WO2003101425A2 - Articles polymeres contenant des agents therapeutiques - Google Patents

Articles polymeres contenant des agents therapeutiques Download PDF

Info

Publication number
WO2003101425A2
WO2003101425A2 PCT/US2003/017536 US0317536W WO03101425A2 WO 2003101425 A2 WO2003101425 A2 WO 2003101425A2 US 0317536 W US0317536 W US 0317536W WO 03101425 A2 WO03101425 A2 WO 03101425A2
Authority
WO
WIPO (PCT)
Prior art keywords
nanoarticle
hydrogel
scaffold
building blocks
nano
Prior art date
Application number
PCT/US2003/017536
Other languages
English (en)
Other versions
WO2003101425A3 (fr
WO2003101425A8 (fr
Inventor
Stephen E. Barry
Andrew A. Goodwin
Rachel Decor
Original Assignee
Alnis Biosciences, Inc.
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 Alnis Biosciences, Inc. filed Critical Alnis Biosciences, Inc.
Priority to AU2003273272A priority Critical patent/AU2003273272A1/en
Priority to EP03741863A priority patent/EP1531868A2/fr
Priority to CA002487720A priority patent/CA2487720A1/fr
Priority to JP2004508783A priority patent/JP2005535604A/ja
Publication of WO2003101425A2 publication Critical patent/WO2003101425A2/fr
Priority to US10/995,880 priority patent/US20050129769A1/en
Publication of WO2003101425A3 publication Critical patent/WO2003101425A3/fr
Publication of WO2003101425A8 publication Critical patent/WO2003101425A8/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/51Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C323/57Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being further substituted by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C323/58Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being further substituted by nitrogen atoms, not being part of nitro or nitroso groups with amino groups bound to the carbon skeleton
    • C07C323/59Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being further substituted by nitrogen atoms, not being part of nitro or nitroso groups with amino groups bound to the carbon skeleton with acylated amino groups bound to the carbon skeleton
    • 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
    • A61K47/51Medicinal 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 non-active ingredient being a modifying agent
    • A61K47/56Medicinal 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 non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/61Medicinal 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 non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule the organic macromolecular compound being a polysaccharide or a derivative thereof
    • 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
    • A61K47/6931Medicinal 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 the material constituting the nanoparticle being a polymer
    • 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
    • A61K47/6931Medicinal 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 the material constituting the nanoparticle being a polymer
    • A61K47/6933Medicinal 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 the material constituting the nanoparticle being a polymer the polymer being obtained by reactions only involving carbon to carbon, e.g. poly(meth)acrylate, polystyrene, polyvinylpyrrolidone or polyvinylalcohol
    • 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
    • A61K47/6931Medicinal 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 the material constituting the nanoparticle being a polymer
    • A61K47/6935Medicinal 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 the material constituting the nanoparticle being a polymer the polymer being obtained otherwise than by reactions involving carbon to carbon unsaturated bonds, e.g. polyesters, polyamides or polyglycerol
    • 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
    • A61K47/6931Medicinal 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 the material constituting the nanoparticle being a polymer
    • A61K47/6939Medicinal 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 the material constituting the nanoparticle being a polymer the polymer being a polysaccharide, e.g. starch, chitosan, chitin, cellulose or pectin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/167Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction with an outer layer or coating comprising drug; with chemically bound drugs or non-active substances on their surface

