Next Article in Journal
Water Soluble Usnic Acid-Polyacrylamide Complexes with Enhanced Antimicrobial Activity against Staphylococcus epidermidis
Next Article in Special Issue
Growth and Potential Damage of Human Bone-Derived Cells on Fresh and Aged Fullerene C60 Films
Previous Article in Journal
A Role for Protein Phosphatase 2A in Regulating p38 Mitogen Activated Protein Kinase Activation and Tumor Necrosis Factor-Alpha Expression during Influenza Virus Infection
Previous Article in Special Issue
Trends in Nanopharmaceutical Patents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Magnetic Drug Targeting Reduces the Chemotherapeutic Burden on Circulating Leukocytes

1
Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nuremberg, Ulmenweg 18, Erlangen 91054, Germany
2
Department of Otorhinolaryngology, Head and Neck Surgery, Section for Experimental Oncology and Nanomedicine (SEON), University Hospital Erlangen, Waldstrasse 1, Erlangen 91054, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors contributed equally to senior authorship.
Int. J. Mol. Sci. 2013, 14(4), 7341-7355; https://doi.org/10.3390/ijms14047341
Submission received: 3 December 2012 / Revised: 8 March 2013 / Accepted: 8 March 2013 / Published: 2 April 2013
(This article belongs to the Special Issue Bioactive Nanoparticles 2012)

Abstract

:
Magnetic drug targeting (MDT) improves the integrity of healthy tissues and cells during treatment with cytotoxic drugs. An anticancer drug is bound to superparamagnetic iron oxide nanoparticles (SPION), injected into the vascular supply of the tumor and directed into the tumor by means of an external magnetic field. In this study, we investigated the impact of SPION, mitoxantrone (MTO) and SPIONMTO on cell viability in vitro and the nonspecific uptake of MTO into circulating leukocytes in vivo. MDT was compared with conventional chemotherapy. MTO uptake and the impact on cell viability were assessed by flow cytometry in a Jurkat cell culture. In order to analyze MTO loading of circulating leukocytes in vivo, we treated tumor-bearing rabbits with MDT and conventional chemotherapy. In vitro experiments showed a dose-dependent MTO uptake and reduction in the viability and proliferation of Jurkat cells. MTO and SPIONMTO showed similar cytotoxic activity. Non-loaded SPION did not have any effect on cell viability in the concentrations tested. Compared with systemic administration in vivo, MDT employing SPIONMTO significantly decreased the chemotherapeutic load in circulating leukocytes. We demonstrated that MDT spares the immune system in comparison with conventional chemotherapy.

1. Introduction

Together with surgery and radiotherapy, chemotherapy is still a mainstay of cancer treatment today. Its main disadvantage lies in the severe systemic adverse effects inherent in this mode of therapy. In order to kill tumor cells directly, anticancer drugs are usually given at the maximum dose tolerated by the patient [1], which means that the dose necessary for complete tumor eradication is barely achieved. Targeted drug delivery to the tumor may overcome this dilemma [2], and nanotechnology provides new possibilities in this rapidly growing medical field, e.g., in otorhinolaryngology [3]. Magnetic drug targeting (MDT), a new and innovative approach, has been developed to concentrate anticancer drugs in the tumor and reduce leakage into healthy tissues and cells. Superparamagnetic iron oxide nanoparticles (SPION) are functionalized with a chemotherapeutic agent, injected into the vascular supply of the tumor, and directed into the tumor itself by means of an external magnetic field (Figure 1) [4].
Systemic distribution and adverse drug reactions are thereby decreased, at the same time as the local impact on the tumor is increased. Lower doses of the anticancer drug can be administered, as enhancement rates are 26 times higher with MDT than with conventional systemic chemotherapy [5]. Previous animal studies with an experimental VX-2 squamous cell carcinoma in rabbits successfully demonstrated MDT-induced tumor remission using nanoparticle-coupled mitoxantrone (MTO) [6]. MTO acts against a variety of hematologic and solid tumors by impairing DNA replication, transcription and repair [7], and may cause adverse effects such as nausea, fever, anemia, immunosuppression, and cardiotoxicity [8].
The immune system plays a key role in carcinogenesis and cancer therapy. The initiation, growth and metastasis of a tumor require “cancer immunoediting” [9]. In addition, more and more attention is being paid to the immunological aspects of cancer therapy: antibodies [10], vaccines [11], and the induction of anti-tumor immunity during radiotherapy and chemotherapy [12]. This pilot study aimed to quantify MTO in peripheral blood leukocytes (PBL) using its inherent fluorescent properties, since PBL are crucial components of the immune system. We observed that targeted delivery of MTO by MDT reduces the cytostatic burden on circulating leukocytes.

