WO2014031727A1 - Ferritin-based tumor targeting agent, and imaging and treatment methods - Google Patents

Ferritin-based tumor targeting agent, and imaging and treatment methods Download PDF

Info

Publication number
WO2014031727A1
WO2014031727A1 PCT/US2013/055955 US2013055955W WO2014031727A1 WO 2014031727 A1 WO2014031727 A1 WO 2014031727A1 US 2013055955 W US2013055955 W US 2013055955W WO 2014031727 A1 WO2014031727 A1 WO 2014031727A1
Authority
WO
WIPO (PCT)
Prior art keywords
ferritin
tumor
agent
cells
iron
Prior art date
Application number
PCT/US2013/055955
Other languages
French (fr)
Inventor
William Keun Chan Park
David R. Mills
Edward G. Walsh
Original Assignee
Brown University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Brown University filed Critical Brown University
Priority to CA2921493A priority Critical patent/CA2921493A1/en
Publication of WO2014031727A1 publication Critical patent/WO2014031727A1/en
Priority to US14/626,071 priority patent/US20150224212A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/18Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes
    • A61K49/1818Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles
    • A61K49/1821Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles
    • A61K49/1824Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles
    • A61K49/1827Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle
    • A61K49/1875Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by a special physical form, e.g. emulsions, microcapsules, liposomes particles, e.g. uncoated or non-functionalised microparticles or nanoparticles coated or functionalised microparticles or nanoparticles coated or functionalised nanoparticles having a (super)(para)magnetic core, being a solid MRI-active material, e.g. magnetite, or composed of a plurality of MRI-active, organic agents, e.g. Gd-chelates, or nuclei, e.g. Eu3+, encapsulated or entrapped in the core of the coated or functionalised nanoparticle coated or functionalised with an antibody
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57492Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds localized on the membrane of tumor or cancer cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57496Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving intracellular compounds
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups

Definitions

  • Magnetic resonance imaging is a versatile medical imaging modality capable of providing both structural and functional information using a variety of contrast weightings.
  • soft tissue contrast is produced by exploiting differences in Tj, T 2 , or T 2 * between different tissues (for example, between grey and white matter in the brain via Ti weighting).
  • Tj structural
  • T 2 contrast-related tissue contrast
  • T 2 * contrast-related tissues
  • contrast agents use high atomic number nuclei to increase attenuation and thereby reveal locations of contrast agent accumulation.
  • contrast agents are used to reduce Tj, T 2 or T 2 * (or some combination of the three) to produce contrast in structures where the agent accumulates.
  • the first approved MRI contrast agents were gadolinium chelates (e.g Gd-DTPA) which act as T, agents.
  • Applications include brain tumor diagnosis: the contrast agent, normally restricted by the intact blood-brain barrier, passes out of the leaky vasculature of a malignant tumor and enters the interstitial space. Ti weighted imaging 10-20 minutes post injection shows significantly increased signal intensity from the tumor owing to Tj reduction whereas normal brain tissue with intact barrier does not show significant changes owing to the restriction of the contrast agent to the vascular compartment.
  • a second class of approved contrast agents has been developed around iron oxide (Fe 3 0 4 ) nanoparticles. These superparamagnetic particles produce primarily T 2 and T 2 * (susceptibility) contrast although some Ti effect has been demonstrated. Since Fe30 4 is isoelectric at physiologic pH, a coating is required to maintain monodispersion. Dextran was the first coating used for an approved agent. Other coatings, such as polyethylene glycol (PEG) have been demonstrated useful for the purpose.
  • PEG polyethylene glycol
  • An important characteristic for any contrast agent is the ability for detection at low concentrations. In this respect, the iron oxide particle agents demonstrate considerably higher relaxivity (defined as the change in relaxation rate per unit agent concentration) than those observed for the gadolinium chelates.
  • Ferritins are iron storage proteins that play a role in the maintenance of iron homeostasis. They function by converting soluble iron into a ferric complex (hydrite) that is stored in an internal cavity of the protein forming in essence, an iron nanocore (Chasteen and Harrison 1999: Harrison and Arosio 1996).
  • Initial work involved the use of endogenous ferritin for estimation of iron concentrations in spleen, pancreas and liver as a means of assessing organ function using T 2 weighted image acquisitions.
  • Natural ferritin complexes however, have been shown to have rl and r2 values too low to act as effective contrast agents (Uchida et al. 2008 and our measurements reported in the discussion of Example 1, below).
  • modified forms have been developed with the aim of encapsulating more iron than in natural forms, with resultant improvements in relaxivity.
  • One such form is the genetically engineered ferritin cage derived from Archaeoglobus fulgidiis developed by Swift and Sana (Swift et al. 2009; Sana et al. 2010).
  • This is a self- assembling spherical cage consisting of 24 subunits which is capable of storing on the order of 7000 Fe atoms per cage in a cavity approximately 8 nm in diameter with an overall hydrodynamic diameter of 14 nm for the entire complex.
  • Advantages of using this complex include a very narrow distribution of particle size (Yoshimura 2006), relaxation enhancement through protein-associated water molecules (Aime et al. 2002), and biocompatibility and stability in biological systems (Mulder et al. 2006).
  • BDEC rat bile duct epithelial cells
  • Necl-5 a cell surface glycoprotein that has been shown to promote cellular proliferation, migration and invasion of transformed cell lines. While Necl-5 is barely detectable in normal epithelial cells, it is dramatically upregulated in many carcinomas including prostate, colorectal, lung, hepatocellular carcinoma (HCC) and other epithelial cancers (Paris et al. 1990; Chadeneau et al. 1991 ;
  • Necl-5 in the rat PEC cell line makes it an attractive target for the development of future cancer detection and therapeutic strategies targeting CD 155 or other human cancer markers.
  • a contrast agent for enhanced imaging comprises metal-loaded, e.g., iron- or manganese-loaded synthetic ferritin nanoparticles coupled with a targeting agent, for example conjugated to an antibody, wherein the antibody or agent targets specific cells, e.g., tumor cells of a known type.
  • a targeting agent for example conjugated to an antibody
  • the antibody or agent targets specific cells, e.g., tumor cells of a known type.
  • Targeting involves binding to a receptor or surface molecule that is up-regulated in the cells, such that the contrast agent specifically or preferentially and effectively adheres to the cells and accumulates at the tumor; the MRI response of the metal-loaded ferritin provides enhanced imaging of the tumor.
  • MRI imaging thus amounts to identifying or diagnosing tumor tissue in a subject or in an in vitro culture.
  • antibody-linked iron-loaded ferritin material is administered to a subject or applied to a cell culture before imaging to enhance MRI imaging of the cells.
  • the method may further include the step of confirming and/or quantifying the ferritin accumulation at the tumor (as evidenced, for example, by reduced T2 and T2 * as compared to a baseline scan), and/or may further include the step of applying an externally-applied stimulus, such as a suitable magnetic field, in a region of the tumor, to locally elevate the temperature and/or promote release of toxic iron from the ferritin, thereby effectively and selectively treating or killing the tumor cells.
  • an externally-applied stimulus such as a suitable magnetic field
  • the magnetic field may be of a strength and be reversed at a frequency effective to promote hyperthermia from energy absorption and Neel relaxation in the iron core nanoparticles.
  • an external magnetic field or other stimulus may be applied in a manner to cause the localized release of ionic iron held in the ferritin cage.
  • the ferritin material is preferably an engineered material with a high capacity for holding iron, and may be further engineered to possess one or more large-dimension pores to enable enhanced release of iron therefrom, e.g., to increase the rate of release as a function of temperature or other stimulation or to initiate release at a high rate upon a relatively modest elevation of temperature.
  • This aspect of the invention also contemplates external stimulation by nonmagnetic means, such as by focused ultrasound, to promote the release of iron at the target tissue.
  • FIGURE 1 schematically illustrates preparation of the ferritin/ mAb Necl-5
  • nanoconjugate ferritin material 6 used in the examples herein wherein individual steps are described under "Methods and Materials”. This diagram depicts the relative length and size of the components, ferritin (about 12.5 nm in diameter), antibody (about 10 nm in length) and the linker (about 1 nm);
  • FIGURE 2 shows images confirming tumor targeting activity of the material of FIGURE l ;
  • FIGURE 3 shows MRI images of uniformly distributed conventional ferritin and the imaging ferritin of this invention at various concentrations;
  • FIGURE 4 shows MRI images of the ferritin attached to tumor cells confirming attachment and enhanced imaging properties
  • FIGURE 5 shows iron assay results quantifying conjugates/cell data for the imaged sample
  • FIGURE 6 shows signal contrast and intensity values.
  • the invention will be understood from the following description of an exemplary embodiment and measurement results obtained therewith, together with discussion of the observed binding, magnetic and imaging characteristics reported below and their use in imaging, diagnosing and treating tissue conditions such as cancer.
  • the invention provides a new MRI contrast agent, namely cell-targeting ferritin cage nanoparticles loaded with iron or other magnetic or paramagnetic metal.
  • the invention also provides diagnostic and treatment methods using the contrast agent.
  • the ferritin used in the present study is a genetically engineered ferritin obtained from
  • Transformed rat PEC were maintained in a 1 : 1 mixture of RPMI 1640 (Gibco, Carlsbad, CA) and MCDB 1 53 (Sigma-Aldrich, St. Louis, MO) supplemented with sodium bicarbonate (1.9 g/L), sodium pyruvate (0.5%), fetal bovine serum (FBS) (5%, Hyclone, Logan, UT), epidermal growth factor (0.02 ⁇ g/ml, BD Biosciences, San Jose, CA), bovine pituitary extract (5 g/ml, BD Biosciences), dexamethasone (2 mM in 95% EtOH), glutamine (1%), gentamycin (0.1 mg/ml, Gibco), ITS (0.25%, BD Biosciences), forskolin (2.5 ⁇ g/ml, Calbiochem, San Diego, CA) and Normocin and incubated at 37°C in a 5% C0 2 humidified atmosphere.
  • RPMI 1640 Gibco,
  • HBSS Hanks Balanced Salt Solution
  • a targeted nanoconjugate version of the ferritin construct was prepared for in vitro testing by binding a monoclonal antibody targeting the Necl-5 glycoprotein, expressed by many epithelial carcinomas, as shown in FIGURE 1.
  • Transformed rat prostate epithelial cells (2.0 x 10 7 cells per sample) were incubated with the targeted form of the ferritin nanoconjugate at three dose levels: 50, 100, and 200 ⁇ g conjugate per ml. After the incubation (37°C, 45 minutes), the samples were washed and centrifuged for three cycles. All of the washes including unbound conjugates were collected and analyzed for iron content using the bathophenanthroline disulfonic acid/sodium dithionate method described earlier (Bonomi and Pagani 1986).
  • iron loaded ferritin cages loaded to 6700 Fe/cage were uniformly dispersed in 1% agarose gel at concentrations of 1, 2, 5, 10, 20, 50, 100, 200, 500 and 1000 nM.
  • Corresponding phantoms were prepared using natural horse ferritin.
  • the gels were contained in 1.5 ml vials for scanning. Scans were acquired using a 3 Tesla Siemens Tim Trio system. A 32-channel head resonator was used for signal receive. Field shimming to second order was performed prior to acquisition of mapping scans.
  • the ferritin vials, along with controls (agarose gel alone) were placed horizontally in a holder within the head resonator.
  • Tomographic images 2 mm thick were acquired of the vials in cross-section with an in-plane resolution of 0.4 mm.
  • T 2 a multi-spin echo sequence was used with a repetition time of 1500 ms and 12 echo times ranging from 10 ms to 120 ms in 10 ms steps.
  • Inversion recovery was used for estimation of Ti with a repetition time of 4000 ms and 12 inversion times ranging from 100 ms to 2400 ms.
  • Relaxation time maps were formed by fitting signal intensity vs echo time (or inversion time) to the relevant signal equations using three-parameter nonlinear least squares fitting routines (Matlab). Relaxivity was determined using a linear fit for relaxation rate vs ferritin
  • T 2 , and T 2 * were visible when the ferritins were evenly distributed in an agarose gel (FIGURE 3 A).
  • T 2 and T 2 * weightings contrast is evident at the shortest echo times (10 ms and 4 ms, respectively).
  • the horse ferritin which is here taken as indicative of endogenous ferritin or a conventional natural ferritin, did not show any significant contrast in the images, although a slight effect was noted in the T 2 and T 2 * maps (FIGURE 3B) while Ti effect is negligible.
  • Relaxivit (rj, r 2 ) was calculated as the slope of the line resulting from a linear fit of relaxation rate vs concentration.
  • FIGURE 4 illustrates T 2 (top panel) and T 2 * (bottom panel) relaxation time maps of the nanoconjugate when bound to target rat prostate epithelial cells. It was observed that the Tj effect was negligible, whereas in the uniformly distributed case the Ti effect was clearly seen. This may relate to the heterogeneous particle distribution resulting in static dephasing (Bowen et al. 2002).
  • Mean relaxation time values were determined for regions of interest taken from the center 80 pixels of the in vitro sample images, and are shown in TABLE 1. The entries are mean + standard deviation of relaxation times for the in vitro study. Circular regions of interest ( 100 pixels) were taken from the center of the vials. SA denotes soft agar.
  • FIGURE 5 is a plot of the conjugate retention vs dose for the in vitro preparation.
  • the clear linear dependence indicates that receptor saturation was not reached even at the highest dose, and that greater binding is possible for this preparation with doses beyond 200 ⁇ / ⁇ 1.
  • Assay results for the in-vitro preparation of Iron per cell are shown in TABLE 2. The iron concentrations were estimated based on the volume of the cell pellets, number of cells per pellet and quantity of iron per cell.
  • FIGURE 6 shows the corresponding signal intensity and contrast curves to illustrate the optimum echo times based on the doses. Contrast is defined as the difference between the signal intensity curve at each concentration subtracted from the control. It was observed that as the dose level increases (and T 2 *) decreases, that peak contrast increases, and the echo time corresponding to peak contrast decreases. The echo times for peak contrast vs. the control occur at 25 ms (50 ⁇ g ml), 23 ms (100 ⁇ g/ml) and 19 ms (200 ⁇ ).
  • compartmentalization causes the assumptions behind the quantum solution to fail, an effect previously described in cell-based studies (Weissleder et al. 1997; Majmudar et al. 1989). Compartmentalization is also accompanied by a substantial increase in the ratio R 2 */R 2 which is not predicted by the quantum solution.
  • the quantum solution assumes the extreme motional narrowing condition, in which water diffusion between superparamagnetic particles is occurring on a time scale significantly shorter than the peak frequency offset and identical values for R 2 and R 2 * are predicted. Compartmentalization of the particles results in bulk susceptibility producing local field inhomogeneities that render the assumption invalid.
  • modified ferritin as a Ti agent appears to be restricted to cases where the particles are maintained in an unbound state such that free water access to the ferritin channels is maximized.
  • One example would be application as a blood pool agent for angiography studies where passage out of the microvasculature into the interstitial space is not desired. In such an application, a targeting ligand would not be required.
  • Rat high passage PEC (p93) cells and soft agar infiltrating (SAI)-selected prostate epithelial cells (PEC) were tumorigenic when injected into immunodeficient beige/nude mice.
  • Tumor size was evaluated at four weeks post-injection for the high passage cells, and three weeks for the SAI-derived PEC tumors.
  • Indirect immunofluorescence imaging and western blotting each demonstrated that high passage (pi 02) and SAI-selected rat PRC expressed high levels of the cell surface glycoprotein Necl-5.
  • Example 2 thus extends the results to in vivo application of an anti-Necl-5/ferritin nanoconjugate for imaging rat prostate epithelial cell tumors, and shows a time-dependent but dramatic difference in MRl response and imaging characteristics.
  • Methods of imaging therefore advantageously include or are preceded by a preliminary time series dose/response sequence of measurements to acquire MRl characteristic data to optimize the interval between administration of the agent and imaging of the tumor.
  • the metal-filled ferritin cages once bound to the target tissue, are caused to release the paramagnetic or superparamagnetic metal contents from their core.
  • This process may be initiated or accelerated by heating, for example by applying a quickly-alternating magnetic field to generate heat, or by applying focused ultrasound to heat the particles and open pores of the ferritin cages.
  • the high valence metal ions thus released from the core of the ferritin cages result in a locally toxic concentration of metal ions.
  • imaging allows the treating physician to coordinate the excitation of the tumor-bound agent and release of the ferritin-caged metal to treat the tumor.
  • the enhanced imaging characteristics enable earlier detection than would otherwise be possible, increasing the effectiveness of such a localized toxic treatment.
  • prostate cancer diagnosis relies on the use of prostate specific antigen (PSA) as a prostate tumor marker that has also served as a target for functionalized nanoparticle detection studies (Taylor et al. 201 1).
  • PSA prostate specific antigen
  • BPH benign prostatic hyperplasia
  • basing a diagnosis on PSA results in over-diagnosis and leads to unnecessary treatment
  • CD155 the human homologue of rat Necl-5
  • the in vitro data indicates that the modified ferritin conjugate has utility as both a T 2 and T 2 * contrast agent when conjugated to an antibody of interest for targeting and imaging antigen-specific tissues.
  • the antigen-specific tissues may be cancer cells or other diseased cells that express a specific cell surface molecule. Many such molecules have been characterized and associated with specific cancers or tissue pathologies; the antibody employed for targeting the ferritin nanoparticles may be an antibody to such a characterizing molecule, or may be an antibody to a relevant portion thereof.
  • the ferritin being conjugated to an antibody
  • equivalent specificity and effective accumulation and concentration at the relevant cells can be expected if the ferritin is clothed with the epitope, or active portion of the antibody responsible for binding.
  • the entire ferritin-epitope construct may be genetically engineered as a fusion protein.
  • the targeted surface molecules may be a molecule that is specific to a highly invasive cell line, so that MRI images reveal specific information as to tumor type.
  • Example 2 reports in vivo results imaging highly invasive tumors grown from soft agar infiltrating prostate epithelial cells. By specifically identifying surface markers and employing targeting antibodies for such cells, the techniques of the invention significantly advance early detection and treatment.
  • the magnitude of the relevant magnetic resonance parameters described above further indicates that other targeting functionalities - such as cloaking the ferritin in a targeting functionalized phospholipid or nanoemuision as the delivery vehicle - can also be applied to advantage to achieve for in vivo delivery to tumor sites.
  • a targeted nanoemuision for in vivo use is compounded to allow the agent to circulate in the bloodstream sufficiently many times to accumulate specifically at the targeted tissue.
  • T 2 and T 2 * values are determined, further baseline studies may be performed for a given targeting agent and target cell line to determine the optimum interval required after administering the ferritin nanoparticles for effective tissue binding to occur, so that diagnostic imaging and/or metal ion release therapy can be efficiently performed without taking multiple or comparative sets of before/after MRI scans.
  • Comparison of pre- and post- administration MRI image data indicate tumorous regions of ferritin accumulation, and imaging protocols that display the difference will provide high contrast, tumor-specific imaging. For example, since the Ti effect in EXAMPLE 1 was seen only when particles were uniformly suspended and unbound, so detection of a tumor would be revealed by T 2 and T 2 * weighting. Once a baseline scan is acquired of the suspect region, tumor presence is revealed by reduction of T 2 and T 2 * relative to the baseline scan when the contrast agent has been administered.
  • a tumor-targeting agent e.g., an antibody
  • Coupling a tumor-targeting agent to the nanoparticle ferritin contrast agent in the present invention assures that the agent binds to the relevant tissue with high efficiency and specificity, so that while a dose/response relationship governs the image, only very small amounts of the contrast agent are needed for diagnostic imaging.
  • Glahn RP Gangloff MB, van Campen DR, Miller DD, Wien EM, Norvell WA, 1995.
  • Bathophenanthrolene disulfonic acid and sodium dithionite effectively remove surface-bound iron from caco-2 cell monolayers. J Nutr 125: 1833-1840.
  • TuAg. l is the liver isoform of the rat colon tumor-associated antigen pE4 and a member of the immunoglobulin-like supergene family. Cancer Research 56: 43034.
  • cholangiocytes determines susceptibility to activated ErbB-2/Neu. Experimental and Molecular Pathology 89: 248-259.
  • Multifunctional iron platinum stealth innumomicelles targeted detection of human prostate cancer cells using both fluorescence and magnetic resonance imaging. J Nanopart Res 13:4717- 4729.
  • Magnetically labeled cells can be detected by MR imaging. J Magn Reson Imaging 7:258-263.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • Nanotechnology (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biochemistry (AREA)
  • Oncology (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Hospice & Palliative Care (AREA)
  • General Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Veterinary Medicine (AREA)
  • Microbiology (AREA)
  • Public Health (AREA)
  • Analytical Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Epidemiology (AREA)
  • Radiology & Medical Imaging (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

