Skip to main content
Top
Published in: Molecular Cancer 1/2014

Open Access 01-12-2014 | Research

HDAC-inhibition counteracts everolimus resistance in renal cell carcinoma in vitro by diminishing cdk2 and cyclin A

Authors: Eva Juengel, Snigdha Nowaz, Jasmina Makarevi, Iyad Natsheh, Isabella Werner, Karen Nelson, Michael Reiter, Igor Tsaur, Jens Mani, Sebastian Harder, Georg Bartsch, Axel Haferkamp, Roman A Blaheta

Published in: Molecular Cancer | Issue 1/2014

Login to get access

Abstract

Background

Targeted therapies have improved therapeutic options of treating renal cell carcinoma (RCC). However, drug response is temporary due to resistance development.

Methods

Functional and molecular changes in RCC Caki-1 cells, after acquired resistance to the mammalian target of rapamycin (mTOR)-inhibitor everolimus (Cakires), were investigated with and without additional application of the histone deacetylase (HDAC)-inhibitor valproic acid (VPA). Cell growth was evaluated by MTT assay, cell cycle progression and apoptosis by flow cytometry. Target molecules of everolimus and VPA, apoptotic and cell cycle regulating proteins were investigated by western blotting. siRNA blockade was performed to evaluate the functional relevance of the proteins.

Results

Everolimus resistance was accompanied by significant increases in the percentage of G2/M-phase cells and in the IC50. Akt and p70S6K, targets of everolimus, were activated in Cakires compared to drug sensitive cells. The most prominent change in Cakires cells was an increase in the cell cycle activating proteins cdk2 and cyclin A. Knock-down of cdk2 and cyclin A caused significant growth inhibition in the Cakires cells. The HDAC-inhibitor, VPA, counteracted everolimus resistance in Cakires, evidenced by a significant decrease in tumor growth and cdk2/cyclin A.

