Skip to main content
Erschienen in: Breast Cancer Research and Treatment 1/2014

01.02.2014 | Preclinical study

A combination treatment with DNA methyltransferase inhibitors and suramin decreases invasiveness of breast cancer cells

verfasst von: Sahra Borges, Heike R. Döppler, Peter Storz

Erschienen in: Breast Cancer Research and Treatment | Ausgabe 1/2014

Einloggen, um Zugang zu erhalten

Abstract

The treatment of patients with invasive breast cancer remains a major issue because of the acquisition of drug resistance to conventional chemotherapy. Here we propose a new therapeutic strategy by combining DNA methyltransferase inhibitors (DMTIs) with suramin. Cytotoxic effects of suramin or combination treatment with DMTIs were determined in highly invasive breast cancer cell lines MDA-MB-231, BT-20 and HCC1954, or control cells. In addition, effects on cell invasion were determined in 3-dimensional cell culture assays. DMTI-mediated upregulation of Protein Kinase D1 (PKD1) expression was shown by Western blotting. Effects of suramin on PKD1 activity was determined in vitro and in cells. The importance of PKD1 in mediating the effects of such combination treatment in cell invasion was demonstrated using 3D cell culture assays. A proof of principal animal experiment was performed showing that PKD1 is critical for breast cancer growth. We show that when used in combination, suramin and DMTIs impair the invasive phenotype of breast cancer cells. We show that PKD1, a kinase that previously has been described as a suppressor of tumor cell invasion, is an interface for both FDA-approved drugs, since the additive effects observed are due to DMTI-mediated re-expression and suramin-induced activation of PKD1. Our data reveal a mechanism of how a combination treatment with non-toxic doses of suramin and DMTIs may be of therapeutic benefit for patients with aggressive, multi-drug resistant breast cancer.
Literatur
1.
Zurück zum Zitat Karahoca M, Momparler RL (2013) Pharmacokinetic and pharmacodynamic analysis of 5-aza-2′-deoxycytidine (decitabine) in the design of its dose-schedule for cancer therapy. Clin Epigenet 5:3CrossRef Karahoca M, Momparler RL (2013) Pharmacokinetic and pharmacodynamic analysis of 5-aza-2′-deoxycytidine (decitabine) in the design of its dose-schedule for cancer therapy. Clin Epigenet 5:3CrossRef
2.
Zurück zum Zitat Singh V, Sharma P, Capalash N (2013) DNA methyltransferase inhibitors as epigenetic therapy for cancer. Curr Cancer Drug Targets 13:379–399PubMedCrossRef Singh V, Sharma P, Capalash N (2013) DNA methyltransferase inhibitors as epigenetic therapy for cancer. Curr Cancer Drug Targets 13:379–399PubMedCrossRef
3.
Zurück zum Zitat Karpf AR, Moore BC, Ririe TO, Jones DA (2001) Activation of the p53 DNA damage response pathway after inhibition of DNA methyltransferase by 5-aza-2′-deoxycytidine. Mol Pharmacol 59:751–757PubMed Karpf AR, Moore BC, Ririe TO, Jones DA (2001) Activation of the p53 DNA damage response pathway after inhibition of DNA methyltransferase by 5-aza-2′-deoxycytidine. Mol Pharmacol 59:751–757PubMed
4.
Zurück zum Zitat Eiseler T, Doppler H, Yan IK, Goodison S, Storz P (2009) Protein kinase D1 regulates matrix metalloproteinase expression and inhibits breast cancer cell invasion. Breast Cancer Res 11:R13PubMedCentralPubMedCrossRef Eiseler T, Doppler H, Yan IK, Goodison S, Storz P (2009) Protein kinase D1 regulates matrix metalloproteinase expression and inhibits breast cancer cell invasion. Breast Cancer Res 11:R13PubMedCentralPubMedCrossRef
5.
Zurück zum Zitat Borges S, Doppler H, Perez EA, Andorfer CA, Sun Z et al (2013) Pharmacologic reversion of epigenetic silencing of the PRKD1 promoter blocks breast tumor cell invasion and metastasis. Breast Cancer Res 15:R66PubMedCrossRef Borges S, Doppler H, Perez EA, Andorfer CA, Sun Z et al (2013) Pharmacologic reversion of epigenetic silencing of the PRKD1 promoter blocks breast tumor cell invasion and metastasis. Breast Cancer Res 15:R66PubMedCrossRef
6.