Definitions

  • the present invention is directed to the field of therapeutic entities. More specifically, this invention relates to polymeric articles incorporating bioactive components.
  • Small molecule drugs are often comprised of substantial hydrophobic structures that have limited water and blood solubility. These compounds will interact strongly with biomembranes, will rapidly diffuse through biomembranes, and thus will readily exit the bloodstream and nonspecifically penetrate tissues. Such undesirable distribution can lead to high dosing requirements, rapid clearance, and toxic side effects. In fact, small molecule drugs can pose toxicity challenges regardless of hydrophobic or hydrophilic nature. To improve performance, drugs have been covalently conjugated to water-soluble, synthetic polymers, such as N-(2-hydroxypropyl)methacrylamide (HPMA) (Buttles et al., Hum. Exp.
  • HPMA N-(2-hydroxypropyl)methacrylamide
  • Such drug-polymer conjugates thus reside in the bloodstream longer in comparison to the drug alone and may therefore be passively targeted to tissue that has a leaky vasculature.
  • Active targeting of drugs has been achieved through the use of ligands constructs capable of binding to specific biomolecular structures, such as receptors overexpressed on certain cancer cells, for the purpose of localizing the bioactive agents to a desired type of cell or cellular component, type of tissue or tissue component, or organ.
  • the HPMA-drug conjugates have been further functionalized with monoclonal antibodies that target receptors overexpressed on certain cancers.
  • drug molecules have been directly conjugated to monoclonal antibodies (mAb).
  • Seattle Genetics Seattle, WA
  • the terms “articles” and “nano-articles” are used interchangeably.
  • This invention is directed to hydrophilic polymeric nano-articles comprising i) a scaffold comprised of crosslinked hydrophilic building blocks and ii) one or more therapeutic agents, such as drug or drug-conjugate molecules, covalently attached to the scaffold.
  • the nano-articles may further optionally comprise one or more recognition elements (REs) to facilitate targeting and/or delivery.
  • the nano-articles may also optionally be comprised of polyethylene glycol (PEG)-based molecules.
  • the PEG chains may serve as linkers or tethers, with one end attached to the nano-article surface and the other end functionalized with an RE, and/or the PEG chains may provide other useful or desirable characteristics to the hydrophilic nano-articles.
  • the invention is further directed to methods of synthesizing these nano-articles and to the various applications for which they may be used.
  • the nano-articles of the invention are preferably from about 5 nm to about 100 nm in diameter, more preferably from about 10 nm to about 70 nm.
  • the size of the nano-articles allows their use as bioactive entities in mammals.
  • nano-articles are preferably less than 100 nm.
  • nano- articles are preferably larger than 5 nm. Reverse microemulsion polymerization can yield nano-article scaffolds of the invention.
  • Therapeutic agents are incorporated into the nano-articles through covalent linkage to the hydrogel scaffold.
  • a preferred therapeutic entity is doxorubicin or doxorubicin derivatives.
  • REs serve to bind the nano-article of the invention to desired biomolecules overexpressed or otherwise found on certain cell surfaces or in certain tissues.
  • the number of REs per nano-article can range from 2 to about 1000, preferably from 2 to 500.
  • the nano-articles may optionally further be comprised of more than one type of RE.
  • a RE "type" is defined as an RE of a specific molecular structure.
  • the nano-article scaffolds are comprised of crosslinked hydrophilic building blocks.
  • the building blocks are crosslinked in the dispersed aqueous phase of a reverse microemulsion.
  • Carbohydrates and carbohydrate derivatives are preferably used as building blocks.
  • water-soluble is meant, herein and in the appended claims, having a solubility in water of greater that 10 mg/mL, and preferably greater than 50 mg/mL.
  • the nano-articles of this invention are comprised of two types of molecular structures: scaffolds and therapeutic agents.
  • the nano-articles also comprise a third molecular structure: REs that bind to biomolecules expressed on certain cells or in certain tissues,
  • the REs and the therapeutic agents are covalently attached to the nano-article scaffold.
  • the terms "nano-article scaffold”, “hydrogel scaffold” and “scaffold” are used interchangeably and refer to the portion of the nano-article (the polymeric matrix structure) that is formed prior to attachment of bioactive agents and recognition elements.
  • the scaffold of the present invention is a chemically crosslinked, nanoscopic hydrogel structure.
  • the nano-articles are hydrophilic and intended for use in mammals.
  • each nano-article is functionalized with two or more recognition elements that possess high affinity to biomolecular targets, the recognition elements being covalently linked to the nano-article polymeric matrix structure.
  • the invention is further directed to methods of synthesizing these polymeric nano-articles.
  • the nano-articles of the invention may range in size from about 5 nm to about 1000 nm, more preferably from about 5 nm to about 100 nm in diameter, and most preferably about 10 nm to about 70 nm. Nano-articles in the 10 to 70 nm size range may effectively avoid renal clearance and uptake by the reticuloendothelial system. Additionally, such nano-articles may advantageously exit the blood stream to reach desired cell, tissue, or organ targets.
  • the nano-articles of the present invention may, in particular, be advantageously used in the treatment of cancer.
  • the leaky vasculature found in tumors may allow these nano-articles to leave the blood stream and concentrate in tumors.
  • This effect described as enhanced permeability and retention (EPR) for macromolecular agents, has been observed to be universal in solid tumors (Maeda H., et al., J. Controlled Release, 2000, 65, p.271 - 284).
  • the key mechanism for the EPR effect for macromolecules is retention, whereas low- molecular-weight substances are not retained but are returned to circulating blood by diffusion. Nano-articles of diameters from 5 to 100 nm can thus accumulate in solid tumors.
  • Hydrophilic building blocks with polymerizable groups are employed to form the hydrogel scaffold.
  • the building blocks are crosslinked in the dispersed aqueous phase of reverse microemulsions.
  • the number of polymerizable groups attached to one single building block can range, for example, from about one to three for low molecular weight building blocks, to ten or more for polymeric building blocks. Building blocks that contain more than one polymerizable group can act as crosslinking agents and enable the formation of a hydrogel scaffold.
  • polymerizable groups include, but are not limited to, acrylate, acrylamide, methacrylate, methacrylamide, vinyl ether, styryl, epoxide, maleic acid derivative, diene, substituted diene, thiol, alcohol, amine, hydroxyamine, carboxylic acid, carboxylic anhydride, carboxylic acid halide, aldehyde, ketone, isocyanate, succinimide, carboxylic acid hydrazide, glycidyl ether, siloxane, alkoxysilane, alkyne, azide, 2'-pyridyldithiol, phenylglyoxal, iodo, maleimide, imidoester, dibromopropionate, and io
  • Free-Radical Polymerization Preferred polymerizable functionalities are acrylate, acrylamide, methacrylate, and methacrylamide moieties. Such moieties are amenable to free-radical polymerization. Free-radical polymerization can be readily achieved through the combination of UV light and photoinitiators, redox-coupled free-radical initiators, or heat and heat-activated initiators.
  • Building blocks that may be used to form the article scaffold include small molecules with one polymerizable group or multiple polymerizable moieties that can act as small molecule crosslinkers.
  • Exemplary building blocks include acrylamide, sodium acrylate (NaA), diacetone acrylamide (DAA), malonate acrylamide (MalAc), levulinic acrylamide, methylene bisacrylamide, ammonium 2,2-bisacrylamidoacetate, 2-acrylamidoglycolic acid, 2-aminoethyl methacrylate, N-(3-aminopropyl)methacrylamide (APMA), ornithine mono- acrylamide, ornithine diacrylamide sodium salt, N-acryloyltris(hydroxymethyl)-methylamine, hydroxyethylacrylate, N-(2-hydroxypropyl)methacrylamide, N-(2-hydroxypropyl)acrylamide, 2-sulfoethylmeth-acrylate, 2-methacryloylethyl gluco
  • Building blocks are chosen to achieve a desired content of certain functionalities in the article scaffold. Such functionalities can improve solubility and may also be used as points of attachment for therapeutic agents and REs.
  • APMA may be used to introduce amines
  • sodium acrylate may be used to introduce carboxylates
  • DAA may be used to incorporate ketones.
  • at least some of the building blocks are a N,N'-cystinebisacrylamide (CiBA) monomer, which has the following formula I:
  • CiBA may be prepared by reacting L-cystine (II) with 2 equivalents of acryloyl chloride (III), according to the following reaction scheme:
  • CiBA is water-soluble and is capable of polymerizing with other scaffold building blocks, such as acrylate-functionalized carbohydrates, to form the hydrogel scaffold.
  • its disulfide linkage provides, after reduction, free thiols for linker attachment.
  • at least some of the building blocks are carbohydrates.
  • the carbohydrate region is comprised of a plurality of hydroxyl groups, wherein at least one hydroxyl group is modified to include at least one polymerizable group.
  • the carbohydrate region of the carbohydrate building block may include a carbohydrate or carbohydrate derivative.
  • the carbohydrate region may be derived from a simple sugar, such as N-acetylglucosamine, N-acetylgalctosamine, N- acetylneuraminic acid, neuraminic acid, galacturonic acid, glucuronic acid, ioduronic acid, glucose, ribose, arabinose, xylose, lyxose, allose, altrose, apiose, mannose, gulose, idose, galactose, fucose, fructose, fructofuranose, rhamnose, arabinofuranose, and talose; a disaccharide, such as maltose, sucrose, lactose, or trehalose; a trisaccharide; a polysaccharide, such as cellulose, starch, glycogen, alginates, inulin, pull
  • carbohydrate-based building blocks may be prepared from the carbohydrate precursor (e.g. sucrose, inulin, dextran, pullulan, etc.) by coupling technologies known in the art of bioorganic chemistry (see, for example, G Hermanson, Bioconjugation Techniques, Academic Press, San Diego, 1996, pp 27-40,155, 183-185, 615-617; and S.
  • carbohydrate precursor e.g. sucrose, inulin, dextran, pullulan, etc.
  • a crosslinkable group can be attached to a carbohydrate via the dropwise addition of acryloyl chloride to an amine-functionalized sugar.
  • Amine-functionalized sugars can be prepared by the reaction of ethylene diamine (or other amines) with 1 ,1'-carbonyldiimidazole-activated sugars.
  • Ester-linked reactive groups can be synthesized through the reaction of acrylic or methacrylic anhydrides with the hydroxyl group of a carbohydrate such as inulin in pyridine.
  • Carbohydrate-based building blocks may also be prepared by the partial (or complete) functionalization of the carbohydrate with moieties that are known to polymerize under free radical conditions.
  • methacrylic esters may be placed on a carbohydrate at varying substitution levels by the reaction of the carbohydrate with methacrylic anhydride or glycidyl methacrylate (Vervoort L., et al., International Journal of Pharmaceutics, 1998, 172, 127-135).
  • the building blocks are inulin multi-methacrylate (IMMA) monomer, which has the following formula IV:
  • n in formula IN is from about 5 to about 50.
  • inulin with an average degree of polymerization (DOP) of about 10 to about 20 is used.
  • n is from about 8 to about 18.
  • DOP average degree of polymerization
  • the extent to which inulin is functionalized with methacrylate moieties is a statistical process governed by the concentrations and weight ratios of inulin and methacrylic anhydride starting material.
  • the extent of functionalization may range from one methacrylate for every 1 to 100 monosaccharide repeat units, more preferably one methacrylate for every 3 to 20 monosaccharide repeat units.
  • the ester linkage to inulin may advantageously function as a site of degradation in vivo, allowing the article to degrade and be cleared from the body.
  • Dextran multimethacrylamide and pullulan multimethacrylamide are additional preferred building blocks that may be prepared using similar methods.
  • Carbohydrate-based building blocks may also be prepared by chemoenzymatic methods (Martin B.D., et al., Macromolecules, 1992, 25, 7081), for example in which Pseudomonas cepacia catalyzes the transesterification of monosaccharides with vinyl acrylate in pyridine or by the direct addition of an acrylate (Piletsky S., et al., Macromolecules, 1999, 32, 633-636).
  • Other functional groups may be present, as numerous derivatized carbohydrates are known to those familiar with the art of carbohydrate chemistry.
  • the carbohydrate structures are chosen in part for their hydrophilicity. ⁇ ano-articles that incorporate substantially hydrophobic drugs must possess highly hydrophilic scaffolds in order that high water solubility be maintained after functionalization with the drug.
  • the nano-articles of the present invention that are comprised of IMMA have a high water content.
  • “High water content”, as used herein and the appended claims, means an article comprised of about 65 to about 98 wt% water, more preferably about 75 to about 98 wt% water, and most preferably about 80 to 97 wt% water.
  • the high water content scaffolds also can reduce immunogenicity, because there are fewer surfaces for immune system components to interact with.
  • acrylate- or acrylamide-derivatized polymeric building blocks include polyethylene glycol-based molecules, such as polyethyleneglycol diacrylate, with molecular weights ranging from 200 to 40,000 daltons.
  • degradable linkages are included within the crosslinked scaffold.
  • Degradable linkages can be included through the use of polylactide, polyglycolide, poly(lactide-co-glycolide), polyphosphazine, polyposphate, polycarbonate, polyamino acid, polyanhydride, and polyorthoester - based building blocks, among others.
  • degradable linkages may be used to attach polymerizable moieties to carbohydrates.
  • IMMA contains ester moieties that connect the inulin carbohydrate backbone to the alkyl chain that is formed upon free radical polymerization used to generate the scaffold of the present invention.
  • small molecule crosslinking agents containing similar hydrolyzable moieties as the polymers such as carbonates, esters, urethanes, orthoesters, amides, and phosphates may be used as building blocks.
  • these building blocks must be functionalized with two or more polymerizable moieties.
  • polyglycolide diacrylate, polyorthoester diacrylate and acrylate-substituted polyphosphazine, acrylate-substituted polyamino acid, or acrylate-substituted polyphosphate polymers can be used as degradable building blocks.
  • Methacrylate or acrylamide moieties can be employed instead of acrylate moieties in the above examples.
  • small molecules containing a hydrolyzable segment and two or more acrylates, methacrylates, or acrylamides may be used.
  • Such degradable polymers and small molecule building blocks may be functionalized with acrylate, methacrylate, acrylamide or similar moieties by methods known in the art.
