Cargando…

Nanoparticles carrying paclitaxel and anti-miR-221 for breast cancer therapy triggered by ultrasound

Nanomaterials have been well demonstrated to have the potential to be used for tumor cell-targeted drug delivery. Targeted inhibition of miR-221 was proved to promote the sensitivity of triple genitive breast cancer (TNBC) cells to chemo-drugs. In order to improve the chemotherapeutic effect in TNBC...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Libo, Ren, Zhen, Lü, Jinhui, Mo, Xinhai, Lin, Jie, Li, Ya, Ma, Wenjing, Liu, Pengfei, Shen, Yajing, Zhao, Qian, Qian, Lu, Cheng, Xiaoxin, Yu, Zuoren, Zhang, Bo
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10427607/
https://www.ncbi.nlm.nih.gov/pubmed/37582832
http://dx.doi.org/10.1038/s41420-023-01594-9
_version_ 1785090275455008768
author Zhang, Libo
Ren, Zhen
Lü, Jinhui
Mo, Xinhai
Lin, Jie
Li, Ya
Ma, Wenjing
Liu, Pengfei
Shen, Yajing
Zhao, Qian
Qian, Lu
Cheng, Xiaoxin
Yu, Zuoren
Zhang, Bo
author_facet Zhang, Libo
Ren, Zhen
Lü, Jinhui
Mo, Xinhai
Lin, Jie
Li, Ya
Ma, Wenjing
Liu, Pengfei
Shen, Yajing
Zhao, Qian
Qian, Lu
Cheng, Xiaoxin
Yu, Zuoren
Zhang, Bo
author_sort Zhang, Libo
collection PubMed
description Nanomaterials have been well demonstrated to have the potential to be used for tumor cell-targeted drug delivery. Targeted inhibition of miR-221 was proved to promote the sensitivity of triple genitive breast cancer (TNBC) cells to chemo-drugs. In order to improve the chemotherapeutic effect in TNBC, herein, we developed a novel kind of nanoparticles shelled with PLGA and loaded with perfluoropentane (PFP), paclitaxel (PTX), and anti-miR-221 inhibitor, which was named PANP. Ultrasound-triggered vaporization of PFP in PANPs was utilized for real-time imaging track of the nanoparticles in vivo. In addition, macrophages were applied for the internalization of PANPs to form RAW-PANP with strong chemotaxis to accumulate around cancer cells. Nanoparticles with different contents did not cause M2 polarization compared with the control group but caused polarization toward M1. We compared the inherent tumor-homing behavior of macrophages containing different contents with that of normal macrophages and no significant abnormalities were observed. After injection into the tumor-burden mice, RAW-PANPs showed enrichment within tumor tissues. Upon the ultrasound cavitation-triggered burst, PTX was released in the tumor. Meanwhile, the release of anti-miR-221 improved the sensitivity of tumor cells to PTX. As a result, RAW-PANPs showed high efficiency in suppressing TNBC cell proliferation in vitro and inhibiting tumor growth and progression in vivo. The treatments did not induce liver, heart, or kidney injury. In conclusion, the current study not only developed a macrophage-carried, ultrasound-triggered, cancer cell-targeted chemotherapeutic system, but also demonstrated a miRNA-based technique to promote drug sensitivity of cancer cells, which holds strong potential to treat patients with TNBC, especially for those suffering drug-resistance. The innovation of this study is to use macrophages to deliver nanoparticles to the tumors and then use ultrasound locally to burst the nanoparticles to release the miRNA and PTX.
