O,O'-diethyl-(S,S)-ethylenediamine-N,N'-di-2-(3-cyclohexyl) propanoate dihydrochloride enhances influx of effective NK and NKT cells in murine breast cancer

  • Milena Jurišević University of Kragujevac, Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research
  • Nikola Jagić University of Kragujevac, Faculty of Medical Sciences, Department of Radiology
  • Nevena Gajović University of Kragujevac, Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research
  • Aleksandar Arsenijević University of Kragujevac, Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research
  • Milan Jovanović Military Medical Academy, Department of Abdominal Surgery; University of Defence, Faculty of Medicine of the Military Medical Academy
  • Marija Milovanović University of Kragujevac, Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research
  • Jelena Pantić University of Kragujevac, Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research
  • Ivan Jovanović University of Kragujevac, Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research
  • Tibor Sabo University of Belgrade, Faculty of Chemistry
  • Gordana Radosavljević University of Kragujevac, Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research
  • Nebojša Arsenijević University of Kragujevac, Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research
Keywords: breast neoplasms;, carcinoma;, mice, inbred balbc;, antineoplastic agents

Abstract


Background/Aim. O,O'-diethyl-(S,S)-ethylenediamine-N,N'-di-2-(3-cyclohexyl)propanoate dihydrochloride (DE-EDCP) has been found to possess promising cytotoxic activity against various tumor cell lines. Also, DE-EDCP reduces tumor progression by several mechanisms such as triggering tumor cell death and inhibition of cell proliferation. The aim of present study was to further evaluate anti-tumor activity of DE-EDCP by investigating effects on migratory potential of tumor cells and anti-tumor immune response. Methods. Migratory potential of DE-EDCP was evaluated by scratch wound assay. Female BALB/c mice were inoculated with 4T1 breast cancer cells and treatment with DE-EDCP started five days following orthotopic tumor implantation. The frequency and phenotype of tumor-infiltrating natural killer (NK) and natural killer T (NKT) cells were analyzed by flow cytometry. Results. DE-EDCP inhibited migratory potential of highly metastatic 4T1 cells. DE-EDCP facilitated accumulation of CD3+CD49+ NKT cells and CD3-CD49+ NK cells in tumor microenvironment. DE-EDCP treatment led to significant decrement of tumor infiltrating anergic NKT cells expressing cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), killer cell lectin like receptor G1 (KLRG-1) and programmed cell death protein-1 (PD-1). Mice given DE-EDCP had significantly increased percentages of tumoricidal fas ligand (FasL) positive NK cells. Conclusion. DE-EDCP inhibits murine breast cancer progression through direct effects on tumor cells and by facilitating anti-tumor immunity. DE-EDCP enhances accumulation, promotes tumoricidal phenotype and maintenances responsiveness of NK and NKT cells in 4T1 murine breast cancer.

References

Lazić JM, Vucićević L, Grgurić-Sipka S, Janjetovic K, Kaluderovic GN, Misirkic M, et al. Synthesis and in vitro anticancer activi-ty of octahedral platinum(IV) complexes with cyclohexyl-functionalized ethylenediamine-N,N’-diacetate- type ligands. Chem Med Chem 2010; 5(6): 881–9.

Misirlić Denčić S, Poljarević J, Isakovic AM, Marković I, Sabo TJ, Grgurić-Šipka S. Antileukemic action of novel diamine Pt(II) halogenido complexes: Comparison of the representative nov-el Pt(II) with corresponding Pt(IV) complex. Chem Biol Drug Des 2017; 90(2): 262–71.

Misirlic Dencic S, Poljarevic J, Vilimanovich U, Bogdanovic A, Isa-kovic AJ, Kravic Stevovic T, et al. Cyclohexyl analogues of eth-ylenediamine dipropanoic acid induce caspase-independent mitochondrial apoptosis in human leukemic cells. Chem Res Toxicol 2012; 25(4):931–9.

Jurisevic M, Arsenijevic A, Pantic J, Gajovic N, Milovanovic J, Mi-lovanovic M, et al. The organic ester O,O'-diethyl-(S,S)-ethylenediamine-N,N'-di-2-(3-cyclohexyl)propanoate dihy-drochloride attenuates murine breast cancer growth and me-tastasis. Oncotarget 2018; 9(46): 28195–212.

Isakovic AM, Petricevic SM, Ristic SM, Popadic DM, Kravic-Stevovic TK, Zogovic NS, et al. In vitro and in vivo antimelanoma effect of ethyl ester cyclohexyl analog of ethylenediamine dipropano-ic acid. Melanoma Res 2018; 28(1): 8–20.

Vujanovic, NL, Basse P, Herberman RB, Whiteside TL. Anti-tumor functions of natural killer cells and control of metasta-sis. Methods 1996; 9(2): 394–408.

Vivier E, Ugolini S, Blaise D, Chabannon C, Brossay L. Targeting natural killer cells and natural killer T cells in cancer. Nat Rev Immunol 2012; 12(4): 239–52.

Morvan MG, Lanier LL. NK cells and cancer: you can teach innate cells new tricks. Nat Rev Cancer 2016; 16(1): 7–19.

Brennan PJ, Brigl M, Brenner MB. Invariant natural killer T cells: an innate activation scheme linked to diverse effector functions. Nat Rev Immunol 2013; 13(2): 101–17.

Gebremeskel S, Clattenburg DR, Slauenwhite D, Lobert L, Johnston B. Natural killer T cell activation overcomes immunosuppres-sion to enhance clearance of postsurgical breast cancer metas-tasis in mice. Oncoimmunology 2015; 4(3): e995562.

Jovanovic IP, Pejnovic NN, Radosavljevic GD, Pantic JM, Milovanovic MZ, Arsenijevic NN, et al. Interleukin-33/ST2 axis promotes breast cancer growth and metastases by facilitating intra-tumoral accumulation of immunosuppressive and innate lym-phoid cells. Int J Cancer 2014; 134(7): 1669–82.

Jiang Q, Pan Y, Cheng Y, Li H, Liu D, Li H. Lunasin suppress-es the migration and invasion of breast cancer cells by inhibit-ing matrix metalloproteinase-2/-9 via the FAK/Akt/ERK and NF-κB signaling pathways. Oncol Rep 2016; 36(1): 253–62.

Xu L, Deng X. Protein kinase Ciota promotes nicotine-induced migration and invasion of cancer cells via phosphory-lation of micro- and m-calpains. J Biol Chem 2006; 281(7): 4457–66.

Valaee S, Yaghoobi MM, Shamsara M. Metformin inhibits gas-tric cancer cells metastatic traits through suppression of epi-thelial-mesenchymal transition in a glucose-independent man-ner. PLoS One 2017; 12(3): e0174486.

Milosavljevic MZ, Jovanovic IP, Pejnovic NN, Mitrovic SL, Arseni-jevic NN, Simovic Markovic BJ, et al. Deletion of IL-33R attenu-ates VEGF expression and enhances necrosis in mammary car-cinoma. Oncotarget 2016; 7(14): 18106–15.

Gajovic N, Jurisevic M, Pantic J, Radosavljevic G, Arsenijevic N, Lukic ML, et al. Attenuation of NK cells facilitates mammary tumor growth in streptozotocin-induced diabetes in mice. En-docr Relat Cancer 2018; 25(4): 493–507.

Oppenheimer SB. Cellular basis of cancer metastasis: A review of fundamentals and new advances. Acta Histochem 2006; 108(5): 327–34.

Kaur P, Nagaraja GM, Zheng H, Gizachew D, Galukande M, Krishnan S, et al. A mouse model for triple-negative breast cancer tumor-initiating cells (TNBC-TICs) exhibits similar ag-gressive phenotype to the human disease. BMC Cancer 2012; 12: 120.

Teng Y, Ross JL, Cowell JK. The involvement of JAK-STAT3 in cell motility, invasion, and metastasis. JAKSTAT 2014; 3(1): e28086.

You J, Shi X, Liang H, Ye J, Wang L, Han H, et al. Cystathi-onine- γ-lyase promotes process of breast cancer in association with STAT3 signaling pathway. Oncotarget 2017; (39): 65677–686.

Lu X, Mazur SJ, Lin T, Appella E, Xu Y. The pluripotency factor nanog promotes breast cancer tumorigenesis and metas-tasis. Oncogene. 2014; 33(20): 2655–64.

Siu MK, Wong ES, Kong DS, Chan HY, Jiang L, Wong OG, et al. Stem cell transcription factor NANOG controls cell migration and invasion via dysregulation of outcome in ovarian cancers. Oncogene 2013; 32(30): 3500–9.

Mukherjee P, Gupta A, Chattopadhyay D, Chatterji U. Modula-tion of SOX2 expression delineates an end-point for paclitax-el-effectiveness in breast cancer stem cells. Sci Rep 2017; 7(1): 9170.

Miljković D, Poljarević JM, Petković F, Blaževski J, Momčilović M, Nikolić I, et al. Novel octahedral Pt(IV) complex with di-n-propyl-(S,S)-ethylenediamine-N,N'-di-2-(3-cyclohexyl)propanoato ligand exerts potent immunomodula-tory effects. Eur J Med Chem 2012; 47(1): 194–201.

Villegas FR, Coca S, Villarrubia VG, Jimenez R, Chillon MJ, Jareño J, et al. Prognostic significance of tumor infiltrating natural killer cells subset CD57 in patients with squamous cell lung cancer. Lung Cancer 2002; 35(1): 23–8.

Hayakawa Y, Takeda K, Yagita H, Smyth MJ, Van Kaer L, Oku-mura K, et al. IFN-γ- mediated inhibition of tumor angiogene-sis by natural killer T-cell ligand, alphagalactosylceramide. Blood 2002; 100(5): 1728–33.

Gebremeskel S, Slauenwhite D, Johnston B. Reconstitution models to evaluate natural killer T cell function in tumor control. Immunol Cell Biol 2016; 94(1): 90–100.

Schneiders FL, de Bruin RC, van den Eertwegh AJ, Scheper RJ, Leemans CR, Brakenhoff RH, et al. Circulating invariant natu-ral killer T-cell numbers predict outcome in head and neck squamous cell carcinoma: updated analysis with 10-year fol-low-up. J Clin Oncol 2012; 30(5): 567–70.

Godfrey DI, Kronenberg M. Going both ways: immune regulation via CD1d-dependent NKT cells. J Clin Invest 2004; 114(10): 1379–88.

Ioachim HL, Decuseara R, Giancotti F, Dorsett BH. FAS and FAS-L expression by tumor cells and lymphocytes in breast carcinomas and their lymph node metastases. Pathol Res Pract 2005; 200(11–12): 743–51.

Wajant H. The Fas signaling pathway: more than a paradigm. Science 2002; 296(5573): 1635–6.

Haraguchi K, Takahashi T, Nakahara F, Matsumoto A, Kurokawa M, Ogawa S, et al. CD1d expression level in tumor cells is an important determinant for anti-tumor immunity by natural killer T cells. Leuk Lymphoma 2006; 47(10): 2218–23.

Teng MW, Sharkey J, McLaughlin NM, Exley MA, Smyth MJ. CD1d-based combination therapy eradicates established tu-mors in mice. J Immunol 2009; 183(3): 1911–20.

Metelitsa LS, Naidenko OV, Kant A, Wu HW, Loza MJ, Perussia B, et al. Human NKT cells mediate antitumor cytotoxicity di-rectly by recognizing target cell CD1d with bound ligand or indirectly by producing IL-2 to activate NK cells. J Immunol 2001; 167(6): 3114–22.

Jovanovic I, Radosavljevic G, Mitrovic M, Juranic VL, McKenzie AN, Arsenijevic N, Jonjic S, Lukic ML. ST2 deletion enhances innate and acquired immunity to murine mammary carcinoma. Eur J Immunol 2011; 41(7): 1902–12.

