Cargando…

A New Pharmacokinetic Model Describing the Biodistribution of Intravenously and Intratumorally Administered Superparamagnetic Iron Oxide Nanoparticles (SPIONs) in a GL261 Xenograft Glioblastoma Model

BACKGROUND: Superparamagnetic iron oxide nanoparticles (SPIONs) have displayed multifunctional applications in cancer theranostics following systemic delivery. In an effort to increase the therapeutic potential of local therapies (including focal hyperthermia), nanoparticles can also be administered...

Descripción completa

Detalles Bibliográficos
Autores principales: Klapproth, Alexander P, Shevtsov, Maxim, Stangl, Stefan, Li, Wei Bo, Multhoff, Gabriele
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Dove 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7335747/
https://www.ncbi.nlm.nih.gov/pubmed/32669844
http://dx.doi.org/10.2147/IJN.S254745
_version_ 1783554189737066496
author Klapproth, Alexander P
Shevtsov, Maxim
Stangl, Stefan
Li, Wei Bo
Multhoff, Gabriele
author_facet Klapproth, Alexander P
Shevtsov, Maxim
Stangl, Stefan
Li, Wei Bo
Multhoff, Gabriele
author_sort Klapproth, Alexander P
collection PubMed
description BACKGROUND: Superparamagnetic iron oxide nanoparticles (SPIONs) have displayed multifunctional applications in cancer theranostics following systemic delivery. In an effort to increase the therapeutic potential of local therapies (including focal hyperthermia), nanoparticles can also be administered intratumorally. Therefore, the development of a reliable pharmacokinetic model for the prediction of nanoparticle distribution for both clinically relevant routes of delivery is of high importance. MATERIALS AND METHODS: The biodistribution of SPIONs (of two different sizes – 130 nm and 60 nm) radiolabeled with zirconium-89 or technetium-99m following intratumoral or intravenous injection was investigated in C57/Bl6 mice bearing subcutaneous GL261 glioblastomas. Based on PET/CT biodistribution data, a novel pharmacokinetic model was established for a better understanding of the pharmacokinetics of the SPIONs after both administration routes. RESULTS: The PET image analysis of the nanoparticles (confirmed by histology) demonstrated the presence of radiolabeled nanoparticles within the glioma site (with low amounts in the liver and spleen) at all investigated time points following intratumoral injection. The mathematical model confirmed the dynamic nanoparticle redistribution in the organism over a period of 72 h with an equilibrium reached after 100 h. Intravenous injection of nanoparticles demonstrated a different distribution pattern with a rapid particle retention in all organs (particularly in liver and spleen) and a subsequent slow release rate. CONCLUSION: The mathematical model demonstrated good agreement with experimental data derived from tumor mouse models suggesting the value of this tool to predict the real-time pharmacokinetic features of SPIONs in vivo. In the future, it is planned to adapt our model to other nanoparticle formulations to more precisely describe their biodistribution in in vivo model systems.
format Online
Article
Text
id pubmed-7335747
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Dove
record_format MEDLINE/PubMed
spelling pubmed-73357472020-07-14 A New Pharmacokinetic Model Describing the Biodistribution of Intravenously and Intratumorally Administered Superparamagnetic Iron Oxide Nanoparticles (SPIONs) in a GL261 Xenograft Glioblastoma Model Klapproth, Alexander P Shevtsov, Maxim Stangl, Stefan Li, Wei Bo Multhoff, Gabriele Int J Nanomedicine Original Research BACKGROUND: Superparamagnetic iron oxide nanoparticles (SPIONs) have displayed multifunctional applications in cancer theranostics following systemic delivery. In an effort to increase the therapeutic potential of local therapies (including focal hyperthermia), nanoparticles can also be administered intratumorally. Therefore, the development of a reliable pharmacokinetic model for the prediction of nanoparticle distribution for both clinically relevant routes of delivery is of high importance. MATERIALS AND METHODS: The biodistribution of SPIONs (of two different sizes – 130 nm and 60 nm) radiolabeled with zirconium-89 or technetium-99m following intratumoral or intravenous injection was investigated in C57/Bl6 mice bearing subcutaneous GL261 glioblastomas. Based on PET/CT biodistribution data, a novel pharmacokinetic model was established for a better understanding of the pharmacokinetics of the SPIONs after both administration routes. RESULTS: The PET image analysis of the nanoparticles (confirmed by histology) demonstrated the presence of radiolabeled nanoparticles within the glioma site (with low amounts