Design Maps for the Hyperthermic Treatment of Tumors with Superparamagnetic Nanoparticles

A plethora of magnetic nanoparticles has been developed and investigated under different alternating magnetic fields (AMF) for the hyperthermic treatment of malignant tissues. Yet, clinical applications of magnetic hyperthermia are sporadic, mostly due to the low energy conversion efficiency of the metallic nanoparticles and the high tissue concentrations required. Here, we study the hyperthermic performance of commercially available formulations of superparamagnetic iron oxide nanoparticles (SPIOs), with core diameter of 5, 7 and 14 nm, in terms of absolute temperature increase ΔT and specific absorption rate (SAR). These nanoparticles are operated under a broad range of AMF conditions, with frequency f varying between 0.2 and 30 MHz; field strength H ranging from 4 to 10 kA m−1; and concentration cMNP varying from 0.02 to 3.5 mg ml−1. At high frequency field (∼30 MHz), non specific heating dominates and ΔT correlates with the electrical conductivity of the medium. At low frequency field (<1 MHz), non specific heating is negligible and the relaxation of the SPIO within the AMF is the sole energy source. We show that the ΔT of the medium grows linearly with cMNP, whereas the SARMNP of the magnetic nanoparticles is independent of cMNP and varies linearly with f and H2. Using a computational model for heat transport in a biological tissue, the minimum requirements for local hyperthermia (Ttissue >42°C) and thermal ablation (Ttissue >50°C) are derived in terms of cMNP, operating AMF conditions and blood perfusion. The resulting maps can be used to rationally design hyperthermic treatments and identifying the proper route of administration – systemic versus intratumor injection – depending on the magnetic and biodistribution properties of the nanoparticles.

[1]  M. Ferrari,et al.  Discoidal Porous Silicon Particles: Fabrication and Biodistribution in Breast Cancer Bearing Mice , 2012, Advanced functional materials.

[2]  D. Wenzel,et al.  Analysis of trajectories for targeting of magnetic nanoparticles in blood vessels. , 2012, Molecular pharmaceutics.

[3]  T. Park,et al.  Chitosan oligosaccharide-stabilized ferrimagnetic iron oxide nanocubes for magnetically modulated cancer hyperthermia. , 2012, ACS nano.

[4]  M. Garcia,et al.  Acute and Long-Term Effects of Hyperthermia in B16-F10 Melanoma Cells , 2012, PloS one.

[5]  Liberato Manna,et al.  Water-soluble iron oxide nanocubes with high values of specific absorption rate for cancer cell hyperthermia treatment. , 2012, ACS nano.

[6]  Anne L. van de Ven,et al.  Rapid tumoritropic accumulation of systemically injected plateloid particles and their biodistribution. , 2012, Journal of controlled release : official journal of the Controlled Release Society.

[7]  Andrew Tsourkas,et al.  ICP-MS analysis of lanthanide-doped nanoparticles as a non-radiative, multiplex approach to quantify biodistribution and blood clearance. , 2012, Biomaterials.

[8]  Glenn P. Goodrich,et al.  MR temperature imaging of nanoshell mediated laser ablation , 2011, International journal of hyperthermia : the official journal of European Society for Hyperthermic Oncology, North American Hyperthermia Group.

[9]  Yu-feng Li,et al.  Fate and toxicity of metallic and metal-containing nanoparticles for biomedical applications. , 2011, Small.

[10]  Wei Lu,et al.  Effects of photoacoustic imaging and photothermal ablation therapy mediated by targeted hollow gold nanospheres in an orthotopic mouse xenograft model of glioma. , 2011, Cancer research.

[11]  C. Kumar,et al.  Magnetic nanomaterials for hyperthermia-based therapy and controlled drug delivery. , 2011, Advanced drug delivery reviews.

[12]  Morteza Mahmoudi,et al.  Magnetic fluid hyperthermia: focus on superparamagnetic iron oxide nanoparticles. , 2011, Advances in colloid and interface science.

[13]  Jung-tak Jang,et al.  Exchange-coupled magnetic nanoparticles for efficient heat induction. , 2011, Nature nanotechnology.

[14]  M. Aggag,et al.  Magnetic nanoparticle-induced hyperthermia treatment under magnetic resonance imaging. , 2011, Magnetic resonance imaging.

[15]  Nishanth Krishnamurthy,et al.  Tumor ablation and nanotechnology. , 2010, Molecular pharmaceutics.

[16]  Florence Gazeau,et al.  Degradability of superparamagnetic nanoparticles in a model of intracellular environment: follow-up of magnetic, structural and chemical properties , 2010, Nanotechnology.

[17]  Masahiro Hiraoka,et al.  Magnetite nanoparticles with high heating efficiencies for application in the hyperthermia of cancer , 2010 .

[18]  Frank Rattay,et al.  Mathematical spatio-temporal model of drug delivery from low temperature sensitive liposomes during radiofrequency tumour ablation , 2010, International journal of hyperthermia : the official journal of European Society for Hyperthermic Oncology, North American Hyperthermia Group.

[19]  Raju V. Ramanujan,et al.  Modeling the performance of magnetic nanoparticles in multimodal cancer therapy , 2010 .

[20]  Dewei Jia,et al.  Current devices for high-performance whole-body hyperthermia therapy , 2010, Expert review of medical devices.

[21]  J. West,et al.  Near-infrared-resonant gold/gold sulfide nanoparticles as a photothermal cancer therapeutic agent. , 2010, Small.

[22]  S. Curley,et al.  Targeted hyperthermia using metal nanoparticles. , 2010, Advanced drug delivery reviews.

[23]  A. Hamler,et al.  Determination of the Heating Effect of Magnetic Fluid in Alternating Magnetic Field , 2010, IEEE Transactions on Magnetics.