2. Results and Discussion

2.1. Analysis of SPION by Flow Cytometry

SPION can be detected by flow cytometry, as shown in Figure 2A. We found a clear distinction from Jurkat cells in size and granularity. Forward and side scatter analysis (FSC/SSC blots) showed SPION as a cloud of events two orders of magnitudes smaller and less granular than the cells. This difference allowed us to gate cells without contaminating SPION.
Since MTO is inherently fluorescent, it is possible to detect and quantify MTO bound to SPION (SPIONMTO) by flow cytometry. The flow cytometer uses separate fluorescence channels to detect emitted light: MTO was excited with 638 nm and the fluorescent emission was recorded with a 725 ± 10 nm band pass detector in channel 7 [FL-7]. Binding MTO to SPION resulted in a moderate increase in the SPION FSC and SSC signals (Figure 2B, upper panel) and caused an MTO dose-dependent shift in the FL-7 signal when compared with non-loaded SPION (Figure 2B, lower panel).
High performance liquid chromatography (HPLC) analysis of detached MTO is an established method of analyzing drug loading efficiency on SPION. We found a clear correlation (R2 = 0.94) between concentrations of SPIONMTO calculated by HPLC and the mean fluorescence intensity (MFI) values determined by flow cytometry. Dilution of SPIONMTO decreased the number of events gated by FSC/SSC and reduced the MFI (Figure 2C).

2.2. Uptake of MTO and SPIONMTO by Jurkat Cells

To gain information about the in vitro uptake of MTO in cell culture, we treated Jurkat cells with MTO and SPIONMTO in various concentrations and used flow cytometry to examine viable and dead Jurkat cells (Figure 3). Cells were classified as viable or dead, according to their morphological properties. SPION-treated cells, both viable and dead, exhibited only low autofluorescence. When we incubated viable cells with various concentrations of pure MTO, we found a dose-dependent increase in MFI [FL-7]. Whereas dead cells displayed a flat MFI signal over time, fluorescence increased in viable cells for at least 24 h. Most importantly, there was no relevant difference between cells incubated with pure MTO and those with SPIONMTO. The MFI signals were strongly dependent on the MTO concentration in both cases, demonstrating that MTO and SPIONMTO display similar cytotoxicity in vitro.

2.3. Proliferation of Jurkat Cells Treated with SPION, MTO and SPIONMTO

We then analyzed the proliferation rate of Jurkat cells treated with SPION, MTO and SPIONMTO using flow cytometry (Figure 4). Viable and dead cells were distinguished by their morphological properties (absolute values are shown).
Untreated cells show marked proliferation. Treatment with 0.001 μg/mL pure MTO or SPIONMTO affected cell proliferation slightly, while 0.005 μg/mL pure MTO or SPIONMTO had severe effects. MTO or SPIONMTO at a concentration of 0.01 μg/mL completely abolished proliferation. Incubation of Jurkat cells with pure MTO or SPIONMTO concentrations above 0.05 μg/mL resulted in cell death starting 24 h after treatment. In summary, MTO and SPIONMTO induced similar proliferation and cell death rates in vitro.

2.4. Apoptosis and Necrosis of Jurkat Cells Treated with SPION, MTO and SPIONMTO

In viable cells, phosphatidylserine (PS) is found mainly in the inner leaflet of the plasma membrane. During apoptosis, PS is exposed on the cell surface. Redistribution of PS in apoptosis and the loss of plasma membrane integrity in necrosis were monitored by annexinV-FITC/propidium iodide (AxV/PI) staining.
We incubated Jurkat cells with SPION, MTO and SPIONMTO in various concentrations for 48 h and analyzed the rate of viable (AxV−/PI−), apoptotic (AxV+/PI−) and necrotic cells (PI+) at several points in time (Figure 5). Mock-treated cells served as controls. Most untreated and SPION-treated cells maintained a viable phenotype throughout the experiment. Neither MTO nor SPIONMTO induced cell death in Jurkat cells at concentrations of MTO ≤ 0.01 μg/mL, whereas both induced apoptosis and secondary necrosis at concentrations of 0.05 μg/mL and above (Figure 5).

2.5. MDT Reduced the MTO Load of Circulating Leukocytes in Vivo

Exposing healthy tissues and cells to anticancer drugs is a crucial problem with conventional chemotherapy and causes multiple adverse reactions (e.g., leukopenia). To study this effect, we investigated MTO contamination in PBL during conventional chemotherapy and MDT in an in vivo rabbit tumor model. Using flow cytometry, we distinguished granulocytes, monocytes, and lymphocytes by their morphological features and quantified the MTO content as MFI [FL-7] (Figure 6A). As shown previously [5], MDT with a 20% therapeutic dose (TD20) of SPIONMTO reduces the chemotherapeutic burden on healthy tissue and enhances MTO in the tumor (Table 1, Figure 6B).
We detected increasing MTO concentrations in all leukocytes immediately after starting the infusion. MTO concentrations reached their peak after 10 min. MTO loading started to decline as soon as the MTO infusion finished (after 20 min). Comparing conventional chemotherapy in rabbits given a 100% therapeutic dose (TD100) of MTO (n = 4), TD10 MTO (n = 2), or Ringer’s solution (n = 2) intravenously, we found that MTO uptake in PBL clearly depended on the systemic dose administered. Granulocytes, monocytes, and lymphocytes displayed similar MTO profiles (Figure 6C).
Next, we studied the uptake of SPIONMTO into PBL. We compared the MTO content of PBL after intravenous injection of TD10 MTO with that found after intra-arterial injection of TD10 SPIONMTO. PBL had a reduced MTO load after SPIONMTO, but the difference was not statistically significant (Figure 6D).
An important step in MDT is focusing SPIONMTO on the target area with an external magnetic field. After injecting TD10 SPIONMTO into an artery directly supplying the tumor, MDT was applied to one group of rabbits (MDT), while the second group was not exposed to the magnetic field. As shown in Figure 6E, MTO uptake by PBL decreased even further. Compared with conventional chemotherapy, MDT resulted in lower MTO loading of circulating PBL—A difference that is statistically significant (p = 0.014, paired Student’s t-test).
Figure 6F summarizes the mean MTO loads of PBL in the different groups of rabbits. We compared TD100 MTO i.v. (n = 4), TD10 MTO i.v. (n = 2), TD10 SPIONMTO i.a. (n = 3), TD10 SPIONMTO i.a. MDT (n = 2) and controls treated with Ringer’s solution (n = 2). We demonstrated that PBL take up MTO in a dose-dependent manner, and that coupling MTO to SPION decreases the (unwanted) MTO load in PBLs, especially when MDT is applied. In summary, MDT significantly reduced the MTO burden of healthy PBL in comparison with conventional chemotherapy.

