Cargando…

MR-labelled liposomes and focused ultrasound for spatiotemporally controlled drug release in triple negative breast cancers in mice

Rationale: Image-guided, triggerable, drug delivery systems allow for precisely placed and highly localised anti-cancer treatment. They contain labels for spatial mapping and tissue uptake tracking, providing key location and timing information for the application of an external stimulus to trigger...

Descripción completa

Detalles Bibliográficos
Autores principales: Amrahli, Maral, Centelles, Miguel, Cressey, Paul, Prusevicius, Martynas, Gedroyc, Wladyslaw, Xu, Xiao Yun, So, Po-Wah, Wright, Michael, Thanou, Maya
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7806456/
https://www.ncbi.nlm.nih.gov/pubmed/33457192
http://dx.doi.org/10.7150/ntno.52168
_version_ 1783636526133936128
author Amrahli, Maral
Centelles, Miguel
Cressey, Paul
Prusevicius, Martynas
Gedroyc, Wladyslaw
Xu, Xiao Yun
So, Po-Wah
Wright, Michael
Thanou, Maya
author_facet Amrahli, Maral
Centelles, Miguel
Cressey, Paul
Prusevicius, Martynas
Gedroyc, Wladyslaw
Xu, Xiao Yun
So, Po-Wah
Wright, Michael
Thanou, Maya
author_sort Amrahli, Maral
collection PubMed
description Rationale: Image-guided, triggerable, drug delivery systems allow for precisely placed and highly localised anti-cancer treatment. They contain labels for spatial mapping and tissue uptake tracking, providing key location and timing information for the application of an external stimulus to trigger drug release. High Intensity Focused Ultrasound (HIFU or FUS) is a non-invasive approach for treating small tissue volumes and is particularly effective at inducing drug release from thermosensitive nanocarriers. Here, we present a novel MR-imageable thermosensitive liposome (iTSL) for drug delivery to triple-negative breast cancers (TNBC). Methods: A macrocyclic gadolinium-based Magnetic Resonance Imaging (MRI) contrast agent was covalently linked to a lipid. This was incorporated at 30 mol% into the lipid bilayer of a thermosensitive liposome that was also encapsulating doxorubicin. The resulting iTSL-DOX formulation was assessed for physical and chemical properties, storage stability, leakage of gadolinium or doxorubicin, and thermal- or FUS-induced drug release. Its effect on MRI relaxation time was tested in phantoms. Mice with tumours were used for studies to assess both tumour distribution and contrast enhancement over time. A lipid-conjugated near-infrared fluorescence (NIRF) probe was also included in the liposome to facilitate the real time monitoring of iTSL distribution and drug release in tumours by NIRF bioimaging. TNBC (MDA-MB-231) tumour-bearing mice were then used to demonstrate the efficacy at retarding tumour growth and increasing survival. Results: iTSL-DOX provided rapid FUS-induced drug release that was dependent on the acoustic power applied. It was otherwise found to be stable, with minimum leakage of drug and gadolinium into buffers or under challenging conditions. In contrast to the usually suggested longer FUS treatment we identified that brief (~3 min) FUS significantly enhanced iTSL-DOX uptake to a targeted tumour and triggered near-total release of encapsulated doxorubicin, causing significant growth inhibition in the TNBC mouse model. A distinct reduction in the tumours' average T(1) relaxation times was attributed to the iTSL accumulation. Conclusions: We demonstrate that tracking iTSL in tumours using MRI assists the application of FUS for precise drug release and therapy.
format Online
Article
Text
id pubmed-7806456
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-78064562021-01-14 MR-labelled liposomes and focused ultrasound for spatiotemporally controlled drug release in triple negative breast cancers in mice Amrahli, Maral Centelles, Miguel Cressey, Paul Prusevicius, Martynas Gedroyc, Wladyslaw Xu, Xiao Yun So, Po-Wah Wright, Michael Thanou, Maya Nanotheranostics Research Paper Rationale: Image-guided, triggerable, drug delivery systems allow for precisely placed and highly localised anti-cancer treatment. They contain labels for spatial mapping and tissue uptake tracking, providing key location and timing information for the application of an external stimulus to trigger drug release. High Intensity Focused Ultrasound (HIFU or FUS) is a non-invasive approach for treating small tissue volumes and is particularly effective at inducing drug release from thermosensitive nanocarriers. Here, we present a