WO2011022350A1 - Nanoparticules de zinc pour le traitement d’infections et du cancer - Google Patents

Nanoparticules de zinc pour le traitement d’infections et du cancer Download PDF

Info

Publication number
WO2011022350A1
WO2011022350A1 PCT/US2010/045688 US2010045688W WO2011022350A1 WO 2011022350 A1 WO2011022350 A1 WO 2011022350A1 US 2010045688 W US2010045688 W US 2010045688W WO 2011022350 A1 WO2011022350 A1 WO 2011022350A1
Authority
WO
WIPO (PCT)
Prior art keywords
zinc
nanoparticle
cancer
subject
tumor
Prior art date
Application number
PCT/US2010/045688
Other languages
English (en)
Inventor
Maulik R. Shah
Mary K. Hoyer
Claudette Klein
Joseph Baldassare
Original Assignee
Saint Louis Unversity
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 Saint Louis Unversity filed Critical Saint Louis Unversity
Publication of WO2011022350A1 publication Critical patent/WO2011022350A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N59/00Biocides, pest repellants or attractants, or plant growth regulators containing elements or inorganic compounds
    • A01N59/16Heavy metals; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/30Zinc; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5146Organic macromolecular compounds; Dendrimers obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyamines, polyanhydrides

Definitions

  • the present invention relates generally to the fields of oncology, infectious disease and medicine. More particularly, it provides novel pharmaceutical formulations of zinc nanoparticles for the treatment of cancer and infections.
  • Zn is an essential nutrient and is present throughout the body. It is required as either a catalytic, co-catalytic or structural component for a large number of enzymes and, as such, contributes to a wide variety of important biological processes including gene expression, replication, hormonal storage and release, neurotransmission, and memory.
  • Zinc has also been shown to impact the invasive capabilities of cancer cells. Zinc ions had the ability to suppress invasion of cancer cells in Matrigel. In studies, evaluating intracellular zinc levels within SCCHN, it was shown that tumor levels of zinc were associated with outcome. Higher zinc levels resulted in greater survival presumably by inhibition of tumor metastasis.
  • Zinc treatment has long been recognized to inhibit tumor development and growth and the reverse, zinc deficiency has been associated with the development of many tumor types.
  • An early 1918 version of the Merck Manual lists various zinc salts as treatment methods for different cancers. Zinc metal readily dissolves into zinc salts in acid conditions. Zinc salts have long been recognized as an astringent and are used as a fixative for skin lesions in Moh's surgery. This astringency effect is modulated by extracellular zinc. Intracellular zinc accumulation has been reported by numerous groups to be cytotoxic in a wide number of cell types. This effect is further corroborated in the inventors' studies using pyrithione, a zinc ionophore.
  • Zinc-oxide nanoparticles have been shown to accumulate in both endosomes and lysozomes depending on the cell type and extracellular conditions
  • nosocomial infections are caused by the contamination of medical devices resulting in serious hospital-acquired infections.
  • Nosocomial pneumonias are the second most common nosocomial infections, and are associated with the highest attributable mortality and morbidity.
  • Recent data have shown that at least 300,000 episodes of nosocomial pneumonia occur annually in the United States (Official Statement, American Thoracic Society). The attributable mortality of this infection is 33-50%, hence, around 100,000 patients die annually because of nosocomial pneumonia (CDC, 1993; Leu et al., 1989).
  • the risk of nosocomial pneumonia increases 6- to 20-fold from the use of mechanical ventilation (Official Statement, American Thoracic Society).
  • Antibiotics and antiseptics have been used to impregnate vascular catheters.
  • the concern with the use of antibiotics has been that resistance might develop to antibiotics, preventing their use therapeutically and systemically in hospitalized patients.
  • the durability of the existing antiseptics has been limited. What is needed is an effective antiseptic having broad spectrum activity against resistant staphylococci, vancomycin-resistant enterococci, resistant Pseudomonas aeruginosa and Candida species, to be used in conjunction with indwelling devices that will inhibit or prevent the nosocomial infections typically associated with the use of these indwelling devices.
  • Nanocomposites with 20% and 30% by weight of ZnO nanoparticles in TiO 2 solgel matrix inhibited 40 to 95% of both antibacterial proliferation from different batch of nanocomposite products. Both nanocomposites selectively inhibited E. coli compared with S. aureus. A clear dose- dependent response between TiO 2 -ZnO20 and TiO 2 -ZnO30 was recorded in S. aureus assay.
  • a zinc nanoparticle composition comprising (a) a zinc core substantially free of other metals or metal oxides; and (b) a non-zinc shell or coating.
  • the zinc nanoparticle may be dispersed in a pharmaceutically acceptable buffer, carrier or diluent.
  • the non-zinc shell may comprise a material permitting conjugation of an agent to said zinc nanoparticle, such as a thiolic acid.
  • the zinc nanoparticle may further comprise an agent conjugated to said zinc nanoparticle.
  • the agent may be a cell or tissue targeting agent, a diagnostic agent, or a therapeutic agent.
  • the non-zinc shell or coating comprises a biologically inert gel, polymer or matrix.
  • the zinc nanoparticle may be between about 5 nm and 100 nm, or between 10 nm and 50 nm.
  • thre is provided a method of treating a subject with a solid tumor comprising administering to said subject a zinc nanoparticle composition comprising (a) a zinc core substantially free of other metals or metal oxides; and (b) a non-zinc shell or coating.
  • the solid tumor may be prostate tumor, basal cell carcinoma tumor, squamous cell carcinoma tumor, or melanoma tumor.
  • the subject may be a human, a non-human primate, a dog, a cat, a horse, a cow, a goat, a rat, a pig, a rabbit, or a mouse.
  • the administering may comprise systemic administration, intravenous injection, or loco-regional administration, such as intratumoral injection, injection into tumor vasculature, or injection regional to said tumor.
  • the method may further comprise administering to said subject a second anticancer therapy, such as radiotherapy, chemotherapy, immunotherapy, gene therapy, hormone therapy, and antibiotic therapy.
  • the cancer may be recurrent, metastatic or multi-drug resistant.
  • a method of treating a subject with an infection comprising administering to said subject a zinc nanoparticle composition comprising (a) a zinc core substantially free of other metals or metal oxides; and (b) a non-zinc shell or coating.
  • the infection may be bacterial, viral, fungal or parasitic.
  • the subject may be a human, a non-human primate, a dog, a cat, a horse, a cow, a goat, a rat, a pig, a rabbit, or a mouse.
  • the administering may comprise systemic administration, intravenous injection, or loco-regional administration, such as topical administration or injection regional to said infection.
  • the method may further comprise administering to said subject a second anti-biotic therapy.
  • the infection may be caused by a drug resistant organism.
  • a method of imaging a site in a subject comprising (i) administering to said subject a zinc nanoparticle composition comprising (a) a zinc core substantially free of other metals or metal oxides; and (b) an agent that targets said zinc nanoparticle to said site, and (ii) imaging said zinc nanoparticle in said subject.
  • the zinc nanoparticle may be further comprise a detectable label, such as a fluorescent, chemilluminescent or radiolabel, or may further comprise a therapeutic agent, such as an antibiotic, an anti-fungal, and anti-parasitic, an anti-viral, a chemotherapeutic, or a radiotherapeutic.
  • the subject may have a tumor or an infection.
  • the shell or coating is composed of a polymer.
  • polymeric surfaces include polyvinyl chloride, polyurethane, polyethylene, silastic elastomers, polytetrafluoroethylene, dacron, collodion, carboethane or nylon, esters of polylactide, polyglycolide, polycapro lactone, polyethelyene glycol, polyethylene oxide and all copolymers of the above derivatives.
  • the surface may be composed of silicone or silk.
  • a medical device coated with zinc nanoparticle composition comprising zinc nanoparticles comprising a zinc core substantially free of other metals or metal oxides.
  • the device may be an implanted or indwelling device, such as a catheter, stent, pump, or suture.
  • the zinc nanoparticle may further comprise a non-zinc shell or coating, such as a biologically inert gel, polymer or matrix.
  • the nanoparticle may be between about 5 nm and 100 nm, or between about 10 nm and 50 nm.
  • the non-zinc shell or coating may comprise a material permitting conjugation of a therapeutic agent to said zinc nanoparticle, such as a thiolic acid.
  • the zinc nanoparticles can be use to impregnate an inorganic surface.
  • inorganic surfaces include floors, table-tops, counter-tops, surfaces of a hospital equipment, wheelchair surfaces, etc.
  • Virtually any surface comprising a material that is capable of being coated by, impregnated with, absorbing or otherwise retaining the antiseptic compounds of the invention may be disinfected and/or sterilized using the present antiseptic compounds and their compositions.
  • the antiseptic compound of the invention can be used to disinfect, sanitize and sterilize a wide variety of surfaces.
  • the invention also provides medical devices coated with zinc nanoparticles.
  • Examples of medical devices include endotracheal tubes, a vascular catheter, an urinary catheter, a nephrostomy tube, a biliary stent, a peritoneal catheter, an epidural catheter, a central nervous system catheter, an orthopedic device, a prosthetic valve, and a medical implant.
  • the vascular catheter may be a central venous catheter, an arterial line, an pulmonary artery catheter, and a peripheral venous catheter.
  • the central nervous system catheter may be an intraventricular shunt.