3. Experimental Section

3.1. Tumor Model

An experimental VX-2 tumor was implanted subcutaneously into the left hind limb of female New Zealand white rabbits (Charles River Laboratories, Sulzfeld, Germany), aged 18–22 weeks and weighing 3.1–3.9 kg. After four to six weeks, the tumor had grown to a size that made intervention necessary.

3.2. Chemotherapeutic Agent

Mitoxantrone (Mitoxantrone NC 2 mg/mL; NeoCorp AG, Weilheim, Germany) is an anticancer drug of the anthracenedione class. MTO acts by intercalating (hydrogen bonds) into DNA, inducing crosslinks and strand breaks [13]; it also inhibits the enzyme topoisomerase II [14].

3.3. Nanoparticles

The superparamagnetic iron oxide nanoparticles (SPION), composed of maghemite (Fe2O3) and magnetite (Fe3O4), were produced by SEON. Alkaline precipitation was followed by self-assembling functionalization with lauric acid and MTO [15]. The hydrodynamic diameter was set at approximately 100 nm using dynamic light scattering. MTO was bound to SPION (SPIONMTO) in an aqueous solution (ferrofluid). To analyze the MTO-content of the ferrofluid, MTO was extracted from 100 μL ferrofluid by the addition of 900 μL 1N HCl and incubation in a Thermomixer® comfort (Eppendorf AG, Hamburg, Germany) at 600 rpm and room temperature for 1 h. After centrifuging with a Minispin® plus (Eppendorf AG, Hamburg, Germany) at 14,500 rpm for 10 min, the supernatant was measured with HPLC, using a Waters Alliance model consisting of a separation module (2695 series), a dual wavelength absorbance detector (2487 series), and a 3.0 × 100 mm X-Bridge phenyl column (Waters, Germany). The mobile phase consisted of formiate buffer (pH 3.0) and methanol (80:20 v/v) [16].

3.4. Mode of Administration

Therapy was given as a single treatment cycle. We administered conventional chemotherapy intravenously into an ear vein, while MDT required intra-arterial access. For this purpose, the rabbits were taken to an animal operation theatre and the femoral artery prepared under sterile conditions. Pure MTO and drug-loaded SPIONs, as well as Ringer’s solution, were injected via a 24 G i.v. catheter (B. Braun, Melsungen, Germany) over 20 min. In one group, an electromagnet (Siemens Healthcare, Erlangen, Germany) with a magnetic field gradient of 72 T/m located directly beneath the pole tip [17] attracted the drug-loaded nanoparticles to the tumor during the 20 min of drug administration and for a further 15 min afterwards. Blood was taken before administration (−60 min), during the infusion (−10 min, 0 min, +10 min), and afterwards (+20 min, +30 min, +40 min, +50 min, +60 min, +120 min). For whole blood analyses, the erythrocytes in rabbit EDTA-blood were subjected to hypotonic lysis twice before flow cytometry.

3.5. Cells and Culture Conditions

To investigate the effects of SPION, MTO and SPIONMTO on mammalian cells, we used the non-adherent human T cell leukemia cell line Jurkat (ACC 282, DSMZ, Braunschweig, Germany). Cell culture was performed at 37 °C and 5.5% CO2 in R10 medium (RPMI 1640 medium supplemented with 10% FCS, 1% glutamine, 1% penicillin-streptomycin (all from Life Technologies GmbH, Darmstadt, Germany), and 1% HEPES (10 mM, pH 7.2) (Merck KGaA, Darmstadt, Germany)). Cells were counted and adjusted to a density of 0.5 × 106 cells/mL in R10 medium for these experiments.

3.6. Measurement of Cellular Morphology

Dying cells change in morphology, and this can be detected by scatter alterations in flow cytometry [18]. Apoptosis is indicated by cell shrinkage, increased cytoplasmic granularity and vacuolization. Apoptotic cells are therefore detected as a population with decreased FSC and increased SSC [19].

3.7. Detection of Exposed Cell Phosphatidylserine and Analysis of Membrane Integrity

Recombinant chicken annexin V (AxV) (responsif GmbH, Erlangen, Germany) was labeled with fluorescein isothiocyanate (FITC) according to the manufacturer’s instructions (Sigma-Aldrich GmbH, Taufkirchen, Germany). AxV-FITC binds to exposed PS on the membranes of cells undergoing apoptosis. Before flow cytometry, 100 μL cell suspension was incubated with 400 μL freshly prepared staining solution, containing 1 μg/mL AxV-FITC and 20 μg/mL propidium iodide (PI) (Sigma-Aldrich GmbH, Taufkirchen, Germany) in Ringer’s solution, for 30 min at 4 °C. The PI was added to differentiate between apoptotic (AxV positive, PI negative), necrotic (AxV positive, PI positive) and viable (AxV negative, PI negative) cells [20].

