CA2628106A1 - Magnetic nanoscale particle compositions, and therapeutic methods related thereto - Google Patents

Magnetic nanoscale particle compositions, and therapeutic methods related thereto Download PDF

Info

Publication number
CA2628106A1
CA2628106A1 CA002628106A CA2628106A CA2628106A1 CA 2628106 A1 CA2628106 A1 CA 2628106A1 CA 002628106 A CA002628106 A CA 002628106A CA 2628106 A CA2628106 A CA 2628106A CA 2628106 A1 CA2628106 A1 CA 2628106A1
Authority
CA
Canada
Prior art keywords
marker
antibody
combination
antigen
magnetic
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
CA002628106A
Other languages
French (fr)
Inventor
Robert Ivkov
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Triton Biosystems Inc
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2628106A1 publication Critical patent/CA2628106A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K41/00Medicinal preparations obtained by treating materials with wave energy or particle radiation ; Therapies using these preparations
    • A61K41/0052Thermotherapy; Hyperthermia; Magnetic induction; Induction heating therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6923Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being an inorganic particle, e.g. ceramic particles, silica particles, ferrite or synsorb
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • 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
    • 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
    • A61K49/1887Agglomerates, clusters, i.e. more than one (super)(para)magnetic microparticle or nanoparticle are aggregated or entrapped in the same maxtrix
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/12Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules
    • A61K51/1241Preparations containing radioactive substances for use in therapy or testing in vivo characterised by a special physical form, e.g. emulsion, microcapsules, liposomes, characterized by a special physical form, e.g. emulsions, dispersions, microcapsules particles, powders, lyophilizates, adsorbates, e.g. polymers or resins for adsorption or ion-exchange resins
    • A61K51/1255Granulates, agglomerates, microspheres
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N1/00Electrotherapy; Circuits therefor
    • A61N1/40Applying electric fields by inductive or capacitive coupling ; Applying radio-frequency signals
    • A61N1/403Applying electric fields by inductive or capacitive coupling ; Applying radio-frequency signals for thermotherapy, e.g. hyperthermia
    • A61N1/406Applying electric fields by inductive or capacitive coupling ; Applying radio-frequency signals for thermotherapy, e.g. hyperthermia using implantable thermoseeds or injected particles for localized hyperthermia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N2/00Magnetotherapy
    • A61N2/002Magnetotherapy in combination with another treatment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N2/00Magnetotherapy
    • A61N2/02Magnetotherapy using magnetic fields produced by coils, including single turn loops or electromagnets
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/10X-ray therapy; Gamma-ray therapy; Particle-irradiation therapy
    • A61N5/1001X-ray therapy; Gamma-ray therapy; Particle-irradiation therapy using radiation sources introduced into or applied onto the body; brachytherapy
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/902Specified use of nanostructure
    • Y10S977/904Specified use of nanostructure for medical, immunological, body treatment, or diagnosis

Abstract

Disclosed are thermotherapeutic compositions for treating disease material, and methods of targeted therapy utilizing such compositions. These compositions comprise a) stable single domain magnetic particles; b) magnetic nanoparticles comprising aggregates of superparamagnetic grains; or c) magnetic nanoparticles comprising aggregates of stable single magnetic domain crystals and superparamagnetic grains. These compositions may also comprise a radio isotope, potential radioactive isotope, chemotherapeutic agent. These methods comprise the administration to a patient's body, body part, body fluid, or tissue of bioprobes (energy susceptive materials attached to a target-specific ligand), and the application of energy to the bioprobes so as to destroy, rupture, or inactivate the target in the patient. Energy forms, such as AMF, are utilized to provide the energy. The disclosed methods may be useful in the treatment of a variety of indications, including cancers, diseases of the immune system, central nervous system and vascular system, and pathogen-borne diseases.

Description

MAGNETIC NANOSCALE PARTICLE COMPOSITIONS, AND THERAPEUTIC
METHODS RELATED THERETO

CROSS REFERENCE TO RELATED APPLICATIONS
[0001.] This Application claims priority to U.S. Provisional Applieatipn. No.
60i732,368 filed November 1, 2005 titled "MAGNETIC NANOSCALE PARTICLE COMPOSITIONS, AND
THERAPEUTIC ME'I'HODS RELATED THERETO", the contents of which are incorporated herein by reference.

Technical Field 100021 The present invention relates generally to targeted magnetic nanotherapy compositions and methods, and specifically, to magnetic nanoscale particle compositions that comprise an energy susceptive material that is attached to a target-specific ligand, and therapeutic methods that comprise the administration of such compositions to a patient's body, body part, tissue, or body fluid, and the administration of energy from an energy source, so as to destroy or inactivate the target.

BACKGROUND
[0003] The time between the onset of disease in a patient and the conclusion of a successful course of therapy is often unacceptably long. Many diseases remain asymptomatic and evade detection ,Arhile progressing to advanced, and often terminal, stages. In addition, this period may be marked by significant psychological and physical trauma for the patient due to the unpleasant side effects of even correctly prescribed treatments. Even diseases that are detected early may be most effectively treated only by therapies that disrupt the normal functions of healthy tissue or have other unwanted side effects.
(0004] One such disease is cancer. Despite considerable research effort and some success, cancer is still the second leading cause of death in the United States, claiming more than 500,000 lives each year according to American Cancer Society estimates. Traditional treatments are invasive a.ndlar are attended by harm#'ul side effects (e.g., toxicity to healthy cells), often making for a traumatic course of therapy with only modest succcss. Early detection, a result of better _1_ diagnostic practices and technology, has improved the prognosis for inany patients. However, the suffering that many patients must endure makes for a more stressful course of therapy and may complicate patient compliance with prescribed therapies. F'urther, some cancers defy currently available treatment options, despite improvements in disease detection. Of the many forms ofcan.cer that still pose a medical challenge, prostate, breast, lung, and liver claim the vast majority of lives each year. Colorectal cancer, ovarian cancer, gastric cancer, leukemia, lymphoma, melanoma, and their metastases may also be li.fe threatening.
[0005] Conventional treatments for breast cancer, for example, typically include surgery followed by radiation and/or chemotherapy. 'I'hese techniques are not always effective, and even if effective, they suffer from certain deficiencies, Surgical procedures range Irom removal of only the tumor (lumpectomy) to complete removal of the breast. In early stage cancer, complete removal of the breast may provide an assurance against recurrence, but is disfiguring and requires the patient to make a very difficult choice. Lumpectomy is less disfiguring, but can be associated with a greater risk of cancer recurrence. Radiation therapy and chemotherapy are arduous and are not completely effective against recurrence.
[0006] Treatment of pathogen-based diseases is also not without complications.
Patients presenting symptoms of systemic infection are often mistakenly treated with broad-spectrnm antibiotics as a first step. This course of action is completely ineffective when the invading organism is viral. Even if a bacterium (e.g., E. coli) is the culprit, the antibiotic therapy eliminates not only the offending bacteria, but also benign intestinal flora in the gut that are necessary for proper digestion of food. I-Ience, patients treated in this manner often experience gastrointestinal distress until the benign bacteria can repopulate. In other instances, antibiotic-resistant bacteria may not respond to antibiotic treatment. Therapies for viral diseases often target only the invading viruses themselves. However, the cells that the viruses have invaded and "hijacked" for use in making additional copies of the virus remain viable.
Hence, progression of the disease is delayed, rather than halted.
[0007] For these reasons, it was desirable to provide improved and alternative techniques for treating disease, particularly techniques that are less invasive and traulnatic to the patient than the existing techniques, and effective only locally at targeted sites, such as diseased tissue, pathogens, or other undesirable matter in the body. It was also desirable to provide techniques capable of being perforrned in a single or very few treatment sessions (minimizing patient non-_2_ compliance), with minimal toxicity to the patient, and which could be targeted to the diseased tissues without requiring significant operator skill and input.
(0008] One such alternative technique is immunotherapy, which is a rapidly expanding type of therapy used for treating a variety of human diseases including cancer, for example. The FDA
has approved anum.ber of antibody-based cancer therapeutics. The ability to engineer antibodies, antibody fragments, and peptides with altered properties (e.g., antigen binding affinity, molecular architecture, specificity, valence, etc.) has enhanced their use in therapies.
Cancer immunotherapeutics have made use of advances in the chimerization and humanization of murine antibodies to reduce immunogenic responses in humans. High affinity human antibodies have also been obtained from transgenic animals that contain many human irnmunoglobulin genes. In addition, phage display technology, ribosome display, and DNA
shuffling have allowed for the discovery of antibody fragments and peptides with high affinity and low immunogenicity for use as targeting ligands. All of these advances have made it possible to design an immunotherapy that has a desired antigen binding affinity and specificity, and minimal immune response.
100091 The field of cancer immunotherapy makes use of markers that are over-expressed by cancer cells (relative to normal cells) or expressed only by cancer cells. The identification of such markers is ongoing and the choice of a ligand/marker combination is critical to the success of any immunotherapy. Immunotherapeutics fall into at least three classes: (1) deployment of antibodies that, themselves, target growth receptors, disrupt cytokine pathways, or induce complement or antibody-dependent cytotoxicity; (2) direct arming of antibodies with a toxin, a radionuclide, or a cytokine; (3) indirect arming of antibodies by attaching them to immunoliposomes used to deliver a toxin or by attaching them to an immunological cell effector (bispecific antibodies). Although armed antibodies have shown potent tumor activity in clinical trials, they have also exhibited unacceptably high levels of toxicity to patients.
100101 The disadvantage of therapies that rely on delivery of immunotoxins or radionuclides (i, e., direct and indirect arming) has been that, once administered to the patient, these agents are active at all times. These therapies often cause damage to non-tumor cells and present toxicity issues and delivery challenges. For example, cancer cells commonly shed surface-expressed antigens (targeted by immunotherapeutics) into the blood stream. Immune complexes can be formed between the immunotherapeutic and the shed antigen. As a result, many antibody-based therapies are diluted due to the interaction of the antibody with these shed antigens rather than interacting with the cancer cells, and thereby redu.cing the true delivered dose. Thus, a''th.erapy-on-deanan.d" approach that minimizes adverse side effects and improves efficacy would be preferable.
100111 With thermot.berapy, temperatures in a range from about 40 C to about (hyperthertnia) can cause irreversible damage to disease cells. However, healthy cells are capable of surviving exposure to temperatures up to around 46.5 C. Elevating the temperature ofitadividual cells in diseased tissue to a lethal level (cellular thermotherapy) may provide a superior treatment option. Pathogens implicated in disease and other u.ndesirable matter in the body can also be destroyed via exposure to locally high temperatures.
100121 Temperatures greater than 46 C may also be effective for the treatment of cancer and other diseases by causing an instantaneous thermo-ablative response. 1-lowever, accurate and precise targeting is necessary to ensure that aminirnal amount of healthy tissue is exposed to such temperatures. Failure to achieve such a level of targeting may produce increased detrimental side effects, and thereby reducing the benefits of the treatment.
[00131 Hyperthermia may hold promise as a treatment for cancer and other diseases because it induces instantaneous necrosis (typically referred to as "thermo-ablation") and/or a heat-shock response in cells (classical hyperthermia), leading to cell death via a series of biochemical changes within the cell. State-of-the-art systems that employ microwave or radio frequency (RF) hyperthermia, such as annular phased array systems (APAS), attempt to tune energy for regional heating of deep-seated tumors. Such techniques are limited by the heterogeneities of tissue electrical conductivities and that of highly perfused tissue. Tbis leads to the as-yet-unsolved problems of "hot spot" phenomena in untargeted tissue with concomitant under-dosage in the desired areas. The result is often a lower than expected therapeutic ratio, and an inherent difficulty to determine with adequate precision the heat dose delivered to the desired area. The latter precludes the development of prescriptive clinical protocols, which are necessary to ensure reproducible and predictable patient benefits following treatment. All of these factors make selective heating of specific regions with such systems very difficult.
100141 Another strategy that utilizes RF hyperthermia requires surgical implantation of microwave or RF based antennae or self-regulating thernal seeds. While this approach avoids problems related to dose determination and some of the problems associated with targeting, it ..4_ . . .............. ... ....... ........... . ....

requires an invasive procedure to implant the thermal seeds. In addition to its invasiveness, this approach provides few (if any) options for treatment of metastases because it requires knowledge of the precise location of the primary tumor. The seed implantation strategy is thus incapable of targeting undetected individual cancer cells or cell clusters not immediately adjacent to the primary tumor site. Clinical success of this strategy is hampered by problems with the targeted generation of heat at the desired tumor tissues.
t00151 A. strategy for treating a disease by generating heat within a tumor using superparamagnetic paa-ticles (having characteristic relaxation time,7--10"9 sec) that are suspended in a suitable medium, referred to as magnetic fluids, and exposing the patient to an alternating magnetic field (AMF) has been proposed (see e.g., US 6,541,039 to Lesniak et al, and US
6,470,220 to Kraus, ct aL). While some variations exist, generally the methods disclosed in the prior art involve the introduction of the magnetic fluid directly into the region to be treated and heating the particles by exposing a significant portion of the patient to low amplitude (less than 16kA/m) alternating magnetic fields with frequency of between 50 kHz and 200 kHz, including the region of interest. It is well established that exposing a significant portion of a patient to an AMF will increase tissue temperature over the whole region exposed, and even the core body temperature, significantly because of the eddy currents generated by the interaction of the AMF
with tissues. Indeed, this is the general strategy used with antennae-based or annular phased array RF devices described above. A cancer tumor located within this region would thus experience an elevated temperature even without a magnetic fluid. Tumor temperature increases to a range of about 40 C to 43 "C are reported in some cases. Such tumor temperatures seem low when one considers the relatively large amounts, about 10 mg to 100 mg particles per gram of tumor, of superpararnagnetic particles injected directly into the tumor.
This suggests that a significant portion of the heat is the result of direct AMF effects on tissue (eddy current), with a lesser degree of heat contributed by the presence of the particles.
100161 The magnetic fluids as described comprise non-interacting superparamagnetic particles, whicb are stated to be preferred because of their decreased tendency to aggregate.
Because the magnetic particles comprising the fluid are superparamagnetic, viscous heating is the mechanism giving rise to particle rotation that deposits energy into the medium due to its viscosity, i,e.,. Brownian relaxation. As disclosed in the prior art, superparamagnetic particles are preferred because they will bave zero, or near zero, remanence, and thus a reduced tendency to aggregate, which occurs when their magnetic moments are non-interacting.
Heating via Neel relaxation (magnetic hysteresis) is precluded in this instance, unless the AMF
period is significantly shorter (less than I0-9 sec) than the characteristic relaxation time of the particle magnetic moments. Thus, magnetic hysteresis heating with an AMF is only possible if the AMF
frequency is greater than I GHz. For methods involving the compositions of the magnetic (superparamagnetic) fluids described, and the typical AMF frequencies disclosed therein (about I00 Wz), there is no possible contribution of heating via Neel relaxation.

SUMMARY OF THE INVENTION
[0017) Hyperthermia for treatment of disease using magnetic fluids exposed to RF fields has been recognized for several decades. However, a major problem with magnetic fluid hypertberm.ia has been the inability to selectively deliver a lethal dose of particles to the cells or pathogens of interest, particularly when the composition is limited to particles possessing characteristic relaxation times much shorter than the period of the applied RF.
100181 The biology of heat damage to cells is well understood, as is the clinical potential that a suitably targeted heating approach holds for the treatment of disease. This results from either the cytotoxic effect of heat, or the enhanced cytotoxic effect of radiation or chemotherapy resulting from heat sensitization of the cell when heat is com.biiied with these treatments, or it can a combination of these. Heat applied to a cell in combination with ionizing radiation, such as ultraviolet, x-ray, gamma, beta, alpha, neutron, etc., or chemotherapy often results in an enhanced cytotoxic effect that may be significantly greater than expected from an additive combination of the ionizing energy or chemotherapy doses. Often, a cell will exhibit a high level of susceptibility to an otherwise sub-lethal dose of either chemotherapeutic agent or ionizing radiation when that dose is combined with heat, also at sub-lethal dose, in some combination.
Such a combination therapy has a demonstrable significant clinical potential because damaging side effects from a dose of either heat or ionizing radiation may be avoided.
Suitably targeting the combined form oftrcatn-ient, r'.e., in the bioprobes, thus has significant advantages over untargeted applications of either treatment modality or their combination.
[00191 In view of the above, there is a need for thermotherapeutic compositions for treating diseased tissue, pathogens, or other undesirable matter, that comprise a) stable single domain magnetic particles (characteristic relaxation time greater than l09 see); b) magnetic nanoparticles comprising aggregates of superparamagnetic grains where the interacting magnetic moments create a collective state possessing characteristic relaxation times that are matched to the period of magnetic fields applied to a target within a patient's body; or c} magnetic nanoparticles comprising aggregates of stable single magnetic domain crystals and superparamagnetic grains, where the interactions of the stable single domain and super.pararrsagnetic znagnetic moments result in a collective state that increases the superparainagnetic characteristic relaxation time to a value much greater than I O"9 sec. It is also desirable to have hyperthermia-based treatment methods that incorporate selective delivery of such thermotherapeutic compositions, and that are safe and effective, short in duration, and require minimal invasion.
(00201 It is, therefore, an object of the present invention to provide a thermotherapeutic magnetic composition for treating disease material that comprises a) stable single domain magnetic particles (characteristic relaxation time greater than 109 sec}; b) magnetic nanoparticles comprising aggregates of superpararnagiietic grains where the interacting magnetic moments create a collective state possessing characteristic relaxation times that are matched to the period of magnetic fields applied to a target within a patient's body; or c) magnetic nanoparticles comprising aggregates of stable single magnetic domain crystals and superparatnagnetic grains, where the interactions of the stable single domain and superparamagnetic magnetic moments result in a collective state that increases the superparamagnetic characteristic relaxation time to a value much greater than IO-9 sec.
100211 It is anotber object of the present invention to provide a method for treating disease material, that comprises selective delivery of such tlicrmotherapeutic compositions, and that are safe and effective, short in duration., and require minimal invasion.
100221 It is another bb,ject of the present invention to provide a treatment method that involves the administration of a magnetic material composition, that comprises stable single domain magnetic grains, or aggregates of magnetically coupled superparamagnetic grains, with characteristic relaxation time of the collective magnetic state appropriately tuned for heating via magnetic hysteresis losses, or aggregates of magnetically coupled stable single domain and superparamagnetic grains, attached to a target-specific li.gand., to a patient, and the application of an alternating magnetic field to inductively heat the magnetic material composition via magnetic hysteresis losses, e.g., Neel relaxation.

..7..