  • the nano-article scaffolds and the scaffold breakdown products of this invention are designed to be non-toxic and eliminated from the body. They may have degradable, preferably carbohydrate-based, polyamino acid-based, polyester-based, or PEG-based cores, with the rate of degradation controlled by the identity of the sugar, crosslink density, and other features.
  • degradable preferably carbohydrate-based, polyamino acid-based, polyester-based, or PEG-based cores
  • the rate of degradation controlled by the identity of the sugar, crosslink density, and other features.
  • Chemoselective building blocks may also be used to form the scaffold.
  • a representative example of this strategy may be the use of a polysaccharide that has been partially oxidized to contain numerous aldehydes within a reverse microemulsion.
  • a di(amino-oxy) containing compound such as that made from reacting ethylene diamine with the NHS ester of Boc-amino-oxyacetic acid (see the following Reaction Scheme), can then be used as a crosslinking agent through the reaction of the aldehydes of the oxidized sugar reacting with the amino-oxy functionalities.
  • Articles of the present invention are fabricated by first forming nanoscopic hydrogel scaffolds through the crosslinking of hydrophilic building blocks solubilized in the dispersed water phase of a reverse microemulsion. The organic solvent and non-reactive surfactants are removed after polymerization to yield crosslinked, water-soluble nanoscopic articles.
  • Reverse microemulsions for scaffold fabrication are formed by combining aqueous buffer or water, building blocks, organic solvent, surfactants and initiators in the appropriate ratios to yield a stable phase of surfactant-stabilized aqueous nanodroplets dispersed in a continuous oil phase.
  • Stable reverse microemulsion formulations can be found using known methods by those skilled in the art. They are discussed, for example, in
  • an aqueous phase with solubilized hydrophilic building blocks is added to an organic solvent containing one or more solubilized surfactants to form a reverse microemulsion.
  • the dispersed aqueous phase contains hydrophilic building blocks solubilized at about 5 to about 65 wt%, preferably about 5 to about 25 wt%, most preferably 10 to 20 wt%.
  • hydrophilic building blocks solubilized at about 5 to about 65 wt%, preferably about 5 to about 25 wt%, most preferably 10 to 20 wt%.
  • the high water content also provides compliancy through a more flexible scaffold.
  • the articles when attaching to cell surface receptors, the articles are able to conform to the cell surface, allowing more surface receptors to be bound. Binding more receptors may allow the article to better function as an antagonist.
  • article cell surface coverage can inhibit other cell signaling pathways.
  • Polymerization of the building blocks in the nanodroplets of the dispersed aqueous phase of the reverse microemulsion follows procedures known to those skilled in the art (see, for example, Odian G.G.; Principles of Polymerization, 3 rd Ed., Wiley, New York, 1991 ; L.H. Sperling, Introduction to Physical Polymer Science, Chapter 1 , pp. 1-21 , John Wiley and Sons, New York, 1986; and R.B. Seymour and C.E. Carraher, Polymer Chemistry, Chapters 7-11 , pp. 193-356, Dekker, New York, 1981).
  • the size of the nanodroplets of the dispersed aqueous phase is determined by the relative amounts of water, surfactant and oil phases employed.
  • Surfactants are utilized to stabilize the reverse microemulsion. These surfactants do not include crosslinkable moieties; they are not building blocks.
  • Surfactants that may be used include commercially available surfactants such as Aerosol OT (AOT), polyethyleneoxy(n)nonylphenol (IgepalTM, Rhodia Inc.
  • sorbitan esters including sorbitan monooleate (Span ® 80), sorbitan monolaurate (Span ® 20), sorbitan monopalmitate (Span ® 40), sorbitan monostearate (Span ® 60), sorbitan trioleate (Span ® 85), and sorbitan tristearate (Span ® 65), which are available, for example, from Sigma (St Louis, MO).
  • Sorbitan sesquioleate (Span ® 83) is available from Aldrich Chemical Co., Inc. (Milwaukee, Wl).
  • Other surfactants that may be used include polyoxyethylenesorbitan (Tween ® ) compounds.
  • cosurfactants include polyoxyethylenesorbitan monolaurate
  • Tween ® 20 and Tween ® 21 polyoxyethylenesorbitan monooleate (Tween ® 80 and Tween ® 80R), polyoxyethylenesorbitan monopalmitate (Tween ® 40), polyoxyethylenesorbitan monostearate (Tween ® 60 and Tween ® 61), polyoxyethylenesorbitan trioleate (Tween ® 85), and polyoxyethylenesorbitan tristearate (Tween ® 65), which are available, for example, from Sigma (St Louis, MO).
  • exemplary commercially available surfactants include polyethyleneoxy(40)-sorbitol hexaoleate ester (Atlas G-1086, ICI Specialties, Wilmington DE), hexadecyltrimethylammonium bromide (CTAB, Aldrich), and linear alkylbenzene sulfonates (LAS, Ashland Chemical Co., Columbus, OH).
  • exemplary surfactants include fatty acid soaps, alkyl phosphates and dialkylphosphates, alkyl sulfates, alkyl sulfonates, primary amine salts, secondary amine salts, tertiary amine salts, quaternary amine salts, n-alkyl xanthates, n-alkyl ethoxylated sulfates, dialkyl sulfosuccinate salts, n-alkyl dimethyl betaines, n-alkyl phenyl polyoxyethylene ethers, n-alkyl polyoxyethylene ethers, sorbitan esters, polyethyleneoxy sorbitan esters, sorbitol esters and polyethyleneoxy sorbitol esters.
  • lipids such as phospholipids, glycolipids, cholesterol and cholesterol derivatives.
  • exemplary lipids include fatty acids or molecules comprising fatty acids, wherein the fatty acids include, for example, palmitate, oleate, laurate, yristate, stearate, arachidate, behenate, lignocerate, palmitoleate, linoleate, linolenate, and arachidonate, and salts thereof such as sodium salts.
  • the fatty acids may be modified, for example, by conversion of the acid functionality to a sulfonate by a coupling reaction to a small molecule containing that moiety, or by other functional group conversions known to those skilled in the art.
  • polyvinyl alcohol PVA
  • polyvinylpirolidone PVP
  • starch and their derivatives may find use as surfactants in the present invention.
  • Cationic lipids may be used as cosurfactants, such as cetyl trimethylammonium bromide/chloride (CTAB/CTAC), dioctadecyl dimethyl ammonium bromide/chloride (DODAB/DODAC), 1 ,2-diacyl-3-trimethylammonium propane (DOTAP), 1 ,2-diacyl-3- dimethyl ammonium propane (DODAP), [2,3-bis(oleoyl)propyl] trimethyl ammonium chloride (DOTMA), and [N-(N'-dimethylaminoethane)-carbamoyl]cholesterol, dioleoyl) (DC-Choi).
  • CTAB/CTAC cetyl trimethylammonium bromide/chloride
  • DODAB/DODAC dioctadecyl dimethyl ammonium bromide/chloride
  • DOTAP 1,2-diacyl-3-trimethylammonium propane
  • Alcohols may also be used as cosurfactants, such as propanol, butanol, pentanol, hexanol, heptanol and octanol. Other alcohols with longer carbon chains may also be used.
  • cosurfactants such as propanol, butanol, pentanol, hexanol, heptanol and octanol. Other alcohols with longer carbon chains may also be used.
  • Incorporation of Drug Molecules into Articles The terms “drug”, “drug- conjugate”, “bioactive agent” and “therapeutic agent” are used interchangeably herein.
  • drug molecules are covalently linked to the article scaffold.
  • the chemotherapeutic doxorubicin may be attached to the scaffold through a EDC coupling reaction between the amine moiety on doxorubicin and a carboxylic acid moiety included in the hydrogel scaffold, for example by using sodium acrylate (NaA), malonate acrylamide (MalAc) or CiBA as a building block.
  • doxorubicin may be attached via an imine bond by reacting doxorubicin's amine moiety with an aldehyde moiety of the hydrogel scaffold.
  • An aldehyde may be created by first using a carbohydrate- based building block to form the article, and then oxidizing the carbohydrate after the article is formed.
  • doxorubicin may be attached to the article matrix through its ketone moiety.
  • Carbohydrazide or other dihydrazide or di-amino-oxy functionalized structures may be used to link doxorubicin to a scaffold that contains an aldehyde or ketone through the formation of a hydrazone bond.
  • An aldehyde or ketone may be incorporated into the scaffold through the use of a ketone-containing acrylate building block such as DAA.
  • a hydrazone bond may favorably release the therapeutic compound under the mildly acidic physiological conditions encountered upon article endocytosis and entrance into lysosomes.
  • nano-article scaffolds comprised of amino groups, for example through the inclusion of APMA or methacrylate-functionalized short peptide (prepared according to US patent 5,037,883) building blocks, may be used to covalently attach cyclosporins that contain carboxylate linkages.
  • Cyclosporin drugs may find applications for pathologies that benefit from immunosuppression, such as inflammatory diseases, and for organ transplantation.
  • nano-article scaffolds comprised of aldehyde or ketone groups (for example incorporated through the inclusion of DAA, levulinic acrylamide or oxidized carboxylates such as inulin or dextran building blocks) may be used to covalently attach drugs or drug derivatives that contain a moiety, for instance calicheamicin, through the use of a hydrazone coupling scheme.
  • This coupling scheme results in a hydrazone bond that is hydrolytically labile, especially at low pH found in lysosomes (Bernstein I., et al., Bioconjugate Chem., 2002, 13, 40-46).
  • nano-article scaffolds comprised of acid or anhydride groups may be used to covalently attach dexamethasone, through the use of an amide coupling scheme.
  • Nano-article scaffolds comprised of carboxylate groups may be used to covalently attach drugs or drug derivatives that contain an amine moiety, for instance peptide-modified camptothecin (Frigerio E., et al., J. Controlled Release, 2000, 65, 105-119) through an EDC-NHS coupling scheme.
  • the nano-article scaffolds are comprised of aldehyde or ketone groups (which may be incorporated through the use of DAA, levulinic acrylamide or oxidized carboxylates such as inulin or dextran), a drug or drug derivative possessing an amine, such as gemcitabine, may be incorporated through the use of a "Schiff base" coupling scheme.
  • the imide bond formed from the attachment of gemcitabine to DAA can be cleaved in acidic media.
  • the drug is taken up by the cell, where it is exposed to the acidic environment of the lysosome, thereby releasing gemcitabine in its unmodified form.
  • nano-article scaffolds comprised of carboxyl groups may be used to covalently attach drugs or drug derivatives that contain a moiety, for instance salicylic acid, through the use of an EDC-NHS coupling scheme.
  • drugs or drug derivatives that contain a moiety for instance salicylic acid
  • EDC-NHS coupling scheme for instance, the hydroxyl group of salicylic acid will react with the carboxyl group of the CiBA, MalAc or NaA to form an ester link.
  • Hydrolysis or the enzyme esterase will cleave the ester bond between salicylic acid and the carboxylic acid groups of CiBA or NaA, releasing salicylic acid in an unmodified form.
  • a drug structure may be modified to facilitate attachment to a nano-article scaffold.
  • the 2' hydroxyl group of paclitaxel can be reacted with multiple linkers that enable the coupling to nano-article scaffolds.
  • the acid moiety of a resin-immobilized glycine linker can be attached to paclitaxel using a carbodiimide; the resulting compound can be cleaved at the site of the amine using 1% TFA, producing a free amine which can be conjugated with nano-articles possessing carboxylates using an EDC coupling scheme.
  • paclitaxel-2'-succinate (Deutsch H., et al., J Med. Chem., 1989, 32, 788-792) conjugation to the nano-article is possible using a carbodiimide- mediated amide coupling. This coupling occurs between the paclitaxel-2'-succinate group and an amine group of the APMA component of the nano-article to form a labile ester.
  • the nano-article can be directly coupled to paclitaxel by reacting the acid-functionalized (NaA) nano-article to the 2'-hydroxyl group of paclitaxel. This chemical pathway has been previously described using a poly(L-glutamic acid)- paclitaxel conjugate (Li H., et al., Cancer Res., 1998, 58, 2404-2409).
  • nano-article scaffolds containing carboxylic acids may be used to covalently attach drugs or drug derivatives that contain a moiety, for instance 5- flourouracil (5FU) (or derivatives) through the use of an amide forming coupling reaction between an amine-functionalized 5FU derivative and the carboxylic acids located on the nano-article.
  • 5FU 5- flourouracil
  • the synthesis of 1 -alkylcarbonyloxymethyl derivatives of 5FU has been previously described and those materials have been demonstrated to release 5FU in an unmodified form (Taylor H.E.; Sloan K.B., Journal of Pharmaceutical Sciences, 1998, 87, 15).
  • Nano-article scaffolds comprised of acid groups (for example incorporated through the inclusion of CiBA, MalAc and NaA building blocks) may be used to covalently attach drugs or drug derivatives that contain a carboxylate moiety, for instance methotrexate, by first coupling the drug or drug derivative to boc-protected ethanolamine to form an ester, and then coupling to the nano-article through an EDC coupling scheme after deprotecting the modified drug.
  • This ester conjugate is known to hydrolyze at low pH, releasing the drug in its original form (Wilson J.M., et al., Biochem Biophys. Res.
  • Such conditions of low pH are found in cellular lysosomes.
  • These nano-articles may find use in the treatment of multiple pathologies, including cancer and inflammatory conditions such as rheumatoid arthritis and inflammatory bowel disease.
  • platinum conjugates are incorporated onto nano-article scaffolds.
  • the platinum may be in the II nd or IV th oxidation state.
  • Such chelates should be highly water-soluble and stable in an aqueous environment, but sufficiently labile in tumor cells to provide species capable of crosslinking DNA and ultimately causing tumor cell death.
  • the hydrogel network of the nano-articles of the present invention combined with the capacity to fine tune its chemical composition allows for such a high water solubility and flexibility in the way to attach chelates, which constitute a significant improvement over WO 9847537.
  • the platinum is complexed to the hydrogel matrix via O,N-ligation, which is expected to yield a more stable compound.
  • O,N-ligation which is expected to yield a more stable compound.
  • This can be accomplished preferentially for nano-articles obtained by free-radical polymerization, and containing a combination of acid functions (such as from NaA) and amines (such as from APMA) or amides moieties (such as acrylamide), or building blocks carrying both types of functions such as CIBA, MalAc or methacryloylate-functionalized short peptides made according to US 5,037,883.
  • Such moieties provide attachment points to generate a O,N-cis platinum nanoarticle conjugate, leaving open the possibility of targeting the nano-article.
  • Drugs that may find use in the present invention include those that act on the peripheral nerves, adrenergic receptors, cholinergic receptors, nervous system, skeletal muscles, cardiovascular system, smooth muscles, blood circulatory system, synaptic sites, neuro-effector junctional sites, endocrine system, hormone systems, immunological system, reproductive system, skeletal system, autocoid systems, alimentary and excretory systems, histamine systems, respiratory system, reticuloendothelial system, skeletal system, skeletal muscles, smooth muscles, immunological system, reproductive system, cancerous tissues, and the like.
