Nonenzymatic chemiluminescence detection of circulating tumor cells in blood based on Au@luminol nanoparticles, hybridization chain reaction and magnetic isolation

2020 ◽  
Vol 318 ◽  
pp. 128287 ◽  
Author(s):  
Hai-Xia Cao ◽  
Peng-Fei Liu ◽  
Li Wang ◽  
Zhi-Jun Liu ◽  
Shu-Ying Ye ◽  
...  
2019 ◽  
Vol 5 (7) ◽  
pp. eaav9186 ◽  
Author(s):  
Jiantong Dong ◽  
Yu Jen Jan ◽  
Ju Cheng ◽  
Ryan Y. Zhang ◽  
Meng Meng ◽  
...  

Well-preserved mRNA in circulating tumor cells (CTCs) offers an ideal material for conducting molecular profiling of tumors, thereby providing a noninvasive diagnostic solution for guiding treatment intervention and monitoring disease progression. However, it is technically challenging to purify CTCs while retaining high-quality mRNA.Here, we demonstrate a covalent chemistry–based nanostructured silicon substrate (“Click Chip”) for CTC purification that leverages bioorthogonal ligation–mediated CTC capture and disulfide cleavage–driven CTC release. This platform is ideal for CTC mRNA assays because of its efficient, specific, and rapid purification of pooled CTCs, enabling downstream molecular quantification using reverse transcription Droplet Digital polymerase chain reaction. Rearrangements of ALK/ROS1 were quantified using CTC mRNA and matched with those identified in biopsy specimens from 12 patients with late-stage non–small cell lung cancer. Moreover, CTC counts and copy numbers of ALK/ROS1 rearrangements could be used together for evaluating treatment responses and disease progression.


Cancer ◽  
1996 ◽  
Vol 78 (11) ◽  
pp. 2445-2447 ◽  
Author(s):  
Klaus Jung ◽  
Wolfgang Henke ◽  
Michael Lein ◽  
Dietmar Schnorr ◽  
Stefan A. Loening

2021 ◽  
Author(s):  
Pengfei Zhang ◽  
Mohamed S. Draz ◽  
Anwen Xiong ◽  
Wannian Yan ◽  
Huanxing Han ◽  
...  

AbstractHighly efficient capture and detection of circulating tumor cells (CTCs) remain elusive mainly because of their extremely low concentration in the peripheral blood of patients. Herein, we present an approach for the simultaneous capturing, isolation, and detection of CTCs using an immuno-fluorescent magnetic nanobead system (iFMNS) coated with a monoclonal anti-EpCAM antibody. The developed antibody nanobead system allows magnetic isolation and fluorescent-based quantification of CTCs. The expression of EpCAM on the surface of captured CTCs could be directly visualized without additional immune-fluorescent labeling. Our approach is shown to result in a 70 - 95% capture efficiency of CTCs, and 95% of the captured cells remain viable. Using our approach, the isolated cells could be directly used for culture, reverse transcription-polymerase chain reaction (RT-PCR), and immunocytochemistry (ICC) identification. We applied iFMNS for testing CTCs in peripheral blood samples from a lung cancer patient, which suggested that this approach would be a promising tool for CTCs enrichment and detection in one step.


Sign in / Sign up

Export Citation Format

Share Document