Zurück zum Zitat Chopin DK, Caruelle JP, Colombel M, Palcy S, Ravery V et al (1993) Increased immunodetection of acidic fibroblast growth factor in bladder cancer, detectable in urine. J Urol 150:1126–1130PubMed Chopin DK, Caruelle JP, Colombel M, Palcy S, Ravery V et al (1993) Increased immunodetection of acidic fibroblast growth factor in bladder cancer, detectable in urine. J Urol 150:1126–1130PubMed
7.
Zurück zum Zitat Cronauer MV, Hittmair A, Eder IE, Hobisch A, Culig Z et al (1997) Basic fibroblast growth factor levels in cancer cells and in sera of patients suffering from proliferative disorders of the prostate. Prostate 31:223–233PubMedCrossRef Cronauer MV, Hittmair A, Eder IE, Hobisch A, Culig Z et al (1997) Basic fibroblast growth factor levels in cancer cells and in sera of patients suffering from proliferative disorders of the prostate. Prostate 31:223–233PubMedCrossRef
8.
Zurück zum Zitat Ravery V, Jouanneau J, Gil Diez S, Abbou CC, Caruelle JP et al (1992) Immunohistochemical detection of acidic fibroblast growth factor in bladder transitional cell carcinoma. Urol Res 20:211–214PubMedCrossRef Ravery V, Jouanneau J, Gil Diez S, Abbou CC, Caruelle JP et al (1992) Immunohistochemical detection of acidic fibroblast growth factor in bladder transitional cell carcinoma. Urol Res 20:211–214PubMedCrossRef
9.
Zurück zum Zitat Ropiquet F, Giri D, Kwabi-Addo B, Mansukhani A, Ittmann M (2000) Increased expression of fibroblast growth factor 6 in human prostatic intraepithelial neoplasia and prostate cancer. Cancer Res 60:4245–4250PubMed Ropiquet F, Giri D, Kwabi-Addo B, Mansukhani A, Ittmann M (2000) Increased expression of fibroblast growth factor 6 in human prostatic intraepithelial neoplasia and prostate cancer. Cancer Res 60:4245–4250PubMed
10.
Zurück zum Zitat Singh RK, Bucana CD, Gutman M, Fan D, Wilson MR et al (1994) Organ site-dependent expression of basic fibroblast growth factor in human renal cell carcinoma cells. Am J Pathol 145:365–374PubMed Singh RK, Bucana CD, Gutman M, Fan D, Wilson MR et al (1994) Organ site-dependent expression of basic fibroblast growth factor in human renal cell carcinoma cells. Am J Pathol 145:365–374PubMed
11.
Zurück zum Zitat Suzuki K, Tokue A, Kamiakito T, Kuriki K, Saito K et al (2001) Predominant expression of fibroblast growth factor (FGF) 8, FGF4, and FGF receptor 1 in nonseminomatous and highly proliferative components of testicular germ cell tumors. Virchows Arch 439:616–621PubMed Suzuki K, Tokue A, Kamiakito T, Kuriki K, Saito K et al (2001) Predominant expression of fibroblast growth factor (FGF) 8, FGF4, and FGF receptor 1 in nonseminomatous and highly proliferative components of testicular germ cell tumors. Virchows Arch 439:616–621PubMed
12.
Zurück zum Zitat Bernsen HJ, Rijken PF, Peters JP, Bakker JH, Boerman RH et al (1999) Suramin treatment of human glioma xenografts; effects on tumor vasculature and oxygenation status. J Neurooncol 44:129–136PubMedCrossRef Bernsen HJ, Rijken PF, Peters JP, Bakker JH, Boerman RH et al (1999) Suramin treatment of human glioma xenografts; effects on tumor vasculature and oxygenation status. J Neurooncol 44:129–136PubMedCrossRef
13.