An MRI contrast material includes tumor-targeting metal-loaded ferritin nanoparticles constructed with genetically modified ferritin coupled to a target-specific agent. Ferritin derived from Archaeoglobus fulgidus (AfFtn-AA) forms hollow nanocages surrounding paramagnetic or superparamagnetic metal core, storing a significantly greater quantity of iron (approximately 7,000 Fe ions per ferritin cage) or other paramagnetic or superparamagnetic metal than natural ferritins, and is conjugated via a short linker with a monoclonal antibody against a cell surface antigen overexpressed by a cancer, to selectively and efficiently attach to tumor cells to enhance MRI contrast. Significant T2 contrast with diminished T1 effect was observed owing to the heterogeneous nanoconjugate distribution when bound to cells. In a treatment method, after imaging, an external stimulus heats the cell-bound agent to release the metal and selectively destroy the targeted cells. The enhanced imaging and release of toxic metal ions provides simultaneous early detection and treatment.

Description

FERRITIN-BASED TUMOR TARGETING AGENT,
AND
IMAGING AND TREATMENT METHODS
Related Applications
This application is related to and claims the benefit of United States provisional applications serial number 61 /803,955 filed August 21, 2012, and serial number 61/691,346 filed March 21, 2013 entitled, " Ferritin-based tumor targeting agent, and imaging and treatment methods". The full text, including drawings and Appendices of those applications are hereby incorporated herein by reference. In addition, a Bibliography in this specification contains further technical detail regarding the procedures and materials described herein. For brevity, articles in the bibliography are referred to by (Author, year) herein narrative text.
Government Support
This invention was made with government support under grant number P20GM 103421 awarded by the National Institute of General Medical Sciences of the National Institutes of Health. The government has certain rights in the invention.
Background
More than 571,950 people in the U.S. died from common cancers (colorectal, prostate, breast, lung and liver cancers), and more than 1.5 million new cancer cases were diagnosed in 201 1 (American Cancer Society, 201 1). Despite numerous technological and medical breakthroughs made in recent years, effective diagnosis and treatment of these cancers remain elusive. In order to overcome limitations regarding the lack of early detection methods and/or selective tumor-targeting therapeutic agents, current paradigms for cancer research continue to place an emphasis on discovery of improved tumor-specific biomarkers, on development of more sensitive detection/visualization methods for accurately assessing the location and extent of tumors, on treatment options and on selective delivery of anti-tumor agents to primary and secondary metastatic tumors.
Magnetic resonance imaging (MRI) is a versatile medical imaging modality capable of providing both structural and functional information using a variety of contrast weightings. For structural (conventional diagnostic) imaging, soft tissue contrast is produced by exploiting differences in Tj, T2, or T2* between different tissues (for example, between grey and white matter in the brain via Ti weighting). Although many structures can be distinguished using endogenous contrast, it was found that some structures (such as tumors that have Ti very similar to that of surrounding normal tissue) could be better visualized through the use of contrast agents. In X-ray imaging methods, contrast agents use high atomic number nuclei to increase attenuation and thereby reveal locations of contrast agent accumulation. In MRI, contrast agents are used to reduce Tj, T2 or T2* (or some combination of the three) to produce contrast in structures where the agent accumulates. The first approved MRI contrast agents were gadolinium chelates (e.g Gd-DTPA) which act as T, agents. Applications include brain tumor diagnosis: the contrast agent, normally restricted by the intact blood-brain barrier, passes out of the leaky vasculature of a malignant tumor and enters the interstitial space. Ti weighted imaging 10-20 minutes post injection shows significantly increased signal intensity from the tumor owing to Tj reduction whereas normal brain tissue with intact barrier does not show significant changes owing to the restriction of the contrast agent to the vascular compartment.
A second class of approved contrast agents has been developed around iron oxide (Fe304) nanoparticles. These superparamagnetic particles produce primarily T2 and T2* (susceptibility) contrast although some Ti effect has been demonstrated. Since Fe304 is isoelectric at physiologic pH, a coating is required to maintain monodispersion. Dextran was the first coating used for an approved agent. Other coatings, such as polyethylene glycol (PEG) have been demonstrated useful for the purpose. An important characteristic for any contrast agent is the ability for detection at low concentrations. In this respect, the iron oxide particle agents demonstrate considerably higher relaxivity (defined as the change in relaxation rate per unit agent concentration) than those observed for the gadolinium chelates.
Recent efforts have examined the use of ferritin as a potential contrast agent (Uchida et al. 2006, 2008; Swift et al. 2009; Sana et al. 2010; Jordan et al. 2010). Ferritins are iron storage proteins that play a role in the maintenance of iron homeostasis. They function by converting soluble iron into a ferric complex (hydrite) that is stored in an internal cavity of the protein forming in essence, an iron nanocore (Chasteen and Harrison 1999: Harrison and Arosio 1996). Initial work involved the use of endogenous ferritin for estimation of iron concentrations in spleen, pancreas and liver as a means of assessing organ function using T2 weighted image acquisitions. Natural ferritin complexes however, have been shown to have rl and r2 values too low to act as effective contrast agents (Uchida et al. 2008 and our measurements reported in the discussion of Example 1, below). To improve the prospects of using ferritin as a basis for MRI contrast agents, modified forms have been developed with the aim of encapsulating more iron than in natural forms, with resultant improvements in relaxivity. One such form is the genetically engineered ferritin cage derived from Archaeoglobus fulgidiis developed by Swift and Sana (Swift et al. 2009; Sana et al. 2010). This is a self- assembling spherical cage consisting of 24 subunits which is capable of storing on the order of 7000 Fe atoms per cage in a cavity approximately 8 nm in diameter with an overall hydrodynamic diameter of 14 nm for the entire complex. Advantages of using this complex include a very narrow distribution of particle size (Yoshimura 2006), relaxation enhancement through protein-associated water molecules (Aime et al. 2002), and biocompatibility and stability in biological systems (Mulder et al. 2006).
In previous studies, we developed a spontaneous transformation model for rat bile duct epithelial cells (BDEC) that culminated at high passage (p>85) in anchorage independent growth for cells plated on soft agar, and tumorigenicity when injected into immune deficient mice (Rozich et al. 2010). Briefly, by mid-passage (p3 1-85), BDEC showed alterations in morphology, onset of aneuploidy, increased growth rate with growth factor independence, decreased cell adhesion and loss of cholangiocyte markers expressed at low passage (p<30). We have recently developed an in vitro model of spontaneous transformation for rat prostate epithelial cells (PEC) that closely recapitulates many of the molecular and cellular changes observed during spontaneous transformation of rat BDEC. The rat prostate cells were isolated from dorso-lateral prostate lobes from mature Fisher 344 rats without prior carcinogen treatment as described previously (Britt et al. 2004; Mills et al. 2012-Exp Mol Path, in press). The development and characterization of the transformed rat PEC line used in the examples herein will be more fully described elsewhere in a forthcoming publication (Mills et al., manuscript in preparation).
However, as relevant to the present invention, previous studies in our laboratory have demonstrated that the transformed rat PEC used in this study express high levels of the cell adhesion protein, Necl-5, a cell surface glycoprotein that has been shown to promote cellular proliferation, migration and invasion of transformed cell lines (Sloan et al. 2004; Sato et al. 2004; Ikeda et al. 2004). While Necl-5 is barely detectable in normal epithelial cells, it is dramatically upregulated in many carcinomas including prostate, colorectal, lung, hepatocellular carcinoma (HCC) and other epithelial cancers (Paris et al. 1990; Chadeneau et al. 1991 ;
Gromeier et al. 2000; Masson et al. 2001 ). The constitutive over-expression of Necl-5 in the rat PEC cell line makes it an attractive target for the development of future cancer detection and therapeutic strategies targeting CD 155 or other human cancer markers.
Summary