[0023] It is another object of the present invention to provide such a treatment method that comprises the detection of at least one location of accumulation of the magnetic material composition within the patient's body prior to the application of an alternating magnetic field.
[0024] It is another object of the present invention to provide such a treatment method that comprises the application of the alternating magnetic field when the magnetic material composition is outside of the patient's body.
[0025] It is another object of the present invention to provide a treatment method that involves the induction of a desired pathological effect by inductively heating the magnetic material to cause necrosis, apoptosis, or deactivation of disease material.
100261 It is another object of the present invention to provide a composition and a treatment method that combine the benefits of hyperthermia, radiation, chemotherapy within the nanoparticle composition.
100271 It is yet another object of the present invention to provide a method for administration of a magnetic material composition, which may be intraperitoneal injection, intravascular injection, intramuscular injection, subcutaneous injection, topical, inhalation, ingestion, rectal insertion, wash, lavage, rinse, or extracorporeal administration into a patient's bodily materials.
100281 It is a further object of the present invention to provide methods for the treatment of tissue in a safe and effective manner, with minimal invasion, and short treatment periods.
100291 The present invention pertains to thermotherapeutic magnetic compositions for treating disease material. In one embodiment, the composition comprises single-domain magnetic particles (characteristic relaxation time greater than 109 see) attached to a target-specific ligand. In another embodiment, the composition comprises magnetic nanoparticles comprising aggregates of superparamagnetic grains, where the interacting magnetic moments create a collective state possessing characteristic relaxation times that are matched to the period of magnetic fields applied to a target within a patient's body. In another embodiment, the composition comprises magnetic nanoparticles comprising aggregates of stable single magnetic domain crystals and superparamagnetic grains, where the interactions of the stable single domain and superparamagnetic magnetic moments result in a collective state that increases the superparamagnetic characteristic relaxation time to a value much greater than I O') sec.
[00301 The present invention pertains to methods for treating diseased tissue, pathogens, or other undesirable matter in a patient. In one embodiment, the treatment method comprises the administration of a thermotherapeutic magnetic composition that comprises single-domain magnetic particles attached to a target-specific ligand, to a patient, and the application of an alternating magnetic field to inductively heat the magnetic material composition. The thermotlaerapeutic magnetic composition may also be administered by administering the ligand and the magnetic particle separately to the patient, and then combining the ligand and the magnetic particle in the patient's body.
100311 In another embodiment, the treatment method comprises the administration of a thermotherapeutic magnetic compositinn, to a patient, detecting at least one location of accumulation of the magnetic composition within the patient's body, and the application of an alternating magnetic field to inductively heat the magnetic contpositian.
100321 In another embodiment, the treatment method comprises the administration of a thermotherapeutic magnetic composition to a patient, and the application of an alternating magnetic field to induce a desired pathological effect by inductively heating the magnetic composition to cause a necrosis, an apoptosis, or a pathogen deactivation.
[0033] I.n another embodiment, the treatment method comprises the administration of a thermotherapeuYic magnetic composition, which may be via intraperitoneal injection, intravascular injection, intramuscular injection, subcutaneous injection, topical, inhalation, ingestion, rectal insertion, wash, lavage or rinse perisurgically, or extracorporeal administration into patient's bodily materiaJs, 100341 Any of the disclosed embodiments for a treatment method may comprise the monitoring of at least one physical characteristic of a portion of a patient.
100351 In any of the disclosed embodiments for a treatment method, the predetermined target may be associated with diseases, such as cancer, diseases of the immune system, pathogen-borne diseases, and undesirable targets, such as toxins, reactions to organ transplants, hormone-related diseases, and non-cancerous diseased cells or tissue.
[0036] The above summary of the present invention is not intended to describe each illustrated embodiment or every implementation of the present invention. The figures and the detailed description which follow particularly exemplify these embodiments.

BRIEF DESCRIPTION OF THE DRAWINGS

[0037] The invention may be more completely understood in consideration of the following detailed description of various embodiments of the invention in connection with the accompanying drawings, in which:

100381 FIG. 1 schematically illustrates a bioprobe configuration, according to an embodiment of the present invention;
[00391 FIG. 2 schematically illustrates target specific baoprobes bound to a disease cell surface, according to an embodinaent of the present invention;
100401 FIG. 3 schematically illustrates a therapy system, according to an embodiment of the present invention;
[00411 FIG. 4 schematically illustrates a therapy system, according to an embodiment of the present invention; and 100421 FIG. 5 schematically illustrates a cross-sectional view of a solenoid coil used as an AMF energy source.

100431 While the invention is amenable to vaxious modifications and alternative forms, specifics thereof have been shown by way of example in the drawings and will be described in detail. It should be understood, however, that the intention is not to limit the invention to the particular embodiments described. On the contrary, the intention is to cover all modifications, equivalents, and alternatives falling within the spirit and scope of the invention as defined by the appended claims.

DETAILED DESCRIPTION OF EXEMPLARY EMBODIMENTS
100441 The present invention pertains to theamotherapeutic compositions for treating disease material, and methods for treating disease material utilizing such compositions, The compositions and methods of the present invention may be used for the treatment of a variety of indications, including cancer. The methods of therapy using the disclosed compositions comprise the administration to a patient a therapeutic magnetic composition that comprise bioprobes, and the application of an alternating magnetic field to an area of'the patient containing the magnetic particle composition to heat the bioprobes sufficiently to kill targeted cells. The present invention, or aspects thereof, may be amenable to use in conjunction with suitable devices or apparatus.

DEFINITIONS
[0045] The term "AMF" ((an abbreviation for altemating magnetic field), as used herein, refers to a magnetic field that changes the direction of its field vector periodically, typically in a sinusoidal, triangular, rectangular or similar shape pattern, with a frequency of in the range of from about 80 kHz to about 800 kHz. The AMF may also be added to a static magnetic field, such that only the AMF component of the resulting magnetic field vector changes direction. It will be appreciated that an alternating magnetic field is accompanied by an alternating electric field and is electromagnetic in nature.
100461 The term "bioprobe", as used herein, refers to a targeted nanoparticle comprising a magnetic nanoparticle core, coating, linker, and targeting ligand, that is used to selectively treat tissue by heating in response to an altemating magnetic field (AM.F).
Additionally, the bioprobe may comprise a radioactive source or species that may become radioactive when exposed to an appropriate energy source. Yttrium-90 (90Y) is an exemplary radioactive souree. Boron-l0 (t B) is an exemplary species that may become radioactive when exposed to a suitable energy source and thereby becomes radioactive. BoronT10 (10B) is becomes radioactive when exposed to a beam of neutrons because it possesses a high neutron absorption cross-section, and becomes radioactive upon capture of a neutron. The bioprobe may also comprise a chemotherapeutic agent. Doxorubicin is an exemplary chemotherapeutic agent.
[0047] The term "bioprobe system", as used herein, refers to a bioprobe specific to a target that is optionally identified via a marker.
100481 The term "coating", as used herein, refers to a material, combination of materials, or covering of the magnetic nanoparticle, comprising a suitable biocompatible material that serves to affect in vivo transport of the bioprobe throughout the patient, and facilitates uptake and retention by diseased tissues and cell. A combination of dex.tra:n and polyethylene glycol is an exemplary coating.
[0049] The term "disease material", as used herein, refers to tissue or cells associated with cancer of any type, such as bone marrow, lung, vascular, neuro, colon, ovarian, breast and prostate cancer; diseases of the immune system, such as AIDS; pathogen-borne diseases, which can be bacterial, viral, parasitic, or fungal, examples of pathoben-borne diseases include HIV, .. .. ........ ..... . ...... .. .. .. ...

tuberculosis and malaria; hormone-related diseases, such as obesity; vascular system diseases;
central nervous system diseases, such as multiple sclerosis; and undesirable matter, such as adverse angipgenesis, restenosis, amyloidosis, toxins, reaction-by-products associated with organ transplants, and other abtiormal cell or tissue growth.
100501 The tem-a "duty cycle", as used herein, refers to the ratio of the time that the energy source is on to the total time that the energy source is on and off in one on-off cycle.
[00511 The term "energy source", as used herein, refers to a device that is capable of delivering energy, of a form other than A.MF, to the bioprobe for the puupose of activating a potential radioaetive sotiarce comprising the bioprobe.
100521 The term "indication", as used herein, refers to a medical condition, such as a disease.
Breast cancer is an exemplary indication.
100531 The terrn "ligand", as used herein, refers to a molecule or compound that attaches to a bioprobe and targets and attaches to a biological marker. A monoclonal antibody specific for HER-2 (an epidermal growth factor receptor protein) is an exemplary ligand.
100541 The terms "linker" or "linker molecule," as used herein, refer to an agent that targets particular functional groups on a ligand and on a magnetic particle or a coating, and thus forms a covalent link between any two of these.
j00551 The term "magnetic nanoparticle", as used herein, refers to aggregates of magnetically coupled superparamagnetic grains, with characteristic relaxation time of the collective magnetic state appropriately tuned to inductively heat via magnetic hysteresis losses, e.g., Neel relaxation, in aii alternating magnttie field.
100561 The term "marker", as used herein, refers to an antigen or other substance to which the bioprobe ligand is specific. HER-2 protein is an exemplary marker.
[0057] The term "target", as used herein, refers to the matter for which deactivation., rupture, disruption or destruction is desired, such as a di.seased cell, a pathogen, or other undesirable matter. A marker may be attached to the target. Breast cancer cells are exemplary targets.

The Targeted Therapv S s~te,,.rn [0058) The targeted therapy system of the present invention involves the utilization of a bioprobe system in conjunction with at least one energy source to treat an indication.

The Bio rÃabe S -stem.

100591 Various embodiments of the bioprobe system of the present invention are demonstrated via FIG. I and FIG. 2. FIG, I illustrates a bioprobe configuration according to an embodiment of the present invention, wherein a bioprobe 100 comprises a magnetic nanoparticle 110. The magnetic nanoparticle 110 may comprise a coating 120. Heat may be generated in the magnetic nanoparticle 110 when the magnetic nanoparticle 110 is exposed to an AMF source through hysteresis. ln addition, the magnetic nanoparticle may comprise a radioactive element, or a suitable isotope of an element that will become radioactive when exposed to a suitable, non-AMF, energy source.

Magnetic Nanoparticle General Properties of Fine Magnetic Grains: Sin Ie Domains - Stable and Unstable [00601 Nanoscale (having dimensions less than 1 m) magnetic materials possess many enhanced properties, such as ren-ian.ence and coereivity, as compared with their conventional or bulk counterparts. With decreasing grain size,an increasing fraction of atoms comprising the grain lieon the surface and interface regions. This increases the significance of the effect of the surface and interface electronic structure on the magnetic properties. The intrinsic magnetic properties of a material, such as spontaneous magnetization and magnetocrystalline anisotropy, are strongly influenced by the grain (crystal) size. Magnetocrystalline anisotropy is only one contribution to the total anisotropy energy of a single domain grain.
Additional contributions may arise from magnetostatic, shape, stress, and surface anisotropies. The last of these is closely related to the detailed chemical nature of surface and grain boundary that become increasingly important as the size is reduced.

[0061] The total anisotropy energy may increase with decreasing grain size, within a certain size range, because of the growing surface anisotropy contribution. It is many of these enbanced magnetic properties that make nanoscale magnetic materials particularly well suited for a variety of applications. For the invention described herein, the enhanced magnetocrystalline anisotropy contributes to increased hysteretic losses of these materials when they are subjected to alternating magnetic fields (AMF), which in turn, results in much higher specific absorption rates (SAR), or heating. Indeed, nanoscale magnetic materials, within a certain size, can produce significantly higher SAR values for a given mass of material under particular AMF conditioiis because the total. anisotropy energy may increase with decreasing grain size, due to the growing surface anisotropy contribution when compared witli their bulk counterparts.
Single Non-interacting Grains 100621 It is understood that a large magnetic body is divided into tin.iforrnly magnetized regions, referred to as domains, that are separated by domain walls (Bloch walls) in order to minimize its magnetostatic energy. However, the energy that must be minimized is the total energy, including magnetostatic, exchange, and anisotropic components, as well as the domain wall contribution. Therefore, it is the final balance of energies that determines both the number and shape of magnetic domains within a magnetic material. As the size of a material is reduced, the size of domains will also be reduced. In addition, the structure of the domains may be changed as will the domain wall width and structure. There is an energy cost associated with domain wall formation; hence the resulting total energy balance will limit the subdivision of domains to a certain optirnum number and size.
100631 There is a corresponding lower limit in the grain size, below which a sin.gle-domain structure exists, because the energy increase due to the formation of domain walls is higher than the energy decrease obtained by dividing the single domain into smaller domains. For many magnetic materials, the dimensional limit is in the range of from about 2 nm to about 800 nm, depending on the spontaneous magnetization and on the anisotropy and exchange energies.
[00641 For a magnetic body that possesses only a single magnetic domain, i.e., a single magnetic domain grain, the behavior of the magnetic moment, m, a vector defining magnitude and direction of magnetization, of the magnetic domain with respect to time and its environment (temperature, extern.al magnetic field, etc.) is governed by the total anisotropy energy of the magnetic grain. The orientation of the magnetic moment with respect to the crystalline axes is an explicit product of the anisotropy energy and physical environment, both past and present, in which the grain is placed. Among the environmental variables of general interest to the in.vention disclosed herein, are the presence and nature of a time varying magnetic field (AMF) and temperature. Specifically, it is the maniier of response exhibited by the spatial orientation (reversal) of m in a single magnetic domain grain when placed in the presence of an AMF that determines hysteresis losses and consequently heat generatcd (SAR).
f00651 The potential oI'a single magnetic domain grain (crystal) to generate heat via hysteresis losses when exposed to an alternating magnetic field is determined by the balance of 14..

energies within the grain that leads to a total anisotropy energy. "1'his sum of anisotropy energies presents an energy barrier, ER, to changes in orientation of the magnetic moment, m, a vector representing both direction and magnitude of the net magnetism of the grain.
Thus, the stability ot'tn with respect to time increases with increasing values of Eq. The grain volurne, 1; and En combine to define a characteristic relaxation time, rf~, which is the time required for spontaneous fluctuations. or relaxations, in the direction. of m to some beginning value after it has been forcibly reoriented by a sufficiently strong magnetic field. Stated in another way, -cg may be considered an intrinsic property of the grain and depends upon various parameters such as composition, volume, skzape, etc. of the grain, and upon symmetries within the grain and upon the relaxation pathways available to m.
[00661 The amount of heat realized through hysteresis losses of a single domain grain when exposed to an alternating magnetic field is the result of a combination of both the intrinsic properties of the grain and experimental conditions. Experimental temperature will determine the relative difference between Eu and energy available to the system, thus setting an experimental relaxation time, orz This relationship may be defined mathematically as:

r = r. exp( ~~ ). (1) 100671 Thus, the relationship of the period of oscillation of the AMF, l/v, with rbecomes a critical experimentally observable quantity that leads directly to the amount of heat generated through hysteresis losses. Here, v is the frequency of oscillation of the AMF.
For 1 jv z-, the moment appears unblocked and spontaneously overcomes ER and reorients randomly without exhibiting hysteresis losses, i.e., no heat will be generated. Conversely, if 1lv r, the moment appears blocked and resists changes in orientation. With a sufficient magnitude of the AMF, rri is forced to overcome E13 and heat is released during the change.
[0068] Anisotropy energy, or potential hysteretic loss, in a single domain grain is proportional, in first approximation, to the volume of the grain. Thus, for large single magnetic domain grains the anisotropy energy may be so high that the energy barrier for r-nagnetication reversal cannot be overcome by thermal energies for any temperature below the material's Curie temperature. Thermal energy is defined by the product kT where k is the Boltzmann constant and T is temperature in Kelvin. Such a single domain magnetic grain is said to have a stable single domain because magnetic moment does not tluctuate, and it can be said to exhibit intrinsically stable magnetic domain behavior with respect to time.
Magnetization reversal may occur in such an intrinsically stable magnetic single domain if the grain is exposed to an external magnetic field that is sufficiently strong to overcome the anisotropy energy, and force a change or reversal of the ma.gnetization vector (magnetic moment). Because the anisotropy energy represents a barrier to rotation of the magnetic moment, such a spatial change in this vector is accompanied by a release of energy in the form of heat. The amount of heat released is proportional, in a first approximation, to the anisotropy energy.
100691 If the magnetic field is removed, the magnetic moment will retain the orientation imprinted by the magnetic field for a characteristic time. The time required for such an orientation change of the magnetic moment to occur after the field is removed is a relaxation time that is characteristic of the grain and is a consequence of both the anisotropy energy of the grain and U. In the extreme case of intrinsically stable magnetic single domain grains, this time is greater than 10 9 seconds. Hence, the magnetic moment appears blocked because the anisotropy energy presents an insurmountable barrier to spontaneous rotations of the magnetic spin system, for all temperatures up to the material Curie temperature. The Curie (or Neel) temperature is defined as the temperature at which a transition from ferromagnetic to parania.gnetic state occurs.
100701 Conversely, as the volume of a grain decreases within the single domain regime, so does the anisotropy energy. Below a certain characteristic grain size, the anisotropy energy may become so low as to be comparable to or lower than kT for any value of T above zero. This implies that the energy barrier for magnetization reversal may be overcome, and then the total magnetic moment of the grain can thermally fluctuate, similar to a single spin in a paramagnetic material. In this case, the entire spin system may spontaneously rotate, the spins within the single-domain particles remaining magnetically coupled. 'T'he orientation of the magnetization vector with respect to the crystalline axes fluctuates and is intrinsically un.stable,. This is superparamagnetism because of the similarity to paramagnetism observed in bulk materials.
Such a magnetic single domain grain may be said to possess an intrinsically unstable single domain., or be intrinsically superparamagnetie.

lb-[0071] Exposing a superpaxainagnetic grain to an external magnetic field will cause the magnetic moment to align in the direction of the magnetic field vector, but with no concomitant release of energy. Because the anisotropy energy is lower than kT for any experimental temperature measurably above zero, it does not present a barrier to magnetization reversai.
When the magnetic field is removed from the grain, the spontaneous fluctuations of the orientation of the magnetic moment will rapidly destroy any imprint imposed by the external magnetic field. The characteristic relaxation time of an intrinsically superparamagnetic grain is very short, typically of order 1 Q'9 seconds. In contrast to the intrinsically stable single domain grain, the magnetic moment of an intrinsically superparamagnetic material is unblocked at all experimental temperatures, and for all time dependent measurements for which the time is longer than the characteristic relaxation time.
[0072] Betvs.Teen these two extremes lies a range of grain volumes for which the anisotropy energy is intermediate, and thus the time scale of magnetization reversal depends explicitly upon the temperature and time scale. of measurements. Thus, for a given grain volume, the relaxation time is defined by temperature, and hence the magnetic moment may appear blocked if the measurement time is shorter than the characteristic relaxation time. In this case, the material will exhibit behavior similar to a stable single domain and will generate heat if placed in an AMF
with a period that is shorter than the characteristic relaxation time. Such a material may be defined as blocked and apparently stable single domain under these conditions.
[0073] Conversely, if the measurement time, i.e., AMF period, exceeds the characteristic relaxation time of the grain unblocked, or apparently superparamagnetic behavior will be observed. Because the characteristic relaxation time in this instance is much shorter than the time of measurement, or AMF period, magnetization reorientation and even reversal occurs randomly with no apparent impedance due to anisotropy energy barriers, and hence no concomitant release of heat.
[0074] Teniperature is also critically important to distinguishing apparently stable single domain, or blocked, behavior from apparently superpararnagnetic, or unblocked, behavior. "I'bus, by analogy, the characteristic relaxation time of the magnetic moment of a grain with specified volume possessing a single magnetic domain will appear blocked when exposed to an AMF of fixed period if the experimental temperature, Te,,F;, is below a characteristic value. If T,xp is increased to a value above this characteristic temperature, the magnetic moment appears . . .......... .. ... . ....... .. ... ....