Zurück zum Zitat Bhargava S, Hotz B, Hines OJ, Reber HA, Buhr HJ et al (2007) Suramin inhibits not only tumor growth and metastasis but also angiogenesis in experimental pancreatic cancer. J Gastrointest Surg 11:171–178PubMedCrossRef Bhargava S, Hotz B, Hines OJ, Reber HA, Buhr HJ et al (2007) Suramin inhibits not only tumor growth and metastasis but also angiogenesis in experimental pancreatic cancer. J Gastrointest Surg 11:171–178PubMedCrossRef
14.
Zurück zum Zitat Danesi R, Del Bianchi S, Soldani P, Campagni A, La Rocca RV et al (1993) Suramin inhibits bFGF-induced endothelial cell proliferation and angiogenesis in the chick chorioallantoic membrane. Br J Cancer 68:932–938PubMedCentralPubMedCrossRef Danesi R, Del Bianchi S, Soldani P, Campagni A, La Rocca RV et al (1993) Suramin inhibits bFGF-induced endothelial cell proliferation and angiogenesis in the chick chorioallantoic membrane. Br J Cancer 68:932–938PubMedCentralPubMedCrossRef
15.
Zurück zum Zitat Hawking F (1978) Suramin: with special reference to onchocerciasis. Adv Pharmacol Chemother 15:289–322PubMedCrossRef Hawking F (1978) Suramin: with special reference to onchocerciasis. Adv Pharmacol Chemother 15:289–322PubMedCrossRef
16.
Zurück zum Zitat Coffey RJ Jr, Goustin AS, Soderquist AM, Shipley GD, Wolfshohl J et al (1987) Transforming growth factor alpha and beta expression in human colon cancer lines: implications for an autocrine model. Cancer Res 47:4590–4594PubMed Coffey RJ Jr, Goustin AS, Soderquist AM, Shipley GD, Wolfshohl J et al (1987) Transforming growth factor alpha and beta expression in human colon cancer lines: implications for an autocrine model. Cancer Res 47:4590–4594PubMed
17.
Zurück zum Zitat Hosang M (1985) Suramin binds to platelet-derived growth factor and inhibits its biological activity. J Cell Biochem 29:265–273PubMedCrossRef Hosang M (1985) Suramin binds to platelet-derived growth factor and inhibits its biological activity. J Cell Biochem 29:265–273PubMedCrossRef
18.
Zurück zum Zitat Pollak M, Richard M (1990) Suramin blockade of insulinlike growth factor I-stimulated proliferation of human osteosarcoma cells. J Natl Cancer Inst 82:1349–1352PubMedCrossRef Pollak M, Richard M (1990) Suramin blockade of insulinlike growth factor I-stimulated proliferation of human osteosarcoma cells. J Natl Cancer Inst 82:1349–1352PubMedCrossRef
19.
Zurück zum Zitat Williams LT, Tremble PM, Lavin MF, Sunday ME (1984) Platelet-derived growth factor receptors form a high affinity state in membrane preparations. Kinetics and affinity cross-linking studies. J Biol Chem 259:5287–5294PubMed Williams LT, Tremble PM, Lavin MF, Sunday ME (1984) Platelet-derived growth factor receptors form a high affinity state in membrane preparations. Kinetics and affinity cross-linking studies. J Biol Chem 259:5287–5294PubMed
20.
Zurück zum Zitat Hensey CE, Boscoboinik D, Azzi A (1989) Suramin, an anti-cancer drug, inhibits protein kinase C and induces differentiation in neuroblastoma cell clone NB2A. FEBS Lett 258:156–158PubMedCrossRef Hensey CE, Boscoboinik D, Azzi A (1989) Suramin, an anti-cancer drug, inhibits protein kinase C and induces differentiation in neuroblastoma cell clone NB2A. FEBS Lett 258:156–158PubMedCrossRef
21.
Zurück zum Zitat Buchinger B, Spitzer S, Karlic H, Klaushofer K, Varga F (2008) Lysyl oxidase (LOX) mRNA expression and genes of the differentiated osteoblastic phenotype are upregulated in human osteosarcoma cells by suramin. Cancer Lett 265:45–54PubMedCrossRef Buchinger B, Spitzer S, Karlic H, Klaushofer K, Varga F (2008) Lysyl oxidase (LOX) mRNA expression and genes of the differentiated osteoblastic phenotype are upregulated in human osteosarcoma cells by suramin. Cancer Lett 265:45–54PubMedCrossRef
22.
