We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Iron oxide nanoparticle targeting mechanism and its application in tumor magnetic resonance imaging and therapy

    Li Wu

    *Author for correspondence:

    E-mail Address: fdwl525@163.com

    College of Medical Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China

    Department of Radiology, The Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai, 519000, China

    ,
    Chunting Wang‡

    College of Medical Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China

    ‡Chunting Wang and Yu Li contributed equally to this work.

    Search for more papers by this author

    &
    Yu Li‡

    College of Medical Imaging, Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China

    ‡Chunting Wang and Yu Li contributed equally to this work.

    Search for more papers by this author

    Published Online:https://doi.org/10.2217/nnm-2022-0246

    Iron oxide nanoparticles (IONPs) can be applied to targeted drug delivery, targeted diagnosis and treatment of tumors due to their easy preparation, good biocompatibility, low biotoxicity, high imaging quality, high magnetothermal sensitivity and stable targeting after certain surface modifications. However, the complexity of the mechanism of action and their properties has led to there being few clinical applications of IONPs. This review first describes the targeting mechanisms of IONPs and their toxicity issues, then discusses the applications of IONP targeting studies in tumor MRI. Finally, the applications of IONP targeting in tumor therapy are listed. The authors show the advantages of targeting IONPs and hope that the review will increase the possibility of converting IONPs from biomedical applications to clinical applications.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J. Clin. 71(1), 7–33 (2021).
    • 2. Xie M, Zhu Y, Xu S et al. A nanoplatform with tumor-targeted aggregation and drug-specific release characteristics for photodynamic/photothermal combined antitumor therapy under near-infrared laser irradiation. Nanoscale 12(21), 11497–11509 (2020).
    • 3. Lin Y, Zhang K, Zhang R et al. Magnetic nanoparticles applied in targeted therapy and magnetic resonance imaging: crucial preparation parameters, indispensable pre-treatments, updated research advancements and future perspectives. J. Mater. Chem. B 8(28), 5973–5991 (2020). •• Summarizes the main biomedical applications of magnetic nanoparticles and provides a clear understanding of the current research progress of iron oxide nanoparticles (IONPs) and the direction of further research.
    • 4. Zhao S, Yu X, Qian Y, Chen W, Shen J. Multifunctional magnetic iron oxide nanoparticles: an advanced platform for cancer theranostics. Theranostics 10(14), 6278–6309 (2020).
    • 5. Danhier F, Feron O, Préat V. To exploit the tumor microenvironment: passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. J. Control. Release 148(2), 135–146 (2010).
    • 6. Nagy JA, Vasile E, Feng D et al. Vascular permeability factor/vascular endothelial growth factor induces lymphangiogenesis as well as angiogenesis. J. Exp. Med. 196(11), 1497–1506 (2002).
    • 7. Omidi Y, Barar J. Targeting tumor microenvironment: crossing tumor interstitial fluid by multifunctional nanomedicines. Bioimpacts 4(2), 55–67 (2014).
    • 8. Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307(5706), 58–62 (2005).
    • 9. Huang D, Sun L, Huang L, Chen Y. Nanodrug delivery systems modulate tumor vessels to increase the enhanced permeability and retention effect. J. Pers. Med. 11(2), 124 (2021). • Discusses abnormal angiogenesis, abnormal vascular structure, irregular blood flow, extensive permeability of tumor vessels and the enhanced permeability and retention effect from the perspective of tumor vessels and proposes that the enhanced permeability and retention effect could be increased by nanoparticles modulating tumor vessels.
