Innovative substance 2250 as a highly promising anti-neoplastic agent in malignant pancreatic carcinoma - in vitro and in vivo

M Buchholz, B Majchrzak-Stiller, S Hahn, D Vangala, R W Pfirrmann, W Uhl, C Braumann, A M Chromik, M Buchholz, B Majchrzak-Stiller, S Hahn, D Vangala, R W Pfirrmann, W Uhl, C Braumann, A M Chromik

Abstract

Background: Former studies already revealed the anti-neoplastic properties of the anti-infective agent Taurolidine (TRD) against many tumor species in vitro and in vivo. Its anti-proliferative and cell death inducing capacity is largely due to its main derivative Taurultam (TRLT). In this study it could be demonstrated, that substance 2250 - a newly defined innovative structural analogue of TRLT - exhibits an anti-neoplastic effect on malignant pancreatic carcinoma in vitro and in vivo.

Methods: The anti-neoplastic potential of substance 2250 as well as its mode of action was demonstrated in extensive in vitro analysis, followed by successful and effective in vivo testings, using xenograft models derived from established pancreatic cancer cell lines as well as patient derived tissue.

Results: Our functional analysis regarding the role of oxidative stress (ROS) and caspase activated apoptosis showed, that ROS driven programmed cell death (PCD) is the major mechanisms induced by substance 2250 in pancreatic carcinoma. What is strongly relevant towards clinical practice is especially the observed inhibition of patient derived pancreatic cancer tumor growth in mice treated with this new substance in combination with its sharply higher metabolic stability.

Conclusion: These encouraging results provide new therapeutical opportunities in pancreatic cancer treatment and build the basis for further functional analysis as well as first clinical studies for this promising agent.

Keywords: Apoptosis; Cancer; Chemotherapy; Substance 2250; Taurolidine.