  • Other medical devices that can benefit from the present invention include blood exchanging devices, vascular access ports, cardiovascular catheters, extracorpeal circuits, stents, implantable prostheses, vascular grafts, pumps, heart valves, and cardiovascular sutures, to name a few. Regardless of detailed embodiments, applicability of the invention should not be considered limited with respect to the type of medical device, implant location or materials of construction of the device.
  • the invention also provides methods for coating a medical device with a zinc nanoparticles comprising a) immersing a medical device in a solvent comprising a zinc nanoparticle; b) drying the device; and c) washing off excessive composition.
  • the solvent used to immerse the device can be methylene chloride, methanol, or a combination thereof.
  • the invention also provides methods for preventing nosocomial infections in a subject comprising coating a medical device that the subject is required to use with a composition comprising zinc nanoparticles.
  • the subject can be human or an animal model.
  • the type of nosocomial infection that can be prevented by the methods of this invention include, but are not limited to, pneumonia, bacteremia, fungimia, candidemia, a urinary tract infection, a catheter-exit site infection, and a surgical wound infection.
  • the nosocomial infections that can be prevented may be caused by bacteria.
  • the bacteria are drug resistant bacteria.
  • drug resistant bacteria include methicillin-resistant staphylococci, vancomycin-resistant enterococci, and resistant Pseudomonas aeruginosa.
  • the nosocomial infection may be caused by a fungus.
  • the fungal agent is a drug resistant fungi.
  • Examples of a drug resistant fungi include members of the Candida species.
  • Other pathogenic organisms that can cause the nosocomial infections are cited elsewhere in this specification and coating devices and surfaces with the antiseptics of the present invention can prevent infections by these organisms as well.
  • the compositions of the present invention have broad uses including use in healthcare by providing sterile medical devices and surface sterilization and decontamination.
  • FIG. 1 Electron microscopy of zinc nanoparticles (ZNP) of approximately 100 nm size. Courtesy of Hefei Kaier Nanotech. Particles are of various different shapes and are electrodense.
  • ZNP zinc nanoparticles
  • FIG. 2 Appearance of ZNP in solution.
  • ZNP exist as a black/grayish powder in Solution, a optically dense gray solution forms. ZNPs aggregate in the absence of surfactant.
  • FIG. 3 Solubility of ZNP in different solutions.
  • ZNP are highly soluble in aqueous solutions. In all cases, dissolution of ZNP to zinc ions occurs within the first 10 min. ZNP dissolution is dependent on zinc supersaturation of the solution. Provided ample, volume, complete dissolution of ZNPs occurs. ZNPs are highly reactive to protein. A slight delay in ZNP dissolution is seen in solutions with large amounts of proteins such as media or serum.
  • FIG. 4 Solubility of ZNP in BioGel formulations of Pluronic.
  • Pluronic is a block co-polymer that shields the ZNP from dissolution in aqueous environments.
  • ZNPs (10 mg/ml) were formulated in different percentages w/v of Pluronic gel. 100 ⁇ l of gel-ZNP was added to 5 ml of media and zinc was measured by TSQ fluorescence over time. ZNP dissolution was dependent on the shielding ability of the block co-polymer.
  • FIG. 5 Cytotoxicity of ZNPs against cancer cells.
  • ZNPs were added to 70% logarithmic growth cultures of PC3 (prostate cancer), CAL27 (Squamous Cell Carcinoma of the Head and Neck), and SKO V3 (Ovarian Cancer). Cancer cell viability was measured by MTT assay at various time points.
  • FIG. 6 Dose dependent ZNP cytotoxicity against cancer cell lines. ZNPs were added to 70% logarithmic growth cell lines in culture. Cell viability was measured by MTT assay at 18 hrs. There is a threshold phenomenon. At low doses, no cytotoxicity is seen. After a threshold dose, significant cell death is observed.
  • FIG. 7 - Synergy of ZNP with cancer chemotherapeutics.
  • ZNPs were added to PC3 cells either alone (LD50) dose or in combination with the reported LD50 of the cancer chemotherapeutic drug on the X-axis.
  • ZNPs potentiate the cytotoxic effects of chemotherapeutics in a number of different classes.
  • FIG. 8 Efficacy of ZNPs against subcutaneous melanomas.
  • Subcutaneous B16 melanomas were grown on the dorsum of immunocompetent C57B/6 mice. When tumors reached approximately 150 mm 3 , they were directly injected with 100 ⁇ l volume of vehicle (PBS), 30% Pluronic, ZNPs (10 mg/ml in PBS), or 30%
  • Pluronic with 10 mg/ml ZNPs Tumor volume was determined by digital caliper measurement. Pluronic groups show a rapid increase in volume most likely due to the gel itself rather than tumor growth. This is supported by the initial increase in volume by Day 1 followed by a decline by Day 3. Non-Pluronic groups do not show a rapid increase in tumor size. ZNPs did not show anti-tumor effect likely due to rapid dissolution of the ZNPs. In contrast, Pluronic-ZNPs showed a significant decline in tumor size over time.
  • FIG. 9 Survival of animals with melanoma.
  • Subcutaneous B16 melanomas were grown on the dorsum of immunocompetent C57B/6 mice. When tumors reached approximately 150 mm 3 , they were directly injected with 100 ⁇ l volume of vehicle (PBS), 30% Pluronic, ZNPs (10 mg/ml in PBS), or 30% Pluronic with 10 mg/ml ZNPs.
  • FIG. 10 Cytotoxicity of Her2/Neu AB-conjugated ZNPs.
  • Cell lines were plated in 6 well-plates at 70% confluence and treated as given on the X-axis.
  • the Her2 AB concentration was 60 ⁇ g/ml.
  • ZNP were added at 10 ⁇ g/ml and Her2-
  • ZNP ZNP at 5 ⁇ g/ml. Cells were washed 1 hr after addition of treatment and then MTT assay performed at 18 hrs. Her2-conjugated ZNP show highly specific killing of Her2-expressing cells (SKOV3 and SKBR3) but not the Cal27 (Her2 non- expressing). Results demonstrate ZNP cytotoxicity can be controlled through the use of a targeting ligand. Cell killing was dose dependent.
  • FIG. 11 Intracellular zinc ion concentration (Mean Fluorescent Intensity) levels are dependent on ZNP targeting.
  • Mean Fluorescent Intensity (MFI, y-axis) representing intracellular zinc ion concentration was measured by FACS on cell lines expressing HER2 (SKBR3 and SKOV3) or expressing high levels of transferrin receptor (PC3) after incubation with lmg of the designated treatment: Control - no treatment; ZNP - unconjugated ZNP; ZNP-Her2 - Anti-Her2 conjugated ZNP; ZNP-TNF - transferrin conjugated ZNP.
  • FIG. 12 Intracellular zinc ion concentrations rise over time and are directly proportional to cell death.
  • Mean Fluoresent Intensity was measured by FACS after staining cells with zinc ion specific fluorescent probes. MFI represents total histogram fluorescence of gated fluorescent cells.
  • SKBR3 cells were analyzed forintracellular zinc ion concentration.
  • Control (diamond) represent non-treated cells.
  • ZNP (square) - cells treated with unconjugated ZNP
  • ZNP-Her2 diamond
  • ZNP-TNF cross marks
  • FIG. 13 - Efficacy of conjugated ZNPs in killing cells is dose dependent.
  • Cell viability (y-axis) was measured by MTT assay after 18 hours of incubation of the various cell lines (legend) with the treatment group (x-axis) represented.
  • FIG. 14 - Efficacy of conjugated ZNPs in killing cells is dose dependent.
  • Cell viability of SKBR3 cells was measured by MTT assay after incubation with treatment group (legend) at the dosage indicated (x-axis). Only Her2 specific killing is observed in a linear dose response.
  • FIG. 15 - Release of zinc ions extracellularly is dependent on the hydrophobicity of the outer polymer coating.
  • ZNPs were coated with PEG polymers of various chain lengths (legend). The greater the chain length, the higher the hydrophobicity.
  • ZNPs were placed in fetal calf serum and zinc ion concentration measured over time. Concentration adjusted for baseline serum zinc levels. Kinetics of zinc release could be altered and delayed based on the hydrophobicity of the polymer coating. DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS
  • the present invention provides novel pharmaceutical formulations comprising zinc nanoparticles having a core comprised of zinc and essentially free of other metals or metal oxides.
  • the nanoparticles may advantageously comprise an outer shell or coating.
  • This shell or coating can serve one or more purposes, namely, to provide a material to which ancillary agents (targeting, diagnostic, therapeutic) can be attached, and/or to sheild the zinc from the environment and control the release of zinc ions.
  • zinc nanoparticle cytotoxicity was dose-dependent based on the amount of zinc nanoparticles added.
  • the dose range was variable depending on the susceptibility of the cell line to zinc ion toxicity and the volume in which the zinc nanoparticles were dissolved to achieve threshold doses.
  • zinc nanoparticles did not need direct contact with cancer cells to exert cytotoxic effect.
  • the cytotoxic effect was distance dependent from the area of zinc nanoparticle deposition. If threshold zinc ion levels were reached based upon zinc nanoparticle dissolution then all cells showed cytotoxic effect.
  • zinc nanoparticle mediated cell death was directly dependent on contact of the nanoparticle with the cell.
  • Zinc nanoparticles showed no specificity of cell killing based on cellular genetic alterations or rate of growth, nor was there species specificity.
  • the inventors believe that there are two mechanism of cytotoxic action for zinc nanoparticles. The first is through internalization into various cellular vacuoles and release of zinc ions resulting in disruption of cellular homeostasis. The second is through slow dissolution in aqueous solution resulting in release of zinc ions increasing extracellular concentrations to a level that either forces internalization or disrupts cell growth.
  • studies with zinc nanoparticles revealed that there is a dose dependent release of zinc ions into aqueous solutions.
  • Zinc ion release could be impacted by coating of zinc nanoparticles with thiolic acids or encapsulation in bio inert gels.
  • Use of bioinert gels reveals that zinc nanoparticle toxicity can be limited to a surface area consistent with the gel.
  • Pathogens often attach to and proliferate in such devices and eventually invade the patient leading to nosocomial infections.