  • the active drug that can be delivered for acting on these recipients includes, but is not limited to, anticonvulsants, analgesics, anti-parkinsons, anti-inflammatories, calcium antagonists, anesthetics, antimicrobials, antimalarials, antiparasitics, antihypertensives, antihistamines, antipyretics, alpha-adrenergic agonists, alpha-blockers, biocides, bactericides, bronchial dilators, beta-adrenergic blocking drugs, contraceptives, chemotherpeutics, cardiovascular drugs, calcium channel inhibitors, depressants, diagnostics, diuretics, electrolytes, enzymes, hypnotics, hormones, hypoglycemics, hyperglycemics, muscle contractants, muscle relaxants, neoplasties, glycoproteins, nucleoproteins, lipoproteins, ophthalmics, psychic energizers, sedatives, steroids, sympathomimetics, parasy
  • drugs that may be advantageously employed in the present invention include, but are not limited to, chemotherapeutics such as doxorubicin, paclitaxel, gemcitibine, vincristine, cisplatin, carboplatin, chlorambucil, topotecan, methotrexate, derivatives of these compounds, and any other FDA-approved chemotherapeutic, as well as molecules that may act as chemotherapeutics but that are not yet commercialized, and derivatives and analogues of all of the above chemotherapeutics.
  • the therapeutic agent for delivery in this invention can be in various pharmaceutically acceptable forms, such as prodrugs, uncharged molecules, molecular complexes, and pharmacologically acceptable salts. Derivatives of medicines, such as esters, ethers and amides, can be used.
  • the article surface may be functionalized with REs.
  • the REs can be linked either directly or through a linker molecule to the surface of the nanoparticle. In a linker configuration, part or all of the REs are "displayed" at the end terminus of the tether. Therefore, in one application of the invention, the articles consist of REs displayed on a hydrogel scaffold. In another embodiment of the patent, the articles consist of an RE, such as a high affinity peptide, linked to the surface of the article core scaffold via a linker molecule, the linker comprising preferentially polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • the components may be attached to one another in the following sequences.
  • the hydrogel scaffold is first reacted with a di-functional PEG-containing tether, followed by functionalization of the free terminus of a portion of the PEG chain with a RE.
  • the RE is coupled first to the PEG-containing tether, followed by the attachment of the other PEG terminus to the scaffold.
  • the surfactants and the oil phase, residual from the synthesis of the hydrogel scaffold, can be removed through the use (singularly or in combination) of solvent washing, for instance using ethanol to solubilize the surfactant and oil while precipitating the articles; surfactant-adsorbing beads; dialysis; or the use of aqueous systems such as 4M urea.
  • solvent washing for instance using ethanol to solubilize the surfactant and oil while precipitating the articles; surfactant-adsorbing beads; dialysis; or the use of aqueous systems such as 4M urea.
  • Methods for surfactant removal are known in the art.
  • the RE must contain a functionality that allows its attachment to the article. Preferentially, although not necessarily, this functionality is one member of a pair of chemoselective reagents selected to aid the coupling reaction (Lemieux, G., Bertozzi, O, Trends in Biotechnology, 1998, 16, 506-513).
  • a peptide RE may be attached through a sulfhydryl moiety.
  • a sulfhydryl moiety in the RE structure can be accomplished through inclusion of a cysteine residue.
  • Coupling is also possible between a primary amine on the article or the linker terminus and a carboxylic acid on the RE.
  • a carboxylate in the peptide structure can be found either on its terminal amino acid, for linear peptides, or through the inclusion of aspartic or glutamic acid residues.
  • the opposite configuration, where the carboxylic acid is on the article and a primary amine belongs to the peptide, is also easily accessible.
  • Many polymerizable building blocks contain acidic moieties, which are accessible at the surface of the beads after their polymerization.
  • poly(amino acid)-based REs a primary amine function can be found either at its N-terminus (if it is linear) and/or via introduction of a lysine residue.
  • Another example of reactive chemical pairs consists of the coupling of a sulfhydryl with a halo acetal or maleimide moiety.
  • the maleimide function can be easily introduced, either on a peptide, a linker, or the surface of the articles, by reacting other common functionalities (such as carboxylic acids, amines, thiols or alcohols) with linkers through methods known to one of skill in the art, such as described for example by G. T. Hermanson in Bioconjugate Techniques, Academic Press Ed., 1996.
  • the inclusion of CiBA, or other disulfide-containing building blocks, in the scaffold facilitates the attachment of REs through thiol reactive moieties.
  • Linker molecules containing groups that are reactive with thiol such as bromoacetamide or maleimide, are added to the reduced therapeutic agent-containing article to attach the linker to the article scaffold.
  • REs are then added, which react with the free terminus of the linker molecules to give RE-functionalized articles.
  • the RE may be attached to one end of the linker molecule prior to attachment of the linker molecule to the reduced article.
  • Peptides can also be coupled to the article and/or the tether with a reaction between an amino-oxy function and an aldehyde or ketone moiety.
  • amino-oxy moiety (either on the articles or in the peptide) can be introduced, starting from other common functionalities (such as amines for example), by a series of transformations known to those skilled in the art.
  • aldehyde- or ketone-containing articles and aldehyde-containing peptides are readily synthesized by known methods.
  • the resulting RE-functionalized, drug-containing articles may be used immediately, may be stored as a liquid solution, or may be lyophilized for long-term storage.
  • the REs may be any small or large molecular structure that provides the desired binding interaction(s) with the cell surface receptors of the targeted molecule.
  • the number of recognition element moieties per article can range from 2 to about 1000, preferably from 2 to 500, and most preferably from 2 to 100.
  • the articles may optionally further be comprised of more than one type of RE.
  • a RE "type" is defined as a specific molecular structure.
  • REs preferably are comprised of peptides.
  • Peptides used as REs according to this invention will generally possess dissociation constants between 10 "4 and 10 ⁇ 9 M or lower.
  • Such REs may be comprised of known peptide ligands.
  • Phoenix Peptides' peptide ligand-receptor library http.V/www.phoenixpeptide.com/Peptidelibrarylist.htm
  • Phoenix Peptides' peptide ligand-receptor library http.V/www.phoenixpeptide.com/Peptidelibrarylist.htm
  • the peptides may be natural peptides such as, for example, lactams, dalargin and other enkaphalins, endorphins, angiotensin II, gonadotropin releasing hormone, melanocyte- stimulating hormone, thrombin receptor fragment, myelin, and antigenic peptides.
  • Peptide building blocks useful in this invention may be discovered via high throughput screening of peptide libraries (e.g. phage display libraries or libraries of linear sequences displayed on beads) to a protein of interest. Such screening methods are known in the art (for example, see OF. Barbas, D. R. Burton, J. K. Scott, G. J. Silverman, Phage Display, 2001 , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY).
  • the high affinity peptides may be comprised of naturally occuring amino acids, modified amino acids or completely synthetic amino acids.
  • the length of the recognition portion of the peptide can vary from about 3 to about 100 amino acids.
  • the recognition portion of the peptide ranges from about 3 to about 15 amino acids, and more preferably from 3 to 10 amino acids. Shorter sequences are preferred because peptides of less than 15 amino acids may be less immunogenic compared to longer peptide sequences. Small peptides have the additional advantage that their libraries can be rapidly screened. Also, they may be more easily synthesized using solid-state techniques.
  • REs may comprise a variety of other molecular structures, including antibodies, antibody fragments, lectins, nucleic acids, and other receptor ligands. Humanized or fully human antibodies, and humanized or fully human antibody fragments are preferred for use in the present invention.
  • non-protein compounds to be employed as the binding moiety, using techniques known to those working in the area of drug design.
  • Such methods include, but are not limited to, self-consistent field (SCF) analysis, configuration interaction (Cl) analysis, and normal mode dynamics computer programs, all of which are well described in the scientific literature.
  • SCF self-consistent field
  • Cl configuration interaction
  • Preparation of non-protein compounds and moieties will depend on their structure and other characteristics and may normally be achieved by standard chemical synthesis techniques. See, for example, Methods in Carbohydrate Chemistry, Vols. I-VII; Analysis and Preparation of Sugars, Whistler et al., Eds., Academic Press, Inc., Orlando (1962), the disclosures of which are incorporated herein by reference.
  • the use of multiple RE molecules of the same molecular structure or of different molecular structure to make up the article can increase the avidity of the article.
  • "high affinity” means a binding of a single RE to a single target molecule with a binding constant stronger than 10 "4 M, while “avidity” means the binding of two or more such RE units to two or more target molecules on a cell or molecular complex.
  • the REs can target a multitude of disease-associated biomolecules. Tumor- associated targets include erbB1 (for example using the growth factor EGF, or using peptides comprised of the amino acid sequence YCPIWKFPDEECY, or other sequences found in Greene, et.al., J. Biol.
  • Vascular targets associated with multiple pathologies, including cancer include VEGFR-1 , VEGFR-2 (for example, using peptides comprised of the amino acid sequence ATWLPPR, as described in Deplace, et.al., EMBO J., 2000, 19(7), 1525-1533), integrins, including integrin ⁇ v ⁇ 3, and integrin ⁇ v ⁇ l , and to extracellular proteins such as fibrin (which may be targeted using peptides comprised of amino acid sequences disclosed in PCT Publication WO 02/055544).
  • Article Attributes As practiced in the invention, the articles have several attributes that make them excellent therapeutic candidates.
  • the articles can be administered by injection (subcutaneous, intravenous, intramuscular, intradermal, intraperitoneal, intracerebral, or parenteral), with intraveneous injection being a preferred route.
  • the articles may also be suitable for nasal, pulmonary, vaginal, ocular delivery and oral administration.
  • the articles may be suspended in a pharmaceutically acceptable carrier for administration.
  • Reagents and starting materials in some embodiments can be obtained commercially from chemical distributors such as Sigma-Aldrich (St Louise, MO and Milwaukee, Wl), Kodak (Rochester, NY), Fisher (Pittsburgh, PA), Pierce Chemical Company (Rockford, IL), Carbomer Inc. (Westborough, MA), Radcure (Smyrna, GA), and Polysciences (Niles, IL).
  • PEG compounds may be purchased through NOF America Corporation (White Plains, NY), and Nektar (Birmingham, AL).
  • Peptides to be used as REs can be purchased from many sources, one being Bachem (King of Prussia, PA). Proteins may be obtained from sources such as Calbiochem (San Diego, CA).
  • Inulin (4 g) was weighed into a 1 neck round bottom flask. A Teflon-coated stir bar was added and the flask was sealed with a septa. Anhydrous pyridine (approximately 20 mL for each 5 grams of inulin) was then transferred into the flask, keeping the entire system under a blanket of nitrogen. The mixture was stirred until the inulin dissolved, then 1 mL of methacrylic anhydride was added dropwise, using a syringe. The reaction was allowed to continue for 16 hours. After that interval, enough pyridine was removed under vacuum to make a viscous liquid. Toluene (approximately 40 mL) was then added with vigorous mixing to precipitate the crude product.
  • the liquid was then decanted from the precipitate.
  • the solid was dissolved with water, producing a viscous, but free flowing, syrup, which was then precipitated with 2-propanol (approximately 200 mL). This process of water dissolution followed by precipitation was repeated twice more, after which the product was dried under vacuum.
  • the product was then redissolved in 50 mL of water and filtered through a filter paper to remove accumulated dust and other insolubles, and the product was then lyophilized to give inulin multi-methacrylate (IMMA).
  • IMMA identity degree of methacrylate functionalization
  • Diethylaminomalonate hydrochloride (5 g) is weighed into a 1 neck round bottom flask. Add a Teflon-coated stir bar and seal the flask using a septa. Anhydrous dichloromethane (approximately 10 mL for each gram of diethylaminomalonate) is then transferred into the flask, keeping the entire system under a blanket of nitrogen. The mixture is stirred, 3.62 mL triethylamine is added (1.1 eq.) with a syringe, followed by a dropwise addition of 2.12 ml of acryloyl chloride (1.1 eq.) with a syringe. The reaction is allowed to continue for 2 hours.
  • the reaction mixture is extracted three times with water to remove the unreacted products [triethylamine and acrylic acid].
  • the organic phase is dried under vacuum and resuspended in water containing resin with strong acidic residues.
  • the aqueous resin suspension is put on a rotary shaker and the deprotection reaction is allowed to proceed overnight.
  • the reaction mixture is then filtered and lyophilized to yield malonate acrylamide.
  • phosphate buffer (0.15 M)
  • 3-mercaptopropionic acid (0.17 g) in water (10 mL) was added with rapid mixing provided by a stir bar.
  • the pH was adjusted to 8 again by the addition of a 1.0 M NaOH solution. Sixteen hours after the addition, the volume of the reaction was reduced under vacuum to 50 mL.
  • the pH of the solution was adjusted to 2 by adding 1.0 M HCI, and the solution was extracted again with chloroform (3 times 50 mL).
  • Example 8 IMMA-CiBA-NaA scaffold formation via free radical polymerization
  • An aqueous phase was prepared by combining 83 wt% buffer, 14 wt% IMMA, 2 wt% CiBA, 1 wt% sodium acrylate, and Eosin Y (the photoinitiator represents from 0.001 to 0.1 wt% of the monomers mass).
  • An oil+surfactant phase was prepared by mixing 7.3 wt% Igepal CO-210, 9.4 wt% Igepal CO-720, and 83.3 wt% cyclohexane. Three grams (3g) of the aqueous phase was added to 30g of the oil+surfactant phase with vigorous stirring, resulting in the formation of a reverse microemulsion.
  • the reverse microemulsion contains surfactant-stabilized nano-droplets of aqueous phase dispersed in a continuous phase of cyclohexane.
  • the reverse microemulsion was transferred to a 100 mL Schlenk tube and was degassed by briefly pulling a vacuum on the chilled mixture. The contents of the
  • Example 9 IMMA-CiBA-DAA scaffold formation through free radical polymerization An aqueous phase was prepared by combining 83 wt% water, 14 wt% IMMA, 2 wt %
  • An oil+surfactant phase was prepared by mixing Igepal CO-210, Igepal CO-720 and cyclohexane in a weight ratio of 1.0:1.3:9.0. Three grams (3g) of the aqueous phase were mixed with 30g of the oil+surfactant phase, resulting in the formation of a reverse microemulsion.