Zurück zum Zitat Wiese C, Nikolova T, Zahanich I, Sulzbacher S, Fuchs J et al (2011) Differentiation induction of mouse embryonic stem cells into sinus node-like cells by suramin. Int J Cardiol 147:95–111PubMedCrossRef Wiese C, Nikolova T, Zahanich I, Sulzbacher S, Fuchs J et al (2011) Differentiation induction of mouse embryonic stem cells into sinus node-like cells by suramin. Int J Cardiol 147:95–111PubMedCrossRef
23.
Zurück zum Zitat Jiang S, Chen X, Li C, Zhang X, Zhang T et al (2012) Suramin inhibits the growth of nasopharyngeal carcinoma cells via the downregulation of osteopontin. Mol Med Rep 6:1351–1354PubMed Jiang S, Chen X, Li C, Zhang X, Zhang T et al (2012) Suramin inhibits the growth of nasopharyngeal carcinoma cells via the downregulation of osteopontin. Mol Med Rep 6:1351–1354PubMed
24.
Zurück zum Zitat Stein CA (1993) Suramin: a novel antineoplastic agent with multiple potential mechanisms of action. Cancer Res 53:2239–2248PubMed Stein CA (1993) Suramin: a novel antineoplastic agent with multiple potential mechanisms of action. Cancer Res 53:2239–2248PubMed
25.
Zurück zum Zitat Bastea LI, Doppler H, Balogun B, Storz P (2012) Protein kinase D1 maintains the epithelial phenotype by inducing a DNA-bound, inactive SNAI1 transcriptional repressor complex. PLoS ONE 7:e30459PubMedCentralPubMedCrossRef Bastea LI, Doppler H, Balogun B, Storz P (2012) Protein kinase D1 maintains the epithelial phenotype by inducing a DNA-bound, inactive SNAI1 transcriptional repressor complex. PLoS ONE 7:e30459PubMedCentralPubMedCrossRef
26.
Zurück zum Zitat Du C, Zhang C, Hassan S, Biswas MH, Balaji KC (2010) Protein kinase D1 suppresses epithelial-to-mesenchymal transition through phosphorylation of snail. Cancer Res 70:7810–7819PubMedCrossRef Du C, Zhang C, Hassan S, Biswas MH, Balaji KC (2010) Protein kinase D1 suppresses epithelial-to-mesenchymal transition through phosphorylation of snail. Cancer Res 70:7810–7819PubMedCrossRef
27.
Zurück zum Zitat Jaggi M, Rao PS, Smith DJ, Hemstreet GP, Balaji KC (2003) Protein kinase C mu is down-regulated in androgen-independent prostate cancer. Biochem Biophys Res Commun 307:254–260PubMedCrossRef Jaggi M, Rao PS, Smith DJ, Hemstreet GP, Balaji KC (2003) Protein kinase C mu is down-regulated in androgen-independent prostate cancer. Biochem Biophys Res Commun 307:254–260PubMedCrossRef
28.
Zurück zum Zitat Olayioye MA, Barisic S, Hausser A (2013) Multi-level control of actin dynamics by protein kinase D. Cell Signal 25:1739–1747PubMedCrossRef Olayioye MA, Barisic S, Hausser A (2013) Multi-level control of actin dynamics by protein kinase D. Cell Signal 25:1739–1747PubMedCrossRef
29.
Zurück zum Zitat Storz P, Doppler H, Johannes FJ, Toker A (2003) Tyrosine phosphorylation of protein kinase D in the pleckstrin homology domain leads to activation. J Biol Chem 278:17969–17976PubMedCrossRef Storz P, Doppler H, Johannes FJ, Toker A (2003) Tyrosine phosphorylation of protein kinase D in the pleckstrin homology domain leads to activation. J Biol Chem 278:17969–17976PubMedCrossRef
30.