novel MR-imageable thermosensitive liposome (iTSL) for drug delivery to triple-negative breast cancers (TNBC). Methods: A macrocyclic gadolinium-based Magnetic Resonance Imaging (MRI) contrast agent was covalently linked to a lipid. This was incorporated at 30 mol% into the lipid bilayer of a thermosensitive liposome that was also encapsulating doxorubicin. The resulting iTSL-DOX formulation was assessed for physical and chemical properties, storage stability, leakage of gadolinium or doxorubicin, and thermal- or FUS-induced drug release. Its effect on MRI relaxation time was tested in phantoms. Mice with tumours were used for studies to assess both tumour distribution and contrast enhancement over time. A lipid-conjugated near-infrared fluorescence (NIRF) probe was also included in the liposome to facilitate the real time monitoring of iTSL distribution and drug release in tumours by NIRF bioimaging. TNBC (MDA-MB-231) tumour-bearing mice were then used to demonstrate the efficacy at retarding tumour growth and increasing survival. Results: iTSL-DOX provided rapid FUS-induced drug release that was dependent on the acoustic power applied. It was otherwise found to be stable, with minimum leakage of drug and gadolinium into buffers or under challenging conditions. In contrast to the usually suggested longer FUS treatment we identified that brief (~3 min) FUS significantly enhanced iTSL-DOX uptake to a targeted tumour and triggered near-total release of encapsulated doxorubicin, causing significant growth inhibition in the TNBC mouse model. A distinct reduction in the tumours' average T(1) relaxation times was attributed to the iTSL accumulation. Conclusions: We demonstrate that tracking iTSL in tumours using MRI assists the application of FUS for precise drug release and therapy. Ivyspring International Publisher 2021-01-01 /pmc/articles/PMC7806456/ /pubmed/33457192 http://dx.doi.org/10.7150/ntno.52168 Text en © The author(s) This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Amrahli, Maral
Centelles, Miguel
Cressey, Paul
Prusevicius, Martynas
Gedroyc, Wladyslaw
Xu, Xiao Yun
So, Po-Wah
Wright, Michael
Thanou, Maya
MR-labelled liposomes and focused ultrasound for spatiotemporally controlled drug release in triple negative breast cancers in mice
title MR-labelled liposomes and focused ultrasound for spatiotemporally controlled drug release in triple negative breast cancers in mice
title_full MR-labelled liposomes and focused ultrasound for spatiotemporally controlled drug release in triple negative breast cancers in mice
title_fullStr MR-labelled liposomes and focused ultrasound for spatiotemporally controlled drug release in triple negative breast cancers in mice
title_full_unstemmed MR-labelled liposomes and focused ultrasound for spatiotemporally controlled drug release in triple negative breast cancers in mice
title_short MR-labelled liposomes and focused ultrasound for spatiotemporally controlled drug release in triple negative breast cancers in mice
title_sort mr-labelled liposomes and focused ultrasound for spatiotemporally controlled drug release in triple negative breast cancers in mice
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7806456/
https://www.ncbi.nlm.nih.gov/pubmed/33457192
http://dx.doi.org/10.7150/ntno.52168
work_keys_str_mv AT amrahlimaral mrlabelledliposomesandfocusedultrasoundforspatiotemporallycontrolleddrugreleaseintriplenegativebreastcancersinmice
AT centellesmiguel mrlabelledliposomesandfocusedultrasoundforspatiotemporallycontrolleddrugreleaseintriplenegativebreastcancersinmice
AT cresseypaul mrlabelledliposomesandfocusedultrasoundforspatiotemporallycontrolleddrugreleaseintriplenegativebreastcancersinmice
AT pruseviciusmartynas mrlabelledliposomesandfocusedultrasoundforspatiotemporallycontrolleddrugreleaseintriplenegativebreastcancersinmice
AT gedroycwladyslaw mrlabelledliposomesandfocusedultrasoundforspatiotemporallycontrolleddrugreleaseintriplenegativebreastcancersinmice
AT xuxiaoyun mrlabelledliposomesandfocusedultrasoundforspatiotemporallycontrolleddrugreleaseintriplenegativebreastcancersinmice
AT sopowah mrlabelledliposomesandfocusedultrasoundforspatiotemporallycontrolleddrugreleaseintriplenegativebreastcancersinmice
AT wrightmichael mrlabelledliposomesandfocusedultrasoundforspatiotemporallycontrolleddrugreleaseintriplenegativebreastcancersinmice
AT thanoumaya mrlabelledliposomesandfocusedultrasoundforspatiotemporallycontrolleddrugreleaseintriplenegativebreastcancersinmice