonna Kusick, Sadhana Sharma, Vincenzo S. Gilberto, Stephen Brindley, Mikayla Aguilar, Jared M. Brown, Stephanie McGrath, Anushree Chatterjee, Julie A. Moreno, Prashant Nagpal
{"title":"关于炎症靶向纳米配体在大脑和其他靶器官中的安全性、药代动力学和生物分布的大型和小型动物研究","authors":"Sydney Risen, Breonna Kusick, Sadhana Sharma, Vincenzo S. Gilberto, Stephen Brindley, Mikayla Aguilar, Jared M. Brown, Stephanie McGrath, Anushree Chatterjee, Julie A. Moreno, Prashant Nagpal","doi":"10.1021/acsptsci.4c00068","DOIUrl":null,"url":null,"abstract":"Immune malfunction or misrecognition of healthy cells and tissue, termed autoimmune disease, is implicated in more than 80 disease conditions and multiple other secondary pathologies. While pan-immunosuppressive therapies like steroids can offer limited relief for systemic inflammation for some organs, many patients never achieve remission, and such drugs do not cross the blood–brain barrier, making them ineffective for tackling neuroinflammation. Especially in the brain, unintended activation of microglia and astrocytes is hypothesized to be directly or indirectly responsible for multiple sclerosis, amyotrophic lateral sclerosis, Parkinson’s disease, and Alzheimer’s disease. Recent studies have also shown that targeting inflammasomes and specific immune targets can be beneficial for these diseases. Furthermore, our previous studies have shown targeting NF-κB and NLRP3 through brain penetrant Nanoligomer cocktail SB_NI_112 (abbreviated as NI112) can be therapeutic for several neurodegenerative diseases. Here, we show safety-toxicity studies, followed by pharmacokinetics and biodistribution in small- (mice) and large-animal (dog) studies of this inflammasome-targeting Nanoligomer cocktail NI112. We conducted studies using four different routes of administration: intravenous, subcutaneous, intraperitoneal, and intranasal, and identified the drug concentration over time using inductively coupled plasma mass spectrometry in the blood serum, the brain (including different brain regions), and other target organs such as liver, kidney, and colon. Our results indicate that the Nanoligomer cocktail has a strong safety profile and shows high biodistribution (<i>F</i> ∼ 0.98) and delivery across multiple routes of administration. Further analysis showed high brain bioavailability with a ratio of NI112 in brain tissue to blood serum of ∼30%. Our model accurately shows dose scaling, translation between different routes of administration, and interspecies scaling. These results provide an excellent platform for human clinical translation and prediction of therapeutic dosage between different routes of administration.","PeriodicalId":501473,"journal":{"name":"ACS Pharmacology & Translational Science","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Large- and Small-Animal Studies of Safety, Pharmacokinetics, and Biodistribution of Inflammasome-Targeting Nanoligomer in the Brain and Other Target Organs\",\"authors\":\"Sydney Risen, Breonna Kusick, Sadhana Sharma, Vincenzo S. Gilberto, Stephen Brindley, Mikayla Aguilar, Jared M. Brown, Stephanie McGrath, Anushree Chatterjee, Julie A. Moreno, Prashant Nagpal\",\"doi\":\"10.1021/acsptsci.4c00068\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"Immune malfunction or misrecognition of healthy cells and tissue, termed autoimmune disease, is implicated in more than 80 disease conditions and multiple other secondary pathologies. While pan-immunosuppressive therapies like steroids can offer limited relief for systemic inflammation for some organs, many patients never achieve remission, and such drugs do not cross the blood–brain barrier, making them ineffective for tackling neuroinflammation. Especially in the brain, unintended activation of microglia and astrocytes is hypothesized to be directly or indirectly responsible for multiple sclerosis, amyotrophic lateral sclerosis, Parkinson’s disease, and Alzheimer’s disease. Recent studies have also shown that targeting inflammasomes and specific immune targets can be beneficial for these diseases. Furthermore, our previous studies have shown targeting NF-κB and NLRP3 through brain penetrant Nanoligomer cocktail SB_NI_112 (abbreviated as NI112) can be therapeutic for several neurodegenerative diseases. Here, we show safety-toxicity studies, followed by pharmacokinetics and biodistribution in small- (mice) and large-animal (dog) studies of this inflammasome-targeting Nanoligomer cocktail NI112. We conducted studies using four different routes of administration: intravenous, subcutaneous, intraperitoneal, and intranasal, and identified the drug concentration over time using inductively coupled plasma mass spectrometry in the blood serum, the brain (including different brain regions), and other target organs such as liver, kidney, and colon. Our results indicate that the Nanoligomer cocktail has a strong safety profile and shows high biodistribution (<i>F</i> ∼ 0.98) and delivery across multiple routes of administration. Further analysis showed high brain bioavailability with a ratio of NI112 in brain tissue to blood serum of ∼30%. Our model accurately shows dose scaling, translation between different routes of administration, and interspecies scaling. These results provide an excellent platform for human clinical translation and prediction of therapeutic dosage between different routes of administration.\",\"PeriodicalId\":501473,\"journal\":{\"name\":\"ACS Pharmacology & Translational Science\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2024-03-21\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"ACS Pharmacology & Translational Science\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.1021/acsptsci.4c00068\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"ACS Pharmacology & Translational Science","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1021/acsptsci.4c00068","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

