High-resolution 3D visualization of nanomedicine distribution in tumors

To improve the clinical translation of anti-cancer nanomedicines, it is necessary to begin building specific insights into the broad concept of the Enhanced Permeability and Retention (EPR) effect, using detailed investigations of the accumulation, distribution and retention of nanomedicines in soli...

Full description

Saved in:
Bibliographic Details
Published inTheranostics Vol. 10; no. 2; pp. 880 - 897
Main Authors Moss, Jennifer I, Barjat, Hervé, Emmas, Sally-Ann, Strittmatter, Nicole, Maynard, Juliana, Goodwin, Richard J A, Storm, Gert, Lammers, Twan, Puri, Sanyogitta, Ashford, Marianne B, Barry, Simon T
Format Journal Article
LanguageEnglish
Published Australia Ivyspring International Publisher Pty Ltd 2020
Ivyspring International Publisher
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:To improve the clinical translation of anti-cancer nanomedicines, it is necessary to begin building specific insights into the broad concept of the Enhanced Permeability and Retention (EPR) effect, using detailed investigations of the accumulation, distribution and retention of nanomedicines in solid tumors. Nanomedicine accumulation in preclinical tumors has been extensively studied; however, treatment efficacy will be heavily influenced by both the quantity of drug-loaded nanomedicines reaching the tumor as well as their spatial distribution throughout the tumor. It remains a challenge to image the heterogeneity of nanomedicine distribution in 3 dimensions within solid tumors with a high degree of spatial resolution using standard imaging approaches. To achieve this, an ex vivo micro computed tomography (µCT) imaging approach was developed to visualize the intratumoral distribution of contrast agent-loaded PEGylated liposomes. Using this semi-quantitative method, whole 3-dimensional (3D) tumor liposome distribution was determined with 17 µm resolution in a phenotypically diverse panel of four preclinical xenograft and patient-derived explant (PDX) tumor models. High-resolution ex vivo μCT imaging revealed striking differences in liposome distribution within tumors in four models with different vascular patterns and densities, stromal contents, and microenvironment morphologies. Following intravenous dosing, the model with the highest density of pericyte-supported vessels showed the greatest liposome accumulation, while the model with vessels present in regions of high α-smooth muscle actin (αSMA) content presented with a large proportion of the liposomes at depths beyond the tumor periphery. The two models with an unsupported vascular network demonstrated a more restricted pattern of liposome distribution. Taken together, vessel distribution and support (the latter indicative of functionality) appear to be key factors determining the accumulation and distribution pattern of liposomes in tumors. Our findings demonstrate that high-resolution 3D visualization of nanomedicine distribution is a useful tool for preclinical nanomedicine research, providing valuable insights into the influence of the tumor vasculature and microenvironment on nanomedicine localization.
Bibliography:Competing interests: JIM, HB, S-AE, NS, JM, RJAG, SP, MBA, STB are present or past employees and shareholders of AstraZeneca PLC.
Present address: Medicines Discovery Catapult, Alderley Park, United Kingdom
ISSN:1838-7640
1838-7640
DOI:10.7150/thno.37178