Skip to main content
Log in

Aurora kinase inhibitors attached to iron oxide nanoparticles enhances inhibition of the growth of liver cancer cells

  • Research Paper
  • Published:
Journal of Nanoparticle Research Aims and scope Submit manuscript

Abstract

We have developed a novel Aurora kinase inhibitor (AKI) AM-005, an analogue of pan-AKI AT-9283. To improve the intracellular efficacy of AM-005 and AT-9283, we utilized magnetite nanoparticles (NPs) to deliver AM-005 and AT-9283 into human SMMC-7721 and HepG2 liver cancer cells. The drug-loaded NPs were prepared through quasi-emulsion solvent diffusion of magnetite NPs with AM-005 or AT-9283. The encapsulated drugs were readily released from NPs, preferentially at low pHs. Upon exposure, cancer cells effectively internalized drug-loaded NPs into lysosome-like vesicles, which triggered a series of cellular changes, including the formation of enlarged cytoplasm, the significant increase of membrane permeability, and the generation of reactive oxygen species (ROS). The increased ROS synthesis sustained over 72 h, whereas that in the cells treated with free-form drugs declined rapidly after 48 h. However, chemical sequestration of the iron core of NPs had a minor influence on the generation of intracellular ROS. On the other hand, uncoupling of AM-005 uptake with NP internalization into cells failed to induce ROS synthesis. Overall, our approach achieved two-fold increase in suppressing the viability of tumor cells in vitro and the growth of tumors in vivo. We conclude that magnetite NPs can be used as pH responsive nanocarriers that are able to improve the efficacy of AKIs.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  • Arbab AS, Wilson LB, Ashari P, Jordan EK, Lewis BK, Frank JA (2005) A model of lysosomal metabolism of dextran coated superparamagnetic iron oxide (SPIO) nanoparticles: implications for cellular magnetic resonance imaging. NMR Biomed 18:383–389

    Article  Google Scholar 

  • Bao G, Mitragotri S, Tong S (2013) Multifunctional nanoparticles for drug delivery and molecular imaging. Annu Rev Biomed Eng 15:253–282. doi:10.1146/annurev-bioeng-071812-152409

    Article  Google Scholar 

  • Batrakova EV, Gendelman HE, Kabanov AV (2011) Cell-mediated drug delivery. Expert Opin Drug Deliv 8:415–433

    Article  Google Scholar 

  • Beck-Broichsitter M et al (2010) Novel ‘Nano in Nano’Composites for sustained drug delivery: biodegradable nanoparticles encapsulated into nanofiber non-wovens. Macromol Biosci 10:1527–1535

    Article  Google Scholar 

  • Berry CC (2009) Progress in functionalization of magnetic nanoparticles for applications in biomedicine. J Phys D Appl Phys 42:224003

    Article  Google Scholar 

  • Carvajal RD, Tse A, Schwartz GK (2006) Aurora kinases: new targets for cancer therapy. Clin Cancer Res 12:6869–6875

    Article  Google Scholar 

  • Castino R, Fiorentino I, Cagnin M, Giovia A, Isidoro C (2011) Chelation of lysosomal iron protects dopaminergic SH-SY5Y neuroblastoma cells from hydrogen peroxide toxicity by precluding autophagy and Akt dephosphorylation. Toxicol Sci 123:523–541

    Article  Google Scholar 

  • Chen Z et al (2012) Dual enzyme-like activities of iron oxide nanoparticles and their implication for diminishing cytotoxicity. ACS Nano 6:4001–4012

    Article  Google Scholar 

  • Dar AA, Goff LW, Majid S, Berlin J, El-Rifai W (2010) Aurora kinase inhibitors-rising stars in cancer therapeutics? Mol Cancer Ther 9:268–278

    Article  Google Scholar 

  • Dennis M, Davies M, Oliver S, D’Souza R, Pike L, Stockman P (2012) Phase I study of the Aurora B kinase inhibitor barasertib (AZD1152) to assess the pharmacokinetics, metabolism and excretion in patients with acute myeloid leukemia. Cancer Chemother Pharmacol 70:461–469. doi:10.1007/s00280-012-1939-2

    Article  Google Scholar 

  • El-Serag HB, Mason AC (1999) Rising incidence of hepatocellular carcinoma in the United States. N Engl J Med 340:745–750

    Article  Google Scholar 

  • Fu J, Bian M, Jiang Q, Zhang C (2007) Roles of Aurora kinases in mitosis and tumorigenesis. Mol Cancer Res 5:1–10

