Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Iron oxide nanoparticles inhibit tumour growth by inducing pro-inflammatory macrophage polarization in tumour tissues

Abstract

Until now, the Food and Drug Administration (FDA)-approved iron supplement ferumoxytol and other iron oxide nanoparticles have been used for treating iron deficiency, as contrast agents for magnetic resonance imaging and as drug carriers. Here, we show an intrinsic therapeutic effect of ferumoxytol on the growth of early mammary cancers, and lung cancer metastases in liver and lungs. In vitro, adenocarcinoma cells co-incubated with ferumoxytol and macrophages showed increased caspase-3 activity. Macrophages exposed to ferumoxytol displayed increased mRNA associated with pro-inflammatory Th1-type responses. In vivo, ferumoxytol significantly inhibited growth of subcutaneous adenocarcinomas in mice. In addition, intravenous ferumoxytol treatment before intravenous tumour cell challenge prevented development of liver metastasis. Fluorescence-activated cell sorting (FACS) and histopathology studies showed that the observed tumour growth inhibition was accompanied by increased presence of pro-inflammatory M1 macrophages in the tumour tissues. Our results suggest that ferumoxytol could be applied ‘off label’ to protect the liver from metastatic seeds and potentiate macrophage-modulating cancer immunotherapies.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Combining ferumoxytol and macrophages leads to cancer cell apoptosis through the Fenton reaction.
Figure 2: Iron oxide nanoparticles inhibit tumour growth.
Figure 3: Ferumoxytol causes in vivo M1 macrophage polarization.
Figure 4: Systemic delivery of ferumoxytol inhibits liver and lung metastases.
Figure 5: Pretreatment with ferumoxytol inhibits development of liver metastases.
Figure 6: Ferumoxytol alters macrophage polarization in hepatic metastasis in vivo.

Similar content being viewed by others

References

  1. Corot, C., Robert, P., Idee, J. M. & Port, M. Recent advances in iron oxide nanocrystal technology for medical imaging. Adv. Drug Deliv. Rev. 58, 1471–1504 (2006).

    Article  CAS  Google Scholar 

  2. Daldrup-Link, H. E. et al. MRI of tumor-associated macrophages with clinically applicable iron oxide nanoparticles. Clin. Cancer Res. 17, 5695–5704 (2011).

    Article  CAS  Google Scholar 

  3. Klenk, C. et al. Ionising radiation-free whole-body MRI versus 18F-fluorodeoxyglucose PET/CT scans for children and young adults with cancer: a prospective, non-randomised, single-centre study. Lancet Oncol. 15, 275–285 (2014).

    Article  Google Scholar 

  4. Ansari, C. et al. Development of novel tumor-targeted theranostic nanoparticles activated by membrane-type matrix metalloproteinases for combined cancer magnetic resonance imaging and therapy. Small 10, 566–575 (2014).

    Article  CAS  Google Scholar 

  5. Neuwelt, E. A. et al. The potential of ferumoxytol nanoparticle magnetic resonance imaging, perfusion, and angiography in central nervous system malignancy: a pilot study. Neurosurgery 60, 601–611 (2007).

    Article  Google Scholar 

  6. Daldrup-Link, H. & Coussens, L. M. MR imaging of tumor-associated macrophages. Oncoimmunology 1, 507–509 (2012).

    Article  Google Scholar 

  7. Vinogradov, S., Warren, G. & Wei, X. Macrophages associated with tumors as potential targets and therapeutic intermediates. Nanomedicine 9, 695–707 (2014).

    Article  CAS  Google Scholar 

  8. Miller, M. A. et al. Tumour-associated macrophages act as a slow-release reservoir of nano-therapeutic Pt(IV) pro-drug. Nat. Commun. 6, 8692 (2015).

    Article  CAS  Google Scholar 

  9. Weissleder, R., Nahrendorf, M. & Pittet, M. J. Imaging macrophages with nanoparticles. Nat. Mater. 13, 125–138 (2014).

    Article  CAS  Google Scholar 

  10. Li, C. A targeted approach to cancer imaging and therapy. Nat. Mater. 13, 110–115 (2014).

    Article  CAS  Google Scholar 

  11. Futterer, S., Andrusenko, I., Kolb, U., Hofmeister, W. & Langguth, P. Structural characterization of iron oxide/hydroxide nanoparticles in nine different parenteral drugs for the treatment of iron deficiency anaemia by electron diffraction (ED) and X-ray powder diffraction (XRPD). J. Pharm. Biomed. Anal. 86, 151–160 (2013).

    Article  CAS  Google Scholar 

  12. Ittrich, H., Peldschus, K., Raabe, N., Kaul, M. & Adam, G. Superparamagnetic iron oxide nanoparticles in biomedicine: applications and developments in diagnostics and therapy. RoFo 185, 1149–1166 (2013).