  • Microorganisms usually migrate along the surfaces of devices to invade sterile environments, such as the bronchoalveolar space leading to pneumonia, the bloodstream leading to bacteremia, or the urinary bladder leading to urinary tract infections.
  • the zinc nanoparticles of the present invention will be also find use in diagnostic contexts by including a cell, tissue or organ targeting moeity.
  • zinc nanoparticles can imaged based on their autofluorescent properties, and this can be enhanced by the addition of other detectable labels.
  • therapeutic agents can be added to achieve simultaneous imaging and therapy.
  • Zinc nanoparticles or Zinc NanoDots discussed herein are comprised of a core of pure metallic zinc. They are spherical or multi- faceted particles of elemental zinc wit high surface area. ZNP of 20-40 nm have a surface area of 30-50 m 2 /g and
  • Quantum dots 100 nm particles have a surface area of 7 m 2 /g.
  • ZNP and Quantum dots or other therapeutic nanoparticles are purely aggregates of metallic zinc sized in the nano-range. They do not exhibit semiconductor like properties similar to quantum dots.
  • Another important distinction between zinc nanoparticles and quantum dots which also contain zinc-sulfide caps around a metallic core is the difference in optical properties. Quantum dots exhibit fluorescent properties across a spectrum depending on energy absorption. The color range exhibited is dependent on both size and shape. In contrast, zinc nanoparticles do not exhibit the optical properties inherent of quantum dots.
  • ZNP are also comprised of a different form of elemental zinc. As such, their
  • CAS No. is 7440-66-6.
  • the appearance of ZNP is black. They have a molecular weight of 65.37, a density of 7140 kg/m 3 and a melting point of 419.53 0 C. The purity is greater than 99%.
  • ZNP with the cytotoxic properties discussed herein can range from 5 nm to
  • ZNPs are shown in FIG. 1.
  • ZNP do not exist in any standard shapes. The inventors believe that the shape may be varied to optimize binding of targeting ligands and to impact rate of dissolution. The optimal shape for targeted ZNP is likely to be spheroids. ZNPs are shown in FIG. 1.
  • ZNP described herein comprise of pure metallic zinc. As such they freely dissolve in aqueous solution until there is super-saturation of zinc ions. The rate of dissolution is dependent on the aqueous solution, pH, and the saturation of zinc ions.
  • Rate of dissolution can also be impacted by the coatings applied to ZNP (Described later).
  • the solubility of ZNP in aqueous solutions distinguishes them from other zinc based nanoparticles such as zinc-oxide.
  • Zinc-oxide nanoparticles have limited solubility in aqueous solutions.
  • the rate of zinc ion dissolution is given in FIGS. 3 and 4.
  • ZNP are highly reactive with proteins.
  • a number of ligands can be bound to the surface of ZNP including antibodies, peptides, proteins, aptamers, viral particles and nucleic acids.
  • the inventors have conjugated antibodies and proteins to the surface of ZNPs. Efficiency of binding is dependent on size and shape of ZNPs.
  • ZNPs exhibit cytotoxicity against all organisms using nucleic acids for replication. It has cytotoxic properties against enveloped and non-enveloped viruses, bacteria, protozoa, molds and human cells including cancer cells. The mechanism of
  • ZNP cell death varies depending on the organism but includes apoptosis, necrosis and inhibition of cell replication. Cytotoxic activity has been demonstrated against the cell types shown in Table 1. Cytotoxicity is independent of the underlying molecular pathway of transformation. Cell death and kinetics of cell death varied depending on the cancer cell type. The LD 5O of each cell type is given in Table 1. Importantly, the rate of cell death was rapid for each cell line starting in as little as 2 hrs (FIG. 5). Dose response curves show sigmoid kinetics (FIG. 6). The linear range for cytotoxic activity depended on the rate of zinc ion dissolution which in term varied by characteristics of the solution and ZNP size and weight.
  • ZNPs are believed to act synergistically with other pharmaceutical preparations with cytotoxic or cytostatic activity. ZNPs were combined with a number of chemotherapeutic drugs and showed either additive or potentiated cytotoxic cell killing (FIG. 7). The inventors have tested ZNPs with cancer chemotherapeutics of various different classes including nucleoside inhibitors, alkylating agents and topoisomerase inhibitors.
  • ZNP cytotoxic activity is associated with internalization of ZNP within cells and subsequent dissolution and zinc ion release or zinc ion release exterior to cells with subsequent zinc ion internalization.
  • a RNA microarray analysis of zinc ion toxicity upon internalization into cells shows that the mechanism of cytotoxic activity is likely to be disruption of proteins with zinc ion motifs. Since zinc ion motifs are rich in proteins with nucleic acid binding properties like DNA polymerases, RNA polymerases, and tRNAs, there is disruption of replication, mRNA production and amino acid incorporation into nascent growing proteins.
  • ZNP were administered at 20-25 grams/mouse either subcutaneously, intradermally, or intratumorally. ZNPs were administered as high as 5% w/w without undo toxicity to animals. Histological examination of tissues including, brain, liver, spleen, kidneys, and heart did not show any cellular disruption or change in characteristics. Serum zinc levels increased depending on dose and were rapidly cleared within 24 hours. Maximum serum zinc levels were seen within the first 120 min with steady decline to normal levels between 8 and 24 hrs. Tissue ZNP accumulation was minimal. No tissue ZNP was detected 7 days after administration.
  • the present application relates to zinc nanoparticles previously unknown for their use in pharmaceutical contexts. These particles are unique from other zinc- containing nanoparticles that have been used medicinally in that their cores are substantially free of oxides or any other metals than pure elemental zinc. By substantially free, it is meant that at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, at least 99.9% or 100% of the core is element zinc.
  • nanoparticles are defined as having sizes in the sub-micrometer range.
  • the zinc nanoparticles will range in size from 1 nm to 250 nm, more specifically 5 nm to 100 nm, and even more specifically, 10 nm to 50 nm. Sizes of 5 nm, 10 nm, 15 nm, 20 nm, 25 nm, 30 nm, 35 nm, 40 nm, 45 nm, 50 nm, 60 nm, 70 nm, 80 nm, 90 nm, or 100 nm.
  • the zinc nanoparticles are covered, encapsulated or encased by a shell or coating.
  • the ZNPs can be individually “coated” with a shell or outer layher with a chemical entity to protect against bio-erosion or dissolution, and may optionally contain a targeting ligand (antibody, peptide, nucleic acid).
  • the shell in this embodiment is directly linked chemically to the surface of the ZNP.
  • the ZNPs are suspended in a colloidal (biogel, inert gel) solution or coating.
  • the coating in this embodiment is a gel formulation and is not chemically attached or linked to the ZNPs; the ZNPs are suspended in this material as a colloid.
  • the gel/colloid may be formulated to protect the ZNPs from erosion or dissolution.
  • purpose of the shell or coating may be one or both of the following: (i) to shield the zinc from the immediate environment, and thereby regulate the introduction of zinc ions into the environment; and/or (ii) to provide a material to which other agents, such as targeting, diagnostic and therapeutic agents, can be attached.
  • the shell will be a bioinert gels, which may be synthetic or natural material that is polymeric or carbon-based and either biocompatible or biodegradable.
  • the shell or coating is required to provide slow release of zinc ions through controlled bioerosion.
  • the biocompatible polymers will permit a therapeutic range of zinc of ZNP released within a disease microenviornment. Additionally, the biocompatible polymer will have an appropriate viscosity for localizide delivery into a specific disease compartment.
  • the active agent, ZNP are dispersed in a bioinert polmer. Diffusion through the polymer matrix and release rate is dependent on the choice of polymer. Two possible systems are envisioned. In the first system, polymer bioerosion is through the conversion of the polymer from a water insoluble to a water-soluble state. As such, the polymer surrounding ZNPs is degraded and ZNPs are released. In the second system, polymer chains are attached to ZNPs. The chains are degraded through interaction with water or enzymes resluting in release of ZNPs.
  • Biodegradable polymers Both natural and biosynthic polymers will need to be bio-degradable. Most of the polymers will have hetero-atom-containing polymer backbones. Bioerosion and rate of degredation can be controlled through the use of chemical linkages including anhydrides, esters or amide bonds. Additionaly, the metabolic products of these biocompatible polymers will be bioinert meaning having no or limited toxicity and mechanims for either excretion or degredation. Examples of biodegradable polymers for use with ZNPs include Poly-esters based on polylactide (PLA) polyglycolide (PGA) and polycaprolactone (PCL). Other potential polymers incude modifications of polysaccharides including starch, cellulose, and chitosan.
  • PLA polylactide
  • PGA polyglycolide
  • PCL polycaprolactone
  • ZNPs dispersed in a matrix of poly(ethylene glycol) (PEG) such that water dispersion into the matrix is limited over approximately 8 hours.
  • PEG poly(ethylene glycol)
  • This PEG-ZNP matrix is injected into a disease state (tumor bed). Through slow bioerosion of the polymer, ZNPs are contacted with incoming water. Upon contact with water, ZNPs are activated and can contact tumor cells and be internalized to exert toxicity.
  • PEG poly(ethylene glycol)
  • ZNPs are conjugaged with targeting antibody directed to disease state. Based on the conjugation effeciency of the targeted antibody, the remaining open space on the surface of ZNPs is filled with polymer cross-linkers consisting of PEG and/or fatty acids.
  • the cross-linkers provide a hydrophobic surface to slow bio-erosion.
  • the rate of biodegredation of the ZNPs can be contolled by the chain length and composition of the conjugated polymers.
  • An example of composition would be:
  • Targeting agents in accordance with the present invention, provide for the direction of zinc nanoparticles to various cells, tissues or organs within a subject.
  • Targeting agents may be attached using standard technology to the matrices or polymers of the shell or coating.
  • cross-linking reagents are used to form molecular bridges that tie functional groups of two different molecules.
  • hetero-bifunctional cross-linkers can be used that eliminate unwanted homopolymer formation. Table 2 illustrates several cross-linkers.
  • An exemplary hetero-bifunctional cross-linker contains two reactive groups: one reacting with primary amine group (e.g., N-hydroxy succinimide) and the other reacting with a thiol group (e.g., pyridyl disulfide, maleimides, halogens, etc.).
  • primary amine group e.g., N-hydroxy succinimide
  • a thiol group e.g., pyridyl disulfide, maleimides, halogens, etc.
  • the cross-linker may react with the lysine residue(s) of one protein (e.g., the selected antibody or fragment) and through the thiol reactive group, the cross-linker, already tied up to the first protein, reacts with the cysteine residue (free sulfhydryl group) of the other protein (e.g., the selective agent).
  • Linkers that contain a disulfide bond that is sterically hindered may prove to give greater stability in vivo, preventing release of the targeting peptide prior to reaching the site of action. These linkers are thus one group of linking agents.
  • SMPT cross-linking reagent
  • Another cross-linking reagent is SMPT, which is a bifunctional cross-linker containing a disulfide bond that is "sterically hindered" by an adjacent benzene ring and methyl groups. It is believed that steric hindrance of the disulfide bond serves a function of protecting the bond from attack by thiolate anions such as glutathione which can be present in tissues and blood, and thereby help in preventing decoupling of the conjugate prior to the delivery of the attached agent to the target site.
  • thiolate anions such as glutathione which can be present in tissues and blood
  • the SMPT cross-linking reagent lends the ability to cross-link functional groups such as the SH of cysteine or primary amines (e.g., the epsilon amino group of lysine).
  • Another possible type of cross-linker includes the hetero-bifunctional photoreactive phenylazides containing a cleavable disulfide bond such as sulfosuccinimidyl-2-(p-azido salicylamido) ethyl- l,3'-dithiopropionate.
  • the N-hydroxy-succinimidyl group reacts with primary amino groups and the phenylazide (upon photolysis) reacts non-selectively with any amino acid residue.
  • non-hindered linkers also can be employed in accordance herewith.
  • Other useful cross-linkers include SATA, SPDP and 2-iminothiolane (Wawrzynczak & Thorpe, 1987). The use of such cross-linkers is well understood in the art. Another embodiment involves the use of flexible linkers.
  • U.S. Patent 4,680,3308 describes bifunctional linkers useful for producing conjugates of ligands with amine-containing polymers and/or proteins, especially for forming antibody conjugates with chelators, drugs, enzymes, detectable labels and the like.
  • U.S. Patents 5,141,648 and 5,563,250 disclose cleavable conjugates containing a labile bond that is cleavable under a variety of mild conditions. This linker is particularly useful in that the agent of interest may be bonded directly to the linker, with cleavage resulting in release of the active agent.
  • U.S. Patent 5,856,456 provides peptide linkers for use in connecting polypeptide constituents to make fusion proteins, e.g., single chain antibodies.
  • the linker is up to about 50 amino acids in length, contains at least one occurrence of a charged amino acid (preferably arginine or lysine) followed by a proline, and is characterized by greater stability and reduced aggregation.
  • U.S. Patent 5,880,270 discloses aminooxy-containing linkers useful in a variety of immunodiagnostic and separative techniques.
  • the targeting agent may be an antibody, or antigen- binding fragment thereof.
  • the antibody may be directed to a tumor cell surface antigen, or to an antigen expressed on the surface of a pathogen.
  • the antibody may be bivalent or single chain (sc), or an Ab fragment, such as a Fab, an Fv, or scFv.
  • Bevacizumab Avastin®; Genentech/Roche
  • Trastuzumab Herceptin®; Genentech/Roche
  • Omnitarg Geneentech/Roche
  • Cetuximab Erbitux®; Lilly/Imclone
  • Imatinib Gleevec®; Novartis
  • Panitumumab Vectibix®; Amgen
  • Nimotuzumab YM Biosciences
  • Matuzumab Merck KGaA
  • Zalutumumab HumanMax -EGFR; Genmab
  • Another targeting agent is a ligand for a cell surface receptor.
  • ligands often are modified such that they do not activate their cognate receptor, i.e., a receptor-binding peptide.
  • Cancer cell markers suitable as targets for a target-directed zinc nanoparticle therapy include include urinary tumor associated antigen (UTAA), fetal antigen, tyrosinase (p97), gp68, TAG-72, HMFG, Sialyl Lewis Antigen, MucA, MucB, PLAP, estrogen receptor, laminin receptor, erb B and pl55, PSA, CEA, MART, MAGEl, MAGE3, gplOO, BAGE, GAGE, TRP-I, TRP-2, PMSA, Mycobaterium tuberculosis soluble factor (Mtb), phenol soluble modulin (PSM), CMV-G, CMV-M, EBV capsid- EB nuclear antigen (EBNA), gpl20, gp41, tat, rev, gag, toxa antigen, rubella antigen, mumps antigen, ⁇ -fetoprotein (AFP), adenocarcinoma antigen (ART -4), CAMEL
  • Bacterial cell markers suitable as targets for zinc nanoparticles include bacterial outer membrane constituents, such as liposaccharide (LPS) and olicosaccharide cell surfaces, bacterial cell wall components, such as peptidoglycan (NAM, NAG, DAP), cell enevelope, components, such as LPS and techioic acid, and outer envelope structures, including the pilus and fimbria.
  • Viral markers suitable as targets for zinc nanoparticles include viral envelope components, including HIV p24, and viral core proteins, such as HCV NS3.
  • Fungal markers suitable as targets for zinc nanoparticles include fungal cell wall and fungal hyphae structures. Parasitic markers als are contemplated as suitable targets for zinc nanoparticles.
  • Zinc nanoparticles can be produced using a number of different processes.
  • zinc as a mined element exists as pure elemental zinc and needs to be rendered to nanoscale size. This size fractionation is performed by either milling (using very fine instruments) or through laser evaporation followed by condensation on a solid surface or substrate.
  • a second class of method is a "bottom up" approach where the ZNPs are synthesized from zinc ions existing in either gas phase or solution. This methodology is more common for zinc oxide rather than ZNP, but is reported for both. In this methodology, an aqueous solution containing zinc ions is evaporated or otherwise treated to precipitate atomic zinc as a condensate.
  • the initial starting "solution” may be zinc ions in some liquid substrate or may be zinc ions in gas phase.
  • Zinc nanoparticles can be obtained from commercial sources such as Sigma- Aldrich, American Elements, Xuzhou Hongwu Nanometer Material Company and NaBond.
  • product 578002-5G from Sigma-Aldrich is a 5 gram quantity of zinc nanopowder having the properties shown in Table 3.
  • Table 3 TABLE 3 - Properties of Zinc Nanoparticles vapor pressure 1 mm Hg ( 487 0 C)
  • zinc nanoparticle compositions disclosed herein may alternatively be administered parenterally, intravenously, intradermally, intramuscularly, transdermally or even intraperitoneally as described in U.S. Patent 5,543,158; U.S. Patent 5,641,515 and U.S. Patent 5,399,363 (each specifically incorporated herein by reference in its entirety).
  • Solutions of the active compounds as free base or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (U.S. Patent 5,466,468, specifically incorporated herein by reference in its entirety). In all cases the form must be sterile and must be fluid to the extent that easy syringability exists.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils.
  • polyol e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • suitable mixtures thereof e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • vegetable oils e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • sterile aqueous media that can be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermolysis fluid or injected at the proposed site of infusion, (see for example, "Remington's Pharmaceutical Sciences” 15th Edition, pages 1035-1038 and 1570-1580).
  • Some variation in dosage will necessarily occur depending on the condition of the subject being treated.
  • the person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
  • preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biologies standards.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile- filtered solution thereof.
  • the compositions disclosed herein may be formulated in a neutral or salt form.
  • Pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the formulations are easily administered in a variety of dosage forms such as injectable solutions, drug release capsules and the like.
  • carrier includes any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like.
  • carrier includes any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like.
  • the use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
  • phrases "pharmaceutically-acceptable” or “pharmacologically-acceptable” refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to a human.
  • the preparation of an aqueous composition that contains a protein as an active ingredient is well understood in the art.
  • such compositions are prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid prior to injection can also be prepared.
  • the zinc nanoparticles of the present invention find particular use in the treatment of cancer.
  • the tumor will be a solid tumor, thereby permitting targeting of the treatment to a particular site within a subject, all cancers should be amenable to treatment with zinc nanoparticles given the non-specific nature of their toxicity.
  • all cancers should be amenable to treatment with zinc nanoparticles given the non-specific nature of their toxicity.
