Enhanced Antitumor Activity with Combining Effect of mTOR Inhibition and Microtubule Stabilization in Hepatocellular Carcinoma

Qian Zhou, Chi Hang Wong, Cecilia Pik Yuk Lau, Connie Wun Chun Hui, Vivian Wai Yan Lui, Stephen Lam Chan, Winnie Yeo, Qian Zhou, Chi Hang Wong, Cecilia Pik Yuk Lau, Connie Wun Chun Hui, Vivian Wai Yan Lui, Stephen Lam Chan, Winnie Yeo

Abstract

Mammalian target of rapamycin (mTOR) and the microtubules are shown to be potential targets for treating hepatocellular carcinoma (HCC). PI3K/Akt/mTOR activation is associated with resistance to microtubule inhibitors. Here, we evaluated the antitumor activity by cotargeting of the mTOR (using allosteric mTOR inhibitor everolimus) and the microtubules (using novel microtubule-stabilizing agent patupilone) in HCC models. In vitro studies showed that either targeting mTOR signaling with everolimus or targeting microtubules with patupilone was able to suppress HCC cell growth in a dose-dependent manner. Cotargeting of the mTOR (by everolimus) and the microtubules (by patupilone, at low nM) resulted in enhanced growth inhibition in HCC cells (achieving maximal growth inhibition of 60-87%), demonstrating potent antitumor activity of this combination. In vivo studies showed that everolimus treatment alone for two weeks was able to inhibit the growth of Hep3B xenografts. Strikingly, the everolimus/patupilone combination induced a more significant antitumor activity. Mechanistic study demonstrated that this enhanced antitumor effect was accompanied by marked cell apoptosis induction and antiangiogenic activity, which were more significant than single-agent treatments. Our findings demonstrated that the everolimus/patupilone combination, which had potent antitumor activity, was a potential therapeutic strategy for HCC.

Figures

Figure 1
Figure 1
Everolimus inhibited proliferation and mTOR signaling in HCC cell lines. (a) Dose-dependent inhibition of HCC cell proliferation by everolimus. The effect of everolimus on cell viability was assessed by MTT assay. Dose-response curves of everolimus for all HCC cell lines were shown. Similar results were observed in 3 independent experiments. (b) Average IC50 values of everolimus in HCC cell lines. Cumulative results from 3 independent experiments were shown as mean ± SEM. (c) Everolimus inhibited the mTOR pathway in HCC cells. HepG2, Hep3B, and SNU398 cells (3 × 105) were treated with 0.1 μM everolimus (hereafter labeled as Eve) or DMSO control for 48 hrs and 72 hrs. The expression levels of the mTOR pathway components, pi-mTOR (ser2448), mTOR, pi-p70S6K (Thr389), p70S6K, pi-S6 (ser240/244), S6, pi-4E-BP1 (ser65), and 4E-BP1, and actin were examined by western blotting. Similar results were observed in 3 independent experiments.
Figure 2
Figure 2
Patupilone inhibited proliferation in HCC cell lines. (a) Dose-dependent inhibition of HCC cell proliferation by patupilone. The effect of patupilone on cell viability was assessed by MTT assay. Dose-response curves of everolimus for all HCC cell lines were shown. Similar results were observed in 3 independent experiments. (b) Average IC50 values of patupilone in HCC cell lines. Cumulative results from 3 independent experiments were shown as mean ± SEM.
Figure 3
Figure 3
Enhanced antitumor activity of the everolimus/patupilone combination in HCC cell lines. (a) Effects of everolimus/patupilone in HCC cell lines. HepG2, Hep3B, and SNU398 cells (1 × 104) were treated with various concentrations of everolimus in combination with 0.5 nM patupilone (Pat) for 24 hrs. Cell viability was assessed by MTT assay. Cumulative results from 3 independent experiments were shown as mean ± SEM (*P < 0.05, **P < 0.01, ***P < 0.001 versus everolimus-treated group). (b) The mTOR signaling in HCC cells was not further suppressed by the everolimus/patupilone combination treatment. HepG2, Hep3B, and SNU398 cells (3 × 105) were treated with everolimus (0.1 μM) and/or patupilone (Pat) (0.5 nM) for 24 hrs. The everolimus/patupilone combination is abbreviated as Eve/Pat hereafter. The expression levels of the mTOR pathway components, pi-mTOR (ser2448), mTOR, pi-p70S6K (Thr389), p70S6K, pi-S6 (ser240/244), S6, pi-4E-BP1 (ser65), and 4E-BP1, and actin were examined by western blotting. Similar results were observed in 3 independent experiments.
Figure 4
Figure 4
Potent antitumor effects of the everolimus/patupilone combination in in vivo models of HCC. Hep3B cells (3 × 106 cells) were inoculated into nude mice by subcutaneous injection. Drug treatments were started on day 20 after inoculation. Mice received administration of drugs for two weeks (black arrow: patupilone i.p. injection; red arrow: everolimus orally given). (a) Treatment of mice with everolimus, patupilone, or combination suppressed tumor growth in established xenografts of Hep3B. Tumor growth was monitored twice weekly. Arrows indicated time of drug administration. The in vivo antitumor activity of everolimus/patupilone combination was more significant than either agent alone (n = 10 per group, *P < 0.05, **P < 0.01, ***P < 0.001 versus vehicle group). (b) Tumor weight of Hep3B xenografts in each group. The tumor weight of everolimus/patupilone combination group was significantly reduced (n = 10 per group, **P < 0.01 versus vehicle group). (c) The mTOR signaling in HCC cells was not further suppressed by the everolimus/patupilone combination treatment in Hep3B xenograft. Tumor xenografts were harvested, fixed, and stained for pi-mTOR and pi-S6 by immunohistochemistry. Representative images (400x magnification) were shown. Quantitation of pi-mTOR and pi-S6 staining using immunohistochemistry scoring was shown in (B) (n = 10 per group, *P < 0.05, **P < 0.01, ***P < 0.001 versus vehicle group).
Figure 5
Figure 5
Everolimus/patupilone combination induced cell apoptosis and exerted antiangiogenic effect in HCC models. Tumor xenografts were harvested, fixed, and stained for cleaved PARP and CD34 by immunohistochemistry. Representative images (400x magnification) were shown. Quantitation of cleaved PARP and CD34 (microvessel density, MVD) staining was shown in (b) (n = 10 per group, *P < 0.05, **P < 0.01, ***P < 0.001 versus vehicle group).

References

    1. He J, Gu D, Wu X, et al. Major causes of death among men and women in China. New England Journal of Medicine. 2005;353(11):1124–1134.
    1. Parkin DM. Global cancer statistics in the year 2000. Lancet Oncology. 2001;2(9):533–543.
    1. Schwartz M. Liver transplantation for hepatocellular carcinoma. Gastroenterology. 2004;127(5, supplement 1):S268–S276.
    1. Poon RTP, Fan ST, Lo CM, et al. Improving survival results after resection of hepatocellular carcinoma: a prospective study of 377 patients over 10 years. Annals of Surgery. 2001;234(1):63–70.
    1. Cheng AL, Kang YK, Chen Z, et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. The Lancet Oncology. 2009;10(1):25–34.
    1. Llovet JM, Ricci S, Mazzaferro V, et al. Sorafenib in advanced hepatocellular carcinoma. New England Journal of Medicine. 2008;359(4):378–390.
    1. Bjornsti MA, Houghton PJ. The TOR pathway: a target for cancer therapy. Nature Reviews Cancer. 2004;4(5):335–348.
    1. Agarwala SS, Case S. Everolimus (RAD001) in the treatment of advanced renal cell carcinoma: a review. Oncologist. 2010;15(3):236–245.
    1. Yim KL. Everolimus and mTOR inhibition in pancreatic neuroendocrine tumors. Cancer Management and Research. 2012;4:207–214.
    1. Baselga J, Campone M, Piccart M, et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. New England Journal of Medicine. 2012;366(6):520–529.
    1. Curran MP. Everolimus: in patients with subependymal giant cell astrocytoma associated with tuberous sclerosis complex. Pediatric Drugs. 2012;14(1):51–60.
    1. Blaszkowsky LS, Abrams TA, Miksad RA, et al. Phase I/II study of everolimus in patients with advanced hepatocellular carcinoma (HCC) Journal of Clinical Oncology. 2010;(ASCO Annual Meeting)e14542
    1. Mabuchi S, Altomare DA, Connolly DC, et al. RAD001 (Everolimus) delays tumor onset and progression in a transgenic mouse model of ovarian cancer. Cancer Research. 2007;67(6):2408–2413.
    1. Wu C, Wangpaichitr M, Feun L, et al. Overcoming cisplatin resistance by mTOR inhibitor in lung cancer. Molecular Cancer. 2005;4, article 25
    1. Patil MA, Chua MS, Pan KH, et al. An integrated data analysis approach to characterize genes highly expressed in hepatocellular carcinoma. Oncogene. 2005;24(23):3737–3747.
    1. Tung CY, Jen CH, Hsu MT, Wang HW, Lin CH. A novel regulatory event-based gene set analysis method for exploring global functional changes in heterogeneous genomic data sets. BMC Genomics. 2009;10, article 26
    1. Zhou Q, Ching AKK, Leung WKC, et al. Novel therapeutic potential in targeting microtubules by nanoparticle albumin-bound paclitaxel in hepatocellular carcinoma. International Journal of Oncology. 2011;38(3):721–731.
    1. Faried LS, Faried A, Kanuma T, et al. Inhibition of the mammalian target of rapamycin (mTOR) by rapamycin increases chemosensitivity of CaSki cells to paclitaxel. European Journal of Cancer. 2006;42(7):934–947.
    1. VanderWeele DJ, Zhou R, Rudin CM. Akt up-regulation increases resistance to microtubule-directed chemotherapeutic agents through mammalian target of rapamycin. Molecular Cancer Therapeutics. 2004;3(12):1605–1613.
    1. Zhou Q, Lui VW, Lau CP, et al. Sustained antitumor activity by co-targeting mTOR and the microtubule with temsirolimus/vinblastine combination in hepatocellular carcinoma. Biochemical Pharmacology. 2012;83(9):1146–1158.
    1. Nettles JH, Li H, Cornett B, Krahn JM, Snyder JP, Downing KH. The binding mode of epothilone A on α,β-tubulin by electron crystallography. Science. 2004;305(5685):866–869.
    1. Altmann KH, Bold G, Caravatti G, et al. Epothilones and their analogs—potential new weapons in the fight against cancer. Chimia. 2000;54(11):612–621.