Conclusion

It is concluded that non-response to everolimus is characterized by increased cdk2/cyclin A, driving RCC cells into the G2/M-phase. VPA hinders everolimus non-response by diminishing cdk2/cyclin A. Therefore, treatment with HDAC-inhibitors might be an option for patients with advanced renal cell carcinoma and acquired everolimus resistance.
Appendix
Available only for authorised users
Literature
2.
go back to reference Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, Grünwald V, Thompson JA, Figlin RA, Hollaender N, Urbanowitz G, Berg WJ, Kay A, Lebwohl D, Ravaud A: RECORD-1 Study Group: Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet. 2008, 372 (9637): 449-456. 10.1016/S0140-6736(08)61039-9CrossRefPubMed Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, Grünwald V, Thompson JA, Figlin RA, Hollaender N, Urbanowitz G, Berg WJ, Kay A, Lebwohl D, Ravaud A: RECORD-1 Study Group: Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet. 2008, 372 (9637): 449-456. 10.1016/S0140-6736(08)61039-9CrossRefPubMed
3.
go back to reference Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, Grünwald V, Thompson JA, Figlin RA, Hollaender N, Kay A, Ravaud A: RECORD‒1 Study Group: Phase 3 trial of everolimus for metastatic renal cell carcinoma: final results and analysis of prognostic factors. Cancer. 2010, 116 (18): 4256-4265. 10.1002/cncr.25219CrossRefPubMed Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, Grünwald V, Thompson JA, Figlin RA, Hollaender N, Kay A, Ravaud A: RECORD‒1 Study Group: Phase 3 trial of everolimus for metastatic renal cell carcinoma: final results and analysis of prognostic factors. Cancer. 2010, 116 (18): 4256-4265. 10.1002/cncr.25219CrossRefPubMed
4.
go back to reference Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, Staroslawska E, Sosman J, McDermott D, Bodrogi I, Kovacevic Z, Lesovoy V, Schmidt-Wolf IG, Barbarash O, Gokmen E, O'Toole T, Lustgarten S, Moore L, Motzer RJ: Global ARCC Trial: Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med. 2007, 356 (22): 2271-2281. 10.1056/NEJMoa066838CrossRefPubMed Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, Staroslawska E, Sosman J, McDermott D, Bodrogi I, Kovacevic Z, Lesovoy V, Schmidt-Wolf IG, Barbarash O, Gokmen E, O'Toole T, Lustgarten S, Moore L, Motzer RJ: Global ARCC Trial: Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med. 2007, 356 (22): 2271-2281. 10.1056/NEJMoa066838CrossRefPubMed
5.
go back to reference Harada K, Miyake H, Kumano M, Fujisawa M: Acquired resistance to temsirolimus in human renal cell carcinoma cells is mediated by the constitutive activation of signal transduction pathways through mTORC2. Br J Cancer. 2013, 109 (9): 2389-2395. 10.1038/bjc.2013.602PubMedCentralCrossRefPubMed Harada K, Miyake H, Kumano M, Fujisawa M: Acquired resistance to temsirolimus in human renal cell carcinoma cells is mediated by the constitutive activation of signal transduction pathways through mTORC2. Br J Cancer. 2013, 109 (9): 2389-2395. 10.1038/bjc.2013.602PubMedCentralCrossRefPubMed
6.
go back to reference Juengel E, Dauselt A, Makarević J, Wiesner C, Tsaur I, Bartsch G, Haferkamp A, Blaheta RA: Acetylation of histone H3 prevents resistance development caused by chronic mTOR inhibition in renal cell carcinoma cells. Cancer Lett. 2012, 324 (1): 83-90. 10.1016/j.canlet.2012.05.003CrossRefPubMed Juengel E, Dauselt A, Makarević J, Wiesner C, Tsaur I, Bartsch G, Haferkamp A, Blaheta RA: Acetylation of histone H3 prevents resistance development caused by chronic mTOR inhibition in renal cell carcinoma cells. Cancer Lett. 2012, 324 (1): 83-90. 10.1016/j.canlet.2012.05.003CrossRefPubMed
7.