3.8. Flow Cytometry

Flow cytometry was performed with a Gallios cytofluorometer™ (Beckman Coulter, Fullerton, CA, USA). The excitation wavelength for FITC and PI was 488 nm; FITC fluorescence was recorded by the FL-1 sensor (525/38 nm band pass), PI fluorescence by the FL-3 sensor (620/30 nm band pass). MTO fluorescence was excited at 638 nm and recorded by the FL-7 sensor (725/20 nm band pass). Electronic compensation was used to eliminate any fluorescence bleed-through. Data were analyzed with Kaluza™ software (Beckman Coulter, Fullerton, CA, USA).

4. Conclusions

MDT reduces the chemotherapeutic burden of circulating leukocytes. In our experiments, we used SPIONMTO with a high magnetic field gradient of 72 T/m, and compared this approach with conventional chemotherapy. As previously demonstrated using MDT (TD20 SPIONMTO), we decreased the chemotherapeutic burden on healthy tissue and enhanced MTO in the tumor (Table 1, Figure 6B) [5].
Flow cytometry is the most appropriate technique for precise analysis at the single cell level. MTO and the closely related anthracycline antibiotics, daunorubicin, doxorubicin and idarubicin (data not shown) are clearly detectable by this method because of their inherent fluorescence [21]. Over the years, the technique has been improved [22]. In this study, we used flow cytometry to monitor the incorporation of MTO into non-adherent cells in cell cultures in vitro as well as into blood cells in vivo.
Using MFI [FL-7], it was also possible to determine the MTO loading efficiency of SPION. Although flow cytometry cannot detect a single SPION because of its small size, we established a measurement strategy to quantify MTO loading under defined conditions. Depending on the amount of MTO bound to the particles, SPION had a hydrodynamic diameter of approximately 100 nm, as determined by dynamic light scattering. Under defined conditions of SPION content, dilution, and flow rate, quantifying MTO bound to SPION in this way can compete with the established HPLC method used to determine loading efficiency [16].
Using flow cytometry, we found a dose-dependent effect of MTO and SPIONMTO on the proliferation and viability of non-adherent human Jurkat cells in vitro. The two compounds had very similar effects, confirming equal cytotoxic activity of these substances if they are not differentially routed by MDT. This finding confirmed earlier results of real-time cell analysis and lactate dehydrogenase release in adherent cancer cells [23]. As shown in previous studies, low and intermediate concentrations of non-loaded SPIONs did not affect cell viability [23]. In contrast to earlier experiments performed with MCF-7 cells (human breast carcinoma) [23], much lower MTO concentrations induced Jurkat cell death. This confirms the high lymphoblast susceptibility to MTO. AxV/PI staining identified MTO-induced cell death as early apoptosis or the later response of secondary necrosis.
Preservation of the immune system during cancer therapy is extremely important and the cytotoxic load in PBL can be used as a surrogate marker for the immune system burden. Unwanted cytotoxic drug uptake by PBL is a major obstacle in MTO dose escalation. Ballestrero therefore developed a treatment strategy compatible with the rescue of blood progenitor cells. He achieved an acceptable hematologic toxicity while escalating the dose of MTO up to 90 mg/m2[8].
During the past few decades, evidence emerged that low-dose chemotherapy has immunomodulatory or immunostimulatory effects [24]. Interestingly enough, it was the anthracyclines in particular which were reported to induce a specific anticancer immune response [2527]. After the injection of mitoxantrone-treated B16-F1 cells into mice, their splenocytes displayed an increased cytolytic effect on B16-F1 cells [28].
Nanoparticles reportedly influence the immune system, causing either immunostimulation or immunosuppression [29]. CoCr nanoparticles, for example, have been found to inhibit T-cell proliferation, which may explain the fact that patients with metal-on-metal implants have reduced CD8+ T-cell populations [30]. Like chemotherapeutic agents, Fe3O4 (magnetite) magnetic nanoparticles, especially in low-doses, were found to affect immune function in mice [31]. In our in vivo and in vitro settings, SPION displayed no detectable cytotoxicity.
In comparison with conventional routes of administration, spillover of the chemotherapeutic agent MTO into healthy circulating PBL is significantly reduced by MDT-based treatment regimens. MDT reduces the major adverse effects of chemotherapy and preserves PBL, taken as a surrogate population of the immune system. We conclude that MDT saves drug and spares the immune system from cytotoxicity.

Acknowledgments

This work was supported by the DFG (AL 552/3-3), by BMBF-Spitzencluster (01EX1012B), by the Else Kröner-Fresenius-Stiftung, Bad Homburg vor der Höhe, Germany, by SFB 643 TP B5, by the Emerging Fields Initiative of the Friedrich-Alexander-University Erlangen-Nuremberg, by the K&R Wucherpfennigstiftung, and by the Interdisciplinary Center of Clinical Research at the University Hospital Erlangen.

Conflict of Interest

The authors declare no conflict of interest.