  • Cancers and pre-cancerous conditions which can be prevented and/or treated by the methods described herein include, but are not limited to: adeno fibroma; adenoma; agnogenic myeloid metaplasia; AIDS-related malignancies; ameloblastoma; anal cancer; angiofollicular mediastinal lymph node hyperplasia; angiokeratoma; angiolymphoid hyperplasia with eosinophilia; angiomatosis; anhidrotic ectodermal dysplasia; anterofacial dysplasia; apocrine metaplasia; apudoma; asphyxiating thoracic dysplasia; Astrocytoma (including, for example, cerebellar and cerebral); atriodigital dysplasia; atypical melanocytic hyperplasia; atypical metaplasia; autoparenchymatous metaplasia; basal cell hyperplasia; benign giant lymph no
  • the zinc nanoparticles of the present invention are themselves toxic to cancer cells, it may prove useful to add a second therapeutic agent or "payload" to the nanoparticle.
  • agents including chemotherapeutic and radiotherapeutics are listed below.
  • Others include cytokines, toxins (ricin, pertussis toxin, cholera toxin) and hormones.
  • the present invention is contemplated to be used in both systemic and localized administration. With respect to systemic administration, it will be desirable to target the zinc nanoparticles to locations within the body using targeting or "homing" molecules, such as antibodies or other ligands for cell surface receptors.
  • targeting or "homing" molecules such as antibodies or other ligands for cell surface receptors.
  • compositions according to the present invention may be via any common route so long as the target tissue is available via that route. This includes oral, nasal, buccal, rectal, vaginal or topical. Alternatively, administration may be by intradermal, subcutaneous, intramuscular, intraperitoneal or intravenous injection. Such compositions would normally be administered as pharmaceutically acceptable compositions, described supra.
  • Injection of zinc nanoparticle may be by syringe or any other method used for injection of a solution, as long as the agent can pass through the particular gauge of needle required for injection.
  • a needleless injection system has been described (U.S. Patent 5,846,233) having a nozzle defining an ampule chamber for holding the solution and an energy device for pushing the solution out of the nozzle to the site of delivery.
  • a syringe system has also been described for use in gene therapy that permits multiple injections of predetermined quantities of a solution precisely at any depth (U.S. Patent
  • an "anti-cancer” therapy is capable of negatively affecting cancer in a subject, for example, by killing cancer cells, inducing apoptosis in cancer cells, reducing the growth rate of cancer cells, reducing the incidence or number of metastases, reducing tumor size, inhibiting tumor growth, reducing the blood supply to a tumor or cancer cells, promoting an immune response against cancer cells or a tumor, preventing or inhibiting the progression of cancer, or increasing the lifespan of a subject with cancer.
  • Anti-cancer therapies are described below as "standard of care” therapies and include biological agents (biotherapy), chemotherapy agents (including anti-tumor antibiotics), and radiotherapy agents, surgery and immunotherapy.
  • these therapies would be provided in a combined amount to achieve a clinically significant effect.
  • This process may involve contacting the cancer cell with the therapies at the same time, such as by administering a single composition or treatment that includes both agents, or by administering two distinct compositions or treatments at the same time.
  • the zinc nanoparticle therapy may precede or follow the other treatment by intervals ranging from minutes to weeks.
  • the other agent and zinc nanoparticle therapy are applied separately, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the therapy and zinc nanoparticle therapy would still be able to exert an advantageously combined effect.
  • zinc nanoparticle therapy is "A” and the secondary therapy is "B":
  • treatment cycles would be repeated as necessary, including two, three, four, five, six, seven, eight, nine, ten or more cycles.
  • a second anti-cancer therapy is radiotherapy.
  • Radiotherapy includes, for example, fractionated radiotherapy, nonfractionated radiotherapy and hyperfractionated radiotherapy, and combination radiation and chemotherapy.
  • Types of radiation also include ionizing (gamma) radiation, particle radiation, low energy transmission (LET), high energy transmission (HET), ultraviolet radiation, infrared radiation, visible light, and photosensitizing radiation.
  • a second anti-cancer therapy is chemotherapy.
  • the chemotherapy may comprise administration of one or more of: 20-epi-l,25 dihydroxyvitamin D3; (1 aS,8S,8aR,8bS)-6-amino-8-(((aminocarbonyl)oxy)methyl)-l,la, 2,8,8a, 8b-hexahydro- 8a-methoxy-5-methylazirino(2',3':3,4)pyrrolo[l,2-a]in-dole-4,7-dione; (8S-cis)-10- ((3-amino-2, 3,6-trideoxy-alpha-L-lyxo-hexopyranosyl)oxy)-7, 8,-9,1, 0-tetrahydro- 6,8,1 l-trihydroxy-8-(hydroxyacetyl)-l-methoxy-5; (I)-mimosine; l-(2-chloroe
  • M3 carmustine (BCNU or BiCNU); CARN 700; carubicin; carubicin hydrochloride; carzelesin; casein kinase inhibitors (ICOS); castanospermine; CCNU; cecropin B; cedefmgol; celecoxib; cetrorelix; cetuximab; chlorambucil (leukeran); chlorins; chloroethyl nitrosoureas; chloroquinoxaline sulfonamide; chlorotrianisene; CHOP
  • CPT-I l CPT-I l; crambescidin 816; crisnatol; crisnatol mesylate; cryptophycin 8; cryptophycin A derivatives; curacin A; cycloleucine; cyclopentanthraquinones; cyclophosphamide; cyclo-phosphamide; cyclophosphamide (Cytoxan); cyclophosphamide anhydrous; cycloplatam; cypemycin; cytarabine; cytarabine HCl
  • cytosar-u cytarabine ocfosfate; cytochalasin B; cytolytic factor; cytosine arabinoside; cytostatin; dacarbazine; dacliximab; dactinomycin (cosmegen); daunomycin; daunorubicin; Daunorubincin HCl (cerubidine); decarbazine (DTIC- dome); decitabine; dehydrodidemnin B; demecolcine; depsipeptide; deslorelin; dexamethasone; dexormaplatin; dexverapamil; dezaguanine; dezaguanine mesylate; dianhydrogalactitol; diarizidinylspermine; diaziquone; diazooxonorleucine; dibromodulcitol; dibrospidium chloride; dicarbazine; didemnin B; didox; diethylnorspermine; die
  • fludara fludarabine phosphate; fluorocitabine; fluorodaunorunicin hydrochloride; fluoxymesterone (halotestin); flutamide; flutamide (eulexin); fluxuridine; forfenimex; formestane; fosquidone; fostriecin; fostriecin sodium; fotemustine; fulvestrant; gadolinium texaphyrin; galarubicin; gallium nitrate; gallium nitrate (granite); galocitabine; ganirelix; gef ⁇ tinib; gelatinase inhibitors; gemcitabine; gemcitabine
  • gemzar gemcitabine hydrochloride; gemicitabine; gemtuzumab; genistein; glufosfamide; glutamic acid; glutathione inhibitors; goserelin (zoladex); GPXlOO; gramicidin D; hepsulfam; heptaplatin; heregulin; hexamethylene bisacetamide; hexestrol; human chorionic gonadotrophin; hydroxyurea; hydroxyurea (hydra); hypericin; ibandronic acid; ibritumomab; idarubicin; idarubicin (idamycin); idarubicin hydrochloride; idoxifene; idramantone; ifosfagemcitabine; ifosfamide; ifosfamide
  • iflex ifosfamide with mesna (MAID); ilmofosine; ilomastat; imatinib mesylate; imidazoacridones; imiquimod; immunostimulant peptides; improsulfan tosylate; insulin-like growth factor- 1 receptor inhibitor; interferon; interferon a; interferon a-
  • interferon a-2b interferon agonists; interferon a-nl; interferon a-n3; interferon b-
  • interleukin II interleukin II (IL-2, including recombinant interleukin
  • interleukin II including recombinant interleukin II or rIL2
  • interleukin-2 interleukins
  • iobenguane iobenguane iobenguane
  • iododoxorubicin ipomeanol
  • iproplatin irinotecan; irinotecan (camptosar); irinotecan hydrochloride; irofulven; iroplact; irsogladine; isobengazole; isohomohalicondrin B; isotretinoin (accutane); itasetron; jasplakinolide; kahalalide F; ketoconazole; lamellarin-N triacetate; lanreotide; lanreotide acetate; leinamycin; lenalidomide; lenograstim; lentinan; lentinan s
  • DDD or lysodren mitotoxin fibroblast growth factor-saporin; mitoxantrone; mitoxantrone HCl (novantrone); mofarotene; molgramostim; mopidamol; multiple drug resistance gene inhibitor; multiple tumor suppressor 1 -based therapy; mustard anticancer agent; mycaperoxide B; mycobacterial cell wall extract; mycophenolic acid; myriaporone; N-(l-methylethyl)-4-((2-methylhydrazino)methyl)benzamide; N- acetyldinaline; nafarelin; nagrestip; naloxone and pentazocine; napavin; naphterpin; nartograstim; nedaplatin; nelarabine; nemorubicin; neridronic acid; neutral endopeptidase; nicardipine; nilutamide (nilandron); nimustine
  • combination therapies include surgery, hormone therapy, cryotherapy and gene therapy.
  • the present invention provides for the treatment of various infections, including those caused by bacteria, viruses, fungi and parasites.
  • Bacterial pathogens include Gram-positive cocci such as Staphylococcus aureus, coagulase negative staphylocci such as Staphylococcus epidermis, Streptococcus pyogenes (group A), Streptococcus spp.
  • Gram- negative cocci such as Neisseria gonorrhoeae, Neisseria meningitidis, and Branhamella catarrhalis
  • Gram-positive bacilli such as Bacillus anthracis, Corynebacterium diphtheriae and Corynebacterium species which are diptheroids (aerobic and anerobic), Listeria monocytogenes, Clostridium tetani, Clostridium difficile, Escherichia coli, Enterobacter species, Proteus mirablis and other spp., Pseudomonas aeruginosa, Klebsiella pneumoniae, Salmonella, Shigella, Serratia, and Campylobacter jejuni.
  • the antibiotic resistant bacteria that can be killed by the antiseptic coated devices of the present invention include Staphylococci (methicillin- resistant strains), vancomycin-resistant enterococci ⁇ Enterococcus faecium), and resistant Pseudomonas aeruginosa.
  • Fungal infections that may be prevented include fungal infections (mycoses), which may be cutaneous, subcutaneous, or systemic.
  • Superficial mycoses include tinea capitis, tinea corporis, tinea pedis, onychomycosis, perionychomycosis, pityriasis versicolor, oral thrush, and other candidoses such as vaginal, respiratory tract, biliary, eosophageal, and urinary tract candidoses.
  • Systemic mycoses include systemic and mucocutaneous candidosis, cryptococcosis, aspergillosis, mucormycosis (phycomycosis), paracoccidioidomycosis, North American blastomycosis, histoplasmosis, coccidioidomycosis, and sporotrichosis.
  • Fungal infections include opportunistic fungal infections, particularly in immunocompromised patients such as those with AIDS. Fungal infections contribute to meningitis and pulmonary or respiratory tract diseases.