unblocked when exposed to an AMF of the same fixed period. This characteristic temperature may be defined as the blocking temperature, Tj. Thus, when a grain possessing a single magnetic domain is placed within an AMF oftixed frequency, the forced oscillations of the magnetic moment will release heat while the grain temperature is below the blocking temperature. Once the grain temperature exceeds the blocking temperature, the magnetic moment becomes unblocked, and any release of heat with further exposure to the AMF will cease. This is because the thermal energy, defined by kT, exceeds the anisotropy energy, thereby providing an excess of energy to the spin system to surmount the magnetocrystalline energy barrier.
Slystems of Non-interacting Cirair~s 100751 The above describes the behavior of an individual single magnetic domain grain of specified composition and volume. A bioprobe may comprise a magnetic nanoparticle that is itself an aggregate of more than one single domain magnetic grain. Further, the bioprobes may comprise a suspension of more than one bioprobe suspended in a suitable, and preferably non-magnetic, medium. Thus, the bioprobe suspension may be comprised of individual bioprobes of varying size, centered on a mean with a distribution. In turn, each bioprobe may comprise a magnetic nanoparticle comprised of more than one single magnetic domain grain that vary in volume, also centered about a mean with a distribution.
[00761 A full theoretical description of relaxation time and consequent hysteresis losses and generated heat in an applied AMF for a bioprobe suspension will necessitate inclusion of many more factors than those necessary to describe the behavior of individual single magnetic domain grains with specified volume, i.e., ensemble of monodisperse grains with non-interacting moments. Because volume is an intrinsic property of a single magnetic domain grain that directly affects EB a determination of z-,g and z, for an ensemble of grains comprising grains with varying volume requires knowledge of the size distribution. While the mean volume may be associated with a value of E,9 sufficient to block m at a specified temperature and AMF
frequency, there may be a sizable fraction of grains in the ensemble with volume and E,y significantly lower. The net effect may result in a measured heat output that may be significantly lower than that expected from knowledge of the mean volume alone. The converse also may be demonstrated. An ensemble of grains may possess a mean volume for which the value of EB is lower than that required to block m, i.e., it appears super-paramagnetic.
Consequently, the system would not be expected to exhibit hysteresis in an AMF with specified frequency at a specified temperature. However, this ensemble may contain a sizable number of grains with volume significantly greater than the mean. This component of the ensemble may appear blocked, resulting in hysteresis and a measurable quantity of heat that is generated when exposed to the experimental AMF with specific frequency at a specified temperature.
The particular amount of heat generated may be substantial and apparently contradicting what might be predicted from knowledge of only the mean grain volume.
100771 Interparticle interactions is another factor that is necessary to fully describe the hysteresis behavior of an ensemble of bioprobes comprising an ensemble of single magnetic domain grains. Magnetic forces are, by definition, long-range forces. That is, the range of influence may extend far beyond the boundary of a magnctic grain. Thus, a magnetic nanoparticle comprised of more than one single domain magnetic grain may exhibit properties greater than the sum of the magnetic properties of each grain, because of the additional contribution to the anisotropy energy that result from the collective state, i. e., interaction contributions of each domain m with others.
[00781 It has been experimentally established that interaction effects modify anisotropy energies to produce a collective state that may exhibit behavior uncharacteristic of the state of the individual grains, if non-interacting. It has often been observed that the result is an apparently increased EB, resulting in an inhomogeneous blocking process. Thus, a magnetic nanoparticle comprised of a cluster of superparamagnetic grains may appear blocked, and even exhibit hysteresis, under appropriate experimental conditions. Because the blocking process is inhomogencous, the hysteresis behavior may be considerably weaker than a single domain grain of volume comparable to the aggregate. Such an aggregate cannot be defined as either superparamagnetic or stable single domain, because it is neither in a strict sense under all conditions. Further, each of the grains comprising the aggregate is superparamagnetic in all respects and cannot exhibit hysteresis.

100791 Precise definition and full characterization of a magnetic nanoparticle comprised of an aggregate of single magnetic domain grains, i. c., a combination of stable and superparamagnetic, or purely superparamagnetic, rnay be difficult and impractical because many measurement techniques are necessary for cbaracterization. Indeed, results of some of these measurements may be inconclusive or even contradictory. Nevertheless, a practical measure does exist to define apparent behavior for the purpose of the invention described herein. An ensemble of bioprobes, wherein each bioprobe may be coinprised of an ensemble of magnetic nanoparticles, and each magnetic nanoparticle, further comprised of either individual grains or an aggregate of grains, can be defined by the aggregate mean anisotropy energy, which then defines the mean characteristic relaxation time and mean behavior in a specific AMF
given a specific temperature. Therefore, it is possible to define the magnetic properties of such an ensemble of bioprobes in a relatively simple manner. Based on experimental temperatures between. 270 K and 380 K, and exposure to an AMF with a frequency in the range of from about 100 kHz to about 600 kHz and an amplitude in the range of from about 7.98 k.A/m to about 104 kA/m, measurement of the SA.R is used to distinguish the apparently blocked from the apparently unblocked behavior of the entire ensemble. An ensemble of unblocked, or apparently superparamagnetic, particles will generate less than 10 W/g particle under the specified conditions. By comparison, an ensemble of non-interacting intrinsically superparamagnetic nanoparticles will generate exactly 0W/g particles, by definition. Conversely, apparently blocked bioprobes will generate between 10 W/g to 150 W/g particle. Further, an ensemble of intrinsically blocked, or stable single domain, particles will generate greater than 150 W/g particle under the specified conditions via hysteresis heating, even though some supetparamagnetic contamination may exist.
Radioactive Isotopes [00$01 Because of the synergistic effects of radiation and heat for treating a disease, particularly cancer, the effectiveness of targeted thermotherapy may be significantly enhanced if the bioprobes comprise, a radioactive isotope. In this manner, the radiation may be delivered at typical doses, from about 20 Gy to about 60 Gy, or preferably at sub-lethal doses (less than 20 Gy) and become lethal only after the thermotherapy has been initiated or completed. The dose level of radiation may be controlled by controlled incorporation of the radioactive isotope in the bioprobe composition. Further controls of the radiation dose may be achieved via the use of a bioprobe suspension that comprises a mixture of radioactively-labeled bioprobes with "unlabeled" bioprobes. Any radioactive isotope currently in use for the treatment of disease, or those developed in the future, may be suitable for use herein to enhance the therapeutic ratio of the targeted thermotherapy. Examples of suitable radioactive isotopes are, but not limited to, iodine-131, cobalt-60, iridiun-192, yttrium-90, strontium-89, sama.raurn-153, rhenium-186, and technetium-99m.
Potentially Radiaactive Isoto~es 100811 Certain isotopes comprise unstable nuclei (non-radioactive) that possess a high absorption cross-sections for certain subatomic particles, i. e. , neutrons, protons, etc., and for certain forms of ionizing radiation, i.e., x-rays. When the nuclei of these isotopes absorb the radiation or particle, the nucleus becomes unstable and thus emits radiation as it decays. This phenomenon has already been recognized and used for the treatment of cancer.
"I"be most notable example is boron (boron-10) neutron capture therapy for the treatment of cancer.
[0082] Such treatments have not gained wider acceptance because the level of radiation emitted by the isotope may be insufficient to produce a lethal dose to a tumor. However, as a component of a bioprobe, the delivery of the isotope is both targeted and its radiation initiated only in a sequence with thermotherapy that will maximize the synergy of the two energy for-ns.
Consequently, the radiation becomes more cytotoxic when used in combination with thennotherapy.
100831 Other isotopes possessing high neutron absorption cross sections include many of the lanthanides, such as samarium-149, gadolinium-157, and gadolinium-155.
Samarium is particularly advantageous in that it is magnetic, and its incorporation. into the magnetic nanoparticle crystalline structure may enhance the magnetic properties of the nanoparticle.
Chemotherapeutic agents [00841 As with radioactive isotopes, bioprobes comprising chemotherapeutic agents may synergistically combine with the targeted thermotherapy to enhance a therapeutic outcome.
Examples of chemotherapeutic agents suitable for use herein include, but not limited to, doxorubicin, platinum complexes, such as cisplatin, etc.

I171a ln 15otfl es [0085[ There are various techniques of imaging isotopes that are suitable for use herein, partictilarly MRI, PET, SPECT, and Bioimpedance.
[0085] Small paramagnetic or superparamag;netic particles of ferrite (iron oxide Fe304 or Fe?O3) can be used as paramagnetic contrast medium in magnetic resonance imaging (MRI).
These agents exhibit strong Tl relaxation properties, and due to susceptibility differences to their surroundings, they also produce a strongly varying local magnetic field that enhances 'I'2 relaxation to darken the contrast media-containing structures. Very small particles of less than 300 nanometers also remain intravascular for a prolonged period of time. The agents are also referred to as SPIO's ("small particle iron oxides" or "superparamagnetic iron oxides") and USPIO's (':ultrasmall particle iron oxides" or "ultrasmall superparamagnetic iron oxides"). In one embodiment of the present invention, targeted thermotherapy and MRI are combined. MRI
contrast isotopes that target vulnerable plaques, such as Gadolinium-labeled antifibrin nanoparticles, are used. Once these nanoparticles are uptaken by the plaque, AMF is used for destroying the plaque.

[0087] Positron emission tomography (PET) is a technique for measuring the concentrations of positron-emitting radioisotopes within the tissue of living patients. A
wide range of compounds can be used with PET. Tbese positron-emitting radionuclides have short half-lives and high radiation energies. The primary positron- emitting radionuclides used in PET include Carbon-11, Nitrogen- 13, Oxygen- 15, and Fluorine-18, with half-lives of 20 min, 10 m.ixx, 2 m.in, and 110 min, respectively. These compounds are commonly known in PET as tracer compounds.
[0088] Single photon emission computed tomography (SPECT) involves the detection of gamma rays emitted singly from radioactive atoms, called radionuclides, such as Technetium-99m and Tb.allium-20I. A radiopharmaccutical is a protein or an organic rnolecule that has a radionuclide attached to it. The proteins and organic molecules are selected based on their use or absorption properties within the human body. SPECT is used routinely to help diagnose and stage cancer, stroke, liver disease, lung disease and a host of other physiological (functional) abnormalities.
[0089] Radioimrnunological imaging radionuclides, such as Molybdenum-99, Technetium-99m, Chrornium-S1, Copper-64, Dysprosium-165, Ytterbium-169, Indium-111, Iodine-125, Iodine-131, Iridium-192, Iron-59. Phosphorus-32, Potassium-42, Rhodium 186, Rhenium-188, Samarium-153, Selenium-75, Sodium-24, Strontium-89, Xenon-133, Xenon-127, Yttrium-90 or others, are bound to antibodies (sometimes referred to as labeling, tracing or tagging) that will bind to a specific antigenic target. In one embodiment of the present invention, radioimmunological imaging is combined with targeted thermotherapy by attaching the radionuclides directly to the bioprobes. In such a configuration, the uptake process of the bioprobes can be directly imaged.

[0090] Bioimpedance is a measure of how well the body impedes electric current flow. Fat has higb resistivity, blood lower resistivity. Impedance is measured by applying a small electric current, for example, using two electrodes, and measuring the resulting small voltage with another pair of electrodes. The lower the voltage is, the lower the tissue impedance will be for a given current. Tissue consists of cells and mernbranes; membranes are thin but have a high resistivity and electrically behave as small capacitors. At high Frequencies, the result becomes independent of the capacities of the cell membranes. At low frequencies, however, the membranes impede current flow, and the results are dependent on liquids outside the cells.
[00911 In one embodiment of the present invention, one or more of these imaging techniques is used to image the uptake of the bioprobes prior to, during, or after targeted therapy administration.

Crratin~
[00921 Coating 120 may enhance the heating properties of bioprobe 100, particularly if coating 120 is a polymeric material. Coating 120 may also comprise radioactive or potentially radioactive elements.

[0093] Suitable materials for the coating 120 include synthetic and biological polymers, copolymers and polymer blends, and inorganic materials. Polymer materials may include various combinations of polymers of acrylates, siloxanes, styrenes, acetates, akylene glycols, alkylenes, alkylene oxides, parylenes, lactic acid, and glycolic acid. Further suitable coating materials include a hydrogel polymer, a histidine-containing polymer, and a combination of a hydrogel polymer and a histidine-containing polymer.
[0094] Coating materials may also include combinations of biological materials, such as a polysaccharide, a polyaniinoacid, a protein, a lipid, a glycerol, and a fatty acid. Examples of other biological materials suitable for use herein include heparin, heparin sulfate, chondroitin sulfate, chitin, chitosan, ccllulose, dextran, alginate, starch, carbohydrate, and glycosaminoglycan. Examples of proteins useful herein include an extracellular matrix protein, proteoglycan, glycoprotein, albumin, peptide, and gelatin. These materials may also be used in combination with any suitable synthetic polymer material.
[0095[ Inorganic coating materials may include any cnmbination of a metal, a metal alloy, and a ceramic. Examples of ceramic materials suitable for use herein include a hydroxyapatite, silicon carbide, carboxylate, sulfonate, phosphate, ferrite, phosphonate, and oxides of Group IV

_2; _ elements of the Periodic Table of Elements. These materials may form a composite coating that may also contain one or more biological or synthetic polymers. Where the magnetic particle 110 is formed from a magnetic material that is bioctimpatible, the surface of the particle itself operates as the biocompatible coating.
100961 The coating material may also serve to facilitate transport of the bioprobe 100 into a cell, a process knoNYn as transfection. Such coating materials, referred to as transfection agents, include vectors, prions, polyaminoacids, cationic liposomes, amphiphiles, and non-liposomal lipids or any combination thereof. A suitable vector may be a plasmid, a virus, a phage, a viron, a viral coat. The bioprobe coating may be a composite of any cornbination of transfection agent with organic and inorganic materials, such that the particular combination may be tailored for a particular type of a diseased rnaterial and a specific location within a patient's body.

Markers 10097] The choice of a marker (antigen) 160 and 1.70, as illustrated in FIG.
2, may be important in the targeted therapy methods of the present invention. Although not limited thereto, use and selection of markers is most prevalent in cancer immunotherapy. For breast cancer and its metastases, a specific marker or markers may be selected from cell surface markers such as, for example, members of the MUC-type mucin family, an epit.belial growth factor (EGFR) receptor, a carcinoembryonic antigen (CEA), a human carcinoma antigen, a vascular endothelial grow-th factor (VEGF) antigen, a melanoma antigen (MAGE) gene, family antigen, a T/Tn antigen, a hormone receptor, growth factor receptors, a cluster designation/differentiation (CD) antigen, a tumor suppressor gene, a cell cycle regulator, an oncogene, an oncogene receptor, a proliferation marker, an adhesion molecule, a proteinase involved in degradation of extracellular matrix, a malignant transformation related factor, an apoptosis related factor, a human carcinoma antigen, glycoprotein antigens, DF3, 4F2, MGFM antigens, breast tumor antigen CA 15-3, calponin, cathepsin, CD 31 antigen, proliferating cell nuclear antigen 10 (PC
10), ancl pS2.
[0098] For other forms of cancer and their metastases, a specific marker or markers may be selected from cell surface markers such as, for exarnple, a mez-nber of vascular endothelial growth factor receptor (VEGFR) family, a member of carci;loetnbryonic antigen (CEA) family, a type of anti-idiotypic mAB, a type of ganglioside mimic, a member of cluster designationr'differentiation antigens, a member of epidennal grow-th factor receptor (EGFR) family, a type of a cellular adhesion molecule, a member of MUCTtype mucin family, a type of cancer antigen (CA), a type of a matrix metalloproteinase, a type of glycoprotein antigen, a type of melanoma associated antigen {MA.A}, a proteolytic enzyme, a calmodulin, a member of tumor necrosis factor (TNF) receptor farnily, a type of angiogenesis marker, a melanoma antigen recognized by T cells (MART) antigen, a member of melanoma antigen encoding gene (MAGE) family, a prostate membrane specific antigen (PMSA), a small cell lung carcinoma antigen (SCLCA), a T/Tn antigen, a hormone receptor, a tumor suppressor gene antigen, a cell cycle regulator antigen, an onedgene antigen, an oncogene receptor antigen, a proliferation marker, a proteinase involved in degradation of extracellular matrix, a malignant transformation related factor, an apoptosis-related factor, a type of human carcinoma antigen.
100991 In one embodiment of the invention, a bioprobe includes one or more ligands 130 targeting the MUC- I receptor of the mucin family. In yet another embodiment, a bioprobe has one or more ligands 130 targeting at least one of the EGFR family, such as HER-1, HER-2, HER-3 and HER-4. MUC-1 (Human epithelial mucin, CD277), is a high molecular weight, transmembrane mucin glycoprotein expressed by most glandular and epithelial cell lineages. In addition, MUC- I has a large extracellular domain, portions of which may be shed into the bloodstream. MUC-1 may have a protective role, as its extracellular domain forms elongated rigid structures extending above other molecules on the cell. MUC-1 also plays a role in cell-cell and cell-substrate adhesion. MUC- l is highly expressed in many human adenocarcinomas, including 80% of breast cancers, and is associated with poor prognosis. Mucin (MUC-1 and MUC-2) expression is associated with tumor invasiveness. MUC-1 and MUC-2 expression is associated with invasive ductive carcinoma of the breast. MUC-1 is also present at high levels on. rnany mylonaas. Different tissues/cells produce differing glycoforms of MUC-l.
Glycosylation of MUC-1 in malignant cells is often altered compared to normal tissue. MUC41 is considered a truly tumor specific antigen, although it is also found on normal cells, its aberrant glycosylation on tumors creates new epitopes for targeting. The extracellular domain of MUC-I
may be shed into the blood streatn. The ligand 130 may target the unshed remainder of the MUC-1 expressed on the cell surface.
[01001 Overexpression of growth factor receptors such as the EGFR family is indicated in turnors and has been associated with increased cell resistance to the cytotoxic effects of macrophages and cytotoxic factors, such as 1:~F (tumor necrosis factor), which can lead to tumor gromh. I'he protein encoded by the HER-1'neu gene is a 170,000 Dalton protein, refez-red to as HERp I. The protein encoded by the HER-2/neu gene is a 185,000 Dalton protein referred to as I-IER-2. Bath. proteins have an intracellular domain, a transmembrane domain that includes two cysteine-rich repeat clusters, and an intracellular kinase domain. The extracellular domain of HER-2 may be shed into the bloQdstream. Thus, ligand 130 may target the unshed remainder of the HER-2 expressed on the surface of the cell.
[01011 For ovarian cancers and their metastases, a specific marker or markers may be selected from cell surface markers such as, for example, one of ERBB2 (HER-2) antigen and CD64 antigen. For ovarian and/'or gastric cancers and their metastases, a specific marker or markers may be selected from cell surface markers such as, for exariiple, a polymorphic epithelia] mucin (PEM). For ovarian cancers and their metastases, a specific marker or markers may be selected from cell surface markers such as, for example, one of cancer antigen 125 (CA125) or matrix metalloproteinase 2(IvIMP-2). For gastric cancers and their metastases, a specific marker or markers may be selected from cell surface markers such as, for exatnple, one of CA 19-9 antigen and CA242 antigen.
[01021 For non small-cell lung cancer (NSCLC), colorectal cancer (CRC) and their metastases, a specific marker or markers may be selected from cell surface markers such as, for example, vascular endothelial growth factor receptor (VEGFR), anti-idiotypic mAb, and carcinoembryonic antigen (CEA) mimic. For at least one of small-cell lung cancer (SCLC), malignant melanoma, and their metastases, a specific marker or markers may be selected from cell surface markers such as, for example, anti-idiotypic anAB or GD3 ganglioside mirriic. For melanoma cancers and their metastases, a specific marker or markers may be selected from cell surface markers such as, for example, a melanoma associated antigen (MAA). For small cell lung cancers and their metastases, a specific marker or markers may be selected from cell surface markers such as, for example, a sinall cell lung carcinoma antigen (SCLCA).
101031 For colorectal cancer (CRC) and/or locally advanced or metastatic head and,'or neck cancer, a specific rnarker or markers may be selected from cell surface markers such as, for example, epidennal growth factor receptor (EGFR). For Duke's colorectal cancer (CRC) and its metastases, a specific marker or niarkers may be selected from cell surface markers such as, for example, Ep-CAM antigen.
101041 For non-Hodgkin's lymphoma (NHI.) and its metastases, a specitic marker or markers may be selected from cell surface markers such as, for example, cluster designation/