    Article  Google Scholar 

  • Gadea BB, Ruderman JV (2005) Aurora kinase inhibitor ZM447439 blocks chromosome-induced spindle assembly, the completion of chromosome condensation, and the establishment of the spindle integrity checkpoint in Xenopus egg extracts. Mol Biol Cell 16:1305–1318

    Article  Google Scholar 

  • Gautier J et al (2012) A pharmaceutical study of doxorubicin-loaded PEGylated nanoparticles for magnetic drug targeting. Int J Pharm 423:16–25

    Article  Google Scholar 

  • Gerweck LE, Vijayappa S, Kozin S (2006) Tumor pH controls the in vivo efficacy of weak acid and base chemotherapeutics. Mol Cancer Ther 5:1275–1279

    Article  Google Scholar 

  • Guo M, Yan Y, Liu X, Yan H, Liu K, Zhang H, Cao Y (2010) Multilayer nanoparticles with a magnetite core and a polycation inner shell as pH-responsive carriers for drug delivery. Nanoscale 2:434–441

    Article  Google Scholar 

  • Gurtler U, Tontsch-Grunt U, Jarvis M, Zahn S, Boehmelt G, Adolf G, Solca F (2010) Effect of BI 811283, a novel inhibitor of Aurora B kinase, on tumor senescence and apoptosis. J Clin Oncol 28:e13632

    Google Scholar 

  • Hamacher-Brady A et al (2011) Artesunate activates mitochondrial apoptosis in breast cancer cells via iron-catalyzed lysosomal reactive oxygen species production. J Biol Chem 286:6587–6601

    Article  Google Scholar 

  • Hetland TE et al (2013) Aurora B expression in metastatic effusions from advanced-stage ovarian serous carcinoma is predictive of intrinsic chemotherapy resistance. Hum Pathol 44:777–785. doi:10.1016/j.humpath.2012.08.002

    Article  Google Scholar 

  • Hillaireau H, Couvreur P (2009) Nanocarriers’ entry into the cell: relevance to drug delivery. Cell Mol Life Sci 66:2873–2896. doi:10.1007/s00018-009-0053-z

    Article  Google Scholar 

  • Hosseinkhani H, Tabata Y (2006) Self assembly of DNA nanoparticles with polycations for the delivery of genetic materials into cells. J Nanosci Nanotechnol 6:2320–2328

    Article  Google Scholar 

  • Huck JJ et al (2010) MLN8054, an inhibitor of Aurora A kinase induces senescence in human tumor cells both in vitro and in vivo. Mol Cancer Res 8:373–384

    Article  Google Scholar 

  • Ikeda K et al (1993) A multivariate analysis of risk factors for hepatocellular carcinogenesis: a prospective observation of 795 patients with viral and alcoholic cirrhosis. Hepatology 18:47–53

    Article  Google Scholar 

  • Kantarjian HM et al (2013) Stage I of a phase 2 study assessing the efficacy, safety, and tolerability of barasertib (AZD1152) versus low-dose cytosine arabinoside in elderly patients with acute myeloid leukemia. Cancer 119:2611–2619. doi:10.1002/cncr.28113

    Article  Google Scholar 

  • Kievit FM et al (2011) Doxorubicin loaded iron oxide nanoparticles overcome multidrug resistance in cancer in vitro. J Control Release 152:76–83

    Article  Google Scholar 

  • Kim H-J, Cho JH, Quan H, Kim J-R (2011) Down-regulation of Aurora B kinase induces cellular senescence in human fibroblasts and endothelial cells through a p53-dependent pathway. FEBS Lett 585:3569–3576

    Article  Google Scholar 

  • Kimura S (2010) AT-9283, a small-molecule multi-targeted kinase inhibitor for the potential treatment of cancer. Curr Opin Investig Drugs 11:1442–1449 (London, England: 2000)

    Google Scholar 

  • Kohler N, Sun C, Wang J, Zhang M (2005) Methotrexate-modified superparamagnetic nanoparticles and their intracellular uptake into human cancer cells. Langmuir 21:8858–8864

    Article  Google Scholar 

  • Gerweck LE (1998) Tumor pH: implications for treatment and novel drug design. Semin Radiat Oncol 3:176–182 Elsevier

    Article  Google Scholar 

  • Li J-j et al (2006) Role of oxidative stress in the apoptosis of hepatocellular carcinoma induced by combination of arsenic trioxide and ascorbic acid. Acta Pharmacol Sin 27:1078–1084

    Article  Google Scholar 

  • Lin Z-Z et al (2009) The Aurora kinase inhibitor VE-465 has anticancer effects in pre-clinical studies of human hepatocellular carcinoma. J Hepatol 50:518–527

