Cargando…

Carrier-free multifunctional nanomedicine for intraperitoneal disseminated ovarian cancer therapy

BACKGROUND: Ovarian cancer is the most lethal gynecological cancer which is characterized by extensive peritoneal implantation metastasis and malignant ascites. Despite advances in diagnosis and treatment in recent years, the five-year survival rate is only 25–30%. Therefore, developing multifunctio...

Descripción completa

Detalles Bibliográficos
Autores principales: Huang, Xiuyu, Qiu, Miaojuan, Wang, Tianqi, Li, Binbin, Zhang, Shiqiang, Zhang, Tianzhi, Liu, Peng, Wang, Qiang, Qian, Zhi Rong, Zhu, Chengming, Wu, Meiying, Zhao, Jing
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8864853/
https://www.ncbi.nlm.nih.gov/pubmed/35193583
http://dx.doi.org/10.1186/s12951-022-01300-4
_version_ 1784655537035542528
author Huang, Xiuyu
Qiu, Miaojuan
Wang, Tianqi
Li, Binbin
Zhang, Shiqiang
Zhang, Tianzhi
Liu, Peng
Wang, Qiang
Qian, Zhi Rong
Zhu, Chengming
Wu, Meiying
Zhao, Jing
author_facet Huang, Xiuyu
Qiu, Miaojuan
Wang, Tianqi
Li, Binbin
Zhang, Shiqiang
Zhang, Tianzhi
Liu, Peng
Wang, Qiang
Qian, Zhi Rong
Zhu, Chengming
Wu, Meiying
Zhao, Jing
author_sort Huang, Xiuyu
collection PubMed
description BACKGROUND: Ovarian cancer is the most lethal gynecological cancer which is characterized by extensive peritoneal implantation metastasis and malignant ascites. Despite advances in diagnosis and treatment in recent years, the five-year survival rate is only 25–30%. Therefore, developing multifunctional nanomedicine with abilities of promoting apoptosis and inhibiting migration on tumor cells would be a promising strategy to improve the antitumor effect. METHODS AND RESULTS: In this study, we developed a novel ACaT nanomedicine composed of alendronate, calcium ions and cyclin-dependent kinase 7 (CDK7) inhibitor THZ1. With the average size of 164 nm and zeta potential of 12.4 mV, the spherical ACaT nanoparticles were selectively internalized by tumor cells and effectively accumulated in the tumor site. Results of RNA-sequencing and in vitro experiments showed that ACaT promoted tumor cell apoptosis and inhibited tumor cell migration by arresting the cell cycle, increasing ROS and affecting calcium homeostasis. Weekly intraperitoneally administered of ACaT for 8 cycles significantly inhibited the growth of tumor and prolonged the survival of intraperitoneal xenograft mice. CONCLUSION: In summary, this study presents a new self-assembly nanomedicine with favorable tumor targeting, antitumor activity and good biocompatibility, providing a novel therapeutic strategy for advanced ovarian cancer. GRAPHICAL ABSTRACT: [Image: see text] SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12951-022-01300-4.
format Online
Article
Text
id pubmed-8864853
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-88648532022-02-28 Carrier-free multifunctional nanomedicine for intraperitoneal disseminated ovarian cancer therapy Huang, Xiuyu Qiu, Miaojuan Wang, Tianqi Li, Binbin Zhang, Shiqiang Zhang, Tianzhi Liu, Peng Wang, Qiang Qian, Zhi Rong Zhu, Chengming Wu, Meiying Zhao, Jing J Nanobiotechnology Research BACKGROUND: Ovarian cancer is the most lethal gynecological cancer which is characterized by extensive peritoneal implantation metastasis and malignant ascites. Despite advances in diagnosis and treatment in recent years, the five-year survival rate is only 25–30%. Therefore, developing multifunctional nanomedicine with abilities of promoting apoptosis and inhibiting migration on tumor cells would be a promising strategy to improve the antitumor effect. METHODS AND RESULTS: In this study, we developed a novel ACaT nanomedicine composed of