Characterisation and manipulation of docetaxel resistant prostate cancer cell lines

Amanda J O'Neill, Maria Prencipe, Catherine Dowling, Yue Fan, Laoighse Mulrane, William M Gallagher, Darran O'Connor, Robert O'Connor, Aoife Devery, Claire Corcoran, Sweta Rani, Lorraine O'Driscoll, John M Fitzpatrick, R William G Watson, Amanda J O'Neill, Maria Prencipe, Catherine Dowling, Yue Fan, Laoighse Mulrane, William M Gallagher, Darran O'Connor, Robert O'Connor, Aoife Devery, Claire Corcoran, Sweta Rani, Lorraine O'Driscoll, John M Fitzpatrick, R William G Watson

Abstract

Background: There is no effective treatment strategy for advanced castration-resistant prostate cancer. Although Docetaxel (Taxotere®) represents the most active chemotherapeutic agent it only gives a modest survival advantage with most patients eventually progressing because of inherent or acquired drug resistance. The aims of this study were to further investigate the mechanisms of resistance to Docetaxel. Three Docetaxel resistant sub-lines were generated and confirmed to be resistant to the apoptotic and anti-proliferative effects of increasing concentrations of Docetaxel.

Results: The resistant DU-145 R and 22RV1 R had expression of P-glycoprotein and its inhibition with Elacridar partially and totally reversed the resistant phenotype in the two cell lines respectively, which was not seen in the PC-3 resistant sublines. Resistance was also not mediated in the PC-3 cells by cellular senescence or autophagy but multiple changes in pro- and anti-apoptotic genes and proteins were demonstrated. Even though there were lower basal levels of NF-κB activity in the PC-3 D12 cells compared to the Parental PC-3, docetaxel induced higher NF-κB activity and IκB phosphorylation at 3 and 6 hours with only minor changes in the DU-145 cells. Inhibition of NF-κB with the BAY 11-7082 inhibitor reversed the resistance to Docetaxel.

Conclusion: This study confirms that multiple mechanisms contribute to Docetaxel resistance and the central transcription factor NF-κB plays an immensely important role in determining docetaxel-resistance which may represent an appropriate therapeutic target.

Trial registration: ClinicalTrials.gov NCT00571675.