    Article  Google Scholar 

  • Lipp JJ, Hirota T, Poser I, Peters J-M (2007) Aurora B controls the association of condensin I but not condensin II with mitotic chromosomes. J Cell Sci 120:1245–1255

    Article  Google Scholar 

  • Liu Q, Ruderman JV (2006) Aurora A, mitotic entry and spindle bipolarity. Proc Natl Acad Sci USA 103:5811–5816

    Article  Google Scholar 

  • Lunov O et al (2010) The effect of carboxydextran-coated superparamagnetic iron oxide nanoparticles on c-Jun N-terminal kinase-mediated apoptosis in human macrophages. Biomaterials 31:5063–5071

    Article  Google Scholar 

  • Maeng JH et al (2010) Multifunctional doxorubicin loaded superparamagnetic iron oxide nanoparticles for chemotherapy and magnetic resonance imaging in liver cancer. Biomaterials 31:4995–5006

    Article  Google Scholar 

  • Malumbres M, Perez de Castro I (2014) Aurora kinase A inhibitors: promising agents in antitumoral therapy. Expert Opin Ther Targets 9:1–17. doi:10.1517/14728222.2014.956085

    Google Scholar 

  • Marumoto T, Zhang D, Saya H (2005) Aurora-A—a guardian of poles. Nat Rev Cancer 5:42–50

    Article  Google Scholar 

  • Mazak K, Noszal B (2014) Drug delivery: a process governed by species-specific lipophilicities. Eur J Pharm Sci 62:96–104. doi:10.1016/j.ejps.2014.05.017

    Article  Google Scholar 

  • Noh Y-W, Jang Y-S, Ahn K-J, Lim YT, Chung BH (2011) Simultaneous in vivo tracking of dendritic cells and priming of an antigen-specific immune response. Biomaterials 32:6254–6263

    Google Scholar 

  • Palombo M, Deshmukh M, Myers D, Gao J, Szekely Z, Sinko PJ (2014) Pharmaceutical and toxicological properties of engineered nanomaterials for drug delivery. Annu Rev Pharm Toxicol 54:581–598. doi:10.1146/annurev-pharmtox-010611-134615

    Article  Google Scholar 

  • Pankhurst Q, Thanh N, Jones S, Dobson J (2009) Progress in applications of magnetic nanoparticles in biomedicine. J Phys D Appl Phys 42:224001

    Article  Google Scholar 

  • Qi W, Liu X, Cooke LS, Persky DO, Miller TP, Squires M, Mahadevan D (2012) AT9283, a novel aurora kinase inhibitor suppresses tumor growth in aggressive B-cell lymphomas. Int J Cancer 130:2997–3005. doi:10.1002/ijc.26324

    Article  Google Scholar 

  • Rabanel JM, Aoun V, Elkin I, Mokhtar M, Hildgen P (2012) Drug-loaded nanocarriers: passive targeting and crossing of biological barriers. Curr Med Chem 19:3070–3102

    Article  Google Scholar 

  • Sasai K et al (2004) Aurora-C kinase is a novel chromosomal passenger protein that can complement Aurora-B kinase function in mitotic cells. Cell Motil Cytoskelet 59:249–263

    Article  Google Scholar 

  • Shan XH et al (2012) Targeting Glut1-overexpressing MDA-MB-231 cells with 2-deoxy-d-glucose modified SPIOs. Eur J Radiol 81:95–99

    Article  Google Scholar 

  • Soenen SJ, De Cuyper M (2010) Assessing iron oxide nanoparticle toxicity in vitro: current status and future prospects. Nanomedicine 5:1261–1275

    Article  Google Scholar 

  • Soenen SJ, Himmelreich U, Nuytten N, De Cuyper M (2011) Cytotoxic effects of iron oxide nanoparticles and implications for safety in cell labelling. Biomaterials 32:195–205

    Article  Google Scholar 

  • Stohs S, Bagchi D (1995) Oxidative mechanisms in the toxicity of metal ions. Free Radic Biol Med 18:321–336

    Article  Google Scholar 

  • Subarsky P, Hill RP (2003) The hypoxic tumour microenvironment and metastatic progression. Clin Exp Metastasis 20:237–250

    Article  Google Scholar 

  • Torti S, Torti F, Whitman S, Brechbiel M, Park G, Planalp R (1998) Tumor cell cytotoxicity of a novel metal chelator. Blood 92:1384–1389

    Google Scholar 

  • Vader G, Lens S (2008) The Aurora kinase family in cell division and cancer. Biochim Biophys Acta-Rev Cancer 1786:60–72