    1. Kowalski RJ, Giannakakou P, Hamel E. Activities of the microtubule-stabilizing agents epothilones A and B with purified tubulin and in cells resistant to paclitaxel (Taxol) Journal of Biological Chemistry. 1997;272(4):2534–2541.
    1. Mok TSK, Choi E, Yau D, et al. Effects of patupilone (epothilone B; EPO906), a novel chemotherapeutic agent, in hepatocellular carcinoma: an in vitro study. Oncology. 2007;71(3-4):292–296.
    1. Rubin EH, Rothermel J, Tesfaye F, et al. Phase I dose-finding study of weekly single-agent patupilone in patients with advanced solid tumors. Journal of Clinical Oncology. 2005;23(36):9120–9129.
    1. Osterlind K, Sanchez JM, Zatloukal P, et al. Phase I/II dose escalation trial of patupilone every 3 weeks in patients with non-small cell lung cancer. Journal of Clinical Oncology. 2005;23(16):p. 647S.
    1. Smit WM, Sufliarsky J, Spanik S, et al. Phase I/II dose-escalation trial of patupilone every 3 weeks in patients with resistant/refractory ovarian cancer. European Journal of Cancer. 2005;3(2):261–262.
    1. Lui VWY, Boehm AL, Koppikar P, et al. Antiproliferative mechanisms of a transcription factor decoy targeting signal transducer and activator of transcription (STAT) 3: the role of STAT1. Molecular Pharmacology. 2007;71(5):1435–1443.
    1. Lui VWY, Yau DM, Wong EY, et al. Cucurbitacin I elicits anoikis sensitization, inhibits cellular invasion and in vivo tumor formation ability of nasopharyngeal carcinoma cells. Carcinogenesis. 2009;30(12):2085–2094.
    1. Lui VWY, Yau DMS, Cheung CSF, et al. FGF8b oncogene mediates proliferation and invasion of Epstein-Barr virus-associated nasopharyngeal carcinoma cells: implication for viral-mediated FGF8b upregulation. Oncogene. 2011;30(13):1518–1530.
    1. Hu L, Hofmann J, Lu Y, Mills GB, Jaffe RB. Inhibition of phosphatidylinositol 3′-kinase increases efficacy of paclitaxel in in vitro and in vivo ovarian cancer models. Cancer Research. 2002;62(4):1087–1092.
    1. Chou TC, Zhang XG, Balog A, et al. Desoxyepothilone B: an efficacious microtubule-targeted antitumor agent with a promising in vivo profile relative to epothilone B. Proceedings of the National Academy of Sciences of the United States of America. 1998;95(16):9642–9647.
    1. Tam KH, Yang ZF, Lau CK, Lam CT, Pang RWC, Poon RTP. Inhibition of mTOR enhances chemosensitivity in hepatocellular carcinoma. Cancer Letters. 2009;273(2):201–209.
    1. Wang L, Shi WY, Wu ZY, et al. Cytostatic and anti-angiogenic effects of temsirolimus in refractory mantle cell lymphoma. Journal of Hematology & Oncology. 2010;3, article 30
    1. Mok TSK, Choi E, Yau D, et al. Effects of patupilone (epothilone B; EPO906), a novel chemotherapeutic agent, in hepatocellular carcinoma: an in vitro study. Oncology. 2007;71(3-4):292–296.
    1. Marimpietri D, Brignole C, Nico B, et al. Combined therapeutic effects of vinblastine and rapamycin on human neuroblastoma growth, apoptosis, and angiogenesis. Clinical Cancer Research. 2007;13(13):3977–3988.
    1. Campostrini N, Marimpietri D, Totolo A, et al. Proteomic analysis of anti-angiogenic effects by a combined treatment with vinblastine and rapamycin in an endothelial cell line. Proteomics. 2006;6(15):4420–4431.
    1. Zhu AX, Abrams TA, Miksad R, et al. Phase 1/2 study of everolimus in advanced hepatocellular carcinoma. Cancer. 2011;117(22):5094–5102.
    1. Venook AP, Poon R, Kang YK, et al. Evaluation of patupilone as monotherapy in patients with advanced hepatocellular carcinoma (HCC) Journal of Clinical Oncology. 2007;25(ASCO Annual Meeting Proceedings)(18S)15055

Source: PubMed

3
Tilaa