  • pathogenic organisms that may be prevented from causing the infections include dermatophytes ⁇ Microsporum canis and other M. spp.; and Trichophyton spp. such as T. rubrum, and T. mentagrophytes), yeasts ⁇ e.g., Candida albicans, C. Parapsilosis, C. glabrata, C.Tropicalis, or other Candida species including drug resistant Candida species), Torulopsis glabrata, Epidermophytonfloccosum, Malassezia fuurfur (Pityropsporon orbiculare, or P.
  • Anti-bacterial antibiotics can be categorized based on their target specificity:
  • “narrow-spectrum” antibiotics target particular types of bacteria, such as Gram- negative or Gram-positive bacteria, while broad-spectrum antibiotics affect a wide range of bacteria.
  • Antibiotics which target the bacterial cell wall penicillins, cephalosporins), or cell membrane (polymixins), or interfere with essential bacterial enzymes (quinolones, sulfonamides) usually are bactericidal in nature.
  • Those which target protein synthesis such as the aminoglycosides, macrolides and tetracyclines are usually bacteriostatic.
  • three new classes of antibiotics have been brought into clinical use.
  • cyclic lipopeptides cyclic lipopeptides
  • tigecycline glycylcyclines
  • linearzolid oxazolidinones
  • Antibiotics for use in conjunction with the present invention include amikacin, gentamicin, kanamycin, neomycin, netilmicin, streptomycin, tobramycin, paramomycin, geldanaymicin, herbimycin, loracarbef, ertapenem, doripenem, impenem, meropenem, cefadroxil, cefazolin, cefalotin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cef ⁇ xime, cefdinir, cefditoren, cefoperazon, cefotaxime, cefpodoxime, ceftazidime, cef ⁇ buten, ceftizoxime, ceftriaxone, cefepime, ceftobiprole, teicoplanin, vancomycin, azithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin,
  • the present invention is contemplated to be used in both systemic and localized administration. With respect to systemic administration, it will be desirable to target the zinc nanoparticles to locations within the body using targeting or
  • “homing” molecules such as antibodies or other ligands for pathogen surface receptors.
  • compositions according to the present invention may be via any common route so long as the target tissue is available via that route. This includes oral, nasal, buccal, rectal, vaginal or topical. Alternatively, administration may be by intradermal, subcutaneous, intramuscular, intraperitoneal or intravenous injection. Such compositions would normally be administered as pharmaceutically acceptable compositions, described supra. D. Combination Therapies
  • an "anti-pathogen” therapy is capable of negatively affecting the pathogen infestation in a subject, for example, by directly killing pathogen or pathogen-infected cells, by reducing the growth rate or reproduction of pathogens, or by rendering the pathogen or pathogen-infected cells more susceptible to host defenses. More generally, these therapies would be provided in a combined amount effective to produce a clinically significant result in the subject.
  • This process may involve contacting the host with the therapies at the same time, such as by administering a single composition or treatment that includes both agents, or by administering two distinct compositions or treatments at the same time.
  • the zinc nanoparticle therapy may precede or follow the other treatment by intervals ranging from minutes to weeks.
  • the other agent and zinc nanoparticle therapy are applied separately, one would generally ensure that a significant period of time did not expire between the time of each delivery, such that the therapy and zinc nanoparticle therapy would still be able to exert an advantageously combined effect on the cell.
  • zinc nanoparticle therapy is "A” and the secondary therapy is "B":
  • compositions with activity against various nosocomial microorganisms that inhabit medical devices including resistant bacteria and fungi.
  • the compositions can be used against resistant staphylococci, vancomycin-resistant enterococci, resistant Pseudomonas aeruginosa and Candida species.
  • These compositions can be used to coat of various polymers, such as polyvinyl chloride, polyethylene, silastic elastomers, polytetrafluoroethylene, dacron, collodion, carboethane, nylon, polymers used in the formation of endotracheal tubes, silicone and polyurethane polymers used in the formation of vascular catheters and surgical silk sutures.
  • polymers such as polyvinyl chloride, polyethylene, silastic elastomers, polytetrafluoroethylene, dacron, collodion, carboethane, nylon, polymers used in the formation of endotracheal tubes, silicone and polyurethane polymers used in the formation of vascular catheters
  • the antiseptic compound can be applied on the surface of a device by simply immersing the device in solution containing the nanoparticles, air drying and washing out excessive antiseptic.
  • Another method used to coat surfaces of medical devices with antibiotics involves first coating the selected surfaces with benzalkonium chloride followed by ionic bonding of the antibiotic composition (Solomon and Sherertz, 1987; U.S. Patent 4,442,133).
  • Other methods of coating surfaces of medical devices with antibiotics are taught in U.S. Patent 4,895,566 (a medical device substrate carrying a negatively charged group having a pH of less than 6 and a cationic antibiotic bound to the negatively charged group); U.S.
  • Patent 4,917,686 antibiotics are dissolved in a swelling agent which is absorbed into the matrix of the surface material of the medical device
  • U.S. Patent 4,107,121 constructing the medical device with ionogenic hydrogels, which thereafter absorb or ionically bind antibiotics
  • U.S. Patent 5,013,306 laminating an antibiotic to a polymeric surface layer of a medical device
  • U.S. Patent 4,952,419 applying a film of silicone oil to the surface of an implant and then contacting the silicone film bearing surface with antibiotic powders.
  • the invention also provides methods to generate a wide variety of antiseptic medical devices.
  • Some examples include antiseptic endotracheal tubes, antiseptic vascular catheters, including central venous catheters, arterial lines, pulmonary artery catheters, and peripheral venous catheters, antiseptic urinary catheters, antiseptic nephrostomy tubes, antiseptic biliary stents, antiseptic peritoneal catheters, antiseptic epidural catheters, antiseptic central nervous system catheters, including intraventricular shuts and devices, antiseptic prosthetic valves, orthopedic implants and antiseptic sutures.
  • Zinc nanoparticles of the present invention involve diagnostics and imaging.
  • Zinc nanoparticles have an inherent auto-fluorescent capacity and thus can be used, when coupled to a targeting agent, to image sites within a subject. Such sites may comprise focal infections or tumors.
  • diagnostic labels include enzymes, radiolabels, haptens, fluorescent labels, phosphorescent molecules, chemilluminescent molecules, chromophores, photoaff ⁇ nity molecules, colored particles or ligands, such as biotin, paramagnetic ions, radioactive isotopes, fluorochromes, NMR-detectable substances, and X-ray imaging agents.
  • paramagnetic ions such as chromium (III), manganese (II), iron (III), iron (II), cobalt (II), nickel (II), copper (II), neodymium (III), samarium (III), ytterbium (III), gadolinium (III), vanadium (II), terbium (III), dysprosium (III), holmium (III) and/or erbium (III), with gadolinium being particularly preferred.
  • Ions useful in other contexts, such as X-ray imaging include but are not limited to lanthanum (III), gold (III), lead (II), and especially bismuth (III).
  • radioactive isotopes for therapeutic and/or diagnostic application, one might mention astatine 211 , 14 carbon, 51 chromium, 36 chlorine, 57 cobalt, 58 cobalt, copper 67 , 152 Eu, gallium 67 , 3 hydrogen, iodine 123 , iodine 125 , iodine 131 , indium 111 , 59 iron, 32 phosphorus, rhenium 186 , rhenium 188 , 75 selenium, 35 sulphur, technicium 99m and/or yttrium 90 .
  • 125 I is often being preferred for use in certain embodiments, and technicium 99m and/or indium 111 are also often preferred due to their low energy and suitability for long range detection.
  • fluorescent labels contemplated for use as conjugates include Alexa 350, Alexa 430, AMCA, BODIPY 630/650, BODIPY 650/665, BODIPY-FL, BODIP Y-R6G, BODIPY-TMR, BODIPY-TRX, Cascade Blue, Cy3, Cy5,6-FAM, Fluorescein Isothiocyanate, HEX, 6-JOE, Oregon Green 488, Oregon Green 500, Oregon Green 514, Pacific Blue, REG, Rhodamine Green, Rhodamine Red, Renographin, ROX, TAMRA, TET, Tetramethylrhodamine, and/or Texas Red.
  • Radiographic images Two forms of radiographic images are in use in medical imaging; projection radiography and fluoroscopy, with latter useful for intraoperative and catheter guidance. These 2D techniques are still in wide use despite the advance of 3D tomography due to the low cost, high resolution, and depending on application, lower radiation dosages.
  • This imaging modality utilizes a wide beam of x rays for image acquisition and is the first imaging technique available in modern medicine.
  • Fluoroscopy produces real-time images of internal structures of the body in a similar fashion to radiography, but employs a constant input of x-rays, at a lower dose rate. Contrast media, such as barium, iodine, and air are used to visualize internal organs as they work. Fluoroscopy is also used in image-guided procedures when constant feedback during a procedure is required. An image receptor is required to convert the radiation into an image after it has passed through the area of interest. Early on this was a fluorescing screen, which gave way to an Image Amplifier (IA) which was a large vacuum tube that had the receiving end coated with cesium iodide, and a mirror at the opposite end. Eventually the mirror was replaced with a TV camera.
  • IA Image Amplifier
  • Radio-opaque contrast media such as barium
  • they can also be used to visualize the structure of the stomach and intestines - this can help diagnose ulcers or certain types of colon cancer.
  • Gamma cameras are used in nuclear medicine to detect regions of biological activity that are often associated with diseases.
  • a short lived isotope, such as 123 I is administered to the patient. These isotopes are more readily absorbed by biologically active regions of the body, such as tumors or fracture points in bones.