alendronate, calcium ions and cyclin-dependent kinase 7 (CDK7) inhibitor THZ1. With the average size of 164 nm and zeta potential of 12.4 mV, the spherical ACaT nanoparticles were selectively internalized by tumor cells and effectively accumulated in the tumor site. Results of RNA-sequencing and in vitro experiments showed that ACaT promoted tumor cell apoptosis and inhibited tumor cell migration by arresting the cell cycle, increasing ROS and affecting calcium homeostasis. Weekly intraperitoneally administered of ACaT for 8 cycles significantly inhibited the growth of tumor and prolonged the survival of intraperitoneal xenograft mice. CONCLUSION: In summary, this study presents a new self-assembly nanomedicine with favorable tumor targeting, antitumor activity and good biocompatibility, providing a novel therapeutic strategy for advanced ovarian cancer. GRAPHICAL ABSTRACT: [Image: see text] SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12951-022-01300-4. BioMed Central 2022-02-22 /pmc/articles/PMC8864853/ /pubmed/35193583 http://dx.doi.org/10.1186/s12951-022-01300-4 Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open AccessThis 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 licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence 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 licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Huang, Xiuyu
Qiu, Miaojuan
Wang, Tianqi
Li, Binbin
Zhang, Shiqiang
Zhang, Tianzhi
Liu, Peng
Wang, Qiang
Qian, Zhi Rong
Zhu, Chengming
Wu, Meiying
Zhao, Jing
Carrier-free multifunctional nanomedicine for intraperitoneal disseminated ovarian cancer therapy
title Carrier-free multifunctional nanomedicine for intraperitoneal disseminated ovarian cancer therapy
title_full Carrier-free multifunctional nanomedicine for intraperitoneal disseminated ovarian cancer therapy
title_fullStr Carrier-free multifunctional nanomedicine for intraperitoneal disseminated ovarian cancer therapy
title_full_unstemmed Carrier-free multifunctional nanomedicine for intraperitoneal disseminated ovarian cancer therapy
title_short Carrier-free multifunctional nanomedicine for intraperitoneal disseminated ovarian cancer therapy
title_sort carrier-free multifunctional nanomedicine for intraperitoneal disseminated ovarian cancer therapy
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8864853/
https://www.ncbi.nlm.nih.gov/pubmed/35193583
http://dx.doi.org/10.1186/s12951-022-01300-4
work_keys_str_mv AT huangxiuyu carrierfreemultifunctionalnanomedicineforintraperitonealdisseminatedovariancancertherapy
AT qiumiaojuan carrierfreemultifunctionalnanomedicineforintraperitonealdisseminatedovariancancertherapy
AT wangtianqi carrierfreemultifunctionalnanomedicineforintraperitonealdisseminatedovariancancertherapy
AT libinbin carrierfreemultifunctionalnanomedicineforintraperitonealdisseminatedovariancancertherapy
AT zhangshiqiang carrierfreemultifunctionalnanomedicineforintraperitonealdisseminatedovariancancertherapy
AT zhangtianzhi carrierfreemultifunctionalnanomedicineforintraperitonealdisseminatedovariancancertherapy
AT liupeng carrierfreemultifunctionalnanomedicineforintraperitonealdisseminatedovariancancertherapy
AT wangqiang carrierfreemultifunctionalnanomedicineforintraperitonealdisseminatedovariancancertherapy
AT qianzhirong carrierfreemultifunctionalnanomedicineforintraperitonealdisseminatedovariancancertherapy
AT zhuchengming carrierfreemultifunctionalnanomedicineforintraperitonealdisseminatedovariancancertherapy
AT wumeiying carrierfreemultifunctionalnanomedicineforintraperitonealdisseminatedovariancancertherapy
AT zhaojing carrierfreemultifunctionalnanomedicineforintraperitonealdisseminatedovariancancertherapy