Figures

Figure 1
Figure 1
Effects of Docetaxel on apoptosis and proliferation in the PC-3, DU-145 and 22RV1 Docetaxel resistant sublines. A. The PC-3 Ag, PC-3 D8, PC-3 D12, DU-145 Ag, DU-145 R, 22RV1 Ag and 22RV1 R cell lines were treated with Docetaxel (20 and 80 nM) for 48 hrs. Apoptosis was assessed by propidium iodide DNA staining and flow cytometry. Values are expressed as mean ± standard deviation. An independent samples t-test was used to compare the mean percentage apoptosis for control, 20 nM and 80 nM for each cell line. **p < 0.001. B. Cell proliferation was measured using the MTT Assay. The PC-3 sublines (PC-3 Ag, PC-3 D8 and PC-3 D12) were treated with increasing doses of Docetaxel (10, 20, 50 & 100 nM) for 24, 48 and 72 hrs. Data shown is representative of three independent experiments.
Figure 2
Figure 2
P-gp expression levels in the Doctaxel resistant cell lines. A. Total cellular protein was extracted from the cells and 50 μg was assessed by western blotting. P-gp was not expressed in the PC-3 Ag, PC-3 D8, PC-3 D12 sublines or the DLKP (40 μg) negative control cell line, but was expressed in the DLKP-A (5 μg) drug resistant variant of the DLKP cell line. β-actin the house keeping protein was used as loading control for P-Gp. B. The PC-3 Ag, PC-3 D12 and NCI-ADR/RES cells lines were treated with Docetaxel (20 nM) alone or in the presence of the P-gylcoprotein inhibitor Elacridar (0.25 and 0.5 μM) and Docetaxel (20 nM) and assessed for apoptosis by flow cytometry. An independent samples t-test was used to compare the average percentage of apoptosis between the control groups and each of the treated groups for the PC-3 Ag, PC-3 D12 and NCI-ADR/RES cell lines *p- < 0.05. C. Total cellular protein was extracted from the cells and 50 μg was assessed by western blotting. P-Gp was weakly expressed in the DU-145 R and strongly expressed in the 22RV1 R sublines. No expression was seen in the Aged matched. β-actin the house keeping protein was used as loading control for p-glycoprotein. D. The DU-145 Ag, DU-145 R and 22RV1 Ag and 22RV1 R cells lines were treated with Docetaxel (20 nM) alone, or the P-Gp inhibitor Elacridar (0.25 μM) alone or in the presence of Elacridar (0.25 μM) and Docetaxel (20 nM) and assessed for apoptosis by flow cytometry. Blots shown are representative of three independent experiments.
Figure 3
Figure 3
Cellular sensecence in response to Docetaxel treatment. The PC-3 Ag, PC-3 D8 and PC-3 D12 cells were plated in 6 well plates at 300,000/well and grown for 24 hours. The cells were then treated with Docetaxel (100 nM) for 48 hours and fixed with 2% formaldehyde and 0.2% glutaraldehyde. Senescent cells were then assessed by staining cells for β-galactosidase expression. Positive cells were counted and expressed as a percentage of the total number of cells in the well. Pictures are a representation of 5 independent experiments.
Figure 4
Figure 4
Autophagy as a mechanisms of Docetaxel resistance. Following treatment of the Aged matched controls (Upper panel) and PC-3 D12 resistant sublines (Bottom panel) with 50 nM of Docetaxel for 1, 3, 6, 12 and 24 hrs, total cellular protein was extracted and assessed for LC3II by western blotting using the relevant anti-body and conditions as described in the methods section. For positive controls cells were either treated with Rapamycin (R, 0.2 μM) for 4 hrs, Starvation (S, the cells were starved in EBSS (Earls Balanced Salt Solution) medium for 2 hrs), or Starvation plus Bafilomycin (S+B, 20 nM) for 2 hrs. β-actin the house keeping protein was used as loading control. Blots shown are representative of three independent experiments.
Figure 5
Figure 5
Validation of Low Density Arrays. Total cellular protein was extracted from the PC-3 sublines (PC-3 Ag, PC-3 D8 & PC-3 D12) and 50 μg was loaded onto a western blot to assess the expression of Clusterin, HSP90, Id-1, Bcl-2, Bcl-xL, Bax, and Bid using their respective antibodies as described in the Method section. The house keeping protein β-actin was used as a loading control. Blots shown are representative of three independent experiments.
Figure 6
Figure 6
NF-κB transcriptional activity and Baseline NFkB subunit activity. A. PC-3 Ag and PC-3 D12 cells were plated at 100,000/per well and grown for 24 hours. The baseline NFκB activity was assessed by luciferase assay, using a Dual Luciferase Assay from Promega following the manufactures instructions. Averages from 3 independent experiments are shown. Mean values were compared using t-test assuming equal variances. B. The PC-3 Ag and PC-3 D12 cells were grown to confluency and nuclear extracts prepared as per the TransAM™ Assay Kit manual. Averages from 3 independent experiments are shown. Mean values were compared using t-test assuming equal variances.
Figure 7
Figure 7
NF-κB transcriptional activity following docetaxel treatment. A. The PC-3 and DU-145 aged matched and Docetaxel resistant sublines were treated with 50 uM concentrations of Docetaxel for 3, 6 and 24 hours at which time NFκB activity was assessed by luciferase assay, using a Dual Luciferase Assay from Promega following the manufactures instructions. Graphs are generated from 3 independent experiments. Mean values were compared using t-test assuming equal variances. * p < 0.01; ** p < 0.0001. B. Cells were treated as above and at the different time points total cellular protein was extracted and 50 ug loaded onto western blots and assessed for P-IkBα and IkBα. β-actin was used as an additional loading control. Blots are representative of three independent experiments.
Figure 8
Figure 8
NF-κB inhibitor studies following docetaxel treatment. The PC-3 sublines where pre-treated with the NFκB inhibitor (BAY 11-7082, 5 μM) for 24 hours followed by Docetaxel (20 nM) for a further 48 hours and assessed for apoptosis by flow cytometry. Graphs represent the results of 3 independent experiments. Mean values were compared using t-test assuming equal variances. * p < 0.05.