differentiation (CD) 20 antigen or CD22 antigen. For B-cell chronic lymphocytic leukaemia and associated metastases, a specific marker or rnarkers -nay be selected from cell surface markers such as, for example, CD52 antigen. For acute myelogenous leukaemia and its metastases, a specific marker or markers may be selected from cell surface markers such as, for exaniple, CD33 antigen.
101051 For prostate cancers and their metastases, a specific marker or markers may be selected from cell surface markers such as, for example, prostate membrane specific antigen (PMSA). For carcinomatous meningitis and their metastases, a specific marker or markers may be selected from cell surface markers such as, for example, one of a vascular endothelial growth factor receptor (VEGFR) or an epithelial associated glycoprotein, for example, HMFGI (human milk fat globulin) antigen.
101061 For lung, ovarian, colon, and melanoma cancers and their metastases, a specific marker or markers may be selected from cell surface markers such as, for example, B7-H1 protein. For cnlon, breast, lung, stomach, cervix, and uterine cancers and their metastases, a specific marker or markers may be selected from cell surface markers such as, for example, TRAIL Receptor-1 protein, a member of the tumor necrosis factor receptor family of proteins.
For ovarian, pancreatic, non-small cell lung, breast, and head anad neck cancers and their metastases, a specific marker or markers may be selected from cell surface markers such as, for example, EGFR (epidermal growth factor receptpr).
[0107] For anti-angiogenesis targeting of tumor blood supply, a specific marker or markers may be selected from cell surface markers such as, for example, Integrin avP3, a cell surface marker specific to endothelial cells of growing blood vessels.
101081 For targeting of colon and bladder cancer and their metastases, a specific rnarker or markers may be selected from cell surface markers such as, for example, RAS, a signaling molecule that transmits signals from the external environment to the nucleus.
A mutated form of RAS is found in many cancers.
[0109] In one embodiment of the present invention, ligand 130 taxgets a predetermined target associated with a disease of the patient's imn-iune system. The particular target 1.50 and one or more ligands 130 may be specific to, but not limited to, the type of the immune disease. The ligand 130 may have an affinity for a cell marker or markers of interest. 'fhe marker or niarkers 160/170 may be selected such that they represent a viable target on T cells or B cells of the patieiit's immune system. The ligand 130 may have an affinity for a target associated with a disease of the patient's immune system such as, for example, a protein, a cytokine, a chemokine, an infections organism, and the like.
101101 For diseases of the patient's immune system, a specific marker or markers 160/170 may be selected from cell surface markers. The targeted cells may be T or B
cells of the immune system. For rheumatoid arthritis, a specific marker or markers may be selected from cell surface markers such as, for example, one of CD52 antigen, tumor necrosis factor (TNF), and CD2S
antigen. For rheumatoid arthritis and/or vasculitis, a specific marker or markers may be selected from cell surface markers such as, for example, CD4 antigen. For vasculitis, a specific marker or markers may be selected from cell surface markers such as, for example, CD 18 antigen. For multiple sclerosis, a specific marker or markers may be selected from cell surface markers such as, for example, CD52 antigen.
101111 Iri another embodiment, ligand 130 targets a predetermined target 150 associated with a pathogen-borne condition. The particular target 150 and ligand 130 may be specific to, but not limited to, the type of the pathogen-borne condition. A pathogen is detined as any disease-producing agent such as, for example, a bacterium, a virus, a microorganism, a fungus, and a parasite. The ligand 130 may have an affinity for the pathogen or pathogen associated matter.
The ligand 130 may have an affinity for a cell marker or markers associated with a pathogen-borne condition. The marker or markers may be selected such that they represent a viable target on infected cells.
101121 For a pathogen-borne condition, the ligand 130 for therapy utilizing bioprobes may be selected to target the pathogen itself. For a bacterial condition, a predetermined target may be the bacteria itself, for example, one of Escherichia coli or Bacillus anthracis. For a viral condition, a predetermined target may be the virus itself, for example, one of Cytomegalovirus (CMV), Epstein-Barr virus (EBV), a hepatitis virus, such as Hepatitis B virus, human immunodeficiency virus, such as IIIV, HIV-1, or I-IIV-2, or a herpes virus, such as Herpes virus 6. For a parasitic condition, a predetermined target may be the parasite itself for exarnple, one of Trypanasoma cruzi, Kinetoplastid, Schistosorna rnansQni, Schrstasorrta japonicurn or Schistos ma brucei. For a fungal condition, a predetermined target may be the fungus itself, for example, one of Aspergillus, C,'ryptacoccus neofarmans or Rhizomucor.

101131 For a pathagen-borne condition, the ligand 130 for therapy utilizing bioprobes may be selected to target cell markers of pathogen infected cells. For the HIV virus, the predetermined target may be CTLA4 expressed on the surface of HIV infected T cells. CTLA4 migrates to the infected cell's outer surface when the 141V virus is ready to be released.
[01I4] In anotber embodiment, ligaiid 130 targets a predetermined target associated witb an undesirable target.material. The particular target 150 and ligand 130 may be specific to, but not limited to, the type of the undesirable target. An undesirable target is a target that may be an undesirable material. Undesirable material is material associated with a disease or an undesirable condition, but which may also be present in a normal condition.
For example, the undesirable material may be present at elevated concentrations or otherwise be altered in the disease or undesirable state. The ligand 130 may have an affinity for the undesirable target or for biological molecular pathways related to the undesirable target. The li.gand 130 may have an affinity for a cell marker or markers associated va=ith the undesirable target material.
[0115] For an undesirable target, the selection of a prcdeterrnined target 150 may be important in the therapeutic methods of the present invention. Ligand 130 is selected to target biological matter associated with a disease or undesirable condition. For arteriosclerosis, a predetermined target may be, for example, apolipoprotein B on low density lipoprotein. (LDL).
An undesirable material may be adipose tissue or cellulite for obesity, associated with obesity, or a precursor to obesity. A predetermined marker or markers for obesity maybe selected from cell surface markers such as, for example, one of gastric inhibitory polypeptide receptor and CD36 antigen. Another undesirable predetermined target may be clotted blood.
101161 In another embodiment, ligand 130 targets a predetermined target associated with a reaction to an organ transplanted into the patient. The particular target 150 and ligand 130 m.ay be specific to, but not limited to, the type of organ transplant. The ligand 100 may have an affinity for a biological molecule associated with a reaction to an organ transplant. 'I'he ligand 130 may have an affinity for a cell marker or markers associated with a reaction to an organ transplant. The marker or markers may be selected such that they represent a viable target on T
cells or B cells of the patient's immune system.
101171 For reaction to a transplanted organ, ligand 130 is selected to target the immune response to a transplant. The transplanted organ may be treated before or after transplantation.
For kidney transplantation, a predetennined marker or markers may be selected from cell surface markers such as, for example,l'iuman T cell receptor (CD3) antigen or CD1:8 antigen. For kidney and bone marrow transplantation, a predetermined marker or markers may be selected from cell surface markers such as, for example, CD52 antigen. For liver and bone marrow transplantation, a predetermined marker or markers may be selected from cell surface markers such as, for example, one of CD1S4 antigen and CDS antigen. For transplantation tolerance, a predetermined marker or markers may be selected from cell surface markers such as, for example, CD4. For bone marrow, a predetermined marker or markers may be selected from cell surface markers such as, for example, CD52 antigen for effieient depletion of'I" cells from bone marrow before transplantation in order to avoid graft versus host disease. For xenotransplantation or xenografting, a predetermined marker or markers may be, for example, galactose. Galactose is known to be on pig organs, but is not present in humans.
[0118] In another embodiment, ligand 130 targets a predetermined target associated with a toxin in the patient. A toxin is defined as any poison produced by an organism including, but not limited to, baeteria] toxins, plant toxins, insect toxin, animal toxins, and man-made toxins. The particular target 150 and ligand 130 may be specific to, but not limited to, the type of toxin. The ligand 130 may have an affinity for the toxin or a biological molecule associated with a reaction to the toxin. The ligand 130 may have an affinity for a cell marker or markers associated with a reaction to the toxin.
[011.9] For a toxin in the patient, ligand 130 is selected to target the toxin. A bacterial toxin target may be, for example, one of Cholera toxin, Diphtheria toxin, and Clostridium botulinus toxin. An insect toxin may be, for example, bee venom. An animal toxin may be, for example, snake toxin, for exarnple, Cratalais durissus terrificus venom.
101201 In another embodiment, ligand 130 targets a predetermined target associated with a hormone-related disease. The particular target 150 and ligand 130 may be specific to, but not limited to, a particular hormone disease. The ligand 130 may have an affinity for a hormone or a biological molecule associated with the hormone pathway. The ligand 130 may have an affinity for a cell maTTer or markers associated with the hormone disease.
101211 For a hormone related diseases, ligand 130 is selected to target a cell marker or markers. For estrogen-related disease or conditions, a predetermined target may be, for example, estrogen or cell surface marker or markers such as, for example, estrogen receptor. For human growth hormone disease, the predetermined target may be, for example, human growth hormone.

101221 In another embodirnent, ligand 130 targets a predetermined target associated with non-cancerous disease material. The particular target 150and ligand 130 may be specific to, but not limited to, a particular non-cancerous disease material. The ligand 1.30 may have an affinity for a biological molecule associated with the non-cancerous disease material.
The ligand 130 may have an affinity for a cell marker or markers associated with the non-cancerous disease material.
[01231 For non-eancerous disease material, the ligand 130 is selected to target a predetermined target such as, for example, one of non-cancerous diseased deposits and precursor deposits. For Alzheimer's disease, a predetermined target may be, for exatnple, amyloid B
protein and its deposits, or apolipoprotein and its deposits.
[0124] In another embodiment, ligand 130 targets a proteinaceous pathogen. The particular target 150 and ligand 130 may be specific to, but not limited to, a particular proteinaceous pathogen. The ligand 130 may have an affinity for a proteinaceous pathogen or a biological molecule associated. with the proteinaceous pathogen. The ligand 130 may have an affinity for a cell marker or markers associated with the proteinaceous pathogen. For prion diseases also known as transmissible spongiform encephalopathies, a predetermined target may be, for example, Prion protein 3F4.

Ligands [0125] In one embodiment of the present invention, at least one targeting ligand 130, such as, but not limited to, an antibody, is located on an exterior portion of bioprobe 100, as illustrated in FIG. 1. Targeting ligand 130 is selected to seek out and attach to a target 150.
101261 FIG. 2 illustrates an embodiment wherein a bioprobe 100, comprising a susceptor 1i0, that comprises a coating 120, is attached to a target (such as a cell) 150 by one or more targeting ligands 130. Ligands 130 may also comprise radioactive or potentially radioactive elements.
101271 Cell comprising target 150 may express several types of markers 160 and 170. The specificity of bioprobe 100 is represented by its attachment to targeted marker 160 over the many other markers or molecules 160 on cell comprising target 150. One or more bioprobes 100 may attach to the cell via ligand 130. Ligand 130 may be adapted, and bioprobe 100 may be designed such that bioprobe 100 remains externally on cell 150 or may be internalized into cell comprising target 150. Once bound to cell 150, the magnetic nanoparticle I 10 heats in response to the _,I_ energy absorbed. For example, the magnetic nanoparticle 110 may heat through hysteresis losses in response to wi AMF. The heat may pass through coating 120 or through interstitial regions to the cell 150, for example via canvection, conduction, radiation, or any combination of these heat transfer mechanisms. The heated cell 150 becomes damaged, preferably in a manner that causes irreparable damage. When bioprobe 100 becomes intemalized within cell comprising target 150, bioprobe 100 may heat cell 150 internally via convection, conduction, radiation, or any combination of these heat transfer znechanisms. When a sufficient amount of energy is transferred by bioprobe 100 to cell 150, cell 150 dies via necrosis, apoptosis or another mechanism.
[01281 Suitable ligands for use herein include, but are not limited to, proteins, peptides, antibodies, antibody fragments, saccharides, carbohydrates, glycans, cytokines, chemokines, nucleotides, lectins, lipids, receptors, steroids, neurotransmitters, Cluster DesignationiDifferen.tiation (CD) markers, and imprinted polymers and the like. The preferred protein ligands include, for exa.rz-tple, cell surface proteins, membrane proteins, proteoglycans, glycoproteins, peptides and the like. The preferred nucleotide ligands include, for exaniple, complete nucleotides, complimentary nucleotides, and nucleotide fragrnents.
The preferred lipid ligands include, for example, phospholipids, glycolipids, and the lilce.
[0129] In one embodiment of the present invention, the ligand 130 may be covalently bonded to or phvsically interacted with the magnetic particle 110 or the coating 120.
'Fhe ligand 130 may be bound covalently or by physical interaction directly to an uncoated portion of the magnetic particle 110. The ligand 130 may be bound covalently or by physical interaction directly to an uncoated portion of the magnetic particle 110 and partially covered by the coating 120. The ligand 130 may be bound covalently or by physical interaction to a coated portion of the bioprobe 100. The ligand 130 may be intercalated to the coated portion of bioprobe 100.
[01301 Covalent bonding may be achieved with a linker molecule. Examples of functional groups used in linking reactions include amines, sulfhydryls, carbohydrates, carboxyls, hydroxyls and the like. The linking agent may be a homobifunctional or heterobifunctional crosslinking reagent, for example, carbodiimides, sulfo-NHS esters linkers and the like. The linking agent may also be an aldehyde crosslinking rcagent such as glutaraldehyde. The linking agent may be selected to link ligand 100 to the magnetic particle 110 or the coating 120 in a preferable orientation, specifically with the active region of the ligand 150 available I-or .........

targeting. I'bysicai interaction does not require the linking maIecule and the ligand 100 be bound directly to the magnetic particle 110 or to the coating 120 by non-covalent means such as, for example, absorption, adsorption, or intercalation, 101311 Some exemplary embodiments of the bioprobe system, along with associated indications for which they may be utilized, are presented in Table 1, Bioprobe Systems and Indications BIOPROBE SYSTEM INI?ICATION
TARGET MARKER LIGAND
Endothelial cells of Integrin 93 Ber .FP4 antibody Metastatic breast cancer, metastatic growing blood LM609 antibody colon carcinoma vessels of Integrin antagonist metastatic cancer cells Cancer cells Unglycosylated Anti-DF3 antibody Breast cancer DF3 anti en Cancer cells Kallikreins ArÃti-kallikrein Ovarian and prostate cancer antibody Cancer cells ErbB2 (HE'R-2/neu) Anti-ErbB2 antibOdy, Breast and ovarian cancers and scFv (F5), IDM-I
(aka MDX-2 10) variants Cancer cells Prostate specific MDX-070 and 7EI 1- Prostate cancer membrane antigen C5.3 antibodies (PSMA) MCF-7 breast 43 Kd membrane 323/A3 antibody tireast cancer cancer cells assoCiated I co rotein Receptor tyrosine Vascular endothelial kinases-- growth factor FLTI (VEGF) and VEGFB Anti-FLTI antibody Tumour angiogenesis FLKI and placental growth Anti-FLKI antibody, factor receptors 2C3 antibody 1'umQur angiogenesis (PGFR) Metastatic CAR. (coxsackie Anti-CAR antibody Metastatic prostate cancer cancer cells adetiovirus cell-surface receptor) Vascular smonth Urokinase type Urokinase type Cancer muscle cells of plasminogen plasminogen activator cancer cells activator receptor (uPA) (uPAR
Blood vessels of I'lasminp~;en Anti-PAI-I ant~body Breast cancer cancer cells activatÃ~r inhibitor antibody 1(I'AI-1) Epithelial ovarian Matrix Anti-MM.P-9 antibody varian carcinotrtas with lymph tumour cells metaloproteinase 9 node rnetastasis.
(i~iEMP-9) [
Cancer cells Cyclin A Anti-cyclin A a.ntibody Squamous cell carcinoma of the ton ue Cancer cells Cyclin D A~ti-cyclirr L3(I,2,3) Malignant breast cancer, head and antibody neck squamous cell carcinomas, mantle cell carcinomas, laryngeal s uamous cel[ carcinoznas Kidney cortex tissue Cvclin E Anti-c clin E antibody iluman renal cell carcinoma Ttt.rnorigenic human [ Cyclin E Anti-cyclin E antibody Breast cancer breast epithelial celis Malignant epithelial Cyc[in E Anti-cyclin E arttibCdy Transitional cell carcinonia of the bladder tissue urinar bladder Cancer cells Cdc2 Anti-cdc 2 antibod Breast cancer Malignarit epithelial P27 Anti-phospho p27 Transitional cell carcinoma of the bladder tissue antibody urina bladder Cancer cells P73 Anti-p73 antibody Lung carciiiogenesis, bladder carcinogenesis, neuroblastoma, breast cancer Cancer cells Ras Anti-ras antibodv Breast cancer Caneer cells c-m c Anti C-m c antibod Breast cancer Cancer cells c-fm.s nti-c-fms antibod Breast cancer Cancer cells Hepatocyte growth Anti-HGFR antibody Colorectal cancer factor receptor IiGFR
Cancer cells c-met Anti-c-met antibody Gastric and colon cancers, bepatomas, ovarian cancer, skin cancer Large granular Apoptosis related Anti-CD95 (Fas) Leukaemia, prostate cancer lymphocyte (LGL) factors: antibody leukaemia cells Fas Fast, Cancer cells Non-receptor protein Anti c-src-polyctonal Metastatic colorectal cancer, and tyrosine kinase V- antibody late stage breast cancer Src and C-Src Cancer cell CAR (coxsackie Onyx-015 adenovirus Lung, ovarian, other cancers ~~~
aderroviriis eel!
surface rece tor}
Cancer cell Epidernial grorvth Molecule 225 antibody Cancer factorreceptor EGFR) Cancer ce[ls I36 antigen Anti-D6 antibody Vascular tumours including Ka osi's sarcoma Cancer cells 2C4 anti ~en Anti-2C4 antibody Breas prostate, otber cancers Cancer cells Cytokeratin S5A10-2 antibody Non-small cell lung cancer epithelial marker and.-or telomerase reverse t_rans cr~tase Cncer cells Carcin~~ern_bryonic MFE 23 scFv of ar~ti-~ Iorectal eancer a_nti eg n(CEA) CEA antibody Cancer cells Proliferating cell Anti-PCNA antibody Breast cancer E nuclear antigen 1'CNA
E f )__ Cancer cells Neu 3, a membrane Anti-neu 3 sialidase Colon cancer associated sialidase antibod Cancer cells Pl 3KC2 beta (cancer Anti-P13KC2beta Lung cancer cell signal mediator) antibc~d Cancer cells Guanylyl cyclase-C Anti-GC-C antibody~ Esophageal or gastric cancer (GC-C) receptor Cancer cells Transforrning Anti-4"GFP antibody Breast cancer growth factor beta (TGFB) rece tor Cancer cells Platelet derived growth factor receptor (PDGFR) PDGFR-A (alpha) Anti-PDGF-A Lung cancer antibody PDGFR-B (beta) Anti-PDGF-B antibod Bone cancer Cancer cells and Vascular endothelial Tiel Cancer =
blood vessels growth factors Tie2 Cancer VEGFR [ an iE> oieti:n Cancer cells Mucin family of Anti-MUC-I antibody, Colorectal and ovarian carcinomas receptors 12E antibody 3D antibody A5 antibody Cancer cells TAG-72 B723 antibody Breast and lung cancers Cancer cells Human milk fat NCL-HMFG I and Breast, lung, colon, and prostate globule receptor NCL-IIMFG2 [ cancers antibodies Methionine synthase Cobalamin receptor B i2 (riboflavin, and Breast, lung, colon, sarcomatous and L- variants) cobalamin thyroid or central nervous system tnethylmalonyl-C A and variants, such as malignancies cancer mutase adenosyicobalamin transcobalamin Cancer cells Glioma chloride Scorpion toxin-- Gliomas channel chlorotoxin and chlorotoxin-like molecules Cancer cells 40 kD glycoprotein NR-LU-10 antibody Small cel.l lung cancer antiQen CNS cells and tissue Brain-specific chondroitin sulphate proteoglycan Brain enriched Anti-BEHAB antibody Gliomas hyaluronan binding protein (BEI-{AB-aka brevican Cancer celis Catenins Alpha catenin. Anti-alpha catenin Colorectal carcinorna, non-small antibody cell lung cancer Beta cateniil Anti-beta catenin Breast cancer antibody Gamm.a catenin Anti-gamma catenin ?'hyroid cancer antibody Cancer cells Interleukin (IL) rece tors ILl.3 receptor ILI3-PP38 antibody Kidney, brain, breast, and head and neck cancers, and Ka osi's sarcoma Cancer cells Mesothelin receptor Anti-mesothelin antibody, and Mesotheliomas E
SSI(dsFv) variant Ovarian cancer and mesothelio-nas Cancer cells CD44 surface Anti-CD44 antibodv Prostate cancer [ adhesion molecule Cancer cells EGFRvIII Ua30:2 antibody Brain, colorectal, pancreatic,billary~
L8A4 antibody liver cancers and soft tissue DN8.3 antibody sarcomas.
1IC6 antiboqy Receptor tyrosine Vascular endothelial Anti-FLT] antibody Atherosclerotic plaques kinases FLT I growth factor (VEGF) and VEGFB
Srnoot~musclc cells Basic fibroblast Anti-bFGF antibody Restenosis in the lumen of growth factor blood vessels rece tor bfiGFR
Vuln.erable plaque Oxidized low density Oxidation-specific Atherosclerosis and vascular lipoprotein (OxLDL) antibodies (Ox-AB) disease MDA-2 antibody Vulnerable plaque Malondialdehyde- IK17 antibody Atherosclerosis and vascuiar modified LDL
(MDA-LDL) = disease M. Tuberculosis APA-antigen Anti-APA antibody Tuberculosis bacilli Retrovirus infected "I'GFA (alpha) Anti-TGFA antibody HN
cells Leukocytes Alpha4 subunit of Antegren Multiple sclerosis alpha4betal -integin (VLA-4) and alpha4beta7-integrin Receptor tyrosine Vascular endothelial Anti-FL'fI antibody Autoimmune joint destruction kinases FL7'I growth factor (VEGF) and VEGFB (arthritis, iupus, etc) Plasmodium Apical membrane Anti-AMA- I antibody Malaria falci arurn antipen-1 (AMA-1) [0132] The methods of the present invention may be used to treat a variety of indications which include, but are not limited to, cancer of any type, such as bone marrow, lung, vascular, neuro, colon, ovarian, breast and prostate cancer, diseases of the immune system, such as AIDS
and autoimmune conditions, and pathogen-borne diseases, such as HIV, malaria and tuberculosis, and undesirable matter, such as adverse angiogenesis, amyloidosis, restenosis, vascular conditions, obesity, toxins and other abnor.nial cell or tissue growth. Tbe bioprobe systems described herein may be used to treat other indications than the associated indications listed in Table 1.

