Fermented mistletoe extract as a multimodal antitumoral agent in gliomas

Oliver Podlech, Patrick N Harter, Michel Mittelbronn, Simone Pöschel, Ulrike Naumann, Oliver Podlech, Patrick N Harter, Michel Mittelbronn, Simone Pöschel, Ulrike Naumann

Abstract

In Europe, commercially available extracts from the white-berry mistletoe (Viscum album L.) are widely used as a complementary cancer therapy. Mistletoe lectins have been identified as main active components and exhibit cytotoxic effects as well as immunomodulatory activity. Since it is still not elucidated in detail how mistle toe extracts such as ISCADOR communicate their effects, we analyzed the mechanisms that might be responsible for their antitumoral function on a molecular and functional level. ISCADOR-treated glioblastoma (GBM) cells down-regulate central genes involved in glioblastoma progression and malignancy such as the cytokine TGF-β and matrix-metalloproteinases. Using in vitro glioblastoma/immune cell co-cultivation assays as well as measurement of cell migration and invasion, we could demonstrate that in glioblastoma cells, lectin-rich ISCADOR M and ISCADOR Q significantly enforce NK-cell-mediated GBM cell lysis. Beside its immune stimulatory effect, ISCADOR reduces the migratory and invasive potential of glioblastoma cells. In a syngeneic as well as in a xenograft glioblastoma mouse model, both pretreatment of tumor cells and intratumoral therapy of subcutaneously growing glioblastoma cells with ISCADOR Q showed delayed tumor growth. In conclusion, ISCADOR Q, showing multiple positive effects in the treatment of glioblastoma, may be a candidate for concomitant treatment of this cancer.