go back to reference Tsaur I, Makarević J, Hudak L, Juengel E, Kurosch M, Wiesner C, Bartsch G, Harder S, Haferkamp A, Blaheta RA: The cdk1-cyclin B complex is involved in everolimus triggered resistance in the PC3 prostate cancer cell line. Cancer Lett. 2011, 313 (1): 84-90. 10.1016/j.canlet.2011.08.026CrossRefPubMed Tsaur I, Makarević J, Hudak L, Juengel E, Kurosch M, Wiesner C, Bartsch G, Harder S, Haferkamp A, Blaheta RA: The cdk1-cyclin B complex is involved in everolimus triggered resistance in the PC3 prostate cancer cell line. Cancer Lett. 2011, 313 (1): 84-90. 10.1016/j.canlet.2011.08.026CrossRefPubMed
8.
go back to reference Santoni M, Pantano F, Amantini C, Nabissi M, Conti A, Burattini L, Zoccoli A, Berardi R, Santoni G, Tonini G, Santini D, Cascinu S: Emerging strategies to overcome the resistance to current mTOR inhibitors in renal cell carcinoma. Biochim Biophys Acta. 2014, 1845 (2): 221-231.PubMed Santoni M, Pantano F, Amantini C, Nabissi M, Conti A, Burattini L, Zoccoli A, Berardi R, Santoni G, Tonini G, Santini D, Cascinu S: Emerging strategies to overcome the resistance to current mTOR inhibitors in renal cell carcinoma. Biochim Biophys Acta. 2014, 1845 (2): 221-231.PubMed
9.
go back to reference Chen CH, Chen MC, Wang JC, Tsai AC, Chen CS, Liou JP, Pan SL, Teng CM: Synergistic Interaction between the HDAC Inhibitor, MPT0E028, and Sorafenib in Liver Cancer Cells In Vitro and In Vivo. Clin Cancer Res. 2014, 20 (5): 1274-1287. 10.1158/1078-0432.CCR-12-3909PubMedCentralCrossRefPubMed Chen CH, Chen MC, Wang JC, Tsai AC, Chen CS, Liou JP, Pan SL, Teng CM: Synergistic Interaction between the HDAC Inhibitor, MPT0E028, and Sorafenib in Liver Cancer Cells In Vitro and In Vivo. Clin Cancer Res. 2014, 20 (5): 1274-1287. 10.1158/1078-0432.CCR-12-3909PubMedCentralCrossRefPubMed
10.
go back to reference Hrabeta J, Stiborova M, Adam V, Kizek R, Eckschlager T: Histone deacetylase inhibitors in cancer therapy. A review. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. in press, Hrabeta J, Stiborova M, Adam V, Kizek R, Eckschlager T: Histone deacetylase inhibitors in cancer therapy. A review. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. in press,
11.
go back to reference Ellis L, Ku SY, Ramakrishnan S, Lasorsa E, Azabdaftari G, Godoy A, Pili R: Combinatorial antitumor effect of HDAC and the PI3K-Akt-mTOR pathway inhibition in a Pten defecient model of prostate cancer. Oncotarget. 2013, 4 (12): 2225-2236.PubMedCentralCrossRefPubMed Ellis L, Ku SY, Ramakrishnan S, Lasorsa E, Azabdaftari G, Godoy A, Pili R: Combinatorial antitumor effect of HDAC and the PI3K-Akt-mTOR pathway inhibition in a Pten defecient model of prostate cancer. Oncotarget. 2013, 4 (12): 2225-2236.PubMedCentralCrossRefPubMed
12.
go back to reference de Jong E, Winkel P, Poelstra K, Prakash J: Anticancer effects of 15d-prostaglandin-J2 in wild-type and doxorubicin-resistant ovarian cancer cells: novel actions on SIRT1 and HDAC. PLoS One. 2011, 6: e25192- 10.1371/journal.pone.0025192PubMedCentralCrossRefPubMed de Jong E, Winkel P, Poelstra K, Prakash J: Anticancer effects of 15d-prostaglandin-J2 in wild-type and doxorubicin-resistant ovarian cancer cells: novel actions on SIRT1 and HDAC. PLoS One. 2011, 6: e25192- 10.1371/journal.pone.0025192PubMedCentralCrossRefPubMed
13.
go back to reference Stronach EA, Alfraidi A, Rama N, Datler C, Studd JB, Agarwal R, Guney TG, Gourley C, Hennessy BT, Mills GB, Mai A, Brown R, Dina R, Gabra H: HDAC4-regulated STAT1 activation mediates platinum resistance in ovarian cancer. Cancer Res. 2011, 71: 4412-4422. 10.1158/0008-5472.CAN-10-4111PubMedCentralCrossRefPubMed Stronach EA, Alfraidi A, Rama N, Datler C, Studd JB, Agarwal R, Guney TG, Gourley C, Hennessy BT, Mills GB, Mai A, Brown R, Dina R, Gabra H: HDAC4-regulated STAT1 activation mediates platinum resistance in ovarian cancer. Cancer Res. 2011, 71: 4412-4422. 10.1158/0008-5472.CAN-10-4111PubMedCentralCrossRefPubMed