in the liver and spleen) at all investigated time points following intratumoral injection. The mathematical model confirmed the dynamic nanoparticle redistribution in the organism over a period of 72 h with an equilibrium reached after 100 h. Intravenous injection of nanoparticles demonstrated a different distribution pattern with a rapid particle retention in all organs (particularly in liver and spleen) and a subsequent slow release rate. CONCLUSION: The mathematical model demonstrated good agreement with experimental data derived from tumor mouse models suggesting the value of this tool to predict the real-time pharmacokinetic features of SPIONs in vivo. In the future, it is planned to adapt our model to other nanoparticle formulations to more precisely describe their biodistribution in in vivo model systems. Dove 2020-06-30 /pmc/articles/PMC7335747/ /pubmed/32669844 http://dx.doi.org/10.2147/IJN.S254745 Text en © 2020 Klapproth et al. http://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/). 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
Klapproth, Alexander P
Shevtsov, Maxim
Stangl, Stefan
Li, Wei Bo
Multhoff, Gabriele
A New Pharmacokinetic Model Describing the Biodistribution of Intravenously and Intratumorally Administered Superparamagnetic Iron Oxide Nanoparticles (SPIONs) in a GL261 Xenograft Glioblastoma Model
title A New Pharmacokinetic Model Describing the Biodistribution of Intravenously and Intratumorally Administered Superparamagnetic Iron Oxide Nanoparticles (SPIONs) in a GL261 Xenograft Glioblastoma Model
title_full A New Pharmacokinetic Model Describing the Biodistribution of Intravenously and Intratumorally Administered Superparamagnetic Iron Oxide Nanoparticles (SPIONs) in a GL261 Xenograft Glioblastoma Model
title_fullStr A New Pharmacokinetic Model Describing the Biodistribution of Intravenously and Intratumorally Administered Superparamagnetic Iron Oxide Nanoparticles (SPIONs) in a GL261 Xenograft Glioblastoma Model
title_full_unstemmed A New Pharmacokinetic Model Describing the Biodistribution of Intravenously and Intratumorally Administered Superparamagnetic Iron Oxide Nanoparticles (SPIONs) in a GL261 Xenograft Glioblastoma Model
title_short A New Pharmacokinetic Model Describing the Biodistribution of Intravenously and Intratumorally Administered Superparamagnetic Iron Oxide Nanoparticles (SPIONs) in a GL261 Xenograft Glioblastoma Model
title_sort new pharmacokinetic model describing the biodistribution of intravenously and intratumorally administered superparamagnetic iron oxide nanoparticles (spions) in a gl261 xenograft glioblastoma model
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7335747/
https://www.ncbi.nlm.nih.gov/pubmed/32669844
http://dx.doi.org/10.2147/IJN.S254745
work_keys_str_mv AT klapprothalexanderp anewpharmacokineticmodeldescribingthebiodistributionofintravenouslyandintratumorallyadministeredsuperparamagneticironoxidenanoparticlesspionsinagl261xenograftglioblastomamodel
AT shevtsovmaxim anewpharmacokineticmodeldescribingthebiodistributionofintravenouslyandintratumorallyadministeredsuperparamagneticironoxidenanoparticlesspionsinagl261xenograftglioblastomamodel
AT stanglstefan anewpharmacokineticmodeldescribingthebiodistributionofintravenouslyandintratumorallyadministeredsuperparamagneticironoxidenanoparticlesspionsinagl261xenograftglioblastomamodel
AT liweibo anewpharmacokineticmodeldescribingthebiodistributionofintravenouslyandintratumorallyadministeredsuperparamagneticironoxidenanoparticlesspionsinagl261xenograftglioblastomamodel
AT multhoffgabriele anewpharmacokineticmodeldescribingthebiodistributionofintravenouslyandintratumorallyadministeredsuperparamagneticironoxidenanoparticlesspionsinagl261xenograftglioblastomamodel
AT klapprothalexanderp newpharmacokineticmodeldescribingthebiodistributionofintravenouslyandintratumorallyadministeredsuperparamagneticironoxidenanoparticlesspionsinagl261xenograftglioblastomamodel
AT shevtsovmaxim newpharmacokineticmodeldescribingthebiodistributionofintravenouslyandintratumorallyadministeredsuperparamagneticironoxidenanoparticlesspionsinagl261xenograftglioblastomamodel
AT stanglstefan newpharmacokineticmodeldescribingthebiodistributionofintravenouslyandintratumorallyadministeredsuperparamagneticironoxidenanoparticlesspionsinagl261xenograftglioblastomamodel
AT liweibo newpharmacokineticmodeldescribingthebiodistributionofintravenouslyandintratumorallyadministeredsuperparamagneticironoxidenanoparticlesspionsinagl261xenograftglioblastomamodel
AT multhoffgabriele newpharmacokineticmodeldescribingthebiodistributionofintravenouslyandintratumorallyadministeredsuperparamagneticironoxidenanoparticlesspionsinagl261xenograftglioblastomamodel