  • Computed tomography is a medical imaging method employing tomography. Digital geometry processing is used to generate a three-dimensional image of the inside of an object from a large series of two-dimensional X-ray images taken around a single axis of rotation. Computed tomography was originally known as the "EMI scan” as it was developed at a research branch of EMI, a company best known today for its music and recording business. It was later known as computed axial tomography (CAT or CT scan) and body section r ⁇ ntgenography.
  • EMI scan computed axial tomography
  • CT produces a volume of data which can be manipulated, through a process known as "windowing,” in order to demonstrate various bodily structures based on their ability to block the X-ray/R ⁇ ntgen beam.
  • Positron emission tomography is primarily used to detect diseases of the brain and heart. Similarly to nuclear medicine, a short-lived isotope, such as F, is incorporated into a substance used by the body such as glucose which is absorbed by the tumor of interest. PET scans are often viewed alongside computed tomography scans, which can be performed on the same equipment without moving the patient. This allows the tumors detected by the PET scan to be viewed next to the rest of the patient,s anatomy detected by the CT scan.
  • Another 3D tomographic technique is SPECT but uses gamma camera-like method for reconstruction.
  • MRI uses three electromagnetic fields: a very strong (on the order of units of teslas) static magnetic field to polarize the hydrogen nuclei, called the static field; a weaker time-varying (on the order of 1 kHz) field(s) for spatial encoding, called the gradient field(s); and a weak radio-frequency (RF) field for manipulation of the hydrogen nuclei to produce measurable signals, collected through an RF antenna.
  • a very strong (on the order of units of teslas) static magnetic field to polarize the hydrogen nuclei called the static field
  • a weaker time-varying (on the order of 1 kHz) field(s) for spatial encoding called the gradient field(s)
  • RF radio-
  • MRI Like CT, MRI traditionally creates a two dimensional image of a thin "slice" of the body and is therefore considered a tomographic imaging technique. Modern MRI instruments are capable of producing images in the form of 3D blocks, which may be considered a generalisation of the single-slice, tomographic, concept. Unlike CT, MRI does not involve the use of ionizing radiation and is therefore not associated with the same health hazards. For example, because MRI has only been in use since the early 1980s, there are no known long-term effects of exposure to strong static fields and therefore there is no limit to the number of scans to which an individual can be subjected, in contrast with X-ray and CT. However, there are well-identified health risks associated with tissue heating from exposure to the RF field and the presence of implanted devices in the body, such as pace makers. These risks are strictly controlled as part of the design of the instrument and the scanning protocols used.
  • CT and MRI are sensitive to different tissue properties, the appearance of the images obtained with the two techniques differ markedly.
  • X- rays must be blocked by some form of dense tissue to create an image, so the image quality when looking at soft tissues will be poor.
  • MRI while any nucleus with a net nuclear spin can be used, the proton of the hydrogen atom remains the most widely used, especially in the clinical setting, because it is so ubiquitous and returns a large signal. This nucleus, present in water molecules, allows the excellent soft-tissue contrast achievable with MRI.
  • Example 1 ZNP as drug carriers
  • ZNPs vs. quantum dots as drug delivery vehicles is their safety and complete dissolution in the body. As such they can be used to alter the pharmacodynamics of drugs conjugated to their surface.
  • the surface of ZNPs can be protected from the aqueous environment through the use of surface polymers. Based on the level of surface protection and the composition of the coating the rate of dissolution and hence drug delivery can be altered.
  • Envisioned is a systemic method of increasing the half- life of pharmaceutical compositions.
  • An example would be the conjugation of cancer chemotherapeutics such as doxorubicin to ZNPs.
  • the chemical composition would consist of a specific molar ratio of ZNPs and doxorubicin.
  • non- drug bound surface of the ZNPs would be coated with polymers such as Poly-ethylene glycol.
  • polymers such as Poly-ethylene glycol.
  • cytotoxic drugs attached to ZNPS Both the cytotoxic drug and ZNPs would accumulate within a tumor environment and act synergistically to improve efficacy by increasing cell killing.
  • An example would be ZNP conjugated with Rapamycin.
  • Cells highly responsive to mTOR signaling would be treated through the administration of ZNP conjugated Rapamycin.
  • Rapamycin would exert cytotoxic effect.
  • targeting ligands In addition, it is anticipated that through the use of targeting ligands, highly specific delivery of both ZNP, Drug Conjugate or both can be used to improve efficacy with improved biosafety.
  • the targeting ligand would result in highly specific delivery of drug conjugate directly to cells resulting in reduced metabolic breakdown and improved pharmacokinetics and improved efficacy.
  • Neoadjuvant therapy is used for the reduction of tumor size prior to systemic therapy.
  • ZNPs are ideally suited as neoadjuvant agents for the reduction of tumor size.
  • the inventors formulated inert biogels with ZNPs.
  • Pluronic gels were mixed with ZNPs to make formulations between 1-30% w/v.
  • Pluronic is a block co-polymer that can retard delivery of drugs.
  • the rate of ZNP dissolution was altered to result in slow release of zinc ions. The rate of release was dependent on the w/v ratio of ZNPs and the percent formulation of the gel.
  • In vitro kinetics of zinc ion release is shown in FIG. 4 (solubility of ZNP in BioGell formulations of pluronic).
  • ZNP Cytotoxicity is enhanced by internalization within cells. Specificity of delivery to cells (bacterial, cancer, etc.) can be controlled through the conjugation of specific targeting ligands.
  • the inventors used two cancer cell lines SKO V3 (ovarian cancer) and SKBR3 (breast adenocarcinoma) which overexpress Her2-neu. These cell lines were treated with ZNPs or ZNPs conjugated with antibody against Her2-neu (Herceptin analogue) Since ZNPs exhibit cytotoxic effect after 2 hrs to distinguish the effect of conjugated versus non-conjugated ZNPs, cells were treated for only 1 hr and then extensively washed to remove extracellular ZNPs and zinc ions (FIG. 10).
  • Cytotoxicity was measured at 18 hrs by standard MTT assay. Conjugated ZNPs were internalized while non-conjugated ZNPs were not. Conjugated ZNPs showed 100% cytotoxic killing of both Her2-Neu expressing cell lines but no effect against non- Her2-Neu expressing cells showing that internalization and cytotoxic effect or specificity was dependent on the targeting ligand (FIG. 10). The Her2-Neu antibody showed no effect over this short period of time indicating that cell death required both cell receptor specific targeting through the use of a binding ligand and ZNPs.
  • the inventors used three different methods of conjugating antibodies and proteins to the surface of zinc nanoparticles.
  • the first is ligand binding with zero-length cross-linkers.
  • EDC EDC reacts with carboxyl groups to form reactive amines.
  • Sulfo-NHS Sulfo-NHS
  • ligands were bound to the EDC-coated zinc nanoparticles at the carboxyl group.
  • the second is polymer coating.
  • zinc nanoparticles were coated with PEG of various molecular weights 1000-20000. ZNP maintain a slight negative charge in acidic environments allowing for electrostatic binding of reactive polymers.
  • PEO and vinyl esters were bound in a similar manner.
  • the third is ligand binding with monofuntional and bifunctional crosslinkers.
  • antibodies to Her2 were conjugated to the reactive groups to result in antibodies attached monofunctionally or heterobifunctionally.
  • Heterobifunctional ligand attachment resulted in greater zinc deliver intracellularly to cells.
  • Polysaccharide polymer binding to ZNP was performed by physical adsorption under alkaline conditions.
  • PEG Sigma Aldrich
  • PEO Sigma Aldrich
  • polyacrylic acids b polystyrenes Sigma Aldrich
  • the PEG, PEO and vinyl-esters were purchased with functional amide groups for subsequent binding of antibody by chemical reduction.
  • the chain length of the coatings varied between 1000-20000 and for the amphiphilic block copolymers between 30-136 units.
  • ZNPs (5mg) were suspended in neutral citrate buffer (ph 7.4) and then dried. Dry PEG, PEO, Esters were added and the mixture rested for 2 hours. The mixture was then hydrated with water, washed, centrifuged, and then resuspended in 100 mM MES buffer (ph 5.0) and stored prior to experimentation.
  • Antibody or ligand attachment was performed as described above.
  • ZNP can be functionalized using standard chemistries used for nanoparticle binding and conjugation. ZNPs show similar efficiencies in ligand binding to iron-oxide and zinc-oxide particles of similar size. (See Figures 1,2, 3, 4) Example 6 - Uptake data
  • the inventors treated prostate cancer cells, ovarian cancer cells and breast cancer cells with HER2 or transferrin-targeted ZNP (FIGS. 13 and 14).
  • the prostate cancer cells, PC3, are known to express transferrin receptor but not HER2 while both the ovarian cancer cells (SKOV3) and breast cancer cells (SKBR3) are known to express HER2.
  • Two separate HER2 antibodies were used.
  • the inventors evaluated cell specificity. Prostate cancer cells were only killed by transferrin-targeted ZNPs while the ovarian and breast cancer cells showed specificity for the HER2 targeted ZNPs.
  • ZNPs were coated with different polymers including PEG and vinyl esters (see conjugation methodology). Based on the length of the polymer, the degree of hydrophobicity could be altered. Zinc ion release (half-life) of ZNPs were dependent on the degree of hydrophobicity of the polymer coating (FIG. 15).
  • Zinc ion concentration was measured upon placing ZNPs in heat-inactivated fetal calf serum. Zinc ion concentration was measured by fluorescent spectroscopy of removed aliquots with the fluorescent molecular probe Fluozin3.
  • compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims. VII. References

