Skip to content
BY-NC-ND 3.0 license Open Access Published by De Gruyter March 16, 2017

Magnetic targeting with superparamagnetic iron oxide nanoparticles for in vivo glioma

  • Marina Fontes de Paula Aguiar

    Marina Fontes de Paula Aguiar received her bachelor degree in Biology from UNESP, Brazil, in 2011 and her Master Degree in Neuroscience from UNIFESP, Brazil, in 2015 that resulted in a national patent. She has experience in Nanotechnology and Cellular and Molecular biology with emphasis in therapeutic strategies for glioblastoma brain tumor using nanobiotechnological tools. Her researches included the use of glioma animal models to develop strategies for treatment of brain tumors using iron oxide nanoparticles and magnetic targeting.

    , Javier Bustamante Mamani

    Javier Bustamante Mamani obtained his PhD from the University of São Paulo in Brazil in 2009. He is currently doing a postdoctoral study on nanobiotechnology at the Israelite Albert Einstein Hospital. His research interests include the elaboration, the physicochemical and biological studies, and the in vivo application of iron oxide nanoparticles for magnetic hyperthermia.

    , Taylla Klei Felix

    Taylla Klei Felix received his Technologist in Radiology degree from the Ipiranga Integrated Colleges in 2012. Nowadays, he conducts research at the Hospital Israelita Albert Einstein in the Brain Institute, developing research in animal models using iron oxide nanoparticles to application in glioblastomas with magnetic hyperthermia therapy, analyzing the efficiency by bioluminescence techniques, MRI, and histology.

    , Rafael Ferreira dos Reis

    Rafael Ferreira dos Reis has a Biomedical degree from the Mogi das Cruzes University (2013). He had his internship in the lab and has experience in cell culture and scaffolds of carbon nanotubes from the Hospital Albert Einstein from 2012 to 2013. He received his Master’s degree in Sciences from UNIFESP in 2016. His research involved the study on labeling stem cells with iron oxide nanoparticles for tracking of celullar therapy in the AVC animal model. He has experience in the use of nanomaterials and stem cell for regeneration tissues.

    , Helio Rodrigues da Silva

    Helio Rodrigues da Silva received his Bachelor’s degree in Biomedicine from UMESP, Brazil, in 2005 and his Doctorate Degree in Health Sciences from FCMSCSP, Brazil, in 2016. He has experience in nanotechnology and animal experimentation, with emphasis in therapeutic strategies for stroke using nanobiotechnological tools.

    , Leopoldo Penteado Nucci

    Leopoldo Penteado Nucci is a dentist, specializing in periodontology and intensive care and has a Master and PhD degrees in Science from the University of São Paulo, Brazil. He has experience in preclinical and structural neuroimaging and nanobiotechnology applied in Parkinson’s disease, stroke, and brain tumor animal models. He has worked as a research collaborator in nanobiotechnology research group and dentistry in the Hospital Israelita Albert Einstein. His current research interest is in developing novel therapy methods associated with nanobiotechnology for neurodegenerative disease.

    , Mariana Penteado Nucci-da-Silva

    Mariana Penteado Nucci-da-Silva graduated as a Physiotherapist from Santa Cecília University in 2000. She obtained her PhD in Science from the University of São Paulo, Brazil, in 2014, and became Scientific Researcher III of the Laboratory of Medical Investigation in Magnetic Resonance at Radiology Institute of Faculty of Medicine – University of São Paulo. She has experience in functional magnetic resonance image technique in neurologic disease, mainly hypoxic events and nanobiotechnology that can be applied in stroke and brain tumor animal models of neurologic disease, as well as animal behavior analyses and neuroimage analysis in animal models.

    and Lionel Fernel Gamarra

    Lionel Fernel Gamarra obtained his Bachelor’s degree in Physics from the National University Federico Villareal (1998) and his Master’s degree and PhD in Physics from the University of São Paulo. He has a postdoctoral at the Hospital Israelita Albert Einstein, where he is currently the principal investigator of the Neuronanobiotechnology lab. His research group develops novel and effective nanobiotechnological tools for diagnosis and/or treatment of brain cancers or stroke using stem cells, therapeutics, nanoparticles, imaging, and/or diagnosis of neurological diseases and the effect of magnetic hyperthermia in brain cancers.

    EMAIL logo
From the journal Nanotechnology Reviews

Abstract

The purpose of this study was to review the use of the magnetic targeting technique, characterized by magnetic driving compounds based on superparamagnetic iron oxide nanoparticles (SPIONs), as drug delivery for a specific brain locus in gliomas. We reviewed a process mediated by the application of an external static magnetic field for targeting SPIONs in gliomas. A search of PubMed, Cochrane Library, Scopus, and Web of Science databases identified 228 studies, 23 of which were selected based on inclusion criteria and predetermined exclusion criteria. The articles were analyzed by physicochemical characteristics of SPIONs used, cell types used for tumor induction, characteristics of experimental glioma models, magnetic targeting technical parameters, and analysis method of process efficiency. The study shows the highlights and importance of magnetic targeting to optimize the magnetic targeting process as a therapeutic strategy for gliomas. Regardless of the intensity of the patterned magnetic field, the time of application of the field, and nanoparticle used (commercial or synthesized), all studies showed a vast advantage in the use of magnetic targeting, either alone or in combination with other techniques, for optimized glioma therapy. Therefore, this review elucidates the preclinical and therapeutic applications of magnetic targeting in glioma, an innovative nanobiotechnological method.

1 Introduction

Gliomas are the most common tumors in the central nervous system [1], [2], [3], [4], [5]. Of these, astrocytomas, including glioblastoma multiforme (GBM), are the most prevalent and the most aggressive [3], [5], [6] representing 76% of all gliomas [7]. The classification is made in accordance with the degree of malignancy, ranging from grades I to IV. Astrocytomas types I and II are tumors that grow slowly and may be present in the brain of patients for many years without symptomatic progression, while astrocytomas types III and IV are considered the most aggressive and malignant cancers [8]. They arise from the glial cells and grow rapidly, reaching regions of the brain and spinal cord. Despite the substantial increase in basic and clinical studies over the past decades, the median survival of patients with high-grade glioma remains about a year; as a result, it is one of the most devastating and deadly of all human cancers.

The prognosis is still very limited, and current treatment involves surgery, chemotherapy and radiation therapy sessions in order to eliminate the infiltrating cells in healthy tissue [9], [10], [11], [12], [13], [14]. However, unlike other cancer therapies, such as the total surgical removal of the tumor with a margin of normal tissue, this cannot be applied in brain tumors, as each brain region has a vital role.

Traditional intravenous chemotherapy has many negative effects, and low concentration of these drugs stream through the blood-brain barrier [15], [16], [17]. The passive biodistribution used for systemic administration usually results in sub-therapeutic doses to the tumor region [18], which not only leads to elimination of the lesion but also can stimulate growth and resistance of malignant cells [19]. Another disadvantage is that chemotherapy is not selective for tumor cells, and increasing the dose may generate systemic toxicity [20]. Furthermore, because of the high molecular heterogeneity of these tumors, they remain, in most cases, refractory to treatment [21], [22], [23].

In this context, studies focused on the use of nanotechnology resource assisting the diagnosis and treatment of brain tumors are of the utmost importance. Among these, superparamagnetic iron oxide nanoparticles (SPIONs) have shown to play an important role in the diagnosis of brain tumors by magnetic resonance imaging (MRI) allowing early detection of disease, more accurate prognostic, and personalized treatments, besides the monitoring capacity and effectiveness of localized treatments [24], [25].

In a study published in 2015, Gobbo et al. [26] affirmed that the major advantage of theranostic nanomedicine in cancer treatment is the rapid evaluation of treatment results and support for therapeutic planning of patients in a personalized way.

SPIONs have magnetic properties [27], as they are composed of an iron oxide core and a polymer coating establishing important biological properties such as biocompatibility, internalization, and viability. Based on these magnetic nanoparticles, a promising approach has been widely explored using an external magnetic field of these nanoparticles for targeting to the tumor region and consequently optimize the diagnosis and/or therapy. The technique, called magnetic targeting, allows active targeting of the glioma tumor. This is a potentially interesting strategy because it is non-invasive and does not interfere with normal brain function [28].

There are two ways for nanoparticle targeting. The first is the passive targeting of nanoparticles to tumor, the cluster of nanoparticles into the microenvironment tumor due to increased permeation effect caused by increased endocytic transport, vascular exudate, and decreased lymphatic drainage [29]. However, this process does not ensure the passive intake of nanoparticles into tumor cells. Another target or driving method with nanoparticles is functional coating with specific ligand to active target in tumor cells [30]. SPION functionalization has been improved in recent years, but optimization in kinetics and biodistribution of nanoparticles are not well established for cancer treatment [30].

Therefore, other driving processes such as ultrasound [31] or magnetic field to efficient targeting of tumor have shown good results, but the efficiency of the application of the external magnetic field for SPION targeting depends on the magnetic field gradient [32]. Some clinical trials have used SPION and magnetic targeting to other tumors, but no studies have reported its use for gliomas, such as the one described in a review carried out by Lubbe et al. (2001) [33], [34].

Among the factors that may influence technical efficiency of this approach are the intensity of the applied magnetic field, the physicochemical properties of nanoparticles, cytotoxicity, their stability in the bloodstream, and the route of administration of these SPION. The magnetic targeting process is shown in Figure 1. Following the route of administration of SPION, a static external magnetic field is applied to the tumor target of these compounds; thus, this process can be monitored in real time by MRI.

Figure 1: Representation of in vivo magnetic targeting using superparamagnetic iron oxide nanoparticles (SPIONs). The process begins with the SPIONs being administered caudally (I) or by carotid artery (II) and being transported by the blood flow (III). An external magnetic field (ExMF) is applied to target the tumor with SPION (IV), which are concentrated in the tumor tissue (V). MRI showing SPION accumulation (arrow) in tumor with (A) and without (B) ExMF. The illustrations are not drawn to scale.
Figure 1:

Representation of in vivo magnetic targeting using superparamagnetic iron oxide nanoparticles (SPIONs). The process begins with the SPIONs being administered caudally (I) or by carotid artery (II) and being transported by the blood flow (III). An external magnetic field (ExMF) is applied to target the tumor with SPION (IV), which are concentrated in the tumor tissue (V). MRI showing SPION accumulation (arrow) in tumor with (A) and without (B) ExMF. The illustrations are not drawn to scale.

Therefore, considering the importance of understanding of new approaches for the diagnosis and/or treatment of glioma, we conducted a systematic review of magnetic targeting technique. This technique is presented as a possible alternative to enhance the treatment of brain tumors using nanobiotechnology resources. This systematic review was conducted in preclinical studies as well as mimic key features of human disease, such as tumor progression kinetics and anti-tumor responses of the immune system in the presence of GBM [35], [36]. It allows the observation of the advantages and disadvantages of magnetic targeting technique.

2 Materials and methods

2.1 Search strategy

We included studies published between June 1999 and October 2016 that were found in the following databases: Cochrane Library, PubMed, SCOPUS, and Web of Science. A Boolean strategy was applied. The following sequence of keywords and Boolean operators was used: (DecS/MeSH): [(iron oxide) OR nanoparticle OR SPIO OR SPION] AND [(magnetic field) OR (magnetic targeting) OR magnetofection NOT AMF NOT (alternating magnetic field) NOT (AC magnetic field)] AND [(brain tumor) OR glioma OR glioblastoma OR gbm OR (giant cell glioblastoma) OR gliosarcoma OR astrocytoma OR (gliomatosis ependymoma) OR ependymoma OR oligodendroglioma OR oligoastrocytoma OR astroblastoma OR gangliocytoma OR ganglioma].

The classification of tumors of the central nervous system, including all glial tumors, standardized by the World Health Organization [8], was sought.

2.2 Data extraction

In this review, seven of the authors independently applied the search strategy and randomly selected reports for each disease in the databases cited above.

MFA, JBM, and LFG independently searched the studies. Discrepancies in the selection of studies between the two reviewers were discussed with a third reviewer, and concensus was reached. The analysis process and table plots of this study were carried out by full consensus of coauthors, respecting the distribution above. In cases of disagreement, a third independent author resolved the differences by data addition or subtraction. Next, JBM and LFG reviewed the nanoparticle characteristics, MFA and RFR reviewed the in vitro characteristics, MFA, MPNS, LPN, and HRS reviewed the animal model characteristics or in vivo assays, and MFA, TKF, JBM, MPNS, and LFG reviewed magnetic targeting methods used to assess therapeutic efficacy. In cases of disagreement, a third independent and senior author decided by data addition or subtraction.

2.3 Selection criteria

This review included only original articles written in the English language that reported on in vivo magnetic targeting for SPION targeting in gliomas or on the mediation of SPION targeting on the influence of an external magnetic field. Publications indexed in more than one database (duplicate mentions), incomplete data, conference presentation, book chapters, and reports written in a language other than English, as well as studies that did not use SPION, were excluded from this review.

3 Results

After application to the search strategy, 228 original articles were identified. Of these, 23 articles were selected after exclusion of the following articles: those that appeared in more than one database; those indexed in different databases; those with incomplete data; those appearing as a presentation at a conference, congress, or symposium; those published as book chapters; or those that did not use SPION; those that did not perform in vivo studies, those not containing data from the external magnetic field, or did not contain data on gliomas (Figure 2).