[01331 Targets, markers and ligands for use in the present invention include, but not limited to, those disclosed hereinabove, those listed in Table 1, as well as those disclosed in related patent applications having U.S.S.N. 10/176,950 and 10/200,082, xvhich are incorporated herein by reference.

Administration of Bio robes 101341 A method of administering the bioprobes 1.00 to the desired area for treatment and the dosage may depend upon, but is not limited to, the type and location of the diseased rnaterial.
The size range of the bioprobes 100 allows for microfiltration for sterilization. An administration method may be, for example, wash, lavage, as a rinse With sponge, or other surgical cloth as a perisurgical administration technique. Other methods of administration may include intravascular injection, intravenous injection, intraperitaneal irijection, subcutaneous injection, and intramuscular injection. The bioprobes 100 may be fortnulated in an injectable fornat (suspension, emulsion) in a medium such as, for example, water, saline, Ringer's solution, dextrose, albumin solution, and oils. The bioprobes 100 may also be administered to the patient through topical application via a salve or lotion, transdermally through a patch, orally ingested as a pill or capsule or suspended in a liquid or rectally inserted in suppository form.
Bioprobes 100 may also be suspended in an aerosol or pre-aerosol formulation suitable for inhalation via the mouth or nose. Once administered to the patient, delivery of the bioprobes 100 to the target site may be assisted by an applied static magnetic field due to the magnetic nature of the bioprobes 100. Assisted delivery may depend on. the location of the targeted cell. The bioprobes may also be delivered to the patient utilizing other methods. For example, the bioprobes 100 may be admiraistered to the patient orally, or may be administered rectally.

The Energy. Source Energy Sources for Energizing Bioprobes ( C[?RRESPONDING ENERGY ENERGY TIHERAPEUTIC
SECTION BELOW FORM SOURCE _--- i.-?VIECHANISM
2.3.1 1 Ionizing Nuclear reactor, particle Combination radiation -- accelerators, radioactive Mechanism -L
Neutron, alpha, materials, cyclotrons, pulsed Damage to genetic beta, amrna, voltage (above 40 kV) material with heat ..3'7_ X-ray sotarce.
2.3.2 AMF Power Generator/Inductor Hysteresis (Induction,) Heatin i 2.4 AMF, Ionizing Extracorporeal 'radiation i _ ~~
Targeted The.rnaotbera in Cornbiriation urith Radiation Th.era r 101351 Radiotherapy, also referred to as radiation therapy, is the treatment of cancer and other diseases utilizing ionizing radiation. Ionizing radiation deposits energy that injures or destroys cells in the area being treated (the "target tissue") by damaging their genetic material, making it impossible for these cells to continue to grow. Although radiation damages both cancer cells and normal cells, the latter are able to repair themselves and function properly.
Radiotherapy may be used to treat localized solid tumors, such as cancers of the skin, tongue, larynx, brain, breast, or uterine cervix. It can also be used to treat leukemia and lymphoma (cancers of the blood-forrning cells and lymphatic system, respectively). In one embodiment of the present invention, radiotherapy or radiation therapy is used in combination with the targeted therrnotberapy methods disclosed herein. Radiotherapy is applied at least once prior to, or at least partly during, or at least once after targeted therapy administration, or any combination thereof.
101361 One type of radiation therapy commonly used involves x-rays or gamma rays. X-rays were the first form of photon radiation to be used to treat cancer. Depending on the amount of energy they possess, the rays can be used to destroy cancer cells on the surface of or deeper in the body. The higher the energy of the x-ray beam, the deeper the penetration of the x-rays into the target tissue. Linear accelerators and betatrons are machines that produce x-rays of increasingly greater energy. The use of machines to focus radiation (such as x-rays) on a cancer site is referred to as external beam radiotherapy. These beams are shielded from the outside world and special shielding is used for "focusing" these beams onto defined body areas. In one embodiment of the invention, external beam radiotherapy is used in combination with the targeted thermotherapy methods disclosed herein. If both the targeted thermotherapy and radiotherapy methods are used simultaneously, the AMI- system may comprise a separate opening for the beam to enter. Alternatively, the beam may be directed through the patient's opening (patient gantry). Intraoperative irradiation is a technique in which a large dose of external radiation is directed at the tumor and surrounding tissue during surgery.
101371 Gamma rays are produced spontaneously as certain elements (such as radium, uranium, and cobalt 60) release radiation as they decompose or decay. Each element decays at a specific rate and emits energy in the fonn of gamma rays and other particles.
X-rays and gamma rays generally have the same effect on cancer cells.
[0I38) Another investigational approach is particle beam radiation therapy.
This type of therapy uses fast-moving subatomic particles to treat localized cancers.
Particle accelerators are used to produce and accelerate the particles required for this procedure. Some particles (neutrons, pions, and heavy ions) deposit more energy than x-rays or gamma rays along the path they take through tissue, thus causiz-ig more damage to the cells they contact. This type of radiation is often referred to as high linear energy transfer (high LET) radiation. In one embodiment of the invention, high LET therapy is used in combination with the targeted tb.erm.otherapy methods disclosed herein.
[01391 Another technique for delivering radiation to cancer cells is to place radioactive implants directly in a tumor or in a body cavity. This is referred to as internal radiotherapy.
(Brachytherapy, interstitial irradiation, and intracavitary irradiation are types of internal radiotherapy.) During this treatment, the radiation dose is concentrated in a small area, and the procedure may require the patient to stay in the hospital for a few days. In one embodiment of the invention, internal radiotherapy is used in combination with the targeted thermotherapy methods disclosed herein. The implant comprises a material that heats during the targeted therapy administration by eddy current or hysteretic heating, or comprises a material that does not heat under AMF exposure, such as plastic, ceramic, glass, or transplanted human tissue.
[0140[ In one embodiment of the invention, radiolabled antibodies deliver doses of radiation directly to the cancer site (radioimmirnotherapy) in combination with targeted thermotherapy. At least one radioisotope 140 is attached to bioprobe 100, as illustrated in F.1G. 1. Such a bioprobe can be a dual therapy bioprobe. Once injected into the body, the antibodies actively seek out the cancer cells, which are destroyed by the cell-killing (cytotoxic) action of the radiation.
[0141] Examples of radioisotopes suitable for use herein are:

= Molybdenurn.-99: Used as the 'parent' in a generator to produce technetium-99m, the niost widel.y used isotope in nuclear medicine.

W,~_ = Technetium-99m: Used particularly for imaging the skeleton and heart muscle, and for imaging the brain, thyroid, lungs (perfusion and ventilation), liver, spleen, kidney (structure and filtration rate), gall bladder, bone marrow, salivary and lacrimal glands, heart blood pool, infection and numerous specialized medical studies.
= Chromium-S 1: Used for labeling red blood cells and quantifying gastro-intestinal protein loss.
= Cobalt-60: Used for extemal beam radiotherapy.

= Copper-64: Used for studying genetic diseases affecting copper metabolism, such as Wilson's and Menke's diseases.
= Dysprosium-165: Used as an aggregated hydroxide for synovectomy treatment of arthritis.
= Ytterbium- 169: Used for cerebrospinal fluid studies in the brain.

= Iodine-125: Used in cancer brachytherapy (prostate and brain), also used for diagnostic evaluation of the kidney filtration rate and for diagnosing deep vein thrombosis in the leg. It is also widely used in radioimmuno assays to show the presence of hormones in small quantities.
= Iodine-131: Widely used in treating thyroid cancer and in imaging the thyroid; also used in the diagnosis of abnormal liver function, renal (kidney) blood flow and urinary tract obstruction. Although it is a strong gamma emitter, it is used for beta therapy.

= lridium-192: Supplied in wire form for use as an internal radiotherapy source for cancer treatment.
= lron-59: Used for studying iron metabolism in the spleen.

= Phosphorus-32: Used in the treatment of polycythemia vera (excess red blood cells). It is a beta emitter.
= 1'otassium-42: Used for the determination of exchangeable potassium in coronary blood flow.
= Rhenium-188 (derived from "1'ungsten-188): Used for beta irradiating coronary arteries from aii angioplasty balloon.

= Sanzarium-153: Very effective in relievin.g the pain of secondary cancers lodged in the bone. It is commercially available as Quadramet'rm. Also, it is very effective for prostate and breast cancer. It is a beta emitter.

= Selenium-75: Used in the form of seleno-methionine to study the production of digestive enzymes.

= Sodium-24: Used for studies of electrolytes within the body.

= Strontium-89: Very effective in reducing the pain of prostate cancer. Beta emitter.
= Xenon-133, Xenon-127: Used for pulmonary (lung) ventilation studies.

= Yttrium-90: Used for cancer therapy and as silicate colloid for the treatment of arthritis in larger joints. It is a beta emitter.
101421 Radiation therapy in combination with targeted thermotherapy may also be used alone, or in combination with chemotherapy, surgery or both.
101431 The energy source for use in the present invention includes any device that is able to provide AMF at the appropriate frequency, or microwave at the appropriate frequency to the bioprobe so that it can convert that energy to heat. In one embodiment of the present invention, energy is delivered to the bioprobe, which then transmits the heat to the targeted cell(s) and cells or tissue that surround the targeted cell(s). In another embodiment, an additional energy source is used in combination AMF or Microware for converting a bioprobe into a source of ionising radiation (neutron, alpha, beta, gamma, etc.). FIG. 3 schematically illustrates an energy source that transmits energy to a subject's body or a body part. Some exemplary energy forms and energy sources useful herein are listed in Table 2. The different forms of energy, for example AMF, or a combination comprising AMF, may be created using a variety of mechanisms, such as those listed in Table 2. The table also lists those sections of the following descriptions that are pertinent to the different energy forms and therapeutic mechanisms.
[01441 In general, as illustrated in FIG. 3, operator 270 controls an energy generating device 250, for example via a console 260, which delivers energy, for example via a cable 220, to an energy source 210. Energy source 210 transmits energy to the bioprobe's susceptor to heat or otherwise affect the targeted cell, and cells or tissue that surround the bioprobe in a subject 230.
[0145] It will be appreciated that the AMF energy sources described herein may also be used for heating other types of bioprobes, for example, the bioprobes disclosed in patent applications having U.S.S.N. 10/I76,950 and 10/200,082. It will further be appreciated that the energy sources disclosed in patent applications having U.S.S.N. 10/176,950 and 10/200,082 may also be used for heating the bioprobes of the present invention.

Alternatin Ma netic Field AMF Heatin [01461 In some embodiments of the present invention, AMF energy may be used with a bioprobe to produce therapeutic heating t.hrough hysteresis loss mechanisms in the magnetic nanoparticles of the bioprobes.
101471 In one embodiment of the present invention, as illustrated in FIG. 4, the therapeutic system 300 comprises an AMF generator, which is located for example within a cabinet 370, designed to produce an AMF that may be guided to a specific location within a subject 310 by a magnetic circuit 330. Subject 310 is placed upon an. X-Y horizontal and vertical axis positioning bed 340. Positioning bed 340 can be positioned horizontally and vertically via a bed controller 350. The AMF generator produces an AMF in magnetic circuit 330 that exits magnetic circuit 330 at one pole face 320, passing through the air gap and the desired treatment area of subject 310, and reenters magnetic circuit 330 through the opposing pole face 320, thus completing the circuit. An operator or medical technician preferably controls and monitors the AMF
characteristics and bed positioning via a control panel 360. When the AMF is generated by an RF generator, the frequency of the AMh' is preferably in the range of about 80 kH.z to about 800 kHz.
1014$1 Other approaches may be used to generate the AMF, and may provide a focused and/or a homogeneous field. In one embodiment, a magnetic solenoid coil 410 may be particularly useful for heating bioprobes in tissue having high length to diameter ratios, such as human limbs or small animals. This is illustrated in FIG. 5, whieh shows a cross-sectional view of the magnetic solenoid coil 410. The coil 410 comprises a circular, doughnut shaped ring 420 of low reluctance magnetic material, which may be specifically formulated for magnetic cores operating at a desired frequency, for example about 150 kHz. One example of such a low reluctance magnetic material is FluxtrolTM (commercially available from Fluxtrol Manufacturing Inc., Auburn Hills, MI, USA).
101491 A magnetic flux focusing bar 400, fabricated from a length of a low reluctance magnetic material may be positioned so as to surround about 25% of the circumference of the outer diameter of solenoid coil 410 and to stretch from the ring 420 to the opposite end of solenoid coil 410. 'I'he magnetic flux focusing bar 400 may be fabricated from the same material as the ring 420, or from a different material. For example, the bar 400 may be fabricated from FerrotronTM {c4tnmercially available from Fluxtrol Manufacturing Inc.}.
101501 The ring 420 and focusing bar 400 direct a xnagnetic flux 430 in a pattern that exposes a reduced cross-section of a human or animal body or body part to the magnetic field. Because eddy current heating is proportional to the square of the cross-section of the exposed tissue in magnetic flux 430, it is advantageous to reduce the size of the exposed cross-section. This approach allows for higher magnetic field strengths for application to the subject with reduced eddy current heating. In addition, circular doughnut shaped ring 420 and focusing bar 400 cause the field strength to drop off significantly outside of solenoid coil 400.
Magnetic solenoid coil 410 focuses the AMF while protecting the non-targeted parts of the subject, such as the head and vital organs.
[0151] The magnetic susceptors for use herein typically are susceptible to AMF
energy supplied by the energy source, and heat when exposed to AMF energy; are biocompatible; and have surfaces that have (or can be modified to have) functional groups to which ligands can be chemically or physically attached. In one embodiment of the present invention, a bioprobe having a magnetic nanoparticle core is surrounded by a biocompatible coating material. There are many possible combinations of core-coating materials. For example, gold as a coating material is particularly advantageous because it forms a protective coating to prevent a chemical change, such as oxidation, in the core material, while being biocompatible. A
gold coating can also be chemically modified to include groups for ligand linking. Further, gold may serve as a good conductor for enhancing eddy current heating associated with AMF heating.
101521 Types of magnetic susceptor cores that require a protective coating include iron, cobalt, other magnetic metals, and their less stable oxides. An example of the latter is magnetite, Fe3O4, which will undergo further oxidation to form maghemite (y-Fe203) and eventually/or hematite (a-FeA). Iron and cobalt, for example, are susceptible to chemical changes, such as oxidation, and possess magnetic properties that are significantly changed due to oxidation. The use of a protective coating is especially preferred in embodiments where the core material may pose a toxic risk to humans and animals in vivo. Thus, the use of a gold coating material is particularly preferred to protect the core material from chemical attack, and to protect the subject from toxic effects of the core material.

[01531 In one embodiment of the present invention, the gold coating is chemically modified via thiol chemistry such that a chemical link is forrned between the gold surface and a suitable ligand. For example, an organic thiol moiety can be attached to the gold, folloived by linking the ligand to the organic thiol moiety using at least one silane, carboxyl, amine, or hydroxyl group, or a combination thereof Other chemical methods for modifying the surface of the coating material may also be utilized.
101541 In another embodiment, nitrogen-doped Mn clusters are used as magnetic susceptors.
'I'hese nitrogen-doped Mn clusters, such as MnN and MnNy. Nvhere x and v are nonzero numbers, are ferromagnetic and comprise large magnetic moments. Calculations based on density-functional theory show that the stability and magnetic properties of small Mn clusters can be fundamentally altered by the presence of nitrogen. Not only are their binding energies substantially enhanced, but also the coupling between the magnetic moments at Mn sites remains ferromagnetic regardless of their size or shape.
[0155] In another embodiment, Nd1_hCaFeO3 is used as a magnetic susceptor. The spontaneous magnetization of the weak ferromagnetism decreases with increasing Ca content or increasing particle size.
101561 Other materials, such as aggregates of superparamagnetic grains af Co36C64, Bi3FesO]2, BaFe]2O19), NiFe, CaNiFe. Co-Fe304, and FePt-Ag, where the collective state of the aggregate induces magnetic blocking, may also be used as susceptors in the present invention, Microwave Resonance Heatin~
101571 It is well knowm that atoms, molecules, and crystals possess resonance frequencies at which energy absorption is effectively achieved. In general, resonance heating offers significant advantages because the targeted material absorbs large quantities of energy from a relatively low power source. Thus, non-targeted materials, including body tissue, the resonant frequency of which differs from that of the targeted material, do not heat to the same extent. Accordingly, materials may be selected to take advantage of a particular resonant frequency in the electromagnetic energy spectrum. A susceptor material may be selected such that the internal chemical bonds of the material may resonate at a particular frequency.
101581 Resonance heating can also be achieved by exploiting interactions of the microwave energy with materials that possess magnetic, electrical, or electric dipole structures on the atomic, i-nolecular, or macroscopic length scales. In addition to the direct modes of heating.

described above, resonance heating may be used indirectly. In one embodiment of the present invention, materials for use as bioprobes are selected such that they possess magnetic or electric properties that will induce a shift in the resonance frequency ol'the tissue to which they become attached. Thus, the molecules of the tissue in close proximity to the bioprobes will heat preferentially in an applied energy field tuned to the appropriate frequency.
[0159] The energy can be applied to a targeted cell, targeted tissue, to the entire body, extracorporeally (outside of the subject's body), or in any cornbination thereof.
Extracorporeal Therar~y [0160] In one embodiment of the present invention, a subject is treated via extracorporeal therapy. The bioprobes may be used to lyse, denature, or otherwise damage the disease material by removing material from the subject, exposing the material to an energy source, and returning the material to the body. The bioprobes may be introduced into the subject's body or body part, and then removed from the subject along with the material that is being extracted. The bioprobes may be separated f'rom the material that is extracted after the treatment. In another embodiment, the bioprobes are introduced to the extracted material while the extracted material is outside of the subject's body or body part. For example, where the extracted material is the subject's blood, the bioprobes may be introduced to the vascular circulating system or into the blood circulating outside of the body, prior to exposure to an energy source.
[0161] In embodiments where the bioprobe/target complexes that are carried primarily in the blood serum or blood plasma are targeted, the blood serum or blood plasma may be separated extracorporeally from the other blood components, exposed to an energy source so as to destroy or inactivate the target, and recombined with the other blood components prior to returning the blood to the subject's body. The bioprobes may be introduced into the vascular circulating system, the blood circulating outside of the body, or the blood serum or blood plasma after it is separated.
[0162] In another ernbodirnent, the bioprobes may be contained in a vessel or column through which the blood circulating outside of the body or the blood serum or blood plasma flows. The vessel or column may be exposed to an energy source so as to destroy or inactivate the targeted cells or antigens prior to returning the blood to the subject',s body.
[0163] The advantages of providing energy to the bioprobes extracorporeally include the ability to heat to higher temperatures and/or heat more rapidly to enhance efficacy while minimizing heating and damage to surrounding body tissue, and the ability to reduce exposure of the body to the energy from the energy source. In embodiments where the bioprobes are introduced into the blood circulating outside of a subject's body, the blood serum, or blood plasma that is extracted from the body, bioprobes need not be directly introduced into the body, and higher concentrations of bioprobes can be introduced to target. Further, the portion of the subject that is being treated extracorporeally can be cooled externally, using a number of applicable methods, while energy is provided to the bioprobes without mitigating the therapeutic effect. In addition, the cooling may take place before, and/or after the administration of energy.
[01641 The treated bioprobes and the associated targets need not be returned to the subject's body. For example, if the bioprobes and the associated targets are contained in blood extracted from a subject, the treated bioprobes and the associated targets may be separated from the blood prior to returning the blood to the subject's body. In embodiments where the bioprobes contain a magnetic component, the bodily fluids containing the bioprobes and associated targets are passed through a magnetic field gradient in order to separate the bioprobes and the associated targets from the extracted bodily materials. In doing so, the amount of susceptors and treated disease material returned to the subject's body is reduced.
[0165] In another embodiment of extracorporeal treatment, the tissue selected for heating is completely or partially removed from a subject's body, e.g., during an open surgical procedure.
The tissue can remain connected to the body or can be dissected and reattached after the tlaerapy.
In yet another embodiment, the tissue is removed from the body or body part of one donor subject and transplanted to that of a recipient subject after the therapy.
[0166] While the above description of the invention has been presented in terms of a human subject, it is appreciated that the invention may also be applicable to treating other subjects, such as mammals, cadavers and the Iike.
101671 As noted above, the present invention is applicable to thermotherapeutic compositions for treating disease material, and methods of targeted therapy utilizing such compositions. The present invention should not be considered limited to the particular embodiments described above, but rather should be understood to cover all aspects of the invention as fairly set out in the attached claims. Various modifications, equivalent processes, as well as numerous structures to which the present invention may be applicable will be readily apparent to those skilled in the art to which the present invention is directed upon review of the present specification. The claims are intended to cover such modifications and devices.

Claims

1. A magnetic composition, comprising:
a) a magnetic nanoparticle that comprises at least one stable single magnetic domain grain that heats when subjected to an alternating magnetic field with frequency in the range of from about 80 kHz to 800 kHz, amplitude in the range of from about 7.98 kA/m and 104 kA/m, and at a temperature in the range of from about 270 to 380 K; and b) a coating.

3. A magnetic composition according to claim 1, wherein the composition further comprises a ligand.

4. A magnetic composition according to claim 1, wherein the coating comprises dextran.

5. A magnetic composition according to claim 1, further comprising a radio isotope, potential radioactive isotope, a chemotherapeutic agent, or any combination thereof.

6. A magnetic composition according to claim 4, wherein the radioactive isotope is iodine-131, cobalt-60, iridium- 192, yttrium-90, strontium-89, samarium- 153, rhenium-186, technetium-99m, or any combination thereof.

7. A magnetic composition according to claim 4, wherein the potentially radioactive isotope is an isotope possessing a high absorption cross-section to neutrons, protons, electrons, or high energy photons.

8. A magnetic composition according to claim 6, wherein the potentially radioactive isotope is an isotope possessing has a high absorption cross-section to neutrons, and is one of boron-10, a lanthanide such as samarium-149, gadolinium-157, and gadolinium-155 or any combination thereof.

9. A magnetic composition according to claim 7, wherein the desired treatment area is exposed to neutrons, protons, electrons, or high energy photons to activate the potentially radioactive isotope in combination with AMF thermotherapy.

10. A magnetic composition according to claim 4, wherein the chemotherapeutic agent is one of doxorubicin, cisplatin, or any combination thereof.

11. A magnetic composition, comprising:
a) a magnetic nanoparticle that comprises either i) an aggregate of superparamagnetic grains that exhibits a collective magnetic state such that it is apparently blocked when subjected to an alternating magnetic field with frequency in the range of from about 80 kHz to 800 kHz, amplitude in the range of from about 7.98 kA/m and kA/m, and at a temperature in the range of from about 270 to 380 K, or ii) an aggregate of stable single magnetic domain grains and superparamagnetic grains, that exhibits a collective magnetic state such that characteristic relaxation time of the superparamagnetic grains is increased, and the aggregate is apparently blocked when subjected to an alternating magnetic field with frequency in the range of from about 80 kHz to 800 kHz, amplitude in the range of from about 7.98 kA/m and kA/m, and at a temperature in the range of from about 270 to 380 K; and b) a coating.

12. A magnetic composition according to claim 10, wherein the composition further comprises a ligand.

13. A magnetic composition according to claim 11, wherein the coating comprises dextran.

14. A magnetic composition according to claim 10, further comprising a radio isotope, potential radioactive isotope, a chemotherapeutic agent, or any combination thereof.

15. A magnetic composition according to claim 13, wherein the radioactive isotope is iodine-131, cobalt-60, iridium-192, yttrium-90, strontium-89, samarium-153, rhenium-186, technetium-99m, or any combination thereof.

16. A magnetic composition according to claim 13, wherein the potentially radioactive isotope is an isotope possessing a high absorption cross-section to neutrons, protons, electrons, or high energy photons.

17. A magnetic composition according to claim 15, wherein the potentially radioactive isotope has a high absorption cross-section to neutrons, and is one of boron-10, a lanthanide such as samarium-149, gadolinium-157, and gadolinium-155 or any combination thereof.

18. A magnetic composition according to claim 16, wherein the desired treatment area is exposed to neutrons, protons, electrons, or high energy photons to activate the potentially radioactive isotope in combination with AMF thermotherapy.

19. A magnetic composition according to claim 13, wherein the chemotherapeutic agent is one of doxorubicin, cisplatin, or any combination thereof.

20. A method for treating a patient, comprising:
a) administering the magnetic composition of claim 1 to at least a portion of a patient, and b) applying an alternating magnetic field (AMF) to the magnetic composition to inductively heat the magnetic material.

21. A method according to claim 19, wherein the magnetic composition of claim 1 is administered directly to the diseased tissue.

22. A magnetic composition according to claim 19, wherein the composition further comprises a ligand specific to a predetermined target in the patient.

23. A method according to claim 21, wherein the target is associated with a cancer, a disease of the subject's vascular system, a disease-causing pathogen, multiple sclerosis, or non-cancerous disease material.

24. A method according to claim 22, wherein the target associated with a cancer comprises a marker, and wherein the marker is a) a member of vascular endothelial growth factor receptor (VEGFR) family; b) a member of carcinoembryonic antigen (CEA) family;
c) unglycosylated DF3 antigen; d) a member of epidermal growth factor receptor (EGFR) family; e) a cellular adhesion molecule; f) a matrix metalloproteinase; g) a glycoprotein antigen; h) an angiogen; i) a prostate specific membrane antigen (PSMA); j) a small cell lung carcinoma antigen (SCLCA); k) a hormone receptor; l) a tumor suppressor gene antigen; m) a cell cycle regulator antigen; n) an oncogene antigen; o) an oncogene receptor antigen; p) a proliferation marker; q) a malignant transformation related factor;
r) an apoptosis-related factor; s) a human carcinoma antigen; t) an integrin;
u) a kallikrein; v) a placental growth factor receptor (PGFR); w) an adenovirus-cell surface receptor; x) a hepatocyte growth factor receptor (HGFR); y) a tyrosine kinase;
z) a cytokeratin epithelial marker; aa) a proliferating cell nuclear antigen (PCNA); bb) a membrane associated sialidase; cc) a cancer cell signal mediator; dd) a cyclase-C
receptor; ee) a transforming growth factor receptor (TGFR); ff) a platelet derived growth factor receptor (PDGFR); gg) a cobalamin receptor; hh) a glioma channel; ii) a brain specific chondroitin sulphate proteoglycan; jj) a catenin; kk) a member of MUC-type mucin family receptors; ll) a member of cluster designation/differentiation (CD) antigen family; mm) a protein antigen; nn) a cytokine receptor; oo) a mesothelin receptor; or pp) any combination of a) through oo).

25. A method according to claim 21, wherein the ligand to the marker is a) a polyclonal antibody; b) a monoclonal antibody; c) a chimeric antibody; d) a humanized antibody; e) a human antibody; f) a recombinant antibody; g) a bispecific antibody; h) an antibody fragment; i) a recombinant single chain antibody fragment; or j) any combination of a) through i).

26. A method according to claim 21, wherein the marker epidermal growth factor receptor (EGFR) comprises HER-1, HER-2, HER-3, HER-4, EGFRvIII, or any combination thereof.

27. A method according to claim 25, wherein the ligand is an antibody to marker HER-2, a variant of antibody to marker HER-2, or any combination thereof.

28. A method according to claim 26, wherein the variant of antibody to marker HER-2 is F5 scFv, IDM-1 (MDX-210), or any combination thereof.

29. A method according to claim 24, wherein the ligand is an antibody to marker EGFRvIII, a variant of antibody to marker EGFRvIII, or any combination thereof.

30. A method according to claim 28, wherein the variant of antibody to marker EGFRvIII is Ua30:2, L8A4, DH8.3, 81C6, or any combination thereof.

31. A method according to claim 23, wherein the marker MUC-type mucin family receptors comprises MUC-1, MUC-2, MUC-3, TAG-72, human milk fat globule receptor, or any combination thereof.

32. A method according to claim 24, wherein the ligand is an antibody to marker MUC-1, a variant of antibody to marker MUC-1, or any combination thereof.

33. A method according to claim 24, wherein the ligand is an antibody to marker TAG-72, a variant of antibody to marker TAG-72, or any combination thereof.

34. A method according to claim 32, wherein the variant of antibody to marker TAG-72 is B72.3.

35. A method according to claim 24, wherein the ligand is an antibody to mark-er CEA, a variant of antibody to marker CEA, or any combination thereof.

36. A method according to claim 34, wherein the variant of antibody to marker CEA is MFE-23 scFv.

37. A method according to claim 23, wherein the marker designation/differentiation protein comprises CD44, and wherein CD44 serves as a cellular adhesion molecule.

38. A method according to claim 23, wherein the marker cytokine receptor comprises at least one member of the interleukin (IL) family.

39. A method according to claim 24, wherein the ligand is an antibody to marker IL13, a variant of antibody to marker IL13, or any combination thereof.

40. A method according to claim 23, wherein the marker matrix metalloproteinase comprises matrix metalloproteinase 9 (MMP-9) 41. A method according to claim 23, wherein the marker glycoprotein antigen comprises a 43 kD membrane associated glycoprotein antigen, a 40 kD glycoprotein antigen, or any combination thereof.

42. A method according to claim 24, wherein the ligand is an antibody to marker 43 kD
membrane associated glycoprotein antigen, a variant of antibody to marker 43 kD
membrane associated glycoprotein antigen, or any combination thereof.

43. A method according to claim 41, wherein the variant of antibody to marker 43 kD
membrane associated glycoprotein antigen is 323/A3.

44. A method according to claim 24, wherein the ligand is an antibody to marker 40 kD
glycoprotein antigen, a variant of antibody to marker 40 kD glycoprotein antigen, or any combination thereof.

45. A method according to claim 43, wherein the variant of antibody to marker 40 kD
glycoprotein antigen is NR-LU- 10.

46. A method according to claim 23, wherein the marker angiogen comprises a vascular endothelial growth factor receptor (VEGFR), integrin av.beta.3, a urokinase type plasminogen activator receptor (uPAR), a plasminogen activator inhibitor 1(PAI-1), VEGFR 2(KDR/Flk-1), or any combination thereof.

47. A method according to claim 45, wherein the vascular endothelial growth factor receptor (VEGFR) comprises FLT1, FLK1, Tie1, Tie2, or any combination thereof.

48. A method according to claim 24, wherein the ligand is an antibody to marker integrin av.beta.3, a variant of antibody to integrin av.beta.3, or any combination thereof.

49. A method according to claim 47, wherein the variant of antibody to marker integrin av.beta.3 is Ber EP4, LM609, 2C3, or any combination thereof.

50. A method according to claim 24, wherein the ligand is an antibody to marker prostate specific membrane antigen, a variant of antibody to marker prostate specific membrane antigen, or any combination thereof.

51. A method according to claim 24, wherein the ligand is an antibody to marker prostate specific membrane antigen, a variant of antibody to marker prostate specific membrane antigen, or any combination thereof.

52. A method according to claim 50, wherein the variant of antibody to marker prostate specific membrane antigen is MID-070, 7E11-C5.3, or any combination thereof.

53. A method according to claim 23, wherein the marker adenovirus-cell surface receptor comprises coxsackie adenovirus cell surface receptor (CAR).

54. A method according to claim 23, wherein the marker cell cycle regulator comprises cyclin A, cyclin D, cyclin E, cdc2, or any combination thereof.

55. A method according to claim 23, wherein the marker oncogene comprises ras.

56. A method according to claim 23, wherein the marker apoptosis related factor comprises Fas, FasL, or any combination thereof.

57. A method according to claim 23, wherein the marker protein tyrosine kinase comprises VSrc, C-Src, or any combination thereof.

58. A method according to claim 23, wherein the marker cancer cell signal mediator comprises P13KC2.

59. A method according to claim 23, wherein the marker cyclase-C receptor comprises guanylyl cyclase-C (GC-C) receptor.

60. A method according to claim 23, wherein the marker platelet derived growth factor receptor (PDGFR) comprises PDGFR-alpha, PDGFR-beta, or any combination thereof.

61. A method according to claim 23, wherein the marker the cobalamin receptor comprises methionine synthase, L-methylmalonyl-CoA mutase, or any combination thereof.

62. A method according to claim 23, wherein the marker glioma channel comprises glioma chloride channel.

63. A method according to claim 23, wherein the marker brain-specific chondroitin sulphate proteoglycan comprises brain enriched hyaluronan binding (BEHAB) protein receptor.

64. A method according to claim 23, wherein the marker catenin comprises alpha catenin, beta catenin, gamma catenin, or any combination thereof.

65. A method according to claim 23, wherein the marker protein antigen comprises p27, p73, or any combination thereof.

66. A method according to claim 24, wherein the ligand is an antibody to marker human milk fat globule receptor (HMFGR), a variant of antibody to marker HMFGR, or any combination thereof.

67. A method according to claim 65, wherein the variant of antibody to marker HMFGR is NCL-HMFG1, NCL-HMFG2, or any combination thereof.

68. A method according to claim 22, wherein the target associated with a disease of the subject's vascular system comprises a marker, and wherein the marker is an antigen associated with an apolipoprotein, a lipoprotein, a vascular endothelial growth factor receptor (VEGFR), basic fibroblast growth factor receptor (bFGFR), or my combination thereof.

69. A method according to claim 67, wherein the marker lipoprotein comprises oxidized low density lipoprotein (OxLDL), malondialdehyde-modified LDL (MDA-LDL), or any combination thereof.

70. A method according to claim 68, wherein the ligand is an antibody to marker OxLDL, a variant of antibody to marker OxLDL, or any combination thereof.

71. A method according to claim 69, wherein the variant of antibody to marker OxLDL is MDA-2.

72. A method according to claim 68, wherein the ligand is an antibody to marker MDA-LDL, a variant of antibody to marker MDA-LDL, or any combination thereof.

73. A method according to claim 71, wherein the variant of antibody to marker MDA-LDL is IK17.

74. A method according to claim 19, wherein the target associated with disease causing pathogen target is a virus, and wherein the virus is associated with tuberculosis.

75. A method according to claim 73, wherein target associated with tuberculosis comprises a marker, and wherein the marker is an antigen associated with APA.

76. A method according to claim 19, wherein the target associated with disease causing pathogen is a virus, and wherein the virus is associated with human immunodeficiency virus (HIV).

77. A method according to claim 75, wherein the target comprises a marker, and wherein the marker is T growth factor receptor alpha (TGFR-A) antigen associated with an HIV
infected cell.

78. A method according to claim 19, wherein the target associated with disease causing pathogen target is a virus, and wherein the virus is associated with malaria.

79. A method according to claim 19, wherein the target associated with a central nervous system disease is multiple sclerosis, and wherein the target comprises a marker, and wherein the marker is an .alpha.4-subunit of .alpha.4.beta.p, -integrin (VLA-4), an .alpha.4-subunit of .alpha.4.beta.7-integrin, or any combination thereof.

80. A method according to claim 19, wherein the target associated with disease causing pathogen comprises a marker, and wherein the marker is an apical membrane antigen-1 (AMA-1) on Plasmodium falciparum.

81. A method according to claim 19, wherein the target associated with non-cancerous disease material comprises a marker, and wherein the marker comprises a non-cancerous disease deposit, a non-cancerous disease precursor deposit, or any combination thereof.

82. A method according to claim 19, wherein the target associated with non-cancerous disease material is a vascular endothelial growth factor receptor associated with autoimmune joint degradation.

83. A method according to claim 20, wherein the AMF is pulsed.

84. A method according to claim 82, wherein the AMF 'on' pulse times are in the range of from about 0.1 seconds to about 1200 seconds, and the 'off' pulse times are in the range of from about 0.1 seconds to about 1200 seconds.

85. A method according to claim 20, wherein the administering utilizes a method that is intraperitoneal injection, intravascular injection, intramuscular injection, subcutaneous injection, topical, inhalation, ingestion, rectal insertion, wash, lavage or rinse perisurgically, extracorporeal, or any combination thereof.

86. A method according to claim 84, wherein the at least a portion of the subject is extracted from the subject's body prior to extracorporeal administration of AMF, and wherein the extracted portion is returned to the subject's body or is transplanted to a recipient's body after the administration of AMF.

87. A method according to claim 85, wherein the extracted portion of the subject is cooled before, during or after the administration of AMF.

88. A method according to claim 85, wherein the magnetic nanoparticle is removed from the extracted portion via a magnetic force after the administration of AMF.

89. A method for treating a patient, comprising:
a) administering the magnetic composition of claim 10 to at least a portion of a patient and b) applying an alternating magnetic field (AMF) to the magnetic composition combined with the predetermined target to inductively heat the magnetic material.

90. A method according to claim 88, wherein the magnetic composition of claim 10 is administered directly to the diseased tissue.

91. A magnetic composition according to claim 89, wherein the composition further comprises a ligand specific to a predetermined target in the patient.

92. A method according to claim 89, wherein the magnetic composition further comprises a radio isotope, potential radioactive isotope, a chemotherapeutic agent, or any combination thereof.

93. A method according to claim 90, wherein the target is associated with a cancer, a disease of the subject's vascular system, a disease-causing pathogen, multiple sclerosis, or non-cancerous disease material.

94. A method according to claim 92, wherein the target associated with a cancer comprises a marker, and wherein the marker is a) a member of vascular endothelial growth factor receptor (VEGFR) family; b) a member of carcinoembryonic antigen (CEA) family;
c) unglycosylated DF3 antigen; d) a member of epidermal growth factor receptor (EGFR) family; e) a cellular adhesion molecule; f) a matrix metalloproteinase; g) a glycoprotein antigen; h) an angiogen; i) a prostate specific membrane antigen (PSMA); j) a small cell lung carcinoma antigen (SCLCA); k) a hormone receptor; l) a tumor suppressor gene antigen; m) a cell cycle regulator antigen; n) an oncogene antigen; o) an oncogene receptor antigen; p) a proliferation marker; q) a malignant transformation related factor;
r) an apoptosis-related factor; s) a human carcinoma antigen; t) an integrin;
u) a kallikrein; v) a placental growth factor receptor (PGFR); w) an adenovirus-cell surface receptor; x) a hepatocyte growth factor receptor (HGFR); y) a tyrosine kinase;
z) a cytokeratin epithelial marker; aa) a proliferating cell nuclear antigen (PCNA); bb) a membrane associated sialidase; cc) a cancer cell signal mediator; dd) a cyclase-C
receptor; ee) a transforming growth factor receptor (TGFR); ff) a platelet derived growth factor receptor (PDGFR); gg) a cobalamin receptor; hh) a glioma channel; ii) a brain specific chondroitin sulphate proteoglycan; jj) a catenin; kk) a member of MUC-type mucin family receptors; ll) a member of cluster designation/differentiation (CD) antigen family; mm) a protein antigen; nn) a cytokine receptor; oo) a mesothelin receptor; or pp) any combination of a) through oo).

95. A method according to claim 93, wherein the ligand to the marker is a) a polyclonal antibody; b) a monoclonal antibody; c) a chimeric antibody; d) a humanized antibody; e) a human antibody; f) a recombinant antibody; g) a bispecific antibody; h) an antibody fragment; i) a recombinant single chain antibody fragment; or j) any combination of a) through i).

96. A method according to claim 94, wherein the ligand is an antibody to marker human milk fat globule receptor (1-HMFGR), a variant of antibody to marker HMFGR, or any combination thereof.

97. A method according to claim 94, wherein the ligand is an antibody to marker EGFRvIII, a variant of antibody to marker EGFRvIII, or any combination thereof.

98. A method according to claim 94, wherein the ligand is an antibody to marker OxLDL, a variant of antibody to marker OxLDL, or any combination thereof.

99. A method according to claim 94, wherein the ligand is an antibody to marker MDA-LDL, a variant of antibody to marker MDA-LDL, or any combination thereof 100. A method according to claim 90, wherein the AMF is pulsed.

101. A method according to claim 90, wherein the administering utilizes a method that is intraperitoneal injection, intravascular injection, intramuscular injection, subcutaneous injection, topical, inhalation, ingestion, rectal insertion, wash, lavage or rinse perisurgically, extracorporeal, or any combination thereof.

102. A method for imaging a patient, comprising:
a) administering the magnetic composition of claim 10 to at least a portion of a patient; and b) applying a magnetic field to the magnetic composition to enhance the image of the portion of the patient containing the magnetic material.

103. A method according to claim 101, wherein the magnetic composition of claim 10 is administered directly to the target tissue.

104. A magnetic composition according to claim 101, wherein the composition further comprises a ligand specific to a predetermined target in the patient.

105. A method according to claim 101, wherein the magnetic composition further comprises a radio isotope or a potential radioactive isotope or any combination thereof to enhance the contrast of the target tissue.
CA002628106A 2005-11-01 2006-11-01 Magnetic nanoscale particle compositions, and therapeutic methods related thereto Abandoned CA2628106A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US73236805P 2005-11-01 2005-11-01
US11/264,680 2005-11-01
US11/264,680 US7731648B2 (en) 2001-07-25 2005-11-01 Magnetic nanoscale particle compositions, and therapeutic methods related thereto
US60/732,368 2005-11-01
PCT/US2006/060419 WO2007079276A2 (en) 2005-11-01 2006-11-01 Magnetic nanoscale particle compositions, and therapeutic methods related thereto

Publications (1)

Publication Number Publication Date
CA2628106A1 true CA2628106A1 (en) 2007-07-12

Family

ID=38228900

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002628106A Abandoned CA2628106A1 (en) 2005-11-01 2006-11-01 Magnetic nanoscale particle compositions, and therapeutic methods related thereto

Country Status (5)

Country Link
US (1) US7731648B2 (en)
EP (1) EP1945159A4 (en)
JP (1) JP2009513722A (en)
CA (1) CA2628106A1 (en)
WO (1) WO2007079276A2 (en)

Families Citing this family (98)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7731648B2 (en) * 2001-07-25 2010-06-08 Aduro Biotech Magnetic nanoscale particle compositions, and therapeutic methods related thereto
US7951061B2 (en) 2001-07-25 2011-05-31 Allan Foreman Devices for targeted delivery of thermotherapy, and methods related thereto
JP4583372B2 (en) * 2003-04-15 2010-11-17 コーニンクレッカ フィリップス エレクトロニクス エヌ ヴィ Method and apparatus for improved identification of the spatial distribution of non-aggregated magnetic particles in an examination region
JP5010914B2 (en) * 2003-04-15 2012-08-29 コーニンクレッカ フィリップス エレクトロニクス エヌ ヴィ Method for determining spatial distribution of magnetic particles and composition for administering magnetic particles
ITRM20030376A1 (en) 2003-07-31 2005-02-01 Univ Roma PROCEDURE FOR THE ISOLATION AND EXPANSION OF CARDIOC STAMIN CELLS FROM BIOPSIA.
US20050090732A1 (en) * 2003-10-28 2005-04-28 Triton Biosystems, Inc. Therapy via targeted delivery of nanoscale particles
US8060179B1 (en) 2006-11-16 2011-11-15 Scientific Nanomedicine, Inc. Biomagnetic detection and treatment of Alzheimer's Disease
US8118754B1 (en) 2007-11-15 2012-02-21 Flynn Edward R Magnetic needle biopsy
US7365289B2 (en) * 2004-05-18 2008-04-29 The United States Of America As Represented By The Department Of Health And Human Services Production of nanostructures by curie point induction heating
CN1733314A (en) * 2004-08-11 2006-02-15 张阳德 Process for preparing cerebrose albumin magnetic adriamycin nanometer particle
US11660317B2 (en) 2004-11-08 2023-05-30 The Johns Hopkins University Compositions comprising cardiosphere-derived cells for use in cell therapy
US9964469B2 (en) 2005-02-28 2018-05-08 Imagion Biosystems, Inc. Magnetic needle separation and optical monitoring
WO2007035871A1 (en) * 2005-09-21 2007-03-29 Massachusetts Institute Of Technology Systems and methods for tuning properties of nanoparticles
US7744596B2 (en) * 2005-10-13 2010-06-29 Boston Scientific Scimed, Inc. Magnetically augmented radio frequency ablation
US20070140974A1 (en) * 2005-12-15 2007-06-21 General Electric Company Targeted nanoparticles for magnetic resonance imaging
US20080187487A1 (en) * 2006-05-03 2008-08-07 Gustavo Larsen Methods for producing multilayered particles, fibers and sprays and methods for administering the same
WO2008060393A2 (en) * 2006-10-24 2008-05-22 Iso Therapeutics Group Llc The use of materials and external stimuli for synovectomy
US8447379B2 (en) 2006-11-16 2013-05-21 Senior Scientific, LLC Detection, measurement, and imaging of cells such as cancer and other biologic substances using targeted nanoparticles and magnetic properties thereof
US8333993B1 (en) 2006-12-29 2012-12-18 University Of Central Florida Research Foundation, Inc. Synthesis of polymer coated ceria nanoparticles for biomedical applications
WO2008084477A2 (en) * 2007-01-08 2008-07-17 Yossi Gross In-situ filter
KR20100014809A (en) * 2007-01-19 2010-02-11 트리톤 바이오시스템즈, 인코포레이티드 Thermotherapy susceptors and methods of using same
KR101043251B1 (en) * 2007-04-12 2011-06-21 연세대학교 산학협력단 Magnetic Resonance Imaging Contrast Agents Comprising Zinc Containing Magnetic Metal Oxide Nanoparticles
US20090306646A1 (en) * 2007-05-14 2009-12-10 Bsd Medical Corporation Apparatus and method for injection enhancement of selective heating of a deposit in tissues in a body
US9387036B2 (en) * 2007-05-14 2016-07-12 Pyrexar Medical Inc. Apparatus and method for selectively heating a deposit in fatty tissue in a body
EP2005973A1 (en) * 2007-06-22 2008-12-24 nanoPET Pharma GmbH Compositions containing positron emitting inorganic particles and their use in medicine, in particular for diagnostic procedures
US9119391B1 (en) 2007-07-16 2015-09-01 University Of Central Florida Research Foundation, Inc. Polymer coated ceria nanoparticles for selective cytoprotection
AU2008288082B2 (en) * 2007-08-15 2013-01-10 Yeda Research And Development Co. Ltd. Regulators of MMP-9 and uses thereof
US8467842B2 (en) * 2010-02-10 2013-06-18 Tokitae Llc Systems, devices, and methods including multi-harmonic optical detection of hemozoin nanoparticles
US8888674B2 (en) 2007-12-11 2014-11-18 University Of Maryland College Park Methods and systems for magnetic focusing of therapeutic, diagnostic or prophylactic agents to deep targets
US8385997B2 (en) 2007-12-11 2013-02-26 Tokitae Llc Spectroscopic detection of malaria via the eye
KR100998569B1 (en) * 2008-03-31 2010-12-07 한국원자력연구원 A radioimmunoconjugate for diagnosis and treatment of cancer or metastasis and development of cancer or cancer metastasis inhibitor using thereof
US9011913B2 (en) * 2008-04-04 2015-04-21 The Regents Of The University Of California Use of functionalized magnetic nanoparticles in cancer detection and treatment
JP2011517604A (en) * 2008-04-09 2011-06-16 コーネル・ユニバーシティー Nanoparticle-mediated microwave treatment
EP2281004A4 (en) * 2008-04-14 2012-02-15 Proscan Rx Pharma Inc Prostate specific membrane antigen antibodies and antigen binding fragments
CN102067250B (en) * 2008-04-16 2014-08-27 干细胞技术公司 Magnetic particles
US8579787B2 (en) 2008-05-19 2013-11-12 University Of Maryland College Park Methods and systems for using therapeutic, diagnostic or prophylactic magnetic agents
KR101101832B1 (en) * 2008-05-20 2012-01-05 연세대학교 산학협력단 Heat Generating Nanomaterials
WO2010042555A2 (en) * 2008-10-06 2010-04-15 The Brigham And Women's Hospital, Inc. Particles with multiple functionalized surface domains
US20130023714A1 (en) * 2008-10-26 2013-01-24 Board Of Regents, The University Of Texas Systems Medical and Imaging Nanoclusters
WO2010057050A1 (en) * 2008-11-14 2010-05-20 Columbia University Applying torque to paramagnetic structures in bodies using dual magnetic fields
EP2387787B1 (en) 2008-12-19 2018-09-12 Ferronova Pty Ltd Magnetic nanoparticles
US20120315480A1 (en) * 2009-07-31 2012-12-13 Universidad Del Pais Vasco Copper nanoparticles with magnetic properties
EP2470131B1 (en) * 2009-08-24 2014-11-12 New Phase Ltd Phase-change materials
TW201114437A (en) * 2009-10-20 2011-05-01 Univ Nat Chiao Tung Magnetic nanoparticles for magnetic resonace imaging
WO2011050049A2 (en) * 2009-10-21 2011-04-28 The University Of North Carolina At Chapel Hill Delivery systems for brachytherapy, and associated methods
US8565892B2 (en) 2009-10-31 2013-10-22 Qteris, Inc. Nanoparticle-sized magnetic absorption enhancers having three-dimensional geometries adapted for improved diagnostics and hyperthermic treatment
CN102695542B (en) * 2009-11-02 2015-08-12 脉冲治疗公司 For magnetic potential stator system and the method for controlled in wireless magnet rotor
US10194825B2 (en) 2009-11-06 2019-02-05 Imagion Biosystems Inc. Methods and apparatuses for the localization and treatment of disease such as cancer
KR20120087165A (en) 2009-11-06 2012-08-06 사이언티픽 나노메디슨, 아이엔씨. Detection, measurement, and imaging of cells such as cancer and other biologic substances using targeted nanoparticles and magnetic properties thereof
US8781184B2 (en) 2010-02-10 2014-07-15 Tokitae Llc Systems, devices, and methods for detection of malaria
US9044141B2 (en) * 2010-02-10 2015-06-02 Tokitae Llc Systems, devices, and methods including a dark-field reflected-illumination apparatus
EP2558108A1 (en) * 2010-04-12 2013-02-20 Ramot at Tel-Aviv University Ltd Iron oxide nanoparticles for use in treating non-infectious inflammatory disorders
US9845457B2 (en) 2010-04-30 2017-12-19 Cedars-Sinai Medical Center Maintenance of genomic stability in cultured stem cells
US9249392B2 (en) 2010-04-30 2016-02-02 Cedars-Sinai Medical Center Methods and compositions for maintaining genomic stability in cultured stem cells
WO2011156471A2 (en) * 2010-06-08 2011-12-15 Northwestern University Compositions and methods for thermoradiotherapy
DE102010050644A1 (en) * 2010-11-09 2012-05-10 Studiengesellschaft Kohle Mbh Process for the preparation of carbon-protected superparamagnetic or magnetic nanospheres
US20120197063A1 (en) * 2011-01-31 2012-08-02 Rainer Meinke Systems and Methods Which Remove Material From Blood Vessel Walls
US8197471B1 (en) * 2011-02-14 2012-06-12 Samuel Harry Tersigni Core-excited nanoparticles and methods of their use in the diagnosis and treatment of disease
KR101468769B1 (en) * 2011-03-04 2014-12-08 사회복지법인 삼성생명공익재단 Magnetic nanocomposite specific for thyroid cancer and use thereof
US20120283503A1 (en) * 2011-04-29 2012-11-08 The Johns Hopkins University Nanoparticle loaded stem cells and their use in mri guided hyperthermia
US10773095B2 (en) * 2011-06-21 2020-09-15 Lockheed Martin Corporation Direct magnetic imaging with metamaterial for focusing and thermal ablation using SPION nanoparticles for cancer diagnosis and treatment
JP5844902B2 (en) 2011-08-10 2016-01-20 マグフォース アーゲー Aggregation of magnetic nanoparticles coated with alkoxysilanes
US9005151B2 (en) 2011-09-07 2015-04-14 Choon Kee Lee Thermal apparatus
AU2012358364B2 (en) 2011-12-21 2017-08-10 Iso Therapeutics Group Llc Radioactive compositions and methods for their therapeutic use
DE112013001753T5 (en) * 2012-03-29 2015-02-19 Spiration, Inc. Devices, methods and systems for the identification and treatment of lung tissue
US9883878B2 (en) 2012-05-15 2018-02-06 Pulse Therapeutics, Inc. Magnetic-based systems and methods for manipulation of magnetic particles
US9884076B2 (en) 2012-06-05 2018-02-06 Capricor, Inc. Optimized methods for generation of cardiac stem cells from cardiac tissue and their use in cardiac therapy
JP6433896B2 (en) 2012-08-13 2018-12-05 シーダーズ−サイナイ・メディカル・センターCedars−Sinai Medical Center Exosomes and microribonucleic acids for tissue regeneration
KR101409296B1 (en) * 2012-09-07 2014-06-24 서울대학교산학협력단 Method of selective activation for magnetic nanoparticle and selectively activated magnetic nanoparticle
KR101868675B1 (en) * 2012-10-05 2018-06-18 연세대학교 산학협력단 Compositions for Hyperthermia Therapy comprising Sensitizing Material
WO2014074584A1 (en) 2012-11-06 2014-05-15 Sakhrat Khizroev On-demand drug release using magneto-electric nanoparticles
US9895549B2 (en) 2012-11-06 2018-02-20 The Florida International University Board Of Trustees On-demand drug release using magneto-electric nanoparticles
US10406228B2 (en) * 2012-11-29 2019-09-10 The Johns Hopkins University Process for making iron oxide nanoparticle preparations for cancer hyperthermia
US9409148B2 (en) 2013-08-08 2016-08-09 Uchicago Argonne, Llc Compositions and methods for direct capture of organic materials from process streams
US20150092811A1 (en) * 2013-09-24 2015-04-02 Worcester Polytechnic Institute Nanoparticles-Based Taggant Systems and Methods
GB201412040D0 (en) * 2014-07-07 2014-08-20 Nottingham University Hospitals Nhs Trust Magnetic resonance imaging methods for the study of gastronintestinal transit
US10188731B2 (en) * 2014-08-14 2019-01-29 Ping Liang Methods for killing cancer cells and cellular imaging using magneto-electric nano-particles and external magnetic field
WO2016054591A1 (en) 2014-10-03 2016-04-07 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of muscular dystrophy
EP3226819B1 (en) 2014-11-25 2018-10-24 New Phase Ltd. Phase-change nanoparticle
EP3389630B1 (en) 2015-12-16 2023-11-08 Gritstone bio, Inc. Neoantigen identification, manufacture, and use
US11253551B2 (en) 2016-01-11 2022-02-22 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of heart failure with preserved ejection fraction
WO2017210360A1 (en) 2016-05-31 2017-12-07 Cardiovax, Llc Methods for diagnosing and treating systemic lupus erythematosus
US10858422B2 (en) 2016-05-31 2020-12-08 Abcentra, Llc Methods for treating systemic lupus erythematosus with an anti-apolipoprotein B antibody
WO2017210652A1 (en) 2016-06-03 2017-12-07 Cedars-Sinai Medical Center Cdc-derived exosomes for treatment of ventricular tachyarrythmias
WO2018013935A1 (en) 2016-07-14 2018-01-18 The Board Of Regents Of The University Of Texas System Methods, apparatuses, and systems for inductive heating of foreign metallic implants
EP3515459A4 (en) 2016-09-20 2020-08-05 Cedars-Sinai Medical Center Cardiosphere-derived cells and their extracellular vesicles to retard or reverse aging and age-related disorders
AU2018255346A1 (en) 2017-04-19 2019-11-07 Capricor, Inc. Methods and compositions for treating skeletal muscular dystrophy
JP7336994B2 (en) 2017-06-13 2023-09-01 シャー,ホウン・サイモン Compositions and methods for enhancing hyperthermia treatment
GB2567132A (en) * 2017-08-10 2019-04-10 Hipermag Ltd Magnetic structures
WO2019038219A1 (en) * 2017-08-21 2019-02-28 INSERM (Institut National de la Santé et de la Recherche Médicale) New prognostic method of pancreatic cancer
JP7227237B2 (en) 2017-10-10 2023-02-21 グリットストーン バイオ インコーポレイテッド Identification of neoantigens using hotspots
CA3083097A1 (en) 2017-11-22 2019-05-31 Gritstone Oncology, Inc. Reducing junction epitope presentation for neoantigens
WO2019120489A1 (en) * 2017-12-19 2019-06-27 Medical Development Technologies S.A. Heatable implant device for tumor treatment
US11660355B2 (en) 2017-12-20 2023-05-30 Cedars-Sinai Medical Center Engineered extracellular vesicles for enhanced tissue delivery
AU2019200986A1 (en) 2018-02-22 2019-09-05 Robert E. Sandstrom Magnetic Field Enhancement of Chemotherapy for Tumor Treatment
US11918315B2 (en) 2018-05-03 2024-03-05 Pulse Therapeutics, Inc. Determination of structure and traversal of occlusions using magnetic particles
CN112538519B (en) * 2019-09-20 2022-09-27 华中农业大学 Method for detecting food-borne pathogenic bacteria by enzymatic low-field nuclear magnetic resonance DNA sensor
CN117794618A (en) * 2021-06-14 2024-03-29 诺沃库勒有限责任公司 Methods for treating and preventing cancer using alternating electric fields, radioactive particles and systemic therapies

Family Cites Families (96)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CU22545A1 (en) * 1994-11-18 1999-03-31 Centro Inmunologia Molecular OBTAINING A CHEMICAL AND HUMANIZED ANTIBODY AGAINST THE RECEPTOR OF THE EPIDERMAL GROWTH FACTOR FOR DIAGNOSTIC AND THERAPEUTIC USE
US4106488A (en) * 1974-08-20 1978-08-15 Robert Thomas Gordon Cancer treatment method
US4303636A (en) * 1974-08-20 1981-12-01 Gordon Robert T Cancer treatment
US4119102A (en) * 1975-07-11 1978-10-10 Leveen Harry H Radio frequency treatment of tumors while inducing hypotension
FR2421628A1 (en) * 1977-04-08 1979-11-02 Cgr Mev LOCALIZED HEATING DEVICE USING VERY HIGH FREQUENCY ELECTROMAGNETIC WAVES, FOR MEDICAL APPLICATIONS
US4323056A (en) 1980-05-19 1982-04-06 Corning Glass Works Radio frequency induced hyperthermia for tumor therapy
US4392040A (en) * 1981-01-09 1983-07-05 Rand Robert W Induction heating apparatus for use in causing necrosis of neoplasm
US4569836A (en) * 1981-08-27 1986-02-11 Gordon Robert T Cancer treatment by intracellular hyperthermia
US4574782A (en) * 1981-11-16 1986-03-11 Corning Glass Works Radio frequency-induced hyperthermia for tumor therapy
US4454234A (en) * 1981-12-30 1984-06-12 Czerlinski George H Coated magnetizable microparticles, reversible suspensions thereof, and processes relating thereto
US4452773A (en) * 1982-04-05 1984-06-05 Canadian Patents And Development Limited Magnetic iron-dextran microspheres
US4622952A (en) * 1983-01-13 1986-11-18 Gordon Robert T Cancer treatment method
EP0330801A1 (en) * 1983-02-08 1989-09-06 Schering Aktiengesellschaft Ferromagnetic, diamagnetic or paramagnetic particles useful in the diagnosis and treatment of disease
US4735796A (en) * 1983-12-08 1988-04-05 Gordon Robert T Ferromagnetic, diamagnetic or paramagnetic particles useful in the diagnosis and treatment of disease
US4545368A (en) * 1983-04-13 1985-10-08 Rand Robert W Induction heating method for use in causing necrosis of neoplasm
US4662359A (en) * 1983-08-12 1987-05-05 Robert T. Gordon Use of magnetic susceptibility probes in the treatment of cancer
US4590922A (en) * 1983-08-19 1986-05-27 Gordon Robert T Use of ferromagnetic, paramagnetic and diamagnetic particles in the treatment of infectious diseases
DE3332843A1 (en) 1983-09-12 1985-04-04 Broers, Dieter, 8079 Pfalzpaint DEVICE FOR THE TREATMENT OF LIVING TISSUE WITH ELECTROMAGNETIC WAVES FOR THE PURPOSE OF THE THERAPEUTIC INFLUENCATION IN THE EVENT OF DISEASES
US6054561A (en) * 1984-02-08 2000-04-25 Chiron Corporation Antigen-binding sites of antibody molecules specific for cancer antigens
US5169774A (en) * 1984-02-08 1992-12-08 Cetus Oncology Corporation Monoclonal anti-human breast cancer antibodies
US4753894A (en) * 1984-02-08 1988-06-28 Cetus Corporation Monoclonal anti-human breast cancer antibodies
US4610241A (en) * 1984-07-03 1986-09-09 Gordon Robert T Atherosclerosis treatment method
US4767611A (en) * 1984-07-03 1988-08-30 Gordon Robert T Method for affecting intracellular and extracellular electric and magnetic dipoles
US5087438A (en) * 1984-07-03 1992-02-11 Gordon Robert T Method for affecting intracellular and extracellular electric and magnetic dipoles
US4889120A (en) * 1984-11-13 1989-12-26 Gordon Robert T Method for the connection of biological structures
US4983159A (en) * 1985-03-25 1991-01-08 Rand Robert W Inductive heating process for use in causing necrosis of neoplasms at selective frequencies
US4708718A (en) * 1985-07-02 1987-11-24 Target Therapeutics Hyperthermic treatment of tumors
US4678667A (en) * 1985-07-02 1987-07-07 501 Regents of the University of California Macrocyclic bifunctional chelating agents
US5776093A (en) * 1985-07-05 1998-07-07 Immunomedics, Inc. Method for imaging and treating organs and tissues
US4758429A (en) * 1985-11-04 1988-07-19 Gordon Robert T Method for the treatment of arthritis and inflammatory joint diseases
DE3719705A1 (en) * 1986-06-13 1987-12-17 Olympus Optical Co DEVICE FOR HYPERTHERMAL HEATING THE BODY
US4950221A (en) * 1986-07-18 1990-08-21 Gordon Robert T Process for affecting molecules in tissue
US4923437A (en) * 1986-07-18 1990-05-08 Gordon Robert T Process for applying a localized magnetic or electric field
US4813399A (en) * 1986-07-18 1989-03-21 Gordon Robert T Process for the treatment of neurological or neuromuscular diseases and development
US4996991A (en) * 1986-07-18 1991-03-05 Gordon Robert T Method for following the distribution of particles in neurological or neuromuscular tissue and cells
WO1989006692A1 (en) * 1988-01-12 1989-07-27 Genentech, Inc. Method of treating tumor cells by inhibiting growth factor receptor function
EP0333381A3 (en) 1988-03-16 1990-07-04 Metcal Inc. Thermal seed for treatment of tumors
US5300750A (en) * 1988-03-16 1994-04-05 Metcal, Inc. Thermal induction heater
US5612019A (en) * 1988-12-19 1997-03-18 Gordon, Deceased; David Diagnosis and treatment of HIV viral infection using magnetic metal transferrin particles
US5128147A (en) * 1989-01-06 1992-07-07 Thermal Developments, Inc. Heat intensifier and localizer for radiofrequency thermotherapy
DK0474727T3 (en) * 1989-05-19 1998-01-12 Genentech Inc HER2 extracellular domain
US5099756A (en) 1989-06-01 1992-03-31 Harry H. Leveen Radio frequency thermotherapy
US5705157A (en) * 1989-07-27 1998-01-06 The Trustees Of The University Of Pennsylvania Methods of treating cancerous cells with anti-receptor antibodies
US5547682A (en) * 1989-12-22 1996-08-20 Bioquest, Incorporated Preparation and use of novel injectable RES avoiding inorganic particles for medical application
US5935866A (en) * 1989-12-22 1999-08-10 Binax Nh, Inc. Preparation of sub 100 A magnetic particles and magnetic molecular switches
US5441746A (en) * 1989-12-22 1995-08-15 Molecular Bioquest, Inc. Electromagnetic wave absorbing, surface modified magnetic particles for use in medical applications, and their method of production
US5067952A (en) * 1990-04-02 1991-11-26 Gudov Vasily F Method and apparatus for treating malignant tumors by local hyperpyrexia
US5203782A (en) * 1990-04-02 1993-04-20 Gudov Vasily F Method and apparatus for treating malignant tumors by local hyperpyrexia
US5834229A (en) * 1991-05-24 1998-11-10 Genentech, Inc. Nucleic acids vectors and host cells encoding and expressing heregulin 2-α
JPH06254168A (en) * 1991-10-29 1994-09-13 Tanaka Kikinzoku Kogyo Kk Inside-tissue heating/warming therapy
EP0543498B1 (en) 1991-11-20 1998-04-08 Kabushiki Kaisha Riken Injectable powder for the treatment of cancer
US5413588A (en) * 1992-03-06 1995-05-09 Urologix, Inc. Device and method for asymmetrical thermal therapy with helical dipole microwave antenna
EP0636029B1 (en) * 1992-04-13 2000-11-08 Dana-Farber Cancer Institute, Inc. Antibodies specific for carcinoma-associated antigens
US5342757A (en) 1992-11-13 1994-08-30 Ludwig Institute For Cancer Research Monoclonal antibodies which specifically binds to endosialin, a 165 Kd glycoprotein found on tumor vascular endothelium, and uses thereof
ATE156706T1 (en) * 1993-03-17 1997-08-15 Silica Gel Gmbh SUPERPARAMAGNETIC PARTICLES, METHOD FOR THE PRODUCTION AND USE OF THE SAME
US5411730A (en) * 1993-07-20 1995-05-02 Research Corporation Technologies, Inc. Magnetic microparticles
US5429583A (en) * 1993-12-09 1995-07-04 Pegasus Medical Technologies, Inc. Cobalt palladium seeds for thermal treatment of tumors
AU2194695A (en) * 1994-03-28 1995-10-17 Regents Of The University Of California, The Method for preparing radionuclide-labeled chelating agent-ligand complexes
WO1997003701A2 (en) * 1995-07-14 1997-02-06 Glycotech Corp. Compounds and methods for treatment of egf receptor associated cancers and purification of the egf receptor
US5658234A (en) * 1995-07-24 1997-08-19 J. D. Technologies, Inc. Method for treating tumors
US6190870B1 (en) * 1995-08-28 2001-02-20 Amcell Corporation Efficient enrichment and detection of disseminated tumor cells
AUPN568095A0 (en) * 1995-09-27 1995-10-26 Austin Research Institute, The Anti-Galalpha(1,3)Gal antibody binding peptides
DE19612001A1 (en) * 1996-03-18 1997-09-25 Silica Gel Gmbh Adsorptions Te Superparamagnetic particles with increased R¶1¶ relaxivity, process for their production and their use
US5968511A (en) * 1996-03-27 1999-10-19 Genentech, Inc. ErbB3 antibodies
AUPN978296A0 (en) * 1996-05-10 1996-05-30 Gray, Bruce N Targeted hysteresis hyperthermia as a method for treating cancer
US5922845A (en) * 1996-07-11 1999-07-13 Medarex, Inc. Therapeutic multispecific compounds comprised of anti-Fcα receptor antibodies
US5976067A (en) * 1997-05-28 1999-11-02 Ablation Technologies, Inc. Combination radioactive and temperature self-regulating thermal seed implant for treating tumors
DE19726282A1 (en) * 1997-06-20 1998-12-24 Inst Neue Mat Gemein Gmbh Nanoscale particles with an iron oxide-containing core surrounded by at least two shells
US6165440A (en) * 1997-07-09 2000-12-26 Board Of Regents, The University Of Texas System Radiation and nanoparticles for enhancement of drug delivery in solid tumors
WO1999019000A1 (en) 1997-10-11 1999-04-22 The Research Foundation Of State University Of New York Controlled size polymeric microspheres with superparamagnetic cores
AUPP008197A0 (en) 1997-10-29 1997-11-20 Paragon Medical Limited Improved targeted hysteresis hyperthermia as a method for treating diseased tissue
US6242196B1 (en) * 1997-12-11 2001-06-05 Dana-Farber Cancer Institute Methods and pharmaceutical compositions for inhibiting tumor cell growth
US6037129A (en) * 1998-05-28 2000-03-14 Medical University Of South Carolina Multi-marker RT-PCR panel for detecting metastatic breast cancer
US6252050B1 (en) * 1998-06-12 2001-06-26 Genentech, Inc. Method for making monoclonal antibodies and cross-reactive antibodies obtainable by the method
US6391026B1 (en) * 1998-09-18 2002-05-21 Pro Duct Health, Inc. Methods and systems for treating breast tissue
US6387888B1 (en) * 1998-09-30 2002-05-14 American Foundation For Biological Research, Inc. Immunotherapy of cancer through expression of truncated tumor or tumor-associated antigen
AUPP899899A0 (en) * 1999-03-03 1999-03-25 Paragon Medical Limited Magnetic material
US6470220B1 (en) * 1999-03-29 2002-10-22 The Regents Of The University Of California Diagnosis and treatment of cancers using in vivo magnetic domains
DE19937493C2 (en) * 1999-08-07 2001-06-07 Mfh Hyperthermiesysteme Gmbh Magnetic field applicator for heating magnetic or magnetizable substances or solids in biological tissue
DE19937492C2 (en) 1999-08-07 2001-08-23 Mfh Hyperthermiesysteme Gmbh Magnetic field applicator for heating magnetic or magnetizable substances or solids in biological tissue
DE19940220B4 (en) * 1999-08-19 2007-05-03 Magforce Nanotechnologies Ag Medical preparation for the treatment of osteoarthritis, arthritis and other rheumatic joint diseases
SE9903185D0 (en) 1999-09-08 1999-09-08 Europ I Of Science Ab Therapeutic method and device based on magnetism
US6514481B1 (en) * 1999-11-22 2003-02-04 The Research Foundation Of State University Of New York Magnetic nanoparticles for selective therapy
US6347633B1 (en) * 2000-01-14 2002-02-19 First Circle Medical, Inc. Treatment of hepatitis C using hyperthermia
DE10109105C2 (en) * 2001-02-24 2003-01-09 Mfh Hyperthermiesysteme Gmbh Magnetic coil arrangement of a magnetic field applicator for heating magnetic or magnetizable substances or solids in biological tissue
AU2002320058A1 (en) * 2001-06-06 2002-12-16 The General Hspital Corporation Magnetic-nanoparticle conjugates and methods of use
US7731648B2 (en) * 2001-07-25 2010-06-08 Aduro Biotech Magnetic nanoscale particle compositions, and therapeutic methods related thereto
US7074175B2 (en) * 2001-07-25 2006-07-11 Erik Schroeder Handy Thermotherapy via targeted delivery of nanoscale magnetic particles
US6997863B2 (en) * 2001-07-25 2006-02-14 Triton Biosystems, Inc. Thermotherapy via targeted delivery of nanoscale magnetic particles
DE10156790A1 (en) 2001-11-19 2003-06-18 Chemagen Biopolymer Technologi Device and method for treating magnetic particles
WO2003047633A2 (en) 2001-12-04 2003-06-12 Nanospectra Biosciences, Inc. Treatment of angiogenesis disorders using targeted nanoparticles
BR0307206A (en) * 2002-01-24 2004-12-21 Barnes Jewish Hospital Integrin-directed Imaging Agents
JP2004105722A (en) 2002-08-29 2004-04-08 Kansai Tlo Kk Heating element for thermotherapy and manufacturing method therefor
DE10331439B3 (en) * 2003-07-10 2005-02-03 Micromod Partikeltechnologie Gmbh Magnetic nanoparticles with improved magnetic properties
FR2855315B1 (en) * 2003-05-23 2005-08-19 Centre Nat Rech Scient NEUTRAL-STABLE FERROFLUIDS AND MODIFIED FERROFLUIDS OBTAINED BY MODIFICATION OF THE PARTICLE SURFACE OF THESE FERROFLUIDS
US7842281B2 (en) * 2004-05-10 2010-11-30 The Florida State University Research Foundation Magnetic particle composition for therapeutic hyperthermia

Also Published As

Publication number Publication date
EP1945159A4 (en) 2011-12-14
US7731648B2 (en) 2010-06-08
WO2007079276A3 (en) 2008-02-21
US20060142749A1 (en) 2006-06-29
WO2007079276A2 (en) 2007-07-12
US20070112339A9 (en) 2007-05-17
JP2009513722A (en) 2009-04-02
EP1945159A2 (en) 2008-07-23

Similar Documents

Publication Publication Date Title
US7731648B2 (en) Magnetic nanoscale particle compositions, and therapeutic methods related thereto
US20050090732A1 (en) Therapy via targeted delivery of nanoscale particles
Soetaert et al. Cancer therapy with iron oxide nanoparticles: Agents of thermal and immune therapies
US20080213382A1 (en) Thermotherapy susceptors and methods of using same
US6997863B2 (en) Thermotherapy via targeted delivery of nanoscale magnetic particles
IP Soares et al. Application of hyperthermia for cancer treatment: recent patents review
Datta et al. Magnetic nanoparticle-induced hyperthermia with appropriate payloads: Paul Ehrlich’s “magic (nano) bullet” for cancer theranostics?
Mornet et al. Magnetic nanoparticle design for medical diagnosis and therapy
Natarajan et al. NanoFerrite particle based radioimmunonanoparticles: binding affinity and in vivo pharmacokinetics
US7074175B2 (en) Thermotherapy via targeted delivery of nanoscale magnetic particles
Li et al. Research perspectives: gold nanoparticles in cancer theranostics
US20060246143A1 (en) Targeted therapy via targeted delivery of energy susceptible nanoscale magnetic particles
US20050271745A1 (en) Magnetic nanoparticle compositions, and methods related thereto
Nikiforov et al. Biomedical applications of magnetic nanoparticles
Chatterjee et al. Convergence of nanotechnology with radiation therapy—insights and implications for clinical translation
Kut et al. Preliminary study of injury from heating systemically delivered, nontargeted dextran–superparamagnetic iron oxide nanoparticles in mice
US20040156846A1 (en) Therapy via targeted delivery of nanoscale particles using L6 antibodies
US20040156852A1 (en) Therapy via targeted delivery of nanoscale particles
EP3727579B1 (en) Heatable implant device for tumor treatment
Zolata et al. Triple therapy of HER2+ cancer using radiolabeled multifunctional iron oxide nanoparticles and alternating magnetic field
Sharma et al. Temperature and magnetic resonance characteristics of zinc, manganese, gadolinium, gold, iron magnetic nanoparticles and cytokine synergy in hyperthermia
Joerg et al. Nanoparticle Thermotherapy: A New Approach in Cancer Therapy
Class et al. Patent application title: Process for Making Iron Oxide Nanoparticle Preparations for Cancer Hyperthermia Inventors: Robert Ivkov (Ellicott City, MD, US) Yit Wooi Goh (Fairfield, Vic, AU) Meng Tack Ng (Singapore, SG) Zhigang Shen (Singapore, SG) Sung Lai Jimmy Yun (Singapore, SG) Assignees: NANOMATERIALS TECHNOLOGY PTE LTD THE JOHNS HOPKINS UNIVERSITY
Ivkov et al. Development of antibody directed nanoparticles for cancer therapy
Etheridge et al. Andreas Jordan

Legal Events

Date Code Title Description
EEER Examination request
FZDE Dead

Effective date: 20140925