    • 10. Alphandéry E. Biodistribution and targeting properties of iron oxide nanoparticles for treatments of cancer and iron anemia disease. Nanotoxicology 13(5), 573–596 (2019).
    • 11. Wang D, Fei B, Halig LV et al. Targeted iron-oxide nanoparticle for photodynamic therapy and imaging of head and neck cancer. ACS Nano 8(7), 6620–6632 (2014).
    • 12. Dhas N, Kudarha R, Pandey A et al. Stimuli responsive and receptor targeted iron oxide based nanoplatforms for multimodal therapy and imaging of cancer: conjugation chemistry and alternative therapeutic strategies. J. Control. Release 333, 188–245 (2021). •• Introduces and describes the particular concept of stimuli-responsiveness, including endogenous, exogenous and dual-/multi-stimulus-based delivery platforms.
    • 13. Friedman AD, Claypool SE, Liu R. The smart targeting of nanoparticles. Curr. Pharm. Des. 19(35), 6315–6329 (2013).
    • 14. Hadjipanayis CG, Machaidze R, Kaluzova M et al. EGFRvIII antibody-conjugated iron oxide nanoparticles for magnetic resonance imaging-guided convection-enhanced delivery and targeted therapy of glioblastoma. Cancer Res. 70(15), 6303–6312 (2010).
    • 15. Farran B, Montenegro RC, Kasa P et al. Folate-conjugated nanovehicles: strategies for cancer therapy. Mater. Sci. Eng. C. Mater. Biol. Appl. 107, 110341 (2020).
    • 16. Nehoff H, Parayath NN, Domanovitch L, Taurin S, Greish K. Nanomedicine for drug targeting: strategies beyond the enhanced permeability and retention effect. Int. J. Nanomed. 9, 2539–2555 (2014).
    • 17. Lima-Tenório MK, Pineda EA, Ahmad NM, Fessi H, Elaissari A. Magnetic nanoparticles: in vivo cancer diagnosis and therapy. Int. J. Pharm. 493(1–2), 313–327 (2015).
    • 18. Al-Jamal KT, Bai J, Wang JT et al. Magnetic drug targeting: preclinical in vivo studies, mathematical modeling, and extrapolation to humans. Nano Lett. 16(9), 5652–5660 (2016).
    • 19. Cole AJ, David AE, Wang J, Galbán CJ, Hill HL, Yang VC. Polyethylene glycol modified, cross-linked starch-coated iron oxide nanoparticles for enhanced magnetic tumor targeting. Biomaterials 32(8), 2183–2193 (2011).
    • 20. Lee N, Yoo D, Ling D, Cho MH, Hyeon T, Cheon J. Iron oxide based nanoparticles for multimodal imaging and magnetoresponsive therapy. Chem. Rev. 115(19), 10637–10689 (2015).
    • 21. Latunde-Dada GO. Ferroptosis: role of lipid peroxidation, iron and ferritinophagy. Biochim. Biophys. Acta Gen. Subj. 1861(8), 1893–1900 (2017).
    • 22. Tan HY, Wang N, Li S, Hong M, Wang X, Feng Y. The reactive oxygen species in macrophage polarization: reflecting its dual role in progression and treatment of human diseases. Oxid. Med. Cell Longev. 2016, 2795090 (2016).
    • 23. Low LE, Lim HP, Ong YS et al. Stimuli-controllable iron oxide nanoparticle assemblies: Design, manipulation and bio-applications. J. Control. Release 345, 231–274 (2022).
    • 24. Aboushoushah S, Alshammari W, Darwesh R, Elbaily N. Toxicity and biodistribution assessment of curcumin-coated iron oxide nanoparticles: multidose administration. Life Sci. 277, 119625 (2021).
    • 25. Huang X, Lin C, Luo C et al. Fe(3)O(4)@M nanoparticles for MRI-targeted detection in the early lesions of atherosclerosis. Nanomedicine-UK 33, 102348 (2021).
    • 26. Huang Y, Hsu JC, Koo H, Cormode DP. Repurposing ferumoxytol: diagnostic and therapeutic applications of an FDA-approved nanoparticle. Theranostics 12(2), 796–816 (2022).
    • 27. Avasthi A, Caro C, Pozo-Torres E, Leal MP, García-Martín ML. Magnetic nanoparticles as MRI contrast agents. Top. Curr. Chem. 378(3), 40 (2020). •• Summarizes in vivo studies of IONPs in animal models, with special emphasis on tumor model-related studies, and clearly compares untargeted and targeted IONPs and describes the advantages of targeting IONPs and current research progress.