Figure 2: Flowchart of selection process of studies included in the review.
Figure 2:

Flowchart of selection process of studies included in the review.

3.1 Physicochemical characteristics of SPION

The physicochemical characteristics of SPIONs used in selected studies are summarized in Table 1. Of the 23 selected studies, 10 studies used SPIONs synthesized in the laboratory (“in lab”) [37], [38], [40], [41], [42], [45], [46], [49], [50], [55], nine used commercial SPIONs [39], [43], [44], [47], [51], [53], [57], [58], [59], and four studies looked at commercial and synthesized SPIONs [48], [52], [54], [56]. Most publications reporting on commercial SPIONs used FluidMAG [43], [47], [48], [51], [52], [53], [56], [57], [58] (Chemicell, Berlin, Germany), all used the FluidMAG-D product, only one study [51] used FluidMAG-ARA, two studies [52], [56] used FluidMAG-CMX, and one used FluidMAG-Heparin and FluidMAG-DEAE [56]. The other commercial SPIONs used were Magnetic Fluid-Amine [39], Advanced Nanotech [44], Resovist [54], and Ferrofluidics [59].

Table 1:

Physicochemical characteristics of SPION used in the magnetic targeting process.

Refs.Iron oxide(Fe) (mg/ml)Core/hydrodynamic size (nm)CoatingMs (Emu/g Fe)R2 relaxivity (s−1mm−1)Tb (K)ζ potential (mV)Manufacture
[37]Fe3O48.12NA/35.7DextranSDMBNANANANAMagnetic Fluid-Aminea
[38]Fe3O4NA11.5/28.7PEG/PEI/Ps 8047.388.56NA19Synthesized
12.3/58DOX@PEG/PEI/Ps 8024.1NANA28
[39]γ-Fe2O3NA8/13ai0.259NANASynthesized
[40]Fe3O4NA5/100–150Lf-MDCsNANANANASynthesized
[41]NA50267.3S-PEG-βgluNANANA−4.3Synthesized
[42]Fe3O42.4NA/164.9DNPH 164.43NANA−29.1
2.4NA/166.6DNPH 265.42−20.3
2.4NA/162.2DNPH 362.87−8.9Synthesized
2.4NA/171.4DNPH 466.59−3.6
[43]Fe3O44012/251βglu-s-IO60118.3NA+18.5G100 (FluidMAG-D)b
[44]NA0.686–10/700–4000Dox-SPIO0.014107.3NANAAdvanced Nanotech (Taiwan)
[45]Fe3O414.15/215–230NA164
18.85/NABCNU57183NANASynthesized
28.25/180–205NA208
[46]NANA8/97Dx-F-RGDNANANANASynthesized
[47]Fe3O42512/110Starch9443.8160−12G100 (FluidMAG-D)b
[48]Fe3O47512/104Starch98.643.8160−12G100 (FluidMAG-D)b
12/142Starch-PEG (5 kDa)103.1NANA+24.4Synthesized
12/168Starch-PEG (20 kDa)104.2NANA+25.6Synthesized
[49]Fe3O4NANA/89SPAnNa-BCNU (9.6%)62.5NANA−19.6Synthesized
SPAnNa-BCNU (19.7%)37.6−33.1
SPAnNa-BCNU (14.4%)55.9−35.1
[50]Fe3O4NANA/10–2066.2NANANASynthesized
SPAnH37.6
[51]Fe3O42512/110Starch12543.8NA−12G100 (FluidMAG-D)b
NA10/225GPEI93NANA37.2Gara (FluidMAG-ARA)b
[52]Fe3O425NA/100CMD94NANANAFluidMAG-CMXb
25NA/100Starch9443.8160−12FluidMAG-Db
0.79/40–280CMD69.4NANA−19.4Synthesized
[53]Fe3O42512/110Starch9443.8160−12G100 (FluidMAG-D)b
[54]Fe3O427.95.9/63.8Carboxydextran73.798.4NA45Resovist®c
γ-Fe2O3
Fe3O4NA10.9/73.7SPAnH-epirubicin51.830.0NANASynthesized
11.4/75.865.9102.3
12.3/83.481.7185.0
[55]Fe3O4NA8.4/NACMDNANANANASynthesized
[56]Fe3O425NA/100CMD94FluidMAG-CMXb
Fe3O410NA/100Heparin94NANANAFluidMAG-Heparinb
Fe3O425NA/100Dextran-diethylaminoethyl94FluidMAG-DEAEb
Fe3O42512/110Starch9443.8160−12G100 (FluidMAG-D)b
Fe3O41014/118Gum arabic93.1NANANASynthesized
[57]Fe3O42512/110Starch9443160−12G100 (FluidMAG-D)c
[58]Fe3O42512/110Starch9443.8160−12G100 (FluidMAG-D)b
[59]Fe3O4NANA/1000–2000AminodextranNA
NA/1000–2000DextranNANANANANA
10/20NAEMG-111d
  1. SPIO, superparamagnetic iron oxide; Fe, ferro; Ms, saturated magnetization strength; R2, relaxivity transversal; Tb, blocking temperature; Lf-MDCs, lactoferrin-tethered magnetic double emulsion nanocapsules; PEG, poly(ethylene glycol); S-PEG-βglu, starch cross-linked, aminated, PEG modification, conjugation of β-glucosidase; DNPH, starch cross-linked, aminated, PEGylated and heparin-conjugated MNP; βglu-s-IO, β-glucosidase to aminated, starch-coated, iron oxide; DOX, doxorubicin; Dox-SPIO, doxorubicin-SPIO-circulating microbubbles; BCNU, (1,3-bis(2-chloroethyl)-1-nitrosourea)-loaded nanobubbles; Dx-F-RGD, Dextrana-Fluorophore- arginine-glycine-aspartate; S-PEG, starch cross-linked, aminated, and modified with PEG; SPAnNa-BCNU (9.6%), poly[aniline-co-sodium N-(1-one-butyric acid) aniline]+BCNU: 9.6% (w/w) SPAnNa; SPAnH, (poly-[aniline-co-N-(1-one-butyric acid) aniline]); PEI, polyethylenimine; GPEI, PEI-modified magnetic nanoparticles coated with gum arabic polysaccharide; CMD, carboxymethyl-dextran; Ps 80, Polysorbate 80; NA, not identified; SDMB, SPIO-doxorubicin-conjugated microbubbles.

  2. aMagQu Co Ltd (New Taipei city, Taiwan).

  3. aiRhodamine-labeled magnetic-fluid-loaded PEG-ylated liposomes (MFLs).

  4. bChemicell® (Berlim, Germany).

  5. c(Bayer Schering Pharma AG, Berlin, Germany).

  6. dFerrofluid (EMG-111, Ferrofluidics Corp, Nashua, NH, USA).

Eight studies [42], [45], [48], [49], [51], [52], [54], [56] used more than one type of SPIONs for a comparison criterion; four studies [48], [52], [54], [56] compared commercial products with synthesized in lab, and four studies compared only SPION synthesized [42], [45], [49], [50]. One study compared only commercial SPIONs [51]. The other studies did not compare the effect of the different SPION types and used only one type in the study, either commercial [39], [43], [44], [47], [53], [57], [58] or synthesized [37], [38], [40], [41], [46], [55]. It is noteworthy that the Pulfer study [59] compared the EMG-111 commercial nanoparticle with microspheres containing SPIONs. However, because the particle size was 1000–2000 nm, this study was not included for this review, in accordance with the previously established selection criteria.

In 14 studies [37], [38], [40], [41], [42], [45], [46], [48], [49], [50], [52], [54], [55], [56], the nanoparticles were synthesized by several chemical methods for SPION production in magnetic targeting. Among the chemical methods used were microemulsions [40], single-phase reaction (one-pot synthesis) [42], [45], hydrothermal reaction [46], sonochemical reaction [52], [55], and the coprecipitation method [49], [50], [56]. The SPION core chemical process for coating particles and conjugations with various materials to produce biocompatible particle was identified in only 14 studies [37], [38], [40], [41], [42], [45], [46], [48], [49], [50], [52], [54], [55], [56], and the crystal phase of iron oxide was identified in 12 studies [37], [38], [40], [42], [45], [48], [49], [50], [52], [54], [55], [56], 11 studies used a magnetite (Fe3O4), one study identified the crystalline phase as maghemite [37]; and two studies [41], [46] did not indicate which of the iron oxide crystalline phase was obtained.

The main coating material for the commercial SPIONs used was starch [43], [47], [48], [51], [52], [53], [56], [57], [58]. Other materials were dextran [39], DOX@PEG/PEI/Ps80 (doxorubicin/poly-ethylene glycol/polyethylenimine/Polysorbate 80) [38], carboxymethyldextran (CMD) [52], [56], carboxydextran [54], heparin, and dextran-diethylaminoethyl [56]. Among the studies summarizing the SPIONs used, the coating materials included DNPH (starch cross-linked, aminated, poly(ethylene glycol) (PEG)ylated and heparin-conjugated magnetic nanoparticles) [42], BCNU (1,3-bis(2-chloroethyl)-1-nitrosourea) [45], Dextrana-Fluorophore-arginine-glycine-aspartate (Dx-F-RGD) [46], SPAnNa-BCNU (poly[aniline-co-sodiumN-(1-one-butyricacid) aniline]-BCNU) [49], SPAnH(poly-[aniline-co-N-(1-one-butyric acid) aniline]) [50], SPAnH-epirubicin [54], and gum arabic [56]. One study [37] used SPION complex to make rhodamine-labeled magnetic fluid-loaded PEG-ylated liposome (MFL) nanoparticle. Some studies have analyzed commercial SPIONs [43], [44], [51], [59] or involved synthesized SPIONs that used a combination of materials to form biocompatible coverage. Fan [39] used commercial SPION coating with dextran to develop SPIO-doxorubicin-conjugated microbubbles. Pulfer [59] used dextran particles with aminodextran and coverage, but as mentioned above, these are treated microspheres and were not considered in this review.

The physicochemical characteristics of the commercial SPIONs used were (I) Magnetic Fluid-Amine (MagQu Co Ltd, New Taipei City, Taiwan) with a magnetite core, average hydrodynamic size of 35.7 nm, coated with dextran, and provided at an iron concentration of 8.12 mg/ml; (II) Fl1uidMAG-d (Chemicell®, Berlin, Germany) with a magnetite core, average core diameter of 12 nm, average hydrodynamic size of 50–251 nm, coated with starch, and provided at an iron concentration of 25 mg/ml; (III) FluidMAG-CMX (Chemicell®, Berlin, Germany) with a magnetite core, an average hydrodynamic diameter of 100 nm, covered with carboxymethyldextran, and provided at an iron concentration of 25 mg/ml; (IV) FluidMAG-ARA (Chemicell®, Berlin, Germany) with a magnetite core, average core diameter of 10 nm and 225 nm average hydrodynamic size, covered and provided with glucuronic acid at an iron concentration of 25 mg/ml; (V) Heparin-FluidMAG (Chemicell®, Berlin, Germany) with a magnetite core, average hydrodynamic size of 100 nm, coated with heparin, and provided at an iron concentration of 10 mg/ml; (VI) FluidMAG DEAE (Chemicell®, Berlin, Germany) with a magnetite core, average hydrodynamic size of 100 nm, coated with diethylaminoethyl-dextran, and provided at an iron concentration of 25 mg/ml; (VII) Resovist® (Bayer Schering Pharma AG, Berlin, Germany), which are nanoparticles of a crystalline phase mixture of magnetite and magnetite, average core diameter of 5.9 nm and 63.8 nm average hydrodynamic size, coated with carboxydextran, and provided at an iron concentration of 27.9 mg/ml; (VIII) Advanced Nanotech (Taiwan), with a core diameter of 6–10 nm and 700–4000 nm hydrodynamic size, and provided at an iron concentration of 0.68 mg/ml; (IX) EMG-111 (Ferrofluidics, Corp. Nashua, NH, USA) with an average core diameter of 10 nm and 20 nm average hydrodynamic size.

3.2 Characteristics of cells used for tumor induction

The characteristics of cells used for tumor induction for further in vivo studies are summarized in Table 2. Most of the selected studies used glial cells of origin [41], [42], [43], [44], [46], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], with only four studies [37], [40], [45], [59] specifically using cells that originated from a glioblastoma rat, called RG21. Among all the studies, only that by Fu [46] and Marie [37] used human cells, designated U87MG, and capable of originating a grade IV astrocytoma or glioblastoma. All other studies used cells from Rattus norvegicus [38], [39], [40], [41], [42], [43], [44], [45], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59] species. Of these, the most widely used cell line was 9L, identified in 11 studies [41], [42], [43], [47], [48], [51], [52], [53], [56], [57], [58].

Table 2:

Cell characteristics of tumor induction.

