Spheroid-plug model as a tool to study tumor development, angiogenesis, and heterogeneity in vivo

Tumour Biol. 2016 Feb;37(2):2481-96. doi: 10.1007/s13277-015-4065-z. Epub 2015 Sep 18.

Abstract

Subcutaneous injection of the tumor cell suspension is a simple and commonly used tool for studying tumor development in vivo. However, subcutaneous models poorly resemble tumor complexity due to the fast growth not reflecting the natural course. Here, we describe an application of the new spheroid-plug model to combine the simplicity of subcutaneous injection with improved resemblance to natural tumor progression. Spheroid-plug model relies on in vitro formation of tumor spheroids, followed by injection of single tumor spheroid subcutaneously in Matrigel matrix. In spheroid-plug model, tumors grow slower in comparison to tumors formed by injection of cell suspension as assessed by 3D ultrasonography (USG) and in vivo bioluminescence measurements. The slower tumor growth rate in spheroid-plug model is accompanied by reduced necrosis. The spheroid-plug model ensures increased and more stable vascularization of tumor than classical subcutaneous tumor model as demonstrated by 3D USG Power Doppler examination. Flow cytometry analysis showed that tumors formed from spheroids have enhanced infiltration of endothelial cells as well as hematopoietic and progenitor cells with stem cell phenotype (c-Kit(+) and Sca-1(+)). They also contain more tumor cells expressing cancer stem cell marker CXCR4. Here, we show that spheroid-plug model allows investigating efficiency of anticancer drugs. Treatment of spheroid-plug tumors with known antiangiogenic agent axitinib decreased their size and viability. The antiangiogenic activity of axitinib was higher in spheroid-plug model than in classical model. Our results indicate that spheroid-plug model imitates natural tumor growth and can become a valuable tool for cancer research.

Keywords: B16 melanoma; Cancer stem cells; Lewis lung carcinoma (LLC); Necrosis; Tumor complexity; Tumor infiltration; Tumor vascularization.

MeSH terms

  • Animals
  • Antineoplastic Agents / pharmacology
  • Axitinib
  • Biomarkers, Tumor / metabolism
  • Carcinogenesis / metabolism
  • Carcinogenesis / pathology*
  • Cell Proliferation / drug effects
  • Cell Survival / drug effects
  • Collagen / metabolism
  • Drug Combinations
  • Endothelial Cells / metabolism
  • Endothelial Cells / pathology
  • Hematopoietic Stem Cells / metabolism
  • Hematopoietic Stem Cells / pathology
  • Imidazoles / pharmacology
  • Indazoles / pharmacology
  • Injections, Subcutaneous / methods
  • Laminin / metabolism
  • Melanoma, Experimental / metabolism
  • Melanoma, Experimental / pathology
  • Mice
  • Mice, Inbred C57BL
  • Neoplastic Stem Cells / drug effects
  • Neoplastic Stem Cells / metabolism
  • Neoplastic Stem Cells / pathology
  • Neovascularization, Pathologic / metabolism
  • Neovascularization, Pathologic / pathology*
  • Proteoglycans / metabolism
  • Receptors, CXCR4 / metabolism
  • Spheroids, Cellular / metabolism
  • Spheroids, Cellular / pathology*
  • Tumor Cells, Cultured

Substances

  • Antineoplastic Agents
  • Biomarkers, Tumor
  • CXCR4 protein, human
  • Drug Combinations
  • Imidazoles
  • Indazoles
  • Laminin
  • Proteoglycans
  • Receptors, CXCR4
  • matrigel
  • Collagen
  • Axitinib