In a first embodiment of the invention, a contrast agent for enhanced imaging, comprises metal-loaded, e.g., iron- or manganese-loaded synthetic ferritin nanoparticles coupled with a targeting agent, for example conjugated to an antibody, wherein the antibody or agent targets specific cells, e.g., tumor cells of a known type. Targeting involves binding to a receptor or surface molecule that is up-regulated in the cells, such that the contrast agent specifically or preferentially and effectively adheres to the cells and accumulates at the tumor; the MRI response of the metal-loaded ferritin provides enhanced imaging of the tumor. By pro viding a tissue-specific change in magnetic response properties, MRI imaging thus amounts to identifying or diagnosing tumor tissue in a subject or in an in vitro culture. In an exemplary imaging method using the contrast agent, antibody-linked iron-loaded ferritin material is administered to a subject or applied to a cell culture before imaging to enhance MRI imaging of the cells. When administered to a subject, either systemically or by local injection to a tumor site, the method may further include the step of confirming and/or quantifying the ferritin accumulation at the tumor (as evidenced, for example, by reduced T2 and T2* as compared to a baseline scan), and/or may further include the step of applying an externally-applied stimulus, such as a suitable magnetic field, in a region of the tumor, to locally elevate the temperature and/or promote release of toxic iron from the ferritin, thereby effectively and selectively treating or killing the tumor cells. The magnetic field may be of a strength and be reversed at a frequency effective to promote hyperthermia from energy absorption and Neel relaxation in the iron core nanoparticles. Alternatively, or in addition, an external magnetic field or other stimulus may be applied in a manner to cause the localized release of ionic iron held in the ferritin cage. The ferritin material is preferably an engineered material with a high capacity for holding iron, and may be further engineered to possess one or more large-dimension pores to enable enhanced release of iron therefrom, e.g., to increase the rate of release as a function of temperature or other stimulation or to initiate release at a high rate upon a relatively modest elevation of temperature. This aspect of the invention also contemplates external stimulation by nonmagnetic means, such as by focused ultrasound, to promote the release of iron at the target tissue.
Brief Description of the Drawings
These and other features of the invention will be understood from the description and claims hereof, taken together with the Drawings, wherein:
FIGURE 1 schematically illustrates preparation of the ferritin/ mAb Necl-5
nanoconjugate ferritin material 6 used in the examples herein wherein individual steps are described under "Methods and Materials". This diagram depicts the relative length and size of the components, ferritin (about 12.5 nm in diameter), antibody (about 10 nm in length) and the linker (about 1 nm);
FIGURE 2 shows images confirming tumor targeting activity of the material of FIGURE l ; FIGURE 3 shows MRI images of uniformly distributed conventional ferritin and the imaging ferritin of this invention at various concentrations;
FIGURE 4 shows MRI images of the ferritin attached to tumor cells confirming attachment and enhanced imaging properties;
FIGURE 5 shows iron assay results quantifying conjugates/cell data for the imaged sample; and
FIGURE 6 shows signal contrast and intensity values.
Detailed Description
The invention will be understood from the following description of an exemplary embodiment and measurement results obtained therewith, together with discussion of the observed binding, magnetic and imaging characteristics reported below and their use in imaging, diagnosing and treating tissue conditions such as cancer. Briefly, the invention provides a new MRI contrast agent, namely cell-targeting ferritin cage nanoparticles loaded with iron or other magnetic or paramagnetic metal. The invention also provides diagnostic and treatment methods using the contrast agent.
Initially we describe in detail the preparation of an iron-loaded, cancer-targeting ferritin nanoparticle contrast agent and its properties. METHODS AND MATERIALS
Ferritin:
The ferritin used in the present study is a genetically engineered ferritin obtained from
Archaeoglobus fulgidus. Cloning, expression and purification were performed following the methods previously described in Sana et al. (2010). Enrichment of the ferritin with iron (II I) ions and the analysis of iron loading were achieved by following the methods reported in Liu et al.
2003; Glahn et al. 1995; and Bonomi and Pagani 1986. The process was repeated three times, and the average value for the number of iron (III) ion per each ferritin was determined to be
6,700. It was observed that iron loading beyond 7000 Fe/cage resulted in some difficulty in maintaining monodispersion in suspension, with precipitation possible due to aggregation. Conjugation of iron-enriched ferritin and
anti-Necl-5 monoclonal antibody (MAb Necl-5):
Generation and characterization of the Necl-5 specific mouse IgG monoclonal antibody
(MAb 324.5) has been described previously (Hixson et al. 1986; Faris et al. 1990; Lim et al.
1996). To prepare the contrast agent, the two components, mAb Necl-5 and Fe(IiI)-enriched ferritin, were tethered by a convergent method, which is schematically illustrated in the schema Succinimidyl S-Acetylthioacetate, 4 equiv. ThermoScientific) in a HEPES buffer solution (pH 7.5), which result in 3j a thioacetyl acetamide elongation. Separately, lysine residues of 2, Fe(Ill)-enriched ferritin were treated with Sulfo-SMCC (succinimidyl 4-[N-maleimidomethyl] cyclohexane-l-carboxylate, 4 equiv. ThermoScientific) in the same buffer solution to yield 5. which in turn, reacted with 4, the deacetylated thiol form of 3. The reaction of 4 with 5 proceeded in the presence of EDTA in order to suppress the disulfide formation between two 4 molecules, and the desired conjugate 6 was obtained and isolated by a size exclusion chromatography (SEC) Superdex 200 10/300 GL column (GE Healthcare, Buckinghamshire, UK).
In vitro preparation of cells:
Transformed rat PEC were maintained in a 1 : 1 mixture of RPMI 1640 (Gibco, Carlsbad, CA) and MCDB 1 53 (Sigma-Aldrich, St. Louis, MO) supplemented with sodium bicarbonate (1.9 g/L), sodium pyruvate (0.5%), fetal bovine serum (FBS) (5%, Hyclone, Logan, UT), epidermal growth factor (0.02 μg/ml, BD Biosciences, San Jose, CA), bovine pituitary extract (5 g/ml, BD Biosciences), dexamethasone (2 mM in 95% EtOH), glutamine (1%), gentamycin (0.1 mg/ml, Gibco), ITS (0.25%, BD Biosciences), forskolin (2.5 μg/ml, Calbiochem, San Diego, CA) and Normocin and incubated at 37°C in a 5% C02 humidified atmosphere. Cells were grown to approximately 75-80% confluence, and were trypsinized and washed in Hanks Balanced Salt Solution (HBSS; Sigma-Aldrich). Cell suspensions were incubated in the presence or absence of Necl-5 nanoconjugate in I X PBS supplemented with 0.5% BSA at 4°C for 1 hr with gentle rotation. Following two 5 min washes in HBSS, cells suspensions were mixed 1 : 1 with 1 % SeaPlaque low melting temperature agarose (Lonza, Rockland, ME) in 2 ml conical vials for subsequent imaging. Cell preparations in 2 ml vials (along with an undosed control cell sample) were scanned using the same procedure as for the uniform dispersion gel samples with relaxation rates and relaxivities calculated in the same manner. Each cell pellet contained approximately 2xl07 cells.
EXAMPLE 1 ,
A targeted nanoconjugate version of the ferritin construct was prepared for in vitro testing by binding a monoclonal antibody targeting the Necl-5 glycoprotein, expressed by many epithelial carcinomas, as shown in FIGURE 1. Transformed rat prostate epithelial cells (2.0 x 107 cells per sample) were incubated with the targeted form of the ferritin nanoconjugate at three dose levels: 50, 100, and 200 μg conjugate per ml. After the incubation (37°C, 45 minutes), the samples were washed and centrifuged for three cycles. All of the washes including unbound conjugates were collected and analyzed for iron content using the bathophenanthroline disulfonic acid/sodium dithionate method described earlier (Bonomi and Pagani 1986).
Magnetic Resonance Assessment of Relaxivity
For MR relaxivity measurements, iron loaded ferritin cages loaded to 6700 Fe/cage were uniformly dispersed in 1% agarose gel at concentrations of 1, 2, 5, 10, 20, 50, 100, 200, 500 and 1000 nM. Corresponding phantoms were prepared using natural horse ferritin. The gels were contained in 1.5 ml vials for scanning. Scans were acquired using a 3 Tesla Siemens Tim Trio system. A 32-channel head resonator was used for signal receive. Field shimming to second order was performed prior to acquisition of mapping scans. The ferritin vials, along with controls (agarose gel alone) were placed horizontally in a holder within the head resonator. Tomographic images 2 mm thick were acquired of the vials in cross-section with an in-plane resolution of 0.4 mm. For estimation of T2 a multi-spin echo sequence was used with a repetition time of 1500 ms and 12 echo times ranging from 10 ms to 120 ms in 10 ms steps. In addition, gradient echo images were acquired to give an indication of susceptibility contrast (TR=1500 ms, TE=4-24 ms, six echoes). Inversion recovery was used for estimation of Ti with a repetition time of 4000 ms and 12 inversion times ranging from 100 ms to 2400 ms.
Relaxation time maps were formed by fitting signal intensity vs echo time (or inversion time) to the relevant signal equations using three-parameter nonlinear least squares fitting routines (Matlab). Relaxivity was determined using a linear fit for relaxation rate vs ferritin
concentration.
Results and discussion
Indirect immunofluorescence imaging demonstrated strong reactivity of the ferritin/mAb Necl-5 nanoconjugate (FIGURE 2, top left) against transformed Necl-5 positive rat prostate epithelial cells that was comparable to anti-Necl-5 antibody alone (FIGURE 2, top right). Furthermore, transmission electron microscopy (TEM) showed that the nanoconjugate binds to the rat prostate epithelial cells in a manner comparable to gold conjugated anti-Necl-5 antibody (FIGURE 2, lower panel). These in vitro studies indicate that conjugation of the modified ferritin cage to anti-Necl-5 antibody did not affect the targeting specificity or reactivity of the antibody against the Necl-5 antigen.
MRI imaging of phantoms made evident that contrast effects of all three weightings (Ti ,
T2, and T2*) were visible when the ferritins were evenly distributed in an agarose gel (FIGURE 3 A). For the T2 and T2* weightings, contrast is evident at the shortest echo times (10 ms and 4 ms, respectively). The horse ferritin, which is here taken as indicative of endogenous ferritin or a conventional natural ferritin, did not show any significant contrast in the images, although a slight effect was noted in the T2 and T2* maps (FIGURE 3B) while Ti effect is negligible. Relaxivit (rj, r2) was calculated as the slope of the line resulting from a linear fit of relaxation rate vs concentration. The values for the ferritin loaded to 6700 Fe/cage were ^=1290 raM"' s"1 and r2=5742 mM"1 s"1. These values were significantly higher than those obtained from the horse ferritin
Figure imgf000009_0001
mM'1 s"1, r2=95.54 mM'1 s"1). This result compares favorably to commercial superparamagnetic iron oxide nanoparticle (SPION) imaging preparations as well as micelle-contained FePt variants (Taylor et al. 2011).