Refs.Tumor cell lineCell typeOrganismTumor typeMedium culture
[37]C6Glial CellRat/RNGliomaDMEM/F12; 10% FBS; 1% Pen Strep; 1.2 g NaHCO3
[38]C6Glial CellRat/RNGliomaH-DMEM; 10% FBS
[39]U87MGGlial cellHumanGlioblastomaDMEM; 10% FBS; 1% Pen Strep Amp-B
[40]RG2GlioblastomaRat/RNDifferentiated malignant gliomaDMEM; 10% FBS; 1% Pen Strep
[41]9LGlial cellRat/RNGliosarcomaDMEM; 10% FBS; 100 IU/ml Pen; 100 mg/ml Strep
[42]9LGlial cellRat/RNGliosarcomaDMEM; 10% FBS; 1% Pen Strep
[43]9LGlial cellRat/RNGliosarcomaDMEM; 10% FBS; 100 IU/ml Pen; 100 mg/ml Strep; 0.29 mg l-glut
[44]C6Glial cellRat/RNGliomaDMEM/F12; 10% FBS; 1% Pen Strep
[45]RG2GlioblastomaRat/RNDifferentiated malignant gliomaRPMI 1640/FCS
[46]U87MGGlial cellHumanGlioblastoma, AstrocytomaNA
[47]9LGlial cellRat/RNGliosarcomaNA
[48]9LGlial cellRat/RNGliosarcomaDMEM; 10% FBS; 1% antibiotics; 0.29 mg l-glut
[49]C6Glial cellRat/RNGliomaMEM; 10% FBS; 1% Pen Strep
[50]C6Glial cellRat/RNGliomaMEM; 10% FBS; 1% Pen Strep
[51]9LGlial cellRat/RNGliosarcomaDMEM; 10% FBS; 100 IU/ml Pen; 100 μg/ml Strep; 0.29 mg l-glut
[52]9LGlial cellRat/RNGliosarcomaDMEM; 10% FBS; 100 IU/ml Pen; 100 μg/ml Strep; 0.29 mg l-glut
[53]9LGlial cellRat/RNGliosarcomaDMEM; 10% FBS; 100 IU/ml Pen; 100 μg/ml Strep; 0.29 mg l-glut
[54]C6Glial cellRat/RNGliomaNA
[55]C6Glial cellRat/RNGliomaMEM; 10% FBS; 1% Pen Strep
[56]9LGlial cellRat/RNGliosarcomaDMEM; 10% FBS; 100 IU/ml Pen; 100 μg/ml Strep; 0.29 mg l-glut
[57]9LGlial cellRat/RNGliosarcomaDMEM; 10% FBS; 100 IU/ml Pen; 100 μg/ml Strep; 0.29 mg l-glut
[58]9LGlial cellRat/RNGliosarcomaDMEM; 10% FBS; 100 IU/ml Pen; 100 μg/ml Strep; 0.29 mg l-glut
[59]RG2GlioblastomaRat/RNDifferentiated malignant gliomaDMEM; 10% FBS
  1. DMEM, Dulbecco’s modified Eagle’s medium; DMEM/F12, DMEM containing growth fator F12; H-DMEM, DMEM, high glucose; MEM, minimum essential medium; EMEM, Eagle’s minimum essential médium; FBS, fetal bovine serum; Pen Strep, penicillin streptomycin; Amp-B, amphotericin B; L-glut, L-glutamine; FCS, fetal calf serum; RN, Rattus norvegicus; NA, not identified.

The 9L cells originate the gliosarcoma tumor (more aggressive) in rats, mimicking important characteristics of human tumors, such as the pattern of tumor growth, growth factor expression, and metastasis. Another cell line widely used was the C6 (more solid and circumscribed with rat tumor), reported in seven studies [38], [39], [44], [49], [50], [54], [55]. These cells were cloned rat glioma induced by N-nitrosomethylurea [60] and have been widely used for a variety of studies on tumor growth, invasion, migration, neovascularization, growth factor regulation, and biochemical studies because they are morphologically very similar to glioblastoma multiforme [61], [62].

3.3 In vivo experimental model of glioma

The experimental model characteristics, as well as tumor induction parameters, are described in Table 3. All studies selected for this review are in preclinical testing and have used experimental models of rats or mice and clinical trials that have been performed. The small number of clinical studies on this subject shows that progress is early, and more work is needed on the nanobiotechnological tools for the diagnosis and treatment of gliomas.

Table 3:

Experimental design of glioma in vivo study.

Refs.Animal descriptionTumor induction
AnimalSexAge/weight (week/g)T/NGCell typeCell numberAV (μl)VehicleAT (min)LocalizationCoordinates (D; A; L/mm)
[37]Sprague-DawleyMaleNA/20–2516/4C67×1057PBS10Str4.5; 0.5; 3
[38]Sprague-DawleyMaleNA/25–3548/12C61×10610PBS10RH5.0; NA; 3
[39]Swiss nude miceFemale8/21–2712/6U87MG5×1045NANAStr (RH)3; 0.5; 2
[40]BALB/c nude miceFemale3–5/NA15/3RG25×10750PBSNASC (Lf and Rf)NA
[41]Black C57BL6 miceNANA/20–25NA9L2×10650Serum-free DMEMNASC (Rf)NA
[42]Nude miceMaleNA/18–22NA9L109100Serum-free DMEMNASC (Lf)NA
[43]Nude miceNANANA9L2×10650Serum-free DMEMNASC (Rf)NA
[44]Rat/SDMaleNA/200–25043/NAC62.5×1035NA10NA4.5; 0.5; 3
[45]Rat/WistarNANA/NA48/24RG104–105100SalineNASC (right leg)NA
Nude miceFemale24/NA20/52SC
[46]SCID mouseMaleNA/28NAU872×10720NANASCNA
MG
[47]Rat/Fisher344MaleNA/125–150NA9L10610Serum-free DMEMNARF3; NA; NA
[48]Rat/Fisher344MaleNA/125–150NA/4–59L10510Serum-free DMEMNARF3–4; 1; 5
[49]Rat/SDMale14–18/250–30024/6C65×1055MEM10Str4.5; 0.5; 3
[50]Rat/SDMale14–18/320–35064/3–6 or 14C65×1055MEM10Str4.5; 0.5; 3
[51]Rat/Fisher344MaleNA/125–150NA9L10610Serum-free DMEMNARF3; NA; NA
[52]Rat/Fisher344MaleNA/150NA9L10610Serum-free DMEM10RF3; 1; 5
[53]Rat/Fisher344MaleNA/125–150NA/5–89L10610Serum-free DMEMNARF3; 1; 5
[54]Rat/SDNANA/300–40014/NAC6106NANANANANA
[55]Rat/SDMale14–18/350NAC65×1055MEM10Str4.5; 0.5; 3
[56]Rat/Fisher344NANA/200NA/3–49L10510Serum-free DMEMNARF3–4; NA; NA
[57]Rat/Fisher344MaleNA/125–150NA/69L10610Serum-free DMEM10RF3; 1; 5
[58]Rat/Fisher344MaleNA/125–150NA/69L10610Serum-free DMEM10RF3; NA; NA
[59]Rat/Fisher344MaleNA/225/250NA/3RG22×1032DMEM0.5RF4.5; NAa; 2
  1. SD, Sprague Dawley; T, total number of animals; NG, number of animals/group; AT, administration time, AV, administration volume; SC, subcutaneously; Lf, left flank; Rf, right flank; RF, right forebrain; RH, right Hemisphere; Str, striatum; D, depth; A, anterior from the bregma; L, lateral from the bregma; DMEM, Dulbecco’s modified Eagle’s medium; MEM, minimum essential medium; PBS, phosphate-buffered saline; NA, not identified.

Among the rodents identified, seven studies used mice [37], [40], [41], [42], [43], [45], [46], 17 used rats in their experiments [38], [39], [44], [45], [47], [48], [49], [50], [51], [52], [53], [54], [55], [56], [57], [58], [59], and Huang used both rats and mice [45]. From the studies that were based on rat models, the most commonly used species was Fisher 344, identified in nine studies [47], [48], [51], [52], [53], [56], [57], [58], [59]. The second most used species were the Sprague-Dawley rats, reported in seven studies [38], [39], [44], [49], [50], [54], [55]. Only one study [45] used Wistar rats. With regard to studies that used mice, the main species was nude mice, identified in four studies [37], [40], [42], [43]. Only one study [46] used SCID mice, and another study [41] used BLACK C57BL6 species.

The weights of the animals used in the various studies varied widely. For studies that used Fisher 344 rats, the weights ranged from 125 to 150 g in seven studies [47], [48], [51], [52], [53], [57], [58], 200 g in the study by Zhang [56], and ranged from 225 to 250 g in the study by Pulfer [59]. Among the studies that used male Sprague-Dawley rats, the weights varied between 200 and 250 g in Fan’s study [44], 250 and 300 g in Hua’s study [49], 320 and 350 g in the study by Chen [50], and 300 and 400 g in other studies [54], [55]. Huang [45] used Wistar rats and mice, both nude, in his study but did not provide the selected weight. Of the other three studies that used nude mice, the weight ranged from 18 to 22 g in Zhang’s study [42] but was not reported in the studies of Fang [40] and Zhou [43]. Only one study [41] that used C57BL6 black mice selected animals weighing 20–25 g, and Fu [46], the only study using SCID mice, considered animals weighing 28 g.

Regarding the sex of the rodents used, three studies [37], [40], [45] used females, while most selected male rats or mice [38], [39], [42], [44], [46], [47], [48], [49], [50], [51], [52], [53], [55], [57], [58], [59] and four studies [41], [43], [54], [56] did not provide this information. A few studies have reported the total number of animals and number of animals used per group. Among the studies that did provide this information, the variability was wide, ranging from three to eight animals per group [38], [39], [40], [45], [48], [49], [50], [53], [56], [57], [58], [59].

The choice of animal model is often directly related to the selected cell line. Thus, most studies that have opted for cell line 9L inoculated these cells in Fisher 344 rats [47], [48], [51], [52], [53], [56], [57], [58]. However, two studies inoculated 9L cells in nude mice [42], [43], and one study did so in black C57BL6 mice [41]. However, the study by Pulfer [59] used the cell line RG2 in Fisher 344 rats, unlike Fang [40] and Huang [45], who used this same lineage but in BALB/c nude mice and Wistar rats, respectively. All authors who used the C6 cell line chose the Sprague-Dawley [38], [39], [44], [49], [50], [54], [55] species. The only study that used a human cell line (RG2) inoculated these cells in SCID mice [46], which are useful as models for the lack of immunity of B and T cells and, hence, are susceptible to human disease in a chronic state.

The number of inoculated cells for tumor induction also differed in relation to both the cell chosen as the site of the tumor and the species used. Studies induced subcutaneous tumor inoculated [40], [41], [42], [43], [44], [45], [46], [47], in general, proportionally inoculated more cells. Among the studies that implanted tumor cells in the mouse flank, two used 2×106 9L cells [41], [43], two studies used 5×107 and 104–105 RG2 cells [40], [45], one inoculated 109 9L cells [42], one study used 2×107 U87MG cells [46]. Another site frequently used for tumor induction was the right cerebral hemisphere, identified in 11 studies [37], [39], [47], [48], [51], [52], [53], [56], [57], [58], [59]. In this place of glioma induction, the studies that used rats as an experimental model inoculated 106 9L cells [47], [51], [52], [53], [57], [58], 105 9L cells [46], [56], and 2×103 RG2 cells [59]. The striatum was also used as a place of tumor induction and was chosen only for the rat model whose tumors were induced between 104 and 106 C6 cells [37], [38], [39], [49], [50], [55]. Fan [44] inoculated 2.5×103 9L cells but did not report the site of induction. Liu [42] also did not provide the site of tumor induction but reported the use of 106 C6 cells to induce the tumor. Therefore, the major model used in the selected studies was lineage 106 9L cells inoculated into the right cerebral hemisphere (striatum) of male Fisher 344 rats weighing 125–150 g.

The vehicle suspension for inoculation of the tumor cells in animals was mostly DMEM fetal bovine serum free, identified in 11 studies [41], [42], [43], [47], [48], [51], [52], [53], [56], [57], [58]. Pulfer’s study [59] also used DMEM as a vehicle but did not specify whether it was fetal bovine serum free. Other vehicle suspensions were PBS [38], [39], [40] and saline [45] and MEM [49], [50], [55], while three studies [44], [46], [54] did not report the vehicle of choice. A few studies reported the duration of administration of the cells to the tumor site. When mentioned, the duration was 10 min in nine studies [38], [39], [44], [49], [50], [52], [55], [57], [58] and 30 s in one study [59].

The coordinates used for tumor induction also varied among the studies. In studies that inoculated cells subcutaneously, this information does not apply; however, the studies that induced glioma into the right hemisphere of the brain or into the striatum region were based on depth coordinates that were latero-lateral and anterior-posterior in relation to the stereotactic atlas. The average depth was 4.5 mm in five studies [44], [49], [50], [55], [59] and 3 mm in the other 10 studies [38], [39], [47], [48], [51], [52], [53], [56], [57], [58]. Only Liu [54] and Marie [39] did not report the depth used for inoculation of the tumor cells. In relation to the anteroposterior coordinate (axis), some studies reported that the cells were inoculated in the right hemisphere at 1 mm anterior to the bregma [48], [52], [53], [57] or 2 mm posterior to the bregma [59]; others did not provide this information [47], [51], [56], [58]. The latero-lateral coordinate used in the studies that deployed the cells in the right hemisphere was 5 mm [48], [52], [53], [57], 2 mm [59], or not reported [47], [51], [56], [58]. Among the studies [37], [38], [39], [49], [50], [55] whose administration of the cells was into the striatum site, the anteroposterior coordinate was 0.5 mm and 3 mm latero-lateral in relation to the bregma.

3.4 Experimental design of magnetic targeting

The parameters of the transfection process mediated by nanoparticles, also called magnetic targeting, are described in Table 4. For this review, we selected studies that performed magnetic targeting by imposing an external static magnetic field for more efficient and specific targeting of SPION at the tumor region.