Zurück zum Zitat Cowell CF, Doppler H, Yan IK, Hausser A, Umezawa Y et al (2009) Mitochondrial diacylglycerol initiates protein-kinase D1-mediated ROS signaling. J Cell Sci 122:919–928PubMedCrossRef Cowell CF, Doppler H, Yan IK, Hausser A, Umezawa Y et al (2009) Mitochondrial diacylglycerol initiates protein-kinase D1-mediated ROS signaling. J Cell Sci 122:919–928PubMedCrossRef
31.
Zurück zum Zitat Guo H, Liu W, Ju Z, Tamboli P, Jonasch E et al (2012) An efficient procedure for protein extraction from formalin-fixed, paraffin-embedded tissues for reverse phase protein arrays. Proteome Sci 10:56PubMedCentralPubMedCrossRef Guo H, Liu W, Ju Z, Tamboli P, Jonasch E et al (2012) An efficient procedure for protein extraction from formalin-fixed, paraffin-embedded tissues for reverse phase protein arrays. Proteome Sci 10:56PubMedCentralPubMedCrossRef
32.
Zurück zum Zitat Vignon F, Prebois C, Rochefort H (1992) Inhibition of breast cancer growth by suramin. J Natl Cancer Inst 84:38–42PubMedCrossRef Vignon F, Prebois C, Rochefort H (1992) Inhibition of breast cancer growth by suramin. J Natl Cancer Inst 84:38–42PubMedCrossRef
33.
Zurück zum Zitat Song S, Yu B, Wei Y, Wientjes MG, Au JL (2004) Low-dose suramin enhanced paclitaxel activity in chemotherapy-naive and paclitaxel-pretreated human breast xenograft tumors. Clin Cancer Res 10:6058–6065PubMedCrossRef Song S, Yu B, Wei Y, Wientjes MG, Au JL (2004) Low-dose suramin enhanced paclitaxel activity in chemotherapy-naive and paclitaxel-pretreated human breast xenograft tumors. Clin Cancer Res 10:6058–6065PubMedCrossRef
34.
Zurück zum Zitat Waldron RT, Rozengurt E (2003) Protein kinase C phosphorylates protein kinase D activation loop Ser744 and Ser748 and releases autoinhibition by the pleckstrin homology domain. J Biol Chem 278:154–163PubMedCrossRef Waldron RT, Rozengurt E (2003) Protein kinase C phosphorylates protein kinase D activation loop Ser744 and Ser748 and releases autoinhibition by the pleckstrin homology domain. J Biol Chem 278:154–163PubMedCrossRef
35.
Zurück zum Zitat Gschwendt M, Kittstein W, Johannes FJ (1998) Differential effects of suramin on protein kinase C isoenzymes. A novel tool for discriminating protein kinase C activities. FEBS Lett 421:165–168PubMedCrossRef Gschwendt M, Kittstein W, Johannes FJ (1998) Differential effects of suramin on protein kinase C isoenzymes. A novel tool for discriminating protein kinase C activities. FEBS Lett 421:165–168PubMedCrossRef
36.
Zurück zum Zitat Matthews SA, Rozengurt E, Cantrell D (1999) Characterization of serine 916 as an in vivo autophosphorylation site for protein kinase D/Protein kinase Cmu. J Biol Chem 274:26543–26549PubMedCrossRef Matthews SA, Rozengurt E, Cantrell D (1999) Characterization of serine 916 as an in vivo autophosphorylation site for protein kinase D/Protein kinase Cmu. J Biol Chem 274:26543–26549PubMedCrossRef
37.
Zurück zum Zitat Storz P, Doppler H, Copland JA, Simpson KJ, Toker A (2009) FOXO3a promotes tumor cell invasion through the induction of matrix metalloproteinases. Mol Cell Biol 29:4906–4917PubMedCentralPubMedCrossRef Storz P, Doppler H, Copland JA, Simpson KJ, Toker A (2009) FOXO3a promotes tumor cell invasion through the induction of matrix metalloproteinases. Mol Cell Biol 29:4906–4917PubMedCentralPubMedCrossRef
38.