FIGURE 4 illustrates T2 (top panel) and T2* (bottom panel) relaxation time maps of the nanoconjugate when bound to target rat prostate epithelial cells. It was observed that the Tj effect was negligible, whereas in the uniformly distributed case the Ti effect was clearly seen. This may relate to the heterogeneous particle distribution resulting in static dephasing (Bowen et al. 2002). Mean relaxation time values were determined for regions of interest taken from the center 80 pixels of the in vitro sample images, and are shown in TABLE 1. The entries are mean + standard deviation of relaxation times for the in vitro study. Circular regions of interest ( 100 pixels) were taken from the center of the vials. SA denotes soft agar.
TABLE 1
Figure imgf000009_0002
FIGURE 5 is a plot of the conjugate retention vs dose for the in vitro preparation. The clear linear dependence indicates that receptor saturation was not reached even at the highest dose, and that greater binding is possible for this preparation with doses beyond 200μ /ιη1. Assay results for the in-vitro preparation of Iron per cell are shown in TABLE 2. The iron concentrations were estimated based on the volume of the cell pellets, number of cells per pellet and quantity of iron per cell.
For the T2* values determined in TABLE 1, FIGURE 6 shows the corresponding signal intensity and contrast curves to illustrate the optimum echo times based on the doses. Contrast is defined as the difference between the signal intensity curve at each concentration subtracted from the control. It was observed that as the dose level increases (and T2*) decreases, that peak contrast increases, and the echo time corresponding to peak contrast decreases. The echo times for peak contrast vs. the control occur at 25 ms (50 μg ml), 23 ms (100 μg/ml) and 19 ms (200 μ^ηιΐ). These magnitudes imply that for an image (pixel) signal-to-noise ratio of 20 in the baseline image, the contrast change will be detectable with a dose of 20 μg/ml for the in vitro preparation described above. That dose would correspond to approximately 0.62 pg/cell iron loading.
Figure imgf000010_0001
The high ratio of R2*/R2 is indicative of static dephasing (Bowen et al. 2002) resulting from local accumulations of particles as opposed to uniform distribution. Dependence of Tj and T2 in the presence of superparamagnetic nanoparticles has been described for uniform distribution using modified forms of the Solomon-Bloembergen-Morgan equations (Koenig et al. 1995; Bulte et al. 1999). These calculations predicted superparamagnetic particles as having a much smaller effect on T| than on T2 owing to the large magnetic moment. This observation was confirmed in the uniform distribution measurements and may be the result of diffusion of associated water molecules through the ferritin channels (Aime et al. 2002). With respect to R2 and R2*, compartmentalization causes the assumptions behind the quantum solution to fail, an effect previously described in cell-based studies (Weissleder et al. 1997; Majmudar et al. 1989). Compartmentalization is also accompanied by a substantial increase in the ratio R2*/R2 which is not predicted by the quantum solution. The quantum solution assumes the extreme motional narrowing condition, in which water diffusion between superparamagnetic particles is occurring on a time scale significantly shorter than the peak frequency offset and identical values for R2 and R2* are predicted. Compartmentalization of the particles results in bulk susceptibility producing local field inhomogeneities that render the assumption invalid. Monte Carlo simulations of water diffusing through local dipolar fields however, have been successfully employed in predicting the relationship between R2 and R2* for the case of particle compartmentalization (Weisskoff et al. 1994; Muller et al. 1991 ; Hardy and Hendelman 1989; Fisel et al. 1991 ; Majmudar and Gore 1988).
Changes in T2 and T2* were clearly distinguished in the in vitro preparation at a concentration (in the cell pellet) of 103 nMol. The minimum detectable concentration for the agent depends on a number of factors including cell density, magnetic field shim conditions in the region of the tissue binding the agent, the scan type (spin vs gradient echo) and scan parameters (repetition time, echo time, and geometric factors affecting signal to noise ratio). As seen from the binding assay (FIGURE 5, TABLE 2) it appears likely that concentrations in excess of 400nM can be produced in this in vitro preparation or an in vivo case with similar cell density, which would thus result in a very substantial contrast effect.
The foregoing experimental results establish the effective targeting and imaging of a specific protein by a ferritin construct, and quantification of the relevant MRI imaging and dosing parameters in an in vitro experimental model. In the study reported by Sana et al. (2010), a clear Ti effect was observed at a field strength of 3 Tesla, the same field strength used in this study. This was verified in examples herein with the preparation in which ferritin particles were uniformly distributed in agarose gel. The lack of Ti effect in the in vitro experiment may be the result of a reduced ability for free water to access the channels of the bound ferritin. If this is the case, use of the modified ferritin as a Ti agent appears to be restricted to cases where the particles are maintained in an unbound state such that free water access to the ferritin channels is maximized. One example would be application as a blood pool agent for angiography studies where passage out of the microvasculature into the interstitial space is not desired. In such an application, a targeting ligand would not be required.
EXAMPLE 2
Rat high passage PEC (p93) cells and soft agar infiltrating (SAI)-selected prostate epithelial cells (PEC) were tumorigenic when injected into immunodeficient beige/nude mice.
Tumor size was evaluated at four weeks post-injection for the high passage cells, and three weeks for the SAI-derived PEC tumors. SAI-derived tumors showed a shorter latency period than high passage derived tumors, and the average weight of removed tumors at the time of sacrifice was 0.2 grams (n=3, 4 weeks) and 0.76 grams (n=5, 3 weeks), for high pass and SAI injected cells, respectively. Indirect immunofluorescence imaging and western blotting each demonstrated that high passage (pi 02) and SAI-selected rat PRC expressed high levels of the cell surface glycoprotein Necl-5. To evaluate the ferritin-based contrast agent, in vivo MRI imaging of immunodeficient mice previously injected with PEC SAI cells was performed at 4 and at 24 hours after injection of anti-Necl-5/ferritin or ferritin alone, and was compared to baseline values taken before the ferritin injections. The nanoconj ugate targeted tumor showed significant reduction of T2 signal at 4 hours post-injection, and a substantially lesser reduction of T2 at 24 hours, while the control, and regions of muscle tissue in both sets of mice were not substantially affected by either the targeted or the non-targeting ferritin.
Example 2 thus extends the results to in vivo application of an anti-Necl-5/ferritin nanoconjugate for imaging rat prostate epithelial cell tumors, and shows a time-dependent but dramatic difference in MRl response and imaging characteristics. Methods of imaging therefore advantageously include or are preceded by a preliminary time series dose/response sequence of measurements to acquire MRl characteristic data to optimize the interval between administration of the agent and imaging of the tumor.
EXAMPLE 3
In accordance with a further aspect of the invention the metal-filled ferritin cages, once bound to the target tissue, are caused to release the paramagnetic or superparamagnetic metal contents from their core. This process may be initiated or accelerated by heating, for example by applying a quickly-alternating magnetic field to generate heat, or by applying focused ultrasound to heat the particles and open pores of the ferritin cages. The high valence metal ions thus released from the core of the ferritin cages result in a locally toxic concentration of metal ions. Thus, imaging allows the treating physician to coordinate the excitation of the tumor-bound agent and release of the ferritin-caged metal to treat the tumor. The enhanced imaging characteristics enable earlier detection than would otherwise be possible, increasing the effectiveness of such a localized toxic treatment.
GENERAL CONSIDERATIONS
The development of targeted imaging contrast agents with high specificity is an important step in the advancement of cancer diagnostics. Yet the diagnostic indicators for some cancers are relatively non-specific. For example, prostate cancer diagnosis relies on the use of prostate specific antigen (PSA) as a prostate tumor marker that has also served as a target for functionalized nanoparticle detection studies (Taylor et al. 201 1). However, it was recently found that benign prostatic hyperplasia (BPH) also produces PSA, so that basing a diagnosis on PSA results in over-diagnosis and leads to unnecessary treatment (Chou et al. 2011). In accordance with the present invention, by targeting CD155, the human homologue of rat Necl-5, this diagnostic ambiguity would be eliminated. In examples herein we have demonstrated targeting of a ferritin-based metal complex to Necl-5 in a transformed rat prostate epithelial cell line model. A clear effect was seen for changes in T2 and T2* as would be reflected in spin echo and gradient echo imaging, respectively. The agent produced a visible effect (compared to a control) at a concentration of 102nM Fe in the in vitro study along with an indication of the feasibility of binding to produce a concentration in excess of 400 nM. This is believed to be the first description of use of the modified ferritin complex as a contrast agent for targeting of a specific protein in an in vitro experimental model. As shown here, the in vitro data indicates that the modified ferritin conjugate has utility as both a T2 and T2* contrast agent when conjugated to an antibody of interest for targeting and imaging antigen-specific tissues. The antigen-specific tissues may be cancer cells or other diseased cells that express a specific cell surface molecule. Many such molecules have been characterized and associated with specific cancers or tissue pathologies; the antibody employed for targeting the ferritin nanoparticles may be an antibody to such a characterizing molecule, or may be an antibody to a relevant portion thereof.