Table 4:

Experimental design of magnetic targeting.

Refs.Nanoparticle administrationMagnetic field
Days after inductionTumor size (mm3)NanoparticleDose (mg/kg)VehicleRAAT (min)MF (T)Time of field (h)MagnetApplication
[37]1022.9SD-MB20SalineJugular veinNA0.483Placed tightly to the scalp of the animal’s left headTargeted drug delivery
[38]14~7.5PEG/PEI/Ps 80-MP

DOX@PEG/PEI/Ps 80-MP
2; 6PBSTail veinNA0.31NdFeB disk (2 cm D)Theranostic treatment
[39]144.3MFLs29.8PBSTail veinNA0.44NdFeB (8 mm D, 4 mm height)Targeting of malignant brain tumors
[40]7–1450–100Lf-MDCs-MP; Lf-Cur-MDCS-MP; Lf-Dox-MDCs-MP; Lf-Dox/Cur-MDCs-MPNASalineTail veinNA0.26Wrapped using a bandageTargeted drug delivery
[41]NA300–400S-PEG-β-Glu-MP12NATail veinNA0.321NdFeB cylindrical on 3 tandem DYOYO-52Targeted drug delivery
[42]NA300–500DNPH 3-MP4; 8; 12; 16; 20NATail veinNA0.320.25; 0.5; 0.75; 1l 1.53 DYOYO-N52 cylindrical (51 mm D)+1 D48-N52 cylindrical (6.4 mm D)Novel nano-platform for MRI and MT
[43]NA100–200β-Glu-S-MP12NATail veinNA0.321NdFeB cylindrical (9 mm D) on 3 tandem DYOYO-52Targeted drug delivery
[44]1019.50±9.92DOX-SPIO-MPNASalineJugular veinNANA0.16NAImproving therapeutic efficacy of chemo agents
[45]7, 10, or 14NABCNU-MP12NATail veinNA1.180.08; 0.5; 3; 6; 12; 24; 48Focused MF (1 cm D)Nanobubble system as a cancer therapy
[46]10NADx-F-RGD -MPNANATail veinNA0.20.16; 0.2; 2NdFeB N52 rectangular bar (7.5×7.5×22 mm3)Early cancer detection and targeted therapy
[47]1070–90Starch-MP12PBSTail vein; carotid arteryNA0.15; 0.350.5Dipole electromagnet+cylindrical NdFeB (9 mm D)Targeted drug delivery
[48]1150–100Starch-MP; Starch-PEG (5 kDa)-MP; Starch-PEG (20 kDa)-MP12PBSTail veinNA0.21Cylindrical NdFeB (9 mm D) secured to a 40 mm D poleTargeted drug delivery
[49]10NASPAnNa-BCNU-MP10.5NAJugular veinNA0.3>24NdFeB fixed to the cranium of ratsTargeted drug delivery
[50]17NASPAnH-MP0.5; 1; 5; 8SalineJugular veinNA0.3>24NdFeB fixed to the cranium of ratsEffective and tolerable tumor treatment
[51]1070–90Starch-MP

GPEI-MP
12PBSTail vein; carotid arteryNA0.350.5NdFeB (9 mm D) attached to the pole of dipole electromagnetTargeted vascular drug/gene delivery
[52]1070Starch-MP

CMD-MP
12NATail veinNA0.40.5NATargeted drug delivery
[53]1050–70Starch-MP12–25PBSTail veinNA0.40.5NAApplicability of ICP-OES and ESR in MP biodistribution study
[54]10NACarboxydextran-MP; SPAnH-Epirubicin-MPNANAJugular veinNA0.2; 0.4; 0.553–24Attached to the animal’s scalp, and tightened using a plastic beltDelivery of nanoparticles to the brain
[55]10NACMD-MP10NAJugular veinNA0.32; 24Magnet fixed securely to the craniumTargeted drug delivery
[56]10NAGum Arabic -MP12PBSTail veinNANA0.5NATargeted drug delivery
[57]1050–70Starch-MP12PBSTail vein100.40.5NATargeted drug delivery
[58]1050–70Starch-MP12PBSTail vein100.40.5NATargeted drug delivery
[59]14–21NAAminodextran-MP; Dextran-MP4Tween 80/salineCarotid artery20.60.5NATargeted drug delivery
  1. SPION, superparamagnetic iron oxide nanoparticles; MP, magnetic nanoparticles; AT, administration time; MF, magnetic field; Lf, lactoferrin; Dox, doxorubicin; Cur, curcumin; MDCs, magnetic emulsion nanocapsules; NdFeB, neodymium iron-boron permanent magnet; RA, route administration; S-PEG-β-Glu, starch cross-linked, aminated, PEG modification, conjugation of β-glucosidase; DNPH, aminated, PEGylated, and heparinized D; MT, magnetic targeting; β Glu-S, β-glucosidase to aminated, starch-coated; D, diameter; Dox-SPIO-MBs, doxorubicin-SPIO-circulating microbubbles; BCNU, bischloroethylnitrosourea-loaded nanobubbles; Dx-F-RGD/RGA, Dextrana-Fluorophore-arginine-glycine-aspartate/RGA; SPAnNa, poly[aniline-co-sodium N-(1-one-butyric acid) aniline]; SPAnH, (poly-[aniline-co-N-(1-one-butyric acid) aniline]); PEI, polyethylenimine; PEG, poly(ethylene glycol); Ps 80, Polysorbate 80; GPEI, PEI-modified magnetic nanoparticles coated with gum arabic polysaccharide; CMD, carboxymethyl-dextran; T, Tesla; PBS, phosphate-buffered saline; MRI, magnetic resonance image; ICP-OES, inductively coupled plasma optical emission spectroscopy; ESR, electron spin resonance spectroscopy; NA, not identified; SD-MB, SPIO-doxorubicin-conjugated microbubbles; MFLs, rhodamine-labeled magnetic-fluid-loaded PEG-ylated liposomes.

The magnetic targeting process consists in SPION systemic injection for later magnetic targeting. Most studies administered SPIONs on the 10th day after tumor induction [44], [46], [47], [49], [51], [52], [53], [54], [55], [56], [57], [58] (described in Table 3). Two other studies administered SPIONs 7–14 days after tumor induction [40], [45]. Pulfer’s study [59] performed the procedure 14–21 days after induction. The other studies [41], [42], [43] did not specify this information. However, the appropriate day to perform magnetic targeting process was reported as that on which the tumor volume (mm3) is 300–400 [41], 300–500 [42], or 100–200 [43]. Even among articles that determine the day of SPION administration in animals, most take into account tumor volume [40], [44], [47], [48], [51], [52], [53], [57], [58].

The concentration of the nanoparticles (mg Fe/kg body weight) administered in the studies varied. The concentration used most often was 12 mg Fe/kg [41], [43], [45], [47], [48], [51], [52], [56], [57], [58]. Hua [49] used a concentration of 10.5 mg Fe/kg and Huang [55] used 10 mg Fe/kg, while Pulfer [59] administered nanoparticles at a concentration of 4 mg Fe/kg. Zhang [42], Chen [50], and Chertok [53] used different concentrations of nanoparticles ranging from 4 to 20 mg Fe/kg, 0.5 to 8 mg Fe/kg, and 12 to 25 mg Fe/kg, respectively. Marie’s [37] study had the highest concentration of iron (29 mg Fe/kg). The other studies [40], [44], [46], [54] did not report this information. Regarding the suspension vehicle used to administer the SPION in different routes, most of the studies reporting this information used saline [39], [40], [44], [50] or PBS [37], [38], [47], [48], [51], [53], [56], [57], [58]; Pulfer [59] used Tween 80/saline suspension as a vehicle. The most frequent administration route was intravenously, identified in 17 studies [37], [38], [39], [40], [41], [42], [43], [45], [46], [47], [48], [51], [52], [53], [56], [57], [58]. Other studies administered nanoparticles via the jugular vein [44], [49], [50], [54], [55] or carotid artery [47], [51], [59]. Of these, Chertok [47], [51] used tail injection and carotid administration in order to compare the efficiency of magnetic targeting process performed in different routes. Only three studies reported the duration of nanoparticle infusion: 10 min in two studies [57], [58] and 2 min in the study by Pulfer [59]. The main route of administration of SPIONs in selected studies was tail vein, with 12 mg Fe/kg in PBS vehicle.

As is known about the in vivo magneto targeting process, all studies used an external and static magnetic field with different characteristics for directing nanoparticles to the tumor region. Regarding the intensity of the field used, four studies [40], [46], [48], [54] used a magnetic field of 0.2 T, nine studies [38], [41], [42], [43], [47], [49], [51], [55] used a magnetic field of 0.3–0.35 T, and five studies [37], [39], [52], [53], [57], [58] used only 0.4–0.5 T. The highest intensity values found were 0.6 T [59] and 1.18 T [45]. Chertok [47] and Liu [54] used different field strengths to promote magnetic targeting: from 0.15 to 0.35 T and from 0.2 to 0.55 T, respectively. Only two studies [44], [56] did not report the intensity of the external magnetic field used. The duration of application of the magnetic field in each study also varied: 0.25–1.5 h [42], 0.08–48 h [45], 0.16–2 h [46], 3–24 h [54], and 2–24 h [55]. Most other studies applied the magnetic field for only 30 min [47], [51], [52], [53], [56], [57], [58], [59], four studies [38], [41], [43], [48] applied it for 1 h, Fang [40] did so for 6 h, and Hua [49] and Chen [50] did so for over 24 h. External and static magneto were used in most studies, by using a magnetic field of 0.3 T and 30 min duration.

To optimize the efficiency of the magnetic field, many studies used adaptations to the field for better targeting. Seven studies [44], [52], [53], [56], [57], [58], [59] did not complement the magnet with any artifice. Among the adjustments used, Fang [40] affixed the magnet to the animal’s head using a bandage, Zhou [41], [43] used a cylindrical neodymium magnet connected to the main magnet, Zhang [42] linked one small cylindrical magnet 6.4 mm in diameter to the other third cylindrical neodymium magnet 51 mm in diameter, Huang [45] used a directed magnetic field 1 cm in diameter, Fu [46] chose a rectangular neodymium magnet with dimensions of 22×7.5×7.5 mm3, Chertok [47], [51] used an electromagnet coupled to a cylindrical neodymium magnet 9 mm in diameter, Cole [48] used a cylindrical neodymium magnet 9 mm in diameter secured to a pole 40 mm in diameter, Hua [49], Chen [50], and Huang [55] set the magnet in the skull of rats, and Liu [42] attached the magnet to the animals’ scalp and secured it with a plastic band.

The main applications of the magnetic targeting process identified by the studies in question were magnetically targeted drug delivery [40], [41], [43], [47], [48], [49], [51], [52], [55], [56], [57], [58], [59], a new platform for monitoring magnetic resonance and targeted drug delivery [42], increased therapeutic efficacy of chemotherapeutic agents [44], development of a nanobubble system for cancer treatment [45], early cancer detection and more targeted treatment [46], more effective and tolerable treatment for cancer [50], applicability of inductively coupled plasma-optical emission spectrometer (ICP-OES) and electron spin resonance (ESR) studies in biodistribution studies of magnetic nanoparticles [53], and effective targeted drug delivery to the brain [54].

Figure 3 shows a schematic of the magnetic targeting process for tumor induction through different routes of SPION administration, the range of the dose used, and its delivery vehicle. In the same figure, the range and time of the external static magnetic field application for further evaluation by different techniques.

Figure 3: Experimental designs of magnetic targeting process used in the studies selected for the review. *Number of repetitions of ExMF. Studies not mentioned in this figure did not present any of the data items outlined.
Figure 3:

Experimental designs of magnetic targeting process used in the studies selected for the review. *Number of repetitions of ExMF. Studies not mentioned in this figure did not present any of the data items outlined.

3.5 Toxicity

Although physicochemical characterization of SPION was described by studies included in our review of stability of SPION in vitro, the viability of the technique to apply SPIONs to tumor was discussed in a few manuscripts as well as the kinetic of biodistribution, elimination of SPION, and in vivo toxicity. These constitute the fundamental elements of the magnetic targeting process [47], [51], [53].

External imposition of a static magnetic field for tumor targeting by SPION is better observed in studies on the distribution of SPION in the organism, highlighting the higher concentration of SPION located in the tumor region and its microenvironment. Such fact proved the efficiency of this technique compared with tumor targeting without external magnetic field. Distribution data suggest, indirectly, the viability of the procedure given that the results showed that the physiological route of iron metabolism is maintained, and SPIONs are pushed by the organism, thus, reducing the long-term accumulation of iron in other organs that would turn them toxic [55], [59], [63].

Some selected studies, in addition to magnetic targeting technique application, have also applied therapeutic techniques using colorimetric and fluorescent assays and have shown a reduction of in vivo or in vitro tumor activity. This reduction was observed after tests with different combinations of SPION with different doses of chemotherapy agents and by imposition of external magnetic fields by alternation (generating heat in the tumor area) or not. These studies suggest that specificity of drug delivery to the local of interest using magnetic targeting resource contributes with efficiency to the therapeutic process [40], [43], [44], [45], [47], [51].

Despite the clinical and scientific knowledge on the advantages of the low toxicity that therapies with SPIONs present, because of its biocompatibility and possibility of controlling its migration by magnetic resonance imaging, further detailed studies are needed on the biological markers of free radical formation and viability [54]. The imposition of external magnetic field increases the speed and concentration in the migration route of SPION by the bloodstream, and this would cause a toxic effect in the organism. For this reason, detailed analyses are required to guarantee procedure precision to the therapy [54].

