Oncotarget

Research Papers:

Adipose-derived mesenchymal stem cells (ASCs) may favour breast cancer recurrence via HGF/c-Met signaling.

Vincenzo Eterno, Alberto Zambelli, Lorenzo Pavesi, Laura Villani, Vittorio Zanini, Gianfranco Petrolo, Stefania Manera, Antonella Tuscano and Angela Amato _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2014; 5:613-633. https://doi.org/10.18632/oncotarget.1359

Metrics: PDF 4249 views  |   HTML 5018 views  |   ?  


Abstract

Vincenzo Eterno1, Alberto Zambelli1,2, Lorenzo Pavesi1,2, Laura Villani4, Vittorio Zanini3, Gianfranco Petrolo3, Stefania Manera1, Antonella Tuscano1, Angela Amato1

1 Laboratory of Experimental Oncology and Pharmacogenomics, IRCCS Salvatore Maugeri Foundation, Pavia

2 Unit of Medical Oncology, IRCCS Salvatore Maugeri Foundation, Pavia

3 Breast Unit, IRCCS Salvatore Maugeri Foundation, Pavia

4 Unit of Pathology, IRCCS Salvatore Maugeri Foundation, Pavia.

Correspondence:

Angela Amato, email:

Keywords: Adipose-derived Mesenchymal Stem Cells (ASCs), Breast Cancer, HGF/c-Met crosstalk, Microenvironment, Neoangiogenesis.

Received: September 05, 2013 Accepted: October 21, 2013 Published: October 23, 2013

Abstract

Adipose tissue is a reservoir of Mesenchymal Stem Cells (Adipose-derived Mesenchymal Stem Cells, ASCs), endowed with regenerative properties. Fat graft was proposed for breast reconstruction in post-surgery cancer patients achieving good aesthetic results and tissues regeneration. However, recent findings highlight a potential tumorigenic role that ASCs may have in cancer recurrence, raising some concerns about their safety in clinical application.

To address this issue, we established a model where autologous ASCs were combined with primary normal or cancer cells from breast of human donors, in order to evaluate potential effects of their interactions, in vitro and in vivo.

Surprisingly, we found that ASCs are not tumorigenic per sè, as they are not able to induce a neoplastic transformation of normal mammary cells, however they could exhacerbate tumorigenic behaviour of c-Met-expressing breast cancer cells, creating an inflammatory microenvironment which sustained tumor growth and angiogenesis.

Pharmacological c-Met inhibition showed that a HGF/c-Met crosstalk between ASCs and breast cancer cells enhanced tumor cells migration, acquiring a metastatic signature, and sustained tumor self-renewal.

The master role of HGF/c-Met pathway in cancer recurrence was further confirmed by c-Met immunostaining in primary breast cancer from human donors, revealing a strong positivity in patients displaying a recurrent pathology after fat grafts and a weak/moderate staining in patients without signs of recurrence.

Altogether our findings, for the first time, suggest c-Met expression, as predictive to evaluate risk of cancer recurrence after autologous fat graft in post-surgery breast cancer patients, increasing the safety of fat graft in clinical application.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 1359