Cargando…

Combined Prussian Blue Nanozyme Carriers Improve Photodynamic Therapy and Effective Interruption of Tumor Metastasis

INTRODUCTION: Photodynamic therapy (PDT) as a new technique for theranostics is to kill tumor cells by activating photosensitizer and interacting with oxygen (O(2)) to produce reactive oxygen species (ROS). However, the hypoxic tumor microenvironment (TME) may constrain the efficacy of PDT. Moreover...

Descripción completa

Detalles Bibliográficos
Autores principales: Shen, Wenhao, Han, Gaohua, Yu, Lei, Yang, Song, Li, Xiangyi, Zhang, Wei, Pei, Pei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Dove 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8964450/
https://www.ncbi.nlm.nih.gov/pubmed/35369032
http://dx.doi.org/10.2147/IJN.S359156
_version_ 1784678221730545664
author Shen, Wenhao
Han, Gaohua
Yu, Lei
Yang, Song
Li, Xiangyi
Zhang, Wei
Pei, Pei
author_facet Shen, Wenhao
Han, Gaohua
Yu, Lei
Yang, Song
Li, Xiangyi
Zhang, Wei
Pei, Pei
author_sort Shen, Wenhao
collection PubMed
description INTRODUCTION: Photodynamic therapy (PDT) as a new technique for theranostics is to kill tumor cells by activating photosensitizer and interacting with oxygen (O(2)) to produce reactive oxygen species (ROS). However, the hypoxic tumor microenvironment (TME) may constrain the efficacy of PDT. Moreover, the lack of O(2) in TME also up-regulates the expression of HIF-1α and promotes tumor metastasis, which is also a leading cause of death for terminal cancer patients. METHODS: Prussian blue (PBs) was firstly synthesized by hydrothermal method, which was then etched by hydrochloric acid to obtained hollow Prussian blue nanoparticles (HPBs). Afterwards, Au-Pt nanozymes were in situ growing on the HPBs by reduction method to prepare Au-Pt@HPBs (APHPBs). Owing to the hollow structure of APHPBs, photosensitizer Ce6 can be easily and efficiently loaded into it to obtain Ce6-Au-Pt@HPBs (Ce6-APHPBs). After ce6-APHPBS regulation, photoacoustic imaging and hypoxic fluorescence imaging were then used to evaluate changes in hypoxic TME in vivo. Finally, under the assistant of Ce6-APHPBs, we evaluated the inhibitory effect of enhanced PDT on primary and metastatic tumors. RESULTS: We first designed and synthesized Ce6 loaded hollow prussian blue nanoparticles with Au-Pt nanozymes grown in situ on it. Both in vitro and in vivo experiments show that the prepared Ce6-APHPBs have good biosafety and could effectively degrade the overexpressed H(2)O(2) in TME to generate O(2), further relieve the hypoxic TME and thus enhance the effect of PDT. At the same time, the increasing O(2) content could also reduce the expression of HIF-1α at the tumor site, which could reduce lung metastasis. CONCLUSION: Ce6-APHPBs designed by us could not only efficiently enhance PDT but also regulate TME to reduce tumor metastasis and prolong survival of mice, which provide a novel idea and strategy for clinical PDT and metastatic tumor.
format Online
Article
Text
id pubmed-8964450
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Dove
record_format MEDLINE/PubMed
spelling pubmed-89644502022-03-31 Combined Prussian Blue Nanozyme Carriers Improve Photodynamic Therapy and Effective Interruption of Tumor Metastasis Shen, Wenhao Han, Gaohua Yu, Lei Yang, Song Li, Xiangyi Zhang, Wei Pei, Pei Int J Nanomedicine Original Research INTRODUCTION: Photodynamic therapy (PDT) as a new technique for theranostics is to kill tumor cells by activating photosensitizer and interacting with oxygen (O(2)) to produce reactive oxygen species (ROS). However, the hypoxic tumor microenvironment (TME) may constrain the efficacy of PDT. Moreover, the lack of O(2) in TME also up-regulates the expression of HIF-1α and promotes tumor metastasis, which is also a leading cause of death for terminal cancer