format Online
Article
Text
id pubmed-10427607
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-104276072023-08-17 Nanoparticles carrying paclitaxel and anti-miR-221 for breast cancer therapy triggered by ultrasound Zhang, Libo Ren, Zhen Lü, Jinhui Mo, Xinhai Lin, Jie Li, Ya Ma, Wenjing Liu, Pengfei Shen, Yajing Zhao, Qian Qian, Lu Cheng, Xiaoxin Yu, Zuoren Zhang, Bo Cell Death Discov Article Nanomaterials have been well demonstrated to have the potential to be used for tumor cell-targeted drug delivery. Targeted inhibition of miR-221 was proved to promote the sensitivity of triple genitive breast cancer (TNBC) cells to chemo-drugs. In order to improve the chemotherapeutic effect in TNBC, herein, we developed a novel kind of nanoparticles shelled with PLGA and loaded with perfluoropentane (PFP), paclitaxel (PTX), and anti-miR-221 inhibitor, which was named PANP. Ultrasound-triggered vaporization of PFP in PANPs was utilized for real-time imaging track of the nanoparticles in vivo. In addition, macrophages were applied for the internalization of PANPs to form RAW-PANP with strong chemotaxis to accumulate around cancer cells. Nanoparticles with different contents did not cause M2 polarization compared with the control group but caused polarization toward M1. We compared the inherent tumor-homing behavior of macrophages containing different contents with that of normal macrophages and no significant abnormalities were observed. After injection into the tumor-burden mice, RAW-PANPs showed enrichment within tumor tissues. Upon the ultrasound cavitation-triggered burst, PTX was released in the tumor. Meanwhile, the release of anti-miR-221 improved the sensitivity of tumor cells to PTX. As a result, RAW-PANPs showed high efficiency in suppressing TNBC cell proliferation in vitro and inhibiting tumor growth and progression in vivo. The treatments did not induce liver, heart, or kidney injury. In conclusion, the current study not only developed a macrophage-carried, ultrasound-triggered, cancer cell-targeted chemotherapeutic system, but also demonstrated a miRNA-based technique to promote drug sensitivity of cancer cells, which holds strong potential to treat patients with TNBC, especially for those suffering drug-resistance. The innovation of this study is to use macrophages to deliver nanoparticles to the tumors and then use ultrasound locally to burst the nanoparticles to release the miRNA and PTX. Nature Publishing Group UK 2023-08-15 /pmc/articles/PMC10427607/ /pubmed/37582832 http://dx.doi.org/10.1038/s41420-023-01594-9 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Zhang, Libo
Ren, Zhen
Lü, Jinhui
Mo, Xinhai
Lin, Jie
Li, Ya
Ma, Wenjing
Liu, Pengfei
Shen, Yajing
Zhao, Qian
Qian, Lu
Cheng, Xiaoxin
Yu, Zuoren
Zhang, Bo
Nanoparticles carrying paclitaxel and anti-miR-221 for breast cancer therapy triggered by ultrasound
title Nanoparticles carrying paclitaxel and anti-miR-221 for breast cancer therapy triggered by ultrasound
title_full Nanoparticles carrying paclitaxel and anti-miR-221 for breast cancer therapy triggered by ultrasound
title_fullStr Nanoparticles carrying paclitaxel and anti-miR-221 for breast cancer therapy triggered by ultrasound
title_full_unstemmed Nanoparticles carrying paclitaxel and anti-miR-221 for breast cancer therapy triggered by ultrasound
title_short Nanoparticles carrying paclitaxel and anti-miR-221 for breast cancer therapy triggered by ultrasound
title_sort nanoparticles carrying paclitaxel and anti-mir-221 for breast cancer therapy triggered by ultrasound
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10427607/
https://www.ncbi.nlm.nih.gov/pubmed/37582832
http://dx.doi.org/10.1038/s41420-023-01594-9
work_keys_str_mv AT zhanglibo nanoparticlescarryingpaclitaxelandantimir221forbreastcancertherapytriggeredbyultrasound
AT renzhen nanoparticlescarryingpaclitaxelandantimir221forbreastcancertherapytriggeredbyultrasound
AT lujinhui nanoparticlescarryingpaclitaxelandantimir221forbreastcancertherapytriggeredbyultrasound
AT moxinhai nanoparticlescarryingpaclitaxelandantimir221forbreastcancertherapytriggeredbyultrasound
AT linjie nanoparticlescarryingpaclitaxelandantimir221forbreastcancertherapytriggeredbyultrasound
AT liya nanoparticlescarryingpaclitaxelandantimir221forbreastcancertherapytriggeredbyultrasound
AT mawenjing nanoparticlescarryingpaclitaxelandantimir221forbreastcancertherapytriggeredbyultrasound
AT liupengfei nanoparticlescarryingpaclitaxelandantimir221forbreastcancertherapytriggeredbyultrasound
AT shenyajing nanoparticlescarryingpaclitaxelandantimir221forbreastcancertherapytriggeredbyultrasound
AT zhaoqian nanoparticlescarryingpaclitaxelandantimir221forbreastcancertherapytriggeredbyultrasound
AT qianlu nanoparticlescarryingpaclitaxelandantimir221forbreastcancertherapytriggeredbyultrasound
AT chengxiaoxin nanoparticlescarryingpaclitaxelandantimir221forbreastcancertherapytriggeredbyultrasound
AT yuzuoren nanoparticlescarryingpaclitaxelandantimir221forbreastcancertherapytriggeredbyultrasound
AT zhangbo nanoparticlescarryingpaclitaxelandantimir221forbreastcancertherapytriggeredbyultrasound