In addition, there is also the need to evaluate if imposition of external magnetic field to tumor targeting with SPIONs, even if static, do not cause excessive increase in temperature of acceptable biological heat band, fluctuations of blood pressure, and other secondary effects, therefore, enabling the continuity of homeostasis of the organism.

3.6 Analysis methods

The analysis of the efficiency magnetic targeting in the tumor region can be performed by a set of techniques such as histological assays, immunohistochemical, in post mortem tissues, or ex vivo and monitoring in vivo by MRI. In short, the selected studies address various neuroimaging techniques, illustrated in Table 5, from in vivo and ex vivo techniques.

Table 5:

Analysis methods.

Refs.Magnetic resonance imagingOther methods
MF (T)SequenceWeighted images: TR(s)/TE (ms)FOV/MT/ST (mm/–/mm)Slice separation (mm)IAT (h)TechniqueAvaliation
[37]7NAT2: 2.54/4134×40/272×320/0.6NA1, 3FUSIn vivo
Multi-echoT2: 2/7.1–11237×60/320×512/0.7NA1, 3HPLC; ICP-OES; EB; HE; PB; fluorescent microscopic imagingEx vivo
[38]3Turbo SET2: 2.3/11056×70/256×256/2.42.91Fluorescence; PB; HE; TUNELEx vivo
[39]7GRE-3DT2*: 4.095/3.12150×150×50/192×144×64/NANA4, 8, 24ESR spectroscopy; confocal fluorescence microscopy; TEM; EFTEMEx vivo
RARET2: 2.5/33200×200/128×128/0.8NA8, 24
[40]NANANANANANAFluorescenceIn vivo
[41]7FSET2: 4/6030×30/128×128/121ESR spectroscopyEx vivo
GRET2: 20×10−3/5
[42]7FSE multi-sliceT2: 4/6030×30/256×256/101ESR spectroscopyEx vivo
[43]7FSET2: 4/6030×30/128×128/121ESR spectroscopy, TEMEx vivo
GRET2: 20×10−3/5
[44]7EchoT2: 30×10−3/1834×40/432×512/0.6NA0.66, 240FUSIn vivo
GRE echo-planarT2*: 1/2535×35/128×128/0.6Fluorometric method, ICP-Mass, EB, HEE× vivo
NAT2: 2.54/4134×40/272×320/0.6
Multi-echo SET2: 2/7.5–12048×65/320×240/0.7
[45]7Turbo RARET2*: 2.5/3340×40/NA/1NA0.5, 3, 6, 12, 24, 48NANA
FlashT2: 9.2×10−3/335.1
[46]NANANANANANAIntravital microscopeIn vivo
[47]7GRET2: 20×10−3/530×30/1220.5ESR spectroscopyEx vivo
FSET2: 4/608×128/1
[48]7GRET2*: 0.18/530×30/128×128/101, 5, 24ESR spectroscopy, PB, NFREx vivo
FSET2: 4/(30 or 60)30×30/256×128/1
[49]3NAT2: 2.51/9439×60/128×256/NANA0, 168TEM, PBEx vivo
[50]3Turbo SET1: 0.421/1139×60/128×256/0.7NA0, 168FUSIn vivo
GRET2: 2.51/9439×60/128×256/0.7TEM, PB, HE, immunohistochemistry, ICP-OESEx vivo
GRET2*: 28×10−3/2043×130/128×384/0.7
[51]7FSET2: 4×10−3/6030×30/128×128/120.5ESR spectroscopyEx vivo
GRET2: 20×10−3/5
[52]7FSET2: (30 or 60)×10−3/430×30/128×128/1NA1, 2, 3, 4, 48, 96, 144, 192, 240NANA
GRET2: 20×10−3/5
[53]7FSET2: 4/6030×30/128×128/120.5ESR spectroscopy, ICP-OESEx vivo
GRET2: 20×10−3/5
[54]3Turbo SET1: 0.78/1539×60/128×256/1.4NA3, 6, 12, 24FUS, confocal/fluorescence microscopyIn Vivo
Double–TE SET2: 3.86/(8/14,28/57,85/228)38×76/128×256/1.4TEM, ICP-OES, PB, NFR, DAPI, EBEx vivo
[55]NANANANANANAICP-OES, HE, PB, NFREx vivo
[56]7FSET2: 4/6030×30/128×128/11.50.5, 240ESR spectroscopyEx vivo
GRET2*: 0.275/15
[57]7FSET2: (30 or 60)×10−3/430×30/128×128/1NA1, 2, 3, 4, 48, 96, 144, 192, 240ESR spectroscopyEx vivo
GRET2: 20×10−3/5
[58]7GRET2: 20×10−3/530×30/128×128/11.51, 2, 3, 4, 48, 96, 144, 192, 240ESR spectroscopy

TEM
Ex vivo
SET2: 4/60
[59]NANANANANANATEMEx vivo
  1. MF, magnetic field; TR, repetition time; TE, echo time; T2, transverse relaxation time; FOV, field-of-view; MT, matrix; ST, Slice thickness; IAT, image acquisition time after magnetofection; FSE, fast-spin-echo; GRE, gradient echo; SE, spin echo; RARE, rapid Imaging with refocused echoes; ESR, electron spin resonance; HPLC, high-performance liquid chromatography; FE-SEM, field emission scanning microscope; TEM, transmission electron microscopy; EFTEM, energy-filtered transmission electron microscopy FUS, focused ultrasound; PB, Prussian blue; NFR, nuclear fast red; HE, hematoxylin-eosin; DAPI, (4′,6-diamidino-2-phenylindole); ICP-Mass, inductively coupled plasma-mass; ICP-OES, inductively coupled plasma optical emission spectroscopy; TUNEL, terminal deoxynucleotidyl transferase dUTP nick-end labeling; NA, not identified.

Most studies evaluated the magnetic targeting efficiency by MRI technique [37], [38], [39], [41], [42], [43], [44], [45], [47], [48], [49], [50], [51], [52], [53], [54], [56], [57], [58]. Of these, 11 studies [39], [41], [42], [43], [47], [48], [51], [53], [56], [57], [58] also used as an analytical method electron spin resonance (ESR) spectroscopy, transmission electron microscopy (TEM) [39], [43], [49], [50], [54], [58], energy-filtered transmission electron microscopy (EFTEM) [39], focused ultrasound (FUS) [37], [44], [50], [54], fluorometry [44], inductively coupled plasma-mass (ICP-Mass) [44], Evans blue (EB) [37], [44], [54], hematoxylin-eosin (HE) [37], [38], [44], [50], Prussian blue (PB) [37], [38], [49], [50], [54], nuclear fast red (NFR) [48], [54], immunohistochemistry [50], inductively coupled plasma optical emission spectroscopy (ICP-OES) [38], [50], [53], [54], high-performance liquid chromatography (HPLC) [38], confocal/fluorescence microscopy [37], [38], [39], [54] or 4′,6-diamidino-2-phenylindole (DAPI) [54], and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) [38]. Only four studies did not use MRI with an analytical method [40], [46], [55], [59]. Of these studies, two [40], [46] used fluorescence tomography, and one [55] used intravital microscopy. Huang [55] reported the use of ICP-OES, HE, PB, and NFR, and another study [59] reported the use of MET. Alternative or complementary methods to MRI were mostly performed ex vivo [37], [38], [39], [40], [41], [42], [43], [44], [47], [48], [49], [50], [51], [53], [54], [55], [56], [57], [58], [59] using tissue samples for ESR spectroscopy, TEM, fluorometry, ICP-mass, Evans blue, HE, PB, NFR, immunohistochemistry, ICP-OES, and DAPI. Some studies also carried out in vivo analyses, such as fluorescence [40], FUS [37], [44], [50], [54], and confocal intravital microscopy [46]/fluorescence microscopy [54].

Among the selected studies, MRI was the main method used for in vivo monitoring and targeting by SPION mediated by an external magnetic field to the tumor region. In addition to the accurate images of the different parts of the body, this technique is also advantageous because it is minimally invasive. The intensity range of the MRI equipment used was from 3.0 T [38], [49], [50], [54] to 7.0 T [37], [39], [41], [42], [43], [44], [45], [47], [48], [51], [52], [53], [56], [57], [58]. The most common sequences used were fast-spin-echo (FSE) [41], [42], [43], [47], [48], [51], [52], [53], [56], [57] and gradient echo (GRE) [39], [41], [43], [44], [47], [48], [50], [51], [52], [53], [56], [57], [58]. Some studies acquired images only 1 h after magnetic targeting [38], [41], [42], [43], while others did temporal measures between 1 and 240 h after targeting [37], [39], [52], [57], [58].

4. Discussion

Brain tumors, especially malignant gliomas, are among the most aggressive human cancers. Despite conventional treatment, which involves surgery, radiation, and chemotherapy, the prognosis is poor, with only 3–5% of patients surviving more than 3 years [64], [65], [66]. Furthermore, conventional chemotherapy has several disadvantages, such as low therapeutic efficacy and severe systemic toxicity due to low selectivity for neoplastic cells [67].

To address these drawbacks, drug delivery systems directed to the area of interest have been investigated [68], [69], with particular attention paid to SPIONs [70], [71]. The biocompatible coverage and high magnetic susceptibility of the core make these important compounds candidates for drug delivery or drug target vehicles. Furthermore, the polymer coating provides the possibility for conjugation of additional functional groups, as well as allows, at the same time, magnetic targeting to tumor and monitoring by MRI [56], [57], [70], [71], [72].

The use of an external magnetic field for targeting compounds to specific sites has been reported for decades; it was initially done with microspheres [73], [74], [75]. The attraction by an external magnetic field allows active targeting into tumor. This is a particularly attractive technique for gliomas because of the difficult location of these tumors. Despite the enhanced permeability and retention effect (EPR) reported in solid tumors, allowing the nanoparticles to accumulate passively in tumor interstice [63], [76], [77], [78], the active targeting significantly increases exposure to the tumor [57].

SPION coating with biomaterials evidence the potential toxicity effects regarding shape, size, and surface characteristics of nanoparticles; however, because of divergence and speculation regarding the analysis of these characteristics, it is very difficult to establish the toxic effects related to these SPION properties [79]. Despite the lack of evidence in toxicity of SPIONs, information regarding the possible risk of SPION exposure to humans is still very limited and conflicting. The potential toxic effects of SPION is mainly focused on evaluating cytotoxic effects, especially changes in viability, cytoskeleton disruptions, ROS production, and in vitro cell cultures [80]. Little is known about the iron toxicity to genetic material, the nervous system, embryonic development, and other endpoints [81]. A review using SPION-labeled cells in regenerative therapies did not report cytotoxic effects for these nanoparticles [82]. In addition, no significant toxicity and successful chondrogenesis occurred in human mesenchymal stem cells incubated for 24 or 72 h with ferucarbotran (a clinically approved and commercially available carboxydextran-coated SPION that is used as a negative MRI contrast agent for hepatic imaging) [83]. These reviews and other studies show lack of evidence to low toxicity of SPIONs in vitro biological assay and in vivo, or in preclinical studies.

In addition, there is scientific and clinical evidence that SPION therapies have advantages such as low toxicity and also present biocompatibility and the possibility of control migration by the MRI [78], [84]. In this context, a search was made for studies that used optimization strategies for the delivery of iron oxide nanoparticles magnetically targeted for tumor lesions using experimental glioma models. Although 23 studies were found within the established prerequisites, they belong to a total of nine research groups. Most publications belong to the group coordinated by Victor Yang (10 out of 23, 46%), followed by Kuo-Wei Chen (4 out of 23, 17%).

Analysis of the accuracy of magnetic targeting process begins with tumor induction in different locations by different cell lines and cell concentrations in selected studies. Afterward, the nanoparticle administration is made by different routes, in different concentrations on different days standardized in each study, for later application of an external magnetic field with different strengths, times, and adjustments, aiming these nanoparticles for targeting to the tumor lesion. Subsequently, it was also compared to the different analysis methods of the targeting efficiency, as is shown in Figure 3.

In this context, and compared with a recent review [62], our review about rat brain tumor models in experimental neuro-oncology, which describes the differences in eight brain tumor lineages in rodent model (C6, 9L, T9, RG2, F98, BT4C, RT-2, CNS-1), allowed to show the difference in route of administration, highlighting that caudal is the most common local used for C6, 9L, BT4C, and RT-2, but intracerebral was the most common for F98 and CNS-1, a result that corroborates with selected studies shown in Figure 3. There are studies with subdermal implantation or animal flank for optimization of SPION magneto driving and tumor growth in other cancer models. However, the blood-brain barrier constitutes a limit for a number of therapeutic drugs and methods because it does not allow infiltration of anything, including drugs. The stereotactic or flank implantation keeps the general principles of tumor cell implantation, but it turns the accuracy of therapeutic outcomes easier or fake in some cancer models. The C6, 9L, and RT-2 are solid and circumscribed tumors. The other tumors are not circumscribed, and they present several degrees of infiltrating capacity of contiguous normal brain with or without islands of tumor cells with distance varying from the core tumor mass. These cancer models pose more difficulty in measuring the accuracy of therapies, including the nanoparticle therapy [62].