References

    1. Mostaghel EA, Page ST, Lin DW, Fazli L, Coleman IM, True LD, Knudsen B, Hess DL, Nelson CC, Matsumoto AM. et al.Intraprostatic androgens and androgen-regulated gene expression persist after testosterone suppression: therapeutic implications for castration-resistant prostate cancer. Cancer Res. 2007;67:5033–5041. doi: 10.1158/0008-5472.CAN-06-3332.
    1. Scher HI, Halabi S, Tannock I, Morris M, Sternberg CN, Carducci MA, Eisenberger MA, Higano C, Bubley GJ, Dreicer R. et al.Design and end points of clinical trials for patients with progressive prostate cancer and castrate levels of testosterone: recommendations of the Prostate Cancer Clinical Trials Working Group. J Clin Oncol. 2008;26:1148–1159. doi: 10.1200/JCO.2007.12.4487.
    1. Raghavan D, Koczwara B, Javle M. Evolving strategies of cytotoxic chemotherapy for advanced prostate cancer. Eur J Cancer. 1997;33:566–574. doi: 10.1016/S0959-8049(96)00510-2.
    1. van Brussel JP, Mickisch GH. Multidrug resistance in prostate cancer. Onkologie. 2003;26:175–181. doi: 10.1159/000071510.
    1. Makarovskiy AN, Siryaporn E, Hixson DC, Akerley W. Survival of docetaxel-resistant prostate cancer cells in vitro depends on phenotype alterations and continuity of drug exposure. Cell Mol Life Sci. 2002;59:1198–1211. doi: 10.1007/s00018-002-8498-3.
    1. Shen MM, Abate-Shen C. Pten inactivation and the emergence of androgen-independent prostate cancer. Cancer Res. 2007;67:6535–6538. doi: 10.1158/0008-5472.CAN-07-1271.
    1. Feldman BJ, Feldman D. The development of androgen-independent prostate cancer. Nat Rev Cancer. 2001;1:34–45. doi: 10.1038/35094009.
    1. McEleny K, Coffey R, Morrissey C, Williamson K, Zangemeister-Wittke U, Fitzpatrick JM, Watson RW. An antisense oligonucleotide to cIAP-1 sensitizes prostate cancer cells to fas and TNFalpha mediated apoptosis. Prostate. 2004;59:419–425. doi: 10.1002/pros.10371.
    1. Gibbons NB, Watson RW, Coffey RN, Brady HP, Fitzpatrick JM. Heat-shock proteins inhibit induction of prostate cancer cell apoptosis. Prostate. 2000;45:58–65. doi: 10.1002/1097-0045(20000915)45:1<58::AID-PROS7>;2-#.
    1. Sternberg CN, Dumez H, Van Poppel H, Skoneczna I, Sella A, Daugaard G, Gil T, Graham J, Carpentier P, Calabro F. et al.Docetaxel plus oblimersen sodium (Bcl-2 antisense oligonucleotide): an EORTC multicenter, randomized phase II study in patients with castration-resistant prostate cancer. Ann Oncol. 2009;20:1264–1269. doi: 10.1093/annonc/mdn784.
    1. De Marzo AM, Platz EA, Sutcliffe S, Xu J, Gronberg H, Drake CG, Nakai Y, Isaacs WB, Nelson WG. Inflammation in prostate carcinogenesis. Nat Rev Cancer. 2007;7:256–269. doi: 10.1038/nrc2090.
    1. McEleny K, Coffey R, Morrissey C, Fitzpatrick JM, Watson RW. Caffeic acid phenethyl ester-induced PC-3 cell apoptosis is caspase-dependent and mediated through the loss of inhibitors of apoptosis proteins. BJU Int. 2004;94:402–406. doi: 10.1111/j.1464-410X.2004.04936.x.
    1. Domingo-Domenech J, Oliva C, Rovira A, Codony-Servat J, Bosch M, Filella X, Montagut C, Tapia M, Campas C, Dang L. et al.Interleukin 6, a nuclear factor-kappaB target, predicts resistance to docetaxel in hormone-independent prostate cancer and nuclear factor-kappaB inhibition by PS-1145 enhances docetaxel antitumor activity. Clin Cancer Res. 2006;12:5578–5586. doi: 10.1158/1078-0432.CCR-05-2767.