14.
go back to reference Fong JT, Jacobs RJ, Moravec DN, Uppada SB, Botting GM, Nlend M, Puri N: Alternative Signaling Pathways as Potential Therapeutic Targets for Overcoming EGFR and c-Met Inhibitor Resistance in Non-Small Cell Lung Cancer. PLoS One. 2013, 8 (11): e78398- 10.1371/journal.pone.0078398PubMedCentralCrossRefPubMed Fong JT, Jacobs RJ, Moravec DN, Uppada SB, Botting GM, Nlend M, Puri N: Alternative Signaling Pathways as Potential Therapeutic Targets for Overcoming EGFR and c-Met Inhibitor Resistance in Non-Small Cell Lung Cancer. PLoS One. 2013, 8 (11): e78398- 10.1371/journal.pone.0078398PubMedCentralCrossRefPubMed
15.
go back to reference Sakai I, Miyake H, Fujisawa M: Acquired resistance to sunitinib in human renal cell carcinoma cells is mediated by constitutive activation of signal transduction pathways associated with tumour cell proliferation. BJU Int. 2013, 112 (2): E211-E220. 10.1111/j.1464-410X.2012.11655.xCrossRefPubMed Sakai I, Miyake H, Fujisawa M: Acquired resistance to sunitinib in human renal cell carcinoma cells is mediated by constitutive activation of signal transduction pathways associated with tumour cell proliferation. BJU Int. 2013, 112 (2): E211-E220. 10.1111/j.1464-410X.2012.11655.xCrossRefPubMed
16.
go back to reference Liu J, Xiao Z, Wong SK, Tin VP, Ho KY, Wang J, Sham MH, Wong MP: Lung cancer tumorigenicity and drug resistance are maintained through ALDH(hi)CD44(hi) tumor initiating cells. Oncotarget. 2013, 4 (10): 1698-1711.PubMedCentralCrossRefPubMed Liu J, Xiao Z, Wong SK, Tin VP, Ho KY, Wang J, Sham MH, Wong MP: Lung cancer tumorigenicity and drug resistance are maintained through ALDH(hi)CD44(hi) tumor initiating cells. Oncotarget. 2013, 4 (10): 1698-1711.PubMedCentralCrossRefPubMed
17.
go back to reference Lin A, Piao HL, Zhuang L, Sarbassov DD, Ma L, Gan B: FoxO transcription factors promote AKT Ser473 phosphorylation and renal tumor growth in response to pharmacological inhibition of the PI3K-AKT pathway. Cancer Res. 2014, 74 (6): 1682-1693. 10.1158/0008-5472.CAN-13-1729PubMedCentralCrossRefPubMed Lin A, Piao HL, Zhuang L, Sarbassov DD, Ma L, Gan B: FoxO transcription factors promote AKT Ser473 phosphorylation and renal tumor growth in response to pharmacological inhibition of the PI3K-AKT pathway. Cancer Res. 2014, 74 (6): 1682-1693. 10.1158/0008-5472.CAN-13-1729PubMedCentralCrossRefPubMed
18.
go back to reference Juengel E, Makarević J, Tsaur I, Bartsch G, Nelson K, Haferkamp A, Blaheta RA: Resistance after chronic application of the HDAC-inhibitor valproic acid is associated with elevated Akt activation in renal cell carcinoma in vivo. PLoS One. 2013, 8 (1): e53100- 10.1371/journal.pone.0053100PubMedCentralCrossRefPubMed Juengel E, Makarević J, Tsaur I, Bartsch G, Nelson K, Haferkamp A, Blaheta RA: Resistance after chronic application of the HDAC-inhibitor valproic acid is associated with elevated Akt activation in renal cell carcinoma in vivo. PLoS One. 2013, 8 (1): e53100- 10.1371/journal.pone.0053100PubMedCentralCrossRefPubMed
19.
go back to reference Joeckel E, Haber T, Prawitt D, Junker K, Hampel C, Thüroff JW, Roos FC, Brenner W: High calcium concentration in bones promotes bone metastasis in renal cell carcinomas expressing calcium-sensing receptor. Mol Cancer. 2014, 13 (1): 42- 10.1186/1476-4598-13-42PubMedCentralCrossRefPubMed Joeckel E, Haber T, Prawitt D, Junker K, Hampel C, Thüroff JW, Roos FC, Brenner W: High calcium concentration in bones promotes bone metastasis in renal cell carcinomas expressing calcium-sensing receptor. Mol Cancer. 2014, 13 (1): 42- 10.1186/1476-4598-13-42PubMedCentralCrossRefPubMed