In other embodiments, rather than the ferritin being conjugated to an antibody, equivalent specificity and effective accumulation and concentration at the relevant cells can be expected if the ferritin is clothed with the epitope, or active portion of the antibody responsible for binding. For example, the entire ferritin-epitope construct may be genetically engineered as a fusion protein. Furthermore, the targeted surface molecules may be a molecule that is specific to a highly invasive cell line, so that MRI images reveal specific information as to tumor type. Example 2 reports in vivo results imaging highly invasive tumors grown from soft agar infiltrating prostate epithelial cells. By specifically identifying surface markers and employing targeting antibodies for such cells, the techniques of the invention significantly advance early detection and treatment.
The magnitude of the relevant magnetic resonance parameters described above further indicates that other targeting functionalities - such as cloaking the ferritin in a targeting functionalized phospholipid or nanoemuision as the delivery vehicle - can also be applied to advantage to achieve for in vivo delivery to tumor sites. A targeted nanoemuision for in vivo use is compounded to allow the agent to circulate in the bloodstream sufficiently many times to accumulate specifically at the targeted tissue.
Once the relevant T2 and T2* values are determined, further baseline studies may be performed for a given targeting agent and target cell line to determine the optimum interval required after administering the ferritin nanoparticles for effective tissue binding to occur, so that diagnostic imaging and/or metal ion release therapy can be efficiently performed without taking multiple or comparative sets of before/after MRI scans. Comparison of pre- and post- administration MRI image data indicate tumorous regions of ferritin accumulation, and imaging protocols that display the difference will provide high contrast, tumor-specific imaging. For example, since the Ti effect in EXAMPLE 1 was seen only when particles were uniformly suspended and unbound, so detection of a tumor would be revealed by T2 and T2* weighting. Once a baseline scan is acquired of the suspect region, tumor presence is revealed by reduction of T2 and T2* relative to the baseline scan when the contrast agent has been administered.
Coupling a tumor-targeting agent (e.g., an antibody) to the nanoparticle ferritin contrast agent in the present invention assures that the agent binds to the relevant tissue with high efficiency and specificity, so that while a dose/response relationship governs the image, only very small amounts of the contrast agent are needed for diagnostic imaging.
The foregoing describes a tissue-targeting nanoparticle MR! contrast agent and confirmatory measurements and observations that confirm its improved imaging characteristics, as well as its utility in methods of diagnosis and of treatment of specific diseased tissue or cancer conditions. The invention and illustrative methods being thus described, further variations and modifications will occur to those skilled in the art, and all such variations and modifications are understood to be within the scope of the invention and claims appended hereto. REFERENCES
Aime S, Frullano L, Crich SG, 2002. Compartmentalization of a gadolinium complex in the apoferritin cavity: a route to obtain high relaxivity contrast agents for magnetic resonance imaging. Angew. Chem. Int. Ed. 41 : 101 7-1019.
American Cancer Society, 201 1. Cancer facts and figures 201 1. Atlanta GA.
Bonomi F, Pagani S, 1986. Removal of ferritin-bound iron by DL-dihydrolipoate and DL- dihydrolipoamide. Eur J Biochem 155:295-300.
Bowen CV, Zhang X, Saab G, Gareau PJ, Rutt B , 2002. Application of the static dephasing regime theory to superparamagnetic iron-oxide loaded cells. Magn Reson Med 48:52-61.
Britt DE, Yang DF, Yang DO, Flanagan D, Callanan H, Lim YP, Lin SH, Hixson DC, 2004. Identification of a novel protein, LYRIC, localized to tight junctions of polarized epithelial cells. Experimental Cell Research 300: 134-148.
Chadeneau C, Denis MG, Blottiere HM, Gregoire M, Douillard JY, Meflah K, 1991.
Characterization, isolation and amino terminal sequencing of a rat colon carcinoma-associated antigen. Int J Cancer 47:903-908.
Chasteen ND, Harrison PM, 1999. Mineralization in ferritin: an efficient means of iron storage. J Struct Biol 126: 182-194. Chou R, Croswell JM, Dana T, Bougatsos, Blazine I, Fu R, Gleitsmann K, Koenig HC, Lam C, Maltz A, Rugge JB, Lin , 201 1. Screening for prostate cancer - a review of the evidence for the U.S. preventive services task force. Ann Inter Med 155:762-771. Erickson BM, Thompson NL, Hixon DC, 2006. Tightly regulated induction of the adhesion molecule necl-5/CD155 during rat liver regeneration and acute liver injury. Hepatology 43:325- 334.
Fan's RA, McEntire KD, Thompson NL, Hixson DC. 1990. Identification and characterization of a rat hepatic oncofetal membrane glycoprotein. Cancer Research 50:4755.
Fisel CR, Ackerman JL, Buxton RB. Garrido L, Belliveau JW, Rosen BR, Brady TJ, 1991. MR contrast due to microscopically heterogeneous magnetic susceptibility: numerical simulations and applications to cerebral physiology. Magn Reson Med 17:336-347.
Glahn RP, Gangloff MB, van Campen DR, Miller DD, Wien EM, Norvell WA, 1995.
Bathophenanthrolene disulfonic acid and sodium dithionite effectively remove surface-bound iron from caco-2 cell monolayers. J Nutr 125: 1833-1840.
Gromeier M, Lachmann S, Rosenfeld MR, Gut in PH, Wimmer E. 2000. Intergeneric poliovirus recombinants for the treatment of malignant glioma. Proc Natl Acad Sci U S A 97: 6803-6808.
Hardy PA, Henkelman RM, 1989. Transverse relaxation rate enhancement caused by magnetic particulates. Magn Reson Imaging 7:265-275.
Harrison P, Arosio P, 1996. The ferritins: molecular properties, iron storage function and cellular regulation. Biochem Biophys Acta Bioenergetics 1275: 161-203. Hixson DC, McEntire K, Chesner J, Faris R, Weltman J, Marceau N. 1986. Monoclonal antibody (MAb) recognizing a glycoprotein absent from normal tissues but present on transplantable (THC) and primary (PHC) hepatocellular carcinomas induced by azo dye.
Proceedings of AACR 27:365.
Ikeda W, Kakunaga S, Takekuni K, Shingai T, Satoh ft, et al. 2004. Nectin-like
molecule-5/Tage4 enhances cell migration in an integrin-dependent, Nectin-3
independent manner. J Biol Chem 279: 18015-18025.
Jordan VC, Caplan MR, BennettKM, 2010. Simplified synthesis and relaxometry of magnetoferritin for magnetic resonance imaging. J Magn Med 64: 1260-1266.
Lim YP, Fowler LC, Hixson DC, Wehbe T, Thompson NL, 1996. TuAg. l is the liver isoform of the rat colon tumor-associated antigen pE4 and a member of the immunoglobulin-like supergene family. Cancer Research 56: 43034.
Liu X, Jin W, Theil EC, 2001. Opening protein pores with chaotropes enhances Fe reduction an dchelation of Fe from the ferritin biomaterial. Proc Natl Acad Sci USA 100:3653-3658.
Majmudar S, Zoghbi SS, Gore JC, 1989, The influence of pulse sequences on the relaxation effects of superparamagnetic iron oxide contrast agents. Magn Reson Med 10:289-301.
Majmudar S, Gore JC, 1988. Studies of diffusion in random fields produced by variations in susceptibility. J Magn Reson 78:41-55. Masson D, Jarry A, Baury B, Blanchardie P, Laboisse C, et al. 2001. Overexpression of the CD 155 gene in human colorectal carcinoma. Gut 49: 236-240. Mulder WJM, Strijkers GJ, van Tilborg GAF, Griffioen AW, Nicolay K, 2006. Lipid-based nanoparticles for contrast-enhanced MRI and molecular imaging. NMR Biomed 19: 142-164.
Muller RN, Gillis P, Moiny F, Roch A, 1991 . Transverse relaxivity of particulate MRI contrast media: from theories to experiments. Magn Reson Med 22: 178- 182.
Rozich RA, Mills DR, Brilliant KE, Callanan HM, Yang DQ, Tantravahi U, Hixson DC. 2010. Accumulation of neoplastic traits prior to spontaneous in vitro transformation of rat
cholangiocytes determines susceptibility to activated ErbB-2/Neu. Experimental and Molecular Pathology 89: 248-259.
Sana B, Johnson E, Sheah K, Poh CL, Lim S, 2010. Iron-based ferritin nanocore as a contrast agent. Biointerphases 5(3):FA48-FA52. Sana B, Poh CL, Lim S, 201 1. A manganese-ferritin nanocomposite as an ultrasensitive T2 contrast agent. Chem. Commun., 2012, 48, 862-864.
Sato T, Irie K, Ooshio T, Ikeda W, Takai Y. 2004. Involvement of heterophilic trans-interaction of Necl-5/Tage4/PVR/CD155 with nectin-3 in formation of nectin- and cadherin-based adherens junctions. Genes Cells 9: 791-799.
Sloan KE, Eustace BK, Stewart JK, Zehetmeier C, Torella C, et al. 2004. CD155/PVR plays a key role in cell motility during tumor cell invasion and migration. BMC Cancer 4: 73. Swift J, Butts CA, Cheung-Liu J, Yerubandi V, Dmochowski, 2009. Efficient self-assembly of archaeoglobus fulgidus ferritin around metallic cores. Langmuir 25:5219-5225.
Taylor RM, Huber DL, Monson TC, Abdul-Medhi A, Bisoffe M, Sillerud LO, 201 1.
Multifunctional iron platinum stealth innumomicelles: targeted detection of human prostate cancer cells using both fluorescence and magnetic resonance imaging. J Nanopart Res 13:4717- 4729.
Uchida M, Terashima M, Cunningham CH, Suzuki Y, Willits DA, Willis AF, Yang PC, Tsao PS, McConnell MV, Young MJ, Douglas T, 2008. A human ferritin iron oxide nano-composite magnetic resonance contrast agent. Magn Reson Med 60: 1073-1081.
Uchida M, Flenniken ML, Allen M, Willits DA, Crowley BE, Brumfield S, Willis AF, Jackiw L, Jiilita M, Young MJ, Douglas T, 2006. Targeting of cancer cells with ferromagnetic ferritin cage nanoparticles. J Am Chem Soc 128: 16626-16633.
Weisskoff RM, Zuo CS, Boxerman JL, Rosen BR, 1994. Microscopic susceptibility variation and transverse relaxation: theory and experiment. Magn Reson Med 31 :601-610.
Weissleder R, Cheng HC, Bogdanova A, Bogdanov Jr A, 1997. Magnetically labeled cells can be detected by MR imaging. J Magn Reson Imaging 7:258-263.
Yoshimura H, 2006. Protein-assisted nanoparticle synthesis. Colloids Surf A 282-283:464-470.