摘要

免疫功能失调或对健康细胞和组织的错误识别被称为自身免疫性疾病,与 80 多种疾病和其他多种继发性病症有关。虽然类固醇等泛免疫抑制疗法可以有限地缓解某些器官的全身炎症,但许多患者的病情从未得到缓解,而且这类药物不能穿过血脑屏障,因此无法有效解决神经炎症问题。特别是在大脑中,小胶质细胞和星形胶质细胞的意外激活被认为是多发性硬化症、肌萎缩侧索硬化症、帕金森病和阿尔茨海默病的直接或间接原因。最近的研究也表明,靶向炎性体和特定免疫靶点对这些疾病有益。此外,我们之前的研究表明,通过脑穿透纳米配体鸡尾酒 SB_NI_112(缩写为 NI112)靶向 NF-κB 和 NLRP3 可以治疗多种神经退行性疾病。在此,我们展示了该炎症体靶向纳米配体鸡尾酒 NI112 的安全性-毒性研究,以及在小动物(小鼠)和大动物(狗)中的药代动力学和生物分布研究。我们使用四种不同的给药途径进行了研究:静脉注射、皮下注射、腹腔注射和鼻内注射,并使用电感耦合等离子体质谱法确定了血清、大脑(包括不同脑区)和其他靶器官(如肝脏、肾脏和结肠)中的药物浓度。我们的研究结果表明,纳米配体鸡尾酒具有很高的安全性,并在多种给药途径中显示出较高的生物分布(F ∼ 0.98)和给药能力。进一步的分析表明,NI112 在脑组织和血清中的生物利用率高达 30%。我们的模型准确地显示了剂量比例、不同给药途径之间的转换以及种间比例。这些结果为不同给药途径之间的人体临床转化和治疗剂量预测提供了一个极好的平台。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

摘要图片

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
Large- and Small-Animal Studies of Safety, Pharmacokinetics, and Biodistribution of Inflammasome-Targeting Nanoligomer in the Brain and Other Target Organs
Immune malfunction or misrecognition of healthy cells and tissue, termed autoimmune disease, is implicated in more than 80 disease conditions and multiple other secondary pathologies. While pan-immunosuppressive therapies like steroids can offer limited relief for systemic inflammation for some organs, many patients never achieve remission, and such drugs do not cross the blood–brain barrier, making them ineffective for tackling neuroinflammation. Especially in the brain, unintended activation of microglia and astrocytes is hypothesized to be directly or indirectly responsible for multiple sclerosis, amyotrophic lateral sclerosis, Parkinson’s disease, and Alzheimer’s disease. Recent studies have also shown that targeting inflammasomes and specific immune targets can be beneficial for these diseases. Furthermore, our previous studies have shown targeting NF-κB and NLRP3 through brain penetrant Nanoligomer cocktail SB_NI_112 (abbreviated as NI112) can be therapeutic for several neurodegenerative diseases. Here, we show safety-toxicity studies, followed by pharmacokinetics and biodistribution in small- (mice) and large-animal (dog) studies of this inflammasome-targeting Nanoligomer cocktail NI112. We conducted studies using four different routes of administration: intravenous, subcutaneous, intraperitoneal, and intranasal, and identified the drug concentration over time using inductively coupled plasma mass spectrometry in the blood serum, the brain (including different brain regions), and other target organs such as liver, kidney, and colon. Our results indicate that the Nanoligomer cocktail has a strong safety profile and shows high biodistribution (F ∼ 0.98) and delivery across multiple routes of administration. Further analysis showed high brain bioavailability with a ratio of NI112 in brain tissue to blood serum of ∼30%. Our model accurately shows dose scaling, translation between different routes of administration, and interspecies scaling. These results provide an excellent platform for human clinical translation and prediction of therapeutic dosage between different routes of administration.
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
自引率
0.00%
发文量
0
期刊最新文献
The Hidden Hand in White Matter: Pericytes and the Puzzle of Demyelination Bovine Respiratory Syncytial Virus Nanovaccine Induces Long-Lasting Humoral Immunity in Mice Determination of Protein–Ligand Binding Affinities by Thermal Shift Assay Development of Pro-resolving and Pro-efferocytic Nanoparticles for Atherosclerosis Therapy Translational Preclinical PET Imaging and Metabolic Evaluation of a New Cannabinoid 2 Receptor (CB2R) Radioligand, (Z)-N-(3-(2-(2-[18F]Fluoroethoxy)ethyl)-4,5-dimethylthiazol-2(3H)-ylidene)-2,2,3,3-tetramethylcyclopropane-1-carboxamide
×
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