Zurück zum Zitat Bowden CJ, Figg WD, Dawson NA, Sartor O, Bitton RJ et al (1996) A phase I/II study of continuous infusion suramin in patients with hormone-refractory prostate cancer: toxicity and response. Cancer Chemother Pharmacol 39:1–8PubMedCrossRef Bowden CJ, Figg WD, Dawson NA, Sartor O, Bitton RJ et al (1996) A phase I/II study of continuous infusion suramin in patients with hormone-refractory prostate cancer: toxicity and response. Cancer Chemother Pharmacol 39:1–8PubMedCrossRef
39.
Zurück zum Zitat Dreicer R, Smith DC, Williams RD, See WA (1999) Phase II trial of suramin in patients with metastatic renal cell carcinoma. Invest New Drugs 17:183–186PubMedCrossRef Dreicer R, Smith DC, Williams RD, See WA (1999) Phase II trial of suramin in patients with metastatic renal cell carcinoma. Invest New Drugs 17:183–186PubMedCrossRef
40.
Zurück zum Zitat Falcone A, Pfanner E, Cianci C, Danesi R, Brunetti I et al (1995) Suramin in patients with metastatic colorectal cancer pretreated with fluoropyrimidine-based chemotherapy. A phase II study. Cancer 75:440–443PubMedCrossRef Falcone A, Pfanner E, Cianci C, Danesi R, Brunetti I et al (1995) Suramin in patients with metastatic colorectal cancer pretreated with fluoropyrimidine-based chemotherapy. A phase II study. Cancer 75:440–443PubMedCrossRef
41.
Zurück zum Zitat George S, Dreicer R, Au JJ, Shen T, Rini BI et al (2008) Phase I/II trial of 5-fluorouracil and a noncytotoxic dose level of suramin in patients with metastatic renal cell carcinoma. Clin Genitourin Cancer 6:79–85PubMedCentralPubMedCrossRef George S, Dreicer R, Au JJ, Shen T, Rini BI et al (2008) Phase I/II trial of 5-fluorouracil and a noncytotoxic dose level of suramin in patients with metastatic renal cell carcinoma. Clin Genitourin Cancer 6:79–85PubMedCentralPubMedCrossRef
42.
Zurück zum Zitat Lustberg MB, Pant S, Ruppert AS, Shen T, Wei Y et al (2012) Phase I/II trial of non-cytotoxic suramin in combination with weekly paclitaxel in metastatic breast cancer treated with prior taxanes. Cancer Chemother Pharmacol 70:49–56PubMedCentralPubMedCrossRef Lustberg MB, Pant S, Ruppert AS, Shen T, Wei Y et al (2012) Phase I/II trial of non-cytotoxic suramin in combination with weekly paclitaxel in metastatic breast cancer treated with prior taxanes. Cancer Chemother Pharmacol 70:49–56PubMedCentralPubMedCrossRef
43.
Zurück zum Zitat Villalona-Calero MA, Otterson GA, Wientjes MG, Weber F, Bekaii-Saab T et al (2008) Noncytotoxic suramin as a chemosensitizer in patients with advanced non-small-cell lung cancer: a phase II study. Ann Oncol 19:1903–1909PubMedCrossRef Villalona-Calero MA, Otterson GA, Wientjes MG, Weber F, Bekaii-Saab T et al (2008) Noncytotoxic suramin as a chemosensitizer in patients with advanced non-small-cell lung cancer: a phase II study. Ann Oncol 19:1903–1909PubMedCrossRef
44.
Zurück zum Zitat Villalona-Calero MA, Wientjes MG, Otterson GA, Kanter S, Young D et al (2003) Phase I study of low-dose suramin as a chemosensitizer in patients with advanced non-small cell lung cancer. Clin Cancer Res 9:3303–3311PubMed Villalona-Calero MA, Wientjes MG, Otterson GA, Kanter S, Young D et al (2003) Phase I study of low-dose suramin as a chemosensitizer in patients with advanced non-small cell lung cancer. Clin Cancer Res 9:3303–3311PubMed
45.
Zurück zum Zitat Song S, Wientjes MG, Gan Y, Au JL (2000) Fibroblast growth factors: an epigenetic mechanism of broad spectrum resistance to anticancer drugs. Proc Natl Acad Sci U S A 97:8658–8663PubMedCentralPubMedCrossRef Song S, Wientjes MG, Gan Y, Au JL (2000) Fibroblast growth factors: an epigenetic mechanism of broad spectrum resistance to anticancer drugs. Proc Natl Acad Sci U S A 97:8658–8663PubMedCentralPubMedCrossRef
46.