The magnetic targeting technique is not new, as the first identified publication was in 1999 [59]. In the selected publications, the use of commercial compounds was equilibrated with the use of the synthesized compounds in the laboratory. Among the studies that used commercial compounds, the majority used FluidMAG-D, coated with starch (15 out of 23, 67%). Another interesting fact is that almost half of the studies used more than one type of nanoparticle to compare the targeting efficiency (9 out of 23, 75%). Of these, about half (4 out of 9, 45%) compared the efficiency of commercial compounds with nanoparticles synthesized by the group, 45% (4 out of 9) compared different synthesized compounds, and only one study compared the efficiency of only commercial compounds. All used nanoparticles had magnetite core and generally had small hydrodynamic size (most with 100–110 nm; 14 out of 23, 64%).

The main cell line used for tumor induction was 9L cells, described in all Yang’s studies (11 out of 23, 45%). These cells are able to generate a gliosarcoma tumor in rats, mimicking important human tumor characteristics, such as the pattern of tumor growth and metastasis. The second most used was the C6 cell line, a model widely used for a variety of studies on tumor growth, invasion, migration, neovascularization, growth factor regulation, and biochemical studies, being morphologically similar to glioblastoma multiforme [61], [62]. DMEM was the main culture medium used for cell proliferation for tumor induction (13 out of 23, 55%).

Most studies used rats as an animal model for tumor induction (15 out of 23, 75%); the most commonly used species was the Fisher 344 rat (9 of 23, 45%). The number of cells also varied, with the most-used concentration 106 cells inoculated directly into the right hemisphere of the brain (7 out of 23, 35%). The main vehicle suspension used for tumor cells was DMEM without serum (11 of 23, 55%).

All studies included in our review showed that application of a static external magnetic field at some level of intensity (0.2–1.18 T) and at some period (0.25–24 h) was enough to increase the nanoparticle concentration in the tumor region compared with non-targeted animals.

For Zhou [41], the conjugation with PEG to increase the stability of the nanoparticles and β-glucosidase enzyme with great effect in killing tumor cells led to a 3.6-fold greater accumulation into the tumor region when applying a magnetic field compared with other groups without targeting. These results corroborate with the previous study [43] in which evidence that the nanoparticle accumulation 60 min after administration was selective for tumor site and increased 3.84-fold nanoparticle concentration in targeted groups that compared with animals without magnetic targeting. Cole [48] also showed an increase of 189- to 229-fold more nanoparticles conjugated with PEG by an external magnetic field in comparison to commercial nanoparticles distributed passively to the tumor.

The success of the magnetic targeting is determined by three factors: intensity of the applied magnetic field, physicochemical properties of the nanoparticles, and stability of these compounds in the bloodstream. Based on this, Zhang’s study [42] examined the influence of the administration route on optimizing the magnetic targeting process; they showed greater therapeutic effectiveness when SPIONs are administered by carotid artery. At a dose of 12 mg Fe/ml and after applying a magnetic field for 45 min, a 200-fold greater accumulation in the tumor region was reported compared with the group using a commercial nanoparticle and non-targeted. Chertok [47] also demonstrated the importance of the administration route, reporting greater nanoparticle exposure in tumor vasculature when administered by carotid. In his study, the nanoparticle aggregation in tumor was 1.8-fold higher when these were administered by intra-arterial route compared to intravenous administration, both with magnetic targeting.

Chertok [51] also tested the applicability of nanoparticles conjugated with PEI to increase retention and vascular drug carriers, using carotid artery for administration in order to decrease the loss of the drug. The influence of the administration route was also studied demonstrating an advantage of 30-fold more nanoparticle aggregation in the tumor comparing carotid to intravenous administration, both subjected to magnetic targeting process. The nanoparticles conjugated with PEI also showed advantages compared to the commercial compound, adding a 5.2-fold higher accumulation in the tumor as magnetically targeted.

The study by Fan [44] used a combination of ultrasound technique with the application of a magnetic field and showed a synergistic effect in nanoparticle accumulation (22.4%) compared with the group without a magnetic field (12%). The same was shown by Chen [50]. In his study, when the magnetic field or ultrasound was applied alone, twofold more SPION accumulation occurred in the tumor region. Therefore, the combination of techniques increases aggregation in tumor 26 times. Also in Liu’s study [54], the combination of these techniques showed an increase in nanoparticle concentration of 244.6% into the tumor area rather than into the adjacent brain tissue. Huan’s study [55] showed that as a consequence of magnetic targeting, nanoparticles were more directed to the brain, reducing the dispersion in other organs as the lungs, liver, kidneys, and spleen. Therefore, the use of 0.5 T of magnetic field for 30 min and single ultrasound of 500 KHz were more effective drug or magneto carrier in the selected studies.

Huang’s study [45] evidenced significant tumor reduction associated with the nanoparticle administration with BCNU and together with an external field, showing that the magnetic targeting increases the tumor exposure and decreases its concentration in the bloodstream, also reducing the toxic effects. In addition, there were more nanoparticle retention and internalization in the tumor by applying the magnetic field (two fold more signal strength after 30 s). Fu [46] in his study also showed the effect of magnetic targeting process in tumor regression, which occurred faster with the use of magnetic targeting due to the higher concentration of nanoparticles conjugated with RDG (antitumor agent) into the tumor locus. Hua [49] also observed a tumor inhibition growth when SPION conjugated with BCNU were magnetically targeted to the tumor region. The Increase in the SPION concentration into the tumor locus using an external magnetic field makes it possible to use a lower dose of drug to promote tumor suppression and reducing systemic adverse effects.

Yu [52] also compared the efficiency of magnetic targeting using commercial nanoparticles and synthesized nanoparticles. It presented advantages over commercial compounds due to more mobility with a magnetic field and effective accumulation in the tumor region. Zhang [56] also compared both commercial and synthesized nanoparticles, conjugating gum arabic to stabilize the nanoparticles, and adding the fluorophore rhodamine B. As a result of magnetic targeting, there was eightfold more nanoparticle aggregation in the tumor when compared to the group without magnetic targeting. The oldest selected study also compared different compounds. Contrary to what occurs in the 1000- to 2000-nm microspheres, there was better magnetic targeting in approximately 10–20 nm nanoparticles. In addition, the imposition of an external magnetic field permitted the accumulation of 41–48% of the all iron dose into tissue between 30 min to 6 h of targeting. Without magnetic targeting, only 31–32% of the dose was accumulated in the tumor locus.

In another, the study by Chertok [57] showed that the imposition of a magnetic field allowed an increase of five times the drug concentration into the tumor area compared to non-targeted animals. Furthermore, the targeting of nanoparticles in the tumor locus persisted for approximately 100 min after the magnet removal. The selectivity retention of the glioma locus relative to the contralateral brain region was also analyzed by Chertok [58]. This study found a 9.6-fold higher retention into the glioma site. In another study by Chertok [53], he only tested the ESR and ICP-OES efficiency quantification of nanoparticles into the tumor region after application of an external magnetic field.

The most common analysis method to check the efficiency of magnetic targeting was the MRI technique identified in 16 studies (80%). Among the various forms of in vivo targeting, MRI is considered an important tool that is non-invasive, with high spatial resolution, without exposure to ionizing radiation, and allows information about the nanoparticle accumulation in the tumor region, from T2-weighted. It allows obtaining real-time images with morphological and functional information at the cellular and molecular level, helping in understanding the migration of nanoparticles guided by an external magnetic field. Other methods that have been widely used are ESR spectroscopy (10 out of 23, 48%), MET (6 out of 23, 30%), and ICP-OES (4 out of 23, 20%), important for nanoparticle quantification and visualization. Less than half of the studies used histological methods to verify the magnetic targeting (6 out of 23, 27%).

In clinical trials about magnetic targeting using SPIONs and other magneto nanoparticles for driving or carrier drugs, therapeutic pitfalls or collateral effects were reported lower, and the toxicity was also low [33], [34], [85]. The clinical trial that used SPION in cancer [33] included seven metastatic breast cancer patients who underwent magnetic field (0.4 T) application near the tumor area twice a day, 3 days per week for 6 weeks, presented SPIONs increased in three fields into the tumor. The comparison done without magneto application showed no hematological effects and toxicity. Other studies produced the same results of the first clinical trial, but an increase in concentration into the tumor area and lower SPION concentration were seen using intravenous administration in the area near the tumor.

According to the data presented here, nanobiotechnology is presented as a powerful tool for the development of magnetically guided nanoplatforms, which ensures a more effective treatment that can be directed to high-grade gliomas and together with MRI allows in vivo tracking. However, because of the complexity of the central nervous system, the magnetic targeting process is still a challenge, and tools in the field of nanomedicine to survey and guide these studies are highly desirable.

About the authors

Marina Fontes de Paula Aguiar

Marina Fontes de Paula Aguiar received her bachelor degree in Biology from UNESP, Brazil, in 2011 and her Master Degree in Neuroscience from UNIFESP, Brazil, in 2015 that resulted in a national patent. She has experience in Nanotechnology and Cellular and Molecular biology with emphasis in therapeutic strategies for glioblastoma brain tumor using nanobiotechnological tools. Her researches included the use of glioma animal models to develop strategies for treatment of brain tumors using iron oxide nanoparticles and magnetic targeting.

Javier Bustamante Mamani

Javier Bustamante Mamani obtained his PhD from the University of São Paulo in Brazil in 2009. He is currently doing a postdoctoral study on nanobiotechnology at the Israelite Albert Einstein Hospital. His research interests include the elaboration, the physicochemical and biological studies, and the in vivo application of iron oxide nanoparticles for magnetic hyperthermia.

Taylla Klei Felix

Taylla Klei Felix received his Technologist in Radiology degree from the Ipiranga Integrated Colleges in 2012. Nowadays, he conducts research at the Hospital Israelita Albert Einstein in the Brain Institute, developing research in animal models using iron oxide nanoparticles to application in glioblastomas with magnetic hyperthermia therapy, analyzing the efficiency by bioluminescence techniques, MRI, and histology.

Rafael Ferreira dos Reis

Rafael Ferreira dos Reis has a Biomedical degree from the Mogi das Cruzes University (2013). He had his internship in the lab and has experience in cell culture and scaffolds of carbon nanotubes from the Hospital Albert Einstein from 2012 to 2013. He received his Master’s degree in Sciences from UNIFESP in 2016. His research involved the study on labeling stem cells with iron oxide nanoparticles for tracking of celullar therapy in the AVC animal model. He has experience in the use of nanomaterials and stem cell for regeneration tissues.

Helio Rodrigues da Silva

Helio Rodrigues da Silva received his Bachelor’s degree in Biomedicine from UMESP, Brazil, in 2005 and his Doctorate Degree in Health Sciences from FCMSCSP, Brazil, in 2016. He has experience in nanotechnology and animal experimentation, with emphasis in therapeutic strategies for stroke using nanobiotechnological tools.

Leopoldo Penteado Nucci

Leopoldo Penteado Nucci is a dentist, specializing in periodontology and intensive care and has a Master and PhD degrees in Science from the University of São Paulo, Brazil. He has experience in preclinical and structural neuroimaging and nanobiotechnology applied in Parkinson’s disease, stroke, and brain tumor animal models. He has worked as a research collaborator in nanobiotechnology research group and dentistry in the Hospital Israelita Albert Einstein. His current research interest is in developing novel therapy methods associated with nanobiotechnology for neurodegenerative disease.

Mariana Penteado Nucci-da-Silva

Mariana Penteado Nucci-da-Silva graduated as a Physiotherapist from Santa Cecília University in 2000. She obtained her PhD in Science from the University of São Paulo, Brazil, in 2014, and became Scientific Researcher III of the Laboratory of Medical Investigation in Magnetic Resonance at Radiology Institute of Faculty of Medicine – University of São Paulo. She has experience in functional magnetic resonance image technique in neurologic disease, mainly hypoxic events and nanobiotechnology that can be applied in stroke and brain tumor animal models of neurologic disease, as well as animal behavior analyses and neuroimage analysis in animal models.

Lionel Fernel Gamarra

Lionel Fernel Gamarra obtained his Bachelor’s degree in Physics from the National University Federico Villareal (1998) and his Master’s degree and PhD in Physics from the University of São Paulo. He has a postdoctoral at the Hospital Israelita Albert Einstein, where he is currently the principal investigator of the Neuronanobiotechnology lab. His research group develops novel and effective nanobiotechnological tools for diagnosis and/or treatment of brain cancers or stroke using stem cells, therapeutics, nanoparticles, imaging, and/or diagnosis of neurological diseases and the effect of magnetic hyperthermia in brain cancers.

Acknowledgments

This study was supported by the Albert Einstein Jewish Institute for Education and Research (Instituto Israelita de Ensino e Pesquisa Albert Einstein – IIEPAE), the National Council for Scientific and Technological Development (Conselho Nacional de Desenvolvimento Científico e Tecnológico – CNPq), the Studies and Projects Financing Agency (Financiadora de Estudos e Projetos – FINEP), the Brazilian Federal Agency for the Support and Evaluation of Graduate Education (Coordenação de Aperfeiçoamento de Pessoal de Nível Superior – CAPES), and the São Paulo State Research Support Foundation (Fundação de Amparo à Pesquisa do Estado de São Paulo – FAPESP).

  1. Competing interest of statement: The authors declare no competing interests. All authors read and approved the final manuscript.

