Suicide HSVtk gene delivery by neurotensin-polyplex nanoparticles via the bloodstream and GCV Treatment specifically inhibit the growth of human MDA-MB-231 triple negative breast cancer tumors xenografted in athymic mice

PLoS One. 2014 May 13;9(5):e97151. doi: 10.1371/journal.pone.0097151. eCollection 2014.

Abstract

The human breast adenocarcinoma cell line MDA-MB-231 has the triple-negative breast cancer (TNBC) phenotype, which is an aggressive subtype with no specific treatment. MDA-MB-231 cells express neurotensin receptor type 1 (NTSR1), which makes these cells an attractive target of therapeutic genes that are delivered by the neurotensin (NTS)-polyplex nanocarrier via the bloodstream. We addressed the relevance of this strategy for TNBC treatment using NTS-polyplex nanoparticles harboring the herpes simplex virus thymidine kinase (HSVtk) suicide gene and its complementary prodrug ganciclovir (GCV). The reporter gene encoding green fluorescent protein (GFP) was used as a control. NTS-polyplex successfully transfected both genes in cultured MDA-MB-231 cells. The transfection was demonstrated pharmacologically to be dependent on activation of NTSR1. The expression of HSVtk gene decreased cell viability by 49% (P<0.0001) and induced apoptosis in cultured MDA-MB-231 cells after complementary GCV treatment. In the MDA-MB-231 xenograft model, NTS-polyplex nanoparticles carrying either the HSVtk gene or GFP gene were injected into the tumors or via the bloodstream. Both routes of administration allowed the NTS-polyplex nanoparticles to reach and transfect tumorous cells. HSVtk expression and GCV led to apoptosis, as shown by the presence of cleaved caspase-3 and Apostain immunoreactivity, and significantly inhibited the tumor growth (55-60%) (P<0.001). At the end of the experiment, the weight of tumors transfected with the HSVtk gene was 55% less than that of control tumors (P<0.05). The intravenous transfection did not induce apoptosis in peripheral organs. Our results offer a promising gene therapy for TNBC using the NTS-polyplex nanocarrier.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Apoptosis / genetics
  • Cell Line, Tumor
  • Cell Proliferation / drug effects
  • Cell Survival / genetics
  • Female
  • Ganciclovir / pharmacology*
  • Gene Transfer Techniques
  • Genes, Transgenic, Suicide / genetics*
  • Genetic Therapy / methods*
  • Humans
  • Mice
  • Mice, Nude
  • Microscopy, Electron, Scanning
  • Nanoparticles / metabolism
  • Neurotensin / metabolism
  • Simplexvirus / enzymology
  • Thymidine Kinase / genetics*
  • Thymidine Kinase / metabolism
  • Transplantation, Heterologous / methods*
  • Triple Negative Breast Neoplasms / physiopathology*
  • Triple Negative Breast Neoplasms / therapy*

Substances

  • Neurotensin
  • Thymidine Kinase
  • Ganciclovir

Grants and funding

This work was supported by Instituto de Ciencia y Tecnología del Distrito Federal (ICyT-DF), and Agence Internationale de la Recherche Blanc International (ANR-10-INTB-1503) NTS-Polyplex. Consejo Nacional de Ciencia y Tecnología (CONACYT) #142947, supported the fellowships of RC-R and DH-B. Evaluation-orientation de la Coopération Scientifique (ECOS Nord) #M07-S01 supported travel missions and RC-R travel fellowships. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.