Cargando…

Biomimetic nanobubbles for triple-negative breast cancer targeted ultrasound molecular imaging

Triple-negative breast cancer (TNBC) is a highly heterogeneous breast cancer subtype with poor prognosis. Although anatomical imaging figures prominently for breast lesion screening, TNBC is often misdiagnosed, thus hindering early medical care. Ultrasound (US) molecular imaging using nanobubbles (N...

Descripción completa

Detalles Bibliográficos
Autores principales: Jugniot, Natacha, Massoud, Tarik F., Dahl, Jeremy J., Paulmurugan, Ramasamy
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9185914/
https://www.ncbi.nlm.nih.gov/pubmed/35689262
http://dx.doi.org/10.1186/s12951-022-01484-9
_version_ 1784724822854467584
author Jugniot, Natacha
Massoud, Tarik F.
Dahl, Jeremy J.
Paulmurugan, Ramasamy
author_facet Jugniot, Natacha
Massoud, Tarik F.
Dahl, Jeremy J.
Paulmurugan, Ramasamy
author_sort Jugniot, Natacha
collection PubMed
description Triple-negative breast cancer (TNBC) is a highly heterogeneous breast cancer subtype with poor prognosis. Although anatomical imaging figures prominently for breast lesion screening, TNBC is often misdiagnosed, thus hindering early medical care. Ultrasound (US) molecular imaging using nanobubbles (NBs) capable of targeting tumor cells holds great promise for improved diagnosis and therapy. However, the lack of conventional biomarkers in TNBC impairs the development of current targeted agents. Here, we exploited the homotypic recognition of cancer cells to synthesize the first NBs based on TNBC cancer cell membrane (i.e., NB(CCM)) as a targeted diagnostic agent. We developed a microfluidic technology to synthesize NB(CCM) based on the self-assembly property of cell membranes in aqueous solutions. In vitro, optimal NB(CCM) had a hydrodynamic diameter of 683 ± 162 nm, showed long-lasting US contrast enhancements and homotypic affinity. In vivo, we demonstrated that NB(CCM) showed increased extravasation and retention in a TNBC mouse model compared to non-targeted NBs by US molecular imaging. Peak intensities and areas under the curves from time-intensity plots showed a significantly enhanced signal from NB(CCM) compared to non-targeted NBs (2.1-fold, P = 0.004, and, 3.6-fold, P = 0.0009, respectively). Immunofluorescence analysis further validated the presence of NB(CCM) in the tumor microenvironment. Circumventing the challenge for universal cancer biomarker identification, our approach could enable TNBC targeting regardless of tumor tissue heterogeneity, thus improving diagnosis and potentially gene/drug targeted delivery. Ultimately, our approach could be used to image many cancer types using biomimetic NBs prepared from their respective cancer cell membranes. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12951-022-01484-9.
format Online
Article
Text
id pubmed-9185914
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-91859142022-06-11 Biomimetic nanobubbles for triple-negative breast cancer targeted ultrasound molecular imaging Jugniot, Natacha Massoud, Tarik F. Dahl, Jeremy J. Paulmurugan, Ramasamy J Nanobiotechnology Research Triple-negative breast cancer (TNBC) is a highly heterogeneous breast cancer subtype with poor prognosis. Although anatomical imaging figures prominently for breast lesion screening, TNBC is often misdiagnosed, thus hindering early medical care. Ultrasound (US) molecular imaging using nanobubbles (NBs) capable of targeting tumor cells holds great promise for improved diagnosis and therapy. However, the lack of conventional biomarkers in TNBC impairs the development of current targeted agents. Here, we exploited the homotypic recognition of cancer cells to synthesize the first NBs based on TNBC cancer cell membrane (i.e., NB(CCM)) as a targeted diagnostic agent. We developed a microfluidic technology to synthesize NB(CCM) based on the self-assembly property of cell membranes in aqueous solutions. In vitro, optimal NB(CCM) had a hydrodynamic diameter of 683 ± 162 nm, showed long-lasting US contrast enhancements and homotypic affinity. In vivo, we demonstrated that NB(CCM) showed increased extravasation and retention in a TNBC mouse model compared to non-targeted NBs by US molecular imaging. Peak intensities and areas under the curves from time-intensity plots showed a significantly enhanced signal from NB(CCM) compared to non-targeted NBs (2.1-fold, P = 0.004, and, 3.6-fold, P = 0.0009, respectively). Immunofluorescence analysis further validated the presence of NB(CCM) in the tumor microenvironment. Circumventing the challenge for universal cancer biomarker identification, our approach could enable TNBC targeting regardless of tumor tissue heterogeneity, thus improving diagnosis and potentially gene/drug targeted delivery. Ultimately, our approach could be used to image many cancer types using biomimetic NBs prepared from their respective cancer cell membranes. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12951-022-01484-9. BioMed Central 2022-06-10 /pmc/articles/PMC9185914/ /pubmed/35689262 http://dx.doi.org/10.1186/s12951-022-01484-9 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
Jugniot, Natacha
Massoud, Tarik F.
Dahl, Jeremy J.
Paulmurugan, Ramasamy
Biomimetic nanobubbles for triple-negative breast cancer targeted ultrasound molecular imaging
title Biomimetic nanobubbles for triple-negative breast cancer targeted ultrasound molecular imaging
title_full Biomimetic nanobubbles for triple-negative breast cancer targeted ultrasound molecular imaging
title_fullStr Biomimetic nanobubbles for triple-negative breast cancer targeted ultrasound molecular imaging
title_full_unstemmed Biomimetic nanobubbles for triple-negative breast cancer targeted ultrasound molecular imaging
title_short Biomimetic nanobubbles for triple-negative breast cancer targeted ultrasound molecular imaging
title_sort biomimetic nanobubbles for triple-negative breast cancer targeted ultrasound molecular imaging
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9185914/
https://www.ncbi.nlm.nih.gov/pubmed/35689262
http://dx.doi.org/10.1186/s12951-022-01484-9
work_keys_str_mv AT jugniotnatacha biomimeticnanobubblesfortriplenegativebreastcancertargetedultrasoundmolecularimaging
AT massoudtarikf biomimeticnanobubblesfortriplenegativebreastcancertargetedultrasoundmolecularimaging
AT dahljeremyj biomimeticnanobubblesfortriplenegativebreastcancertargetedultrasoundmolecularimaging
AT paulmuruganramasamy biomimeticnanobubblesfortriplenegativebreastcancertargetedultrasoundmolecularimaging