References

[1] Stylli SS, Luwor RB, Ware TM, Tan F, Kaye AH. Mouse models of glioma. J. Clin. Neurosci. 2015, 22, 619–626.10.1016/j.jocn.2014.10.013Search in Google Scholar PubMed

[2] Lim SK, Llaguno SR, McKay RM, Parada LF. Glioblastoma multiforme: a perspective on recent findings in human cancer and mouse models. BMB Rep. 2011, 44, 158–164.10.5483/BMBRep.2011.44.3.158Search in Google Scholar PubMed

[3] de Vries NA, Beijnen JH, van Tellingen O. High-grade glioma mouse models and their applicability for preclinical testing. Cancer Treat. Rev. 2009, 35, 714–723.10.1016/j.ctrv.2009.08.011Search in Google Scholar PubMed

[4] Hu X, Holland EC. Applications of mouse glioma models in preclinical trials. Mutat. Res. 2005, 576, 54–65.10.1016/j.mrfmmm.2004.08.023Search in Google Scholar PubMed

[5] Burton EC, Prados MD. Malignant gliomas. Curr. Treat. Options Oncol. 2000, 1, 459–468.10.1007/s11864-000-0073-2Search in Google Scholar PubMed

[6] Adamson DC, Rasheed BA, McLendon RE, Bigner DD. Central nervous system. Cancer Biomark. 2010, 9, 193–210.10.3233/CBM-2011-0177Search in Google Scholar PubMed

[7] CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2007–2011. 2014, 16.10.1093/neuonc/nou223Search in Google Scholar

[8] Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007, 114, 97–109.10.1007/s00401-007-0243-4Search in Google Scholar PubMed

[9] Castro MG, Cowen R, Williamson IK, David A, Jimenez-Dalmaroni MJ, Yuan X, Bigliari A, Williams JC, Hu J, Lowenstein PR. Current and future strategies for the treatment of malignant brain tumors. Pharmacol. Ther. 2003, 98, 71–108.10.1016/S0163-7258(03)00014-7Search in Google Scholar PubMed

[10] Chiocca EA. Oncolytic viruses. Nat. Rev. Cancer 2002, 2, 938–950.10.1038/nrc948Search in Google Scholar PubMed

[11] Curtin JF, King GD, Candolfi M, Greeno RB, Kroeger KM, Lowenstein PR, Castro MG. Combining cytotoxic and immune-mediated gene therapy to treat brain tumors. Curr. Top. Med. Chem. 2005, 5, 1151–1170.10.2174/156802605774370856Search in Google Scholar PubMed

[12] Gomez-Manzano C, Yung WK, Alemany R, Fueyo J. Genetically modified adenoviruses against gliomas: from bench to bedside. Neurology 2004, 63, 418–426.10.1212/01.WNL.0000133302.15022.7FSearch in Google Scholar PubMed

[13] Castro MG, Candolfi M, Kroeger K, King GD, Curtin JF, Yagiz K, Mineharu Y, Assi H, Wibowo M, Ghulam Muhammad AK, Foulad D, Puntel M, Lowenstein PR. Gene therapy and targeted toxins for glioma. Curr Gene Ther. 2005, 5, 535–557.10.2174/156652305774964631Search in Google Scholar PubMed

[14] Prados MD, Levin V. Biology and treatment of malignant glioma. Semin. Oncol. 2000, 27, 1–10.Search in Google Scholar PubMed

[15] Muldoon LL, Soussain C, Jahnke K, Johanson C, Siegal T, Smith QR, Hall WA, Hynynen K, Senter PD, Peereboom DM, Neuwelt EA. Chemotherapy delivery issues in central nervous system malignancy: a reality check. J. Clin. Oncol. 2007, 25, 2295–2305.10.1200/JCO.2006.09.9861Search in Google Scholar PubMed

[16] Juillerat-Jeanneret L. The targeted delivery of cancer drugs across the blood-brain barrier: chemical modifications of drugs or drug-nanoparticles? Drug Discov. Today 2008, 13, 1099–1106.10.1016/j.drudis.2008.09.005Search in Google Scholar PubMed

[17] Sheikov N, McDannold N, Sharma S, Hynynen K. Effect of focused ultrasound applied with an ultrasound contrast agent on the tight junctional integrity of the brain microvascular endothelium. Ultrasound Med. Biol. 2008, 34, 1093–1104.10.1016/j.ultrasmedbio.2007.12.015Search in Google Scholar PubMed

[18] Motl S, Zhuang Y, Waters CM, Stewart CF. Pharmacokinetic considerations in the treatment of CNS tumours. Clin. Pharmacokinet. 2006, 45, 871–903.10.2165/00003088-200645090-00002Search in Google Scholar PubMed

[19] Heimberger AB, Archer GE, McLendon RE, Hulette C, Friedman AH, Friedman HS, Bigner DD, Sampson JH. Temozolomide delivered by intracerebral microinfusion is safe and efficacious against malignant gliomas in rats. Clin. Cancer Res. 2000, 6, 4148–4153.Search in Google Scholar PubMed

[20] Gutman RL, Peacock G, Lu DR. Targeted drug delivery for brain cancer treatment. J. Control. Release 2000, 65, 31–41.10.1016/S0168-3659(99)00229-1Search in Google Scholar PubMed

[21] Louis DN, Pomeroy SL, Cairncross JG. Focus on central nervous system neoplasia. Cancer Cell 2002, 1, 125–128.10.1016/S1535-6108(02)00040-5Search in Google Scholar PubMed

[22] Ohgaki H, Kleihues P. Population-based studies on incidence, survival rates, and genetic alterations in astrocytic and oligodendroglial gliomas. J. Neuropathol. Exp. Neurol. 2005, 64, 479–489.10.1093/jnen/64.6.479Search in Google Scholar PubMed

[23] Riemenschneider MJ, Jeuken JW, Wesseling P, Reifenberger G. Molecular diagnostics of gliomas: state of the art. Acta Neuropathol. 2010, 120, 567–584.10.1007/s00401-010-0736-4Search in Google Scholar PubMed PubMed Central

[24] Meng XX, Wan JQ, Jing M, Zhao SG, Cai W, Liu EZ. Specific targeting of gliomas with multifunctional superparamagnetic iron oxide nanoparticle optical and magnetic resonance imaging contrast agents. Acta Pharmacologica 2007, 28, 2019–2007.10.1111/j.1745-7254.2007.00661.xSearch in Google Scholar PubMed

[25] Bakhtiary Z, Saei AA, Hajipour MJ, Raoufi M, Vermesh O, Mahmoudi M. Targeted superparamagnetic iron oxide nanoparticles for early detection of cancer: possibilities and challenges. Nanomed.: Nanotechnol. Biol. Med. 2016, 12, 287–307.10.1016/j.nano.2015.10.019Search in Google Scholar PubMed

[26] Gobbo OL, Sjaastad K, Radomski MW, Volkov Y, Prina-Mello A. Magnetic nanoparticles in cancer theranostics. Theranostics 2015, 5, 1249.10.7150/thno.11544Search in Google Scholar PubMed PubMed Central

[27] Bull E, Madani SY, Sheth R, Seifalian A, Green M, Seifalian AM. Stem cell tracking using iron oxide nanoparticles. Int. J. Nanomedicine 2014, 9, 1641–1653.10.2147/IJN.S48979Search in Google Scholar PubMed PubMed Central

[28] Wegscheid ML, Morshed RA, Cheng Y, Lesniak MS. The art of attraction: applications of multifunctional magnetic nanomaterials for malignant glioma. Exp. Opin. Drug Deliv. 2014, 11, 957–975.10.1517/17425247.2014.912629Search in Google Scholar PubMed

[29] Wanga M, Thanoua M. Targeting nanoparticles to cancer. Pharmacol. Res. 2010, 62, 90–99.10.1016/j.phrs.2010.03.005Search in Google Scholar PubMed

[30] Bazak R, Houri M, El Achy S, Kamel S, Refaat T. Cancer active targeting by nanoparticles: a comprehensive review of literature. J. Cancer Res. Clin. Oncol. 2015, 141, 769–784.10.1007/s00432-014-1767-3Search in Google Scholar PubMed PubMed Central

[31] Yeh JS, Sennoga CA, McConnell E, Eckersley R, Tang MX, Nourshargh S, Seddon JM, Haskard DO, Nihoyannopoulos P. A targeting microbubble for ultrasound molecular imaging. PLoS One 2015, 10, e0129681.10.1371/journal.pone.0129681Search in Google Scholar PubMed PubMed Central

[32] Sensenig R, Sapir Y, MacDonald C, Cohen S, Polyak B. Magnetic nanoparticle-based approaches to locally target therapy and enhance tissue regeneration in vivo. Nanomedicine (Lond) 2012, 7, 1425–1442.10.2217/nnm.12.109Search in Google Scholar PubMed PubMed Central

[33] Lübbe AS, Bergemann C, Riess H, Schriever F, Reichardt P, Possinger K, Matthias M, Dörken B, Herrmann F, Gürtler R, Hohenberger P, Haas N, Sohr R, Sander B, Lemke AJ, Ohlendorf D, Huhnt W, Huhn D. Clinical experiences with magnetic drug targeting: a phase I study with 4′-epidoxorubicin in 14 patients with advanced solid tumors. Cancer Res. 1996, 56, 4686–4693.Search in Google Scholar PubMed

[34] Lübbe AS, Alexiou C, Bergemann C. Clinical applications of magnetic drug targeting. J. Surg. Res. 2001, 95, 200–206.10.1006/jsre.2000.6030Search in Google Scholar PubMed

[35] Maher EA, Furnari FB, Bachoo RM, Rowitch DH, Louis DN, Cavenee WK, DePinho RA. Malignant glioma: genetics and biology of a grave matter. Genes Dev. 2001, 15, 1311–1333.10.1101/gad.891601Search in Google Scholar PubMed

[36] Kleihues P, Zülch KJ, Matsumoto S, Radke U. Morphology of malignant gliomas induced in rabbits by systemic application of N-methyl-N-nitrosourea. Z. Neurol. 1970, 198, 65–78.10.1007/BF00316136Search in Google Scholar PubMed

[37] Fan C, Cheng Y, Ting C, Ho Y, Hsu P, Liu H. Ultrasound/magnetic targeting with SPIO-DOX-microbubble complex for image-guided drug delivery in brain tumors. Theranostics 2016, 6, 1542–1556.10.7150/thno.15297Search in Google Scholar PubMed PubMed Central

[38] Xu H, Mao K, Huang Y, Yang J, Xu J, Chen P, Fan ZL, Zou S, Gao ZZ, Yin JY, Xiao J, Lu CT, Zhang BL, Zhao YZ. Glioma-targeted superparamagnetic iron oxide nanoparticles as drug-carrying vehicles for theranostic effects. Nanoscale 2016, 8, 14222–14236.10.1039/C6NR02448CSearch in Google Scholar PubMed

[39] Marie H, Lemaire L, Franconi F, Lajnef S, Frapart Y, Nicolas V, Frébourg G, Trichet M, Ménager C, Lesieur S. Superparamagnetic liposomes for MRI monitoring and external magnetic field-induced selective targeting of malignant brain tumors. Adv. Funct. Mater. 2015, 25, 1258–1269.10.1002/adfm.201402289Search in Google Scholar

[40] Fang JH, Lai YH, Chiu TL, Chen YY, Hu SH, Chen SY. Magnetic core-shell nanocapsules with dual targeting capabilities and co delivery of multiple drugs to treat brain gliomas. Adv. Healthc. Mater. 2014, 3, 1250–1260.10.1002/adhm.201300598Search in Google Scholar PubMed

[41] Zhou J, Zhang J, Gao W. Enhanced and selective delivery of enzyme therapy to 9L-glioma tumor via magnetic targeting of PEG-modified, β-glucosidase-conjugated iron oxide nanoparticles. Int. J. Nanomedicine 2014, 9, 2905–2917.10.2147/IJN.S59556Search in Google Scholar PubMed PubMed Central

[42] Zhang J, Shin MC, Yang VC. Magnetic targeting of novel heparinized iron oxide nanoparticles evaluated in a 9L-glioma mouse model. Pharm, Res. 2014, 31, 579–592.10.1007/s11095-013-1182-5Search in Google Scholar PubMed PubMed Central

[43] Zhou J, Zhang J, David AE, Yang VC. Magnetic tumor targeting of β-glucosidase immobilized iron oxide nanoparticles. Nanotechnology 2013, 24, 375102.10.1088/0957-4484/24/37/375102Search in Google Scholar PubMed PubMed Central

[44] Fan CH, Ting CY, Lin HJ, Wang CH, Liu HL, Yen TC, Yeh CK. SPIO-conjugated, doxorubicin-loaded microbubbles for concurrent MRI and focused-ultrasound enhanced brain-tumor drug delivery. Biomaterials 2013, 34, 3706–3715.10.1016/j.biomaterials.2013.01.099Search in Google Scholar PubMed

[45] Huang HY, Hu SH, Hung SY, Chiang CS, Liu HL, Chiu TL, Lai HY, Chen YY, Chen SY. SPIO nanoparticle-stabilized PAA-F127 thermosensitive nanobubbles with MR/US dual-modality imaging and HIFU-triggered drug release for magnetically guided in vivo tumor therapy. J. Control. Release 2013, 172, 118–127.10.1016/j.jconrel.2013.07.029Search in Google Scholar PubMed

