Modulation of tumor cell growth in vivo by extracellular matrix metalloprotease inducer

Arch Otolaryngol Head Neck Surg. 2008 Nov;134(11):1218-24. doi: 10.1001/archotol.134.11.1218.

Abstract

Objective: To investigate if loss of extracellular matrix metalloprotease inducer (EMMPRIN) will inhibit the growth of head and neck squamous cell carcinoma (HNSCC) tumor cell lines in vivo. Tumor cell-derived EMMPRIN is highly overexpressed in HNSCC and is thought to be induced by surrounding fibroblasts to stimulate matrix metalloproteases, which modulate tumor cell invasion, growth, and angiogenesis.

Design: In vivo study using FaDu tumor xenografts.

Setting: Academic research facility.

Subjects: Severe combined immunodeficiency (SCID) mice.

Interventions: The HNSCC cell line FaDu was transfected with EMMPRIN (FaDu/E), control vector (FaDu), or plasmid-expressing small-interfering RNA against EMMPRIN (FaDu/siE). Tumor cells combined with fibroblast cells were xenografted onto the flank of SCID mice. Tumors were measured biweekly over 4 weeks, at which time the mice were killed, and tumor samples were analyzed for proliferation (Ki-67 immunohistochemical analysis), vascularization (factor VIII staining), and apoptosis (TUNEL [terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick end labeling] assay).

Main outcome measure: Growth of head and neck cancer cell lines genetically engineered to express variable levels of EMMPRIN.

Results: Tumor growth positively correlated and animal survival negatively correlated with increasing EMMPRIN expression. FaDu/E tumor growth was significantly larger at 4 weeks compared with FaDu tumors (P = .006). Similarly, the control vector-transfected FaDu tumors were significantly larger than FaDu/siE (P < .001). Immunohistochemical analysis demonstrated increased Ki-67 in EMMPRIN-transfected cells, without a significant change in the rate of apoptosis between groups. Vascular density and tumor formation rate also increased significantly with EMMPRIN expression.

Conclusion: This study suggests that anti-EMMPRIN-targeted therapy may prove to be a novel treatment option in HNSCC.

Publication types

  • Research Support, N.I.H., Extramural

MeSH terms

  • Animals
  • Basigin / genetics*
  • Blotting, Western
  • Carcinoma, Squamous Cell / blood supply
  • Carcinoma, Squamous Cell / genetics*
  • Carcinoma, Squamous Cell / pathology*
  • Cell Line, Tumor
  • Cell Proliferation*
  • Cell Transformation, Neoplastic / genetics
  • Cell Transformation, Neoplastic / pathology
  • Gene Expression Regulation, Neoplastic / genetics
  • Humans
  • Immunoenzyme Techniques
  • In Situ Nick-End Labeling
  • Ki-67 Antigen / genetics
  • Male
  • Matrix Metalloproteinase 9 / genetics
  • Mice
  • Mice, SCID
  • Neoplasm Invasiveness / genetics
  • Neoplasm Transplantation
  • Neovascularization, Pathologic / genetics
  • Neovascularization, Pathologic / pathology
  • Otorhinolaryngologic Neoplasms / blood supply
  • Otorhinolaryngologic Neoplasms / genetics*
  • Otorhinolaryngologic Neoplasms / pathology*
  • RNA, Small Interfering
  • Transfection
  • Transplantation, Heterologous
  • Tumor Burden / genetics
  • Vascular Endothelial Growth Factor A / genetics

Substances

  • BSG protein, human
  • Ki-67 Antigen
  • RNA, Small Interfering
  • Vascular Endothelial Growth Factor A
  • Basigin
  • Matrix Metalloproteinase 9