    • 28. Geraldes CFGC, Laurent S. Classification and basic properties of contrast agents for magnetic resonance imaging. Contrast Media Mol. Imaging 4(1), 1–23 (2009).
    • 29. Peng Y, Tsang SCE, Chou P. Chemical design of nanoprobes for T1-weighted magnetic resonance imaging. Mater. Today 19(6), 336–348 (2016).
    • 30. Chiarelli PA, Revia RA, Stephen ZR et al. Nanoparticle biokinetics in mice and nonhuman primates. ACS Nano 11(9), 9514–9524 (2017).
    • 31. Xie W, Guo Z, Gao F et al. Shape-, size- and structure-controlled synthesis and biocompatibility of iron oxide nanoparticles for magnetic theranostics. Theranostics 8(12), 3284–3307 (2018).
    • 32. Koikkalainen J, Rhodius-Meester H, Tolonen A et al. Differential diagnosis of neurodegenerative diseases using structural MRI data. NeuroImage: Clin. 11, 435–449 (2016).
    • 33. Currie S, Hoggard N, Craven IJ, Hadjivassiliou M, Wilkinson ID. Understanding MRI: basic MR physics for physicians. Postgrad. Med. J. 89(1050), 209–223 (2013).
    • 34. Jeon M, Halbert MV, Stephen ZR, Zhang M. Iron oxide nanoparticles as T1 contrast agents for magnetic resonance imaging: fundamentals, challenges, applications, and prospectives. Adv. Mater. 33(23), 1906539 (2021).
    • 35. Wang J, Huang Y, David AE et al. Magnetic nanoparticles for MRI of brain tumors. Curr. Pharm. Biotechnol. 13(12), 2403–2416 (2012).
    • 36. Wu Y, Lu Z, Li Y, Yang J, Zhang X. Surface modification of iron oxide-based magnetic nanoparticles for cerebral theranostics: application and prospection. Nanomaterials 10(8), 1441 (2020).
    • 37. Zhang L, Yu F, Cole AJ et al. Gum arabic-coated magnetic nanoparticles for potential application in simultaneous magnetic targeting and tumor imaging. AAPS J. 11(4), 693–699 (2009).
    • 38. Chertok B, Moffat BA, David AE et al. Iron oxide nanoparticles as a drug delivery vehicle for MRI monitored magnetic targeting of brain tumors. Biomaterials 29(4), 487–496 (2008).
    • 39. Kiessling F, Mertens ME, Grimm J, Lammers T. Nanoparticles for imaging: top or flop? Radiology 273(1), 10–28 (2014).
    • 40. Yang R, Fu C, Li N et al. Glycosaminoglycan-targeted iron oxide nanoparticles for magnetic resonance imaging of liver carcinoma. Mater. Sci. and Eng.: C Mater. Biol. Appl. 45, 556–563 (2014).
    • 41. Liang J, Zhang X, Miao Y, Li J, Gan Y. Lipid-coated iron oxide nanoparticles for dual-modal imaging of hepatocellular carcinoma. Int. J. Nanomed. 12, 2033–2044 (2017).
    • 42. Yan C, Wu Y, Feng J et al. Anti-αvβ3 antibody guided three-step pretargeting approach using magnetoliposomes for molecular magnetic resonance imaging of breast cancer angiogenesis. Int. J. Nanomed. 8, 245–255 (2013).
    • 43. Yang RM, Fu CP, Fang JZ et al. Hyaluronan-modified superparamagnetic iron oxide nanoparticles for bimodal breast cancer imaging and photothermal therapy. Int. J. Nanomed. 12, 197–206 (2017).
    • 44. Li L, Wu C, Pan L et al. Bombesin-functionalized superparamagnetic iron oxide nanoparticles for dual-modality MR/NIRFI in mouse models of breast cancer. Int. J. Nanomed. 14, 6721–6732 (2019).
    • 45. Artemov D, Mori N, Okollie B, Bhujwalla ZM. MR molecular imaging of the Her-2/neu receptor in breast cancer cells using targeted iron oxide nanoparticles. Magn. Reson. Med. 49(3), 403–408 (2003).
    • 46. Zou Q, Zhang CJ, Yan YZ, Min ZJ, Li CS. MUC-1 aptamer targeted superparamagnetic iron oxide nanoparticles for magnetic resonance imaging of pancreatic cancer in vivo and in vitro experiment. J. Cell. Biochem. 120(11), 18650–18658 (2019).
    • 47. Wang L, Yin H, Bi R, Gao G, Li K, Liu HL. ENO1-targeted superparamagnetic iron oxide nanoparticles for detecting pancreatic cancer by magnetic resonance imaging. J. Cell. Mol. Med. 24(10), 5751–5757 (2020).
    • 48. Ren S, Song L, Tian Y et al. Emodin-conjugated PEGylation of Fe3O4 nanoparticles for FI/MRI dual-modal imaging and therapy in pancreatic cancer. Int. J. Nanomed. 16, 7463–7478 (2021).