Figures

Fig. 1
Fig. 1
Molecular structure of substances TRLT and 2250. Substance 2250, 1.4.5-oxathiazan-dioxid-4.4, is an oxathiazine derivative like TRLT with a moleculare weight of 137.15 g/mol
Fig. 2
Fig. 2
Effects of 2250 in different malignant cell lines measured by MTT-assay. AsPC-1 (a), BxPC-3 (b), MiaPaca-2 (c), Panc-1 (d) and Panc TuI cells (e) were incubated with 2250 (100, 200, 500, 1000, 1500, 2000 μmol/l) and ddH2O (control) for 24 h and submitted to a MTT-assay. Values are means ± SEM of 6 replicates of three independent experiments with consecutive passages. Asterisk symbols indicate differences between control, which was adjusted to 100% and 2250 treatment. *** p ≤ 0.001, ** p ≤ 0.01, * p ≤ 0.05, n.s. p > 0.05 (one-way ANOVA followed by Tukey’s post-hoc test)
Fig. 3
Fig. 3
Effects of Taurolidine (TRD) on cell proliferation in different malignant cell lines measured by BrdU-assay. AsPC-1 (a), BxPC-3 (b), MiaPaca-2 (c), Panc-1 (d) and Panc TuI cells (e) were incubated with 2250 (100, 200, 500, 1000, 1500, 2000 μmol/l) and ddH2O (control) for 6 h and submitted to a BrdU-assay. Values are means ± SEM of 8 replicates of three independent experiments with consecutive passages. Asterisk symbols on columns indicate differences between control, which was adjusted to 100% and 2250 treatment. *** p ≤ 0.001, ** p ≤ 0.01, * p ≤ 0.05, n.s. p > 0.05 (one-way ANOVA followed by Tukey’s post-hoc test)
Fig. 4
Fig. 4
Effects of 1000 μmol/l and 1500 μmol/l 2250 on viability, apoptosis and necrosis in different malignant pancreatic cell lines measured by FACS analysis. AsPC-1 (a), BxPC-3 (b) and Panc-TuI (c) cells were incubated with 2250 (1000 and 1500 μmol/l) and ddH2O (control) for 24 h. The percentages of viable, apoptotic and necrotic cells were determined by FACS-analysis with Annexin V-FITC and Propidiumiodide. Values are means ± SEM of 4–6 independent experiments with three consecutive passages. Asterisk symbols on columns indicate differences between control and 2250 treatment. *** p ≤ 0.001, ** p ≤ 0.01, * p ≤ 0.05, n.s. p > 0.05 (one-way ANOVA followed by Tukey’s post-hoc test)
Fig. 5
Fig. 5
Effects of NAC on 2250 induced cell death in different malignant cell lines measured by MTT (a-e); impact of 2250 on the cellular level of ROS (f). AsPC-1 (a), BxPC-3 (b), MiaPaca-2 (c), Panc-1 (d) and Panc TuI (e) cells were incubated with either 2250 (500, 1000 μmol/l), NAC (5 mmol/l) or the combination of both agents (2250 500, 1000 μmol/l + NAC 5 mmol/l) and ddH2O as control for 24 h and submitted to a MTT-assay. The level of ROS was analyzed in untreated compared to 2250 treated cells, additional NAC treatment served as a neg. Control (f). Values are means ± SEM of 6 replicates of three independent experiments with consecutive passages. Asterisk symbols indicate differences between control, which was adjusted to 100% and 2250 treatment. *** p ≤ 0.001, ** p ≤ 0.01, * p ≤ 0.05, (one-way ANOVA followed by Tukey’s post-hoc test)
Fig. 6
Fig. 6
Effects of NAC on 2250 induced cell death in cell line AsPC-1 measured by FACS analysis. AsPC-1 cells were incubated with either 2250 (500, 1000 μmol/l), NAC (5 mmol/l) or the combination of both agents (2250 500, 1000 μmol/l + NAC 5 mmol/l) and ddH2O as control for 24 h. The percentage of viable (a), apoptotic (b) and necrotic (c) cells were determined by FACS analysis. Values are means ± SEM of 4–6 replicates of three independent experiments with consecutive passages. Asterisk symbols indicate differences between control, which was adjusted to 100% and 2250 treatment. *** p ≤ 0.001, ** p ≤ 0.01, * p ≤ 0.05, n.s. p > 0.05 (one-way ANOVA followed by Tukey’s post-hoc test)
Fig. 7
Fig. 7
calibration curve of metabolic half-life measurement. The calibration curve consisting of five different concentrations of substance 2250 served as a standard for the calculation of the metabolic half-life of substance 2250 in nude mice, which resulted in a value of 13.8 h. The according measured parameters are shown in Table 2
Fig. 8
Fig. 8
Effects of 500 mg/kg*BW 2250 on the subcutaneous tumor growth in nude mice in vivo. Nude mice with tumors of MiaPaca-2 (a), Panc-TuI (c) or patient tissue (b, d) were incubated with 2250 (500 mg/kg*BW) and ddH2O (control) for up to 3 weeks. The tumor volume was measured on alternating days for up to 3 weeks. Asterisk symbols indicate differences between control and 2250 treatment. *** p ≤ 0.001, ** p ≤ 0.01, * p ≤ 0.05, n.s. p >0.05 (one-way ANOVA followed by Tukey’s post-hoc test)