    1. Gill C, Dowling C, O'Neill AJ, Watson RW. Effects of cIAP-1, cIAP-2 and XIAP triple knockdown on prostate cancer cell susceptibility to apoptosis, cell survival and proliferation. Mol Cancer. 2009;8:39. doi: 10.1186/1476-4598-8-39.
    1. Prencipe M, Fitzpatrick P, Gorman S, Tosetto M, Klinger R, Furlong F, Harrison M, O'Connor D, Roninson IB, O'Sullivan J, McCann A. Cellular senescence induced by aberrant MAD2 levels impacts on paclitaxel responsiveness in vitro. Br J Cancer. 2009;101:1900–1908. doi: 10.1038/sj.bjc.6605419.
    1. Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, Medrano EE, Linskens M, Rubelj I, Pereira-Smith O. et al.A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci USA. 1995;92:9363–9367. doi: 10.1073/pnas.92.20.9363.
    1. Maria McCrohan A, Morrissey C, O'Keane C, Mulligan N, Watson C, Smith J, Fitzpatrick JM, Watson RW. Effects of the dual 5 alpha-reductase inhibitor dutasteride on apoptosis in primary cultures of prostate cancer epithelial cells and cell lines. Cancer. 2006;106:2743–2752. doi: 10.1002/cncr.21938.
    1. Walsh S, Gill C, O'Neill A, Fitzpatrick JM, Watson RW. Hypoxia increases normal prostate epithelial cell resistance to receptor-mediated apoptosis via AKT activation. Int J Cancer. 2009;124:1871–1878. doi: 10.1002/ijc.24145.
    1. Collins DM, Crown J, O'Donovan N, Devery A, O'Sullivan F, O'Driscoll L, Clynes M, O'Connor R. Tyrosine kinase inhibitors potentiate the cytotoxicity of MDR-substrate anticancer agents independent of growth factor receptor status in lung cancer cell lines. Invest New Drugs. pp. 433–444.
    1. Breedveld P, Beijnen JH, Schellens JH. Use of P-glycoprotein and BCRP inhibitors to improve oral bioavailability and CNS penetration of anticancer drugs. Trends Pharmacol Sci. 2006;27:17–24. doi: 10.1016/j.tips.2005.11.009.
    1. Hyafil F, Vergely C, Du Vignaud P, Grand-Perret T. In vitro and in vivo reversal of multidrug resistance by GF120918, an acridonecarboxamide derivative. Cancer Res. 1209;53:4595–4602.
    1. Nakatogawa H, Suzuki K, Kamada Y, Ohsumi Y. Dynamics and diversity in autophagy mechanisms: lessons from yeast. Nat Rev Mol Cell Biol. 2009;10:458–467. doi: 10.1038/nrm2708.
    1. Morselli E, Galluzzi L, Kepp O, Vicencio JM, Criollo A, Maiuri MC, Kroemer G. Anti- and pro-tumor functions of autophagy. Biochim Biophys Acta. 2009;1793:1524–1532. doi: 10.1016/j.bbamcr.2009.01.006.
    1. Klionsky DJ, Abeliovich H, Agostinis P, Agrawal DK, Aliev G, Askew DS, Baba M, Baehrecke EH, Bahr BA, Ballabio A. et al.Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy. 2008;4:151–175.
    1. Mahon KL, Henshall SM, Sutherland RL, Horvath LG. Pathways of chemotherapy resistance in castration-resistant prostate cancer. Endocr Relat Cancer. 2011;18:103–123. doi: 10.1530/ERC-10-0343.
    1. Takeda M, Mizokami A, Mamiya K, Li YQ, Zhang J, Keller ET, Namiki M. The establishment of two paclitaxel-resistant prostate cancer cell lines and the mechanisms of paclitaxel resistance with two cell lines. Prostate. 2007;67:955–967. doi: 10.1002/pros.20581.
    1. de Bono JS, Oudard S, Ozguroglu M, Hansen S, Machiels JP, Kocak I, Gravis G, Bodrogi I, Mackenzie MJ, Shen L, Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet. pp. 1147–1154.