[46] Fu A, Wilson RJ, Smith BR, Mullenix J, Earhart C, Akin D, Guccione S, Wang SX, Gambhir SS. Fluorescent magnetic nanoparticles for magnetically enhanced cancer imaging and targeting in living subjects. ACS Nano 2012 Aug 28, 6, 6862–6869.10.1021/nn301670aSearch in Google Scholar PubMed PubMed Central

[47] Chertok B, David AE, Yang VC. Brain tumor targeting of magnetic nanoparticles for potential drug delivery: effect of administration route and magnetic field topography. J. Control. Release. 2011, 155, 393–399.10.1016/j.jconrel.2011.06.033Search in Google Scholar PubMed PubMed Central

[48] Cole AJ, David AE, Wang J, Galbán CJ, Yang VC. Magnetic brain tumor targeting and biodistribution of long-circulating PEG-modified, cross-linked starch-coated iron oxide nanoparticles. Biomaterials 2011, 32, 6291–6301.10.1016/j.biomaterials.2011.05.024Search in Google Scholar PubMed PubMed Central

[49] Hua MY, Liu HL, Yang HW, Chen PY, Tsai RY, Huang CY, Tseng IC, Lyu LA, Ma CC, Tang HJ, Yen TC, Wei KC. The effectiveness of a magnetic nanoparticle-based delivery system for BCNU in the treatment of gliomas. Biomaterials 2011, 32, 516–527.10.1016/j.biomaterials.2010.09.065Search in Google Scholar PubMed

[50] Chen PY, Liu HL, Hua MY, Yang HW, Huang CY, Chu PC, Lyu LA, Tseng IC, Feng LY, Tsai HC, Chen SM, Lu YJ, Wang JJ, Yen TC, Ma YH, Wu T, Chen JP, Chuang JI, Shin JW, Hsueh C, Wei KC. Novel magnetic/ultrasound focusing system enhances nanoparticle drug delivery for glioma treatment. Neuro Oncol. 2010, 12, 1050–1060.10.1093/neuonc/noq054Search in Google Scholar PubMed PubMed Central

[51] Chertok B, David AE, Yang VC. Polyethyleneimine-modified iron oxide nanoparticles for brain tumor drug delivery using magnetic targeting and intra-carotid administration. Biomaterials 2010, 31, 6317–6324.10.1016/j.biomaterials.2010.04.043Search in Google Scholar PubMed PubMed Central

[52] Yu F, Zhang L, Huang Y, Sun K, David AE, Yang VC. The magnetophoretic mobility and superparamagnetism of core-shell iron oxide nanoparticles with dual targeting and imaging functionality. Biomaterials 2010, 31, 5842–5848.10.1016/j.biomaterials.2010.03.072Search in Google Scholar PubMed PubMed Central

[53] Chertok B, Cole AJ, David AE, Yang VC. Comparison of electron spin resonance spectroscopy and inductively-coupled plasma optical emission spectroscopy for biodistribution analysis of iron-oxide nanoparticles. Mol. Pharm. 2010, 7, 375–385.10.1021/mp900161hSearch in Google Scholar PubMed PubMed Central

[54] Liu HL, Hua MY, Yang HW, Huang CY, Chu PC, Wu JS, Tseng IC, Wang JJ, Yen TC, Chen PY, Wei KC. Magnetic resonance monitoring of focused ultrasound/magnetic nanoparticle targeting delivery of therapeutic agents to the brain. Proc. Natl. Acad. Sci. USA 2010, 107, 15205–15210.10.1073/pnas.1003388107Search in Google Scholar PubMed PubMed Central

[55] Huang BR, Chen PY, Huang CY, Jung SM, Ma YH, Wu T, Chen JP, Wei KC. Bioavailability of magnetic nanoparticles to the brain. J. Magn. Magn. Mater. 2009, 312, 1604–1609.10.1016/j.jmmm.2009.02.095Search in Google Scholar

[56] Zhang L, Yu F, Cole AJ, Chertok B, David AE, Wang J, Yang VC. Gum arabic-coated magnetic nanoparticles for potential application in simultaneous magnetic targeting and tumor imaging. AAPS J. 2009, 11, 693–699.10.1208/s12248-009-9151-ySearch in Google Scholar PubMed PubMed Central

[57] Chertok B, Moffat BA, David AE, Yu F, Bergemann C, Ross BD, Yang VC. Iron oxide nanoparticles as a drug delivery vehicle for MRI monitored magnetic targeting of brain tumors. Biomaterials 2008, 29, 487–496.10.1016/j.biomaterials.2007.08.050Search in Google Scholar PubMed PubMed Central

[58] Chertok B, David AE, Huang Y, Yang VC. Glioma selectivity of magnetically targeted nanoparticles: a role of abnormal tumor hydrodynamics. J. Control. Release 2007, 122, 315–323.10.1016/j.jconrel.2007.05.030Search in Google Scholar PubMed PubMed Central

[59] Pulfer SK, Ciccotto SL, Gallo JM. Distribution of small magnetic particles in brain tumor-bearing rats. J. Neurooncol. 1999, 41, 99–105.10.1023/A:1006137523591Search in Google Scholar PubMed

[60] Benda P, Lightbody J, Sato G, Levine L, Sweet W. Differentiated rat glial cell strain in tissue culture. Science 1968, 161, 370–371.10.1126/science.161.3839.370Search in Google Scholar PubMed

[61] Grobben B, De Deyn PP, Slegers H. Rat C6 glioma as experimental model system for the study of glioblastoma growth and invasion. Cell Tissue Res. 2002, 310, 257–270.10.1007/s00441-002-0651-7Search in Google Scholar PubMed

[62] Barth RF, Kaur B. Rat brain tumor models in experimental neuro-oncology: the C6, 9L, T9, RG2, F98, BT4C, RT-2 and CNS-1 gliomas. J. Neurooncol. 2009, 94, 299–312.10.1007/s11060-009-9875-7Search in Google Scholar PubMed PubMed Central

[63] Maeda H, Bharate GY, Daruwalla J. Polymeric drugs for efficient tumor-targeted drug delivery based on EPR-effect. Eur. J. Pharm. Biopharm. 2009, 71, 409–419.10.1016/j.ejpb.2008.11.010Search in Google Scholar PubMed

[64] Aoki T, Hashimoto N, Matsutani M. Management of glioblastoma. Exp. Opin. Pharmacother. 2007, 8, 3133–3146.10.1517/14656566.8.18.3133Search in Google Scholar PubMed

[65] Maher EA, Mietz J, Arteaga CL, DePinho RA, Mohla S. Brain metastasis: opportunities in basic and translational research. Cancer Res. 2009, 69, 6015–6020.10.1158/0008-5472.CAN-08-4347Search in Google Scholar PubMed

[66] Krex D, Klink B, Hartmann C, von Deimling A, Pietsch T, Simon M, Sabel M, Steinbach JP, Heiese O, Reifenberger G, Weller M, Schackert G. German GliomaNetwork. Long-term survival with glioblastoma multiforme. Brain 2007, 130, 2596–2606.10.1093/brain/awm204Search in Google Scholar PubMed

[67] Lindley C, McCune JS, Thomason TE, Lauder D, Sauls A, Adkins S, Sawyer WT. Perception of chemotherapy side effects cancer versus noncancer patients. Cancer Pract. 1999, 7, 59–65.10.1046/j.1523-5394.1999.07205.xSearch in Google Scholar PubMed

[68] Sawant RM, Hurley JP, Salmaso S, Kale A, Tolcheva E, Levchenko TS, Torchilin VP. “SMART” drug delivery systems: double-targeted pH-responsive pharmaceutical nanocarriers. Bioconjug. Chem. 2006, 17, 943–949.10.1021/bc060080hSearch in Google Scholar PubMed PubMed Central

[69] Winer I, Wang S, Lee YE, Fan W, Gong Y, Burgos-Ojeda D, Spahlinger G, Kopelman R, Buckanovich RJ. F3-targeted cisplatin-hydrogel nanoparticles as an effective therapeutic that targets both murine and human ovarian tumor endothelial cells in vivo. Cancer Res. 2010, 70, 8674–8683.10.1158/0008-5472.CAN-10-1917Search in Google Scholar PubMed PubMed Central

[70] Xie J, Liu G, Eden HS, Ai H, Chen X. Surface-engineered magnetic nanoparticle platforms for cancer imaging and therapy. Acc. Chem. Res. 2011, 44, 883–892.10.1021/ar200044bSearch in Google Scholar PubMed PubMed Central

[71] Reddy LH, Arias JL, Nicolas J, Couvreur P. Magnetic nanoparticles: design and characterization, toxicity and biocompatibility, pharmaceutical and biomedical applications. Chem. Rev. 2012, 112, 5818–5878.10.1021/cr300068pSearch in Google Scholar PubMed

[72] Yoo D, Lee JH, Shin TH, Cheon J. Theranostic magnetic nanoparticles. Acc. Chem. Res. 2011, 44, 863–874. Erratum in: Acc Chem Res. 2012, 45, 1622.10.1021/ar200085cSearch in Google Scholar PubMed

[73] Widder KJ, Senyel AE, Scarpelli GD. Magnetic microspheres: a model system of site specific drug delivery in vivo. Proc. Soc. Exp. Biol. Med. 1978, 158, 141–146.10.3181/00379727-158-40158Search in Google Scholar PubMed

[74] Widder KJ, Senyei AE. Magnetic microspheres: a vehicle for selective targeting of drugs. Pharmacol. Ther. 1983, 20, 377–395.10.1016/0163-7258(83)90033-5Search in Google Scholar PubMed

[75] Gupta PK, Hung CT. Magnetically controlled targeted micro-carrier systems. Life Sci. 1989, 44, 175–186.10.1016/0024-3205(89)90593-6Search in Google Scholar PubMed

[76] Chandran SS, Banerjee SR, Mease RC, Pomper MG, Denmeade SR. Characterization of a targeted nanoparticle functionalized with a urea-based inhibitor of prostate-specific membrane antigen (PSMA). Cancer Biol. Ther. 2008, 7, 974–982.10.4161/cbt.7.6.5968Search in Google Scholar PubMed

[77] Maeda H, Wu J, Sawa T, Matsumura Y, Hori K. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J. Control. Release 2000, 65, 271–284.10.1016/S0168-3659(99)00248-5Search in Google Scholar PubMed

[78] Maier-Hauff K, Ulrich F, Nestler D, Niehoff H, Wust P, Thiesen B, Orawa H, Budach V, Jordan A. Efficacy and safety of intratumoral thermotherapy using magnetic iron-oxide nanoparticles combined with external beam radiotherapy on patients with recurrent glioblastoma multiforme. J. Neurooncol. 2011, 103, 317–324.10.1007/s11060-010-0389-0Search in Google Scholar PubMed PubMed Central

[79] Patil US, Adireddy S, Jaiswal A, Mandava S, Lee BR, Chrisey DB. In vitro/in vivo toxicity evaluation and quantification of iron oxide nanoparticles. Int. J. Mol. Sci. 2015, 16, 24417–24450.10.3390/ijms161024417Search in Google Scholar PubMed PubMed Central

[80] Valdiglesias V, Kiliç G, Costa C, Fernández-Bertólez N, Pásaro E, Teixeira JP, Laffon B. Effects of iron oxide nanoparticles: cytotoxicity, genotoxicity, developmental toxicity, and neurotoxicity. Environ. Mol. Mutagen. 2015, 56, 125–148.10.1002/em.21909Search in Google Scholar PubMed

[81] Au KW, Liao SY, Lee YK, Lai WH, Ng KM, Chan YC, Yip MC, Ho CY, Wu EX, Li RA, Siu CW, Tse HF. Effects of iron oxide nanoparticles on cardiac differentiation of embryonic stem cells. Biochem. Biophys. Res. Commun. 2009, 379, 898–903.10.1016/j.bbrc.2008.12.160Search in Google Scholar PubMed

[82] Alvarim LT, Nucci LP, Mamani JB, Marti LC, Aguiar MF, Silva HR, Silva GS, Nucci-da-Silva MP, DelBel EA, Gamarra LF. Therapeutics with SPION-labeled stem cells for the main diseases related to brain aging: a systematic review. Int. J. Nanomedicine 2014, 9, 3749–3770.10.2147/IJN.S65616Search in Google Scholar PubMed PubMed Central

[83] Yang CY, Hsiao JK, Tai MF, Chen ST, Cheng HY, Wang JL, Liu HM. Direct labeling of hMSC with SPIO: the long-term influence on toxicity, chondrogenic differentiation capacity, and intracellular distribution. Mol. Imaging Biol. 2011, 13, 443–451.10.1007/s11307-010-0360-7Search in Google Scholar PubMed

[84] Kim BYS, Rutka JT, Chan WCW. Nanomedicine. Review Article. N. Engl. J. Med. 2010, 363, 2434–2443.10.1056/NEJMra0912273Search in Google Scholar PubMed

[85] Johnson J, Kent T, Koda J, Peterson C, Rudge S, Tapolsky G. The MTC technology: a platform technology for the site-specific delivery of pharmaceutical agents. Eur. Cells Mater. 2002, 3, 12–15.Search in Google Scholar

Received: 2016-9-28
Accepted: 2017-1-24
Published Online: 2017-3-16
Published in Print: 2017-10-26

©2017 Walter de Gruyter GmbH, Berlin/Boston

This article is distributed under the terms of the Creative Commons Attribution Non-Commercial License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Downloaded on 20.5.2024 from https://www.degruyter.com/document/doi/10.1515/ntrev-2016-0101/html
Scroll to top button