patients. METHODS: Prussian blue (PBs) was firstly synthesized by hydrothermal method, which was then etched by hydrochloric acid to obtained hollow Prussian blue nanoparticles (HPBs). Afterwards, Au-Pt nanozymes were in situ growing on the HPBs by reduction method to prepare Au-Pt@HPBs (APHPBs). Owing to the hollow structure of APHPBs, photosensitizer Ce6 can be easily and efficiently loaded into it to obtain Ce6-Au-Pt@HPBs (Ce6-APHPBs). After ce6-APHPBS regulation, photoacoustic imaging and hypoxic fluorescence imaging were then used to evaluate changes in hypoxic TME in vivo. Finally, under the assistant of Ce6-APHPBs, we evaluated the inhibitory effect of enhanced PDT on primary and metastatic tumors. RESULTS: We first designed and synthesized Ce6 loaded hollow prussian blue nanoparticles with Au-Pt nanozymes grown in situ on it. Both in vitro and in vivo experiments show that the prepared Ce6-APHPBs have good biosafety and could effectively degrade the overexpressed H(2)O(2) in TME to generate O(2), further relieve the hypoxic TME and thus enhance the effect of PDT. At the same time, the increasing O(2) content could also reduce the expression of HIF-1α at the tumor site, which could reduce lung metastasis. CONCLUSION: Ce6-APHPBs designed by us could not only efficiently enhance PDT but also regulate TME to reduce tumor metastasis and prolong survival of mice, which provide a novel idea and strategy for clinical PDT and metastatic tumor. Dove 2022-03-25 /pmc/articles/PMC8964450/ /pubmed/35369032 http://dx.doi.org/10.2147/IJN.S359156 Text en © 2022 Shen et al. https://creativecommons.org/licenses/by-nc/3.0/This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/ (https://creativecommons.org/licenses/by-nc/3.0/) ). By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms (https://www.dovepress.com/terms.php).
spellingShingle Original Research
Shen, Wenhao
Han, Gaohua
Yu, Lei
Yang, Song
Li, Xiangyi
Zhang, Wei
Pei, Pei
Combined Prussian Blue Nanozyme Carriers Improve Photodynamic Therapy and Effective Interruption of Tumor Metastasis
title Combined Prussian Blue Nanozyme Carriers Improve Photodynamic Therapy and Effective Interruption of Tumor Metastasis
title_full Combined Prussian Blue Nanozyme Carriers Improve Photodynamic Therapy and Effective Interruption of Tumor Metastasis
title_fullStr Combined Prussian Blue Nanozyme Carriers Improve Photodynamic Therapy and Effective Interruption of Tumor Metastasis
title_full_unstemmed Combined Prussian Blue Nanozyme Carriers Improve Photodynamic Therapy and Effective Interruption of Tumor Metastasis
title_short Combined Prussian Blue Nanozyme Carriers Improve Photodynamic Therapy and Effective Interruption of Tumor Metastasis
title_sort combined prussian blue nanozyme carriers improve photodynamic therapy and effective interruption of tumor metastasis
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8964450/
https://www.ncbi.nlm.nih.gov/pubmed/35369032
http://dx.doi.org/10.2147/IJN.S359156
work_keys_str_mv AT shenwenhao combinedprussianbluenanozymecarriersimprovephotodynamictherapyandeffectiveinterruptionoftumormetastasis
AT hangaohua combinedprussianbluenanozymecarriersimprovephotodynamictherapyandeffectiveinterruptionoftumormetastasis
AT yulei combinedprussianbluenanozymecarriersimprovephotodynamictherapyandeffectiveinterruptionoftumormetastasis
AT yangsong combinedprussianbluenanozymecarriersimprovephotodynamictherapyandeffectiveinterruptionoftumormetastasis
AT lixiangyi combinedprussianbluenanozymecarriersimprovephotodynamictherapyandeffectiveinterruptionoftumormetastasis
AT zhangwei combinedprussianbluenanozymecarriersimprovephotodynamictherapyandeffectiveinterruptionoftumormetastasis
AT peipei combinedprussianbluenanozymecarriersimprovephotodynamictherapyandeffectiveinterruptionoftumormetastasis