Zurück zum Zitat Song S, Wientjes MG, Walsh C, Au JL (2001) Nontoxic doses of suramin enhance activity of paclitaxel against lung metastases. Cancer Res 61:6145–6150PubMed Song S, Wientjes MG, Walsh C, Au JL (2001) Nontoxic doses of suramin enhance activity of paclitaxel against lung metastases. Cancer Res 61:6145–6150PubMed
47.
Zurück zum Zitat Zhang Y, Song S, Yang F, Au JL, Wientjes MG (2001) Nontoxic doses of suramin enhance activity of doxorubicin in prostate tumors. J Pharmacol Exp Ther 299:426–433PubMed Zhang Y, Song S, Yang F, Au JL, Wientjes MG (2001) Nontoxic doses of suramin enhance activity of doxorubicin in prostate tumors. J Pharmacol Exp Ther 299:426–433PubMed
48.
Zurück zum Zitat Skliris GP, Munot K, Bell SM, Carder PJ, Lane S et al (2003) Reduced expression of oestrogen receptor beta in invasive breast cancer and its re-expression using DNA methyl transferase inhibitors in a cell line model. J Pathol 201:213–220PubMedCrossRef Skliris GP, Munot K, Bell SM, Carder PJ, Lane S et al (2003) Reduced expression of oestrogen receptor beta in invasive breast cancer and its re-expression using DNA methyl transferase inhibitors in a cell line model. J Pathol 201:213–220PubMedCrossRef
49.
Zurück zum Zitat Zhu WG, Hileman T, Ke Y, Wang P, Lu S et al (2004) 5-Aza-2′-deoxycytidine activates the p53/p21Waf1/Cip1 pathway to inhibit cell proliferation. J Biol Chem 279:15161–15166PubMedCrossRef Zhu WG, Hileman T, Ke Y, Wang P, Lu S et al (2004) 5-Aza-2′-deoxycytidine activates the p53/p21Waf1/Cip1 pathway to inhibit cell proliferation. J Biol Chem 279:15161–15166PubMedCrossRef
50.
Zurück zum Zitat Constantinides PG, Jones PA, Gevers W (1977) Functional striated muscle cells from non-myoblast precursors following 5-azacytidine treatment. Nature 267:364–366PubMedCrossRef Constantinides PG, Jones PA, Gevers W (1977) Functional striated muscle cells from non-myoblast precursors following 5-azacytidine treatment. Nature 267:364–366PubMedCrossRef
51.
Zurück zum Zitat Jones PA, Taylor SM (1980) Cellular differentiation, cytidine analogs and DNA methylation. Cell 20:85–93PubMedCrossRef Jones PA, Taylor SM (1980) Cellular differentiation, cytidine analogs and DNA methylation. Cell 20:85–93PubMedCrossRef
52.
Zurück zum Zitat Eiseler T, Doppler H, Yan IK, Kitatani K, Mizuno K et al (2009) Protein kinase D1 regulates cofilin-mediated F-actin reorganization and cell motility through slingshot. Nat Cell Biol 11:545–556PubMedCentralPubMedCrossRef Eiseler T, Doppler H, Yan IK, Kitatani K, Mizuno K et al (2009) Protein kinase D1 regulates cofilin-mediated F-actin reorganization and cell motility through slingshot. Nat Cell Biol 11:545–556PubMedCentralPubMedCrossRef
53.
Zurück zum Zitat Eiseler T, Hausser A, De Kimpe L, Van Lint J, Pfizenmaier K (2010) Protein kinase D controls actin polymerization and cell motility through phosphorylation of cortactin. J Biol Chem 285:18672–18683PubMedCrossRef Eiseler T, Hausser A, De Kimpe L, Van Lint J, Pfizenmaier K (2010) Protein kinase D controls actin polymerization and cell motility through phosphorylation of cortactin. J Biol Chem 285:18672–18683PubMedCrossRef
54.