[24]  Q. Pankhurst,et al.  Progress in applications of magnetic nanoparticles in biomedicine , 2009 .

[25]  J. Bischof,et al.  Nanoparticle enhanced thermal therapies , 2009, 2009 Annual International Conference of the IEEE Engineering in Medicine and Biology Society.

[26]  C. Augustine,et al.  Optimizing regional infusion treatment strategies for melanoma of the extremities , 2009, Expert review of anticancer therapy.

[27]  R Ivkov,et al.  Nearly complete regression of tumors via collective behavior of magnetic nanoparticles in hyperthermia , 2009, Nanotechnology.

[28]  Takashi Nakagawa,et al.  Suitability of commercial colloids for magnetic hyperthermia , 2009 .

[29]  Michael J Sailor,et al.  Computationally guided photothermal tumor therapy using long-circulating gold nanorod antennas. , 2009, Cancer research.

[30]  S. Dutz,et al.  Effects of size distribution on hysteresis losses of magnetic nanoparticles for hyperthermia , 2008, Journal of physics. Condensed matter : an Institute of Physics journal.

[31]  D. Schutt,et al.  Effects of variation in perfusion rates and of perfusion models in computational models of radio frequency tumor ablation. , 2008, Medical physics.

[32]  R. Ivkov,et al.  The influence of magnetic and physiological behaviour on the effectiveness of iron oxide nanoparticles for hyperthermia , 2008 .

[33]  Kevin D Donohue,et al.  Assessment of a non-invasive high-throughput classifier for behaviours associated with sleep and wake in mice , 2008, Biomedical engineering online.

[34]  E. Sigurdson,et al.  Diagnosis and treatment of metastatic disease to the liver. , 2008, Seminars in oncology.

[35]  Jennifer Stanfield,et al.  Selective prostate cancer thermal ablation with laser activated gold nanoshells. , 2008, The Journal of urology.

[36]  Christian Bergemann,et al.  Iron oxide nanoparticles as a drug delivery vehicle for MRI monitored magnetic targeting of brain tumors. , 2008, Biomaterials.

[37]  Nastassja A. Lewinski,et al.  Cytotoxicity of nanoparticles. , 2008, Small.

[38]  Matteo Pasquali,et al.  Carbon nanotube‐enhanced thermal destruction of cancer cells in a noninvasive radiofrequency field , 2007, Cancer.

[39]  J. Bacri,et al.  Size-sorted anionic iron oxide nanomagnets as colloidal mediators for magnetic hyperthermia. , 2007, Journal of the American Chemical Society.

[40]  H. Dai,et al.  Carbon nanotubes as multifunctional biological transporters and near-infrared agents for selective cancer cell destruction. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[41]  D. Huber,et al.  Synthesis, properties, and applications of iron nanoparticles. , 2005, Small.

[42]  Jeff W M Bulte,et al.  Iron oxide MR contrast agents for molecular and cellular imaging , 2004, NMR in biomedicine.

[43]  D. P. O'Neal,et al.  Photo-thermal tumor ablation in mice using near infrared-absorbing nanoparticles. , 2004, Cancer letters.

[44]  H. Honda,et al.  Anticancer effect and immune induction by hyperthermia of malignant melanoma using magnetite cationic liposomes , 2003, Melanoma research.

[45]  R. E. Rosensweig,et al.  Heating magnetic fluid with alternating magnetic field , 2002 .

[46]  P. Wust,et al.  The cellular and molecular basis of hyperthermia. , 2002, Critical reviews in oncology/hematology.

[47]  Seungho Wang,et al.  Effects of hyperthermia on the cytoskeleton and focal adhesion proteins in a human thyroid carcinoma cell line , 1999, Journal of cellular biochemistry.

[48]  W. Linnemans,et al.  The effects of hyperthermia on the cytoskeleton: a review. , 1996, International journal of hyperthermia : the official journal of European Society for Hyperthermic Oncology, North American Hyperthermia Group.

[49]  H. Arkin,et al.  Recent developments in modeling heat transfer in blood perfused tissues , 1994, IEEE Transactions on Biomedical Engineering.

[50]  R. Klausner,et al.  Regulating the fate of mRNA: The control of cellular iron metabolism , 1993, Cell.

[51]  H. H. Pennes Analysis of tissue and arterial blood temperatures in the resting human forearm. 1948. , 1948, Journal of applied physiology.

[52]  Jing Liu,et al.  A review of hyperthermia combined with radiotherapy/chemotherapy on malignant tumors. , 2010, Critical reviews in biomedical engineering.

[53]  P Wust,et al.  The biologic rationale of hyperthermia. , 2007, Cancer treatment and research.

[54]  Robert E Lenkinski,et al.  Radiofrequency ablation: modeling the enhanced temperature response to adjuvant NaCl pretreatment. , 2004, Radiology.

[55]  S. Curley,et al.  New Approaches to the Treatment of Hepatic Malignancies Radiofrequency Ablation of Malignant Liver Tumors , 2003 .

[56]  J. Zee Heating the patient : a promising approach ? , 2002 .

[57]  E. Gerner,et al.  Heat shock-induced shedding of cell surface integrins in A549 human lung tumor cells in culture. , 1994, Experimental cell research.

[58]  Dev P. Chakraborty,et al.  Usable Frequencies in Hyperthermia with Thermal Seeds , 1984, IEEE Transactions on Biomedical Engineering.

[59]  H. H. Pennes Analysis of tissue and arterial blood temperatures in the resting human forearm. , 1948, Journal of applied physiology.

[60]  G. Sreenivasa,et al.  For Personal Use. Only Reproduce with Permission from the Lancet Publishing Group. Review Hyperthermia in Cancer Treatment Hyperthermia in Combined Treatment of Cancer , 2022 .