Abstract

IntroductionIn murine breast cancer models, the two interferon-gamma (IFN-γ) inducible chemokines and CXC-chemokine receptor 3 (CXCR3) receptor ligands, monokine induced by γ-interferon (CXCL9) and interferon-γ-inducible protein-10 (CXCL10) impair tumor growth and metastasis formation through recruitment of natural killer (NK) cells and tumor-suppressive T lymphocytes. In human breast cancer, CXCL9 mRNA overexpression correlates with the number of tumor infiltrating lymphocytes and predicts response to different chemotherapeutic regimens. Raising the intratumoral CXCR3 ligand concentration is therefore a possible way to enhance immune intervention in breast cancer. Little is known, however, about expression levels and regulation of these chemokines in human breast cancer. Since the inhibition of cyclooxygenases (COX) has been shown to reduce tumor growth and incidence of metastases in a lymphocytic and IFN-γ dependent manner, we argued that COX isoenzymes are a pharmacologic target to increase intratumoral CXCR3 ligand concentration in human breast cancer.MethodsCXCL9 was visualized in breast cancer specimens by immunohistochemistry, expression levels of CXCL9 and cyclooxygenases were determined by ELISA and western blotting, respectively. For regulation studies, Michigan Cancer Foundation-7 (MCF-7) and M.D. Anderson - Metastatic Breast 231 (MDA-MB 231) breast cancer cells were stimulated with IFN-γ with or without prostaglandin E2 (PGE2) or COX inhibitors (indomethacin, acetylsalicylic acid (ASA), celecoxib). CXCR3 ligand release from cells was measured by ELISA.ResultsWithin the tumor microenvironment, cancer cells are the major source of CXCL9. PGE2 impairs IFN-γ mediated CXCL9 and CXCL10 release from MCF-7 and MDA-MB 231 cells, and inhibition of endogenous cyclooxygenases by indomethacin or ASA correspondingly increases this secretion. Otherwise, high concentrations of the Cyclooxygenase-2 (COX-2) specific antagonist celecoxib have opposite effects and impair CXCL9 and CXCL10 release. In human breast cancer tissue specimens there is an inverse correlation between COX-2 overexpression and CXCL9 concentration, suggesting that the observed in vitro effects are of importance in vivo as well.ConclusionsSuppressing endogenous PGE2 synthesis by cyclooxygenase inhibition increases CXCL9 and CXCL10 release from breast cancer cells and is therefore a pharmacologic candidate to enhance intratumoral immune infiltration. Yet, to this end the unselective COX inhibitors ASA and indomethacin seem preferable to celecoxib that at higher concentrations reduces CXCR3 ligand release most probably due to COX independent mechanisms.

Highlights

  • In murine breast cancer models, the two interferon-gamma (IFN-g) inducible chemokines and CXCchemokine receptor 3 (CXCR3) receptor ligands, monokine induced by g-interferon (CXCL9) and interferon-ginducible protein-10 (CXCL10) impair tumor growth and metastasis formation through recruitment of natural killer (NK) cells and tumor-suppressive T lymphocytes

  • In human breast cancer tissue specimens there is an inverse correlation between COX-2 overexpression and monokine induced by γ-interferon (CXCL9) concentration, suggesting that the observed in vitro effects are of importance in vivo as well

  • Reagents and cell lines Dulbecco’s modified Eagle’s medium (DMEM), fetal calf serum (FCS), gentamycin, 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES), and glutamine were from Gibco Life Technologies (Gaithersburg, MD, USA); recombinant human interferon gamma (IFN-g) and recombinant human TNF-a were from PeproTech (Hamburg, Germany); prostaglandin E2, indomethacin, and celecoxib were from Cayman Chemicals (Ann Arbor, MI, USA) and reconstituted in dimethyl sulfoxide (DMSO); acetylsalicylic acid (ASA) was from Sigma

Read more

Summary

Introduction

In murine breast cancer models, the two interferon-gamma (IFN-g) inducible chemokines and CXCchemokine receptor 3 (CXCR3) receptor ligands, monokine induced by g-interferon (CXCL9) and interferon-ginducible protein-10 (CXCL10) impair tumor growth and metastasis formation through recruitment of natural killer (NK) cells and tumor-suppressive T lymphocytes. We and others have shown that a high expression of the CXCL9 mRNA correlates with an increased number of infiltrating lymphocytes and a better response to chemotherapy [14,15]. In a mouse model transfection of murine breast cancer cells with CXCL9 increases chemotactic T cell recruitment, impairs tumor growth, prevents lung metastasis formation, and prolongs survival [16]. Origin and regulation of CXCR3 chemokines in human breast cancer are poorly understood

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.