    Article  Google Scholar 

  • Vaupel P (2004) Tumor microenvironmental physiology and its implications for radiation oncology. Semin Radiat Oncol 3:198–206 (Elsevier)

    Article  Google Scholar 

  • Vogt E, Kipp A, Eichenlaub-Ritter U (2009) Aurora kinase B epigenetic state of centromeric heterochromatin and chiasma resolution in oocytes. Reprod Biomed Online 19:352–368

    Article  Google Scholar 

  • Voinov MA, Pagán JOS, Morrison E, Smirnova TI, Smirnov AI (2010) Surface-mediated production of hydroxyl radicals as a mechanism of iron oxide nanoparticle biotoxicity. J Am Chem Soc 133:35–41

    Article  Google Scholar 

  • Wang CY, Hong JM, Chen G, Zhang Y, Gu N (2010) Facile method to synthesize oleic acid-capped magnetite nanoparticles. Chin Chem Lett 21:179–182

    Article  Google Scholar 

  • Wang FS, Fan JG, Zhang Z, Gao B, Wang HY (2014) The global burden of liver disease: the major impact of China. Hepatology. doi:10.1002/hep.27406

  • Wilhelm C, Gazeau F (2008) Universal cell labelling with anionic magnetic nanoparticles. Biomaterials 29:3161–3174

    Article  Google Scholar 

  • Yu MK et al (2008) Drug-loaded superparamagnetic iron oxide nanoparticles for combined cancer imaging and therapy in vivo. Angew Chem Int Ed 47:5362–5365

    Article  Google Scholar 

  • Zheng M, Zheng Y, Xie L, Chang W, Gu N, Ji M (2013a) Orally active Aurora A/B kinase inhibitor, AM-005 suppresses the growth of human colon carcinoma cells. Drug Dev Res 74:272–281. doi:10.1002/ddr.21077

    Article  Google Scholar 

  • Zheng M et al (2013b) Synthesis and quantum chemical studies of new 4-aminoquinazoline derivatives as Aurora A/B kinase inhibitors. Chem Biol Drug Des 81:399–407. doi:10.1111/cbdd.12089

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Min Ji, Ning Gu or Xi Zhan.

Electronic supplementary material

Below is the link to the electronic supplementary material.

11051_2014_2708_MOESM1_ESM.ppt

S-Fig 1. Drug loaded NPs induced apoptosis. (A)HepG2 and SMMC-7721 cells were treated with NPs, AM-005 and AM-NPs, respectively, for 4 days, and stained with annexin V-FITC/PI. The stained cells were analyzed by flow cytometry. Quantification of the data is presented on the right. (B) HepG2 cells were treated as above except that AT-9283 and AT-NPs were used. (C) HepG2 cells were treated with the reagents as indicated for 4 days, and analyzed by Western blot for the expression of α-tubulin and caspase-3. The intensity of each band was converted to digital data and quantified by ImageJ. The value of caspase-3 was normalized to that of corresponding α-tubulin. S-Fig 2. AM-NPs promote LDH release. HepG2 (A) and SMMC-7721 (B) cells were treated with AM-005 and AM-NPs for 48 h at indicated concentrations. The amount of LDH present in the media was measured as described in Materials and Methods. S-Fig 3. AT-NPs increase ROS generation. HepG2 cells were treated with NPs, AT-9283 and AT-NPs, respectively, at either 250 ng/mL (A) or 500 ng/mL (B) for 24h, 48h and 72h as indicated. All the treated cells were subjected to ROS synthesis analysis as described in the legend of Fig 6. The data were presented as the mean ± SEM (n=3) of ROS levels as defined by DCF. **, p < 0.005; and ***; p < 0.0005 (t-test), referring to the differences as indicated. S-Fig 4. Analysis of ROS in tumors. HepG2 tumors were excised at different days after treatment with saline, NPs, AT-9283, and AT-NPs (A) or saline, NPs, AM-005 and AM-NPs (B). Isolated tumor cells were then subjected to ROS analysis by measuring the conversion of H2DCF-DA into DCF as described in the Materials and Methods. S-Fig 5. DSL analysis of stored AM-NPs and NPs. AM-NPs and NPs were prepared and stored in water at 4°C. After 4 months, they were re-analyzed by DLS. Supplementary material 1 (PPT 1796 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, X., Xie, L., Zheng, M. et al. Aurora kinase inhibitors attached to iron oxide nanoparticles enhances inhibition of the growth of liver cancer cells. J Nanopart Res 17, 247 (2015). https://doi.org/10.1007/s11051-014-2708-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s11051-014-2708-4

Keywords

Navigation