References

  1. Shimizu, K; Oku, N. Cancer anti-angiogenic therapy. Biol. Pharm. Bull. 2004, 27, 599–605. [Google Scholar]
  2. Tietze, R; Lyer, S; Durr, S; Alexiou, C. Nanoparticles for cancer therapy using magnetic forces. Nanomedicine (London, England) 2012, 7, 447–457. [Google Scholar]
  3. Durr, S; Tietze, R; Lyer, S; Alexiou, C. Nanomedicine in otorhinolaryngology—Future prospects. Laryngorhinootologie 2012, 91, 6–12. [Google Scholar]
  4. Alexiou, C; Tietze, R; Schreiber, E; Lyer, S. Nanomedicine: Magnetic nanoparticles for drug delivery and hyperthermia—new chances for cancer therapy. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2010, 53, 839–845. [Google Scholar]
  5. Alexiou, C; Jurgons, R; Schmid, R; Erhardt, W; Parak, F; Bergemann, C; Iro, H. Magnetic drug targeting—a new approach in locoregional tumor therapy with chemotherapeutic agents. Experimental animal studies. HNO 2005, 53, 618–622. [Google Scholar]
  6. Alexiou, C; Arnold, W; Klein, R.J; Parak, F.G; Hulin, P; Bergemann, C; Erhardt, W; Wagenpfeil, S; Lubbe, A.S. Locoregional cancer treatment with magnetic drug targeting. Cancer Res. 2000, 60, 6641–6648. [Google Scholar]
  7. Fox, M.E; Smith, P.J. Long-term inhibition of DNA synthesis and the persistence of trapped topoisomerase II complexes in determining the toxicity of the antitumor DNA intercalators mAMSA and mitoxantrone. Cancer Res. 1990, 50, 5813–5818. [Google Scholar]
  8. Ballestrero, A; Ferrando, F; Garuti, A; Basta, P; Gonella, R; Esposito, M; Vannozzi, M.O; Sorice, G; Friedman, D; Puglisi, M.; et al. High-dose mitoxantrone with peripheral blood progenitor cell rescue: Toxicity, pharmacokinetics and implications for dosage and schedule. Br. J. Cancer 1997, 76, 797–804. [Google Scholar]
  9. Dunn, G.P; Bruce, A.T; Ikeda, H; Old, L.J; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar]
  10. Scott, A.M; Wolchok, J.D; Old, L.J. Antibody therapy of cancer. Nat. Rev. Cancer 2012, 12, 278–287. [Google Scholar]
  11. Palucka, K; Ueno, H; Banchereau, J. Recent developments in cancer vaccines. J. Immunol. 2011, 186, 1325–1331. [Google Scholar]
  12. Frey, B; Rubner, Y; Wunderlich, R; Weiss, E.M; Pockley, A.G; Fietkau, R; Gaipl, U.S. Induction of abscopal anti-tumor immunity and immunogenic tumor cell death by ionizing irradiation - implications for cancer therapies. Curr. Med. Chem. 2012, 19, 1751–1764. [Google Scholar]
  13. Kapuscinski, J; Darzynkiewicz, Z; Traganos, F; Melamed, M.R. Interactions of a new antitumor agent, 1,4-dihydroxy-5,8-bis[[2-[(2-hydroxyethyl)amino]-ethyl]amino]-9,10-anthracenedion e, with nucleic acids. Biochem. Pharmacol. 1981, 30, 231–240. [Google Scholar]
  14. Frank, P; Novak, R.F. Mitoxantrone and bisantrene inhibition of platelet aggregation and prostaglandin E2 production in vitro. Biochem. Pharmacol. 1985, 34, 3609–3614. [Google Scholar]
  15. Khalafalla, S.E; Reimers, G.W. Preparation of dilution-stable aqueous magnetic fluids. IEEE Trans. Magn. 1980, 16, 178–183. [Google Scholar]
  16. Tietze, R; Schreiber, E; Lyer, S; Alexiou, C. Mitoxantrone loaded superparamagnetic nanoparticles for drug targeting: A versatile and sensitive method for quantification of drug enrichment in rabbit tissues using HPLC-UV. J. Biomed. Biotechnol. 2010, 597304:1–597304:8. [Google Scholar]
  17. Alexiou, C; Diehl, D; Henninger, P; Iro, H; Rockelein, R; Schmidt, W; Weber, H. A high field gradient magnet for magnetic drug targeting. IEEE Trans Appl. Supercon. 2006, 16, 1527–1530. [Google Scholar]
  18. Elstein, K.H; Zucker, R.M. Comparison of cellular and nuclear flow cytometric techniques for discriminating apoptotic subpopulations. Exp. Cell Res. 1994, 211, 322–331. [Google Scholar]
  19. Hagenhofer, M; Germaier, H; Hohenadl, C; Rohwer, P; Kalden, J.R; Herrmann, M. UV-B irradiated cell lines execute programmed cell death in various forms. Apoptosis 1998, 3, 123–132. [Google Scholar]
  20. Vermes, I; Haanen, C; Steffens-Nakken, H; Reutelingsperger, C. A novel assay for apoptosis. Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled Annexin V. J. Immunol. Methods 1995, 184, 39–51. [Google Scholar]
  21. Smith, P.J; Sykes, H.R; Fox, M.E; Furlong, I.J. Subcellular distribution of the anticancer drug mitoxantrone in human and drug-resistant murine cells analyzed by flow cytometry and confocal microscopy and its relationship to the induction of DNA damage. Cancer Res. 1992, 52, 4000–4008. [Google Scholar]
  22. Chan, A; Weilbach, F.X; Toyka, K.V; Gold, R. Mitoxantrone induces cell death in peripheral blood leucocytes of multiple sclerosis patients. Clin. Exp. Immunol. 2005, 139, 152–158. [Google Scholar]
  23. Durr, S; Lyer, S; Mann, J; Janko, C; Tietze, R; Schreiber, E; Herrmann, M; Alexiou, C. Real-time cell analysis of human cancer cell lines after chemotherapy with functionalized magnetic nanoparticles. Anticancer Res. 2012, 32, 1983–1989. [Google Scholar]
  24. Apetoh, L; Ghiringhelli, F; Tesniere, A; Obeid, M; Ortiz, C; Criollo, A; Mignot, G; Maiuri, M.C; Ullrich, E; Saulnier, P.; et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 2007, 13, 1050–1059. [Google Scholar]
  25. Zitvogel, L; Apetoh, L; Ghiringhelli, F; Andre, F; Tesniere, A; Kroemer, G. The anticancer immune response: Indispensable for therapeutic success? J. Clin. Invest. 2008, 118, 1991–2001. [Google Scholar]
  26. Apetoh, L; Mignot, G; Panaretakis, T; Kroemer, G; Zitvogel, L. Immunogenicity of anthracyclines: Moving towards more personalized medicine. Trends Mol. Med. 2008, 14, 141–151. [Google Scholar]
  27. Galluzzi, L; Senovilla, L; Zitvogel, L; Kroemer, G. The secret ally: Immunostimulation by anticancer drugs. Nat. Rev. Drug Discov. 2012, 11, 215–233. [Google Scholar]
  28. Cao, C; Han, Y; Ren, Y; Wang, Y. Mitoxantrone-mediated apoptotic B16-F1 cells induce specific anti-tumor immune response. Cell Mol. Immunol. 2009, 6, 469–475. [Google Scholar]
