Eight out of 10 breast cancer patients die within 5 years after the primary tumor has spread to the bones. Tumor cells disseminated from the breast roam the vasculature, colonizing perivascular niches around blood capillaries. Slow flows support the niche maintenance by driving the oxygen, nutrients, and signaling factors from the blood into the interstitial tissue, while extracellular matrix, endothelial cells, and mesenchymal stem cells regulate metastatic homing. Here, we show the feasibility of developing a perfused bone perivascular niche-on-a-chip to investigate the progression and drug resistance of breast cancer cells colonizing the bone. The model is a functional human triculture with stable vascular networks within a 3D native bone matrix cultured on a microfluidic chip. Providing the niche-on-a-chip with controlled flow velocities, shear stresses, and oxygen gradients, we established a long-lasting, self-assembled vascular network without supplementation of angiogenic factors. We further show that human bone marrow-derived mesenchymal stem cells, which have undergone phenotypical transition toward perivascular cell lineages, support the formation of capillary-like structures lining the vascular lumen. Finally, breast cancer cells exposed to interstitial flow within the bone perivascular niche-ona- chip persist in a slow-proliferative state associated with increased drug resistance. We propose that the bone perivascular niche-ona- chip with interstitial flow promotes the formation of stable vasculature and mediates cancer cell colonization.

Human bone perivascular niche-on-a-chip for studying metastatic colonization

Raimondi, Manuela Teresa;
2018-01-01

Abstract

Eight out of 10 breast cancer patients die within 5 years after the primary tumor has spread to the bones. Tumor cells disseminated from the breast roam the vasculature, colonizing perivascular niches around blood capillaries. Slow flows support the niche maintenance by driving the oxygen, nutrients, and signaling factors from the blood into the interstitial tissue, while extracellular matrix, endothelial cells, and mesenchymal stem cells regulate metastatic homing. Here, we show the feasibility of developing a perfused bone perivascular niche-on-a-chip to investigate the progression and drug resistance of breast cancer cells colonizing the bone. The model is a functional human triculture with stable vascular networks within a 3D native bone matrix cultured on a microfluidic chip. Providing the niche-on-a-chip with controlled flow velocities, shear stresses, and oxygen gradients, we established a long-lasting, self-assembled vascular network without supplementation of angiogenic factors. We further show that human bone marrow-derived mesenchymal stem cells, which have undergone phenotypical transition toward perivascular cell lineages, support the formation of capillary-like structures lining the vascular lumen. Finally, breast cancer cells exposed to interstitial flow within the bone perivascular niche-ona- chip persist in a slow-proliferative state associated with increased drug resistance. We propose that the bone perivascular niche-ona- chip with interstitial flow promotes the formation of stable vasculature and mediates cancer cell colonization.
2018
Bone perivascular niche; Breast cancer; Drug resistance; Metastatic colonization; Microfluidic chip; Bone Matrix; Bone Neoplasms; Breast Neoplasms; Cell Line, Tumor; Coculture Techniques; Drug Resistance, Neoplasm; Female; Humans; Mesenchymal Stromal Cells; Oxygen; Perfusion; Tissue Scaffolds; Lab-On-A-Chip Devices; Multidisciplinary
File in questo prodotto:
File Dimensione Formato  
post-print.pdf

Accesso riservato

: Publisher’s version
Dimensione 1.86 MB
Formato Adobe PDF
1.86 MB Adobe PDF   Visualizza/Apri

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/11311/1070233
Citazioni
  • ???jsp.display-item.citation.pmc??? 51
  • Scopus 152
  • ???jsp.display-item.citation.isi??? 138
social impact