    • 49. Luo Y, Li Y, Li J, Fu C, Yu X, Wu L. Hyaluronic acid-mediated multifunctional iron oxide-based MRI nanoprobes for dynamic monitoring of pancreatic cancer. RSC Adv. 9(19), 10486–10493 (2019).
    • 50. Yari H, Gali H, Awasthi V. Nanoparticles for targeting of prostate cancer. Curr. Pharm. Des. 26(42), 5393–5413 (2020).
    • 51. Tse BW, Cowin GJ, Soekmadji C et al. PSMA-targeting iron oxide magnetic nanoparticles enhance MRI of preclinical prostate cancer. Nanomedicine 10(3), 375–386 (2015).
    • 52. Zhu Y, Sun Y, Chen Y et al. In vivo molecular MRI imaging of prostate cancer by targeting PSMA with polypeptide-labeled superparamagnetic iron oxide nanoparticles. Int. J. Mol. Sci. 16(5), 9573–9587 (2015).
    • 53. Ahmed M, Salam AB, Yates C et al. Double-receptor-targeting multifunctional iron oxide nanoparticles drug delivery system for the treatment and imaging of prostate cancer. Int. J. Nanomed. 12, 6973–6984 (2017).
    • 54. Vallabani N, Singh S. Recent advances and future prospects of iron oxide nanoparticles in biomedicine and diagnostics. 3 Biotech 8(6), 279 (2018). • Summarizes the role of IONPs in various imaging modalities and for hyperthermia and photothermal therapy, enabling a clearer understanding of the advantages of IONPs and their development prospects.
    • 55. Park K. Facing the truth about nanotechnology in drug delivery. ACS Nano 7(9), 7442–7447 (2013).
    • 56. El-Boubbou K. Magnetic iron oxide nanoparticles as drug carriers: clinical relevance. Nanomedicine 13(8), 953–971 (2018). •• Analyzes clinical progress in the use of IONPs and discusses current problems and aspects of active targeting that need to be addressed.
    • 57. Alexiou C, Arnold W, Hulin P et al. Therapeutic efficacy of ferrofluid bound anticancer agent. Magnetohydrodynamics 37(3), 318–322 (2001).
    • 58. Bañobre-López M, Teijeiro A, Rivas J. Magnetic nanoparticle-based hyperthermia for cancer treatment. Rep. Pract. Oncol. Radiother. 18(6), 397–400 (2013).
    • 59. Kumar CSSR, Mohammad F. Magnetic nanomaterials for hyperthermia-based therapy and controlled drug delivery. Adv. Drug Deliv. Rev. 63(9), 789–808 (2011).
    • 60. Chatterjee DK, Diagaradjane P, Krishnan S. Nanoparticle-mediated hyperthermia in cancer therapy. Ther. Deliv. 2(8), 1001–1014 (2011).
    • 61. Yoo D, Jeong H, Noh S, Lee J, Cheon J. Magnetically triggered dual functional nanoparticles for resistance-free apoptotic hyperthermia. Angew. Chem. Int. Ed. 52(49), 13047–13051 (2013).
    • 62. Obaidat IM, Issa B, Haik Y. Magnetic properties of magnetic nanoparticles for efficient hyperthermia. Nanomaterials (Basel) 5(1), 63–89 (2015).
    • 63. Hayashi K, Nakamura M, Sakamoto W et al. Superparamagnetic nanoparticle clusters for cancer theranostics combining magnetic resonance imaging and hyperthermia treatment. Theranostics 3(6), 366–376 (2013).
    • 64. Johannsen M, Gneveckow U, Taymoorian K et al. Morbidity and quality of life during thermotherapy using magnetic nanoparticles in locally recurrent prostate cancer: results of a prospective phase I trial. Int. J. Hyperthermia 23(3), 315–323 (2007).
    • 65. Revia RA, Zhang M. Magnetite nanoparticles for cancer diagnosis, treatment, and treatment monitoring: recent advances. Mater. Today 19(3), 157–168 (2016).
    • 66. Hu Y, Mignani S, Majoral JP, Shen M, Shi X. Construction of iron oxide nanoparticle-based hybrid platforms for tumor imaging and therapy. Chem. Soc. Rev. 47(5), 1874–1900 (2018).
    • 67. Shen S, Tang H, Zhang X et al. Targeting mesoporous silica-encapsulated gold nanorods for chemo-photothermal therapy with near-infrared radiation. Biomaterials 34(12), 3150–3158 (2013).
    • 68. Bagley AF, Hill S, Rogers GS, Bhatia SN. Plasmonic photothermal heating of intraperitoneal tumors through the use of an implanted near-infrared source. ACS Nano 7(9), 8089–8097 (2013).
    • 69. Zhou Z, Sun Y, Shen J et al. Iron/iron oxide core/shell nanoparticles for magnetic targeting MRI and near-infrared photothermal therapy. Biomaterials 35(26), 7470–7478 (2014).