  • the reverse microemulsion contained surfactant-stabilized nano-droplets of aqueous phase dispersed in a continuous phase of cyclohexane.
  • aqueous solution containing Eosin Y where the photoinitiator represented from 0.001 to 0.1 wt% of the monomers mass.
  • the reverse microemulsion was degassed with freeze-thawing cycles under vacuum, with N 2 gas backfill between cycles. The contents were stirred and irradiated with a UV or visible light source of at least 100 W for 20 min to two hours to polymerize the building blocks.
  • the nanoarticles were precipitated by adding 9 mL of pure ethanol directly to the solution.
  • the nanoarticle-containing pellets were resuspended in water. Residual surfactants and solvents were removed by standard techniques (dialysis, chromatography, etc.). At this point, the aqueous solution of nano- articles may be lyophilized, if desired.
  • An aqueous phase was prepared by combining 83 wt% buffer, 14 wt% IMMA, 2 wt% CiBA, 1 wt% N-(3-aminopropyl)methacrylamide hydrochloride (APMA), and Eosin Y (the photoinitiator represents from 0.001 to 0.1 wt% of the monomers mass).
  • An oil+surfactant phase was prepared by mixing 7.3 wt% Igepal CO-210, 9.4 wt% Igepal CO-720, and 83.3 wt% cyclohexane. Three grams (3g) of the aqueous phase was added to 30g of the oil+surfactant phase with vigorous stirring, resulting in the formation of a reverse microemulsion.
  • the reverse microemulsion contains surfactant-stabilized nano-droplets of aqueous phase dispersed in a continuous phase of cyclohexane.
  • the reverse microemulsion was transferred to a 100 mL Schlenk tube and was degassed by briefly pulling a vacuum on the chilled mixture. The contents of the Schlenk tube were stirred and irradiated with a UV light source for one hour to polymerize the building blocks. Once the polymerization was complete, the nanoarticles were precipitated by adding pure ethanol directly to the solution and were isolated from the reaction mixture by centrifugation. The nanoarticle-containing pellet was resuspended in deionized water. Residual surfactants and solvents were removed by standard techniques (solid phase extraction). At this point, the aqueous solution of nanoarticles were filtered and the product was isolated as a solid product after lyophilization.
  • Example 11 IMMA-MalAc scaffold formation via free radical polymerization
  • An aqueous phase was prepared by combining 80 wt% buffer, 14 wt% IMMA, 6 wt% malonate acrylamide (MalAc) and Eosin Y (the photoinitiator represents from 0.001 to 0.1 wt% of the monomers mass).
  • An oil+surfactant phase was prepared by mixing 7.3 wt% Igepal CO-210, 9.4 wt% Igepal CO-720, and 83.3 wt% cyclohexane. Three grams (3g) of the aqueous phase was added to 30g of the oil+surfactant phase with vigorous stirring, resulting in the formation of a reverse microemulsion.
  • the reverse microemulsion contains surfactant-stabilized nano-droplets of aqueous phase dispersed in a continuous phase of cyclohexane.
  • the reverse microemulsion was transferred to a 100 mL Schlenk tube and was degassed by briefly pulling a vacuum on the chilled mixture. The contents of the Schlenk tube were stirred and irradiated with a UV light source for one hour to polymerize the building blocks. Once the polymerization was complete, the nano-articles were precipitated by adding pure ethanol directly to the solution and were isolated from the reaction mixture by centrifugation. The nanoarticle-containing pellet was resuspended in deionized water. Residual surfactants and solvents were removed by standard techniques (solid phase extraction). At this point, the aqueous solution of nano-articles were filtered and the product was isolated as a lyophilized powder.
  • An aqueous phase was prepared by combining 82 wt% buffer, 14 wt% IMMA, 2 wt% CIBA and 2 wt% N-methacryloylated Gly-Gly peptide (made according to US 5,037,883) with Eosin Y (the photoinitiator represents from 0.001 to 0.1 wt% of the monomers mass).
  • An oil+surfactant phase was prepared by mixing 7.3 wt% Igepal CO-210, 9.4 wt% Igepal
  • the reverse microemulsion contains surfactant-stabilized nano-droplets of aqueous phase dispersed in a continuous phase of cyclohexane.
  • the reverse microemulsion was transferred to a 100 mL Schlenk tube and was degassed by briefly pulling a vacuum on the chilled mixture. The contents of the Schlenk tube were stirred and irradiated with a UV light source for one hour to polymerize the building blocks. Once the polymerization was complete, the nano-articles were precipitated by adding pure ethanol directly to the solution and were isolated from the reaction mixture by centrifugation.
  • the nanoarticle-containing pellet was resuspended in deionized water. Residual surfactants and solvents were removed by standard techniques (solid phase extraction). At this point, the aqueous solution of nanoarticles were filtered and the product was isolated as a lyophilized powder.
  • Example 13 Scaffold formation via reaction of oxidized inulin with a bis amino oxy compound
  • An aqueous phase was prepared by mixing together 85 wt% water and 15 wt% inulin.
  • An oil+surfactant phase was prepared by mixing Igepal CO-210, Igepal CO-720 and cyclohexane in a weight ratio of 1.0:1.3:9.0. Three grams (3g) of the aqueous phase were mixed with 40g of the oil+surfactant phase, resulting in the formation of a reverse microemulsion. One gram of a sodium periodate solution in water was added to the mixture. The in-situ oxidation was allowed to proceed for ca. 10 minutes.
  • a concentrated solution (of at least 1g/mL) of bis[(2-amino-oxy)ethylamido]-(1,3-)propane in an aqueous methanol mixture (50/50 volume) was then added to the microemulsion and allowed to react overnight.
  • the resulting nanoarticles obtained by the crosslinking of the bis[(2-amino- oxy)ethylamido]-(1 ,3)propane with the aldehydes functionalities of oxidized inulin were isolated by precipitation in presence of ethanol and centrifugation and purified from the excess of unreacted reagents by dialysis.
  • Example 14 Scaffold formation via reaction of oxidized dextran with carbo- hydrazide
  • An aqueous phase was prepared by combining 90 wt% water and 10 wt% dextran.
  • An oil+surfactant phase was prepared by mixing Igepal CO-210, Igepal CO-720 and cyclohexane in a weight ratio of 1.0:1.3:9.0. Three grams (3g) of the aqueous phase were mixed with 40g of the oil+surfactant phase, resulting in the formation of a reverse microemulsion. One gram of a sodium periodate solution in water was added to the mixture, so that there was a maximum of one periodate equivalent per glucose monomer unit (from the dextran). The in-situ oxidation proceeded for 5 to 20 minutes.
  • An aqueous phase was prepared by mixing together 85 wt% water and 15 wt% inulin. Two grams (2g) of this aqueous phase were mixed with 30g of an oil+surfactant phase composed of 8.5 wt% Igepal CO-520 in cyclohexane. One gram of a sodium periodate solution in water was added to the mixture, so that there was a maximum of one periodate equivalent per glucose monomer unit (from the inulin). The in-situ oxidation was allowed to proceed for ca. 10 minutes.
  • a concentrated solution of succinic dihydrazine in an aqueous methanol mixture (50/50 volume) was added to the microemulsion and allowed to react for three hours, crosslinking the amino-oxy moieties with the aldehydes functions.
  • the resulting nano-articles were purified by precipitation and resuspended in aqueous solution in the presence of an excess of succinic dihydrazine.
  • the reaction of succinic dihydrazine (with remaining aldehydes on the nanoarticles) was allowed to proceed for 12 hours.
  • Doxorubicin 120 mg was completely dissolved in 15.0 mL water, and 15.0 mL of HEPES buffer, pH 7.5 (0.20 M) was added.
  • HEPES buffer pH 7.5 (0.20 M
  • 160 mg of N-hydroxysuccinimide (NHS) and 50 mg 1-(3-dimethylaminopropyl)-3- ethylcarbodiimide hydrochloride (EDC) were added in succession. Twenty minutes after the addition of EDC, 7.5 mL of the doxorubicin solution was added.
  • a second 50 mg aliquot of EDC was added, followed by a twenty minute delay before the addition of a second quantity of doxorubicin. After another 0.5 hour reaction interval, the addition/delay cycle was repeated twice more so that the entire doxorubicin solution was added. The reaction mixture was then stirred for 24 hours. The unreacted starting materials and side products were separated from the Dox-nano-article product by passage through a HiPrep 26/10 desalting column, and the nanoarticles were isolated as a dry solid by lyophilization. The Dox loading of the resulting articles was approximately 6 to 8 wt%. The doxorubicin content of the nanoarticles can be varied by using smaller quantities of this material during the additions or by decreasing the total number of reaction cycles.
  • Example 17 Doxorubicin attachment via hydrazone linkage (high loading)
  • Example 9 1.0 gram of material from Example 9 was solubilized in 40 mL of 0.1 M ammonium formate, pH 8.5, and added to a concentrated solution of carbohydrazide in excess (100 eq. carbohydrazide to each diacetone acrylamide in the starting material). The reaction was allowed to proceed for 12 hours at 4°C. An hydrazone bond was thus formed between the carboxyhydrazide and the ketone moieties of the nanoarticle. The reaction mixture was purified of the excess carbohydrazide using dialysis or size exclusion chromatography.
  • doxorubicin 200 mg were added to the carbohydrazide- functionalized scaffolds in water buffered by ammonium formate. The reaction was allowed to proceed for 18 to 24 hours at 4°C. A hydrazone bond was thus formed between the ketone moieties on doxorubicin and the carbohydrazide linked to the nano-article. The resulting nano-articles with covalently attached doxorubicin were then purified from unreacted doxorubicin and side products using dialysis or size exclusion chromatography.
  • Example 18 Doxorurubicin attachment via hydrazone linkage (lower loading)
  • Example 9 1.0 gram of material from Example 9 was solubilized in 40 mL of 0.1 M potassium borate buffer, pH 8, and added to a concentrated solution of carbohydrazide in excess (10 eq. carbohydrazide to each diacetone acrylamide in the starting material). The reaction was allowed to proceed for 1 hour. The reaction mixture was purified of the excess carbohydrazide using dialysis or size exclusion chromatography. An hydrazone bond was thus formed between the carboxyhydrazide and the ketone moieties of the nano-article. 50 mg of doxorubicin, diluted in 20 mL water, were added to the carbohydrazide- functionalized nanoarticle in water buffered by ammonium formate.
  • Cyclosporin is mixed with 4-benzoylbenzoic acid (BBa) at a molar ratio of 1 to 2 in benzene.
  • the solution is purged with nitrogen gas and photolysed at a wavelength of 320 nm at room temperature (US patent 5,405,785).
  • benzene is evaporated in a rotary evaporator under vacuum and the dried product is dissolved in methanol.
  • the product is isolated by preparative HPLC.
  • Cyclosporin-BBa is then added to a solution of N- hydroxysuccinimide (NHS) and dicyclohexylcarbodiimide at a molar ratio of approximately 1 to 1.2 to 1 in methanol. The reaction is allowed to run overnight at room temperature.
  • NHS N- hydroxysuccinimide
  • dicyclohexylcarbodiimide dicyclohexylcarbodiimide
  • Activated ester formation can be detected with a neutral Fe-hydroxamate test.
  • the cyclosporin-BBa-NHS can be coupled to a nano-article scaffold comprised of amine moieties (the fabrication of which is described in Example 13) using EDC coupling.
  • Example 20 Calicheamicin attachment via a hydrazone bond
  • N-acetyl-gamma calicheamicin dimethyl hydrazide The synthesis of the calicheamicin derivative, N-acetyl-gamma calicheamicin dimethyl hydrazide has previously been reported (Upeslacis J., et al., Cancer Res., 1993, 53, 3336-3342).
  • 1.0 gram of IMMA-CiBA-DAA scaffold nano-article is solubilized in 40 mL of 0.1 M ammonium formate, pH 8.5 and added to a concentrated solution of N-acetyl- gamma calicheamicin dimethyl hydrazide in excess (100 eq. N-acetyl-gamma calicheamicin dimethyl hydrazide to each diacetone acrylamide in the starting material).
  • the reaction is allowed to proceed for 12 hours at 4°C.
  • a hydrazone bond is thus formed between the N- acetyl-gamma calicheamicin dimethyl hydrazide and ketone moieties of the nano-article.
  • the reaction mixture is purified of excess N-acetyl-gamma calicheamicin dimethyl hydrazide using a 26/10 desalting column.
  • Dexamethasone conjugation to the nano-article is possible via a glycine- functionalized linker.
  • This linker compound is prepared through the use of solid phase synthetic techniques via glycine-functionalized 2-CITrityl resins (2002/2003 NovaBiochem Catalog, page 2.16-2.17, CalBiochem-NovaBiochem Corp., San Diego, USA).
  • the acid moiety of the glycine linker is reacted with the 21 carbon hydroxyl group of dexamethasone using a carbodiimide.
  • An excess of dexamethasone is employed to react all acid groups of the glycine linker.
  • the resulting compound is cleaved at the site of the amine using 1% trifluoroacetic acid (TFA).
  • TFA trifluoroacetic acid
  • Example 9 Dissolve nano-articles (1.0 g) of Example 9 (and preferably containing DAA or oxidized sugars) in 10.0 mL PBS buffer, pH 7.2 (0.2 M). Add 15.5 mg of gemcitabine to the nano-article solution (at 35°C). React for 1 hour before isolating nano-articles via size exclusion chromatography. The aqueous solution of nano-articles can be lyophilized at this point.
  • Methotrexate (100 mg, 0.22 mmol) is dissolved in dimethyl sulfoxide (DMSO) (10 mL) with addition of boc-protected ethanolamine (100 mg, 0.44 mmol), dicyclohexylcarbodiimide (DCC) (91 mg, 0.44 mmol) and 4-dimethylaminopyridine (DMAP) (36 mg, 0.293 mmol) at 0°C.
  • DMSO dimethyl sulfoxide
  • DCC dicyclohexylcarbodiimide
  • DMAP 4-dimethylaminopyridine
  • the boc-protected group is removed by dissolving the product in 50:50 mixture of methylene chloride and TFA and stirring for 3 hrs at room temperature. On evaporating the solvent, the deprotected amino-ester derivative is obtained (Minko T., et al., Cancer Chemother. Pharmacol., 2002, 50, 143-150).
  • the dried powder of the amino-ester derivative (100 mg) is completely dissolved in 2 mL DMSO.
  • a solution of nano-articles of Example 8 1.0 g
  • 10.0 mL HEPES buffer, pH 7.5 (0.20 M) is added 10.0 mL water.
  • the methotrexate solution is added followed by the addition of 160 mg of N-hydroxysuccinimide (NHS) and 50 mg 1- (3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC) in succession. After 30 minutes, a second 50 mg aliquot of EDC is added. After another 0.5 hour reaction interval, the addition/delay cycle is repeated twice more.
  • NHS N-hydroxysuccinimide
  • EDC 1- (3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
  • the reaction mixture is then stirred for 24 hours.
  • the unreacted starting materials and side products are separated from the methotrexate-nano-article product by passage through a HiPrep 26/10 desalting column, and the nano-articles are isolated as a dry solid by lyophilization.
  • succinic anhydride 400 mg is reacted with 2.0 g IMMA (prepared according to Example 1) in pyridine to a level of approximately 1 carboxylic acid moieties per 3 saccharide repeat groups of inulin and isolated according to the method described in Example 1.
  • 1.0 g of this acid modified IMMA (AM-IMMA) is then coupled to 300 mg of salicylic acid in DMF using 1,3-dicyclohexylcarbodiimide(DCC, 500 mg).
  • DCC 1,3-dicyclohexylcarbodiimide
  • the AM-IMMA is dissolved in anhydrous DMF (50 mL) and the DCC is added. After 4 hours, the salicylic acid is added and the reaction is stirred overnight. The following day, the reaction is filtered, and the DMF is removed under vacuum.