Figures

Figure 1
Figure 1
In GBM cells, ISCADOR Q changes the expression of cancer associated genes. (a) PCR-based microarray expression analysis. LNT-229 cells were treated with ISCADOR Q (100 μg/mL) or were left untreated. 24 h later, the cells were harvested, mRNA was prepared, reverse transcribed into cDNA and used for microarray-based expression analysis on a panel of 96 migration-/invasion-angiogenesis-involved genes. The best downregulated genes with a threshold >3 are shown. (b) RT-PCR-based validation of differentially regulated genes detected in (a) at 12 or 24 h of ISCADOR Q treatment (n = 4, SEM). (c) TGF-β ELISA of ISCADOR Q treated LNT-229 cells. The cells were treated with ISCADOR Q (100 μg/mL, 24 h) followed by incubation in serum-free medium for 48 h. Supernatants were harvested and TGF-β 1 or TGF-β 2 was quantified using ELISA (n = 4, SEM). (d) The cells were treated with ISCADOR Q (100 μg/mL, 12 or 24 h). Supernatants were harvested 48 h later. Angiopoietin-1 content was analyzed using ELISA (n = 4, SEM). (e) The cells were treated as in (c). Supernatants were harvested and VEGF content was analyzed using ELISA (n = 4, SEM). (f) Contingency table analysis of the PCR-based microarray expression data together with the corresponding genes of the REMBRANDT database. Nominal scaled response variable (genes significantly up- (1), non- (0), or down- (−1) regulated in glioblastomas versus normal brain tissue expression pattern in the REMBRANDT database) and nominal explanatory variable (same gene up- or downregulated in our PCR-based microarray expression analysis after ISCADOR treatment) were analysed and subsequently tested by the likelihood ratio test (P = 0.0046).
Figure 2
Figure 2
ISCADOR reduces glioma cell growth in a dose dependent manner. LNT-229 (a) or SMA560 cells (b) were treated of 24 or 48 h with increasing concentrations of ISCADOR or were left untreated. Cell density was assessed by crystal violet staining (n = 3, SEM).
Figure 3
Figure 3
ISCADOR does not induce cell death at a concentration of 100 μg/mL. (a) LNT-229 cells were treated with ISCADOR (100 μg/mL, 24 h), or were left untreated, and then allowed to grow. Cell density was assessed by crystal violet staining every 24 h (n = 9 for LNT-229 cells, n = 4 for SMA560 cells, SEM). (b) The cells were treated as in (a). Viable cells were determined by trypan blue staining (n = 3, SEM). (c) Fluorescence based Caspase 3/7 activity assay. The cells were treated as indicated and caspase activity was analyzed 16, 24 and 48 h later using the fluorescent substrate DEVM-amc. As a positive control, the cells were treated with CD95L (100 U/mL + 10 μg/mL CHX) for 4 h (n = 3, SEM).
Figure 4
Figure 4
Treatment of GBM cells with ISCADOR enhances NK cell mediated tumor cell lysis. (a) LNT-229-Luc cells were treated with the appropriated ISCADOR variant (100 μg/mL) for 24 h. The cells were washed and incubated with human PBLs at different E/T cell ratio for 4 h. Cell lysis was measured by quantifying luciferase activity. (b) LNT-229-Luc cells were treated as in (a). PBLs were incubated with neutralizing NKG2D antibody (R&D Systems, Wiesbaden, Germany, 10 μg/mL) or left untreated prior to coincubation with GBM cells at an E/T of 40 : 1. (c). LNT-229-Luc cells were treated as in (a) and coincubated with purified polyclonal NK cells ((a) to (c): one representative out of three independent experiments is shown, bars = SD). (d) CFSE labeled purified NK cells were cocultivated with LNT-229 glioma cells. After extensive washing, NK cells (upper panel) or coculture (lower panels) were microscopically documented (n = 3, one representative experiment is shown). (e) Quantification of NK cell attachment on LNT-229 cells (n = 3, SEM,  *P = 0.035).
Figure 5
Figure 5
ISCADOR reduces cell motility and invasiveness in human and mouse GBM cell lines. (a) Boyden chamber migration assays. The cells were treated for 24 h with ISCADOR P, M, or Q (100 μg/mL). 20.000 treated cells were seeded in upper parts of Boyden chambers. Migrated cells were counted 21 h later (n = 3 for ISCADOR P, n = 5 for ISCADOR M, n = 7 for ISCADOR Q, SEM). (b) Scratch assay. The cells were seeded and treated as in (a) 24 h after treatment a scratch was set and migration of cells were documented photographically every 24 h. For better visualization, migration borders were marked with black lines (n = 3, one representative experiment is shown). (c) Boyden chamber matrigel invasion assay. The cells were treated as in (a) (n = 3, SEM).
Figure 6
Figure 6
ISCADOR Q mediated reduction of cell motility is caused by reduced MMP-expression and activity. (a) LNT-229 cells were treated with ISCADOR Q or were left untreated. 24 h later, cellular supernatants (SN) or lysates were prepared. MMP or TIMP expression was analyzed by immunoblot (Ab; specific antibody, one representative experiment is shown). (b) Quantification of protein expression (n = 3 for MMP-1, n = 9 for MMP-2, n = 3 for MMP-3, n = 8 for MMP-9, n = 2 for MMP-10, n = 4 for MMP-14, n = 5 for TIMP-2). (c) Gelatinase zymogram analyzing MMP activity (n = 10, one representative experiment is shown). (d) Quantification of MMP gelatinase activity (n = 8 for MMP-9, n = 10 for MMP-2, n = 4 for 36 kD active MMP-2; SEM). Note that the activated form of MMP-9 could not be separated strictly from MMP-2. Therefore quantification of MMP-9 activity was only done for proMMP-9.
Figure 7
Figure 7
ISCADOR Q reduces the growth of subcutaneously growing GBM in mice. (a) and (b) The cells were pretreated with ISCADOR Q (100 μg/mL) or were left untreated. One million viable LNT-229 cells were implanted into the right flank of nude mice (a), or one million SMA560 cells into VMDk-mice (b) Tumor size was measured 3 times per week. Inlet: As a control and to avoid the measurement of artifacts induced by ISCADOR Q mediated inhibition of cell growth, proliferation of implanted cells was assessed by crystal violet staining in parallel. (c) One million LNT-229 cells were implanted into the right flank of nude mice and tumors were allowed to grow for six days. At day 7 subcutaneous injections twice a week with PBS or a weekly increasing dose of ISCADOR Q (1 up to 100 μg in total, arrows indicate ISCADOR injections) were given on the contralateral body site. After two weeks of treatment with the highest dosage, injections were stopped and tumor size was measured as in (a). (d) One million SMA560 cells were implanted into the right flank of VMDk mice and tumors were allowed to grow for six day. On day 7, a single intratumoral injection of ISCADOR Q (20 μL, 100 μg/mL, indicated by an arrow) was given into the palpable tumors. Tumor size was measured as in (a).