    • 70. Chu M, Shao Y, Peng J et al. Near-infrared laser light mediated cancer therapy by photothermal effect of Fe3O4 magnetic nanoparticles. Biomaterials 34(16), 4078–4088 (2013).
    • 71. Shen S, Wang S, Zheng R et al. Magnetic nanoparticle clusters for photothermal therapy with near-infrared irradiation. Biomaterials 39, 67–74 (2015).
    • 72. Feng L, Yang D, He F et al. A core-shell-satellite structured Fe3O4@g-C3N4-UCNPs-PEG for T1/T2-weighted dual-modal MRI-guided photodynamic therapy. Adv. Healthcare Mater. 6(18), 1700502 (2017).
    • 73. Li Z, Wang C, Cheng L et al. PG-functionalized iron oxide nanoclusters loaded with chlorin e6 for targeted, NIR light induced, photodynamic therapy. Biomaterials 34(36), 9160–9170 (2013).
    • 74. Shi J, Yu X, Wang L et al. PEGylated fullerene/iron oxide nanocomposites for photodynamic therapy, targeted drug delivery and MR imaging. Biomaterials 34(37), 9666–9677 (2013).
    • 75. Canese R, Vurro F, Marzola P. Iron oxide nanoparticles as theranostic agents in cancer immunotherapy. Nanomaterials (Basel) 11(8), 1950 (2021).
    • 76. Dadfar SM, Roemhild K, Drude NI et al. Iron oxide nanoparticles: diagnostic, therapeutic and theranostic applications. Adv. Drug Deliv. Rev. 138, 302–325 (2019). •• Clear summary of the IONPs currently approved or in clinical trials and of key studies for in vitro and in vivo biomedical applications.
    • 77. Denardo DG, Ruffell B. Macrophages as regulators of tumour immunity and immunotherapy. Nat. Rev. Immunol. 19(6), 369–382 (2019).
    • 78. Pan Y, Yu Y, Wang X, Zhang T. Tumor-associated macrophages in tumor immunity. Front. Immunol. 11, 583084 (2020). • Clearly explains the two subtypes of tumor-associated macrophages (TAMs) and their plasticity in response to changes in the tumor microenvironment or therapeutic intervention; summarizes the challenges in inhibiting the protumor effects of TAMs and discusses the potential use of TAMs as a therapeutic target.
    • 79. Reichel D, Tripathi M, Perez JM. Biological effects of nanoparticles on macrophage polarization in the tumor microenvironment. Nanotheranostics 3(1), 66–88 (2019).
    • 80. Corna G, Campana L, Pignatti E et al. Polarization dictates iron handling by inflammatory and alternatively activated macrophages. Haematologica 95(11), 1814–1822 (2010).
    • 81. Wu X, Zhang H. Therapeutic strategies of iron–based nanomaterials for cancer therapy. Biomed. Mater. 16(3), 32003 (2021).
    • 82. Zanganeh S, Hutter G, Spitler R et al. Iron oxide nanoparticles inhibit tumour growth by inducing pro-inflammatory macrophage polarization in tumour tissues. Nat. Nanotechnol. 11(11), 986–994 (2016).
    • 83. Schwerdt JI, Goya GF, Calatayud MP, Hereñú CB, Reggiani PC, Goya RG. Magnetic field-assisted gene delivery: achievements and therapeutic potential. Curr. Gene Ther. 12(2), 116–126 (2012).
    • 84. Wang X, Zhou Z, Wang Z et al. Gadolinium embedded iron oxide nanoclusters as T1–T2 dual-modal MRI-visible vectors for safe and efficient siRNA delivery. Nanoscale 5(17), 8098–8104 (2013).
    • 85. Miao L, Zhang K, Qiao C et al. Antitumor effect of human TRAIL on adenoid cystic carcinoma using magnetic nanoparticle-mediated gene expression. Nanomedicine 9(1), 141–150 (2013).
    • 86. Jia N, Wu H, Duan J et al. Polyethyleneimine-coated iron oxide nanoparticles as a vehicle for the delivery of small interfering RNA to macrophages in vitro and in vivo. J. Vis. Exp. (144), e58660 (2019).
    • 87. Baghban R, Afarid M, Soleymani J, Rahimi M. Were magnetic materials useful in cancer therapy? Biomed. Pharmacother. 144, 112321 (2021).
    • 88. Dulińska-Litewka J, Bazarczyk A, Hałubiec P, Szafrański O, Karnas K, Karewicz A. Superparamagnetic iron oxide nanoparticles-current and prospective medical applications. Materials (Basel) 12(4), (2019).