  • N- hydroxysuccinimide NHS
  • EDC 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
  • Twenty minutes after the addition of EDC 7.5 mL of the 20-O-peptidyl-camptothecin solution was added.
  • a second 50 mg aliquot of EDC was added, followed by a twenty-minute delay before the addition of a second quantity of 20-O-peptidyl-camptothecin.
  • the addition/delay cycle was repeated twice more so that the entire 20-O-peptidyl-camptothecin solution was added.
  • the reaction mixture was then stirred for 24 hours.
  • the unreacted starting materials and side products were separated from the 20-O-peptidyl-camptothecin nanoparticle product by passage through HiPrep 26/10 desalting column, and the nanoarticles were isolated as a dry solid by lyophilization.
  • Example 26 Cis-diamino-platinum N,O complexed with malonate-containing nanoarticles
  • a solution of 1.0 g nano-articles as described in Example 11 (composed of 80 wt% buffer, 14 wt% IMMA, 6 wt% MalAc, and Eosin Y), in 0.1 M sodium nitrate pH 7.4 is added 26 mL of a 50 mM solution of cis-diaminediaquaplatinum dinitrate in sodium nitrate.
  • the reaction is allowed to proceed for 12 hours at room temperature.
  • the use of nitrate favors the ligand exchange.
  • the O,O-platinum-malonate complex initially formed will rearrange itself into the more stable O,N-malonate complex (the transformation can be followed by Pt NMR).
  • the unreacted platinum derivatives are then separated from the platinum-complexed- nanoarticle product by passage through a HiPrep 26/10 desalting column.
  • the nanoarticles are isolated as a dry solid by lyophilization.
  • Example 27 Cis-diamino-platinum N,O complexed with the amide and acid moieties of CIBA
  • nanoarticles composed of 80 wt% buffer, 14 wt% IMMA, 6 wt% CiBA, and Eosin Y, in 0.1 M sodium nitrate pH 7.4 was added 18 mL of a 100 mM solution of cis-diaminediaquaplatinum dinitrate in sodium nitrate.
  • nitrate favors the ligands exchange.
  • the reaction was allowed to proceed overnight at room temperature.
  • the unreacted platinum derivatives as well as those too loosely complexed were separated from the platinum-complexed-nano-article product by passage through a HiPrep 26/10 desalting column.
  • the nano-articles were isolated as a dry solid by lyophilization.
  • Example 28 Targeted cis-platin nanoparticles [loading post targeting]
  • Nano-articles (composed of 82 wt% buffer, 14 wt% IMMA, 2 wt% CIBA and 2 wt% N-Methacryloylated Gly-Gly peptide, made according to US 5,037,883 with Eosin Y) made according to Example 12 were reduced following the procedure described in Example 30 (DTT reduction), functionalized with PEG dBA (Example 30), and targeted with an ErbB-2 ligand (bromoacetamide functionalized cyclic F[CDGFYAC]YMDV) as described in Example 37.
  • DTT reduction DTT reduction
  • PEG dBA Example 30
  • ErbB-2 ligand bromoacetamide functionalized cyclic F[CDGFYAC]YMDV
  • the chloro-methyl ester of Boc-protected glycine is attached to 5FU following the method of Taylor et. al. (Taylor H.E., Sloan K.B. Journal of Pharmaceutical Sciences, 1998, 87, 15). After deprotection with trifluoroacetic acid (TFA), and removal of excess TFA under vacuum, the amine-functionalized 5FU (made from 198 mg of the Boc-protected starting material) is used immediately for attachment to carboxylic acid containing nano-articles. This material is completely dissolved in 15.0 L water, and 15.0 mL of HEPES buffer, pH 7.5 (0.20 M) is added.
  • TFA trifluoroacetic acid
  • Example 8 To a solution of nano-articles of Example 8 (1.0 g), dissolved in 10.0 mL HEPES buffer, pH 7.5 (0.20 M), 10.0 mL of water is added. To this solution, 160 mg of N-hydroxysuccinimide (NHS) and 50 mg 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC) is added in succession. Twenty minutes after the addition of EDC, 7.5 mL of the amine functionalized 5FU solution is added. The resulting solution is stirred gently for 0.5 hour. A second 50 mg aliquot of EDC is added, followed by a twenty minute delay before the addition of a second quantity of the derivatized 5FU.
  • NHS N-hydroxysuccinimide
  • EDC 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
  • the addition/delay cycle is repeated twice more so that the entire derivatized 5FU solution is added.
  • the reaction mixture is then stirred for 24 hours.
  • the unreacted starting materials and side products are separated from the nano-article product by passage through a HiPrep 26/10 desalting column and the nano-articles are isolated as a dry solid by lyophilization.
  • FPLC Fast Performance Liquid Chromatography
  • the freshly purified nanoarticle solution was added to the PEG-1500 dBA, and the PEG attachment reaction was stirred gently for 2 hours at room temperature under aluminum foil.
  • the nanoarticles were then separated from the other components of the reaction mixture using the same FPLC arrangement described earlier.
  • the pooled fractions containing nanoarticles were used in subsequent procedures, outlined in following examples, in which specific targeting elements are attached.
  • the disulfide-bridged cyclic peptide CTCPIWKFPDEECY(174 m g) wa s added to the pooled fractions eluting off the FPLC that contained the nanoarticles described in Example 16. This reaction was stirred gently for 2 hours under aluminum foil. Separation of the nanoarticles from the other components in the reaction mixture was achieved by the use of an FPLC equipped with a HiPrep26/10 desalting column, using de-ionized water as the solvent. The nanoarticle products may be isolated by lyophilization.
  • the disulfide-bridged cyclic peptide C(dF)CDGF(dY)ACYMDV (the no tation dF representing the D isomer of phenylalanine and dY representing the D isomer of tyrosine, 162 mg) was added to the pooled fractions eluting off the FPLC that contained the nanoarticles described in Example 16. This reaction was stirred gently for 2 hours under aluminum foil. Separation of the nanoarticles from the other components in the reaction mixture was achieved by the use of an FPLC equipped with a HiPrep26/10 desalting column, using de-ionized water as the solvent. The nanoarticle products may be isolated by lyophilization.
  • the peptide ATWLPPRC 120 mg was added to the pooled fractions eluting off the FPLC that contained the nanoarticles described in Example 16. This reaction was stirred gently for 2 hours under aluminum foil. Separation of the nanoarticles from the other components in the reaction mixture was achieved by the use of an FPLC equipped with a HiPrep26/10 desalting column, using de-ionized water as the solvent. The nanoarticle products may be isolated by lyophilization.
  • Example 35 RGD attachment to PEG dBA functionalized articles
  • Cyclic RGD peptide (HAP3C, RGDdFC, or FW 576) (62 mg) was added to a solution containing 355 mg of doxorubicin-containing nanoarticles with attached PEG 400 dBA linker tethers (prepared by functionalizing Dox-articles of Example 17 with PEG 00 dBA following the procedures of Example 30) at 10 mg/mL in PBS, pH 8. The reaction was allowed to run for 2 hours, after which 13 mg of cysteine was added.
  • the reaction with cysteine was allowed to run for 1 hour, after which the nanoarticles were purified by FPLC with a volatile buffer, pH 7 to 9 as eluent to give doxorubicin-containing nanoarticles with RGD attached via PEG 400 dBA chains.
  • the nanoarticles were lyophilized.
  • doxorubicin-containing nanoarticles with attached PEG 0 odBA linker tethers prepared by functionalizing Dox- articles of Example 17 with PEG 400 dBA following the procedures of Example 30
  • the reaction was allowed to run for 4 hours, after which 11 mg of cysteine was added.
  • the reaction with cysteine was allowed to run for 1 hour, after which the nanoarticles were purified by FPLC to give doxorubicin-containing nanoarticles with Erb1 attached via PEG 40 odBA chains.
  • doxorubicin-containing nanoarticles with attached PEG 40 odBA linker tethers prepared by functionalizing Dox-articles of Example 17 with PEG 400 dBA following the procedures of Example 30
  • the reaction was allowed to run for 4 hours, after which 11 mg of cysteine was added.
  • the reaction was allowed to run for 2 hours, after which the nanoarticles were purified by FPLC to give doxorubicin- containing nanoarticles with ErB2 attached via PEG 0 odBA chains.
  • a cysteine end-terminated VEGFR-2 peptide (100 mg) was added to a solution containing 355 mg of doxorubicin-containing nanoarticles with attached PEG 00 dBA linker tethers (prepared by functionalizing Dox-articles of Example 17 with PEG 400 dBA following the procedures of Example 30) at 10 mg/mL in PBS, pH 8.
  • the reaction was allowed to run for 2 hours, after which 13 mg of cysteine was added.
  • the reaction was allowed to run for 1 hour, after which the nanoarticles were purified by FPLC to give doxorubicin-containing nanoarticles with VEGFR-2 attached via PEG 40 odBA chains.
  • mice Nine female SCID/Rag 2M mice (age 9 weeks, weight 18-22 g) were randomly grouped into 3 groups of three mice prior to study initiation. Articles were solubilized in saline solution and injected intravenously into the tail veins of the mice. Intravenous administration of nanoarticles at 100, 200, and 400 mg/kg was well tolerated with no acute or delayed signs of toxicity. Weights were monitored over a period of 16 days. During that time no weight loss was observed. Results are shown in Table 1 :
  • Example 40 PK of FITC-labeled NP matrix
  • FITC-labeled nanoparticles or FITC-labeled nanoparticles conjugated with PEG-400 (NP-PEG) were made using the reverse microemulsion polymerization method described in Example 8, where the aqueous phase was comprised of 83 wt% water, 14 wt% IMMA, 1 wt% APMA, and 1 wt % NOBA.
  • Fluorescein Isothiocyanate (FITC) was attached via linkage between the isothiocyanate of the fluorescein and the amine-containing moiety aminopropyl methacrylamide (APMA) on the nanoparticles.
  • mice Female Balb/c mice were injected intravenously with 50 mg/kg of FITC-labeled nanoparticles (NP) or FITC-labeled nanoparticles conjugated with PEG-400 (NP-PEG). The treatment groups were assessed at 15-minutes, 2-hours, 6-hours, and 24-hours. Blood and tissue samples were collected at the indicated time points. Tissue samples were snap frozen prior to further analysis. From the blood samples plasma was prepared via centrifugation at 1500g for 10 minutes. Plasma and tissues were assayed for the presence of NP or NP-PEG via a fluorescence assay using a microplate fluorescence reader at an excitation wavelength of 485 nm and emission wavelength of 535 nm.
  • Plasma samples were diluted 10-fold dilution with PBS. Final assayed volumes were 100 ⁇ l per well in black- bottom 96-well plates. Tissues were extracted in 2 ml of PBS containing 0.05% Triton X-100 using a tissue homogenizer. Then tissue samples were diluted 5-fold in PBS with a final volume of 100 ⁇ l per well in black-bottom 96-well plates. Plasma elimination of FITC-NP represented in ⁇ g of particles indicated a long half- life in the blood of 6.6 hours for both the bare (non-functionalized) nanoparticles and nanoparticles conjugated with PEG-400.
  • the biodistribution was analyzed by measuring the fluorescence in liver, spleen, kidneys, heart, and lungs. Low accumulation in the liver and spleen was detected for both types of nanoparticles from organ extracts taken at the 4 time points over 24 hours. No nanoparticles were found in the lungs, heart, and kidneys.
  • Example 41 CAM results for RGD-Dox-NPs
  • CAM chorioallantoic membranes
  • Dox-NP nano-article-conjugated doxorubicin
  • Dox-NP of composition 14/1/1 IMMA/CiBA/NaA, 5.6 w% Dox were made following procedures described in Example 16.
  • CL66 murine mammary carcinoma cells were grown to 80-90% confluence in supplemented media, trypsinized for 2 min at 37°C with 1X trypsin, rinsed with Hank's Balanced Salt Solution (w/out calcium and magnesium) and resuspended at 1 x 10 6 cell/ml, with 0.1 ml injected into the mammary fat pad of female Balb/C mice.
  • Dox-NP Lyophilized Dox-NP was reconstituted in sterile PBS at the appropriate concentration and stored at 2-8°C for at least 1 hour, or overnight. Excipient Dox was diluted in PBS at the appropriate concentrations. 200 ⁇ L was injected intravenously through the tail vein of the mice on days 13, 15, and 18 following tumor challenge. Dox was injected at 225 ⁇ g /animal, while Dox-NP was injected at a nano-article level sufficient to deliver Dox at 250, 420 and 560 ⁇ g/animal. Tumor volumes were measured superficially in two dimensions. Weight loss (or gain) and survival were recorded. A significant reduction in tumor size was observed with 250 ⁇ g/animal.
  • the higher doses of nanoparticles containing Dox were toxic, as was the 225 ⁇ g/animal dose of Dox, although the mice injected with Dox had a slightly longer survival time.
  • the lowest dose of Dox-NP had therapeutic activity, resulting in slower tumor growth and thus smaller tumors.
  • the tumors were smaller in the 250 ⁇ g/animal Dox-NP group and their weight was lower than those of the Dox group.
  • the smaller tumors the difference in survival was minimal (post tumor injection) between the 250 ⁇ g/animal Dox-NP and control mice. This suggests that Dox-NP had activity, which was reversed with continued tumor growth in the absence of additional therapy.
  • Example 43 In Vivo Toxicity/Efficacy data for RGD-Dox NPs and Dox NPs
  • Dox-NP and Dox- NP-cRGD The efficacy and toxicity of nano-article-conjugated doxorubicin (Dox-NP and Dox- NP-cRGD)) was studied.
  • Dox-NPs of composition 14/1/1 IMMA/CiBA/NaA, 5.6 w% Dox, PEG 40 o (with and without cRGD) were made following procedures described in Example 16.
  • PEG 0 odBA was linked following procedures described in Example 30.
  • Tumor-bearing mice were inoculated, treated, and observed as described in Example 42 (above). Intravenous injections were given 3 times a week for one week at cumulative doses of 1200 ⁇ g Dox equivalent for Dox-NP and Dox-NP-cRGD.
  • Doxorubicin may be attached to the nano-article scaffold through linkages comprised of covalent bonds that are degraded very slowly or faster in the intracellular environment. For instance, amide bonds are degraded more slowly, while other linkages that are more rapidly degraded through hydrolysis such as linkages comprised of hydrazone bonds.
  • the toxicity of Dox linked to the nano-articles via the amide bond was tested in an in vitro toxicity assay against C-32 cells (human melanoma).
  • Dox-nano-articles of composition 14/2/1 IMMA/CiBA/NaA, were made following procedures described in Example 8.
  • Example 45 In vitro toxicity of Dox-hydrazone linked nanoparticles
  • Dox-nano-articles of composition 14/2/1 IMMA/CiBA/DAA were made following procedures described in Example 9.
  • a 96-well tissue culture microtiter plate 10,000 cells of C-32 per well in culture medium were allowed to attach for 2 hours prior to incubation with varying concentrations of Dox-hydrazone nano-article, and free Dox.
  • Cells were incubated at 37°C, 5% CO 2 in a humidified chamber for 48 hours. Cell proliferation was assayed using the colorimetric MTT kit (CT01 , Chemicon, Int.). The 50% growth inhibition was determined to 0.07 ⁇ g/mL of Dox equivalent compared to 0.8 ⁇ g/mL of free Dox.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Nanotechnology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biophysics (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Medical Informatics (AREA)
  • Molecular Biology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Polysaccharides And Polysaccharide Derivatives (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

La présente invention concerne des articles polymères hydrophiles comprenant une structure polymère et un ou plusieurs agents thérapeutiques, tels que des molécules médicamenteuses ou de conjugués médicamenteux. Les articles peuvent en outre éventuellement comprendre des éléments de reconnaissance qui se fixent à des structures biomoléculaires exprimées sur certaines cellules ou sur certains tissus, pour faciliter le ciblage et/ou l'administration.
PCT/US2003/017536 2002-06-03 2003-06-03 Articles polymeres contenant des agents therapeutiques WO2003101425A2 (fr)

Priority Applications (5)

Application Number Priority Date Filing Date Title
AU2003273272A AU2003273272A1 (en) 2002-06-03 2003-06-03 Therapeutic agent-containing polymeric nanoarticles
EP03741863A EP1531868A2 (fr) 2002-06-03 2003-06-03 Agents therapeutiques contenant des nano-polymeres
CA002487720A CA2487720A1 (fr) 2002-06-03 2003-06-03 Agent therapeutique contenant des nanoarticles polymeriques
JP2004508783A JP2005535604A (ja) 2002-06-03 2003-06-03 治療剤を含むポリマーナノ物品
US10/995,880 US20050129769A1 (en) 2002-06-03 2004-11-22 Polymeric articles for carrying therapeutic agents

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US38553702P 2002-06-03 2002-06-03
US60/385,537 2002-06-03
US46316203P 2003-04-16 2003-04-16
US60/463,162 2003-04-16

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US10/995,880 Continuation-In-Part US20050129769A1 (en) 2002-06-03 2004-11-22 Polymeric articles for carrying therapeutic agents

Publications (3)

Publication Number Publication Date
WO2003101425A2 true WO2003101425A2 (fr) 2003-12-11
WO2003101425A3 WO2003101425A3 (fr) 2005-03-17
WO2003101425A8 WO2003101425A8 (fr) 2005-04-28

Family

ID=29715373

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/017536 WO2003101425A2 (fr) 2002-06-03 2003-06-03 Articles polymeres contenant des agents therapeutiques

Country Status (5)

Country Link
EP (1) EP1531868A2 (fr)
JP (1) JP2005535604A (fr)
AU (1) AU2003273272A1 (fr)
CA (1) CA2487720A1 (fr)
WO (1) WO2003101425A2 (fr)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005027873A2 (fr) * 2003-06-20 2005-03-31 Alnis Biosciences, Inc. Nanoarticles therapeutiques
WO2005065724A1 (fr) * 2003-12-30 2005-07-21 Alnis Biosciences, Inc. Preparations a base de complexes d'ions paramagnetiques
WO2005070471A2 (fr) * 2004-01-20 2005-08-04 Alnis Biosciences, Inc. Articles comprenant un materiau magnetique et des agents bioactifs
JP2008542295A (ja) * 2005-05-27 2008-11-27 ロイヤー バイオメディカル, インク. 生体再吸収性ポリマーマトリックス、ならびにその作製および使用方法
EP2111872A3 (fr) * 2004-07-05 2009-12-23 Complex Biosystems GmbH Hydrogel de conjugués polymères promédicamenteux
CN101864071A (zh) * 2010-05-21 2010-10-20 北京中海康医药科技发展有限公司 聚乙二醇-二硬脂酰磷脂酰乙醇胺衍生物及其制备方法
US7968085B2 (en) 2004-07-05 2011-06-28 Ascendis Pharma A/S Hydrogel formulations
WO2013036931A1 (fr) 2011-09-08 2013-03-14 Western University Of Health Sciences Liposomes ciblés en thérapie cancéreuse
US9023370B2 (en) 2008-12-16 2015-05-05 Fundacio Privada Institut Catala De Nanotecnologia Conjugates comprising nanoparticles coated with platinum containing compounds
US9133276B2 (en) 2010-09-17 2015-09-15 Sanofi-Aventis Deutschland Gmbh Prodrugs comprising an exendin linker conjugate
US9138462B2 (en) 2009-07-31 2015-09-22 Sanofi-Aventis Deutschland Gmbh Prodrugs comprising an insulin linker conjugate
US9265723B2 (en) 2009-07-31 2016-02-23 Sanofi-Aventis Deutschland Gmbh Long acting insulin composition
US9295736B2 (en) 2007-09-24 2016-03-29 Bar Ilan University Polymer nanoparticles coated by magnetic metal oxide and uses thereof
US9434610B2 (en) 2008-10-07 2016-09-06 Rexahn Pharmaceuticals, Inc. HPMA—docetaxel conjugates and uses therefore
WO2017201407A1 (fr) * 2016-05-20 2017-11-23 Massachusetts Institute Of Technology Particule hydrogel d'aldéhyde dextrane réticulé par la cystamine, contenant de la doxorubicine conjuguée à l'imine et un peptide rgd pour la chimiothérapie
US10285943B2 (en) 2010-12-02 2019-05-14 Greenmark Biomedical Inc. Aptamer bioconjugate drug delivery device
US11666515B2 (en) 2018-03-28 2023-06-06 Greenmark Biomedical Inc. Phosphate crosslinked starch nanoparticle and dental treatments

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2010306917B2 (en) * 2009-10-13 2014-11-27 Rexahn Pharmaceuticals, Inc. Polymeric systems for the delivery of anticancer agents
PL221351B1 (pl) * 2012-03-14 2016-03-31 Politechnika Warszawska Sposób otrzymywania nanocząstek polisacharydowych

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5415864A (en) * 1990-04-18 1995-05-16 University Of Utah Research Foundation Colonic-targeted oral drug-dosage forms based on crosslinked hydrogels containing azobonds and exhibiting PH-dependent swelling
US5562099A (en) * 1990-10-05 1996-10-08 Massachusetts Institute Of Technology Polymeric microparticles containing agents for imaging
US5702727A (en) * 1993-02-22 1997-12-30 Alza Corporation Compositions and methods for the oral delivery of active agents
EP0838224A2 (fr) * 1996-10-02 1998-04-29 Kuraray Co., Ltd. Conjugués de polymère - médicament avec in lien clivable par des enzymes
WO2000009088A1 (fr) * 1998-08-14 2000-02-24 Incept Llc Systemes d'administration de medicaments au moyen d'hydrogels composites
WO2001015738A2 (fr) * 1999-09-02 2001-03-08 Rice University Materiaux hydrogels produisant du monoxyde d'azote
WO2001078786A2 (fr) * 2000-04-14 2001-10-25 Alnis Biosciences, Inc. Nanoparticules renfermant des peptides a haute affinite

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5415864A (en) * 1990-04-18 1995-05-16 University Of Utah Research Foundation Colonic-targeted oral drug-dosage forms based on crosslinked hydrogels containing azobonds and exhibiting PH-dependent swelling
US5562099A (en) * 1990-10-05 1996-10-08 Massachusetts Institute Of Technology Polymeric microparticles containing agents for imaging
US5702727A (en) * 1993-02-22 1997-12-30 Alza Corporation Compositions and methods for the oral delivery of active agents
EP0838224A2 (fr) * 1996-10-02 1998-04-29 Kuraray Co., Ltd. Conjugués de polymère - médicament avec in lien clivable par des enzymes
WO2000009088A1 (fr) * 1998-08-14 2000-02-24 Incept Llc Systemes d'administration de medicaments au moyen d'hydrogels composites
WO2001015738A2 (fr) * 1999-09-02 2001-03-08 Rice University Materiaux hydrogels produisant du monoxyde d'azote
WO2001078786A2 (fr) * 2000-04-14 2001-10-25 Alnis Biosciences, Inc. Nanoparticules renfermant des peptides a haute affinite

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
BOUHADIR K H ET AL: "Hydrogels for combination delivery of antineoplastic agents" BIOMATERIALS, ELSEVIER SCIENCE PUBLISHERS BV., BARKING, GB, vol. 22, no. 19, 1 October 2001 (2001-10-01), pages 2625-2633, XP004296082 ISSN: 0142-9612 *
BOUHADIR KAMAL H ET AL: "Sustained and controlled release of daunomycin from cross-linked poly(aldehyde guluronate) hydrogels" JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 89, no. 7, July 2000 (2000-07), pages 910-919, XP002261385 ISSN: 0022-3549 *
CAIOLFA V R ET AL: "Polymer-bound camptothecin: initial biodistribution and antitumour activity studies" JOURNAL OF CONTROLLED RELEASE, ELSEVIER SCIENCE PUBLISHERS B.V., AMSTERDAM, NL, vol. 65, no. 1-2, March 2000 (2000-03), pages 105-119, XP004190316 ISSN: 0168-3659 cited in the application *
DENNY WILLIAM A ET AL: "The design of selectively-activated anti-cancer prodrugs for use in antibody-directed and gene-directed enzyme-prodrug therapies" JOURNAL OF PHARMACY AND PHARMACOLOGY, vol. 50, no. 4, April 1998 (1998-04), pages 387-394, XP009020713 ISSN: 0022-3573 *
GOLDBERG EUGENE P: "POLYMERIC AFFINITY DRUGS FOR CARDIOVASCULAR, CANCER AND UROLITHIASIS THERAPY" 173RD;NEW ORLEANS, LA, USA MAR 20-25 1977, vol. 18, no. 1, March 1977 (1977-03), pages 575-580, XP009020428 Am Chem Soc Div Polym Chem Prepr Mar 1977 *
LLOYD JOHN B ET AL: "SOLUBLE SYNTHETIC POLYMERS AS TARGETABLE AGENTS FOR INTRACELLULAR DRUG RELEASE" PAP PRESENTED AT THE POLYM AS DRUG RELEASE AGENTS SYMP;LONDON, ENGL MAR 23 1983, vol. 15, no. 4, 23 March 1983 (1983-03-23), pages 158-159, XP009020432 Br Polym J Dec 1983 *
VERVOORT LIESBETH ET AL: "Inulin hydrogels as carriers for colonic drug targeting: I. Synthesis and characterization of methacrylated inulin and hydrogel formation" PHARMACEUTICAL RESEARCH (NEW YORK), vol. 14, no. 12, December 1997 (1997-12), pages 1730-1737, XP009020453 ISSN: 0724-8741 *

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005027873A3 (fr) * 2003-06-20 2005-06-23 Alnis Biosciences Inc Nanoarticles therapeutiques
WO2005027873A2 (fr) * 2003-06-20 2005-03-31 Alnis Biosciences, Inc. Nanoarticles therapeutiques
WO2005065724A1 (fr) * 2003-12-30 2005-07-21 Alnis Biosciences, Inc. Preparations a base de complexes d'ions paramagnetiques
WO2005070471A2 (fr) * 2004-01-20 2005-08-04 Alnis Biosciences, Inc. Articles comprenant un materiau magnetique et des agents bioactifs
WO2005070471A3 (fr) * 2004-01-20 2006-03-30 Alnis Biosciences Inc Articles comprenant un materiau magnetique et des agents bioactifs
US9056126B2 (en) 2004-07-05 2015-06-16 Ascendis Pharma A/S Hydrogel formulations
US10029015B2 (en) 2004-07-05 2018-07-24 Ascendis Pharma A/S Hydrogel formulations
EP2111872A3 (fr) * 2004-07-05 2009-12-23 Complex Biosystems GmbH Hydrogel de conjugués polymères promédicamenteux
EP1781335B1 (fr) * 2004-07-05 2021-01-13 Ascendis Pharma GmbH Conjugués d'hydrogel
US7968085B2 (en) 2004-07-05 2011-06-28 Ascendis Pharma A/S Hydrogel formulations
EP2594291A1 (fr) * 2004-07-05 2013-05-22 Ascendis Pharma GmbH Hydrogel
JP2008542295A (ja) * 2005-05-27 2008-11-27 ロイヤー バイオメディカル, インク. 生体再吸収性ポリマーマトリックス、ならびにその作製および使用方法
JP2013155204A (ja) * 2005-05-27 2013-08-15 Royer Biomedical Inc 生体再吸収性ポリマーマトリックス、ならびにその作製および使用方法
US9295736B2 (en) 2007-09-24 2016-03-29 Bar Ilan University Polymer nanoparticles coated by magnetic metal oxide and uses thereof
US9434610B2 (en) 2008-10-07 2016-09-06 Rexahn Pharmaceuticals, Inc. HPMA—docetaxel conjugates and uses therefore
US9023370B2 (en) 2008-12-16 2015-05-05 Fundacio Privada Institut Catala De Nanotecnologia Conjugates comprising nanoparticles coated with platinum containing compounds
US9138462B2 (en) 2009-07-31 2015-09-22 Sanofi-Aventis Deutschland Gmbh Prodrugs comprising an insulin linker conjugate
US9265723B2 (en) 2009-07-31 2016-02-23 Sanofi-Aventis Deutschland Gmbh Long acting insulin composition
US9457066B2 (en) 2009-07-31 2016-10-04 Sanofi-Aventis Deutschland Gmbh Prodrugs comprising an insulin linker conjugate
CN101864071A (zh) * 2010-05-21 2010-10-20 北京中海康医药科技发展有限公司 聚乙二醇-二硬脂酰磷脂酰乙醇胺衍生物及其制备方法
US9133276B2 (en) 2010-09-17 2015-09-15 Sanofi-Aventis Deutschland Gmbh Prodrugs comprising an exendin linker conjugate
US11369570B2 (en) 2010-12-02 2022-06-28 Greenmark Biomedical Inc. Aptamer bioconjugate drug delivery device
US10285943B2 (en) 2010-12-02 2019-05-14 Greenmark Biomedical Inc. Aptamer bioconjugate drug delivery device
EP2763658A4 (fr) * 2011-09-08 2015-08-12 Univ Western Health Sciences Liposomes ciblés en thérapie cancéreuse
US9808425B2 (en) 2011-09-08 2017-11-07 Western University Of Health Sciences Targeted liposomes in cancer therapy
WO2013036931A1 (fr) 2011-09-08 2013-03-14 Western University Of Health Sciences Liposomes ciblés en thérapie cancéreuse
WO2017201407A1 (fr) * 2016-05-20 2017-11-23 Massachusetts Institute Of Technology Particule hydrogel d'aldéhyde dextrane réticulé par la cystamine, contenant de la doxorubicine conjuguée à l'imine et un peptide rgd pour la chimiothérapie
US11666515B2 (en) 2018-03-28 2023-06-06 Greenmark Biomedical Inc. Phosphate crosslinked starch nanoparticle and dental treatments

Also Published As

Publication number Publication date
AU2003273272A1 (en) 2003-12-19
EP1531868A2 (fr) 2005-05-25
WO2003101425A3 (fr) 2005-03-17
JP2005535604A (ja) 2005-11-24
CA2487720A1 (fr) 2003-12-11
WO2003101425A8 (fr) 2005-04-28
AU2003273272A8 (en) 2003-12-19

Similar Documents

Publication Publication Date Title
WO2003101425A2 (fr) Articles polymeres contenant des agents therapeutiques
Zhang et al. Designing polymers with sugar-based advantages for bioactive delivery applications
Khandare et al. Multifunctional dendritic polymers in nanomedicine: opportunities and challenges
Medina et al. Dendrimers as carriers for delivery of chemotherapeutic agents
JP2022191364A (ja) ポリマーナノ粒子およびその調整のプロセス
Liao et al. Dual pH-responsive-charge-reversal micelle platform for enhanced anticancer therapy
Cafaggi et al. Preparation and evaluation of nanoparticles made of chitosan or N-trimethyl chitosan and a cisplatin–alginate complex
Van Der Poll et al. Design, synthesis, and biological evaluation of a robust, biodegradable dendrimer
Mezghrani et al. Hepatocellular carcinoma dually-targeted nanoparticles for reduction triggered intracellular delivery of doxorubicin
US20060127310A1 (en) Amplification of biotin-mediated targeting
Ma et al. Copolymers containing carbohydrates and other biomolecules: design, synthesis and applications
JP2003531930A (ja) 両親媒性大環状誘導体およびそれらの類似体
JPWO2006028129A1 (ja) 医薬品製剤
US20040126900A1 (en) High affinity peptide- containing nanoparticles
Mukwaya et al. Saccharide‐based nanocarriers for targeted therapeutic and diagnostic applications
Hudlikar et al. Controlled multi‐functionalization facilitates targeted delivery of nanoparticles to cancer cells
Nie et al. In vitro and in vivo evaluation of stimuli-responsive vesicle from PEGylated hyperbranched PAMAM-doxorubicin conjugate for gastric cancer therapy
CA2553647A1 (fr) Articles comprenant un materiau magnetique et des agents bioactifs
US20050129769A1 (en) Polymeric articles for carrying therapeutic agents
JPH06505755A (ja) 重合体誘導体結合アンスラサイクリン配糖体を含有する医薬組成物及びその製造方法
US6054492A (en) Fluorinated copolymeric pharmaceutical adjuncts
Wu et al. Enhanced drug delivery to hepatocellular carcinoma with a galactosylated core–shell polyphosphoester nanogel
WO2005065724A1 (fr) Preparations a base de complexes d'ions paramagnetiques
KR20180107745A (ko) 기체 발포형 마이셀 및 이의 제조방법
KR102358116B1 (ko) 기체 발포형 마이셀 및 이의 제조방법

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 10995880

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2487720

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2004508783

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2003741863

Country of ref document: EP

CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i

Free format text: IN PCT GAZETTE 50/2003 UNDER (71) THE NAME/ADDRESS SHOULD READ "ALNIS BIOSCIENCES, INC."

WWP Wipo information: published in national office

Ref document number: 2003741863

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 2003741863

Country of ref document: EP