References

    1. Stupp R, Mason WP, Van Den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. New England Journal of Medicine. 2005;352(10):987–996.
    1. Hermisson M, Klumpp A, Wick W, et al. O6-methylguanine DNA methyltransferase and p53 status predict temozolomide sensitivity in human malignant glioma cells. Journal of Neurochemistry. 2006;96(3):766–776.
    1. Weller M, Rieger J, Grimmel C, et al. Predicting chemoresistance in human malignant glioma cells: the role of molecular genetic analyses. International Journal of Cancer. 1998;79(6):640–644.
    1. Weller M, Fontana A. The failure of current immunotherapy for malignant glioma. Tumor-derived TGF-β, T-cell apoptosis, and the immune privilege of the brain. Brain Research Reviews. 1995;21(2):128–151.
    1. Platten M, Wick W, Weller M M. Malignant glioma biology: role for TGF-beta in growth, motility, angiogenesis, and immune escape. Microscopic Research Technology. 2001;52(4):401–410.
    1. Nikolai G, Friedl P, Werner M, Niggemann B, Zänker KS. Effect of a mistletoe extract (Iscador® QuFrF) on viability and migratory behavior of human peripheral CD4+ and CD8+ T lymphocytes in three-dimensional collagen lattices. In Vitro Cellular and Developmental Biology. 1997;33(9):710–716.
    1. Gren A. Effects of Iscador preparations on the reactivity of mouse immune system. Neuroendocrinology Letters. 2009;30(4):530–534.
    1. Lee CH, Kim JK, Kim HY, Park SM, Lee SM. Immunomodulating effects of Korean mistletoe lectin in vitro and in vivo. International Immunopharmacology. 2009;9(13-14):1555–1561.
    1. Braedel-Ruoff S. Immunomodulatory effects of Viscum album extracts on natural killer cells: review of clinical trials. Forschende Komplementarmedizin. 2010;17(2):63–73.
    1. Stein GM, Büssing A, Schietzel M. Stimulation of the maturation of dendritic cells in vitro by a fermented mistletoe extract. Anticancer Research. 2002;22(6):4215–4219.
    1. Hajtó T, Fodor K, Perjési P, Németh P. Difficulties and perspectives of immunomodulatory therapy with mistletoe lectins and standardized mistletoe extracts in evidence-based medicine. Evidence-Based Complementary and Alternative Medicine. 2011;2011:6 pages.298972
    1. Gardin NE. Immunological response to mistletoe (Viscum album L.) in cancer patients: a four-case series. Phytotherapy Research. 2009;23(3):407–411.
    1. Park HJ, Hong JH, Kwon HJ, et al. TLR4-mediated activation of mouse macrophages by Korean mistletoe lectin-C (KML-C) Biochemical and Biophysical Research Communications. 2010;396(3):721–725.
    1. Stirpe F, Sandvig K, Olsnes S, Pihl A. Action of viscumin, a toxic lectin from mistletoe, on cells in culture. Journal of Biological Chemistry. 1982;257(22):13271–13277.
    1. Büssing A, Schietzel M. Apoptosis-inducing properties of Viscum album L. extracts from different host trees, correlate with their content of toxic mistletoe lectins. Anticancer Research. 1999;19(1):23–28.
    1. Büssing A, Suzart K, Schweizer K. Differences in the apoptosis-inducing properties of Viscum album L. extracts. Anti-Cancer Drugs. 1997;8(1, supplement):S9–S14.
    1. Burger AM, Mengs U, Schüler JB, Fiebig HH. Antiproliferative activity of an aqueous mistletoe extract in human tumor cell lines and xenografts in vitro. Arzneimittel-Forschung. 2001;51(9):748–757.
    1. Park WB, Lyu SY, Kim JH, et al. Inhibition of tumor growth and metastasis by Korean mistletoe lectin is associated with apoptosis and antiangiogenesis. Cancer Biotherapy and Radiopharmaceuticals. 2001;16(5):439–447.
    1. Choi SH, Lyu SY, Park WB. Mistletoe lectin induces apoptosis and telomerase inhibition in human A253 cancer cells through dephosphorylation of Akt. Archives of Pharmacal Research. 2004;27(1):68–76.
    1. Lyu SY, Park WB, Choi KH, Kim WH. Involvement of caspase-3 in apoptosis induced by Viscum album var. coloratum agglutinin in HL-60 cells. Bioscience, Biotechnology and Biochemistry. 2001;65(3):534–541.
