scholarly journals Polyclonal breast cancer metastases arise from collective dissemination of keratin 14-expressing tumor cell clusters

2016 ◽  
Vol 113 (7) ◽  
pp. E854-E863 ◽  
Author(s):  
Kevin J. Cheung ◽  
Veena Padmanaban ◽  
Vanesa Silvestri ◽  
Koen Schipper ◽  
Joshua D. Cohen ◽  
...  

Recent genomic studies challenge the conventional model that each metastasis must arise from a single tumor cell and instead reveal that metastases can be composed of multiple genetically distinct clones. These intriguing observations raise the question: How do polyclonal metastases emerge from the primary tumor? In this study, we used multicolor lineage tracing to demonstrate that polyclonal seeding by cell clusters is a frequent mechanism in a common mouse model of breast cancer, accounting for >90% of metastases. We directly observed multicolored tumor cell clusters across major stages of metastasis, including collective invasion, local dissemination, intravascular emboli, circulating tumor cell clusters, and micrometastases. Experimentally aggregating tumor cells into clusters induced a >15-fold increase in colony formation ex vivo and a >100-fold increase in metastasis formation in vivo. Intriguingly, locally disseminated clusters, circulating tumor cell clusters, and lung micrometastases frequently expressed the epithelial cytoskeletal protein, keratin 14 (K14). RNA-seq analysis revealed that K14+ cells were enriched for desmosome and hemidesmosome adhesion complex genes, and were depleted for MHC class II genes. Depletion of K14 expression abrogated distant metastases and disrupted expression of multiple metastasis effectors, including Tenascin C (Tnc), Jagged1 (Jag1), and Epiregulin (Ereg). Taken together, our findings reveal K14 as a key regulator of metastasis and establish the concept that K14+ epithelial tumor cell clusters disseminate collectively to colonize distant organs.

Cancers ◽  
2021 ◽  
Vol 13 (10) ◽  
pp. 2356
Author(s):  
Carolina Reduzzi ◽  
Serena Di Cosimo ◽  
Lorenzo Gerratana ◽  
Rosita Motta ◽  
Antonia Martinetti ◽  
...  

The clinical relevance of circulating tumor cell clusters (CTC-clusters) in breast cancer (BC) has been mostly studied using the CellSearch®, a marker-dependent method detecting only epithelial-enriched clusters. However, due to epithelial-to-mesenchymal transition, resorting to marker-independent approaches can improve CTC-cluster detection. Blood samples collected from healthy donors and spiked-in with tumor mammospheres, or from BC patients, were processed for CTC-cluster detection with 3 technologies: CellSearch®, CellSieve™ filters, and ScreenCell® filters. In spiked-in samples, the 3 technologies showed similar recovery capability, whereas, in 19 clinical samples processed in parallel with CellSearch® and CellSieve™ filters, filtration allowed us to detect more CTC-clusters than CellSearch® (median number = 7 versus 1, p = 0.0038). Next, samples from 37 early BC (EBC) and 23 metastatic BC (MBC) patients were processed using ScreenCell® filters for attaining both unbiased enrichment and marker-independent identification (based on cytomorphological criteria). At baseline, CTC-clusters were detected in 70% of EBC cases and in 20% of MBC patients (median number = 2, range 0–20, versus 0, range 0–15, p = 0.0015). Marker-independent approaches for CTC-cluster assessment improve detection and show that CTC-clusters are more frequent in EBC than in MBC patients, a novel finding suggesting that dissemination of CTC-clusters is an early event in BC natural history.


Cell ◽  
2014 ◽  
Vol 158 (5) ◽  
pp. 1110-1122 ◽  
Author(s):  
Nicola Aceto ◽  
Aditya Bardia ◽  
David T. Miyamoto ◽  
Maria C. Donaldson ◽  
Ben S. Wittner ◽  
...  

2014 ◽  
Author(s):  
Nicola Aceto ◽  
Aditya Bardia ◽  
Joel A. Spencer ◽  
Ben S. Wittner ◽  
Min Yu ◽  
...  

The Breast ◽  
2015 ◽  
Vol 24 ◽  
pp. S71
Author(s):  
Sara Jansson ◽  
Pär-Ola Bendahl ◽  
Anna-Maria Larsson ◽  
Kristina Aaltonen ◽  
Lisa Rydén

Sign in / Sign up

Export Citation Format

Share Document