Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Anal Chem ; 93(14): 5862-5871, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33797884

ABSTRACT

Breast cancer is one of the leading causes of cancer death in women. Novel in vitro tools that integrate three-dimensional (3D) tumor models with highly sensitive chemical reporters can provide useful information to aid biological characterization of cancer phenotype and understanding of drug activity. The combination of surface-enhanced Raman scattering (SERS) techniques with microfluidic technologies offers new opportunities for highly selective, specific, and multiplexed nanoparticle-based assays. Here, we explored the use of functionalized nanoparticles for the detection of estrogen receptor alpha (ERα) expression in a 3D tumor model, using the ERα-positive human breast cancer cell line MCF-7. This approach was used to compare targeted versus nontargeted nanoparticle interactions with the tumor model to better understand whether targeted nanotags are required to efficiently target ERα. Mixtures of targeted anti-ERα antibody-functionalized nanotags (ERα-AuNPs) and nontargeted (against ERα) anti-human epidermal growth factor receptor 2 (HER2) antibody-functionalized nanotags (HER2-AuNPs), with different Raman reporters with a similar SERS signal intensity, were incubated with MCF-7 spheroids in microfluidic devices and spectroscopically analyzed using SERS. MCF-7 cells express high levels of ERα and no detectable levels of HER2. 2D and 3D SERS measurements confirmed the strong targeting effect of ERα-AuNP nanotags to the MCF-7 spheroids in contrast to HER2-AuNPs (63% signal reduction). Moreover, 3D SERS measurements confirmed the differentiation between the targeted and the nontargeted nanotags. Finally, we demonstrated how nanotag uptake by MCF-7 spheroids was affected by the drug fulvestrant, the first-in-class approved selective estrogen receptor degrader (SERD). These results illustrate the potential of using SERS and microfluidics as a powerful in vitro platform for the characterization of 3D tumor models and the investigation of SERD activity.


Subject(s)
Breast Neoplasms , Metal Nanoparticles , Breast Neoplasms/drug therapy , Cell Line, Tumor , Estrogen Receptor alpha , Female , Fulvestrant , Gold , Humans , MCF-7 Cells , Microfluidics
2.
Analyst ; 145(22): 7225-7233, 2020 Nov 09.
Article in English | MEDLINE | ID: mdl-33164013

ABSTRACT

The detection and identification of estrogen receptor alpha (ERα), one of the main biomarkers in breast cancer, is crucial for the clinical diagnosis and therapy of the disease. Here, we use a non-destructive approach for detecting and localising ERα expression at the single cell level using surface enhanced Raman spectroscopy (SERS) combined with functionalised gold nanoparticles (AuNPs). Antibody functionalised nanotags (ERα-AuNPs) showed excellent biocompatibility and enabled the spatial and temporal understanding of ERα location in breast cancer cell lines with different ERα expression status. Additionally, we developed an approach based on the percentage area of SERS response to qualitatively measure expression level in ERα positive (ERα+) breast cancer cells. Specifically, the calculation of relative SERS response demonstrated that MCF-7 cells (ERα+) exhibited higher nanotag accumulation resulting in a 4.2-times increase in SERS signal area in comparison to SKBR-3 cells (ERα-). These results confirmed the strong targeting effect of ERα-AuNPs towards the ERα receptor. The functionalised ERα-AuNP nanotags were also used to investigate the activity of fulvestrant, the first-in-class approved selective estrogen receptor degrader (SERD). SERS mapping confirmed that ERα degradation occurred after fulvestrant treatment since a weaker SERS signal, and hence accumulation of nanotags, was observed in MCF-7 cells treated with fulvestrant. Most importantly, a correlation coefficient of 0.9 between the SERS response and the ERα expression level, obtained by western blot, was calculated. These results confirmed the strong relationship between the two approaches and open up the possibilities of using SERS as a tool for the estimation of ERα expression levels, without the requirement of destructive and time-consuming techniques. Therefore, the potential of using SERS as a rapid and sensitive method to understand the activity of SERDs in breast cancer is demonstrated.


Subject(s)
Breast Neoplasms , Estrogen Receptor alpha , Metal Nanoparticles , Breast Neoplasms/drug therapy , Cell Line, Tumor , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Gold , Humans , MCF-7 Cells
3.
4.
Chem Sci ; 11(22): 5819-5829, 2020 May 27.
Article in English | MEDLINE | ID: mdl-34094083

ABSTRACT

Gold nanoparticles (AuNPs) are widely used in various applications such as cancer imaging and drug delivery. The functionalisation of AuNPs has been shown to affect their cellular internalisation, accumulation and targeting efficiency. The mechanism of cellular uptake of functionalised AuNPs by different cancer cells is not well understood. Therefore, a detailed understanding of the molecular processes is necessary to improve AuNPs for their selective uptake and fate in specific cellular systems. This knowledge can greatly help in designing nanotags with higher cellular uptake for more selective and specific targeting capabilities with less off-target effects. Here, we demonstrate for the first time a straightforward and non-destructive 3D surface enhanced Raman spectroscopy (SERS) imaging approach to track the cellular uptake and localisation of AuNPs functionalised with an anti-ERα (estrogen receptor alpha) antibody in MCF-7 ERα-positive human breast cancer cells under different conditions including temperature and dynamin inhibition. 3D SERS enabled information rich monitoring of the intracellular internalisation of the SERS nanotags. It was found that ERα-AuNPs were internalised by MCF-7 cells in a temperature-dependent manner suggesting an active endocytosis-dependent mechanism. 3D SERS cell mapping also indicated that the nanotags entered MCF-7 cells using dynamin dependent endocytosis, since dynamin inhibition resulted in the SERS signal being obtained from, or close to, the cell surface rather than inside the cells. Finally, ERα-AuNPs were found to enter MCF-7 cells using an ERα receptor-mediated endocytosis process. This study addresses the role of functionalisation of SERS nanotags in biological environments and highlights the benefits of using 3D SERS for the investigation of cellular uptake processes.

SELECTION OF CITATIONS
SEARCH DETAIL
...