    1. Trougakos IP, So A, Jansen B, Gleave ME, Gonos ES. Silencing expression of the clusterin/apolipoprotein j gene in human cancer cells using small interfering RNA induces spontaneous apoptosis, reduced growth ability, and cell sensitization to genotoxic and oxidative stress. Cancer Res. 2004;64:1834–1842. doi: 10.1158/0008-5472.CAN-03-2664.
    1. Shannan B, Seifert M, Leskov K, Willis J, Boothman D, Tilgen W, Reichrath J. Challenge and promise: roles for clusterin in pathogenesis, progression and therapy of cancer. Cell Death Differ. 2006;13:12–19. doi: 10.1038/sj.cdd.4401779.
    1. Zhang H, Kim JK, Edwards CA, Xu Z, Taichman R, Wang CY. Clusterin inhibits apoptosis by interacting with activated Bax. Nat Cell Biol. 2005;7:909–915. doi: 10.1038/ncb1291.
    1. Steinberg J, Oyasu R, Lang S, Sintich S, Rademaker A, Lee C, Kozlowski JM, Sensibar JA. Intracellular levels of SGP-2 (Clusterin) correlate with tumor grade in prostate cancer. Clin Cancer Res. 1997;3:1707–1711.
    1. Sowery RD, Hadaschik BA, So AI, Zoubeidi A, Fazli L, Hurtado-Coll A, Gleave ME. Clusterin knockdown using the antisense oligonucleotide OGX-011 re-sensitizes docetaxel-refractory prostate cancer PC-3 cells to chemotherapy. BJU Int. 2008;102:389–397. doi: 10.1111/j.1464-410X.2008.07618.x.
    1. Simpson LA, Burwell EA, Thompson KA, Shahnaz S, Chen AR, Loeb DM. The antiapoptotic gene A1/BFL1 is a WT1 target gene that mediates granulocytic differentiation and resistance to chemotherapy. Blood. 2006;107:4695–4702. doi: 10.1182/blood-2005-10-4025.
    1. Modur V, Nagarajan R, Evers BM, Milbrandt J. FOXO proteins regulate tumor necrosis factor-related apoptosis inducing ligand expression. Implications for PTEN mutation in prostate cancer. J Biol Chem. 2002;277:47928–47937. doi: 10.1074/jbc.M207509200.
    1. Yasuda M, Han JW, Dionne CA, Boyd JM, Chinnadurai G. BNIP3alpha: a human homolog of mitochondrial proapoptotic protein BNIP3. Cancer Res. 1999;59:533–537.
    1. Degli-Esposti MA, Dougall WC, Smolak PJ, Waugh JY, Smith CA, Goodwin RG. The novel receptor TRAIL-R4 induces NF-kappaB and protects against TRAIL-mediated apoptosis, yet retains an incomplete death domain. Immunity. 1997;7:813–820. doi: 10.1016/S1074-7613(00)80399-4.
    1. Karin M, Yamamoto Y, Wang QM. The IKK NF-kappa B system: a treasure trove for drug development. Nat Rev Drug Discov. 2004;3:17–26. doi: 10.1038/nrd1279.
    1. Pu YS, Hour TC, Chuang SE, Cheng AL, Lai MK, Kuo ML. Interleukin-6 is responsible for drug resistance and anti-apoptotic effects in prostatic cancer cells. Prostate. 2004;60:120–129. doi: 10.1002/pros.20057.
    1. Venkatraman M, Anto RJ, Nair A, Varghese M, Karunagaran D. Biological and chemical inhibitors of NF-kappaB sensitize SiHa cells to cisplatin-induced apoptosis. Mol Carcinog. 2005;44:51–59. doi: 10.1002/mc.20116.
    1. Hour TC, Chen J, Huang CY, Guan JY, Lu SH, Pu YS. Curcumin enhances cytotoxicity of chemotherapeutic agents in prostate cancer cells by inducing p21(WAF1/CIP1) and C/EBPbeta expressions and suppressing NF-kappaB activation. Prostate. 2002;51:211–218. doi: 10.1002/pros.10089.
    1. Li Y, Ahmed F, Ali S, Philip PA, Kucuk O, Sarkar FH. Inactivation of nuclear factor kappaB by soy isoflavone genistein contributes to increased apoptosis induced by chemotherapeutic agents in human cancer cells. Cancer Res. 2005;65:6934–6942. doi: 10.1158/0008-5472.CAN-04-4604.

Source: PubMed

3
S'abonner