    1. Lyu SY, Park WB. Mistletoe lectin (Viscum album coloratum) modulates proliferation and cytokine expressions in murine splenocytes. Journal of Biochemistry and Molecular Biology. 2006;39(6):662–670.
    1. Thies A, Dautel P, Meyer A, Pfüller U, Schumacher U. Low-dose mistletoe lectin-I reduces melanoma growth and spread in a scid mouse xenograft model. British Journal of Cancer. 2008;98(1):106–112.
    1. Beuth J, Ko HL, Schneider H, et al. Intratumoral application of standardized mistletoe extracts down regulates tumor weight via decreased cell proliferation, increased apoptosis and necrosis in a murine model. Anticancer Research. 2006;26(6):4451–4456.
    1. Pryme IF, Bardocz S, Pusztai A, Ewen SWB. Suppression of growth of tumour cell lines in vitro and tumours in vivo by mistletoe lectins. Histology and Histopathology. 2006;21(1–3):285–299.
    1. Burger AM, Mengs U, Schüler JB, Fiebig HH. Anticancer activity of an aqueous mistletoe extract (AME) in syngeneic murine tumor models. Anticancer Research. 2001;21(3):1965–1968.
    1. Lenartz D, Andermahr J, Plum G, Menzel J, Beuth J. Efficiency of treatment with galactoside-specific lectin from mistletoe against rat glioma. Anticancer Research. 1998;18(2):1011–1014.
    1. Büssing A, Raak C, Ostermann T. Quality of life and related dimensions in cancer patients treated with mistletoe extract (Iscador): a meta-analysis. Evidence Based Complementary and Alternative Medicine. 2012;2012:8 pages.219402
    1. Matthes H, Friedel WE, Bock PR, Zänker KS. Molecular mistletoe therapy: friend or foe in established antitumor protocols? a multicenter, controlled, retrospective pharmaco-epidemiological study in pancreas cancer. Current Molecular Medicine. 2010;10(4):430–439.
    1. Kienle GS, Kiene H. Influence of Viscum album L (European Mistletoe) extracts on quality of life in cancer patients: a systematic review of controlled clinical studies. Integrative Cancer Therapies. 2010;9(2):142–157.
    1. Ostermann T, Raak C, Büssing A. Survival of cancer patients treated with mistletoe extract (Iscador): a systematic literature review. BMC Cancer. 2009;9, article 451
    1. Ziegler R, Grossarth-Maticek R. Individual patient data meta-analysis of survival and psychosomatic self-regulation from published prospective controlled cohort studies for long-term therapy of breast cancer patients with a mistletoe preparation (Iscador) Evidence-Based Complementary and Alternative Medicine. 2010;7(2):157–166.
    1. Lenartz D, Dott U, Menzel J, Schierholz JM, Beuth J. Survival of glioma patients after complementary treatment with galactoside-specific lectin from mistletoe. Anticancer Research. 2000;20(3):2073–2076.
    1. Friese MA, Platten M, Lutz SZ, et al. MICA/NKG2D-mediated immunogene therapy of experimental gliomas. Cancer Research. 2003;63(24):8996–9006.
    1. Antony S, Kuttan R, Kuttan G. Role of natural killer cells in Iscador mediated inhibition of metastasis by adoptive immunotherapy. Immunological Investigations. 2000;29(3):219–231.
    1. Ishii N, Maier D, Merlo A, et al. Frequent co-alterations of TP53, p16/CDKN2A, p14(ARF), PTEN tumor suppressor genes in human glioma cell lines. Brain Pathology. 1999;9(3):469–479.
    1. Vanmeter TE, Rooprai HK, Kibble MM, Fillmore HL, Broaddus WC, Pilkington GJ. The role of matrix metalloproteinase genes in glioma invasion: co-dependent and interactive proteolysis. Journal of Neuro-Oncology. 2001;53(2):213–235.
    1. Wick W, Platten M, Weller M. Glioma cell invasion: regulation of metalloproteinase activity by TGF-β . Journal of Neuro-Oncology. 2001;53(2):177–185.
    1. Pilkington GJ, Darling JL, Lantos PL, Thomas DGT. Cell lines (VMDk) derived from a spontaneous murine astrocytoma. Morphological and immunocytochemical characterization. Journal of the Neurological Sciences. 1983;62(1–3):115–139.
    1. Pilkington GJ, Darling JL, Lantos PL, Thomas DGT. Tumorigenicity of cell lines (VMDk) derived from a spontaneous murine astrocytoma. Histology, fine structure and immunocytochemistry of tumours. Journal of the Neurological Sciences. 1985;71(2-3):145–164.