References

    1. Ryan DP, Hong TS, Bardeesy N. Pancreatic Adenocarcinoma. N Engl J Med. 2014;371:1039–1049. doi: 10.1056/NEJMra1404198.
    1. Malvezzi M, Bertuccio P, Levi F, La Vecchia C, Negri E. European cancer mortality predictions for the year 2013. Ann Oncol. 2013;24:792–800. doi: 10.1093/annonc/mdt010.
    1. Cancer Facts and Figures | American Cancer Society. 2015. .
    1. Stewart BW, Wild CP. World Cancer Report. 2014. ISBN: 978-92-832-0443-5.
    1. Burris HA, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15:2403–2413. doi: 10.1200/JCO.1997.15.6.2403.
    1. Ueno H, Ioka T, Ikeda M, Ohkawa S, Yanagimoto H, Boku N, et al. Randomized phase III study of gemcitabine plus S-1, S-1 alone, or gemcitabine alone in patients with locally advanced and metastatic pancreatic cancer in Japan and Taiwan: GEST study. J Clin Oncol. 2013;31:1640–1648. doi: 10.1200/JCO.2012.43.3680.
    1. Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–1825. doi: 10.1056/NEJMoa1011923.
    1. Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691–1703. doi: 10.1056/NEJMoa1304369.
    1. Jurewitsch B, Lee T, Park J, Jeejeebhoy K. Taurolidine 2% as an antimicrobial lock solution for prevention of recurrent catheter-related bloodstream infections. JPEN J Parenter Enteral Nutr. 1998;22:242–4.
    1. Jacobi CA, Ordemann J, Böhm B, Zieren HU, Sabat R, Müller JM. Inhibition of peritoneal tumor cell growth and implantation in laparoscopic surgery in a rat model. Am J Surg. 1997;174:359–363. doi: 10.1016/S0002-9610(97)00099-8.
    1. McCourt M, Wang JH, Sookhai S, Redmond HP. Taurolidine inhibits tumor cell growth in vitro and in vivo. Ann Surg Oncol. 2000;7:685–691. doi: 10.1007/s10434-000-0685-6.
    1. Rodak R, Kubota H, Ishihara H, Eugster H-P, Könü D, Möhler H, et al. Induction of reactive oxygen intermediates-dependent programmed cell death in human malignant ex vivo glioma cells and inhibition of the vascular endothelial growth factor production by taurolidine. J Neurosurg. 2005;102:1055–1068. doi: 10.3171/jns.2005.102.6.1055.
    1. Sun BS, Wang JH, Liu LL, Gong SL, Redmond HP. Taurolidine induces apoptosis of murine melanoma cells in vitro and in vivo by modulation of the Bcl-2 family proteins. J Surg Oncol. 2007;96:241–248. doi: 10.1002/jso.20827.
    1. Aceto N, Bertino P, Barbone D, Tassi G, Manzo L, Porta C, et al. Taurolidine and oxidative stress: a rationale for local treatment of mesothelioma. Eur Respir J. 2009;34:1399–1407. doi: 10.1183/09031936.00102308.
    1. Chromik AM, Daigeler A, Bulut D, Flier A, May C, Harati K, et al. Comparative analysis of cell death induction by Taurolidine in different malignant human cancer cell lines. J Exp Clin Cancer Res. 2010;29:21. doi: 10.1186/1756-9966-29-21.
    1. Stendel R, Picht T, Schilling A, Heidenreich J, Loddenkemper C, Jänisch W, et al. Treatment of glioblastoma with intravenous taurolidine. First clinical experience. Anticancer Res. 2004;24:1143–7.
    1. Braumann C, Gutt CN, Scheele J, Menenakos C, Willems W, Mueller JM, et al. Taurolidine reduces the tumor stimulating cytokine interleukin-1beta in patients with resectable gastrointestinal cancer: a multicentre prospective randomized trial. World J Surg Oncol. 2009;7:32. doi: 10.1186/1477-7819-7-32.
    1. Gong L, Greenberg HE, Perhach JL, Waldman SA, Kraft WK. The pharmacokinetics of taurolidine metabolites in healthy volunteers. J Clin Pharmacol. 2007;47:697–703. doi: 10.1177/0091270007299929.
    1. Clauss K, Lück E, von Rymon Lipinski GW. Acetosulfam, a new sweetener. 1. Synthesis and properties (author’s transl) Zeitschrift für Leb und -forsch. 1976;162:37–40. doi: 10.1007/BF01104359.
    1. Jacobi CA, Sabat R, Ordemann J, Wenger F, Volk HD, Müller JM. Peritoneal instillation of taurolidine and heparin for preventing intraperitoneal tumor growth and trocar metastases in laparoscopic operations in the rat model. Langenbecks Arch für Chir. 1997;382:S31–S36. doi: 10.1007/PL00014641.
    1. Jacobi CA, Peter FJ, Wenger FA, Ordemann J, Müller JM. New therapeutic strategies to avoid intra- and extraperitoneal metastases during laparoscopy: results of a tumor model in the rat. Dig Surg. 1999;16:393–399. doi: 10.1159/000018754.
    1. Stendel R, Scheurer L, Schlatterer K, Stalder U, Pfirrmann RW, Fiss I, et al. Pharmacokinetics of taurolidine following repeated intravenous infusions measured by HPLC-ESI-MS/MS of the derivatives taurultame and taurinamide in glioblastoma patients. Clin Pharmacokinet. 2007;46:513–524. doi: 10.2165/00003088-200746060-00005.
    1. Gorman SP, McCafferty DF, Woolfson AD, Jones DS. Reduced adherence of micro-organisms to human mucosal epithelial cells following treatment with Taurolin, a novel antimicrobial agent. J Appl Bacteriol. 1987;62:315–320. doi: 10.1111/j.1365-2672.1987.tb04926.x.
    1. Bedrosian I, Sofia RD, Wolff SM, Dinarello CA. Taurolidine, an analogue of the amino acid taurine, suppresses interleukin 1 and tumor necrosis factor synthesis in human peripheral blood mononuclear cells. Cytokine. 1991;3:568–575. doi: 10.1016/1043-4666(91)90483-T.
    1. Leithäuser ML, Rob PM, Sack K. Pentoxifylline, cyclosporine a and taurolidine inhibit endotoxin-stimulated tumor necrosis factor-alpha production in rat mesangial cell cultures. Exp Nephrol. 1997;5:100–4.
    1. Stendel R, Biefer HRC, Dékány GM, Kubota H, Münz C, Wang S, et al. The antibacterial substance taurolidine exhibits anti-neoplastic action based on a mixed type of programmed cell death. Autophagy. 2009;5:194–210. doi: 10.4161/auto.5.2.7404.
    1. Vanden Berghe T, Vanlangenakker N, Parthoens E, Deckers W, Devos M, Festjens N, et al. Necroptosis, necrosis and secondary necrosis converge on similar cellular disintegration features. Cell Death Differ. 2010;17:922–930. doi: 10.1038/cdd.2009.184.
    1. Ouyang L, Shi Z, Zhao S, Wang F-T, Zhou T-T, Liu B, et al. Programmed cell death pathways in cancer: a review of apoptosis, autophagy and programmed necrosis. Cell Prolif. 2012;45:487–498. doi: 10.1111/j.1365-2184.2012.00845.x.
    1. Stendel R, Scheurer L, Stoltenburg-Didinger G, Brock M, Möhler H. Enhancement of Fas-ligand-mediated programmed cell death by taurolidine. Anticancer Res. 2003;23:2309–14.
    1. Shrayer DP, Lukoff H, King T, Calabresi P. The effect of Taurolidine on adherent and floating subpopulations of melanoma cells. Anti-Cancer Drugs. 2003;14:295–303. doi: 10.1097/00001813-200304000-00007.
    1. Nici L, Monfils B, Calabresi P. The effects of taurolidine, a novel antineoplastic agent, on human malignant mesothelioma. Clin Cancer Res. 2004;10:7655–7661. doi: 10.1158/1078-0432.CCR-0196-03.
    1. Möhler H, Pfirrmann RW, Frei K. Redox-directed cancer therapeutics: Taurolidine and Piperlongumine as broadly effective antineoplastic agents. Int J Oncol. 2014;45:1329–1336.
    1. Opitz I, Sigrist B, Hillinger S, Lardinois D, Stahel R, Weder W, et al. Taurolidine and povidone-iodine induce different types of cell death in malignant pleural mesothelioma. Lung Cancer. 2007;56:327–336. doi: 10.1016/j.lungcan.2007.01.024.
    1. Tait SWG, Ichim G, Green DR. Die another way--non-apoptotic mechanisms of cell death. J Cell Sci. 2014;127:2135–2144. doi: 10.1242/jcs.093575.