    Article  CAS  Google Scholar 

  13. Richards, J. M. et al. In vivo mononuclear cell tracking using superparamagnetic particles of iron oxide: feasibility and safety in humans. Circ. Cardiovasc. Imaging 5, 509–517 (2012).

    Article  Google Scholar 

  14. Warheit, D. B. & Hartsky, M. A. Role of alveolar macrophage chemotaxis and phagocytosis in pulmonary clearance responses to inhaled particles: comparisons among rodent species. Microsc. Res. Tech 26, 412–422 (1993).

    Article  CAS  Google Scholar 

  15. Sindrilaru, A. et al. An unrestrained proinflammatory M1 macrophage population induced by iron impairs wound healing in humans and mice. J. Clin. Invest. 121, 985–997 (2011).

    Article  CAS  Google Scholar 

  16. Wodarz, D. & Anton-Culver, H. Dynamical interactions between multiple cancers. Cell Cycle 4, 764–771 (2005).

    Article  CAS  Google Scholar 

  17. Nejadnik, H. et al. Somatic differentiation and MR imaging of magnetically labeled human embryonic stem cells. Cell Transplant. 21, 2555–2567 (2012).

    Article  Google Scholar 

  18. Jahchan, N. S. et al. A drug repositioning approach identifies tricyclic antidepressants as inhibitors of small cell lung cancer and other neuroendocrine tumors. Cancer Discov. 3, 1364–1377 (2013).

    Article  CAS  Google Scholar 

  19. Lu, M., Cohen, M. H., Rieves, D. & Pazdur, R. FDA report: ferumoxytol for intravenous iron therapy in adult patients with chronic kidney disease. Am. J. Hematol. 85, 315–319 (2010).

    CAS  Google Scholar 

  20. Choi, J. Y. et al. Iron intake, oxidative stress-related genes (MnSOD and MPO) and prostate cancer risk in CARET cohort. Carcinogenesis 29, 964–970 (2008).

    Article  CAS  Google Scholar 

  21. Poljak-Blazi, M. et al. Effect of ferric ions on reactive oxygen species formation, cervical cancer cell lines growth and E6/E7 oncogene expression. Toxicol. In Vitro 25, 160–166 (2011).

    Article  CAS  Google Scholar 

  22. Crowe, W. E., Maglova, L. M., Ponka, P. & Russell, J. M. Human cytomegalovirus-induced host cell enlargement is iron dependent. Am. J. Physiol. Cell Physiol. 287, C1023–C1030 (2004).

    Article  CAS  Google Scholar 

  23. Petersen, D. R. Alcohol, iron-associated oxidative stress, and cancer. Alcohol 35, 243–249 (2005).

    Article  CAS  Google Scholar 

  24. Nie, G., Chen, G., Sheftel, A. D., Pantopoulos, K. & Ponka, P. In vivo tumor growth is inhibited by cytosolic iron deprivation caused by the expression of mitochondrial ferritin. Blood 108, 2428–2434 (2006).

    Article  CAS  Google Scholar 

  25. Knobel, Y., Glei, M., Osswald, K. & Pool-Zobel, B. L. Ferric iron increases ROS formation, modulates cell growth and enhances genotoxic damage by 4-hydroxynonenal in human colon tumor cells. Toxicol. In Vitro 20, 793–800 (2006).

    Article  CAS  Google Scholar 

  26. Basel, M. T. et al. Cell-delivered magnetic nanoparticles caused hyperthermia-mediated increased survival in a murine pancreatic cancer model. Int. J. Nanomedicine 7, 297–306 (2012).

    Article  CAS  Google Scholar 

  27. Chung, T. H. et al. Iron oxide nanoparticle-induced epidermal growth factor receptor expression in human stem cells for tumor therapy. ACS Nano 5, 9807–9816 (2011).

    Article  CAS  Google Scholar 

  28. Foy, S. P. & Labhasetwar, V. Oh the irony: iron as a cancer cause or cure? Biomaterials 32, 9155–9158 (2011).

    Article  CAS  Google Scholar 

  29. Reinisch, W., Staun, M., Bhandari, S. & Munoz, M. State of the iron: how to diagnose and efficiently treat iron deficiency anemia in inflammatory bowel disease. J. Crohns Colitis 7, 429–440 (2013).

    Article  Google Scholar 

  30. Lunov, O. et al. Lysosomal degradation of the carboxydextran shell of coated superparamagnetic iron oxide nanoparticles and the fate of professional phagocytes. Biomaterials 31, 9015–9022 (2010).

    Article  CAS  Google Scholar 

  31. Gupta, A. K. & Wells, S. Surface-modified superparamagnetic nanoparticles for drug delivery: preparation, characterization, and cytotoxicity studies. IEEE Trans. Nanobioscience 3, 66–73 (2004).