    1. Naumann U, Kügler S, Wolburg H, et al. Chimeric tumor suppressor 1, a p53-derived chimeric tumor suppressor gene, kills p53 mutant and p53 wild-type glioma cells in synergy with irradiation and CD95 ligand. Cancer Research. 2001;61(15):5833–5842.
    1. Seznec J, Silkenstedt B, Naumann U. Therapeutic effects of the Sp1 inhibitor mithramycin A in glioblastoma. Jorunal of Neurooncology. 2011;101:365–377.
    1. Steinle SA, Li P, Morris DL, et al. Interactions of human NKG2D with its ligands MICA, MICB, and homologs of the mouse RAE-1 protein family. Immunogenetics. 2001;53(4):279–287.
    1. Adamopoulou E, Diekmann J, Tolosa E, et al. Human CD4+ T cells displaying viral epitopes elicit a functional virus-specific memory CD8+ T cell response. Journal of Immunology. 2007;178(9):5465–5472.
    1. Koka V, Potti A, Forseen SE, et al. Role of Her-2/neu overexpression and clinical determinants of early mortality in glioblastoma multiforme. American Journal of Clinical Oncology. 2003;26(4):332–335.
    1. Sasaki T, Lopes MBS, Hankins GR, Helm GA. Expression of survivin, an inhibitor of apoptosis protein, in tumors of the nervous system. Acta Neuropathologica. 2002;104(1):105–109.
    1. Georgescu MM. Pten tumor suppressor network in PI3K-Akt pathway control. Genes and Cancer. 2011;1(12):1170–1177.
    1. Koul D. PTEN signaling pathways in glioblastoma. Cancer Biology and Therapy. 2008;7(9):1321–1325.
    1. Klampfer L. Signal transducers and activators of transcription (STATs): novel targets of chemopreventive and chemotherapeutic drugs. Current Cancer Drug Targets. 2006;6(2):107–121.
    1. Liu C, Sun C, Huang H, Janda K, Edgington T. Overexpression of legumain in tumors is significant for invasion/metastasis and a candidate enzymatic target for prodrug therapy. Cancer Research. 2003;63(11):2957–2964.
    1. Bernardo MM, Fridman R. TIMP-2 (tissue inhibitor of metalloproteinase-2) regulates MMP-2 (matrix metalloproteinase-2) activity in the extracellular environment after pro-MMP-2 activation by MT1 (membrane type 1)-MMP. Biochemical Journal. 2003;374(3):739–745.
    1. Lu KV, Jong KA, Rajasekaran AK, Cloughesy TF, Mischel PS. Upregulation of tissue inhibitor of metalloproteinases (TIMP)-2 promotes matrix metalloproteinase (MMP)-2 activation and cell invasion in a human glioblastoma cell line. Laboratory Investigation. 2004;84(1):8–20.
    1. Deryugina EI, Bourdon MA, Luo GX, Reisfeld RA, Strongin A. Matrix metalloproteinase-2 activation modulates glioma cell migration. Journal of Cell Science. 1997;110(19):2473–2482.
    1. Quo P, Imanishi Y, Cackowski FC, et al. Up-regulation of angiopoietin-2, matrix metalloprotease-2, membrane type 1 metalloprotease, and laminin 5 γ 2 correlates with the invasiveness of human glioma. American Journal of Pathology. 2005;166(3):877–890.
    1. Munaut C, Noël A, Hougrand O, Foidart JM, Boniver J, Deprez M. Vascular endothelial growth factor expression correlates with matrix metalloproteinases MT1-MMP, MMP-2 and MMP-9 in human glioblastomas. International Journal of Cancer. 2003;106(6):848–855.
    1. Lee OH, Xu J, Fueyo J, et al. Expression of the receptor tyrosine kinase Tie2 in neoplastic glial cells is associated with integrin β1-dependent adhesion to the extracellular matrix. Molecular Cancer Research. 2006;4(12):915–926.
    1. Kuttan G, Kuttan R. Immunological mechanism of action of the tumor reducing peptide from mistletoe extract (NSC 635089) cellular proliferation. Cancer Letters. 1992;66(2):123–130.
    1. Schink M, Tröger W, Dabidian A, et al. Mistletoe extract reduces the surgical suppression of natural killer cell activity in cancer patients. A randomized phase III trial. Forschende Komplementarmedizin. 2007;14(1):9–17.
    1. Li MO, Wan YY, Sanjabi S, Robertson AKL, Flavell RA. Transforming growth factor-β regulation of immune responses. Annual Review of Immunology. 2006;24:99–146.

Source: PubMed

3
Subskrybuj