叶酸修饰的三氧化二砷药前脂质体的制备及其抗肝癌活性评价

Q3 Medicine Digital Chinese Medicine Pub Date : 2020-12-01 DOI:10.1016/j.dcmed.2020.12.005
Zhu Jing-Jing , Chen Xiao-Jie , Yao Wen-Dong , Wei Ying-Hui , Zheng Hang-Sheng , Zheng Hong-Yue , Zhu Zhi-Hong , Wang Bin-Hui , Li Fan-Zhu
{"title":"叶酸修饰的三氧化二砷药前脂质体的制备及其抗肝癌活性评价","authors":"Zhu Jing-Jing ,&nbsp;Chen Xiao-Jie ,&nbsp;Yao Wen-Dong ,&nbsp;Wei Ying-Hui ,&nbsp;Zheng Hang-Sheng ,&nbsp;Zheng Hong-Yue ,&nbsp;Zhu Zhi-Hong ,&nbsp;Wang Bin-Hui ,&nbsp;Li Fan-Zhu","doi":"10.1016/j.dcmed.2020.12.005","DOIUrl":null,"url":null,"abstract":"<div><h3>Objective</h3><p>To reduce the toxicity and side effects of arsenic trioxide (ATO) and provide a new approach for the treatment of primary liver cancer, a folic acid-modified calcium arsenite liposomal “target-controlled” drug delivery system (FA-LP-CaAs) was fabricated using the reverse microemulsion method.</p></div><div><h3>Methods</h3><p>A Malvern particle size analyzer and a transmission electron microscope were employed to determine the particle size, distribution, zeta potential and morphology of FA-LP-CaAs. Further, inductively coupled plasma emission spectrometry was employed to determine the drug loading capacity, entrapment efficiency, and <em>in vitro</em> release behavior of FA-LP-CaAs. To determine its toxicity in human hepatoma cells (HepG2) and human normal hepatocytes (LO2) and its effect on HepG2 cell cycle and apoptosis, the MTT method was used. Laser confocal and flow cytometry were also employed to determine the uptake of FA-LP-CaAs by cells. After establishing a mouse liver cancer model, the <em>in vivo</em> distribution of the drug included in the formulation was investigated using <em>in vivo</em> fluorescence. To evaluate the liver cancer targeting and anti-tumor effects of FA-LP-CaAs <em>in vivo</em>, the distribution of ATO in tissues and changes in tumor volume and body weight after liposomal administration were investigated using hematoxylin-eosin (HE)-stained tumor sections.</p></div><div><h3>Results</h3><p>The particle size, zeta potential and PDI of FA-LP-CaAs were (122.67 ± 2.18) nm, (12.81 ± 0.75) mV and 0.22 ± 0.01, respectively, while its drug loading capacity was 18.49% ± 1.14%. <em>In vitro</em> experimental results revealed that FA-LP-CaAs had a strong killing effect on HepG2 cells. Further, the cell uptake capacity of this formulation was found to improve. Based on <em>in vivo</em> assessments, FA-LP-CaAs could significantly increase the distribution of ATO in tumor sites and inhibit tumor growth.</p></div><div><h3>Conclusions</h3><p>Herein, an FA-LP-CaAs formulation was successfully fabricated. This liposomal drug delivery system had a round appearance, uniform particle size, good polydispersity coeffi-cient, evident “core-shell” structure, high drug loading capacity and pH response, tumor targeted drug delivery and sustained drug release. These findings support further research and the application of ATO as an anti-liver cancer prodrug and provide a new method for the treatment of liver cancer.</p></div>","PeriodicalId":33578,"journal":{"name":"Digital Chinese Medicine","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2020-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.dcmed.2020.12.005","citationCount":"2","resultStr":"{\"title\":\"Fabrication of A Folic Acid-Modified Arsenic Trioxide Prodrug Liposome and Assessment of its Anti-Hepatocellular Carcinoma Activity\",\"authors\":\"Zhu Jing-Jing ,&nbsp;Chen Xiao-Jie ,&nbsp;Yao Wen-Dong ,&nbsp;Wei Ying-Hui ,&nbsp;Zheng Hang-Sheng ,&nbsp;Zheng Hong-Yue ,&nbsp;Zhu Zhi-Hong ,&nbsp;Wang Bin-Hui ,&nbsp;Li Fan-Zhu\",\"doi\":\"10.1016/j.dcmed.2020.12.005\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<div><h3>Objective</h3><p>To reduce the toxicity and side effects of arsenic trioxide (ATO) and provide a new approach for the treatment of primary liver cancer, a folic acid-modified calcium arsenite liposomal “target-controlled” drug delivery system (FA-LP-CaAs) was fabricated using the reverse microemulsion method.</p></div><div><h3>Methods</h3><p>A Malvern particle size analyzer and a transmission electron microscope were employed to determine the particle size, distribution, zeta potential and morphology of FA-LP-CaAs. Further, inductively coupled plasma emission spectrometry was employed to determine the drug loading capacity, entrapment efficiency, and <em>in vitro</em> release behavior of FA-LP-CaAs. To determine its toxicity in human hepatoma cells (HepG2) and human normal hepatocytes (LO2) and its effect on HepG2 cell cycle and apoptosis, the MTT method was used. Laser confocal and flow cytometry were also employed to determine the uptake of FA-LP-CaAs by cells. After establishing a mouse liver cancer model, the <em>in vivo</em> distribution of the drug included in the formulation was investigated using <em>in vivo</em> fluorescence. To evaluate the liver cancer targeting and anti-tumor effects of FA-LP-CaAs <em>in vivo</em>, the distribution of ATO in tissues and changes in tumor volume and body weight after liposomal administration were investigated using hematoxylin-eosin (HE)-stained tumor sections.</p></div><div><h3>Results</h3><p>The particle size, zeta potential and PDI of FA-LP-CaAs were (122.67 ± 2.18) nm, (12.81 ± 0.75) mV and 0.22 ± 0.01, respectively, while its drug loading capacity was 18.49% ± 1.14%. <em>In vitro</em> experimental results revealed that FA-LP-CaAs had a strong killing effect on HepG2 cells. Further, the cell uptake capacity of this formulation was found to improve. Based on <em>in vivo</em> assessments, FA-LP-CaAs could significantly increase the distribution of ATO in tumor sites and inhibit tumor growth.</p></div><div><h3>Conclusions</h3><p>Herein, an FA-LP-CaAs formulation was successfully fabricated. This liposomal drug delivery system had a round appearance, uniform particle size, good polydispersity coeffi-cient, evident “core-shell” structure, high drug loading capacity and pH response, tumor targeted drug delivery and sustained drug release. These findings support further research and the application of ATO as an anti-liver cancer prodrug and provide a new method for the treatment of liver cancer.</p></div>\",\"PeriodicalId\":33578,\"journal\":{\"name\":\"Digital Chinese Medicine\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2020-12-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://sci-hub-pdf.com/10.1016/j.dcmed.2020.12.005\",\"citationCount\":\"2\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Digital Chinese Medicine\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://www.sciencedirect.com/science/article/pii/S2589377720300690\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q3\",\"JCRName\":\"Medicine\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Digital Chinese Medicine","FirstCategoryId":"3","ListUrlMain":"https://www.sciencedirect.com/science/article/pii/S2589377720300690","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"Medicine","Score":null,"Total":0}
引用次数: 2