Claims

What is claimed is:
1 . A contrast agent for enhanced imaging, the agent comprising high-capacity ferritin
nanoparticles loaded with metal ions and linked to an antibody or epitope thereof, wherein the antibody targets an antigen up-regulated in tumor cells, such that the contrast agent administered to a subject specifically and effectively accumulates at a tumor and the MR1 response of the metal-loaded ferritin results in enhanced imaging of the tumor.
An imaging method comprising the steps of administering or applying to a subject or a cell culture, iron-loaded ferritin nanoparticles linked to targeting material, wherein the targeting material binds to a cell surface molecule that is up-regulated in tumor cells such that the nanoparticles selectively accumulate in the region of the tumor cells, the iron- loaded ferritin material having characteristic Ti, T2 and/or T2 response to enhance MR1 imaging of the tumor cells.
The method of claim 2, further comprising the step of applying an external perturbation such as an alternating magnetic field in a region of the tumor to locally elevate temperature of iron held within or released by the ferritin thereby selectively killing the tumor cells.
The method of claim 2, further comprising the step of initiating release of cytotoxic iron ions from the ferritin to selectively kill the tumor cells.
The method of claim 4, wherein the step of initiating release comprises applying a magnetic exciting field to a region about the tumor such that magnetic field-induced hyperthermia promotes shedding of ionic iron from the ferritin.
The method of claim 3 or 4, wherein the ferritin is a synthetic ferritin forming a cage structure having one or more large pores to enhance shedding of iron held within in the ferritin, and wherein the magnetic field is adapted to promote one or more effects selected from among Neel relaxation heating and ultrasonic kinetic heating from
Brownian-like motion.
The contrast agent of claim 1 or method of claim 2, wherein the ferritin is a synthetic ferritin forming a nanocage structure that holds over 2000, preferably over 5000 iron ions, and the targeting material is an antibody or epitope of an antibody linked to or conjugated to the ferritin.
8. The contrast agent or method of claim 7, wherein the contrast agent is produced as a recombinant fusion protein of ferritin and the epitope of an antibody, and is loaded with iron to form a monodisperse nanoparticle agent for administration to a subject.
9. The contrast agent or method of claim 8, adapted to selectively enhance image contrast either bound to a target tissue or in a free fluid by application of two or more different imaging protocols.
10. The contrast agent of claim 1 or method of claim 2, wherein the ferritin is administered either systemically or locally injected directly to a tumor site.
1 1. An agent for treatment or diagnosis of a disease condition, the agent comprising high capacity iron-loaded ferritin nanoparticles coupled to a disease-targeting portion, wherein the disease-targeting portion preferentially binds to diseased cells such as specific cancer cells so that the agent effectively accumulates at and binds to the diseased cells providing characteristic MRI enhanced response for detection, diagnosis or imaging of the bound cells.
12. The agent of claim 1 1 , engineered to release toxic iron upon external stimulation so as to selectively destroy the bound cells.
13. The agent of claim 12, wherein the external stimulus effects heating and/or vibration of the ferritin nanoparticle bound to cells.
14. The agent of claim 12, wherein the ferritin is engineered to self-assemble from multiple copies of a basic peptide into a nanocage structure that incorporates ionic iron, and to have pores open enabling enhanced release of the incorporated ionic iron for selective and localized tissue destruction.
1 5. A method of diagnosis or treatment of a disease condition, the method comprising administering to a subject an agent comprising high capacity paramagnetic metal particle-loaded ferritin nanoparticles coupled to a disease -targeting portion, wherein the disease-targeting portion preferentially binds to diseased cells such as specific tumor cells so that the agent effectively accumulates at and binds to the cells providing characteristic MRI enhanced response for detection, diagnosis or imaging of a tumor.
16. The method of claim 15, further comprising the step of applying an external stimulus to release metal ions from the high capacity ferritin so as to attacking the tumor and/or destroy diseased cells to which the nanoparticles are bound.
17. The method of claim 15, wherein the ferritin has a capacity of more than about
500, preferably over 3000, over 5000 or about 7000 iron or paramagnetic metal ions, such as a synthetic ferritin derived from Archaeoglobus fulgidus or an exotic organism, modified to form a self- assembling cage having open pores, recombinant produced and wherein the targeting agent includes one or more of an antibody to a disease marker, or to a characteristic cell surface glycoprotein or other cell-related functional targeting agent.
18. The method of claim 15, comprising the step of MRI imaging to identify presence of the cells targeted by the targeting agent, and further comprising the step of applying energy to the imaged region to release cytotoxic ions from the nanoparticles to selectively destroy the cells, wherein the energy may include a rapidly oscillating magnetic field, ultrasound, and electric force field, and/or wherein the cytotoxic ions may include iron or other paramagnetic metal ions or combinations thereof.
19. A method of early detection or treatment of cancer, the method comprising:
coupling to ferritin nanoparticles an antibody or targeting agent that targets and binds to a marker of an invasive or resistant tumor cell of interest;
administering a formulation of the targeting agent/ferritin to a subject so that it selectively, preferentially or effectively binds to a marker of a type of invasive or resistant tumor cell of interest and changes MRI response characteristics of the invasive or resistant tumor cells of interest; and imaging the subject to thereby identify an invasive or resistant tumor.
20. The method of claim 19, further comprising the step of exciting the formulation bound to the tumor to release material from the nanoparticles and treat the tumor.
21. The method of claim 19 or 20, wherein the targeting agent specifically targets cells of an aggressive or invasive tumor, whereby enhanced MRI imaging enables substantially simultaneous detection and treatment at an early stage or prior to substantial growth or metastasis of the tumor.
A method of treatment of cancer, the method comprising:
coupling to a ferritin nanoparticle loaded with a paramagnetic metal core to an antibody or targeting agent that selectively, preferentially or effectively binds to a marker of an invasive or resistant tumor cell of interest;
administering a formulation of the targeting agent/ferritin to a subject so that it selectively, preferentially or effectively binds to a marker of a type of invasive or resistant tumor cell of interest; and
exciting the ferritin nanoparticles to release locally toxic metal from the core to kill the tumor cells.
23. The contrast agent of claim 1 or method of claim 2, wherein the ferritin is a synthetic ferritin forming a nanocage structure that holds substantially more metal ions than a natural ferritin, such as over 2000, preferably over 5000 iron ions or hundreds of manganese ions, and the targeting material is an antibody or epitope of an antibody linked to or conjugated to the ferritin by a short linker such that the ferritin nanocage effectively binds to a targeted tissue, and the ferritin is adapted to locally release a toxic level of free metal ions when a rapidly alternating magnetic field is applied to the tissue.
PCT/US2013/055955 2012-08-21 2013-08-21 Ferritin-based tumor targeting agent, and imaging and treatment methods WO2014031727A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CA2921493A CA2921493A1 (en) 2012-08-21 2013-08-21 Ferritin-based tumor targeting agent, and imaging and treatment methods
US14/626,071 US20150224212A1 (en) 2012-08-21 2015-02-19 Ferritin-based tumor targeting agent, and imaging and treatment methods

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201261691346P 2012-08-21 2012-08-21
US61/691,346 2012-08-21
US201361803955P 2013-03-21 2013-03-21
US61/803,955 2013-03-21

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/626,071 Continuation US20150224212A1 (en) 2012-08-21 2015-02-19 Ferritin-based tumor targeting agent, and imaging and treatment methods

Publications (1)

Publication Number Publication Date
WO2014031727A1 true WO2014031727A1 (en) 2014-02-27

Family

ID=50150358

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/055955 WO2014031727A1 (en) 2012-08-21 2013-08-21 Ferritin-based tumor targeting agent, and imaging and treatment methods

Country Status (3)

Country Link
US (1) US20150224212A1 (en)
CA (1) CA2921493A1 (en)
WO (1) WO2014031727A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016048246A1 (en) * 2014-09-25 2016-03-31 Nanyang Technological University Targeting of melanocytes for delivering therapeutic or diagnostic agents using protein nanocages
WO2016197093A1 (en) 2015-06-05 2016-12-08 Brown University Heat substrate and/or image enhancement compositions and enhanced tissue ablation methods
WO2016207256A1 (en) * 2015-06-22 2016-12-29 Cellis Sp. Z O.O. [Ltd.] Cellular targeted label delivery system
EP3661968A4 (en) * 2017-08-04 2021-05-12 The Hospital for Sick Children Nanoparticle platform for antibody and vaccine delivery
CN114984217A (en) * 2022-06-21 2022-09-02 南京林业大学 Preparation method and application of aptamer ferritin nanoparticles co-loaded with paclitaxel and manganese phthalocyanine
US12016624B2 (en) 2015-12-23 2024-06-25 Rhode Island Hospital Thermal accelerant compositions and methods of use

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018053434A1 (en) 2016-09-16 2018-03-22 The Johns Hopkins University Protein nanocages with enhanced mucus penetration for targeted tissue and intracellular delivery
WO2022196675A1 (en) * 2021-03-16 2022-09-22 味の素株式会社 Complex or salt thereof, and method for manufacturing same
CN113501883A (en) * 2021-07-05 2021-10-15 北京理工大学 Metal ion delivery carrier and preparation method and application thereof
CN113456836B (en) * 2021-07-07 2022-09-16 中国科学院精密测量科学与技术创新研究院 Manganese-heme coordination polymer nanoparticle and preparation method and application thereof
CN118121714A (en) * 2022-12-02 2024-06-04 中国科学院过程工程研究所 Targeting nanoscale particles, targeting cells, preparation method and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030220230A1 (en) * 2002-03-14 2003-11-27 Children's Hospital & Research Center At Oakland Enhancement of iron chelation therapy
US20030219385A1 (en) * 2002-03-07 2003-11-27 Eric Ahrens Contrast agents for magnetic resonance imaging and methods related thereto

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030219385A1 (en) * 2002-03-07 2003-11-27 Eric Ahrens Contrast agents for magnetic resonance imaging and methods related thereto
US20030220230A1 (en) * 2002-03-14 2003-11-27 Children's Hospital & Research Center At Oakland Enhancement of iron chelation therapy

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DEANS, A. E. ET AL.: "Cellular MRI contrast via coexpression of transferrin receptor and ferritin", MAGNETIC RESONANCE IN MEDICINE, vol. 56, 2006, pages 51 - 59 *
SANA, B. ET AL.: "Iron-based ferritin nanocore as a contrast agent", BIOINTERPHASES, vol. 5, no. 3, 2010, pages FA48 - FA52 *
UCHIDA, M. ET AL.: "Targeting of cancer cells with ferrimagnetic ferritin cage nanoparticles", JACS, vol. 128, 2006, pages 16626 - 16633 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016048246A1 (en) * 2014-09-25 2016-03-31 Nanyang Technological University Targeting of melanocytes for delivering therapeutic or diagnostic agents using protein nanocages
US11202820B2 (en) 2014-09-25 2021-12-21 Nanyang Technological University Targeting of melanocytes for delivering therapeutic or diagnostic agents using protein nanocages
WO2016197093A1 (en) 2015-06-05 2016-12-08 Brown University Heat substrate and/or image enhancement compositions and enhanced tissue ablation methods
US11497554B2 (en) 2015-06-05 2022-11-15 Brown University Heat substrate and/or image enhancement compositions and enhanced tissue ablation methods
WO2016207256A1 (en) * 2015-06-22 2016-12-29 Cellis Sp. Z O.O. [Ltd.] Cellular targeted label delivery system
CN108026514A (en) * 2015-06-22 2018-05-11 塞尔丽思股份有限公司 Cell-targeting marker delivery system
US11464879B2 (en) 2015-06-22 2022-10-11 Cellis Ag Cellular targeted active ingredient delivery system
US12016624B2 (en) 2015-12-23 2024-06-25 Rhode Island Hospital Thermal accelerant compositions and methods of use
EP3661968A4 (en) * 2017-08-04 2021-05-12 The Hospital for Sick Children Nanoparticle platform for antibody and vaccine delivery
EP4074734A1 (en) * 2017-08-04 2022-10-19 The Hospital for Sick Children Nanoparticle platform for antibody and vaccine delivery
CN114984217A (en) * 2022-06-21 2022-09-02 南京林业大学 Preparation method and application of aptamer ferritin nanoparticles co-loaded with paclitaxel and manganese phthalocyanine

Also Published As

Publication number Publication date
US20150224212A1 (en) 2015-08-13
CA2921493A1 (en) 2014-02-27

Similar Documents

Publication Publication Date Title
US20150224212A1 (en) Ferritin-based tumor targeting agent, and imaging and treatment methods
Wang et al. Active targeting theranostic iron oxide nanoparticles for MRI and magnetic resonance-guided focused ultrasound ablation of lung cancer
Shevtsov et al. Ionizing radiation improves glioma-specific targeting of superparamagnetic iron oxide nanoparticles conjugated with cmHsp70. 1 monoclonal antibodies (SPION–cmHsp70. 1)
Shevtsov et al. Granzyme B Functionalized Nanoparticles Targeting Membrane Hsp70‐Positive Tumors for Multimodal Cancer Theranostics
Wickline et al. Molecular imaging and therapy of atherosclerosis with targeted nanoparticles
Xie et al. Lactoferrin-conjugated superparamagnetic iron oxide nanoparticles as a specific MRI contrast agent for detection of brain glioma in vivo
Sipkins et al. Detection of tumor angiogenesis in vivo by αvβ3-targeted magnetic resonance imaging
Soenen et al. Magnetoliposomes: versatile innovative nanocolloids for use in biotechnology and biomedicine
Jang et al. Trastuzumab-conjugated liposome-coated fluorescent magnetic nanoparticles to target breast cancer
Harvell-Smith et al. Magnetic particle imaging: tracer development and the biomedical applications of a radiation-free, sensitive, and quantitative imaging modality
Hultman et al. Magnetic resonance imaging of major histocompatibility class II expression in the renal medulla using immunotargeted superparamagnetic iron oxide nanoparticles
US10987436B2 (en) Superparamagnetic nanoparticles as a contrast agent for magnetic resonance imaging (MRI) of magnetic susceptibility (T2*)
KR101468769B1 (en) Magnetic nanocomposite specific for thyroid cancer and use thereof
Su et al. Multimodality imaging of angiogenesis in a rabbit atherosclerotic model by GEBP11 peptide targeted nanoparticles
JP2009519316A (en) Targeting nanoparticles for magnetic resonance imaging
Kubovcikova et al. Poly-L-lysine designed magnetic nanoparticles for combined hyperthermia, magnetic resonance imaging and cancer cell detection
Deddens et al. MRI of ICAM-1 upregulation after stroke: the importance of choosing the appropriate target-specific particulate contrast agent
Shevtsov et al. Brain tumor magnetic targeting and biodistribution of superparamagnetic iron oxide nanoparticles linked with 70-kDa heat shock protein study by nonlinear longitudinal response
Neumaier et al. MR and iron magnetic nanoparticles. Imaging opportunities in preclinical and translational research
Chen et al. Targeted superparamagnetic iron oxide nanoparticles for in vivo magnetic resonance imaging of T-cells in rheumatoid arthritis
Tsuchiya et al. Atherosclerotic imaging using 4 types of superparamagnetic iron oxides: new possibilities for mannan-coated particles
Dassler et al. Current limitations of molecular magnetic resonance imaging for tumors as evaluated with high-relaxivity CD105-specific iron oxide nanoparticles
Xin et al. Coupling Gd‑DTPA with a bispecific, recombinant protein anti‑EGFR‑iRGD complex improves tumor targeting in MRI
Zhang et al. Magnetic resonance imaging tracking and assessing repair function of the bone marrow mesenchymal stem cells transplantation in a rat model of spinal cord injury
Lahooti et al. Preliminary studies of 68Ga-NODA-USPION-BBN as a dual-modality contrast agent for use in positron emission tomography/magnetic resonance imaging

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13831245

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13831245

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2921493

Country of ref document: CA