Zurück zum Zitat Eiseler T, Schmid MA, Topbas F, Pfizenmaier K, Hausser A (2007) PKD is recruited to sites of actin remodelling at the leading edge and negatively regulates cell migration. FEBS Lett 581:4279–4287PubMedCrossRef Eiseler T, Schmid MA, Topbas F, Pfizenmaier K, Hausser A (2007) PKD is recruited to sites of actin remodelling at the leading edge and negatively regulates cell migration. FEBS Lett 581:4279–4287PubMedCrossRef
55.
Zurück zum Zitat Peterburs P, Heering J, Link G, Pfizenmaier K, Olayioye MA et al (2009) Protein kinase D regulates cell migration by direct phosphorylation of the cofilin phosphatase slingshot 1 like. Cancer Res 69:5634–5638PubMedCrossRef Peterburs P, Heering J, Link G, Pfizenmaier K, Olayioye MA et al (2009) Protein kinase D regulates cell migration by direct phosphorylation of the cofilin phosphatase slingshot 1 like. Cancer Res 69:5634–5638PubMedCrossRef
56.
Zurück zum Zitat Spratley SJ, Bastea LI, Doppler H, Mizuno K, Storz P (2011) Protein kinase D regulates cofilin activity through p21-activated kinase 4. J Biol Chem 286:34254–34261PubMedCrossRef Spratley SJ, Bastea LI, Doppler H, Mizuno K, Storz P (2011) Protein kinase D regulates cofilin activity through p21-activated kinase 4. J Biol Chem 286:34254–34261PubMedCrossRef
57.
Zurück zum Zitat Kim M, Jang HR, Kim JH, Noh SM, Song KS et al (2008) Epigenetic inactivation of protein kinase D1 in gastric cancer and its role in gastric cancer cell migration and invasion. Carcinogenesis 29:629–637PubMedCrossRef Kim M, Jang HR, Kim JH, Noh SM, Song KS et al (2008) Epigenetic inactivation of protein kinase D1 in gastric cancer and its role in gastric cancer cell migration and invasion. Carcinogenesis 29:629–637PubMedCrossRef
58.
Zurück zum Zitat Onishi Y, Kawamoto T, Kishimoto K, Hara H, Fukase N et al (2012) PKD1 negatively regulates cell invasion, migration and proliferation ability of human osteosarcoma. Int J Oncol 40:1839–1848PubMed Onishi Y, Kawamoto T, Kishimoto K, Hara H, Fukase N et al (2012) PKD1 negatively regulates cell invasion, migration and proliferation ability of human osteosarcoma. Int J Oncol 40:1839–1848PubMed
59.
Zurück zum Zitat Isham CR, Tibodeau JD, Jin W, Xu R, Timm MM et al (2007) Chaetocin: a promising new antimyeloma agent with in vitro and in vivo activity mediated via imposition of oxidative stress. Blood 109:2579–2588PubMedCrossRef Isham CR, Tibodeau JD, Jin W, Xu R, Timm MM et al (2007) Chaetocin: a promising new antimyeloma agent with in vitro and in vivo activity mediated via imposition of oxidative stress. Blood 109:2579–2588PubMedCrossRef
60.
Zurück zum Zitat Sundram V, Chauhan SC, Ebeling M, Jaggi M (2012) Curcumin attenuates beta-catenin signaling in prostate cancer cells through activation of protein kinase D1. PLoS ONE 7:e35368PubMedCentralPubMedCrossRef Sundram V, Chauhan SC, Ebeling M, Jaggi M (2012) Curcumin attenuates beta-catenin signaling in prostate cancer cells through activation of protein kinase D1. PLoS ONE 7:e35368PubMedCentralPubMedCrossRef
Metadaten
Titel
A combination treatment with DNA methyltransferase inhibitors and suramin decreases invasiveness of breast cancer cells
verfasst von
Sahra Borges
Heike R. Döppler
Peter Storz
Publikationsdatum
01.02.2014
Verlag
Springer US
Erschienen in
Breast Cancer Research and Treatment / Ausgabe 1/2014
Print ISSN: 0167-6806
Elektronische ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-014-2857-2

Weitere Artikel der Ausgabe 1/2014

Breast Cancer Research and Treatment 1/2014 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.