    Article  Google Scholar 

  32. Mou, W. et al. Expression of Sox2 in breast cancer cells promotes the recruitment of M2 macrophages to tumor microenvironment. Cancer Lett. 358, 115–123 (2015).

    Article  CAS  Google Scholar 

  33. Becker, M., Muller, C. B., De Bastiani, M. A. & Klamt, F. The prognostic impact of tumor-associated macrophages and intra-tumoral apoptosis in non-small cell lung cancer. Histol. Histopathol. 29, 21–31 (2014).

    Google Scholar 

  34. Laskar, A., Eilertsen, J., Li, W. & Yuan, X. M. SPION primes THP1 derived M2 macrophages towards M1-like macrophages. Biochem. Biophys. Res. Commun. 441, 737–742 (2013).

    Article  CAS  Google Scholar 

  35. Solinas, G., Germano, G., Mantovani, A. & Allavena, P. Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation. J. Leukoc. Biol. 86, 1065–1073 (2009).

    Article  CAS  Google Scholar 

  36. Franklin, R. A. et al. The cellular and molecular origin of tumor-associated macrophages. Science 344, 921–925 (2014).

    Article  CAS  Google Scholar 

  37. Edris, B. et al. Antibody therapy targeting the CD47 protein is effective in a model of aggressive metastatic leiomyosarcoma. Proc. Natl Acad. Sci. USA 109, 6656–6661 (2012).

    Article  CAS  Google Scholar 

  38. Shiao, S. L. et al. TH2-polarized CD4+ T cells and macrophages limit efficacy of radiotherapy. Cancer Immunol. Res. 3, 518–525 (2015).

    Article  CAS  Google Scholar 

  39. Hadjipanayis, C. G. et al. EGFRvIII antibody-conjugated iron oxide nanoparticles for magnetic resonance imaging-guided convection-enhanced delivery and targeted therapy of glioblastoma. Cancer Res. 70, 6303–6312 (2010).

    Article  CAS  Google Scholar 

  40. Thorek, D. L. et al. Non-invasive mapping of deep-tissue lymph nodes in live animals using a multimodal PET/MRI nanoparticle. Nat. Commun. 5, 3097 (2014).

    Article  Google Scholar 

  41. Rosner, M. H. & Auerbach, M. Ferumoxytol for the treatment of iron deficiency. Expert Rev. Hematol. 4, 399–406 (2011).

    Article  CAS  Google Scholar 

  42. Cheng, K. et al. Magnetic antibody-linked nanomatchmakers for therapeutic cell targeting. Nat. Commun. 5, 4880 (2014).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors acknowledge support from the National Institute of Health/National Cancer Institute (NIH/NCI), grant numbers R21CA156124 and R21CA176519, and the Department of Defense BCRP Era of Hope Scholar Expansion Award (BC10412). S.Z. was supported by the Stanford Cancer Imaging Training (SCIT) T32 fellowship programme. We also thank the Stanford Center for Innovation and In-Vivo Imaging (SCI 3) supported by the NCI Cancer Center (P30 CA124435–02) and NCI ICMIC (P50 CA114747) for providing the infrastructure for mouse imaging. G.H. was supported by a Swiss National Science Foundation Grant P-155336 (www.snf.ch) and the Novartis Foundation for Medical-Biological Research (www.stiftungmedbiol.novartis.com). In addition, we thank E. Misquez for her excellent administrative assistance throughout this project, D. Yang for assistance with cell culture and M. Winslow's laboratory (Stanford University) for their generous gift of the SCLC KP1-GFP-Luc cell lines.

Author information

Authors and Affiliations

Authors

Contributions

The study concept and design was developed by H.E.D.-L., L.M.C. and S.Z. The acquisition of data was performed by S.Z., R.S., G.H., M.Ma., S.G. and A.S. (cell experiments, tissue experiments and immunohistochemistry), S.Z., J.S.P., O.L. and H.N. (animal experiments), S.Z., O.L., M.Mo. (magnetic resonance imaging), and S.Z., G.H., A.S. (flow cytometry). All authors contributed to the analysis of the data and discussed the results. H.E.D.-L. and S.Z. wrote the manuscript. All authors edited the manuscript and approved the final version. Funding was obtained by H.E.D.-L. and all studies were supervised by H.E.D.-L.

Corresponding author

Correspondence to Heike Elisabeth Daldrup-Link.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary information

Supplementary information (PDF 1038 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zanganeh, S., Hutter, G., Spitler, R. et al. Iron oxide nanoparticles inhibit tumour growth by inducing pro-inflammatory macrophage polarization in tumour tissues. Nature Nanotech 11, 986–994 (2016). https://doi.org/10.1038/nnano.2016.168

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nnano.2016.168

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing