Elsevier

Biomaterials

Volume 127, May 2017, Pages 25-35
Biomaterials

Active targeting theranostic iron oxide nanoparticles for MRI and magnetic resonance-guided focused ultrasound ablation of lung cancer

https://doi.org/10.1016/j.biomaterials.2017.02.037Get rights and content

Abstract

Despite its great promise in non-invasive treatment of cancers, magnetic resonance-guided focused ultrasound surgery (MRgFUS) is currently limited by the insensitivity of magnetic resonance imaging (MRI) for visualization of small tumors, low efficiency of in vivo ultrasonic energy deposition, and damage to surrounding tissues. We hereby report the development of an active targeting nano-sized theranostic superparamagnetic iron oxide (SPIO) platform for significantly increasing the imaging sensitivity and energy deposition efficiency using a clinical MRgFUS system. The surfaces of these PEGylated SPIO nanoparticles (NPs) were decorated with anti-EGFR (epidermal growth factor receptor) monoclonal antibodies (mAb) for targeted delivery to lung cancer with EGFR overexpression. The potential of these targeted nano-theranostic agents for MRI and MRgFUS ablation was evaluated in vitro and in vivo in a rat xenograft model of human lung cancer (H460). Compared with nontargeting PEGylated SPIO NPs, the anti-EGFR mAb targeted PEGylated SPIO NPs demonstrated better targeting capability to H460 tumor cells and greatly improved the MRI contrast at the tumor site. Meanwhile, this study showed that the targeting NPs, as synergistic agents, could significantly enhance the efficiency for in vivo ultrasonic energy deposition in MRgFUS. Moreover, we demonstrated that a series of MR methods including T2-weighted image (T2WI), T1-weighted image (T1WI), diffusion-weighted imaging (DWI) and contrast-enhanced T1WI imaging, could be utilized to noninvasively and conveniently monitor the therapeutic efficacy in rat models by MRgFUS.

Introduction

Magnetic resonance-guided focused ultrasound surgery (MRgFUS), as a promising non-invasive ultrasound thermal treatment for soft tissue lesions, has been utilized in the treatment of prostate, kidney, and liver cancer [1], [2], [3]. In particular, there is precise control of beam direction and ongoing feedback is provided to detail temperature changes at and around the treated tissue in MRgFUS. The main advantages of MRgFUS are that it is non-invasive, able to provide real-time, three-dimensional imaging, and closed-loop MR feedback. Magnetic resonance imaging (MRI) is a clinically useful diagnostic and tumor detection technique and can provide real-time temperature monitoring in MRgFUS. MRgFUS ablation usually involves the use of gradient-echo MR imaging for real-time temperature monitoring, based on spin-lattice (T1) relaxation time and temperature sensitivity of the proton resonance frequency [4]. The tumor ablation efficiency of MRgFUS highly depends on its capacity to deposit energy in tissue. However, when shifting from an in vitro environment to in vivo tissue, there is considerable attenuation of ultrasonic energy emitted by the MR-guided ultrasound transducer [5]. High ultrasound power must be employed to improve the therapeutic efficiency and guidance of conventional MRgFUS in order to achieve reduction effects on a deeper lesion. This high ultrasound power could cause severe side effects, such as skin burns, edema, perforation of intestines, and injury of peripheral nerve surrounding the tumor [6]. Furthermore, the set current is not sensitive enough to visualize small lesions using conventional MRgFUS settings [6], [7], [8], [9].

Due to their excellent biocompatibility and magnetic properties, superparamagnetic iron oxide (SPIO) NPs have been used extensively for drug delivery, MRI probes, and tumor thermotherapy [7], [8], [9], [10], [11], [12], [13], [14]. Previous studies have shown that dipole relaxation induced by an alternating magnetic field can contribute to magnetic hyperthermia of SPIO NPs. During irradiations of near-infrared (NIR) laser light and high intensity focused ultrasound (HIFU) treatment, SPIO NPs were used to induce the thermal therapy effect in tumor tissue [6], [15], [16], [17], [18], [19]. Recently, superparamagnetic iron oxide-polymer composite microcapsules (mean diameter, 587 nm & 885.6 nm) have been proposed for MRgFUS [5], [17]. These microcapsules were demonstrated with the ability to enhance ultrasonic wave absorption and energy deposition in the targeted tissue, thereby enhancing the tumor-ablative effects of MRgFUS [17].

In the present study, we propose to develop an active targeting, nanosized theranostic SPIO nanoplatform to improve the MRI sensitivity and tumor-ablative efficacy of a clinical MRgFUS system. These SPIO nanoparticles were coated with a layer of polyethylene glycol (PEG) to improve their biocompatibility [14], [20]. Epidermal growth factor receptor (EGFR) is a tyrosine kinase cellular transmembrane receptor and is linked to aberrant survival and poor prognosis. It is overexpressed in various epithelial tumors, such as non-small cell lung, kidney, breast, and head-and-neck squamous cell carcinoma [21], [22]. In this study, the surface of these PEGylated SPIO nanoparticles was further decorated with high affinity anti-EGFR monoclonal antibody (Cetuximab) to form a nanocomposite (anti-EGFR-PEG-SPIO) for targeted delivery to EGFR overexpressing H460 lung cancer. The anti-EGFR-PEG-SPIO was physicochemically characterized. Their targeting capability, MRI contrast enhancement and cytotoxicity were studied in H460 lung cancer cells as well as in nude rat models bearing H460 lung cancer xenografts. The capability of anti-EGFR-PEG-SPIO for synergistic MRgFUS treatment of cancer and the applications of a series of MRI approaches for non-invasive monitoring treatment response were demonstrated using nude rat lung tumor models (Scheme shown in Fig. 1a). To the best of our knowledge, this study represents the first evaluation of an active targeting and nano-sized SPIO platform for the enhancement of imaging sensitivity and tumor-ablative efficacy in MRgFUS.

Section snippets

Preparation of Fe3O4 nanoparticles

Iron oxide nanoparticles were produced according to a reported procedure [23], [24]. In brief, Fe(acac)3 (2.12 g, 6 mmol), HOOC-PEG-COOH (0.12 M, M.W. = 2000), and oleylamine (7.90 mL, 24 mmol) were mixed in 100 mL of diphenyl oxide solution. The above solution was heated to reflux under a nitrogen environment. Next, the iron solution was stirred at 400 rpm. In order to monitor the particle growth and formation, different aliquots were extracted during the heating process. Upon addition of a

Synthesis and characterization of anti-EGFR-PEG-SPIO

We were able to successfully synthesize anti-EGFR-PEG-SPIO, and utilize TEM, DLS, XRD, and hysteresis loop to characterize their size, composition and magnetic properties. TEM image of PEGylated SPIO before ligand decoration showed a uniform size distribution with a mean diameter of about 9.2 nm (Fig. S1a). DLS results demonstrated that the hydrodynamic diameter was about 38.5 nm for PEGyloated SPIO and 45.7 nm for EGFR-targeted NPs, respectively (Fig. 1b). Based on the XRD results, we

Discussion

The therapeutic efficacy of MRgFUS is relatively low for large volume tumor masses or deep lesions. With an increase of depth in tumor tissues, the ultrasound energy is attenuated exponentially, and therefore high ultrasound power must be employed to obtain the desired therapeutic efficacy. Such high acoustic energy could cause damage to normal surrounding tissues in the process of ultrasound, resulting in potential complications such as inadvertent injury to hollow viscera adjacent to the

Conclusion

We have successfully developed an anti-EGFR mAb modified PEGylated SPIO nanoparticle as targeting MR imaging contrast agents and synergistic agents for MRgFUS ablation in lung carcinoma. These targeting nanoparticles significantly improved the MRI sensitivity for visualization of EGFR overexpressed lung cancer in a rat model. Furthermore, these nanoparticles are highly desirable for their ability to further enhance MRgFUS efficacy with lower energy levels, which may be able to the reduce side

Acknowledgments

The authors would like to gratefully acknowledge the editorial assistance from Puiyan Ho and the technical support for MRgFUS studies from Na Tang. This work was financially supported by the National Nature Science of China (Grant NO. 81271384 and 81371623 and 81671740), NIH/NCI (R01CA199668) and NIH/NICHD (R01HD086195).

References (42)

  • D. Kopelman et al.

    Magnetic resonance-guided focused ultrasound surgery using an enhanced sonication technique in a pig muscle model

    Eur. J. Radiol

    (2006)
  • C. Liao et al.

    Targeting EGFR-overexpressing tumor cells using Cetuximab-immunomicelles loaded with doxorubicin and superparamagnetic iron oxide

    Eur. J. Radiol

    (2011)
  • H.E. Cline et al.

    Focused US system for MR imaging-guided tumor ablation

    Radiology

    (1995)
  • D. Gianfelice et al.

    MR imaging–guided focused us ablation of breast cancer: histopathologic assessment of effectiveness—initial experience 1

    Radiology

    (2003)
  • F.A. Jolesz et al.

    Magnetic resonance image-guided focused ultrasound surgery

    Cancer J. (Sudbury, Mass.)

    (2001)
  • Y. Ishihara et al.

    A precise and fast temperature mapping using water proton chemical shift

    Magn. Reson. Med.

    (1995)
  • Y. Chen et al.

    Multifunctional mesoporous composite nanocapsules for highly efficient MRI-guided high-intensity focused ultrasound cancer surgery

    Angew. Chem. Int. Ed.

    (2011)
  • C.B. Foldager et al.

    Chondrocyte gene expression is affected by very small iron oxide particles-labeling in long-term in vitro MRI tracking

    J. Magn. Reson. Imaging

    (2011)
  • A. Protti et al.

    Assessment of inflammation with a very small iron-oxide particle in a murine model of reperfused myocardial infarction

    J. Magn. Reson. Imaging

    (2014)
  • M. Wagner et al.

    Coronary MR angiography using citrate-coated very small superparamagnetic iron oxide particles as blood-pool contrast agent: initial experience in humans

    J. Magn. Reson. Imaging

    (2011)
  • G. Fu et al.

    Magnetic Prussian blue nanoparticles for targeted photothermal therapy under magnetic resonance imaging guidance

    Bioconjugate Chem.

    (2014)
  • Cited by (175)

    • Electrochemical biosensors for determination of tumor biomarkers

      2024, Semiconducting Polymer Materials for Biosensing Applications
    View all citing articles on Scopus
    View full text