20.
go back to reference Ou YC, Li JR, Kuan YH, Raung SL, Wang CC, Hung YY, Pan PH, Lu HC, Chen CJ: Luteolin sensitizes human 786-O renal cell carcinoma cells to TRAIL-induced apoptosis. Life Sci. 2014, 100 (2): 110-117. 10.1016/j.lfs.2014.02.002CrossRefPubMed Ou YC, Li JR, Kuan YH, Raung SL, Wang CC, Hung YY, Pan PH, Lu HC, Chen CJ: Luteolin sensitizes human 786-O renal cell carcinoma cells to TRAIL-induced apoptosis. Life Sci. 2014, 100 (2): 110-117. 10.1016/j.lfs.2014.02.002CrossRefPubMed
21.
go back to reference White NM, Masui O, Newsted D, Scorilas A, Romaschin AD, Bjarnason GA, Siu KW, Yousef GM: Galectin-1 has potential prognostic significance and is implicated in clear cell renal cell carcinoma progression through the HIF/mTOR signaling axis. Br J Cancer. 2014, 110 (5): 1250-1259. 10.1038/bjc.2013.828PubMedCentralCrossRefPubMed White NM, Masui O, Newsted D, Scorilas A, Romaschin AD, Bjarnason GA, Siu KW, Yousef GM: Galectin-1 has potential prognostic significance and is implicated in clear cell renal cell carcinoma progression through the HIF/mTOR signaling axis. Br J Cancer. 2014, 110 (5): 1250-1259. 10.1038/bjc.2013.828PubMedCentralCrossRefPubMed
22.
go back to reference El Andalousi J, Li Y, Anand-Srivastava MB: Natriuretic peptide receptor-C agonist attenuates the expression of cell cycle proteins and proliferation of vascular smooth muscle cells from spontaneously hypertensive rats: role of Gi proteins and MAPkinase/PI3kinase signaling. PLoS One. 2013, 8 (10): e76183- 10.1371/journal.pone.0076183PubMedCentralCrossRefPubMed El Andalousi J, Li Y, Anand-Srivastava MB: Natriuretic peptide receptor-C agonist attenuates the expression of cell cycle proteins and proliferation of vascular smooth muscle cells from spontaneously hypertensive rats: role of Gi proteins and MAPkinase/PI3kinase signaling. PLoS One. 2013, 8 (10): e76183- 10.1371/journal.pone.0076183PubMedCentralCrossRefPubMed
23.
go back to reference Voss MH, Bastos DA, Karlo CA, Ajeti A, Hakimi AA, Feldman DR, Hsieh JJ, Molina AM, Patil S, Motzer RJ: Treatment outcome with mTOR inhibitors for metastatic renal cell carcinoma with nonclear and sarcomatoid histologies. Ann Oncol. 2014, 25 (3): 663-668. 10.1093/annonc/mdt578PubMedCentralCrossRefPubMed Voss MH, Bastos DA, Karlo CA, Ajeti A, Hakimi AA, Feldman DR, Hsieh JJ, Molina AM, Patil S, Motzer RJ: Treatment outcome with mTOR inhibitors for metastatic renal cell carcinoma with nonclear and sarcomatoid histologies. Ann Oncol. 2014, 25 (3): 663-668. 10.1093/annonc/mdt578PubMedCentralCrossRefPubMed
24.
go back to reference Arnedos M, Drury S, Afentakis M, A'hern R, Hills M, Salter J, Smith IE, Reis-Filho JS, Dowsett M: Biomarker changes associated with the development of resistance to aromatase inhibitors (AIs) in estrogen receptor-positive breast cancer. Ann Oncol. 2014, 25 (3): 605-610. 10.1093/annonc/mdt575PubMedCentralCrossRefPubMed Arnedos M, Drury S, Afentakis M, A'hern R, Hills M, Salter J, Smith IE, Reis-Filho JS, Dowsett M: Biomarker changes associated with the development of resistance to aromatase inhibitors (AIs) in estrogen receptor-positive breast cancer. Ann Oncol. 2014, 25 (3): 605-610. 10.1093/annonc/mdt575PubMedCentralCrossRefPubMed
25.
