Abstract

Co-culture models are currently bridging the gap between classical cultures and in vivo animal models. Exploring this novel approach unlocks the possibility to mimic the tumor microenvironment in vitro, through the establishment of cancer-stroma synergistic interactions. Notably, these organotypic models offer a perfect platform for the development and pre-clinical evaluation of candidate nanocarriers loaded with anti-tumoral drugs in a high throughput screening mode, with lower costs and absence of ethical issues. However, this evaluation was until now limited to co-culture systems established with precise cell ratios, not addressing the natural cell heterogeneity commonly found in different tumors. Therefore, herein the multifunctional nanocarriers efficiency was characterized in various fibroblast-MCF-7 co-culture systems containing different cell ratios, in order to unravel key design parameters that influence nanocarrier performance and the therapeutic outcome. The successful establishment of the co-culture models was confirmed by the tissue-like distribution of the different cells in culture. Nanoparticles incubation in the various co-culture systems reveals that these nanocarriers possess targeting specificity for cancer cells, indicating their suitability for being used in this illness therapy. Additionally, by using different co-culture ratios, different nanoparticle uptake profiles were obtained. These findings are of crucial importance for the future design and optimization of new drug delivery systems, since their real targeting capacity must be addressed in heterogenous cell populations, such as those found in tumors.

Highlights

  • In the last decade the emerging development of nanomedicine has encouraged its rapid application in the development of numerous strategies to treat impairing diseases, that are still incurable [1]

  • Co-cultures of MCF-7 and human dermal fibroblasts (hFIB) Initially, different breast cancer co-culture models were established using various cell ratios that were previously described in the literature as being descriptive of in vivo-mimicking conditions, namely, 1:1; 1:3; 1:5; 3:1, MCF-7 to hFIB cells [19,39,40,41,42]

  • In cocultures that were established with more fibroblasts than breast cancer cells (Figure 2 B1-B5 and C1-C5), MCF-7 cells developed a slight tendency to form agglomerates surrounded by fibroblasts

Read more

Summary

Introduction

In the last decade the emerging development of nanomedicine has encouraged its rapid application in the development of numerous strategies to treat impairing diseases, that are still incurable [1]. Chitosan has been one of the most extensively used for the synthesis of a variety of nanoparticulated drug delivery systems, due to its unique properties like biocompatibility, stability, ease to be chemically modified and low immunogenicity [5]. These unique characteristics can be further tailored by functionalizing the particle surface with cell-specific molecules such as antibodies, folic acid, biotin, or aptamers [6,7], that significantly increase nanocarrier specificity towards target cells, protecting normal cells against drug-derived toxic side effects [8]

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call