    1. Kroemer G, Martin SJ. Caspase-independent cell death. Nat Med. 2005;11:725–730. doi: 10.1038/nm1263.
    1. Kim E-A, Jang J-H, Lee Y-H, Sung E-G, Song I-H, Kim J-Y, et al. Dioscin induces caspase-independent apoptosis through activation of apoptosis-inducing factor in breast cancer cells. Apoptosis. 2014;19:1165–1175. doi: 10.1007/s10495-014-0994-z.
    1. Leon LJ, Pasupuleti N, Gorin F, Carraway KL. A cell-permeant amiloride derivative induces caspase-independent. AIF-mediated programmed necrotic death of breast cancer cells PLoS One. 2013;8
    1. Braumann C, Ordemann J, Kilian M, Wenger FA, Jacobi CA. Local and systemic chemotherapy with taurolidine and taurolidine/heparin in colon cancer-bearing rats undergoing laparotomy. Clin. Exp. Metastasis. 2003;20:387–394. doi: 10.1023/A:1025402919341.
    1. Opitz I, van der Veen HC, Braumann C, Ablassmaier B, Führer K, Jacobi CA. The influence of adhesion prophylactic substances and taurolidine/heparin on local recurrence and intraperitoneal tumor growth after laparoscopic-assisted bowel resection of colon carcinoma in a rat model. Surg Endosc. 2003;17:1098–1104. doi: 10.1007/s00464-002-9161-7.
    1. Bobrich E, Braumann C, Opitz I, Menenakos C, Kristiansen G, Jacobi CA. Influence of intraperitoneal application of taurolidine/heparin on expression of adhesion molecules and colon cancer in rats undergoing laparoscopy. J Surg Res. 2007;137:75–82. doi: 10.1016/j.jss.2006.07.013.
    1. Braumann C, Stuhldreier B, Bobrich E, Menenakos C, Rogalla S, Jacobi CA. High doses of taurolidine inhibit advanced intraperitoneal tumor growth in rats. J Surg Res. 2005;129:129–135. doi: 10.1016/j.jss.2005.03.012.
    1. Nestler G, Schulz HU, Schubert D, Krüger S, Lippert H, Pross M. Impact of taurolidine on the growth of CC531 coloncarcinoma cells in vitro and in a laparoscopic animal model in rats. Surg Endosc. 2005;19:280–284. doi: 10.1007/s00464-003-9301-8.
    1. Raue W, Kilian M, Braumann C, Atanassow V, Makareinis A, Caldenas S, et al. Multimodal approach for treatment of peritoneal surface malignancies in a tumour-bearing rat model. Int J Color Dis. 2010;25:245–250. doi: 10.1007/s00384-009-0819-7.
    1. Kilian M, Mautsch I, Braumann C, Schimke I, Guski H, Jacobi CA, et al. Effects of taurolidine and octreotide on tumor growth and lipid peroxidation after staging-laparoscopy in ductal pancreatic cancer. Prostaglandins Leukot Essent Fatty Acids. 2003;69:261–267. doi: 10.1016/S0952-3278(03)00108-X.
    1. Braumann C, Jacobi CA, Rogalla S, Menenakos C, Fuehrer K, Trefzer U, et al. The tumor suppressive reagent taurolidine inhibits growth of malignant melanoma--a mouse model. J Surg Res. 2007;143:372–378. doi: 10.1016/j.jss.2007.01.041.
    1. Braumann C, Ordemann J, Wildbrett P, Jacobi CA. Influence of intraperitoneal and systemic application of taurolidine and taurolidine/heparin during laparoscopy on intraperitoneal and subcutaneous tumour growth in rats. Clin Exp Metastasis. 2000;18:547–552. doi: 10.1023/A:1011988923523.
    1. Da Costa ML, Redmond HP, Bouchier-Hayes DJ. Taurolidine improves survival by abrogating the accelerated development and proliferation of solid tumors and development of organ metastases from circulating tumor cells released following surgery. J Surg Res. 2001;101:111–119. doi: 10.1006/jsre.2001.6250.
    1. Darnowski JW, Goulette FA, Cousens LP, Chatterjee D, Calabresi P. Mechanistic and antineoplastic evaluation of taurolidine in the DU145 model of human prostate cancer. Cancer Chemother Pharmacol. 2004;54:249–258. doi: 10.1007/s00280-004-0806-1.

Source: PubMed

3
S'abonner