摘要

目的为降低三氧化二砷(ATO)的毒副作用,为原发性肝癌的治疗提供新途径,采用反相微乳法制备叶酸修饰亚砷酸钙脂质体“靶控”给药体系(FA-LP-CaAs)。方法采用Malvern粒度分析仪和透射电镜对FA-LP-CaAs的粒径、分布、zeta电位和形貌进行测定。采用电感耦合等离子体发射光谱法测定FA-LP-CaAs的载药量、包封效率和体外释放行为。采用MTT法测定其对人肝癌细胞(HepG2)和人正常肝细胞(LO2)的毒性及对HepG2细胞周期和凋亡的影响。用激光共聚焦和流式细胞术测定细胞对FA-LP-CaAs的摄取。建立小鼠肝癌模型后,采用体内荧光法研究制剂所含药物在体内的分布。为了评估FA-LP-CaAs在体内的靶向性和抗肿瘤作用,采用苏木精-伊红(HE)染色肿瘤切片,研究了脂质体给药后ATO在组织中的分布以及肿瘤体积和体重的变化。结果FA-LP-CaAs的粒径为(122.67±2.18)nm, zeta电位为(12.81±0.75)mV, PDI为0.22±0.01,载药量为18.49%±1.14%。体外实验结果显示,FA-LP-CaAs对HepG2细胞具有较强的杀伤作用。此外,发现该配方的细胞摄取能力得到改善。根据体内评估,FA-LP-CaAs可以显著增加ATO在肿瘤部位的分布,抑制肿瘤生长。结论成功制备了FA-LP-CaAs配方。该脂质体给药系统外形圆圆,粒径均匀,多分散系数好,“核-壳”结构明显,载药能力和pH响应高,具有肿瘤靶向给药和药物缓释的特点。这些发现为ATO作为抗肝癌前药的进一步研究和应用提供了依据,为肝癌的治疗提供了一种新的方法。
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
Fabrication of A Folic Acid-Modified Arsenic Trioxide Prodrug Liposome and Assessment of its Anti-Hepatocellular Carcinoma Activity

Objective

To reduce the toxicity and side effects of arsenic trioxide (ATO) and provide a new approach for the treatment of primary liver cancer, a folic acid-modified calcium arsenite liposomal “target-controlled” drug delivery system (FA-LP-CaAs) was fabricated using the reverse microemulsion method.

Methods

A Malvern particle size analyzer and a transmission electron microscope were employed to determine the particle size, distribution, zeta potential and morphology of FA-LP-CaAs. Further, inductively coupled plasma emission spectrometry was employed to determine the drug loading capacity, entrapment efficiency, and in vitro release behavior of FA-LP-CaAs. To determine its toxicity in human hepatoma cells (HepG2) and human normal hepatocytes (LO2) and its effect on HepG2 cell cycle and apoptosis, the MTT method was used. Laser confocal and flow cytometry were also employed to determine the uptake of FA-LP-CaAs by cells. After establishing a mouse liver cancer model, the in vivo distribution of the drug included in the formulation was investigated using in vivo fluorescence. To evaluate the liver cancer targeting and anti-tumor effects of FA-LP-CaAs in vivo, the distribution of ATO in tissues and changes in tumor volume and body weight after liposomal administration were investigated using hematoxylin-eosin (HE)-stained tumor sections.

Results

The particle size, zeta potential and PDI of FA-LP-CaAs were (122.67 ± 2.18) nm, (12.81 ± 0.75) mV and 0.22 ± 0.01, respectively, while its drug loading capacity was 18.49% ± 1.14%. In vitro experimental results revealed that FA-LP-CaAs had a strong killing effect on HepG2 cells. Further, the cell uptake capacity of this formulation was found to improve. Based on in vivo assessments, FA-LP-CaAs could significantly increase the distribution of ATO in tumor sites and inhibit tumor growth.

Conclusions

Herein, an FA-LP-CaAs formulation was successfully fabricated. This liposomal drug delivery system had a round appearance, uniform particle size, good polydispersity coeffi-cient, evident “core-shell” structure, high drug loading capacity and pH response, tumor targeted drug delivery and sustained drug release. These findings support further research and the application of ATO as an anti-liver cancer prodrug and provide a new method for the treatment of liver cancer.

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
Digital Chinese Medicine
Digital Chinese Medicine Medicine-Complementary and Alternative Medicine
CiteScore
1.80
自引率
0.00%
发文量
126
审稿时长
63 days
期刊最新文献
Distribution of traditional Chinese medicine pattern types and prognostic risk factors in patients undergoing percutaneous coronary intervention (PCI): a systematic review and meta-analysis Research status and prospect of tongue image diagnosis analysis based on machine learning Thoughts on the system construction of digital Chinese medicine Differences in pulse manifestations at Cunkou based on simplified modeling of tactile sensing A novel deep learning based cloud service system for automated acupuncture needle counting: a strategy to improve acupuncture safety
×
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