Ito M, Maruyama T, Saito N, Koganei S, Yamamoto K, Matsumoto N. Killer cell lectin-like receptor G1 binds three members of the classical cadherin family to inhibit NK cell cytotoxicity. J Exp Med 2006; 203(2): 289–95.

Shimizu K, Sato Y, Shinga J, Watanabe T, Endo T, Asakura M, et al. KLRG+ invariant natural killer T cells are long-lived effec-tors. Proc Natl Acad Sci U S A 2014; 111(34): 12474–9.

Huntington ND, Tabarias H, Fairfax K, Brady J, Hayakawa Y, Degli Esposti MA, et al. NK cell maturation and peripheral homeostasis is associated with KLRG1 up-regulation. J Im-munol 2007; 178(8): 4764–70.

Müller-Durovic B, Lanna A, Covre LP, Mills RS, Henson SM, Akbar AN. Killer Cell Lectin-like Receptor G1 Inhibits NK Cell Function through Activation of Adenosine 5'-Monophosphate-Activated Protein Kinase. J Immunol 2016; 197(7): 2891–9.

Robbins SH, Nguyen KB, Takahashi N, Mikayama T, Biron CA, Brossay L. Cutting edge: inhibitory functions of the killer cell lectin-like receptor G1 molecule during the activation of mouse NK cells. J Immunol 2002; 168(6): 2585–9.

Nair S, Dhodapkar MV. Natural Killer T Cells in Cancer Im-munotherapy. Front Immunol 2017; 8: 1178.

Sheppard KA, Fitz LJ, Lee JM, Benander C, George JA, Wooters J, et al. PD-1 inhibits T-cell receptor induced phosphorylation of the ZAP70/CD3zeta signalosome and downstream signal-ing to PKCtheta. FEBS Lett 2004; 574(1–3): 37–41.

Dong Y, Sun Q, Zhang X. PD-1 and its ligands are important immune checkpoints in cancer. Oncotarget 2017; 8(2): 2171–86.

Chang WS, Kim JY, Kim YJ, Kim YS, Lee JM, Azuma M, et al. Cutting edge: Programmed death-1/programmed death ligand 1 interaction regulates the induction and maintenance of in-variant NKT cell anergy. J Immunol 2008; 181(10): 6707–10.

Parekh VV, Lalani S, Kim S, Halder R, Azuma M, Yagita H, et al. PD-1/PD-L blockade prevents anergy induction and enhances the anti-tumor activities of glycolipid-activated invariant NKT cells. J Im-munol 2009; 182(5): 2816–26.

Buchbinder EI, Desai A. CTLA-4 and PD-1 Pathways: Similari-ties, Differences, and Implications of Their Inhibition. Am J Clin Oncol 2016; 39(1): 98–106.

Rudd CE, Taylor A, Schneider H. CD28 and CTLA-4 corecep-tor expression and signal transduction. Immunol Rev 2009; 229(1): 12–26.

Beldi-Ferchiou A, Caillat-Zucman S. Control of NK Cell Activa-tion by Immune Checkpoint Molecules. Int J Mol Sci 2017; 18(10): pii: E2129.

an Elsas A, Hurwitz AA, Allison JP. Combination immuno-therapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines in-duces rejection of subcutaneous and metastatic tumors ac-companied by autoimmune depigmentation. J Exp Med 1999; 190: 355–66.

Fan X, Quezada SA, Sepulveda MA, Sharma P, Allison JP. En-gagement of the ICOS pathway markedly enhances efficacy of CTLA-4 blockade in cancer immunotherapy. J Exp Med 2014; 211(4): 715–25.

Kim K, Skora AD, Li Z, Liu Q, Tam AJ, Blosser RL, et al. Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proc Natl Acad Sci U S A 2014; 111(32): 11774–9.

Pentcheva-Hoang T, Simpson TR, Montalvo-Ortiz W, Allison JP. Cytotoxic T lymphocyte antigen-4 blockade enhances anti-tumor immunity by stimulating melanoma-specific T-cell mo-tility. Cancer Immunol Res 2014; 2(10): 970–80.

Published
2021/04/12
Section
Original Paper