go back to reference Deb S, Xu H, Tuynman J, George J, Yan Y, Li J, Ward RL, Mortensen N, Hawkins NJ, McKay MJ, Ramsay RG, Fox SB: RAD21 cohesin overexpression is a prognostic and predictive marker exacerbating poor prognosis in KRAS mutant colorectal carcinomas. Br J Cancer. 2014, 110 (6): 1606-1613. 10.1038/bjc.2014.31PubMedCentralCrossRefPubMed Deb S, Xu H, Tuynman J, George J, Yan Y, Li J, Ward RL, Mortensen N, Hawkins NJ, McKay MJ, Ramsay RG, Fox SB: RAD21 cohesin overexpression is a prognostic and predictive marker exacerbating poor prognosis in KRAS mutant colorectal carcinomas. Br J Cancer. 2014, 110 (6): 1606-1613. 10.1038/bjc.2014.31PubMedCentralCrossRefPubMed
26.
go back to reference Jeong HM, Kwon MJ, Shin YK: Overexpression of Cancer-Associated Genes via Epigenetic Derepression Mechanisms in Gynecologic Cancer. Front Oncol. 2014, 4: 12-PubMedCentralCrossRefPubMed Jeong HM, Kwon MJ, Shin YK: Overexpression of Cancer-Associated Genes via Epigenetic Derepression Mechanisms in Gynecologic Cancer. Front Oncol. 2014, 4: 12-PubMedCentralCrossRefPubMed
27.
go back to reference Li J, Zhu W, Leng T, Shu M, Huang Y, Xu D, Qiu P, Su X, Yan G: Triptolide-induced cell cycle arrest and apoptosis in human renal cell carcinoma cells. Oncol Rep. 2011, 25 (4): 979-987.CrossRefPubMed Li J, Zhu W, Leng T, Shu M, Huang Y, Xu D, Qiu P, Su X, Yan G: Triptolide-induced cell cycle arrest and apoptosis in human renal cell carcinoma cells. Oncol Rep. 2011, 25 (4): 979-987.CrossRefPubMed
28.
go back to reference Klintman M, Strand C, Ahlin C, Beglerbegovic S, Fjällskog ML, Grabau D, Gudlaugsson E, Janssen EA, Lövgren K, Skaland I, Bendahl PO, Malmström P, Baak JP, Fernö M: The Prognostic Value of Mitotic Activity Index (MAI), Phosphohistone H3 (PPH3), Cyclin B1, Cyclin A, and Ki67, Alone and in Combinations, in Node-Negative Premenopausal Breast Cancer. PLoS One. 2013, 8 (12): e81902- 10.1371/journal.pone.0081902PubMedCentralCrossRefPubMed Klintman M, Strand C, Ahlin C, Beglerbegovic S, Fjällskog ML, Grabau D, Gudlaugsson E, Janssen EA, Lövgren K, Skaland I, Bendahl PO, Malmström P, Baak JP, Fernö M: The Prognostic Value of Mitotic Activity Index (MAI), Phosphohistone H3 (PPH3), Cyclin B1, Cyclin A, and Ki67, Alone and in Combinations, in Node-Negative Premenopausal Breast Cancer. PLoS One. 2013, 8 (12): e81902- 10.1371/journal.pone.0081902PubMedCentralCrossRefPubMed
29.
go back to reference Strand C, Ahlin C, Bendahl PO, Fjällskog ML, Hedenfalk I, Malmström P, Fernö M: Combination of the proliferation marker cyclin A, histological grade, and estrogen receptor status in a new variable with high prognostic impact in breast cancer. Breast Cancer Res Treat. 2012, 131 (1): 33-40. 10.1007/s10549-011-1386-5CrossRefPubMed Strand C, Ahlin C, Bendahl PO, Fjällskog ML, Hedenfalk I, Malmström P, Fernö M: Combination of the proliferation marker cyclin A, histological grade, and estrogen receptor status in a new variable with high prognostic impact in breast cancer. Breast Cancer Res Treat. 2012, 131 (1): 33-40. 10.1007/s10549-011-1386-5CrossRefPubMed
30.
go back to reference Ahlin C, Zhou W, Holmqvist M, Holmberg L, Nilsson C, Jirström K, Blomqvist C, Amini RM, Fjällskog ML: Cyclin A is a proliferative marker with good prognostic value in node-negative breast cancer. Cancer Epidemiol Biomarkers Prev. 2009, 18 (9): 2501-2506. 10.1158/1055-9965.EPI-09-0169CrossRefPubMed Ahlin C, Zhou W, Holmqvist M, Holmberg L, Nilsson C, Jirström K, Blomqvist C, Amini RM, Fjällskog ML: Cyclin A is a proliferative marker with good prognostic value in node-negative breast cancer. Cancer Epidemiol Biomarkers Prev. 2009, 18 (9): 2501-2506. 10.1158/1055-9965.EPI-09-0169CrossRefPubMed
31.
go back to reference Migita T, Oda Y, Naito S, Tsuneyoshi M: Low expression of p27(Kip1) is associated with tumor size and poor prognosis in patients with renal cell carcinoma. Cancer. 2002, 94 (4): 973-979. 10.1002/cncr.10338CrossRefPubMed Migita T, Oda Y, Naito S, Tsuneyoshi M: Low expression of p27(Kip1) is associated with tumor size and poor prognosis in patients with renal cell carcinoma. Cancer. 2002, 94 (4): 973-979. 10.1002/cncr.10338CrossRefPubMed