Abstract

La présente invention a trait à la production et à l’utilisation de nanoparticules de zinc pharmaceutiquement acceptables ayant des noyaux comprenant du zinc élémentaire sans quantités significatives d’autres métaux ou oxydes métalliques. La présente invention a également trait à l’utilisation de ces nanoparticules dans le cadre de la thérapie contre le cancer et du traitement de maladies infectieuses, ainsi que de l’imagerie médicale.
PCT/US2010/045688 2009-08-17 2010-08-17 Nanoparticules de zinc pour le traitement d’infections et du cancer WO2011022350A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US23437409P 2009-08-17 2009-08-17
US61/234,374 2009-08-17

Publications (1)

Publication Number Publication Date
WO2011022350A1 true WO2011022350A1 (fr) 2011-02-24

Family

ID=43607298

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/045688 WO2011022350A1 (fr) 2009-08-17 2010-08-17 Nanoparticules de zinc pour le traitement d’infections et du cancer

Country Status (1)

Country Link
WO (1) WO2011022350A1 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2475251C1 (ru) * 2012-02-06 2013-02-20 Владимир Владимирович Алипов Способ комбинированного лечения абсцессов в эксперименте
US20180271904A1 (en) * 2017-03-21 2018-09-27 Auburn University Engineered metal nanoparticles and methods thereof
US10398732B2 (en) 2016-10-13 2019-09-03 Marshall University Research Corporation Compositions and methods for treating striated muscle injury, treating striated muscle atrophy and/or for promoting striated muscle growth
WO2020086014A1 (fr) 2018-10-25 2020-04-30 Yeditepe Universitesi Utilisation de nanoparticules de borate de plomb ciblées par un gène p53 mutant dans le traitement du cancer et procédé de production de ces nanoparticules
US11278512B2 (en) * 2019-08-21 2022-03-22 Brain Chemistry Labs Compositions comprising a metal and L-serine, and uses thereof
WO2022204714A1 (fr) * 2021-03-24 2022-09-29 Kansas State University Research Foundation Composites de physio-nanocomposites à base de zinc et leurs méthodes d'utilisation

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070212331A1 (en) * 2006-03-07 2007-09-13 Baldassare Joseph J Methods and compositions for selectively killing cells
US20070269382A1 (en) * 2004-04-30 2007-11-22 Swadeshmukul Santra Nanoparticles and Their Use for Multifunctional Bioimaging
US20080089836A1 (en) * 2006-10-12 2008-04-17 Nanoprobes, Inc. Functional associative coatings for nanoparticles

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070269382A1 (en) * 2004-04-30 2007-11-22 Swadeshmukul Santra Nanoparticles and Their Use for Multifunctional Bioimaging
US20070212331A1 (en) * 2006-03-07 2007-09-13 Baldassare Joseph J Methods and compositions for selectively killing cells
US20080089836A1 (en) * 2006-10-12 2008-04-17 Nanoprobes, Inc. Functional associative coatings for nanoparticles

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2475251C1 (ru) * 2012-02-06 2013-02-20 Владимир Владимирович Алипов Способ комбинированного лечения абсцессов в эксперименте
US10398732B2 (en) 2016-10-13 2019-09-03 Marshall University Research Corporation Compositions and methods for treating striated muscle injury, treating striated muscle atrophy and/or for promoting striated muscle growth
US20180271904A1 (en) * 2017-03-21 2018-09-27 Auburn University Engineered metal nanoparticles and methods thereof
US10806754B2 (en) 2017-03-21 2020-10-20 Auburn University Engineered metal nanoparticles and methods thereof
WO2020086014A1 (fr) 2018-10-25 2020-04-30 Yeditepe Universitesi Utilisation de nanoparticules de borate de plomb ciblées par un gène p53 mutant dans le traitement du cancer et procédé de production de ces nanoparticules
US11278512B2 (en) * 2019-08-21 2022-03-22 Brain Chemistry Labs Compositions comprising a metal and L-serine, and uses thereof
WO2022204714A1 (fr) * 2021-03-24 2022-09-29 Kansas State University Research Foundation Composites de physio-nanocomposites à base de zinc et leurs méthodes d'utilisation

Similar Documents

Publication Publication Date Title
Zhang et al. Ultrasound-driven biomimetic nanosystem suppresses tumor growth and metastasis through sonodynamic therapy, CO therapy, and indoleamine 2, 3-dioxygenase inhibition
Lu et al. Drug delivery in cancer therapy, quo vadis?
Ni et al. Photothermal-chemotherapy enhancing tumor immunotherapy by multifunctional metal–organic framework based drug delivery system
WO2011022350A1 (fr) Nanoparticules de zinc pour le traitement d’infections et du cancer
Safavy et al. Synthesis and Biological Evaluation of Paclitaxel− C225 Conjugate as a Model for Targeted Drug Delivery1
JP2021503492A (ja) 放射線療法と組み合わせて癌を治療するための粒子
CN108377643A (zh) 使用超小纳米粒子通过铁死亡诱导营养素剥夺癌细胞的细胞死亡的治疗方法
CN107001031A (zh) 用于光动力疗法、x射线诱导的光动力疗法、放射疗法、化学疗法、免疫疗法及其任意组合的纳米颗粒
CA2995029C (fr) Formulation medicamenteuse a base de particules comprenant un conjugue polysaccharide-vitamine
CN108697815A (zh) 具有egfr配体的治疗性纳米颗粒及其制备和使用方法
US20210069115A1 (en) High Surface-Area Lyophilized Compositions Comprising Arsenic For Oral Administration In Patients
CN108473538A (zh) Sstr靶向缀合物及其颗粒和制剂
Yuan et al. Target challenging-cancer drug delivery to gastric cancer tissues with a fucose graft epigallocatechin-3-gallate-gold particles nanocomposite approach
JP2018518509A (ja) 白金化合物、組成物及びその使用
Munoz et al. Metronomic anti-cancer therapy: A multimodal therapy governed by the tumor microenvironment
Kong et al. Injectable and thermosensitive liposomal hydrogels for NIR-II light-triggered photothermal-chemo therapy of pancreatic cancer
Fernandes et al. Metal-organic frameworks applications in synergistic cancer photo-immunotherapy
CN113260653B (zh) 用于递送抗癌剂的立体复合物
Péraudeau et al. Combination of Targeted Therapies for Colorectal Cancer Treatment
BR112020008375A2 (pt) métodos de reduzir efeitos colaterais de terapia conjugada com fármacos de anticorpo anti-cd30
US20220395578A1 (en) Therapeutic annexin-drug conjugates and methods of use
Nasser et al. Combined cancer immunotheranostic nanomedicines: Delivery technologies and therapeutic outcomes
CN102935228B (zh) 用于肿瘤治疗的试剂、其用途及方法
Qian et al. Self-assembled amphiphilic copolymers–doxorubicin conjugated nanoparticles for gastric cancer therapy with low in vivo toxicity and high efficacy
Hu et al. Paclitaxel-loaded tumor cell-derived microparticles improve radiotherapy efficacy in triple-negative breast cancer by enhancing cell killing and stimulating immunity

Legal Events

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

Ref document number: 10810455

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10810455

Country of ref document: EP

Kind code of ref document: A1