  29. Zolnik, B.S; Gonzalez-Fernandez, A; Sadrieh, N; Dobrovolskaia, M.A. Nanoparticles and the immune system. Endocrinology 2010, 151, 458–465. [Google Scholar]
  30. Ogunwale, B; Schmidt-Ott, A; Meek, R.M; Brewer, J.M. Investigating the immunologic effects of CoCr nanoparticles. Clin. Orthop. Relat. Res. 2009, 467, 3010–3016. [Google Scholar]
  31. Chen, B.A; Jin, N; Wang, J; Ding, J; Gao, C; Cheng, J; Xia, G; Gao, F; Zhou, Y; Chen, Y.; et al. The effect of magnetic nanoparticles of Fe3O4 on immune function in normal ICR mice. Int. J. Nanomed. 2010, 5, 593–599. [Google Scholar]
Figure 1. Principles of magnetic drug targeting. Superparamagnetic iron oxide nanoparticles (SPION) coated with lauric acid are loaded with mitoxantrone (SPIONMTO). The solution is injected into the vascular supply of the tumor and accumulated at the intended site by means of an external magnetic field.
Figure 1. Principles of magnetic drug targeting. Superparamagnetic iron oxide nanoparticles (SPION) coated with lauric acid are loaded with mitoxantrone (SPIONMTO). The solution is injected into the vascular supply of the tumor and accumulated at the intended site by means of an external magnetic field.
Ijms 14 07341f1
Figure 2. Analysis of superparamagnetic iron oxide nanoparticles (SPION) by flow cytometry. (A) Morphological analysis of SPION. Because of their small size, SPION can be clearly distinguished from Jurkat cells by forward and side scatter analysis (FSC/SSC) in flow cytometry. 0.5 × 106 Jurkat cells in 500 μL R10 medium and 10 μL of SPION (5.7 μg/μL Fe) were analyzed at a low flow rate setting; (B) Mitoxantrone-loaded SPION (SPIONMTO) show higher MFI [FL-7] values than non-loaded SPION. MTO coupling results in an increase of FSC and SSC (upper panel). The MFI of 10 μL pure SPION and 10 μL SPION loaded with various amounts of MTO in 500 μL phosphate buffered saline (PBS)/20% fetal calf serum (FCS) are shown, and clearly demonstrate that the MFI [FL-7] depends on the MTO content (lower panel). Measurements were performed at a medium flow rate setting; (C) Analysis of MTO loading on SPION by high performance liquid chromatography (HPLC) and flow cytometry. 10 μL SPIONMTO were diluted in 500 μL PBS/20% FCS and the measurement was performed at a medium flow rate setting. There was a good correlation between the quantification of MTO by HPLC and the flow cytometry of SPIONMTO (R2 = 0.94).
Figure 2. Analysis of superparamagnetic iron oxide nanoparticles (SPION) by flow cytometry. (A) Morphological analysis of SPION. Because of their small size, SPION can be clearly distinguished from Jurkat cells by forward and side scatter analysis (FSC/SSC) in flow cytometry. 0.5 × 106 Jurkat cells in 500 μL R10 medium and 10 μL of SPION (5.7 μg/μL Fe) were analyzed at a low flow rate setting; (B) Mitoxantrone-loaded SPION (SPIONMTO) show higher MFI [FL-7] values than non-loaded SPION. MTO coupling results in an increase of FSC and SSC (upper panel). The MFI of 10 μL pure SPION and 10 μL SPION loaded with various amounts of MTO in 500 μL phosphate buffered saline (PBS)/20% fetal calf serum (FCS) are shown, and clearly demonstrate that the MFI [FL-7] depends on the MTO content (lower panel). Measurements were performed at a medium flow rate setting; (C) Analysis of MTO loading on SPION by high performance liquid chromatography (HPLC) and flow cytometry. 10 μL SPIONMTO were diluted in 500 μL PBS/20% FCS and the measurement was performed at a medium flow rate setting. There was a good correlation between the quantification of MTO by HPLC and the flow cytometry of SPIONMTO (R2 = 0.94).
Ijms 14 07341f2
Figure 3. Uptake of MTO from superparamagnetic iron oxide nanoparticles (SPION), mitoxantrone (MTO), and MTO-loaded SPION (SPIONMTO) by Jurkat cells. Cells were classed as viable or dead, depending on their morphological properties. (A) SPION-treated cells showed only autofluorescence; (B) Viable cells treated with MTO showed an increase in MFI [FL-7] with time, while no increase in MFI occurred in dead cells; (C) SPIONMTO-treated cells behaved similarly to cells incubated with pure MTO. In all cases, the MFI [FL-7] was dependent on MTO concentrations. Figures show the mean values of duplicates with standard deviations.
Figure 3. Uptake of MTO from superparamagnetic iron oxide nanoparticles (SPION), mitoxantrone (MTO), and MTO-loaded SPION (SPIONMTO) by Jurkat cells. Cells were classed as viable or dead, depending on their morphological properties. (A) SPION-treated cells showed only autofluorescence; (B) Viable cells treated with MTO showed an increase in MFI [FL-7] with time, while no increase in MFI occurred in dead cells; (C) SPIONMTO-treated cells behaved similarly to cells incubated with pure MTO. In all cases, the MFI [FL-7] was dependent on MTO concentrations. Figures show the mean values of duplicates with standard deviations.
Ijms 14 07341f3
Figure 4. Proliferation of Jurkat cells treated with superparamagnetic iron oxide nanoparticles (SPION), mitoxantrone (MTO), and MTO-loaded SPION (SPIONMTO). Viable and dead cells were distinguished according to their morphological properties (absolute values shown). (A) Pure SPION did not induce cell death in the tested concentrations; (B) Depending on the pure MTO concentration, cells showed reduced proliferation and an increased cell death rate; (C) SPIONMTO demonstrated similar behavior to MTO. Figures show the mean values of duplicates with standard deviations.
Figure 4. Proliferation of Jurkat cells treated with superparamagnetic iron oxide nanoparticles (SPION), mitoxantrone (MTO), and MTO-loaded SPION (SPIONMTO). Viable and dead cells were distinguished according to their morphological properties (absolute values shown). (A) Pure SPION did not induce cell death in the tested concentrations; (B) Depending on the pure MTO concentration, cells showed reduced proliferation and an increased cell death rate; (C) SPIONMTO demonstrated similar behavior to MTO. Figures show the mean values of duplicates with standard deviations.
Ijms 14 07341f4
Figure 5. Cell death of Jurkat cells after treatment with superparamagnetic iron oxide nanoparticles (SPION), mitoxantrone (MTO), and MTO-loaded SPION (SPIONMTO). Cell death was determined by annexinV-FITC/propidium iodide (AxV/PI) staining. AxV−/PI−, AxV+/PI−, and PI+ cells were considered viable, apoptotic and necrotic, respectively. Cells were incubated with various concentrations of SPION, MTO and SPIONMTO and then stained with AxV/PI at various points in time. (A) Mock-treated cells served as control; (B) Non-loaded SPION did not induce cell death, whereas (C) MTO and (D) SPIONMTO induced apoptosis in a dose-dependent manner, with secondary necrosis after prolonged incubation. Figures show the mean values of duplicates with standard deviations.
Figure 5. Cell death of Jurkat cells after treatment with superparamagnetic iron oxide nanoparticles (SPION), mitoxantrone (MTO), and MTO-loaded SPION (SPIONMTO). Cell death was determined by annexinV-FITC/propidium iodide (AxV/PI) staining. AxV−/PI−, AxV+/PI−, and PI+ cells were considered viable, apoptotic and necrotic, respectively. Cells were incubated with various concentrations of SPION, MTO and SPIONMTO and then stained with AxV/PI at various points in time. (A) Mock-treated cells served as control; (B) Non-loaded SPION did not induce cell death, whereas (C) MTO and (D) SPIONMTO induced apoptosis in a dose-dependent manner, with secondary necrosis after prolonged incubation. Figures show the mean values of duplicates with standard deviations.
Ijms 14 07341f5
Figure 6. Magnetic drug targeting (MDT) prevents exposure of healthy circulating leukocytes to mitoxantrone (MTO). Blood was taken before (−60 min), during (−10 min, 0 min, +10 min), and after (+20 min, +30 min, +40 min, +50 min, +60 min, +120 min) administration of the drug. (A) Flow cytometry of rabbit blood. Before flow cytometry, erythrocytes were lysed by hypotonic treatment in rabbit whole blood; granulocytes, monocytes and lymphocytes were distinguished by their morphological features (FSC: size; SSC: granularity). MTO uptake into blood cells was monitored by flow cytometry employing MFI [FL-7]; (B) After MDT, MTO accumulates primarily in the tumor, with MTO concentrations in the central tumor and neighboring lymph nodes being similar. The load was reduced in all other organs; (C) MTO incorporated into circulating leukocytes depended on the systemic dose administered. TD100 MTO (n = 4), TD10 MTO (n = 2), and pure Ringer’s solution (n = 2) were injected intravenously; (D) Coupling MTO to superparamagnetic iron oxide nanoparticles (SPION) and injecting the resultant SPIONMTO into an artery supplying the tumor reduces the amount of MTO in circulating leukocytes. TD10 MTO (n = 2) was given intravenously and TD10 SPIONMTO (n = 3) injected intra-arterially; (E) Routing SPIONMTO into the tumor region with an external magnetic field further reduced circulating leukocyte exposure to MTO. TD10 SPIONMTO (n = 5) was administered directly into the arterial supply of the tumor. One group (n = 2) was exposed to an external magnetic field (MDT); (F) Data on MTO uptake show that MDT significantly reduced the MTO burden of circulating leukocytes: conventional chemotherapy with TD100 MTO i.v. (n = 4), TD10 MTO i.v. (n = 2), TD10 SPIONMTO i.a. (n = 3), TD10 SPIONMTO i.a. with exposure to an external magnetic field (n = 2), and controls treated with Ringer’s solution (n = 2).
Figure 6. Magnetic drug targeting (MDT) prevents exposure of healthy circulating leukocytes to mitoxantrone (MTO). Blood was taken before (−60 min), during (−10 min, 0 min, +10 min), and after (+20 min, +30 min, +40 min, +50 min, +60 min, +120 min) administration of the drug. (A) Flow cytometry of rabbit blood. Before flow cytometry, erythrocytes were lysed by hypotonic treatment in rabbit whole blood; granulocytes, monocytes and lymphocytes were distinguished by their morphological features (FSC: size; SSC: granularity). MTO uptake into blood cells was monitored by flow cytometry employing MFI [FL-7]; (B) After MDT, MTO accumulates primarily in the tumor, with MTO concentrations in the central tumor and neighboring lymph nodes being similar. The load was reduced in all other organs; (C) MTO incorporated into circulating leukocytes depended on the systemic dose administered. TD100 MTO (n = 4), TD10 MTO (n = 2), and pure Ringer’s solution (n = 2) were injected intravenously; (D) Coupling MTO to superparamagnetic iron oxide nanoparticles (SPION) and injecting the resultant SPIONMTO into an artery supplying the tumor reduces the amount of MTO in circulating leukocytes. TD10 MTO (n = 2) was given intravenously and TD10 SPIONMTO (n = 3) injected intra-arterially; (E) Routing SPIONMTO into the tumor region with an external magnetic field further reduced circulating leukocyte exposure to MTO. TD10 SPIONMTO (n = 5) was administered directly into the arterial supply of the tumor. One group (n = 2) was exposed to an external magnetic field (MDT); (F) Data on MTO uptake show that MDT significantly reduced the MTO burden of circulating leukocytes: conventional chemotherapy with TD100 MTO i.v. (n = 4), TD10 MTO i.v. (n = 2), TD10 SPIONMTO i.a. (n = 3), TD10 SPIONMTO i.a. with exposure to an external magnetic field (n = 2), and controls treated with Ringer’s solution (n = 2).
Ijms 14 07341f6aIjms 14 07341f6b
Table 1. Distribution of MTO in organs (μg/g tissue) after conventional chemotherapy (TD100 MTO) and magnetic drug targeting (TD20 SPIONMTO) [5].
Table 1. Distribution of MTO in organs (μg/g tissue) after conventional chemotherapy (TD100 MTO) and magnetic drug targeting (TD20 SPIONMTO) [5].
MTO content (μg/g tissue) after treatment

OrganConventional chemotherapy (TD100 MTO)Magnetic drug targeting (TD20 SPIONMTO)
Tumor0.580.50
Peritumoral0.2520.93
Ipsilateral lymph node1.031.05
Heart1.750.20
Lung1.660.26
Liver0.470.22
Gall bladder4.381.75
Kidney7.290.62
Spleen2.650.28

Share and Cite

MDPI and ACS Style

Janko, C.; Dürr, S.; Munoz, L.E.; Lyer, S.; Chaurio, R.; Tietze, R.; Löhneysen, S.V.; Schorn, C.; Herrmann, M.; Alexiou, C. Magnetic Drug Targeting Reduces the Chemotherapeutic Burden on Circulating Leukocytes. Int. J. Mol. Sci. 2013, 14, 7341-7355. https://doi.org/10.3390/ijms14047341

AMA Style

Janko C, Dürr S, Munoz LE, Lyer S, Chaurio R, Tietze R, Löhneysen SV, Schorn C, Herrmann M, Alexiou C. Magnetic Drug Targeting Reduces the Chemotherapeutic Burden on Circulating Leukocytes. International Journal of Molecular Sciences. 2013; 14(4):7341-7355. https://doi.org/10.3390/ijms14047341

Chicago/Turabian Style

Janko, Christina, Stephan Dürr, Luis E. Munoz, Stefan Lyer, Ricardo Chaurio, Rainer Tietze, Sarah Von Löhneysen, Christine Schorn, Martin Herrmann, and Christoph Alexiou. 2013. "Magnetic Drug Targeting Reduces the Chemotherapeutic Burden on Circulating Leukocytes" International Journal of Molecular Sciences 14, no. 4: 7341-7355. https://doi.org/10.3390/ijms14047341

Article Metrics

Back to TopTop