32.
go back to reference Blaheta RA, Michaelis M, Natsheh I, Hasenberg C, Weich E, Relja B, Jonas D, Doerr HW, Cinatl J: Valproic acid inhibits adhesion of vincristine- and cisplatin-resistant neuroblastoma tumour cells to endothelium. Br J Cancer. 2007, 96: 1699-1706. 10.1038/sj.bjc.6603777PubMedCentralCrossRefPubMed Blaheta RA, Michaelis M, Natsheh I, Hasenberg C, Weich E, Relja B, Jonas D, Doerr HW, Cinatl J: Valproic acid inhibits adhesion of vincristine- and cisplatin-resistant neuroblastoma tumour cells to endothelium. Br J Cancer. 2007, 96: 1699-1706. 10.1038/sj.bjc.6603777PubMedCentralCrossRefPubMed
33.
go back to reference Vallo S, Xi W, Hudak L, Juengel E, Tsaur I, Wiesner C, Haferkamp A, Blaheta RA: HDAC inhibition delays cell cycle progression of human bladder cancer cells in vitro. Anticancer Drugs. 2011, 22: 1002-1009.PubMed Vallo S, Xi W, Hudak L, Juengel E, Tsaur I, Wiesner C, Haferkamp A, Blaheta RA: HDAC inhibition delays cell cycle progression of human bladder cancer cells in vitro. Anticancer Drugs. 2011, 22: 1002-1009.PubMed
34.
go back to reference Wedel S, Hudak L, Seibel JM, Makarević J, Juengel E, Tsaur I, Waaga-Gasser A, Haferkamp A, Blaheta RA: Molecular targeting of prostate cancer cells by a triple drug combination down-regulates integrin driven adhesion processes, delays cell cycle progression and interferes with the cdk-cyclin axis. BMC Cancer. 2011, 11: 375- 10.1186/1471-2407-11-375PubMedCentralCrossRefPubMed Wedel S, Hudak L, Seibel JM, Makarević J, Juengel E, Tsaur I, Waaga-Gasser A, Haferkamp A, Blaheta RA: Molecular targeting of prostate cancer cells by a triple drug combination down-regulates integrin driven adhesion processes, delays cell cycle progression and interferes with the cdk-cyclin axis. BMC Cancer. 2011, 11: 375- 10.1186/1471-2407-11-375PubMedCentralCrossRefPubMed
35.
go back to reference Kumar P, Tripathi S, Pandey KN: Histone Deacetylase Inhibitors Modulate the Transcriptional Regulation of Guanylyl Cyclase/Natriuretic Peptide Receptor-A Gene: Interactive roles of Modified Histones, HATS, p300, and Sp1. J Biol Chem. in press, Kumar P, Tripathi S, Pandey KN: Histone Deacetylase Inhibitors Modulate the Transcriptional Regulation of Guanylyl Cyclase/Natriuretic Peptide Receptor-A Gene: Interactive roles of Modified Histones, HATS, p300, and Sp1. J Biol Chem. in press,
36.
go back to reference Jones J, Juengel E, Mickuckyte A, Hudak L, Wedel S, Jonas D, Blaheta RA: The histone deacetylase inhibitor valproic acid alters growth properties of renal cell carcinoma in vitro and in vivo. J Cell Mol Med. 2009, 13: 2376-2385. 10.1111/j.1582-4934.2008.00436.xCrossRefPubMed Jones J, Juengel E, Mickuckyte A, Hudak L, Wedel S, Jonas D, Blaheta RA: The histone deacetylase inhibitor valproic acid alters growth properties of renal cell carcinoma in vitro and in vivo. J Cell Mol Med. 2009, 13: 2376-2385. 10.1111/j.1582-4934.2008.00436.xCrossRefPubMed
Metadata
Title
HDAC-inhibition counteracts everolimus resistance in renal cell carcinoma in vitro by diminishing cdk2 and cyclin A
Authors
Eva Juengel
Snigdha Nowaz
Jasmina Makarevi
Iyad Natsheh
Isabella Werner
Karen Nelson
Michael Reiter
Igor Tsaur
Jens Mani
Sebastian Harder
Georg Bartsch
Axel Haferkamp
Roman A Blaheta
Publication date
01-12-2014
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2014
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/1476-4598-13-152

Other articles of this Issue 1/2014

Molecular Cancer 1/2014 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine