WO2010075072A2 - Long circulating nanoparticles for sustained release of therapeutic agents - Google Patents

Long circulating nanoparticles for sustained release of therapeutic agents Download PDF

Info

Publication number
WO2010075072A2
WO2010075072A2 PCT/US2009/068028 US2009068028W WO2010075072A2 WO 2010075072 A2 WO2010075072 A2 WO 2010075072A2 US 2009068028 W US2009068028 W US 2009068028W WO 2010075072 A2 WO2010075072 A2 WO 2010075072A2
Authority
WO
WIPO (PCT)
Prior art keywords
therapeutic agent
nanoparticles
nanoparticle composition
patient
biocompatible
Prior art date
Application number
PCT/US2009/068028
Other languages
French (fr)
Other versions
WO2010075072A3 (en
Inventor
Stephen E. Zale
Greg Troiano
Mir Mukkaram Ali
Jeff Hrkach
James Wright
Susan Low
Original Assignee
Bind Biosciences
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 Bind Biosciences filed Critical Bind Biosciences
Priority to JP2011540968A priority Critical patent/JP2012512175A/en
Priority to ES09835578T priority patent/ES2776126T3/en
Priority to EA201100765A priority patent/EA201100765A1/en
Priority to EP09835578.7A priority patent/EP2379064B1/en
Publication of WO2010075072A2 publication Critical patent/WO2010075072A2/en
Publication of WO2010075072A3 publication Critical patent/WO2010075072A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5146Organic macromolecular compounds; Dendrimers obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyamines, polyanhydrides
    • A61K9/5153Polyesters, e.g. poly(lactide-co-glycolide)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/475Quinolines; Isoquinolines having an indole ring, e.g. yohimbine, reserpine, strychnine, vinblastine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • A61K47/54Medicinal 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 compound
    • A61K47/542Carboxylic acids, e.g. a fatty acid or an amino acid
    • 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
    • A61K47/59Medicinal 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 obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/593Polyesters, e.g. PLGA or polylactide-co-glycolide
    • 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
    • A61K47/59Medicinal 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 obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal 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 obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO 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
    • A61K47/6925Medicinal 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 microcapsule, nanocapsule, microbubble or nanobubble
    • 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
    • A61K47/6937Medicinal 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 the polymer being PLGA, PLA or polyglycolic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate 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
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1641Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, poloxamers
    • 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

Definitions

  • Nanoparticles for the delivery of therapeutic agents have the potential to circumvent many challenges associated with traditional delivery approaches including lack of patient compliance to prescribed therapy, adverse effects, and inferior clinical efficacy due to lack of targeted delivery.
  • Important technological advantages of nanoparticles for drug delivery include the ability to deliver water-insoluble and unstable drugs, incorporation of both hydrophobic and hydrophilic therapeutic agents, and ability to utilize various routes of administration. Nanoparticle delivery systems may also facilitate targeted drug delivery and controlled release applications, enhance drug bioavailability at the site of action, reduce dosing frequency, and minimize side effects.
  • nanoparticulate systems have been examined for use as drug delivery vehicles, including polymeric micelles, polymers, liposomes, low-density lipoproteins, dendrimers, hydrophilic drug -polymer complexes, and ceramic nanoparticles.
  • Typical polymeric materials utilized in polymeric particulate drug delivery systems include polylactic acid (PLA), poly(D,L-glycolide) (PLG), and poly(lactide-co- glycolide) (PLGA).
  • PLA and PLGA are listed as Generally Recognized as Safe (GRAS) under Sections 201(s) and 409 of the Federal Food, Drug, and Cosmetic Act, and are approved for use in commercially available microparticulate systems, including Decapeptyl ® , Parlodel LA ® , and Enantone Depot ® , as well as in implant devices, such as Zoladex ® .
  • GRAS Generally Recognized as Safe
  • nanoparticle systems such as liposomes
  • polymeric nanoparticles developed to date have limited effectiveness, in part because such nanoparticles clear from the body quickly once administered and/or may accumulate in healthy tissue where treatment is not needed. Control of delivery of an active agent, using nanosystems, remains a challenge.
  • biocompatible compositions capable of extended delivery of active agents, e.g., anti-neoplastic agents, that provide for prolonged and/or increased plasma drug concentrations in a patient, especially as compared to administration of an active agent alone.
  • active agents e.g., anti-neoplastic agents
  • a nanoparticle composition that includes a biodegradable and/or biocompatible polymer and a therapeutic agent, wherein the biodegradable and/or biocompatible polymer matrix releases the therapeutic agent at a rate allowing controlled release of the agent over at least about 12 hours, or in some embodiments, at least about 24 hours
  • a biocompatible nanoparticle composition comprising a plurality of long circulating nanoparticles, each comprising a biocompatible polymer and a therapeutic agent, said composition providing an elevated plasma concentration of the therapeutic agent for at least 12 hours when the composition is administered to a patient, and an area under the plasma concentration time curve (AUC) that is increased by at least 100% over the AUC provided when the therapeutic agent is administered alone to a patient.
  • AUC area under the plasma concentration time curve
  • a biocompatible nanoparticle composition comprising a plurality of long circulating nanoparticles, each comprising a ⁇ -hydroxy polyester-co-polyether and a therapeutic agent, said composition providing an elevated plasma concentration of the therapeutic agent for at least 6 hours, at least 12 hours, or at least 24 hours or more when the composition is administered to a patient, to provide an area under the plasma concentration time curve (AUC) that is increased by at least 100%, or at least by 150%, over the AUC provided when the therapeutic agent is administered alone to a patient.
  • AUC plasma concentration time curve
  • disclosed nanoparticles may provide an actual peak plasma concentration (C max ) that is at least 10% higher, or even at least 100% higher, as compared to a C max of said therapeutic agent when administered alone.
  • Disclosed nanoparticles may provide a volume of distribution when administered to the patient that is less than or equal to about 5 plasma volumes.
  • dislcosed nanoparticles and/or compositions may decrease the volume of distribution (V 2 ) by at least 50% as compared to the V z of the patient when the therapeutic agent is administered alone.
  • Disclosed biocompatible nanoparticle compositions may include long circulating nanoparticles that may further comprise a biocompatible polymer coupled to a targeting moiety, for example, a targeting moiety that is selected from the group consisting of a protein, peptide, antibody, antibody fragment, saccharide, carbohydrate, small molecule, glycan, cytokine, chemokine, nucleotide, lectin, lipid, receptor, steroid, neurotransmitter, cell surface marker, cancer antigen, or glycoprotein antigen.
  • a targeting moiety may bind to prostate membrane specific antigen (PMSA).
  • a disclosed nanoparticle may include a biocompatible polymer coupled to a targeting moiety, e.g., a nanoparticle may include PLA-PEG-( ⁇ , S-2- ⁇ 3-[ 1 -carboxy-5-amino-pentyl] -ureido ⁇ -pentanedioic acid.
  • Disclosed long circulating nanoparticles may include 1 to about 4% by weight, or 2% to about 4% by weight, of a biocompatible polymer coupled to a targeting moiety [0010]
  • a biocompatible nanoparticle may include a biocompatible polymer such as PLA-PEG.
  • a ⁇ -hydroxy polyester-co-polyether may be polylactic acid -co-polyethylene glycol, and/or a ⁇ -hydroxy polyester-co-polyether comprises about 16kDa polylactic acid and about 5 kDa polyethylene glycol.
  • Disclosed long circulating nanoparticles may be about 80 to about 90 weight percent ⁇ -hydroxy polyester-co-polyether.
  • disclosed long circulating nanoparticles may further comprise a biodegradable polymer, such as poly(lactic) acid.
  • long circulating nanoparticles may have about 40 to about 50 weight percent poly(lactic)acid, and about 40 to about 50 weight percent of ⁇ -hydroxy polyester-co-polyether.
  • compositions that include such biocompatible nanoparticles and a therapeutic agent may provide a peak plasma concentration (C max ) of a therapeutic agent at least 100% higher than the C max of the therapeutic agent when administered alone, and/or the area under the plasma concentration time curve (AUC) mayincreased by at least 200% over the AUC of the therapeutic agent when administered alone to the patient.
  • C max peak plasma concentration
  • AUC area under the plasma concentration time curve
  • Disclosed nanoparticle compositions may include a therapeutic agent such as one selected from the group consisting of chemotherapeutic agents, diagnostic agents, prophylactic agents, nutraceutical agents, nucleic acids, proteins, peptides, lipids, carbohydrates, hormones, small molecules, metals, ceramics, drugs, vaccines, immunological agents, and combinations thereof, for example, a nanoparticle may include an anti-neoplastic agent such as docetaxel, vincristine, methotrexate, paclitaxel, or sirolimus. Disclosed nanoparticle compositions may further include an aqueous solution of a saccharide.
  • a therapeutic agent such as one selected from the group consisting of chemotherapeutic agents, diagnostic agents, prophylactic agents, nutraceutical agents, nucleic acids, proteins, peptides, lipids, carbohydrates, hormones, small molecules, metals, ceramics, drugs, vaccines, immunological agents, and combinations thereof
  • a nanoparticle may include an anti-neo
  • a method of treating a solid tumor cancer comprising administering disclosed nanoparticle composition to a patient (e.g. a mammal or primate) in need thereof.
  • a patient e.g. a mammal or primate
  • Such methods may provide wherein at least 24 hours after administration, a solid tumor has significant concentration of therapeutic agent.
  • Contemplated herein is a method of treating a solid tumor in a mammal in need thereof, comprising administering a nanoparticle composition comprising a plurality of nanoparticles each comprising a ⁇ -hydroxy polyester-co-polyether and a therapeutic agent, wherein the composition has an amount of therapeutic agent effective to inhibit the growth of said tumor, for example, a single dose of said composition may provide extended elevated plasma concentrations of said therapeutic agent in the patient for a least one day, (e.g.. the peak plasma concentration (C max ) of the therapeutic agent after administration of the composition to the mammal is at least 10% higher than the C max of said therapeutic agent if administered in a non-nanoparticle formulation.
  • C max peak plasma concentration
  • a method of minimizing unwanted side effects or toxicity of an active agent in a patient comprising: administering a nanoparticle composition comprising a plurality of nanoparticles each comprising a ⁇ -hydroxy polyester-co-polyether and a therapeutic agent, wherein said composition is capable of delivery a higher plasma concentration of therapeutic agent to the patient as compared to administering the therapeutic agent alone, and wherein upon administering the nanoparticle composition the volume distribution of the active agent in the patient is reduced, as compared to the volume distribution if the therapeutic agent was administered alone.
  • a method for modulating the plasma concentration of a therapeutic agent in a patient e.g. a primate (e.g.
  • FIG. 1 is a schematic illustration of a nanoparticle according to one aspect of the present invention.
  • FIG. 2 is a block diagram of the emulsion process used in the fabrication of nanoparticles in one aspect of the present invention.
  • FIG. 3 depicts the in vitro release of docetaxel from nanoparticles and conventional docetaxel.
  • FIG. 4 depicts the pharmacokinetics of docetaxel encapsulated in nanoparticles and conventional docetaxel in rats.
  • FIG. 5 depicts the distribution of radioactivity determined in selected tissues of rats after IV administration of nanoparticles containing 14 C-targeting polymer ( ), nanoparticles containing 14 C-docetaxel (ss), and conventional 14 C -docetaxel (V).
  • FIG. 6 depicts docetaxel concentration in tumor tissue after administration of docetaxel encapsulated in nanoparticles or conventional docetaxel to LNCaP tumor bearing
  • FIG. 7 depicts the reduction in tumor volume in mice with PSM A-expres sing LNCaP xenografts when treated with docetaxel encapsulated in nanoparticles or conventional docetaxel.
  • FIG. 8 depicts pharmacokinetics of vincristine encapsulated in disclosed nanoparticles and conventional vincristine in rats.
  • FIG. 9 depicts pharmacokinetics of methotrexate encapsulated in disclosed nanoparticles and conventional methotrexate in rats.
  • FIG. 10 depicts pharmacokinetics of paclitaxel encapsulated in disclosed nanoparticles and conventional paclitaxel in rats.
  • FIG. 11 depicts pharmacokinetics of rapamycin (sirolimus) encapsulated in disclosed nanoparticles and conventional rapamycin in rats.
  • FIG. 12 depicts the tumor accumulation of docetaxel in disclosed nanoparticles in a MX-I mouse breast tumor model.
  • FIG. 13 depicts pharmacokinetics of docetaxel in a NHP model using various disclosed nanoparticles.
  • an "effective amount” or “therapeutically effective amount” of a composition, as used herein, is a predetermined amount calculated to achieve a desired effect.
  • long-circulating refers to enhanced stability in the circulatory system of a patient, regardless of biological activity.
  • nanophase and “nanosize” refer to a special state of subdivision implying that a particle has an average dimension smaller than about 1000 nm ( 100Ox 10 9 m) .
  • polyethylene glycol) or "PEG” and “poly(ethylene oxide)” or “PEO” denote polyethers comprising repeat -CH 2 -CH 2 -O- units.
  • PEG and/or PEO can be different polymers depending upon end groups and molecular weights.
  • poly(ethylene glycol) and PEG describes either type of polymer.
  • An " ⁇ -hydroxy polyester” refers to polymers having monomers based on one or more ⁇ -hydroxy acid, such as poly(lactic) acid, poly(glycolic) acid, poly-lactic-co-glycolic acid, polycaprolactone.
  • target refers to the cell type or tissue to which enhanced delivery of the therapeutic agent is desired.
  • diseased tissue may be a target for therapy.
  • terapéutica agent means a compound utilized to image, impact, treat, combat, ameliorate, prevent or improve an unwanted condition or disease of a patient.
  • disclosed long-circulating nanoparticles include a therapeutic agent and biodegradable and/or biocompatible polymeric particles, optionally functionalized with targeting moieties.
  • the nanoparticles are designed to circulate in a vascular compartment of a patient for an extended period of time, distribute and accumulate at a target, and release the encapsulated therapeutic agent in a controlled manner. These characteristics can result in an increased level of therapeutic agent in the target and a potential reduction in off-target exposure.
  • the disclosed nanoparticles remain in circulation longer because, upon administration to a patient (e.g. a mammal, primate (e.g. human)), the disclosed nanoparticles are substantially confined to the vascular compartment of the patient, and are engineered to be cleared very slowly.
  • the activity of many therapeutic agents is dependent on their pharmacokinetic behavior. This pharmacokinetic behavior defines the drug concentrations and period of time over which cells are exposed to the drug. For most therapeutics, e.g. anti-neoplasties, longer exposure times are preferred as this results in increased killing of the cancer cells.
  • peak plasma concentration, or maximum plasma concentration (C max ) and area under the curve (AUC) are examples.
  • AUC is a measure of plasma drug levels over time and provides an indication of the total drug exposure. Generally, plasma concentration and plasma AUC for a therapeutic agent correlate with increased therapeutic efficacy.
  • a biocompatible nanoparticle composition comprising a plurality of long circulating nanoparticles, each comprising a ⁇ -hydroxy polyester-co-polyether and a therapeutic agent.
  • Such compositions may provide a therapeutic effect for at least 12 hours, at least 24 hours, or at least 36 hours, or 48 hours or more, upon administration to a patient.
  • peak plasma concentration (C max ) of the therapeutic agent of such nanoparticles e.g. when the composition is administered to a patient, may be least 10% higher, 20% higher, or about 10% to about 100% higher, or more, than the C max of the same therapeutic agent when administered alone.
  • Actual peak plasma concentration of delivered therapeutic agent includes both agent that is released from the nanoparticle (e.g. after administration) and therapeutic agent remaining in any nanoparticle remaining in the plasma, e.g. at a given time.
  • disclosed nanoparticles may provide upon administration to a patient, an area under the plasma concentration time curve (AUC), that may be increased by at least 100%, at least 200% , or about 100% to about 500% or more, over the AUC of the therapeutic agent when administered alone to the patient.
  • AUC plasma concentration time curve
  • a provided composition that includes disclosed nanoparticles may decrease the volume of distribution(V z ) of distributed active agent, upon administration, in a patient by at least 10%, or by at least 20%, or about 10% to about 100%, as compared to the V z of the patient when the therapeutic agent is administered alone.
  • a provided nanoparticle composition may provide V z in a patient that is on the same order of magnitude that the of plasma volume and/or a volume of distribution less than about 10 plasma volumes.
  • a disclosed nanoparticle composition may provide a Vz that is less than, or about, 2 times the plasma volume, or less than or about 8 plasma volumes.
  • a disclosed nanoparticle composition may provide a V z in a patient that is on about the same order of plasma volume, (e.g. about 5L for an exemplary 70 kg patient), e.g. about a V z that indicates administered nanoparticles are substantially in the patient's plasma and not substantially in other tissues.
  • disclosed nanoparticles may be used as a drug delivery vehicle based on the encapsulation of a therapeutic agent in a polymer matrix with controlled porosity and/or a soluble shell or matrix that upon dissolution releases the therapeutic agent in the immediate vicinity of the targeted area.
  • the protection of the therapeutic agent provided by the polymer shell or matrix allows for the delivery of therapeutic agents that are water- insoluble or unstable.
  • dissolution kinetics of the polymer can be designed to provide sustained release of therapeutic agents at a target for an extended period of time.
  • Disclosed nanoparticles can be used for a variety of applications, such as, without limitation, drug delivery, gene therapy, medical diagnosis, and for medical therapeutics for cancer, pathogen-borne diseases, hormone-related diseases, reaction-by-products associated with organ transplants, and other abnormal cell or tissue growth.
  • a patient e.g. a mammal suffering from cancer, e.g. a solid tumor cancer, prostate cancer, breast cancer or lung cancer using e.g., disclosed nanoparticles.
  • contemplated diseases that may be treated using disclosed nanoparticles include a broad range of diseases and find limitation only by e.g.
  • a nanoparticle delivery system that mitigates against colloidal instability, agglomeration, polydispersity in nanoparticle size and shape, swelling, and leakage of encapsulated materials.
  • nanoparticles for delivery of therapeutic agents are provided that exhibit encapsulation efficiency.
  • Encapsulation efficiency is affected by factors including, for example, material characteristics of the polymer utilized as the carrier matrix, the chemical and physical properties of the therapeutic agent to be encapsulated, and type of solvents used in the nanoparticle fabrication process.
  • polymeric nanoparticles for delivery of therapeutic agents are provided that exhibit particle heterogeneity.
  • Conventional polymeric nanoparticle fabrication techniques generally provide multimodal particle size distributions as a result of self- aggregation during nanoprecipitation of both the polymer and the drug molecules.
  • Polymeric nanoparticles for delivery of therapeutic agents are provided, in an embodiment, that may reduce or eliminate burst release effects.
  • Conventional polymeric nanoparticle carriers frequently exhibit a bimodal drug release pattern with up to about 40-80% or more of the encapsulated drug released during the first several hours. After 24 to 48 hours, drug release is significantly reduced due to the increased diffusion barrier for drug molecules located deep within the polymer matrix.
  • nanoparticle carrier systems poorly encapsulated drug molecules diffuse quickly into solution, which may lead to significant toxicity in vivo. Further, by the time the evacuated nanoparticles arrive and accumulate at the targeted site (e.g., tumor tissue), the nanoparticles generally have little or no remaining therapeutic efficacy.
  • polymeric nanoparticles for delivery of therapeutic agents may evade rapid capture by the reticuloendothelial system (RES), leading to extended circulation time and elevated concentration of the nanoparticles in the blood.
  • Rapid capture and elimination is typically caused by the process of opsonization in which opsonin proteins present in the blood serum quickly bind to conventional nanoparticles, allowing macrophages to easily recognize and remove these particulates before they can perform their designed therapeutic function.
  • the extent and nature of opsonin adsorption at the surface of nanoparticles and their simultaneous blood clearance depend on the physicochemical properties of the particles, such as size, surface charge, and surface hydrophobicity.
  • a nanoparticle composition including a biodegradable and/or biocompatible polymer matrix and a therapeutic agent coupled to the biodegradable and/or biocompatible polymer matrix wherein the clearance rate of said therapeutic agent coupled to the biodegradable and/or biocompatible polymer matrix is lower than the clearance rate of said therapeutic agent when administered alone.
  • methods are provided that mask or camouflage nanoparticles in order to evade uptake by the RES.
  • One such method is the engineering of particles in which polyethers, such as poly(ethylene glycol) (PEG) or PEG containing surfactants, are deployed on the surface of nanoparticles.
  • PEG poly(ethylene glycol)
  • PEG- containing copolymers e.g. on the surface of nanoparticles can result in an increase in the blood circulation half-life of the nanoparticles by several orders of magnitude.
  • This method creates a hydrophilic protective layer around the nanoparticles that is able to repel the absorption of opsonin proteins via steric repulsion forces, thereby blocking and delaying the first step in the opsonization process.
  • FIG. 1 schematically illustrates a nanoparticle according to one aspect of the present invention.
  • docetaxel 100 an anti-neoplastic agent approved for the treatment of hormone refractory prostate cancer (HRPC)
  • HRPC hormone refractory prostate cancer
  • a matrix 110 derived from the biodegradable and/or biocompatible polymers PLA and poly(lactide-b- ethylene glycol) (PLA-PEG).
  • the polymer matrix 110 contains a targeting polymer (PLA-
  • PEG-lys(urea)glu) 120 that is end-functionalized (through the 5 amino moiety) with the lysine - urea-glutamate heterodimer (S,S-2- ⁇ 3-[l-carboxy-5-amino-pentyl]-ureido ⁇ -pentanedioic acid ( lys(urea)glu) 130, a small molecule ligand that selectively binds to PSMA, a clinically relevant prostate cancer cell surface marker.
  • the nanoparticles e.g.
  • the nanoparticle polymer(s) may be biologically degraded by, for example, enzymatic activity or cellular machinery into monomers and/or other moieties that cells can either use or excrete.
  • the dissolution or degradation rate of the nanoparticles is influenced by the composition of the polymer shell or matrix.
  • the half-life of the polymer (the time at which 50% of the polymer is degraded into monomers and/or other nonpolymeric moieties) may be on the order of days, weeks, months, or years, depending on the polymer.
  • nanoparticle delivery characteristics such as water uptake, controlled release of therapeutic agent, and polymer degradation kinetics may be optimized through selection of polymer shell or matrix composition.
  • Suitable polymers that may form some of the disclosed nanoparticles may include, but are not limited to, biodegradable ⁇ -hydroxy polyesters and biocompatible polyethers.
  • exemplary polyesters include, for example, PLA, PLGA, PEG, PEO, PEGylated polymers and copolymers of lactide and glycolide (e.g., PEGylated PLA, PEGylated PGA, PEGylated PLGA), and derivatives thereof.
  • suitable polymers include, for example, polyanhydrides, poly(ortho ester) PEGylated poly(ortho ester), poly(caprolactone), PEGylated poly(caprolactone), polylysine, PEGylated polylysine, poly(ethylene inline), PEGylated poly( ethylene imine), poly(L-lactide-co-L- lysine), poly(serine ester), poly(4-hydroxy-L-proline ester), poly[a-(4-aminobutyl)-L- glycolic acid], and combinations and derivatives thereof.
  • a polymer matrix may comprise one or more acrylic polymers.
  • acrylic polymers include, for example, acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, aminoalkyl methacrylate copolymer, poly( acrylic acid), poly(methacrylic acid), methacrylic acid alkylamide copolymer, poly(methyl methacrylate), poly(methacrylic acid polyacrylamide) copolymer, aminoalkyl methacrylate copolymer, glycidyl methacrylate copolymers, polycyanoacrylates, and combinations thereof.
  • the matrix may include dextran, acylated dextran, chitosan (e.g., acetylated to various levels), poly( vinyl) alcohol (for example, hydrolyzed to various degrees), and/or alginate, e.g. alginate complexed to bivalent cations such as a calcium alginate complex.
  • dextran acylated dextran
  • chitosan e.g., acetylated to various levels
  • poly( vinyl) alcohol for example, hydrolyzed to various degrees
  • alginate e.g. alginate complexed to bivalent cations such as a calcium alginate complex.
  • Nanoparticles disclosed herein include one, two, three or more biocompatible and/or biodegradable polymers.
  • a contemplated nanoparticle may include about 10 to about 99 weight percent of one or more block co-polymers that include a biodegradable polymer and polyethylene glycol, and about 0 to about 50 weight percent of a biodegradable homopolymer.
  • Exemplary therapeutic nanoparticles may include about 40 to about 90 weight percent poly(lactic) acid-poly(ethylene)glycol copolymer or about 40 to about 80 weight percent poly(lactic) acid-poly(ethylene)glycol copolymer.
  • Such poly(lactic) acid- block - poly(ethylene)glycol copolymer may include poly(lactic acid) having a number average molecular weight of about 15 to 20 kDa (or for example about 15 to about 10OkDa, e.g., about 15 to about 8OkDa), and poly(ethylene)glycol having a number average molecular weight of about 2 to about 1OkDa, for example, about 4 to about 6 kDa.
  • a disclosed therapeutic nanoparticle may include about 70 to about 90 weight percent PLA-PEG and about 5 to about 25 weight percent active agent (e.g.
  • PLA-PEG docetaxel
  • PLA ((poly)lactic acid) may have a number average molecular weight of about 5 to about 10 kDa.
  • PLGA poly lactic -co-glycolic acid
  • PLA-PEG copolymers may include a chemical linker, oligomer, or polymer chain between the PLA and PEG blocks, e.g., may include PLA-linker-PEG.
  • disclosed nanoparticles may include about 10 to 15 weight percent active agent (e.g. about 10 weight percent docetaxel), and about 86 to about 90 weight percent PLA-PEG (with e.g. PLA about 16 kDa and PEG about 5kDa, e.g. about 87.5% PLA-
  • a disclosed nanoparticle which may have slow release properties, may include about 42 to about 45 weight percent PLA-PEG (with e.g. PLA about 16 kDa and PEG about 5kDa), (e.g. 43.25% PLA-PEG), about 42 to 45 weight percent PLA (e.g. about 75 kDa) (e.g. 43.25% PLA/75kDa) and about 10 to 15 weight percent active agent (e.g. docetaxel).
  • PLA-PEG with e.g. PLA about 16 kDa and PEG about 5kDa
  • PLA-PEG e.g. 43.25% PLA-PEG
  • PLA e.g. about 75 kDa
  • PLA/75kDa e.g. 43.25% PLA/75kDa
  • active agent e.g. docetaxel
  • disclosed nanoparticles may optionally include about 1 to about 50 weight percent poly(lactic) acid or poly(lactic) acid-co-poly (glycolic) acid (which does not include PEG, e.g a homopolymer of PLA), or may optionally include about 1 to about 50 weight percent, or about 10 to about 50 weight percent or about 30 to about 50 weight percent poly(lactic) acid or poly(lactic) acid-co-poly (glycolic) acid.
  • disclosed nanoparticles may include two polymers, e.g. PLA-PEG and PLA, in a weight ratio of about 30:60 to about 60: 30, e.g, about 40:60, about 60:40, or about 50:50.
  • Such substantially homopolymeric poly(lactic) or poly(lactic)-co-poly(glycolic) acid may have a weight average molecular weight of about 10 to about 130 kDa, for example, about 20 to about 30 kDa, or about 100 to about 130 kDa.
  • Such homopolymeric PLA may have a number average molecule weight of about 5 to about 90 kDa, or about 5 to about 12 kDa, about 15 to about 30 kDa, or about 60 to about 90 kDa.
  • Exemplary homopolymeric PLA may have a number average molecular weight of about 80 kDa or a weight average molecular weight of about 124 kDa.
  • molecular weight of polymers can be related to an inherent viscosity.
  • homopolymer PLA may have an inherent viscosity of about 0.2 to about 0.4, e.g. about 0.3; in other embodiments, PLA may have an inherent viscosity of about 0.6 to about 0.8.
  • Exemplary PLGA may have a number average molecular weight of about 8 to about 12 kDa.
  • modified surface chemistry and/or small particle size of disclosed nanoparticles may contribute to the effectiveness of the nanoparticles in the delivery of a therapeutic agent.
  • nanoparticle surface charge may be modified to achieve slow biodegradation and reduce clearance of the nanoparticles.
  • porosity of the polymer shell or matrix is optimized to achieve extended and controlled release of the therapeutic agent.
  • the nanoparticles may have porosity in the range of about 10 to about 90 percent and/or a pore diameters in the range of about 0.001 to about 0.01 microns.
  • the nanoparticles according to some embodiments of the invention may be able to penetrate the altered and often compromised vasculature of tumors via the enhanced permeability and retention (EPR) effect resulting in preferential accumulation of nanoparticles in tumor interstitium.
  • EPR enhanced permeability and retention
  • therapeutic agents examples include, but are not limited to, chemotherapeutic agents (e.g. anti-cancer agents), diagnostic agents (e.g. contrast agents, radionuclides, and fluorescent, luminescent, and magnetic moieties), prophylactic agents (e.g. vaccines), nutraceutical agents (e.g. vitamins and minerals), nucleic acids (e.g., siRNA, RNAi, and mircoRNA agents), proteins (e.g. antibodies), peptides, lipids, carbohydrates, hormones, small molecules, metals, ceramics, drugs, vaccines, immunological agents, and/or combinations thereof.
  • chemotherapeutic agents e.g. anti-cancer agents
  • diagnostic agents e.g. contrast agents, radionuclides, and fluorescent, luminescent, and magnetic moieties
  • prophylactic agents e.g. vaccines
  • nutraceutical agents e.g. vitamins and minerals
  • nucleic acids e.g., siRNA, RNAi, and mircoRNA agents
  • the active agent or drug may be a therapeutic agent such as an antineoplastic such as a mTor inhibitor (e.g., sirolimus (rapamycin), temsirolimus, or everolimus), vinca alkaloids such as vincristine, a diterpene derivative, a taxane such as paclitaxel (or its derivatives such as DHA-paclitaxel or PG- paxlitaxel), docetaxel, or methatrexate.
  • antineoplastic such as a mTor inhibitor (e.g., sirolimus (rapamycin), temsirolimus, or everolimus), vinca alkaloids such as vincristine, a diterpene derivative, a taxane such as paclitaxel (or its derivatives such as DHA-paclitaxel or PG- paxlitaxel), docetaxel, or methatrexate.
  • the therapeutic agent to be delivered is an agent useful in the treatment of cancer (e.g., a solid tumor cancer e.g., prostate or breast cancer).
  • Such therapeutic agents may include, for example, doxorubicin (adriamycin), gemcitabine (gemzar), daunorubicin, procarbazine, mitomycin, cytarabine, etoposide, methotrexate, 5-fluorouracil (5-FU), vinblastine, vincristine, bleomycin, paclitaxel (taxol), docetaxel (taxotere), mitoxantrone, mitoxantrone hydrochloride, aldesleukin, asparaginase, busulfan, carboplatin, cladribine, camptothecin, CPT-Il, lO-hydroxy-7-ethylcamptothecin (SN38), dacarbazine, S-I capecitabine, ftor
  • contemplated nanoparticles may include more than one therapeutic agent. Such nanoparticles may be useful, for example, in aspects where it is desirable to monitor a targeting moiety as such moiety directs a nanoparticle containing a drug to a particular target in a subject.
  • Disclosed nanoparticles may be formed using an emulsion process, e.g. as presented as a block diagram in FIG. 2. As shown in FIG.
  • an organic polymer/drug solution containing docetaxel, PLA, PLA-PEG, and PLA-PEG-lys(urea)glu dissolved in a co-solvent mixture of ethyl acetate and benzyl alcohol is dispersed in an aqueous solution of sodium cholate, ethyl acetate, and benzyl alcohol to form a coarse emulsion.
  • the conditions under which the emulsion process is performed favor the orientation of the PEG and/or PEG-lys(urea)glu polymer chains toward the particle surface.
  • an orientation is achieved where the PEG is folded within the nanoparticle polymer shell or matrix.
  • a coarse emulsion can be passed through a high pressure homogenizer to reduce the droplet size, forming a fine emulsion.
  • the fine emulsion is diluted into an excess volume of a quench solution of cold water containing polysorbate 80.
  • the presence of polysorbate 80 serves to remove excess therapeutic agent that has not been encapsulated in the nanoparticle.
  • polysorbate 80 may also be adhered or associated with a nanoparticle surfaces. While not wishing to be bound by theory, polysorbate 80 coupled to the nanoparticle surfaces may impact characteristics such as controlled release of therapeutic agent and polymer degradation kinetics.
  • Quenching may be performed at least partially at a temperature of about 5 0 C or less.
  • water used in the quenching may be at a temperature that is less that room temperature (e.g., about 0 to about 1O 0 C, or about 0 to about 5 0 C).
  • not all of the therapeutic agent e.g., docetaxel
  • a drug solubilizer is added to the quenched phase to form a solubilized phase.
  • the drug solubilizer may be for example, Tween 80, Tween 20, polyvinyl pyrrolidone, cyclodextran, sodium dodecyl sulfate, or sodium cholate.
  • Tween-80 may added to the quenched nanoparticle suspension to solubilize the free drug and prevent the formation of drug crystals.
  • a ratio of drug solubilizer to therapeutic agent e.g., docetaxel
  • a ratio of drug solubilizer to therapeutic agent is about 100:1 to about 10:1.
  • Ethyl acetate and benzyl alcohol are extracted from the organic phase droplets, resulting in formation of a hardened nanoparticle suspension.
  • docetaxel or other active agent may be encapsulated at e.g. a loading level of 10% w/w; corresponding to more than 10,000 drug molecules per nanoparticle.
  • the nanoparticle suspension is processed using tangential flow ultrafiltration/diafiltration (UF/DF) with cold water to remove processing aids and to concentrate the nanoparticles to a desired value. Residual precursor materials and excess organics present in unwashed nanoparticle suspensions may have a detrimental impact on biomedical applications as well as undesired toxic effects on the physiological system.
  • the washed nanoparticle suspension is then passed through a prefilter and at least two sterilizing- grade filters.
  • the nanoparticles Once the nanoparticles have been prepared, they may be combined with an acceptable carrier to produce a pharmaceutical formulation, according to another aspect of the invention.
  • the carrier may be selected based on factors including, but not limited to, the route of administration, the location of the targeted disease tissue, the therapeutic agent being delivered, and/or the time course of delivery of the therapeutic agent.
  • a concentrated sucrose solution is aseptically added to the sterile nanoparticle suspension to produce a pharmaceutical formulation.
  • the sucrose serves as a cryoprotectant and a tonicity agent.
  • the resulting pharmaceutical formulation is a sterile, aqueous, injectable suspension of docetaxel encapsulated in nanoparticles comprised of biocompatible and biodegradable polymers.
  • the suspension is assayed for docetaxel content, and may be aseptically diluted to the desired concentration.
  • the particle suspension is aseptically filled and sealed in glass vials.
  • the bulk drug product suspension is stored frozen at -20 0 C + 5 0 C prior to filling into vials.
  • the fabrication methods for the nanoparticles of the invention may be modified in some embodiments to achieve desired drug-delivery features. For example, nanoparticle characteristics such as surface functionality, surface charge, particle size, zeta ( ⁇ ) potential, hydrophobicity, controlled release capability, and ability to control immunogenicity, and the like, may be optimized for the effective delivery of a variety of therapeutic agents.
  • the long-circulating nanoparticles produced according to the emulsion process shown in FIG. 2 are well dispersed and unagglomerated, which facilitates conjugation or functionalization of the nanoparticle surfaces with targeting moieties.
  • Disclosed nanoparticles may include optional targeting moieties, which may be selected to ensure that the nanoparticles selectively attach to, or otherwise associate with, a selected marker or target.
  • targeting moieties may be selected to ensure that the nanoparticles selectively attach to, or otherwise associate with, a selected marker or target.
  • disclosed nanoparticles may be functionalized with an amount of targeting moiety effective for the treatment of prostate cancer in a subject (e.g., a low-molecular weight PSMA ligand).
  • an amount of targeting moiety effective for the treatment of prostate cancer in a subject e.g., a low-molecular weight PSMA ligand.
  • the nanoparticles are effective only at targeted sites, which minimizes adverse side effects and improves efficacy.
  • Targeted delivery also allows for the administration of a lower dose of therapeutic agent, which may reduce undesirable side effects commonly associated with traditional treatments of disease.
  • disclosed nanoparticles may be optimized with a specific density of targeting moities on the nanoparticle surface, such that e.g., an effective amount of targeting moiety is associated with the nanoparticle for delivery of a therapeutic agent.
  • the fraction of the biodegradable and/or biocompatible polymer matrix functionalized with a targeting moiety may be less than 80% of the total.
  • the fraction of the biodegradable and/or biocompatible polymer matrix functionalized with a targeting moiety is less than about 50% of the total.
  • Increased density of the targeting moiety may, in some embodiments, increase target binding (cell binding/target uptake).
  • Exemplary targeting moieties include, for example, proteins, peptides, antibodies, antibody fragments, saccharides, carbohydrates, glycans, cytokines, chemokines, nucleotides, lectins, lipids, receptors, steroids, neurotransmitters and combinations thereof.
  • markers that may be useful in embodiments of the invention include, but are not limited to, cell surface markers, a cancer antigen (CA), a glycoprotein antigen, a melanoma associated antigen (MAA), a proteolytic enzyme, an angiogenesis marker, a prostate membrane specific antigen (PMSA), a small cell lung carcinoma antigen (SCLCA), a hormone receptor, a tumor suppressor gene antigen, a cell cycle regulator antigen, a proliferation marker, and a human carcinoma antigen.
  • exemplary targeting moieties include:
  • a disclosed nanoparticle may include PLA-PEG-targeting moiety, e.g. S,S-2- ⁇ 3-[l-carboxy-5-amino-pentyl]-ureido ⁇ -pentanedioic acid.
  • PLA-PEG-targeting moiety e.g. S,S-2- ⁇ 3-[l-carboxy-5-amino-pentyl]-ureido ⁇ -pentanedioic acid.
  • disclosed nanoparticles may include about 10 to 15 weight percent active agent (e.g. docetaxel), and about 86 to about 90 weight percent PLA-PEG (with e.g. PLA about 16 kDa and PEG about 5kDa), and about 2 to about 3 weight percent PLA-PEG-lys(urea)glu (16kDa/5kDa PLA-PEG).
  • a disclosed nanoparticle may include about 42 to about 45 weight percent PLA-PEG (with e.g. PLA about 16 kDa and PEG about 5kDa) about 42 to 45 weight percent PLA (e.g. about 75 kDa), about 10 to 15 weight percent active agent (e.g. docetaxel), and about about 2 to about 3 weight percent PLA-PEG-lys(urea)glu (16/5 PLA-PEG).
  • PLA-PEG with e.g. PLA about 16 kDa and PEG about 5kDa
  • PLA e.g. about 75 kDa
  • active agent e.g. docetaxel
  • PLA-PEG-lys(urea)glu (16/5 PLA-PEG
  • targeting moieties are targeted to an antigen associated with a disease of a patient's immune system or a pathogen-borne condition.
  • targeting moieties are targeted to cells present in normal healthy conditions. Such targeting moieties may be directly targeted to a molecule or other target or indirectly targeted to a molecule or other target associated with a biological molecular pathway related to a condition.
  • the amount of nanoparticles administered to a patient may vary and may depend on the size, age, and health of the patient, the therapeutic agent to be delivered, the disease being treated, and the location of diseased tissue. Moreover, the dosage may vary depending on the mode of administration.
  • nanoparticles are administered to a subject systemically.
  • methods of administration may include, but are not limited to, intravascular injection, intravenous injection, intraperitoneal injection, subcutaneous injection, and intramuscular injection.
  • the nanoparticles necessitate only a single or very few treatment sessions to provide effective treatment of disease, which ultimately may facilitate patient compliance.
  • administration of the nanoparticles can occur via intravenous infusion once every three weeks.
  • methods of treating solid tumors e.g. prostate, lung, breast or other cancers, comprising administering a disclosed nanoparticle composition to a patient, e.g. a mammal in need thereof.
  • the solid tumor may have significant concentration of therapeutic agent, e.g.
  • a solid tumor in a mammal comprising administering a nanoparticle composition comprising a plurality of nanoparticles each comprising a ⁇ -hydroxy polyester-co-polyether and a therapeutic agent, wherein the composition has an amount of therapeutic agent effective to inhibit the growth of said tumor, for example, wherein single dose of said composition provides extended release of said therapeutic agent for a least one day.
  • active agent e.g. docetaxel
  • Such methods may provide an actual peak plasma concentration (C max ) of the therapeutic agent after administration of the composition to the mammal that is at least 10% higher, or at least 20% higher or 100% higher or more than the C max of said therapeutic agent if administered in a non-nanoparticle formulation.
  • Disclosed methods may provide, upon administration of nanoparticles, an area under the plasma concentration time curve (AUC) in a patient that is increased by at least 100% over the AUC provided when the therapeutic agent is administered alone to a patient.
  • disclosed methods may also, alone or in addition to the above plasma parameters, decrease the volume of distribution (V 2 ) of the therapeutic agent upon administration by at least 50% as compared to the V z of the patient when the therapeutic agent is administered alone.
  • disclosed nanoparticles may, upon administration, provide a higher plasma concentration of therapeutic agent as compared to administering an equivalent dosage of therapeutic agent alone.
  • disclosed nanoparticles may circulate substantially in the vascular compartment, and therefore may not contribute significantly to other areas that may cause toxicity or unwanted side effects.
  • Example 1 In Vitro Release of Docetaxel from Nanoparticles
  • a suspension of docetaxel encapsulated in nanoparticles fabricated according to the emulsion process depicted in FIG. 2 and Example 12 using 87.5 weight percent PLA-PEG, 10 wt.% docetaxel, and 2.5 wt. percent docetaxel (Formulation A) (all docetaxel nanoparticle formulations used in these Examples were in a composition of 5% nanoparticles, 65% water, and 30% sucrose), was placed in a dialysis cassette and incubated in a reservoir of phosphate buffered saline (PBS) at 37 0 C with stirring.
  • PBS phosphate buffered saline
  • Figure 3 presents the in vitro release profile of docetaxel encapsulated in nanoparticles compared to conventional docetaxel. Release of the encapsulated docetaxel from the polymer matrix was essentially linear over the first 24 hours with the remainder gradually released over a period of about 96 hours.
  • Example 2 Single Dose Pharmacokinetic Study of Docetaxel Encapsulated in Nanoparticles and Conventional Docetaxel in Sprague-Dawley Rats
  • LC- MS liquid chromatography-mass spectrometry
  • Figure 4 and Example Table 2.1 present the observed pharmacokinetic profiles and pharmacokinetic parameters, respectively, of docetaxel encapsulated in nanoparticles and conventional docetaxel.
  • Example Table 2.1 further includes data from the preclinical development of TAXOTERE ® for comparative reference (Bissery et al. 1995). The results for conventional docetaxel were consistent with those reported in literature (Bissery et al. 1995), indicating docetaxel was rapidly cleared from the blood and distributed to tissues. The peak plasma concentration (C max ) was observed at the first sampling time point for all treatments. [0088] The C max and AUC of the docetaxel encapsulated in nanoparticles were approximately 100 times higher than that for conventional docetaxel.
  • the difference in the C max may be attributable to having missed the rapid initial tissue distribution phase for conventional docetaxel.
  • the data indicate that the docetaxel encapsulated in nanoparticles largely remains in circulation upon injection and is slowly cleared over a 24 hour period.
  • the data further shows that docetaxel is released from the nanoparticles in a controlled manner during the 24 hour period (e.g., rapid burst release is not observed). If the nanoparticles were very quickly cleared from circulation, the large increase in AUC would not be observed. Similarly, if there was rapid burst release of docetaxel from the nanoparticles, the pharmacokinetic profile would be expected to more closely resemble that of conventional docetaxel.
  • Example Table 2.1 Summary of Docetaxel Encapsulated in Nanoparticles and Conventional Docetaxel Pharmacokinetic Parameters
  • docetaxel was released very quickly from the nanoparticles, it would be expected to be rapidly distributed from the plasma, yielding a profile similar to that of conventional docetaxel. Conversely, if docetaxel was retained in the nanoparticles over this timeframe, the profiles of the docetaxel encapsulated in nanoparticles wherein the ligand of the PLA-PEG-lys(urea)glu targeting polymer was 14 C-labeled and the docetaxel encapsulated in nanoparticles wherein the encapsulated docetaxel was 14 C-labeled would be superimpo sable.
  • Example Tables 3.1, 3.2, and 3.3 present the tissue distribution of radioactivity determined in rats after intravenous (IV) administration of (1) docetaxel encapsulated in nanoparticles in which the ligand of the PLA-PEG-lys(urea)glu targeting polymer was R elabeled, (2) docetaxel encapsulated in nanoparticles in which the encapsulated docetaxel was 14 C-labeled, and (3) 14 C-labeled conventional docetaxel, respectively.
  • Example Figure 5 contains the radioactivity concentration curves of the test articles determined in plasma, liver, spleen, and bone marrow.
  • the concentration of docetaxel encapsulated in nanoparticles was higher in most tissues than conventional docetaxel. After 24 hours, the concentration of docetaxel derived from the nanoparticles was lower than or approximately the same as the concentration of conventional docetaxel in all of the tissues evaluated, except the spleen.
  • Marrow Intestine Intestine (h) (nCi/mL) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g)
  • Marrow Intestine Intestine (h) (nCi/mL) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g)
  • Marrow Intestine Intestine (h) (nCi/mL) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g)
  • Example 4 Tumor Targeting of Docetaxel Encapsulated in Nanoparticles and Conventional Docetaxel after a Single Dose in a Human Tumor Xenograft Model (LNCaP)
  • mice Male severe combined immunodeficiency (SCID) mice were subcutaneously inoculated with human LNCaP prostate cancer cells. Three to four weeks after inoculation, the mice were assigned to different treatment groups such that the average tumor volume in each group was 300 mm 3 . At this time, a single intravenous (IV) dose of 50 mg/kg docetaxel was administered as either docetaxel encapsulated in nanoparticles or conventional docetaxel. The test subjects were sacrificed 2 hour or 12 hour post-dose. The tumors from each group were excised and assayed for docetaxel using liquid chromatography-mass spectrometry (LC-MS).
  • IV intravenous
  • Example Table 4.1 Measured Docetaxel Concentration in Tumors Treated with Docetaxel Encapsulated in Nanoparticles and Conventional Docetaxel
  • mice Male severe combined immunodeficiency (SCID) mice were subcutaneously inoculated with human LNCaP prostate cancer cells. Three to four weeks after inoculation, the mice were assigned to different treatment groups such that the average tumor volume in each group was 250 mm 3 . Subsequently, the mice were treated every other day (Q2D) for four doses, with an eight day holiday, followed by another four doses at the Q2D schedule. [0101] Average tumor volumes for each treatment group are shown in Example Figure 7. Treatment with either conventional docetaxel or docetaxel encapsulated in nanoparticles resulted in appreciable reduction in tumor volume.
  • Tumor volume reduction was greater in test subjects receiving docetaxel encapsulated in nanoparticles compared to conventional docetaxel.
  • Example 9 Pharmacokinetics of paclitaxel passively targeted nanoparticles in Rats [0108] Similar to the procedure in Example 2, rats were intravenously dosed with 1.0 mg/kg with either nanoparticles prepared as in Figure 2 having paclitaxel and PLA-PEG (formulation C) and no specific targeting moiety (passively targeted nanoparticles (PTNP); or paclitaxel alone. The release profiles are shown in Figure 10.
  • Example 10 Pharmacokinetics of rapamycin (sirolimus) passively targeted nanoparticles in rats [0110] Similar to the procedure in Example 2, rats were intravenously dosed with 2.0 mg/kg with either nanoparticles prepared as in Figure 2 and Example 16, having rapamycin and PLA-PEG and no specific targeting moiety (passively targeted nanoparticles (PTNP); or rapamycin alone. The release profiles are shown in Figure 11.
  • Plasma samples were analyzed using LC/MS and the PK analysis was performed using WinNonlin software.
  • a comparison of the pharmacokinetics of the nanoparticles with rapamcyin alone is as follows:
  • Example 11 Tumor Accumulation of docetaxel nanoparticles in MX-I breast tumors in mice.
  • Mice with MX -1 breast tumors were randomized into three groups, receiving docetaxel (3 mice), passively targeted nanoparticles (Formulation A, without a targeting moiety, PTNP), or Formulation A.
  • the average tumor mass was 1.7g (RSD 34%).
  • Mice were then injected with 10 mg/kg of the test article, then euthanized 24 hours later and the tumors were removed and analyzed for docetaxel content using LC/MS/MS. Results are depicted in Figure 12.
  • the percent of injected dose in the tumor was 3% (for docetaxel alone), 30% for PTNP, and 30% Formulation A.
  • Example 12 Pharmacokinetics of docetaxel nanoparticles in primates [0113] Na ⁇ ve non human primates (3 male and 3 female) were administered docetaxel, docetaxel nanoparticles (Formulation A) or docetaxel nanoparticles (Formulation B: 43.25% PLA-PEG(16/5), 43.25% PLA (75kDa), 10% docetaxel, 2.5% PLA-PEG-lys(urea)glu, prepared as in Example 14), using and following appropriate ethical guidelines at all times. 1 male and 1 female were used per dose group.
  • the dosing day was 1 day and the formulations were administered by 30 minute IV infusion at 25 mg/m 2 docetaxel or 50 mg/m 2 docetaxel (animals were randomized and then dosed with 50 mg/m2 on day 29 and PK, hematology and clinical chemistry were measured for 21 days). At the end of the study, PK, hematology and clinical chemistry collected over a 21 day period were assessed.
  • Figure 12 depicts the results of male (M) and female (F) PNP.
  • a comparison of the pharmacokinetics of the nanoparticles of Formulation A (25 mg/m 2 dose) with docetaxel alone is as follows:
  • An organic phase is formed composed of a mixture of docetaxel (DTXL) and polymer (homopolymer, co-polymer, and co-polymer with ligand).
  • the organic phase is mixed with an aqueous phase at approximately a 1:5 ratio (oil phase:aqueous phase) where the aqueous phase is composed of a surfactant and some dissolved solvent.
  • aqueous phase is composed of a surfactant and some dissolved solvent.
  • about 30% solids in the organic phase is used.
  • the primary, coarse emulsion is formed by the combination of the two phases under simple mixing or through the use of a rotor stator homogenizer.
  • the rotor/stator yielded a homogeneous milky solution, while the stir bar produced a visibly larger coarse emulsion. It was observed that the stir bar method resulted in significant oil phase droplets adhering to the side of the feed vessel, suggesting that while the coarse emulsion size is not a process parameter critical to quality, it should be made suitably fine in order to prevent yield loss or phase separation. Therefore the rotor stator is used as the standard method of coarse emulsion formation, although a high speed mixer may be suitable at a larger scale.
  • the primary emulsion is then formed into a fine emulsion through the use of a high pressure homogenizer.
  • the fine emulsion is then quenched by addition to deionized water at a given temperature under mixing.
  • the emulsion is added to a cold aqueous quench under agitation. This serves to extract a significant portion of the oil phase solvents, effectively hardening the nanoparticles for downstream filtration. Chilling the quench significantly improved drug encapsulation.
  • the quench:emulsion ratio is approximately 5:1.
  • Tween 80 A solution of 35% (wt%) of Tween 80 is added to the quench to achieve approximately 2% Tween 80 overall After the emulsion is quenched a solution of Tween-80 is added which acts as a drug solubilizer, allowing for effective removal of unencapsulated drug during filtration. Table B indicates each of the quench process parameters.
  • Table B Summary quench process parameters.
  • the temperature must remain cold enough with a dilute enough suspension (low enough concentration of solvents) to remain below the T g of the particles. If the Q:E ratio is not high enough, then the higher concentration of solvent plasticizes the particles and allows for drug leakage. Conversely, colder temperatures allow for high drug encapsulation at low Q:E ratios (to -3:1), making it possible to run the process more efficiently.
  • the nanoparticles are then isolated through a tangential flow filtration process to concentrate the nanoparticle suspension and buffer exchange the solvents, free drug, and drug solubilizer from the quench solution into water. A regenerated cellulose membrane is used with with a molecular weight cutoffs (MWCO) of 300.
  • MWCO molecular weight cutoffs
  • a constant volume diafiltration (DF) is performed to remove the quench solvents, free drug and Tween-80.
  • buffer is added to the retentate vessel at the same rate the filtrate is removed.
  • Crossflow rate refers to the rate of the solution flow through the feed channels and across the membrane. This flow provides the force to sweep away molecules that can foul the membrane and restrict filtrate flow.
  • the transmembrane pressure is the force that drives the permeable molecules through the membrane.
  • the filtered nanoparticle slurry is then thermal cycled to an elevated temperature during workup.
  • a small portion typically 5-10% of the encapsulated drug is released from the nanoparticles very quickly after its first exposure to 25 0 C.
  • the nanoparticle suspension (concentration 50 mg/ml),is passed through a sterilizing grade filter (0.2 ⁇ m absolute).
  • Pre-filters are used to protect the sterilizing grade filter in order to use a reasonable filtration area/ time for the process. Filtration flow rate is -1.3 L/min/m 2 .
  • the filtration train is Ertel Alsop Micromedia XL depth filter M953P membrane
  • Example 14 Preparation of Nanoparticles with Long Release Properties
  • Nanoparticle batches were prepared using the general procedure of Example 12, with 80% (w/w) Polymer-PEG or Polymer-PEG with homopolymer PLA at 40% (w/w) each, with a batch of % total solids of 5%, 15% and 30%. Solvents used were: 21% benzyl alcohol and 79% ethyl acetate (w/w). For each 2 gram batch size, 400 mg of drug was used and 1.6 g of 16-5 Polymer-PEG or 0.8 g of 16-5 Polymer-PEG + 0.8 g of 10 kDa PLA (homopolymer) was used.
  • the organic phase (drug and polymer) is prepared in 2 g batches: To 20 mL scintillation vial add drug and polymer(s). The mass of solvents needed at % solids concentration is:: 5% solids: 7.98 g benzyl alcohol + 30.02 g ethyl acetate; 30% solids: 0.98 g benzyl alcohol + 3.69 g ethyl acetate.
  • aqueous solution is prepared with 0.5% sodium cholate, 2% benzyl alcohol, and 4% ethyl acetate in water. Add to the bottle 7.5g sodium cholate, 1402.5g of DI water, 30g of benzyl alcohol and 6Og of ethyl acetate, and mix on stir plate until dissolved. [0135] For the formation of emulsion, a ratio of aqueous phase to oil phase is 5: 1. The organic phase is poured into the aqueous solution and homogenized using IKA for 10 seconds at room temperature to form course emulsion.
  • the solution is fed through the homogenizer (110S) at 9 Kpsi (45 psi on gauge) for 2 discreet passes to form nanoemulsion.
  • the emulsion is poured into quench (D.I. water) at ⁇ 5°C while stirring on stir plate. Ratio of quench to emulsion is 8:1. 35% (w/w) Tween 80 is added in water to quench at ratio of 25:1 Tween 80 to drug.
  • the nanoparticles are concentrated through TFF and the quench is concentrated on TFF with 500 kDa Pall cassette (2 membrane) to -100 mL.
  • Diafiltering is used using -20 diavolumes (2 liters) of cold DI water, and the volume is brought down to minimal volume then collect final slurry, -100 mL.
  • the solids concentration of unfiltered final slurry is determined by the using tared 20 mL scintillation vial and adding 4 mL final slurry and dry under vacuum on lyo/oven and the weight of nanoparticles in the 4 mL of slurry dried down is determined.
  • Concentrated sucrose (0.666 g/g) is added to final slurry sample to attain 10% sucrose.
  • Solids concentration of 0.45 um filtered final slurry was determined by filtering about 5mL of final slurry sample before addition of sucrose through 0.45 ⁇ m syringe filter; to tared 20 mL scintillation vial add 4 mL of filtered sample and dry under vacuum on lyo/oven. [0138] The remaining sample of unfiltered final slurry was frozen with sucrose.
  • Drug was dissolved in the inner aqueous phase consisting of water with 1-arginine or NaOH used for solubilizing the drug.
  • the polymer (16-5 PLA-PEG) was dissolved in the oil phase organic solvent system, such as dichloromethane (DCM) at 20% solid concentration.
  • the outer aqueous phase consisted mainly of water with 1% sodium cholate (SC) as surfactant, unless noted otherwise.
  • the w/o emulsion was prepared by adding the inner aqueous phase into the oil phase under rotor stator homogenization or sonication (using Branson Digital Sonifier) at a w/o ratio of 1 : 10.
  • the coarse w/o/w emulsion was also prepared by adding the w/o emulsion into an outer aqueous phase under either rotor stator homogenization or sonication at o/w ratio of 1 : 10.
  • the fine w/o/w emulsion was then prepared by processing the coarse emulsion through a Microfluidics high pressure homogenizer (MHOS pneumatic) at 45000 psi with a 100 ⁇ m Z-interaction chamber.
  • the fine emulsion was then quenched into cold DI water at 10:1 quench: emulsion ratio.
  • Polysorbate 80 (Tween 80) was then added as a process solubilizer to solubilize the unencapsulated drug. No drug precipitation was observed at a drug:Tween 80 ratio of 1:200.
  • the batch was then processed with ultrafiltration followed by diafiltration to remove solvents, unencapsulated drug and solubilizer. The particle size measurements were performed by Brookhaven DLS and/or Horiba laser diffraction. To determine drug load, slurry samples were analyzed by HPLC and solid concentration analysis. The slurry retains were then diluted with sucrose to 10% before freezing. All ratios listed are on a w/w basis, unless specified otherwise.
  • both the inner w/o and outer w/o/w emulsions were formed by rotor stator homogenization followed by 2 passes at 45k psi using a high pressure homogenizer.
  • the nanoparticle suspension was quenched in cold DI water followed by ultrafiltration/diafiltration work-up. HPLC and PSD analysis was used to determine that the drug load stayed at 0.38% for 131 nm particles.
  • An organic phase is formed composed of a mixture of sirolimus and polymer
  • the organic phase is mixed with an aqueous phase at approximately a 1:5 ratio (oil phase:aqueous phase) where the aqueous phase is composed of a surfactant and some dissolved solvent.
  • aqueous phase is composed of a surfactant and some dissolved solvent.
  • about 30% solids in the organic phase is used.
  • the primary, coarse emulsion is formed by the combination of the two phases under simple mixing or through the use of a rotor stator homogenizer.
  • the primary emulsion is then formed into a fine emulsion through the use of a high pressure homogenizer. The process is continued as in Example 12.

Abstract

The present disclosure is directed in part to a biocompatible nanoparticle composition comprising a plurality of non-colloidal long circulating nanoparticles, each comprising a α-hydroxy polyester-co-polyether and a therapeutic agent, wherein such disclosed compositions provide a therapeutic effect for at least 12 hours.

Description

LONG CIRCULATING NANOPARTICLES FOR SUSTAINED RELEASE OF
THERAPEUTIC AGENTS
RELATED APPLICATIONS
[0001] This application claims priority to provisional applications U. S. S.N. 61/122,479, filed December 15, 2008, U.S.S.N. 61/260,200, filed November 11, 2009, and U.S.S.N 61/249,022, filed October 6, 2009, each of which is hereby incorporated by reference in its entirety. BACKGROUND
[0002] Nanoparticles for the delivery of therapeutic agents have the potential to circumvent many challenges associated with traditional delivery approaches including lack of patient compliance to prescribed therapy, adverse effects, and inferior clinical efficacy due to lack of targeted delivery. Important technological advantages of nanoparticles for drug delivery include the ability to deliver water-insoluble and unstable drugs, incorporation of both hydrophobic and hydrophilic therapeutic agents, and ability to utilize various routes of administration. Nanoparticle delivery systems may also facilitate targeted drug delivery and controlled release applications, enhance drug bioavailability at the site of action, reduce dosing frequency, and minimize side effects. [0003] Because of these possible advantages, nanoparticulate systems have been examined for use as drug delivery vehicles, including polymeric micelles, polymers, liposomes, low-density lipoproteins, dendrimers, hydrophilic drug -polymer complexes, and ceramic nanoparticles. Typical polymeric materials utilized in polymeric particulate drug delivery systems include polylactic acid (PLA), poly(D,L-glycolide) (PLG), and poly(lactide-co- glycolide) (PLGA). PLA and PLGA are listed as Generally Recognized as Safe (GRAS) under Sections 201(s) and 409 of the Federal Food, Drug, and Cosmetic Act, and are approved for use in commercially available microparticulate systems, including Decapeptyl®, Parlodel LA®, and Enantone Depot®, as well as in implant devices, such as Zoladex®.
[0004] However, certain nanoparticle systems, such as liposomes, are not amenable for use with certain therapeutic agents. Polymeric nanoparticles developed to date have limited effectiveness, in part because such nanoparticles clear from the body quickly once administered and/or may accumulate in healthy tissue where treatment is not needed. Control of delivery of an active agent, using nanosystems, remains a challenge.
[0005] Therefore there is a need for biocompatible compositions capable of extended delivery of active agents, e.g., anti-neoplastic agents, that provide for prolonged and/or increased plasma drug concentrations in a patient, especially as compared to administration of an active agent alone.
SUMMARY
[0006] In one aspect of the invention, a nanoparticle composition is provided that includes a biodegradable and/or biocompatible polymer and a therapeutic agent, wherein the biodegradable and/or biocompatible polymer matrix releases the therapeutic agent at a rate allowing controlled release of the agent over at least about 12 hours, or in some embodiments, at least about 24 hours For example, provided herein is a biocompatible nanoparticle composition comprising a plurality of long circulating nanoparticles, each comprising a biocompatible polymer and a therapeutic agent, said composition providing an elevated plasma concentration of the therapeutic agent for at least 12 hours when the composition is administered to a patient, and an area under the plasma concentration time curve (AUC) that is increased by at least 100% over the AUC provided when the therapeutic agent is administered alone to a patient. [0007] In an embodiment, disclosed herein is a biocompatible nanoparticle composition comprising a plurality of long circulating nanoparticles, each comprising a α-hydroxy polyester-co-polyether and a therapeutic agent, said composition providing an elevated plasma concentration of the therapeutic agent for at least 6 hours, at least 12 hours, or at least 24 hours or more when the composition is administered to a patient, to provide an area under the plasma concentration time curve (AUC) that is increased by at least 100%, or at least by 150%, over the AUC provided when the therapeutic agent is administered alone to a patient. [0008] In some embodiments, disclosed nanoparticles may provide an actual peak plasma concentration (Cmax) that is at least 10% higher, or even at least 100% higher, as compared to a Cmax of said therapeutic agent when administered alone. Disclosed nanoparticles, for example, may provide a volume of distribution when administered to the patient that is less than or equal to about 5 plasma volumes. Fo example, dislcosed nanoparticles and/or compositions may decrease the volume of distribution (V2) by at least 50% as compared to the Vz of the patient when the therapeutic agent is administered alone. [0009] Disclosed biocompatible nanoparticle compositions may include long circulating nanoparticles that may further comprise a biocompatible polymer coupled to a targeting moiety, for example, a targeting moiety that is selected from the group consisting of a protein, peptide, antibody, antibody fragment, saccharide, carbohydrate, small molecule, glycan, cytokine, chemokine, nucleotide, lectin, lipid, receptor, steroid, neurotransmitter, cell surface marker, cancer antigen, or glycoprotein antigen. An exemplary targeting moiety may bind to prostate membrane specific antigen (PMSA). For example, a disclosed nanoparticle may include a biocompatible polymer coupled to a targeting moiety, e.g., a nanoparticle may include PLA-PEG-(^, S-2- { 3-[ 1 -carboxy-5-amino-pentyl] -ureido } -pentanedioic acid. Disclosed long circulating nanoparticles may include 1 to about 4% by weight, or 2% to about 4% by weight, of a biocompatible polymer coupled to a targeting moiety [0010] In some embodiments, a biocompatible nanoparticle may include a biocompatible polymer such as PLA-PEG. For example, a α-hydroxy polyester-co-polyether may be polylactic acid -co-polyethylene glycol, and/or a α-hydroxy polyester-co-polyether comprises about 16kDa polylactic acid and about 5 kDa polyethylene glycol. [0011] Disclosed long circulating nanoparticles may be about 80 to about 90 weight percent α-hydroxy polyester-co-polyether. [0012] In some embodiments, disclosed long circulating nanoparticles may further comprise a biodegradable polymer, such as poly(lactic) acid. For example, long circulating nanoparticles may have about 40 to about 50 weight percent poly(lactic)acid, and about 40 to about 50 weight percent of α-hydroxy polyester-co-polyether. Compositions that include such biocompatible nanoparticles and a therapeutic agent may provide a peak plasma concentration (Cmax) of a therapeutic agent at least 100% higher than the Cmax of the therapeutic agent when administered alone, and/or the area under the plasma concentration time curve (AUC) mayincreased by at least 200% over the AUC of the therapeutic agent when administered alone to the patient.
[0013] Disclosed nanoparticle compositions may include a therapeutic agent such as one selected from the group consisting of chemotherapeutic agents, diagnostic agents, prophylactic agents, nutraceutical agents, nucleic acids, proteins, peptides, lipids, carbohydrates, hormones, small molecules, metals, ceramics, drugs, vaccines, immunological agents, and combinations thereof, for example, a nanoparticle may include an anti-neoplastic agent such as docetaxel, vincristine, methotrexate, paclitaxel, or sirolimus. Disclosed nanoparticle compositions may further include an aqueous solution of a saccharide. [0014] Also provided herein is a method of treating a solid tumor cancer, comprising administering disclosed nanoparticle composition to a patient (e.g. a mammal or primate) in need thereof. Such methods, may provide wherein at least 24 hours after administration, a solid tumor has significant concentration of therapeutic agent. Contemplated herein is a method of treating a solid tumor in a mammal in need thereof, comprising administering a nanoparticle composition comprising a plurality of nanoparticles each comprising a α-hydroxy polyester-co-polyether and a therapeutic agent, wherein the composition has an amount of therapeutic agent effective to inhibit the growth of said tumor, for example, a single dose of said composition may provide extended elevated plasma concentrations of said therapeutic agent in the patient for a least one day, (e.g.. the peak plasma concentration (Cmax) of the therapeutic agent after administration of the composition to the mammal is at least 10% higher than the Cmax of said therapeutic agent if administered in a non-nanoparticle formulation. ) [0015] Also provided herein is a method of minimizing unwanted side effects or toxicity of an active agent in a patient, comprising: administering a nanoparticle composition comprising a plurality of nanoparticles each comprising a α-hydroxy polyester-co-polyether and a therapeutic agent, wherein said composition is capable of delivery a higher plasma concentration of therapeutic agent to the patient as compared to administering the therapeutic agent alone, and wherein upon administering the nanoparticle composition the volume distribution of the active agent in the patient is reduced, as compared to the volume distribution if the therapeutic agent was administered alone. A method for modulating the plasma concentration of a therapeutic agent in a patient, e.g. a primate (e.g. human) is also provided, comprising: providing a polymeric nanoparticle comprising the therapeutic agent and administering the polymeric nanoparticle to the patient, thereby modulating the plasma concentration of the human patient. [0016] The invention may be more completely understood in consideration of the following detailed description of various embodiments of the invention in connection with the accompanying drawings.
BRIEF DESCRIPTION OF THE DRAWINGS [0017] FIG. 1 is a schematic illustration of a nanoparticle according to one aspect of the present invention.
[0018] FIG. 2 is a block diagram of the emulsion process used in the fabrication of nanoparticles in one aspect of the present invention.
[0019] FIG. 3 depicts the in vitro release of docetaxel from nanoparticles and conventional docetaxel.
[0020] FIG. 4 depicts the pharmacokinetics of docetaxel encapsulated in nanoparticles and conventional docetaxel in rats.
[0021] FIG. 5 depicts the distribution of radioactivity determined in selected tissues of rats after IV administration of nanoparticles containing 14C-targeting polymer ( ), nanoparticles containing 14C-docetaxel (ss), and conventional 14C -docetaxel (V).
[0022] FIG. 6 depicts docetaxel concentration in tumor tissue after administration of docetaxel encapsulated in nanoparticles or conventional docetaxel to LNCaP tumor bearing
SCID mice.
[0023] FIG. 7 depicts the reduction in tumor volume in mice with PSM A-expres sing LNCaP xenografts when treated with docetaxel encapsulated in nanoparticles or conventional docetaxel.
[0024] FIG. 8 depicts pharmacokinetics of vincristine encapsulated in disclosed nanoparticles and conventional vincristine in rats. [0025] FIG. 9 depicts pharmacokinetics of methotrexate encapsulated in disclosed nanoparticles and conventional methotrexate in rats.
[0026] FIG. 10 depicts pharmacokinetics of paclitaxel encapsulated in disclosed nanoparticles and conventional paclitaxel in rats. [0027] FIG. 11 depicts pharmacokinetics of rapamycin (sirolimus) encapsulated in disclosed nanoparticles and conventional rapamycin in rats.
[0028] FIG. 12 depicts the tumor accumulation of docetaxel in disclosed nanoparticles in a MX-I mouse breast tumor model.
[0029] FIG. 13 depicts pharmacokinetics of docetaxel in a NHP model using various disclosed nanoparticles.
DETAILED DESCRIPTION
[0030] It is to be understood that the invention is not limited to the particular processes, compositions, or methodologies described, as these may vary. It is also to be understood that the terminology used in the description is for the purpose of describing particular versions or embodiments only and is not intended to limit the scope of the invention. All of the publications and references mentioned herein are incorporated by reference. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.
[0031] As used herein the singular forms "a", "an" and "the" include plural reference unless the context clearly dictates otherwise. Further, unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art. [0032] As used herein, the term "about" means plus or minus 10% of the numerical value of the number with which it is being used. Therefore, about 50% means in the range of
40%-60%.
[0033] An "effective amount" or "therapeutically effective amount" of a composition, as used herein, is a predetermined amount calculated to achieve a desired effect.
[0034] As used herein, the term "long-circulating" refers to enhanced stability in the circulatory system of a patient, regardless of biological activity.
[0035] As used herein, the prefix "nano" and the terms "nanophase" and "nanosize" refer to a special state of subdivision implying that a particle has an average dimension smaller than about 1000 nm ( 100Ox 109 m) .
[0036] As used herein, the terms "polyethylene glycol)" or "PEG" and "poly(ethylene oxide)" or "PEO" denote polyethers comprising repeat -CH2-CH2-O- units. PEG and/or PEO can be different polymers depending upon end groups and molecular weights. As used herein, poly(ethylene glycol) and PEG describes either type of polymer. [0037] An "α-hydroxy polyester" refers to polymers having monomers based on one or more α-hydroxy acid, such as poly(lactic) acid, poly(glycolic) acid, poly-lactic-co-glycolic acid, polycaprolactone.
[0038] The term "target", as used herein, refers to the cell type or tissue to which enhanced delivery of the therapeutic agent is desired. For example, diseased tissue may be a target for therapy.
[0039] As used herein, the term "therapeutic agent" means a compound utilized to image, impact, treat, combat, ameliorate, prevent or improve an unwanted condition or disease of a patient. [0040] In an embodiment, disclosed long-circulating nanoparticles include a therapeutic agent and biodegradable and/or biocompatible polymeric particles, optionally functionalized with targeting moieties. The nanoparticles are designed to circulate in a vascular compartment of a patient for an extended period of time, distribute and accumulate at a target, and release the encapsulated therapeutic agent in a controlled manner. These characteristics can result in an increased level of therapeutic agent in the target and a potential reduction in off-target exposure. For example, the disclosed nanoparticles remain in circulation longer because, upon administration to a patient (e.g. a mammal, primate (e.g. human)), the disclosed nanoparticles are substantially confined to the vascular compartment of the patient, and are engineered to be cleared very slowly.
[0041] The activity of many therapeutic agents is dependent on their pharmacokinetic behavior. This pharmacokinetic behavior defines the drug concentrations and period of time over which cells are exposed to the drug. For most therapeutics, e.g. anti-neoplasties, longer exposure times are preferred as this results in increased killing of the cancer cells. In general, several parameters are used to describe drug pharmacokinetics. Peak plasma concentration, or maximum plasma concentration (Cmax) and area under the curve (AUC) are examples. AUC is a measure of plasma drug levels over time and provides an indication of the total drug exposure. Generally, plasma concentration and plasma AUC for a therapeutic agent correlate with increased therapeutic efficacy. [0042] The combination of long circulation time, confinement of particles to the vascular compartment and controlled release of drug results in higher circulating drug concentrations for longer periods of time (as evidenced by higher AUC and lower Vd). than drug alone, or, for example, drug in a PLA polymeric nanoparticles that does not include PLA- PEG, or that do not include e.g. PLA alone. [0043] For example, provided herein, in an embodiment, is a biocompatible nanoparticle composition comprising a plurality of long circulating nanoparticles, each comprising a α-hydroxy polyester-co-polyether and a therapeutic agent. Such compositions may provide a therapeutic effect for at least 12 hours, at least 24 hours, or at least 36 hours, or 48 hours or more, upon administration to a patient. In some embodiments, peak plasma concentration (Cmax) of the therapeutic agent of such nanoparticles, e.g. when the composition is administered to a patient, may be least 10% higher, 20% higher, or about 10% to about 100% higher, or more, than the Cmax of the same therapeutic agent when administered alone. Actual peak plasma concentration of delivered therapeutic agent includes both agent that is released from the nanoparticle (e.g. after administration) and therapeutic agent remaining in any nanoparticle remaining in the plasma, e.g. at a given time.
[0044] In another embodiment, disclosed nanoparticles may provide upon administration to a patient, an area under the plasma concentration time curve (AUC), that may be increased by at least 100%, at least 200% , or about 100% to about 500% or more, over the AUC of the therapeutic agent when administered alone to the patient. In another embodiment, a provided composition that includes disclosed nanoparticles may decrease the volume of distribution(Vz) of distributed active agent, upon administration, in a patient by at least 10%, or by at least 20%, or about 10% to about 100%, as compared to the Vz of the patient when the therapeutic agent is administered alone. For example, a provided nanoparticle composition may provide Vz in a patient that is on the same order of magnitude that the of plasma volume and/or a volume of distribution less than about 10 plasma volumes. For example, a disclosed nanoparticle composition may provide a Vz that is less than, or about, 2 times the plasma volume, or less than or about 8 plasma volumes. In an embodiment, a disclosed nanoparticle composition may provide a Vz in a patient that is on about the same order of plasma volume, (e.g. about 5L for an exemplary 70 kg patient), e.g. about a Vz that indicates administered nanoparticles are substantially in the patient's plasma and not substantially in other tissues. [0045] In some embodiments, disclosed nanoparticles may be used as a drug delivery vehicle based on the encapsulation of a therapeutic agent in a polymer matrix with controlled porosity and/or a soluble shell or matrix that upon dissolution releases the therapeutic agent in the immediate vicinity of the targeted area. The protection of the therapeutic agent provided by the polymer shell or matrix allows for the delivery of therapeutic agents that are water- insoluble or unstable. Furthermore, dissolution kinetics of the polymer can be designed to provide sustained release of therapeutic agents at a target for an extended period of time. [0046] Disclosed nanoparticles can be used for a variety of applications, such as, without limitation, drug delivery, gene therapy, medical diagnosis, and for medical therapeutics for cancer, pathogen-borne diseases, hormone-related diseases, reaction-by-products associated with organ transplants, and other abnormal cell or tissue growth. [0047] Provided herein, in an embodiment, are methods for treating a patient e.g. a mammal suffering from cancer, e.g. a solid tumor cancer, prostate cancer, breast cancer or lung cancer using e.g., disclosed nanoparticles. However, contemplated diseases that may be treated using disclosed nanoparticles include a broad range of diseases and find limitation only by e.g. the therapeutic agent, the availability of a marker and/or a targeting ligand for the disease. [0048] In other embodiments, a nanoparticle delivery system is provided that mitigates against colloidal instability, agglomeration, polydispersity in nanoparticle size and shape, swelling, and leakage of encapsulated materials.
[0049] In yet another embodiment, nanoparticles for delivery of therapeutic agents are provided that exhibit encapsulation efficiency. Encapsulation efficiency is affected by factors including, for example, material characteristics of the polymer utilized as the carrier matrix, the chemical and physical properties of the therapeutic agent to be encapsulated, and type of solvents used in the nanoparticle fabrication process.
[0050] In yet another aspect, polymeric nanoparticles for delivery of therapeutic agents are provided that exhibit particle heterogeneity. Conventional polymeric nanoparticle fabrication techniques generally provide multimodal particle size distributions as a result of self- aggregation during nanoprecipitation of both the polymer and the drug molecules. [0051] Polymeric nanoparticles for delivery of therapeutic agents are provided, in an embodiment, that may reduce or eliminate burst release effects. Conventional polymeric nanoparticle carriers frequently exhibit a bimodal drug release pattern with up to about 40-80% or more of the encapsulated drug released during the first several hours. After 24 to 48 hours, drug release is significantly reduced due to the increased diffusion barrier for drug molecules located deep within the polymer matrix. In such conventional nanoparticle carrier systems, poorly encapsulated drug molecules diffuse quickly into solution, which may lead to significant toxicity in vivo. Further, by the time the evacuated nanoparticles arrive and accumulate at the targeted site (e.g., tumor tissue), the nanoparticles generally have little or no remaining therapeutic efficacy.
[0052] In an embodiment, polymeric nanoparticles for delivery of therapeutic agents are provided that may evade rapid capture by the reticuloendothelial system (RES), leading to extended circulation time and elevated concentration of the nanoparticles in the blood. Rapid capture and elimination is typically caused by the process of opsonization in which opsonin proteins present in the blood serum quickly bind to conventional nanoparticles, allowing macrophages to easily recognize and remove these particulates before they can perform their designed therapeutic function. The extent and nature of opsonin adsorption at the surface of nanoparticles and their simultaneous blood clearance depend on the physicochemical properties of the particles, such as size, surface charge, and surface hydrophobicity. In yet another embodiment, a nanoparticle composition is provided including a biodegradable and/or biocompatible polymer matrix and a therapeutic agent coupled to the biodegradable and/or biocompatible polymer matrix wherein the clearance rate of said therapeutic agent coupled to the biodegradable and/or biocompatible polymer matrix is lower than the clearance rate of said therapeutic agent when administered alone.
[0053] In certain embodiments, methods are provided that mask or camouflage nanoparticles in order to evade uptake by the RES. One such method is the engineering of particles in which polyethers, such as poly(ethylene glycol) (PEG) or PEG containing surfactants, are deployed on the surface of nanoparticles. The presence of PEG and/or PEG- containing copolymers, e.g. on the surface of nanoparticles can result in an increase in the blood circulation half-life of the nanoparticles by several orders of magnitude. This method creates a hydrophilic protective layer around the nanoparticles that is able to repel the absorption of opsonin proteins via steric repulsion forces, thereby blocking and delaying the first step in the opsonization process.
[0054] FIG. 1 schematically illustrates a nanoparticle according to one aspect of the present invention. As shown in FIG. 1, docetaxel 100, an anti-neoplastic agent approved for the treatment of hormone refractory prostate cancer (HRPC), is encapsulated in a matrix 110 derived from the biodegradable and/or biocompatible polymers PLA and poly(lactide-b- ethylene glycol) (PLA-PEG). The polymer matrix 110 contains a targeting polymer (PLA-
PEG-lys(urea)glu) 120 that is end-functionalized (through the 5 amino moiety) with the lysine - urea-glutamate heterodimer (S,S-2-{3-[l-carboxy-5-amino-pentyl]-ureido}-pentanedioic acid ( lys(urea)glu) 130, a small molecule ligand that selectively binds to PSMA, a clinically relevant prostate cancer cell surface marker. [0055] Once the nanoparticles, e.g. as provided herein are administered, at least portions of the nanoparticle polymer(s) may be biologically degraded by, for example, enzymatic activity or cellular machinery into monomers and/or other moieties that cells can either use or excrete. In certain aspects of the invention, the dissolution or degradation rate of the nanoparticles is influenced by the composition of the polymer shell or matrix. For example, in some embodiments, the half-life of the polymer (the time at which 50% of the polymer is degraded into monomers and/or other nonpolymeric moieties) may be on the order of days, weeks, months, or years, depending on the polymer. [0056] According to some aspects of the invention, nanoparticle delivery characteristics such as water uptake, controlled release of therapeutic agent, and polymer degradation kinetics may be optimized through selection of polymer shell or matrix composition. [0057] Suitable polymers that may form some of the disclosed nanoparticles may include, but are not limited to, biodegradable α-hydroxy polyesters and biocompatible polyethers. In some aspects, exemplary polyesters include, for example, PLA, PLGA, PEG, PEO, PEGylated polymers and copolymers of lactide and glycolide (e.g., PEGylated PLA, PEGylated PGA, PEGylated PLGA), and derivatives thereof. In other aspects, suitable polymers include, for example, polyanhydrides, poly(ortho ester) PEGylated poly(ortho ester), poly(caprolactone), PEGylated poly(caprolactone), polylysine, PEGylated polylysine, poly(ethylene inline), PEGylated poly( ethylene imine), poly(L-lactide-co-L- lysine), poly(serine ester), poly(4-hydroxy-L-proline ester), poly[a-(4-aminobutyl)-L- glycolic acid], and combinations and derivatives thereof.
[0058] In other aspects, a polymer matrix may comprise one or more acrylic polymers.
Exemplary acrylic polymers include, for example, acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, aminoalkyl methacrylate copolymer, poly( acrylic acid), poly(methacrylic acid), methacrylic acid alkylamide copolymer, poly(methyl methacrylate), poly(methacrylic acid polyacrylamide) copolymer, aminoalkyl methacrylate copolymer, glycidyl methacrylate copolymers, polycyanoacrylates, and combinations thereof. The matrix may include dextran, acylated dextran, chitosan (e.g., acetylated to various levels), poly( vinyl) alcohol (for example, hydrolyzed to various degrees), and/or alginate, e.g. alginate complexed to bivalent cations such as a calcium alginate complex.
[0059] Nanoparticles disclosed herein include one, two, three or more biocompatible and/or biodegradable polymers. For example, a contemplated nanoparticle may include about 10 to about 99 weight percent of one or more block co-polymers that include a biodegradable polymer and polyethylene glycol, and about 0 to about 50 weight percent of a biodegradable homopolymer. Exemplary therapeutic nanoparticles may include about 40 to about 90 weight percent poly(lactic) acid-poly(ethylene)glycol copolymer or about 40 to about 80 weight percent poly(lactic) acid-poly(ethylene)glycol copolymer. Such poly(lactic) acid- block - poly(ethylene)glycol copolymer may include poly(lactic acid) having a number average molecular weight of about 15 to 20 kDa (or for example about 15 to about 10OkDa, e.g., about 15 to about 8OkDa), and poly(ethylene)glycol having a number average molecular weight of about 2 to about 1OkDa, for example, about 4 to about 6 kDa. For example, a disclosed therapeutic nanoparticle may include about 70 to about 90 weight percent PLA-PEG and about 5 to about 25 weight percent active agent (e.g. docetaxel), or about 30 to about 50 weight percent PLA-PEG, about 30 to about 50 weight percent PLA or PLGA, and about 5 to about 25 weight percent active agent (e.g. doxetaxel). Such PLA ((poly)lactic acid) may have a number average molecular weight of about 5 to about 10 kDa. Such PLGA (poly lactic -co-glycolic acid) may have a number average molecular weight of about 8 to about 12 kDa. It should be appreciated that disclosed PLA-PEG copolymers may include a chemical linker, oligomer, or polymer chain between the PLA and PEG blocks, e.g., may include PLA-linker-PEG. [0060] For example, disclosed nanoparticles may include about 10 to 15 weight percent active agent (e.g. about 10 weight percent docetaxel), and about 86 to about 90 weight percent PLA-PEG (with e.g. PLA about 16 kDa and PEG about 5kDa, e.g. about 87.5% PLA-
PEG(16kDa/5kDa)), and optionally e.g. a PLA-PEG-lys(urea)-glu (e.g. at 2.5 weight percent). [0061] Alternatively, a disclosed nanoparticle, which may have slow release properties, may include about 42 to about 45 weight percent PLA-PEG (with e.g. PLA about 16 kDa and PEG about 5kDa), (e.g. 43.25% PLA-PEG), about 42 to 45 weight percent PLA (e.g. about 75 kDa) (e.g. 43.25% PLA/75kDa) and about 10 to 15 weight percent active agent (e.g. docetaxel). For example, disclosed nanoparticles may optionally include about 1 to about 50 weight percent poly(lactic) acid or poly(lactic) acid-co-poly (glycolic) acid (which does not include PEG, e.g a homopolymer of PLA), or may optionally include about 1 to about 50 weight percent, or about 10 to about 50 weight percent or about 30 to about 50 weight percent poly(lactic) acid or poly(lactic) acid-co-poly (glycolic) acid. In an embodiment, disclosed nanoparticles may include two polymers, e.g. PLA-PEG and PLA, in a weight ratio of about 30:60 to about 60: 30, e.g, about 40:60, about 60:40, or about 50:50.
[0062] Such substantially homopolymeric poly(lactic) or poly(lactic)-co-poly(glycolic) acid may have a weight average molecular weight of about 10 to about 130 kDa, for example, about 20 to about 30 kDa, or about 100 to about 130 kDa. Such homopolymeric PLA may have a number average molecule weight of about 5 to about 90 kDa, or about 5 to about 12 kDa, about 15 to about 30 kDa, or about 60 to about 90 kDa. Exemplary homopolymeric PLA may have a number average molecular weight of about 80 kDa or a weight average molecular weight of about 124 kDa. As is known in the art, molecular weight of polymers can be related to an inherent viscosity. In some embodiments, homopolymer PLA may have an inherent viscosity of about 0.2 to about 0.4, e.g. about 0.3; in other embodiments, PLA may have an inherent viscosity of about 0.6 to about 0.8. Exemplary PLGA may have a number average molecular weight of about 8 to about 12 kDa. [0063] In other embodiments, modified surface chemistry and/or small particle size of disclosed nanoparticles may contribute to the effectiveness of the nanoparticles in the delivery of a therapeutic agent. For example, in one disclosed aspect, nanoparticle surface charge may be modified to achieve slow biodegradation and reduce clearance of the nanoparticles. In another aspect, porosity of the polymer shell or matrix is optimized to achieve extended and controlled release of the therapeutic agent. For example, in one embodiment of the invention, the nanoparticles may have porosity in the range of about 10 to about 90 percent and/or a pore diameters in the range of about 0.001 to about 0.01 microns. Further, without wishing to be bound by theory, because of their small size and persistence in the circulation, the nanoparticles according to some embodiments of the invention may be able to penetrate the altered and often compromised vasculature of tumors via the enhanced permeability and retention (EPR) effect resulting in preferential accumulation of nanoparticles in tumor interstitium. [0064] Examples of therapeutic agents that may form part of the disclosed nanoparticles include, but are not limited to, chemotherapeutic agents (e.g. anti-cancer agents), diagnostic agents (e.g. contrast agents, radionuclides, and fluorescent, luminescent, and magnetic moieties), prophylactic agents (e.g. vaccines), nutraceutical agents (e.g. vitamins and minerals), nucleic acids (e.g., siRNA, RNAi, and mircoRNA agents), proteins (e.g. antibodies), peptides, lipids, carbohydrates, hormones, small molecules, metals, ceramics, drugs, vaccines, immunological agents, and/or combinations thereof. For example, the active agent or drug may be a therapeutic agent such as an antineoplastic such as a mTor inhibitor (e.g., sirolimus (rapamycin), temsirolimus, or everolimus), vinca alkaloids such as vincristine, a diterpene derivative, a taxane such as paclitaxel (or its derivatives such as DHA-paclitaxel or PG- paxlitaxel), docetaxel, or methatrexate.
[0065] In some aspects of the invention, the therapeutic agent to be delivered is an agent useful in the treatment of cancer (e.g., a solid tumor cancer e.g., prostate or breast cancer). Such therapeutic agents may include, for example, doxorubicin (adriamycin), gemcitabine (gemzar), daunorubicin, procarbazine, mitomycin, cytarabine, etoposide, methotrexate, 5-fluorouracil (5-FU), vinblastine, vincristine, bleomycin, paclitaxel (taxol), docetaxel (taxotere), mitoxantrone, mitoxantrone hydrochloride, aldesleukin, asparaginase, busulfan, carboplatin, cladribine, camptothecin, CPT-Il, lO-hydroxy-7-ethylcamptothecin (SN38), dacarbazine, S-I capecitabine, ftorafur, 5'deoxyflurouridine, UFT, eniluracil, deoxycytidine, 5-azacytosine, 5-azadeoxycytosine, allopurinol, 2-chloroadenosine, trimetrexate, aminopterin, methylene- 10-deazaaminopterin (MDAM), oxaplatin, picoplatin, tetraplatin, satraplatin, platinum-DACH, ormaplatin, CI-973, JM-216, and analogs thereof, epirubicin, etoposide phosphate, 9- aminocamptothecin, 10,11- methylenedioxycamptothecin, karenitecin, 9-nitrocamptothecin, TAS 103, vindesine, L- phenylalanine mustard, ifosphamidemefosphamide, perfosfamide, trophosphamide carmustine, semustine, epothilones A-E, tomudex, 6-mercaptopurine, 6-thioguanine, amsacrine, etoposide phosphate, karenitecin, acyclovir, valacyclovir, ganciclovir, amantadine, rimantadine, lamivudine, zidovudine, bevacizumab, trastuzumab, rituximab and combinations thereof.
[0066] In some embodiments, contemplated nanoparticles may include more than one therapeutic agent. Such nanoparticles may be useful, for example, in aspects where it is desirable to monitor a targeting moiety as such moiety directs a nanoparticle containing a drug to a particular target in a subject. [0067] Disclosed nanoparticles may be formed using an emulsion process, e.g. as presented as a block diagram in FIG. 2. As shown in FIG. 2, an organic polymer/drug solution containing docetaxel, PLA, PLA-PEG, and PLA-PEG-lys(urea)glu dissolved in a co-solvent mixture of ethyl acetate and benzyl alcohol is dispersed in an aqueous solution of sodium cholate, ethyl acetate, and benzyl alcohol to form a coarse emulsion. In some aspects the conditions under which the emulsion process is performed favor the orientation of the PEG and/or PEG-lys(urea)glu polymer chains toward the particle surface. In other aspects, an orientation is achieved where the PEG is folded within the nanoparticle polymer shell or matrix. [0068] As presented in FIG. 2, a coarse emulsion can be passed through a high pressure homogenizer to reduce the droplet size, forming a fine emulsion. The fine emulsion is diluted into an excess volume of a quench solution of cold water containing polysorbate 80. The presence of polysorbate 80 serves to remove excess therapeutic agent that has not been encapsulated in the nanoparticle. In some aspects of the present invention, polysorbate 80 may also be adhered or associated with a nanoparticle surfaces. While not wishing to be bound by theory, polysorbate 80 coupled to the nanoparticle surfaces may impact characteristics such as controlled release of therapeutic agent and polymer degradation kinetics. Quenching may be performed at least partially at a temperature of about 5 0C or less. For example, water used in the quenching may be at a temperature that is less that room temperature (e.g., about 0 to about 1O0C, or about 0 to about 5 0C).
[0069] In some embodiments, not all of the therapeutic agent (e.g., docetaxel) is encapsulated in the particles at this stage, and a drug solubilizer is added to the quenched phase to form a solubilized phase. The drug solubilizer may be for example, Tween 80, Tween 20, polyvinyl pyrrolidone, cyclodextran, sodium dodecyl sulfate, or sodium cholate. For example, Tween-80 may added to the quenched nanoparticle suspension to solubilize the free drug and prevent the formation of drug crystals. In some embodiments, a ratio of drug solubilizer to therapeutic agent (e.g., docetaxel) is about 100:1 to about 10:1.
[0070] Ethyl acetate and benzyl alcohol are extracted from the organic phase droplets, resulting in formation of a hardened nanoparticle suspension. For example, docetaxel or other active agent may be encapsulated at e.g. a loading level of 10% w/w; corresponding to more than 10,000 drug molecules per nanoparticle.
[0071] The nanoparticle suspension is processed using tangential flow ultrafiltration/diafiltration (UF/DF) with cold water to remove processing aids and to concentrate the nanoparticles to a desired value. Residual precursor materials and excess organics present in unwashed nanoparticle suspensions may have a detrimental impact on biomedical applications as well as undesired toxic effects on the physiological system. The washed nanoparticle suspension is then passed through a prefilter and at least two sterilizing- grade filters. [0072] Once the nanoparticles have been prepared, they may be combined with an acceptable carrier to produce a pharmaceutical formulation, according to another aspect of the invention. As would be appreciated by one of skill in this art, the carrier may be selected based on factors including, but not limited to, the route of administration, the location of the targeted disease tissue, the therapeutic agent being delivered, and/or the time course of delivery of the therapeutic agent. For example, as shown in FIG. 2, a concentrated sucrose solution is aseptically added to the sterile nanoparticle suspension to produce a pharmaceutical formulation. The sucrose serves as a cryoprotectant and a tonicity agent. In this embodiment, the resulting pharmaceutical formulation is a sterile, aqueous, injectable suspension of docetaxel encapsulated in nanoparticles comprised of biocompatible and biodegradable polymers. The suspension is assayed for docetaxel content, and may be aseptically diluted to the desired concentration. In some embodiments, the particle suspension is aseptically filled and sealed in glass vials. In other embodiments, the bulk drug product suspension is stored frozen at -20 0C + 5 0C prior to filling into vials. [0073] The fabrication methods for the nanoparticles of the invention may be modified in some embodiments to achieve desired drug-delivery features. For example, nanoparticle characteristics such as surface functionality, surface charge, particle size, zeta (ζ) potential, hydrophobicity, controlled release capability, and ability to control immunogenicity, and the like, may be optimized for the effective delivery of a variety of therapeutic agents. Furthermore, the long-circulating nanoparticles produced according to the emulsion process shown in FIG. 2 are well dispersed and unagglomerated, which facilitates conjugation or functionalization of the nanoparticle surfaces with targeting moieties.
[0074] Disclosed nanoparticles may include optional targeting moieties, which may be selected to ensure that the nanoparticles selectively attach to, or otherwise associate with, a selected marker or target. For example, in some embodiments, disclosed nanoparticles may be functionalized with an amount of targeting moiety effective for the treatment of prostate cancer in a subject (e.g., a low-molecular weight PSMA ligand). Through functionalization of nanoparticle surfaces with such targeting moieties, the nanoparticles are effective only at targeted sites, which minimizes adverse side effects and improves efficacy. Targeted delivery also allows for the administration of a lower dose of therapeutic agent, which may reduce undesirable side effects commonly associated with traditional treatments of disease. [0075] In certain aspects, disclosed nanoparticles may be optimized with a specific density of targeting moities on the nanoparticle surface, such that e.g., an effective amount of targeting moiety is associated with the nanoparticle for delivery of a therapeutic agent. For example, the fraction of the the biodegradable and/or biocompatible polymer matrix functionalized with a targeting moiety may be less than 80% of the total. According to another embodiment, the fraction of the biodegradable and/or biocompatible polymer matrix functionalized with a targeting moiety is less than about 50% of the total. Increased density of the targeting moiety may, in some embodiments, increase target binding (cell binding/target uptake).
[0076] Exemplary targeting moieties include, for example, proteins, peptides, antibodies, antibody fragments, saccharides, carbohydrates, glycans, cytokines, chemokines, nucleotides, lectins, lipids, receptors, steroids, neurotransmitters and combinations thereof. The choice of a marker may vary depending on the selected target, but in general, markers that may be useful in embodiments of the invention include, but are not limited to, cell surface markers, a cancer antigen (CA), a glycoprotein antigen, a melanoma associated antigen (MAA), a proteolytic enzyme, an angiogenesis marker, a prostate membrane specific antigen (PMSA), a small cell lung carcinoma antigen (SCLCA), a hormone receptor, a tumor suppressor gene antigen, a cell cycle regulator antigen, a proliferation marker, and a human carcinoma antigen. Exemplary targeting moieties include:
Figure imgf000023_0001
-lys-(urea)glu, which may be conjugated to PEG, e.g. a disclosed nanoparticle may include PLA-PEG-targeting moiety, e.g. S,S-2-{3-[l-carboxy-5-amino-pentyl]-ureido}-pentanedioic acid. For example, disclosed nanoparticles may include about 10 to 15 weight percent active agent (e.g. docetaxel), and about 86 to about 90 weight percent PLA-PEG (with e.g. PLA about 16 kDa and PEG about 5kDa), and about 2 to about 3 weight percent PLA-PEG-lys(urea)glu (16kDa/5kDa PLA-PEG). Alternatively, a disclosed nanoparticle may include about 42 to about 45 weight percent PLA-PEG (with e.g. PLA about 16 kDa and PEG about 5kDa) about 42 to 45 weight percent PLA (e.g. about 75 kDa), about 10 to 15 weight percent active agent (e.g. docetaxel), and about about 2 to about 3 weight percent PLA-PEG-lys(urea)glu (16/5 PLA-PEG).
[0077] In other aspects of the invention, targeting moieties are targeted to an antigen associated with a disease of a patient's immune system or a pathogen-borne condition. In yet another aspect, targeting moieties are targeted to cells present in normal healthy conditions. Such targeting moieties may be directly targeted to a molecule or other target or indirectly targeted to a molecule or other target associated with a biological molecular pathway related to a condition.
[0078] The amount of nanoparticles administered to a patient may vary and may depend on the size, age, and health of the patient, the therapeutic agent to be delivered, the disease being treated, and the location of diseased tissue. Moreover, the dosage may vary depending on the mode of administration.
[0079] Various routes of administration are contemplated herein. In a particular aspect, the nanoparticles are administered to a subject systemically. Further, in some aspects, methods of administration may include, but are not limited to, intravascular injection, intravenous injection, intraperitoneal injection, subcutaneous injection, and intramuscular injection.
According to aspects of the present invention, the nanoparticles necessitate only a single or very few treatment sessions to provide effective treatment of disease, which ultimately may facilitate patient compliance. For example, in some aspects, administration of the nanoparticles can occur via intravenous infusion once every three weeks. [0080] Also contemplated herein are methods of treating solid tumors, e.g. prostate, lung, breast or other cancers, comprising administering a disclosed nanoparticle composition to a patient, e.g. a mammal in need thereof. For example, after such administration, e.g. at least after 12 hours, 24 hours, 36 hours, or 48 hours, or more after administration, the solid tumor may have significant concentration of therapeutic agent, e.g. may have an increase in tumor drug concentration of at least about 20%, or at least about 30% or more active agent (e.g. docetaxel) as compared to the amount present in a tumor after administration of (e.g. the same dosage) of therapeutic agent alone (e.g. not in a disclosed nanoparticle composition). [0081] Disclosed herein is a method of treating a solid tumor in a mammal comprising administering a nanoparticle composition comprising a plurality of nanoparticles each comprising a α-hydroxy polyester-co-polyether and a therapeutic agent, wherein the composition has an amount of therapeutic agent effective to inhibit the growth of said tumor, for example, wherein single dose of said composition provides extended release of said therapeutic agent for a least one day. Such methods may provide an actual peak plasma concentration (Cmax) of the therapeutic agent after administration of the composition to the mammal that is at least 10% higher, or at least 20% higher or 100% higher or more than the Cmax of said therapeutic agent if administered in a non-nanoparticle formulation. Disclosed methods may provide, upon administration of nanoparticles, an area under the plasma concentration time curve (AUC) in a patient that is increased by at least 100% over the AUC provided when the therapeutic agent is administered alone to a patient. In some embodiments, disclosed methods may also, alone or in addition to the above plasma parameters, decrease the volume of distribution (V2) of the therapeutic agent upon administration by at least 50% as compared to the Vz of the patient when the therapeutic agent is administered alone. [0082] A method of minimizing unwanted side effects or toxicity of an active or therapeutic agent in a patient is also provided herein. For example, disclosed nanoparticles, may, upon administration, provide a higher plasma concentration of therapeutic agent as compared to administering an equivalent dosage of therapeutic agent alone. However, upon administration, in some embodiments, disclosed nanoparticles circulate substantially in the vascular compartment, and therefore may not contribute significantly to other areas that may cause toxicity or unwanted side effects.
[0083] In order that the invention disclosed herein may be more efficiently understood, examples are provided below. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting the invention in any manner.
EXAMPLES
Example 1 In Vitro Release of Docetaxel from Nanoparticles [0084] A suspension of docetaxel encapsulated in nanoparticles fabricated according to the emulsion process depicted in FIG. 2 and Example 12 using 87.5 weight percent PLA-PEG, 10 wt.% docetaxel, and 2.5 wt. percent docetaxel (Formulation A) (all docetaxel nanoparticle formulations used in these Examples were in a composition of 5% nanoparticles, 65% water, and 30% sucrose), was placed in a dialysis cassette and incubated in a reservoir of phosphate buffered saline (PBS) at 37 0C with stirring. Samples of the dialysate were collected and analyzed for docetaxel using reversed phase high performance liquid chromatography (HPLC). For comparison, conventional docetaxel was analyzed under the same procedure. [0085] Figure 3 presents the in vitro release profile of docetaxel encapsulated in nanoparticles compared to conventional docetaxel. Release of the encapsulated docetaxel from the polymer matrix was essentially linear over the first 24 hours with the remainder gradually released over a period of about 96 hours. Example 2 Single Dose Pharmacokinetic Study of Docetaxel Encapsulated in Nanoparticles and Conventional Docetaxel in Sprague-Dawley Rats
[0086] Six- to eight-week old male Sprague-Dawley rats were administered a single bolus dose (5 mg/kg of docetaxel) of docetaxel encapsulated in nanoparticles or conventional docetaxel via a tail vein. The dose groups consisted of six rats each. Blood was drawn at
0.083, 0.5, 1, 2, 3, 4, 6, and 24 hours post-dosing and processed to plasma. The concentration of total docetaxel in plasma was measured by a liquid chromatography-mass spectrometry (LC- MS) method following extraction with methyl tert-butyl ether (MTBE). The MTBE extraction does not differentiate nanoparticle-encapsulated docetaxel from docetaxel that was released from the nanoparticles into the plasma, and as such, the LC-MS data does not distinguish between the two.
[0087] Figure 4 and Example Table 2.1 present the observed pharmacokinetic profiles and pharmacokinetic parameters, respectively, of docetaxel encapsulated in nanoparticles and conventional docetaxel. Example Table 2.1 further includes data from the preclinical development of TAXOTERE® for comparative reference (Bissery et al. 1995). The results for conventional docetaxel were consistent with those reported in literature (Bissery et al. 1995), indicating docetaxel was rapidly cleared from the blood and distributed to tissues. The peak plasma concentration (Cmax) was observed at the first sampling time point for all treatments. [0088] The Cmax and AUC of the docetaxel encapsulated in nanoparticles were approximately 100 times higher than that for conventional docetaxel. The difference in the Cmax may be attributable to having missed the rapid initial tissue distribution phase for conventional docetaxel. The data indicate that the docetaxel encapsulated in nanoparticles largely remains in circulation upon injection and is slowly cleared over a 24 hour period. The data further shows that docetaxel is released from the nanoparticles in a controlled manner during the 24 hour period (e.g., rapid burst release is not observed). If the nanoparticles were very quickly cleared from circulation, the large increase in AUC would not be observed. Similarly, if there was rapid burst release of docetaxel from the nanoparticles, the pharmacokinetic profile would be expected to more closely resemble that of conventional docetaxel.
Example Table 2.1. Summary of Docetaxel Encapsulated in Nanoparticles and Conventional Docetaxel Pharmacokinetic Parameters
Dose
Species Mnax ^ max AUC o- CL
(mg/kg) (min) (ng/niL) (h) (ng /mL'h) (L/h/kg)
Conventional
Sprague- Docetaxel 5 2 4,100 0.8b 910 5.5 Dawley Rats
(Bissery et al. 1995)
Conventional Sprague-
5 5 600 4.4C 623 2.33 Docetaxel Dawley Rats
Docetaxel
Sprague- Encapsulated in 5 5 54,800 2.6C 57,300 0.01 Dawley Rats Nanoparticles
For each treatment, tmax equals the first sampling time. bThe study duration was 6 hours. cThe half life was determined from 2-12 hours. Example 3. Tissue Distribution Study of Docetaxel Encapsulated in Nanoparticles and Conventional Docetaxel in Sprague-Dawley Rats
[0089] Six- to eight-week old male Sprague-Dawley rats were administered a single bolus intravenous dose of one of the following: (1) docetaxel encapsulated in nanoparticles in which the ligand of the PLA-PEG-lys(urea)glu targeting polymer was 14C-labeled, (2) docetaxel encapsulated in nanoparticles in which the encapsulated docetaxel was 14C-labeled, (3) 14C-labeled conventional docetaxel.
[0090] Blood was drawn at 1, 3, 6, 12, and 24 hours post-dosing and processed to plasma. Immediately following blood collection, the rats were euthanized by CO2 asphyxiation and tissues were immediately collected, blotted, weighed, and frozen on dry ice. Tissue samples were stored frozen (approximately -70 0C) until analysis for radioactivity by liquid scintillation (LS) counting.
[0091] As shown in Figure 5, the docetaxel encapsulated in nanoparticles was gradually cleared from the plasma, exhibiting an approximate 2-fold decrease in plasma concentration over the 24 hour period studied. These results are indicative of limited or delayed nanoparticle clearance via the mononuclear phagocyte system (MPS) relative to that often observed for particulate formulations. Without wishing to be bound by theory, this difference in plasma clearance times may be attributed to certain nanoparticle characteristics, including particle size and surface properties (e.g., surface charge and porosity). [0092] The distinctions in plasma profiles of docetaxel encapsulated in nanoparticles and conventional docetaxel indicate that encapsulation of docetaxel in the nanoparticles prevents it from being rapidly distributed from the plasma compartment, resulting in significantly higher Cmax and AUC values relative to conventional docetaxel. [0093] The differences in the profiles of docetaxel encapsulated in nanoparticles wherein the ligand of the PLA-PEG-lys(urea)glu targeting polymer was 14C-labeled and the docetaxel encapsulated in nanoparticles wherein the encapsulated docetaxel was 14C-labeled are reflective of the controlled release of docetaxel from the polymeric matrix of the nanoparticles. If docetaxel was released very quickly from the nanoparticles, it would be expected to be rapidly distributed from the plasma, yielding a profile similar to that of conventional docetaxel. Conversely, if docetaxel was retained in the nanoparticles over this timeframe, the profiles of the docetaxel encapsulated in nanoparticles wherein the ligand of the PLA-PEG-lys(urea)glu targeting polymer was 14C-labeled and the docetaxel encapsulated in nanoparticles wherein the encapsulated docetaxel was 14C-labeled would be superimpo sable. [0094] Example Tables 3.1, 3.2, and 3.3 present the tissue distribution of radioactivity determined in rats after intravenous (IV) administration of (1) docetaxel encapsulated in nanoparticles in which the ligand of the PLA-PEG-lys(urea)glu targeting polymer was Relabeled, (2) docetaxel encapsulated in nanoparticles in which the encapsulated docetaxel was 14C-labeled, and (3) 14C-labeled conventional docetaxel, respectively. Example Figure 5 contains the radioactivity concentration curves of the test articles determined in plasma, liver, spleen, and bone marrow.
[0095] Lower levels of nanoparticles (i.e., radioactivity from the 14C-labeled targeting polymer) were detected in all tissues relative to plasma except in the spleen, where nanoparticle concentrations were higher than plasma at 12 and 24 hours. It cannot be determined to what extent the radioactivity in tissues reflect the content in blood contained within the tissues versus the tissues themselves, because the tissues were not exsanguinated.
[0096] At time points closely following administration, the concentration of docetaxel encapsulated in nanoparticles was higher in most tissues than conventional docetaxel. After 24 hours, the concentration of docetaxel derived from the nanoparticles was lower than or approximately the same as the concentration of conventional docetaxel in all of the tissues evaluated, except the spleen.
[0097] Although the concentration of docetaxel encapsulated in nanoparticles was higher than conventional docetaxel at early timepoints and throughout the 24 hour period in the spleen, the nanoparticles doped with docetaxel were well tolerated at approximately 10 mg/kg docetaxel dose. In addition, body weight changes and clinical observations in the Sprague- Dawley rats indicate that the docetaxel encapsulated in nanoparticles was tolerated as well as conventional docetaxel through a range of acute doses (5-30 mg/kg docetaxel). Example Table 3.1. Tissue Distribution of Radioactivity Determined in Rats after IV
Administration of Nanoparticles Containing 14 C/- -Targeting Polymer.
Bone Small Large
Time Plasma Liver Spleen Heart Lungs
Marrow Intestine Intestine (h) (nCi/mL) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g)
1 2341 + 168 337+14 829 + 26 180+ 23 294 + 78 109 + 25 56 + 2.5 36 + 3.1
3 2023 + 58 334 + 43 1141+75 190+ 62 264 + 38 191 + 122 50 + 5.4 33 + 3.6
6 2001 + 71 364 + 23 1789+173 174 + 25 263 + 40 372 + 8.7 48 + 8.0 43 + 10
12 1445 + 59 375+41 2079 + 205 151 +21 266 + 24 390 + 58 71+3.6 40 + 6.1
24 998 + 55 398 + 59 2808 + 238 119 + 11 218 + 26 594 + 248 88 + 17 38 + 5.0
Example Table 3.2. Tissue Distribution of Radioactivity Determined in Rats after IV
Administration of Nanoparticles Containing 14 C/- -Docetaxel.
Bone Small Large
Time Plasma Liver Spleen Heart Lungs
Marrow Intestine Intestine (h) (nCi/mL) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g)
1 753 + 149 267 + 45 889 + 43 156 + 15 277 + 27 142 + 20 409 + 158 71 +24
3 265 + 52 127+12 999 + 94 80 + 3.6 154 +9.0 127+17 219 + 30 151+37
6 140 + 38 88+9.7 972 + 44 69 + 9.8 118 + 23 121+5.9 119 + 20 133 + 38
12 24+1.9 47 + 6.3 854 + 56 41 +2.1 58 + 8.4 89 + 9.3 50 + 2.7 98 + 14
24 5.7+1.0 22 + 3.1 634 + 95 23 + 1.3 44 + 2.5 33 + 8.2 43 + 9.3 58+4.9
Example Table 3.3. Tissue Distribution of Radioactivity Determined in Rats after IV
Administration of Conventional 14C-Docetaxel.
Bone Small Large
Time Plasma Liver Spleen Heart Lungs
Marrow Intestine Intestine (h) (nCi/mL) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g) (nCi/g)
1 4.9 + 0.4 78 + 15 100 + 9.4 71 +2.5 82 + 9.6 97 + 4.2 517 + 99 54 + 3.6
3 1.9 + 0.2 49 + 7.3 81+7.5 39+ 1.5 66 + 0.7 83 + 1.0 122 + 43 166 + 37
993 + 62 + 5.2 185 + 82
6 1.6 + 0.5 49 + 11 82 + 4.6 33 + 1.3 78 + 1.9 1605*
1438 + 41 +4.9 83 + 18
12 0.8 + 0.2 55 + 7.4 77 + 11 28 + 1.7 62 + 9.4 1218*
24 0.6 + 0.1 43 + 4.0 85 + 8.6 24 + 2.6 962 + 99* 41 +6.8 47 + 19 48 + 34
*Samples likely contaminated during collection/analysis Example 4. Tumor Targeting of Docetaxel Encapsulated in Nanoparticles and Conventional Docetaxel after a Single Dose in a Human Tumor Xenograft Model (LNCaP)
[0098] Male severe combined immunodeficiency (SCID) mice were subcutaneously inoculated with human LNCaP prostate cancer cells. Three to four weeks after inoculation, the mice were assigned to different treatment groups such that the average tumor volume in each group was 300 mm3. At this time, a single intravenous (IV) dose of 50 mg/kg docetaxel was administered as either docetaxel encapsulated in nanoparticles or conventional docetaxel. The test subjects were sacrificed 2 hour or 12 hour post-dose. The tumors from each group were excised and assayed for docetaxel using liquid chromatography-mass spectrometry (LC-MS). [0099] The measured docetaxel concentrations in tumors excised from the test subjects dosed with docetaxel encapsulated in nanoparticles or conventional docetaxel are presented in Example Table 4.1 and Figure 6. At 12 hours post-dose, the tumor docetaxel concentration in test subjects receiving docetaxel encapsulated in nanoparticles was approximately 7 times higher than in the test subjects receiving conventional docetaxel. These results are consistent with the pharmacokinetic and tissue distribution data as well as the proposed mechanism of action wherein the nanoparticles doped with docetaxel are designed to provide extended particle circulation times and controlled release of docetaxel from the nanoparticles so that particles can be targeted to and bind with a marker or target to increase the amount of docetaxel delivered to the tumor.
Example Table 4.1. Measured Docetaxel Concentration in Tumors Treated with Docetaxel Encapsulated in Nanoparticles and Conventional Docetaxel
Docetaxel Concentration in the Tumor (ng/mg)
„. ,. λ „ .. , „ . , Docetaxel Encapsulated
Time (h) Conventional Docetaxel . . τ ' . . in Nanoparticles
2 12.9 + 7.9 14.8 + 6.5
12 3.6 + 2.1 25.4 + 15.1 Example 5 Anti-tumor Activity of Docetaxel Encapsulated in Nanoparticles and Conventional Docetaxel after Repeated Doses in a Human Tumor Xenograft Model (LNCaP)
[0100] Male severe combined immunodeficiency (SCID) mice were subcutaneously inoculated with human LNCaP prostate cancer cells. Three to four weeks after inoculation, the mice were assigned to different treatment groups such that the average tumor volume in each group was 250 mm3. Subsequently, the mice were treated every other day (Q2D) for four doses, with an eight day holiday, followed by another four doses at the Q2D schedule. [0101] Average tumor volumes for each treatment group are shown in Example Figure 7. Treatment with either conventional docetaxel or docetaxel encapsulated in nanoparticles resulted in appreciable reduction in tumor volume. Tumor volume reduction was greater in test subjects receiving docetaxel encapsulated in nanoparticles compared to conventional docetaxel. These results suggest that the increase in tumor docetaxel concentration in test subjects receiving nanoparticles doped with docetaxel, compared to conventional docetaxel, may result in a more pronounced cytotoxic effect.
Example 6 Acute Dose Range Finding Study of Docetaxel Encapsulated in Nanoparticles in Sprague-Dawley Rats
[0102] Sixty Sprague-Dawley rats (30/sex) were assigned to 10 dose groups (3 rats/sex/group) and were administered a single dose of either docetaxel encapsulated in nanoparticles (5.7, 7.5, 10, 15 or 30 mg/kg body weight) or conventional docetaxel (5.7, 7.5, 10, 15 or 30 mg/kg body weight). The therapeutic compositions were administered by intravenous (IV) infusion over a 30-minute period on Day 1, after which the test subjects were observed for 7 days prior to undergoing a gross necropsy. [0103] All test subjects survived to their scheduled necropsy. Clinical observations considered to be potentially related to administration included piloerection, which appeared near the end of the 7 day observation period, and discharges from the nose and eyes. Piloerection was observed for one male rat dosed with 15 mg/kg of docetaxel encapsulated in nanoparticles, and for 5/9 male rats and 1/9 female rats dosed with 10 mg/kg of conventional docetaxel or higher. The nature and time of appearance of this clinical sign were consistent with toxicity that would be expected from cytotoxic drugs like docetaxel. Nasal and eye discharges appeared with a pattern that was unrelated to dose level, test article, sex of the animals, or time after dosing, and this clinical sign was considered to be possibly related to docetaxel and/or to stress from the dosing procedure. As shown in Example Table 6.1, male and female rats dosed with either conventional docetaxel or docetaxel encapsulated in nanoparticles showed generally minor deficits in body weight gain or actual body weight losses that were considered to be due to docetaxel toxicity. The no-adverse effect level (NOAEL) of nanoparticles doped with docetaxel in this study was considered to be 7.5 mg/kg. Example Table 6.1. Comparison of Body Weight Changes in Males and Females
Dose Docetaxel Encapsulated in Nanoparticles Conventional Docetaxel
Sex (mg/kg) Body Weight Change (%) Body Weight Change (%)
M 5.7 460 8^60
M 7.5 1.67 1.21
M 10 -3.15 -11.55
M 15 -6.23 -9.48
M 30 -7.16 -8.66
F 5.7 3.63 0.34
F 7.5 3.25 -0.11
F 10 -2.49 -0.17
F 15 -2.50 -6.86
F 30 -5.66 -5.89 Example 7 Pharmacokinetics of vincristine passively targeted nanoparticles in rats [0104] Similar to the procedure in Example 2, rats were intravenously dosed with 0.5 mg/kg with either nanoparticles prepared as in Figure 2 and Example 14 having vincristine and PLA-PEG, and no specific targeting moiety (passively targeted nanoparticles (PTNP); or vincristine alone. The release profiles are shown in Figure 8.
[0105] Plasma samples were analyzed using LC/MS and the PK analysis was performed using WinNonlin software. A comparison of the pharmacokinetics of the nanoparticles with vincristine alone is as follows:
Figure imgf000035_0001
Example 8 Pharmacokinetics of methotrexate passively targeted nanoparticles in rats
[0106] Similar to the procedure in Example 2, , rats were intravenously dosed with 0.5 mg/kg with either nanoparticles prepared as in Figure 2 and Example 15 having methotrexate and PLA-PEG, and no specific targeting moiety (passively targeted nanoparticles (PTNP); or methotrexate alone. The release profiles are shown in Figure 9. [0107] Plasma samples were analyzed using LC/MS and the PK analysis was performed using WinNonlin software. A comparison of the pharmacokinetics of the nanoparticles with methotrexate alone is as follows:
Figure imgf000036_0001
Example 9 Pharmacokinetics of paclitaxel passively targeted nanoparticles in Rats [0108] Similar to the procedure in Example 2, rats were intravenously dosed with 1.0 mg/kg with either nanoparticles prepared as in Figure 2 having paclitaxel and PLA-PEG (formulation C) and no specific targeting moiety (passively targeted nanoparticles (PTNP); or paclitaxel alone. The release profiles are shown in Figure 10.
[0109] Plasma samples were analyzed using LC/MS and the PK analysis was performed using WinNonlin software. A comparison of the pharmacokinetics of the nanoparticles with paclitaxel alone is as follows:
Figure imgf000036_0002
Example 10 Pharmacokinetics of rapamycin (sirolimus) passively targeted nanoparticles in rats [0110] Similar to the procedure in Example 2, rats were intravenously dosed with 2.0 mg/kg with either nanoparticles prepared as in Figure 2 and Example 16, having rapamycin and PLA-PEG and no specific targeting moiety (passively targeted nanoparticles (PTNP); or rapamycin alone. The release profiles are shown in Figure 11.
[0111] Plasma samples were analyzed using LC/MS and the PK analysis was performed using WinNonlin software. A comparison of the pharmacokinetics of the nanoparticles with rapamcyin alone is as follows:
Figure imgf000037_0001
Example 11 Tumor Accumulation of docetaxel nanoparticles in MX-I breast tumors in mice. [0112] Mice with MX -1 breast tumors were randomized into three groups, receiving docetaxel (3 mice), passively targeted nanoparticles (Formulation A, without a targeting moiety, PTNP), or Formulation A. The average tumor mass was 1.7g (RSD 34%). Mice were then injected with 10 mg/kg of the test article, then euthanized 24 hours later and the tumors were removed and analyzed for docetaxel content using LC/MS/MS. Results are depicted in Figure 12. The percent of injected dose in the tumor was 3% (for docetaxel alone), 30% for PTNP, and 30% Formulation A. Example 12 Pharmacokinetics of docetaxel nanoparticles in primates [0113] Naϊve non human primates (3 male and 3 female) were administered docetaxel, docetaxel nanoparticles (Formulation A) or docetaxel nanoparticles (Formulation B: 43.25% PLA-PEG(16/5), 43.25% PLA (75kDa), 10% docetaxel, 2.5% PLA-PEG-lys(urea)glu, prepared as in Example 14), using and following appropriate ethical guidelines at all times. 1 male and 1 female were used per dose group. The dosing day was 1 day and the formulations were administered by 30 minute IV infusion at 25 mg/m2 docetaxel or 50 mg/m2 docetaxel (animals were randomized and then dosed with 50 mg/m2 on day 29 and PK, hematology and clinical chemistry were measured for 21 days). At the end of the study, PK, hematology and clinical chemistry collected over a 21 day period were assessed. Figure 12 depicts the results of male (M) and female (F) PNP. A comparison of the pharmacokinetics of the nanoparticles of Formulation A (25 mg/m2 dose) with docetaxel alone is as follows:
Figure imgf000038_0001
[0114] The pharmokinetics were as follows for each NHP group:
A. Docetaxel alone
Figure imgf000039_0001
B. Formulation A
Figure imgf000039_0002
C. Formulation B
Figure imgf000040_0001
Example 13 Preparation of Docetaxel Nanoparticles
[0115] An organic phase is formed composed of a mixture of docetaxel (DTXL) and polymer (homopolymer, co-polymer, and co-polymer with ligand). The organic phase is mixed with an aqueous phase at approximately a 1:5 ratio (oil phase:aqueous phase) where the aqueous phase is composed of a surfactant and some dissolved solvent. In order to achieve high drug loading, about 30% solids in the organic phase is used.
[0116] The primary, coarse emulsion is formed by the combination of the two phases under simple mixing or through the use of a rotor stator homogenizer. The rotor/stator yielded a homogeneous milky solution, while the stir bar produced a visibly larger coarse emulsion. It was observed that the stir bar method resulted in significant oil phase droplets adhering to the side of the feed vessel, suggesting that while the coarse emulsion size is not a process parameter critical to quality, it should be made suitably fine in order to prevent yield loss or phase separation. Therefore the rotor stator is used as the standard method of coarse emulsion formation, although a high speed mixer may be suitable at a larger scale. [0117] The primary emulsion is then formed into a fine emulsion through the use of a high pressure homogenizer.
[0118] After 2-3 passes the particle size was not significantly reduced, and successive passes can even cause a particle size increase. Organic phase was emulsified 5:1 O:W with standard aqueous phase, and multiple discreet passes were performed, quenching a small portion of emulsion after each pass. The indicated scale represents the total solids of the formulation.
[0119] The effect of scale on particle size showed surprising scale dependence. The trend shows that in the 2-1Og batch size range, larger batches produce smaller particles. It has been demonstrated that this scale dependence is eliminated when considering greater than 1Og scale batches. The amount of solids used in the oil phase was about 30%. Figures 8and 9 depicts the effect of solids concentration on particle size and drug loading; with the exception of the 15- 175 series, all batches are placebo. For placebo batches the value for % solids represents the % solids were drug present at the standard 20% w/w. [0120] Table A summarizes the emulsification process parameters.
TABLE A
Figure imgf000042_0001
[0121] The fine emulsion is then quenched by addition to deionized water at a given temperature under mixing. In the quench unit operation, the emulsion is added to a cold aqueous quench under agitation. This serves to extract a significant portion of the oil phase solvents, effectively hardening the nanoparticles for downstream filtration. Chilling the quench significantly improved drug encapsulation. The quench:emulsion ratio is approximately 5:1. [0122] A solution of 35% (wt%) of Tween 80 is added to the quench to achieve approximately 2% Tween 80 overall After the emulsion is quenched a solution of Tween-80 is added which acts as a drug solubilizer, allowing for effective removal of unencapsulated drug during filtration. Table B indicates each of the quench process parameters.
Table B: Summary quench process parameters.
Figure imgf000043_0001
[0123] The temperature must remain cold enough with a dilute enough suspension (low enough concentration of solvents) to remain below the Tg of the particles. If the Q:E ratio is not high enough, then the higher concentration of solvent plasticizes the particles and allows for drug leakage. Conversely, colder temperatures allow for high drug encapsulation at low Q:E ratios (to -3:1), making it possible to run the process more efficiently. [0124] The nanoparticles are then isolated through a tangential flow filtration process to concentrate the nanoparticle suspension and buffer exchange the solvents, free drug, and drug solubilizer from the quench solution into water. A regenerated cellulose membrane is used with with a molecular weight cutoffs (MWCO) of 300.
[0125] A constant volume diafiltration (DF) is performed to remove the quench solvents, free drug and Tween-80. To perform a constant-volume DF, buffer is added to the retentate vessel at the same rate the filtrate is removed. The process parameters for the TFF operations are summarized in Table C. Crossflow rate refers to the rate of the solution flow through the feed channels and across the membrane. This flow provides the force to sweep away molecules that can foul the membrane and restrict filtrate flow. The transmembrane pressure is the force that drives the permeable molecules through the membrane.
Table C: TFF Parameters
Figure imgf000044_0001
[0126] The filtered nanoparticle slurry is then thermal cycled to an elevated temperature during workup. A small portion (typically 5-10%) of the encapsulated drug is released from the nanoparticles very quickly after its first exposure to 250C. By exposing the nanoparticle slurry to elevated temperature during workup, 'loosely encapsulated' drug can be removed and improve the product stability at the expense of a small drop in drug loading.
[0127] After the filtration process the nanoparticle suspension (concentration 50 mg/ml),is passed through a sterilizing grade filter (0.2 μm absolute). Pre-filters are used to protect the sterilizing grade filter in order to use a reasonable filtration area/ time for the process. Filtration flow rate is -1.3 L/min/m2.
[0128] The filtration train is Ertel Alsop Micromedia XL depth filter M953P membrane
(0.2 μm Nominal); Pall SUPRAcap with Seitz EKSP depth filter media (0.1 - 0.3 μm Nominal); Pall Life Sciences Supor EKV 0.65/ 0.2 micron sterilizing grade PES filter. 0.2 m of filtration surface area per kg of nanoparticles for depth filters and 1.3 m2 of filtration surface area per kg of nanoparticles for the sterilizing grade filters can be used.
Example 14 Preparation of Nanoparticles with Long Release Properties [0129] The nanoparticle preparation protocol described in Example 12 was modified to produce slow release nanoparticles.
[0130] A batch of nanoparticles was produced that incorporated a 50:50 ratio of 100 DL
7E PLA (see Table 1) with the 16/5 PLA-PEG copolymer. The addition of high molecular weight PLA is thought to decrease drug diffusion by increasing crystallinity, raising the glass transition temperature, or reducing drug solubility in the polymer. Table 1 : High Molecular Weight PLA Tested
Figure imgf000045_0001
[0131] The addition of high molecular weight PLA resulted in larger particle size when all other formulation variables were kept constant. In order to obtain slow release nanoparticles with comparable sizes as nanoparticles prepared without the high molecular weight PLA, the concentration of solids in the oil phase was reduced and the concentration of sodium cholate in the water phase was increased. Table 2 illustrates the slow release nanoparticle formulation.
Table 2: Slow Release Formulation Summary
Polymers Used % Solids in % Sodium % Drug Particle
Figure imgf000046_0001
Example 14 Nanoparticles with vincristine
[0132] Nanoparticle batches were prepared using the general procedure of Example 12, with 80% (w/w) Polymer-PEG or Polymer-PEG with homopolymer PLA at 40% (w/w) each, with a batch of % total solids of 5%, 15% and 30%. Solvents used were: 21% benzyl alcohol and 79% ethyl acetate (w/w). For each 2 gram batch size, 400 mg of drug was used and 1.6 g of 16-5 Polymer-PEG or 0.8 g of 16-5 Polymer-PEG + 0.8 g of 10 kDa PLA (homopolymer) was used. The diblock polymer 16-5 PLA-PEG or PLGA-PEG (50:50 L:G) was used, and if used, the homopolymer: PLA with a Mn = 6.5 kDa, Mw=IO kDa, and Mw/Mn=1.55. [0133] The organic phase (drug and polymer)is prepared in 2 g batches: To 20 mL scintillation vial add drug and polymer(s). The mass of solvents needed at % solids concentration is:: 5% solids: 7.98 g benzyl alcohol + 30.02 g ethyl acetate; 30% solids: 0.98 g benzyl alcohol + 3.69 g ethyl acetate. [0134] An aqueous solution is prepared with 0.5% sodium cholate, 2% benzyl alcohol, and 4% ethyl acetate in water. Add to the bottle 7.5g sodium cholate, 1402.5g of DI water, 30g of benzyl alcohol and 6Og of ethyl acetate, and mix on stir plate until dissolved. [0135] For the formation of emulsion, a ratio of aqueous phase to oil phase is 5: 1. The organic phase is poured into the aqueous solution and homogenized using IKA for 10 seconds at room temperature to form course emulsion. The solution is fed through the homogenizer (110S) at 9 Kpsi (45 psi on gauge) for 2 discreet passes to form nanoemulsion. [0136] The emulsion is poured into quench (D.I. water) at <5°C while stirring on stir plate. Ratio of quench to emulsion is 8:1. 35% (w/w) Tween 80 is added in water to quench at ratio of 25:1 Tween 80 to drug. The nanoparticles are concentrated through TFF and the quench is concentrated on TFF with 500 kDa Pall cassette (2 membrane) to -100 mL. Diafiltering is used using -20 diavolumes (2 liters) of cold DI water, and the volume is brought down to minimal volume then collect final slurry, -100 mL. The solids concentration of unfiltered final slurry is determined by the using tared 20 mL scintillation vial and adding 4 mL final slurry and dry under vacuum on lyo/oven and the weight of nanoparticles in the 4 mL of slurry dried down is determined. Concentrated sucrose (0.666 g/g) is added to final slurry sample to attain 10% sucrose.
[0137] Solids concentration of 0.45 um filtered final slurry was determined by filtering about 5mL of final slurry sample before addition of sucrose through 0.45 μm syringe filter; to tared 20 mL scintillation vial add 4 mL of filtered sample and dry under vacuum on lyo/oven. [0138] The remaining sample of unfiltered final slurry was frozen with sucrose.
Vincristine Formulations
Figure imgf000047_0001
[0139] Analytical characterization of Vincristine Formulations:
Figure imgf000047_0002
Example 15 Nanoparticles with methotrexate
[0140] Drug was dissolved in the inner aqueous phase consisting of water with 1-arginine or NaOH used for solubilizing the drug. The polymer (16-5 PLA-PEG) was dissolved in the oil phase organic solvent system, such as dichloromethane (DCM) at 20% solid concentration. The outer aqueous phase consisted mainly of water with 1% sodium cholate (SC) as surfactant, unless noted otherwise. The w/o emulsion was prepared by adding the inner aqueous phase into the oil phase under rotor stator homogenization or sonication (using Branson Digital Sonifier) at a w/o ratio of 1 : 10. The coarse w/o/w emulsion was also prepared by adding the w/o emulsion into an outer aqueous phase under either rotor stator homogenization or sonication at o/w ratio of 1 : 10. The fine w/o/w emulsion was then prepared by processing the coarse emulsion through a Microfluidics high pressure homogenizer (MHOS pneumatic) at 45000 psi with a 100 μm Z-interaction chamber. The fine emulsion was then quenched into cold DI water at 10:1 quench: emulsion ratio. These w/o, o/w and emulsion: quench ratios were maintained at 1:10 for all w/o/w experiments, unless noted otherwise. Polysorbate 80 (Tween 80) was then added as a process solubilizer to solubilize the unencapsulated drug. No drug precipitation was observed at a drug:Tween 80 ratio of 1:200. The batch was then processed with ultrafiltration followed by diafiltration to remove solvents, unencapsulated drug and solubilizer. The particle size measurements were performed by Brookhaven DLS and/or Horiba laser diffraction. To determine drug load, slurry samples were analyzed by HPLC and solid concentration analysis. The slurry retains were then diluted with sucrose to 10% before freezing. All ratios listed are on a w/w basis, unless specified otherwise. [0141] Using 16/5 PLA-PEG dissolved in ethyl acetate afforded particles between 77-85 nm in size at < 6% solid concentration in an outer aqueous phase consisting of 1% SC in DI water. Emulsions were formed under sonication at 30% amplitude. Gel formation occurred in the initial w/o emulsion with > 6% solid concentration. The inner aqueous phase MTX concentration was increased to 225 mg/ml using 1-arginine. The batch was made with 20% solids in the oil phase, consisting of 28/5 PLGA-PEG dissolved in DCM. Here, both the inner w/o and outer w/o/w emulsions were formed by rotor stator homogenization followed by 2 passes at 45k psi using a high pressure homogenizer. The nanoparticle suspension was quenched in cold DI water followed by ultrafiltration/diafiltration work-up. HPLC and PSD analysis was used to determine that the drug load stayed at 0.38% for 131 nm particles. [0142] Three different batches can be prepared according to the general procedure with the following modifications; Inner aqueous phase MTX concentration was 225 mg/ml in 0.66N NaOH solution, i.e., a 1-arginine: MTX molar ratio of 1.45:1; Span 80/Tween 80 surfactant mix (HLB = 6.2) was used as the oil phase surfactant; Batch 55-lOlC: 16/5 PLA-PEG was used instead of 28/5 PLGA-PEG. The emulsion process for all three batches remained similar. The highest drug load was obtained for the 16/5 PLA-PEG batch at 2.23% while the drug load was 0.2% and 0.04% for other batches. Example 16 Preparation of Sirolimus nanoparticles
[0143] An organic phase is formed composed of a mixture of sirolimus and polymer
(homopolymer, co-polymer, and co-polymer with ligand). The organic phase is mixed with an aqueous phase at approximately a 1:5 ratio (oil phase:aqueous phase) where the aqueous phase is composed of a surfactant and some dissolved solvent. In order to achieve high drug loading, about 30% solids in the organic phase is used. The primary, coarse emulsion is formed by the combination of the two phases under simple mixing or through the use of a rotor stator homogenizer.
[0144] The primary emulsion is then formed into a fine emulsion through the use of a high pressure homogenizer. The process is continued as in Example 12. Representative Rapamycin (sirolimus) formulations:
Figure imgf000050_0001
[0145] Although the invention has been described in considerable detail with reference to certain preferred aspects thereof, other versions are possible. Therefore the spirit and scope of the appended claims should not be limited to the description and the preferred versions contained within this specification.
Incorporation by Reference
[0146] References and citations to other documents, such as patents, patent applications, patent publications, journals, books, papers, web contents, have been made throughout this disclosure. All such documents are hereby incorporated herein by reference in their entirety for all purposes.
Equivalents [0147] Various modifications of the invention and many further embodiments thereof, in addition to those shown and described herein, will become apparent to those skilled in the art from the full contents of this document, including references to the scientific and patent literature cited herein. The subject matter herein contains important information, exemplification and guidance that can be adapted to the practice of this invention in its various embodiments and equivalents thereof. [0148] What is claimed is :

Claims

CLAIMS 1. A biocompatible nanoparticle composition comprising a plurality of long circulating nanoparticles, each comprising a α-hydroxy polyester-co-polyether and a therapeutic agent, said composition providing an elevated plasma concentration of the therapeutic agent for at least 12 hours when the composition is administered to a patient, to provide an area under the plasma concentration time curve (AUC) that is increased by at least 100% over the AUC provided when the therapeutic agent is administered alone to a patient.
2. The biocompatible nanoparticle composition of claim 1, that provides an actual peak plasma concentration (Cmax) that is at least 10% higher as compared to a Cmax of said therapeutic agent when administered alone.
3. The biocompatible nanoparticle composition of claim 1 or 2, wherein the volume of distribution when administered to the patient is less than or equal to about 5 plasma volumes.
4. The biocompatible nanoparticle composition of any one of claims 1-3, wherein the composition decreases the volume of distribution (V2) by at least 50% as compared to the Vz of the patient when the therapeutic agent is administered alone.
5. The biocompatible nanoparticle composition of any one of claims 1-4, wherein the composition provides an elevated plasma concentration of the therapeutic agent for at least 24 hours
6. The biocompatible nanoparticle composition of any one of claims 1-5, wherein the long circulating nanoparticles each further comprise a biocompatible polymer coupled to a targeting moiety.
7. The biocompatible nanoparticle composition of claim 6, wherein the targeting moiety is selected from the group consisting of a protein, peptide, antibody, antibody fragment, saccharide, carbohydrate, small molecule, glycan, cytokine, chemokine, nucleotide, lectin, lipid, receptor, steroid, neurotransmitter, cell surface marker, cancer antigen, or glycoprotein antigen.
8. The biocompatible nanoparticle composition of claim 7, wherein the targeting moiety binds to prostate membrane specific antigen (PMSA).
9. The biocompatible nanoparticle of any one of claims 6-8, wherein the biocompatible polymer is PLA-PEG.
10. The biocompatible nanoparticle of any one of claims 6-9, wherein the biocompatible polymer coupled to the targeting moiety is PLA-PEG-((5,5-2-{3-[l-carboxy-5-amino-pentyl]- ureido } -pentanedioic acid.
11. The biocompatible nanoparticle composition of any one of claims 6-10, wherein the long circulating nanoparticles comprise about 1 to about 4% by weight of the biocompatible polymer coupled to the targeting moiety.
12. The biocompatible nanoparticle composition of any one of claims 1-11, wherein the OC- hydroxy polyester-co-polyether is polylactic acid -co-polyethylene glycol.
13. The biocompatible nanoparticle composition of any one of claims 1-12, wherein the OC- hydroxy polyester-co-polyether comprises about 16kDa polylactic acid and about 5 kDa polyethylene glycol.
14. The biocompatible nanoparticle composition of any one of claims 1-13, wherein the long circulating nanoparticles are about 80 to about 90 weight percent oc-hydroxy polyester-co- polyether.
15. The biocompatible nanoparticle composition of any one of claims 1-14, wherein the peak plasma concentration (Cmax) of said therapeutic agent at least 100% higher than the Cmax of said therapeutic agent when administered alone.
16. The biocompatible nanoparticle composition of any one of claims 1-15, wherein the area under the plasma concentration time curve (AUC) is increased by at least 150% over the AUC of the therapeutic agent when administered alone to the patient.
17. The biocompatible nanoparticle composition of claims 1-13, wherein the long circulating nanoparticles further comprise a biodegradable polymer.
18. The biocompatible nanoparticle composition of claim 17, wherein the biodegradable polymer is poly(lactic) acid.
19. The biocompatible nanoparticle composition of claims 17 or 18, wherein the long circulating nanoparticles have about 40 to about 50 weight percent poly(lactic)acid, and about 40 to about 50 weight percent of α-hydroxy polyester-co-polyether.
20. The biocompatible nanoparticle composition of any one of claims 17-19, wherein the peak plasma concentration (Cmax) of said therapeutic agent at least 100% higher than the Cmax of said therapeutic agent when administered alone.
21. The biocompatible nanoparticle composition of any one of claims 17-20, wherein the area under the plasma concentration time curve (AUC) is increased by at least 200% over the AUC of the therapeutic agent when administered alone to the patient.
22. The biocompatible nanoparticle composition of any one of claims 1-21, wherein the therapeutic agent is selected from the group consisting of chemotherapeutic agents, diagnostic agents, prophylactic agents, nutraceutical agents, nucleic acids, proteins, peptides, lipids, carbohydrates, hormones, small molecules, metals, ceramics, drugs, vaccines, immunological agents, and combinations thereof.
23. The biocompatible nanoparticle composition of any one of claims 1-22, wherein the therapeutic agent is an anti-neoplastic agent.
24. The biocompatible nanoparticle composition of any one of claims 1-23, wherein the therapeutic agent is chosen from docetaxel, vincristine, methotrexate, paclitaxel, or sirolimus.
25. The biocompatible nanoparticle composition of any one of claims 1-24, further comprising an aqueous solution of a saccharide.
26. A biocompatible nanoparticle composition comprising a plurality of long circulating nanoparticles, each comprising a biocompatible polymer and a therapeutic agent, said composition providing an elevated plasma concentration of the therapeutic agent for at least 12 hours when the composition is administered to a patient, and an area under the plasma concentration time curve (AUC) that is increased by at least 100% over the AUC provided when the therapeutic agent is administered alone to a patient.
27. The biocompatible nanoparticle composition of any one of claims 1-26, wherein the patient is a mammal.
28. The biocompatible nanoparticle composition of any one of claims 1-26, wherein the patient is a primate.
29. A method of treating a solid tumor cancer, comprising administering the nanoparticle composition of any one of claims 1-29 to a patient in need thereof.
30. The method of claim 29, wherein at least 24 hours after administration, the solid tumor has significant concentration of therapeutic agent.
31. A method of treating a solid tumor in a mammal in need thereof, comprising administering a nanoparticle composition comprising a plurality of nanoparticles each comprising a α-hydroxy polyester-co-polyether and a therapeutic agent, wherein the composition has an amount of therapeutic agent effective to inhibit the growth of said tumor.
32. The method of claim 31, wherein a single dose of said composition provides extended eleveated plasma concentrations of said therapeutic agent in the patient for a least one day.
33. The method of claim 32, wherein the peak plasma concentration (Cmax) of the therapeutic agent after administration of the composition to the mammal is at least 10% higher than the Cmax of said therapeutic agent if administered in a non-nanoparticle formulation.
34. A method of minimizing unwanted side effects or toxicity of an active agent in a patient, comprising: administering a nanoparticle composition comprising a plurality of nanoparticles each comprising a α-hydroxy polyester-co-polyether and a therapeutic agent, wherein said composition is capable of delivery a higher plasma concentration of therapeutic agent to the patient as compared to administering the therapeutic agent alone, and wherein upon administering the nanoparticle composition the volume distribution of the active agent in the patient is reduced, as compared to the volume distribution if the therapeutic agent was administered alone.
35. A method for modulating the plasma concentration of a therapeutic agent in a human patient, comprising: providing a polymeric nanoparticle comprising the therapeutic agent and administering the polymeric nanoparticle to the patient, thereby modulating the plasma concentration of the human patient.
PCT/US2009/068028 2008-12-15 2009-12-15 Long circulating nanoparticles for sustained release of therapeutic agents WO2010075072A2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
JP2011540968A JP2012512175A (en) 2008-12-15 2009-12-15 Long-circulating nanoparticles for sustained release of therapeutic agents
ES09835578T ES2776126T3 (en) 2008-12-15 2009-12-15 Long-circulating nanoparticles for sustained release of therapeutic agents
EA201100765A EA201100765A1 (en) 2008-12-15 2009-12-15 Long-term circulation nanoparticles
EP09835578.7A EP2379064B1 (en) 2008-12-15 2009-12-15 Long circulating nanoparticles for sustained release of therapeutic agents

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US12247908P 2008-12-15 2008-12-15
US61/122,479 2008-12-15
US24902209P 2009-10-06 2009-10-06
US61/249,022 2009-10-06
US26020009P 2009-11-11 2009-11-11
US61/260,200 2009-11-11

Publications (2)

Publication Number Publication Date
WO2010075072A2 true WO2010075072A2 (en) 2010-07-01
WO2010075072A3 WO2010075072A3 (en) 2010-10-14

Family

ID=42288375

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/068028 WO2010075072A2 (en) 2008-12-15 2009-12-15 Long circulating nanoparticles for sustained release of therapeutic agents

Country Status (6)

Country Link
US (5) US20100216804A1 (en)
EP (1) EP2379064B1 (en)
JP (4) JP2012512175A (en)
EA (1) EA201100765A1 (en)
ES (1) ES2776126T3 (en)
WO (1) WO2010075072A2 (en)

Cited By (109)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012068531A2 (en) 2010-11-18 2012-05-24 The General Hospital Corporation Novel compositions and uses of anti-hypertension agents for cancer therapy
US8211473B2 (en) 2009-12-11 2012-07-03 Bind Biosciences, Inc. Stable formulations for lyophilizing therapeutic particles
US8246968B2 (en) 2007-03-30 2012-08-21 Bind Biosciences, Inc. Cancer cell targeting using nanoparticles
US8293276B2 (en) 2008-06-16 2012-10-23 Bind Biosciences, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
EP2512459A2 (en) * 2009-12-15 2012-10-24 Bind Biosciences, Inc. Therapeutic polymeric nanoparticles comprising epothilone and methods of making and using same
EP2512487A2 (en) * 2009-12-15 2012-10-24 Bind Biosciences, Inc. Therapeutic polymeric nanoparticles comprising corticosteroids and methods of making and using same
US8318211B2 (en) 2008-06-16 2012-11-27 Bind Biosciences, Inc. Therapeutic polymeric nanoparticles comprising vinca alkaloids and methods of making and using same
US8518963B2 (en) 2009-12-15 2013-08-27 Bind Therapeutics, Inc. Therapeutic polymeric nanoparticle compositions with high glass transition temperature or high molecular weight copolymers
WO2013151736A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics In vivo production of proteins
WO2013151666A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of biologics and proteins associated with human disease
CN103446060A (en) * 2013-09-22 2013-12-18 上海市第八人民医院 Docetaxel polydroxybutyrate nanoparticle, manufacturing method and pisum sativum agglutinin modification method and application
US8623417B1 (en) 2008-06-16 2014-01-07 Bind Therapeutics, Inc. Therapeutic polymeric nanoparticles with mTOR inhibitors and methods of making and using same
US8709483B2 (en) 2006-03-31 2014-04-29 Massachusetts Institute Of Technology System for targeted delivery of therapeutic agents
WO2014124006A1 (en) * 2013-02-05 2014-08-14 The Johns Hopkins University Nanoparticles for magnetic resonance imaging tracking and methods of making and using thereof
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
WO2014152540A1 (en) 2013-03-15 2014-09-25 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US8905997B2 (en) 2008-12-12 2014-12-09 Bind Therapeutics, Inc. Therapeutic particles suitable for parenteral administration and methods of making and using same
US8906381B2 (en) 2008-10-12 2014-12-09 Massachusetts Institute Of Technology Immunonanotherapeutics that provide IGG humoral response without T-cell antigen
US8932595B2 (en) 2008-10-12 2015-01-13 Massachusetts Institute Of Technology Nicotine immunonanotherapeutics
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
WO2015075557A2 (en) 2013-11-22 2015-05-28 Mina Alpha Limited C/ebp alpha compositions and methods of use
WO2015073587A3 (en) * 2013-11-18 2015-08-06 Rubius Therapeutics, Inc. Synthetic membrane-receiver complexes
US9198874B2 (en) 2008-12-15 2015-12-01 Bind Therapeutics, Inc. Long circulating nanoparticles for sustained release of therapeutic agents
WO2016020901A1 (en) 2014-08-07 2016-02-11 Acerta Pharma B.V. Methods of treating cancers, immune and autoimmune diseases, and inflammatory diseases based on btk occupancy and btk resynthesis rate
WO2016073798A1 (en) * 2014-11-05 2016-05-12 Selecta Biosciences, Inc. Methods and compositions related to the use of low hlb surfactants in the production of synthetic nanocarriers comprising a rapalog
US9381477B2 (en) 2006-06-23 2016-07-05 Massachusetts Institute Of Technology Microfluidic synthesis of organic nanoparticles
US9492400B2 (en) 2004-11-04 2016-11-15 Massachusetts Institute Of Technology Coated controlled release polymer particles as efficient oral delivery vehicles for biopharmaceuticals
US9526702B2 (en) 2007-10-12 2016-12-27 Massachusetts Institute Of Technology Vaccine nanotechnology
WO2017053346A1 (en) 2015-09-21 2017-03-30 Teva Pharmaceuticals International Gmbh Sustained release olanzapine formulations
WO2017070626A2 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Respiratory virus vaccines
WO2017070622A1 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Respiratory syncytial virus vaccine
WO2017070620A2 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Broad spectrum influenza virus vaccine
WO2017070601A1 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Nucleic acid vaccines for varicella zoster virus (vzv)
WO2017070623A1 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Herpes simplex virus vaccine
WO2017106630A1 (en) 2015-12-18 2017-06-22 The General Hospital Corporation Polyacetal polymers, conjugates, particles and uses thereof
US9877923B2 (en) 2012-09-17 2018-01-30 Pfizer Inc. Process for preparing therapeutic nanoparticles
US9895378B2 (en) 2014-03-14 2018-02-20 Pfizer Inc. Therapeutic nanoparticles comprising a therapeutic agent and methods of making and using the same
US9987354B2 (en) 2011-04-29 2018-06-05 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for antigen-specific deletion of T effector cells
US10046064B2 (en) 2014-09-07 2018-08-14 Selecta Biosciences, Inc. Methods and compositions for attenuating exon skipping anti-viral transfer vector immune responses
US10047072B2 (en) 2013-09-16 2018-08-14 Astrazeneca Ab Therapeutic polymeric nanoparticles and methods of making and using same
WO2018172850A1 (en) 2017-03-20 2018-09-27 Teva Pharmaceuticals International Gmbh Sustained release olanzapine formulaitons
WO2018213789A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Modified messenger rna comprising functional rna elements
WO2018213731A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
WO2018232006A1 (en) 2017-06-14 2018-12-20 Modernatx, Inc. Polynucleotides encoding coagulation factor viii
WO2019048631A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Hnf4a sarna compositions and methods of use
WO2019048645A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
WO2019104152A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding ornithine transcarbamylase for the treatment of urea cycle disorders
WO2019104160A2 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding phenylalanine hydroxylase for the treatment of phenylketonuria
WO2019104195A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits for the treatment of propionic acidemia
US10335395B2 (en) 2013-05-03 2019-07-02 Selecta Biosciences, Inc. Methods of administering immunosuppressants having a specified pharmacodynamic effective life and therapeutic macromolecules for the induction of immune tolerance
WO2019136241A1 (en) 2018-01-05 2019-07-11 Modernatx, Inc. Polynucleotides encoding anti-chikungunya virus antibodies
WO2019195350A1 (en) 2018-04-03 2019-10-10 Vaxess Technologies, Inc. Microneedle comprising silk fibroin applied to a dissolvable base
WO2019200171A1 (en) 2018-04-11 2019-10-17 Modernatx, Inc. Messenger rna comprising functional rna elements
WO2019226650A1 (en) 2018-05-23 2019-11-28 Modernatx, Inc. Delivery of dna
WO2020023390A1 (en) 2018-07-25 2020-01-30 Modernatx, Inc. Mrna based enzyme replacement therapy combined with a pharmacological chaperone for the treatment of lysosomal storage disorders
WO2020047201A1 (en) 2018-09-02 2020-03-05 Modernatx, Inc. Polynucleotides encoding very long-chain acyl-coa dehydrogenase for the treatment of very long-chain acyl-coa dehydrogenase deficiency
WO2020056155A2 (en) 2018-09-13 2020-03-19 Modernatx, Inc. Polynucleotides encoding branched-chain alpha-ketoacid dehydrogenase complex e1-alpha, e1-beta, and e2 subunits for the treatment of maple syrup urine disease
WO2020056239A1 (en) 2018-09-14 2020-03-19 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
WO2020056147A2 (en) 2018-09-13 2020-03-19 Modernatx, Inc. Polynucleotides encoding glucose-6-phosphatase for the treatment of glycogen storage disease
WO2020069169A1 (en) 2018-09-27 2020-04-02 Modernatx, Inc. Polynucleotides encoding arginase 1 for the treatment of arginase deficiency
WO2020097409A2 (en) 2018-11-08 2020-05-14 Modernatx, Inc. Use of mrna encoding ox40l to treat cancer in human patients
US10729786B2 (en) 2011-02-08 2020-08-04 The Johns Hopkins University Mucus penetrating gene carriers
WO2020208361A1 (en) 2019-04-12 2020-10-15 Mina Therapeutics Limited Sirt1-sarna compositions and methods of use
WO2020227642A1 (en) 2019-05-08 2020-11-12 Modernatx, Inc. Compositions for skin and wounds and methods of use thereof
US10869898B2 (en) 2014-04-01 2020-12-22 Rubius Therapeutics, Inc. Methods and compositions for immunomodulation
WO2020263985A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Messenger rna comprising functional rna elements and uses thereof
WO2020263883A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Endonuclease-resistant messenger rna and uses thereof
EP3641766A4 (en) * 2017-06-20 2021-03-17 Tarveda Therapeutics, Inc. Hsp90 targeted conjugates and particle formulations thereof
WO2021061707A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. Compositions and methods for modulating apolipoprotein b (apob) gene expression
WO2021061815A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. COMPOSITIONS AND METHODS FOR MODULATING HEPATOCYTE NUCLEAR FACTOR 4-ALPHA (HNF4α) GENE EXPRESSION
US11007279B2 (en) 2014-05-12 2021-05-18 The Johns Hopkins University Highly stable biodegradable gene vector platforms for overcoming biological barriers
WO2021183720A1 (en) 2020-03-11 2021-09-16 Omega Therapeutics, Inc. Compositions and methods for modulating forkhead box p3 (foxp3) gene expression
WO2021247507A1 (en) 2020-06-01 2021-12-09 Modernatx, Inc. Phenylalanine hydroxylase variants and uses thereof
WO2021252354A1 (en) 2020-06-12 2021-12-16 University Of Rochester ENCODING AND EXPRESSION OF ACE-tRNAs
EP3971287A1 (en) 2013-07-11 2022-03-23 ModernaTX, Inc. Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use
WO2022104131A1 (en) 2020-11-13 2022-05-19 Modernatx, Inc. Polynucleotides encoding cystic fibrosis transmembrane conductance regulator for the treatment of cystic fibrosis
US11345932B2 (en) 2018-05-16 2022-05-31 Synthego Corporation Methods and systems for guide RNA design and use
WO2022122872A1 (en) 2020-12-09 2022-06-16 Ucl Business Ltd Therapeutics for the treatment of neurodegenerative disorders
WO2022153262A1 (en) 2021-01-18 2022-07-21 Anton Frenkel Pharmaceutical dosage form
EP4039699A1 (en) 2015-12-23 2022-08-10 ModernaTX, Inc. Methods of using ox40 ligand encoding polynucleotides
US11426451B2 (en) 2017-03-11 2022-08-30 Selecta Biosciences, Inc. Methods and compositions related to combined treatment with antiinflammatories and synthetic nanocarriers comprising an immunosuppressant
WO2022204369A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Polynucleotides encoding methylmalonyl-coa mutase for the treatment of methylmalonic acidemia
WO2022204370A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles and polynucleotides encoding ornithine transcarbamylase for the treatment of ornithine transcarbamylase deficiency
WO2022200810A1 (en) 2021-03-26 2022-09-29 Mina Therapeutics Limited Tmem173 sarna compositions and methods of use
WO2022204380A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits and uses thereof
WO2022204390A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding phenylalanine hydroxylase and uses thereof
WO2022204371A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding glucose-6-phosphatase and uses thereof
EP4074834A1 (en) 2012-11-26 2022-10-19 ModernaTX, Inc. Terminally modified rna
WO2022240806A1 (en) 2021-05-11 2022-11-17 Modernatx, Inc. Non-viral delivery of dna for prolonged polypeptide expression in vivo
WO2022266083A2 (en) 2021-06-15 2022-12-22 Modernatx, Inc. Engineered polynucleotides for cell-type or microenvironment-specific expression
WO2022271776A1 (en) 2021-06-22 2022-12-29 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
WO2023283359A2 (en) 2021-07-07 2023-01-12 Omega Therapeutics, Inc. Compositions and methods for modulating secreted frizzled receptor protein 1 (sfrp1) gene expression
WO2023281406A1 (en) 2021-07-06 2023-01-12 Mark Hasleton Treatment of serotonin reuptake inhibitor withdrawal syndrome
EP4144378A1 (en) 2011-12-16 2023-03-08 ModernaTX, Inc. Modified nucleoside, nucleotide, and nucleic acid compositions
EP4159741A1 (en) 2014-07-16 2023-04-05 ModernaTX, Inc. Method for producing a chimeric polynucleotide encoding a polypeptide having a triazole-containing internucleotide linkage
WO2023056044A1 (en) 2021-10-01 2023-04-06 Modernatx, Inc. Polynucleotides encoding relaxin for the treatment of fibrosis and/or cardiovascular disease
US11633350B2 (en) 2014-02-23 2023-04-25 The Johns Hopkins University Hypotonic microbicidal formulations and methods of use
WO2023099884A1 (en) 2021-12-01 2023-06-08 Mina Therapeutics Limited Pax6 sarna compositions and methods of use
WO2023104964A1 (en) 2021-12-09 2023-06-15 Ucl Business Ltd Therapeutics for the treatment of neurodegenerative disorders
WO2023150753A1 (en) 2022-02-07 2023-08-10 University Of Rochester Optimized sequences for enhanced trna expression or/and nonsense mutation suppression
WO2023161350A1 (en) 2022-02-24 2023-08-31 Io Biotech Aps Nucleotide delivery of cancer therapy
EP4242307A2 (en) 2018-04-12 2023-09-13 MiNA Therapeutics Limited Sirt1-sarna compositions and methods of use
WO2023170435A1 (en) 2022-03-07 2023-09-14 Mina Therapeutics Limited Il10 sarna compositions and methods of use
WO2023183909A2 (en) 2022-03-25 2023-09-28 Modernatx, Inc. Polynucleotides encoding fanconi anemia, complementation group proteins for the treatment of fanconi anemia
WO2023196399A1 (en) 2022-04-06 2023-10-12 Modernatx, Inc. Lipid nanoparticles and polynucleotides encoding argininosuccinate lyase for the treatment of argininosuccinic aciduria
WO2023215498A2 (en) 2022-05-05 2023-11-09 Modernatx, Inc. Compositions and methods for cd28 antagonism
WO2023250117A2 (en) 2022-06-24 2023-12-28 Vaxess Technologies, Inc. Applicator for medicament patch
US11884918B2 (en) 2019-01-25 2024-01-30 Synthego Corporation Systems and methods for modulating CRISPR activity
WO2024026254A1 (en) 2022-07-26 2024-02-01 Modernatx, Inc. Engineered polynucleotides for temporal control of expression

Families Citing this family (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007070682A2 (en) 2005-12-15 2007-06-21 Massachusetts Institute Of Technology System for screening particles
US9217129B2 (en) 2007-02-09 2015-12-22 Massachusetts Institute Of Technology Oscillating cell culture bioreactor
EP2144600A4 (en) 2007-04-04 2011-03-16 Massachusetts Inst Technology Poly (amino acid) targeting moieties
CA2807552A1 (en) 2010-08-06 2012-02-09 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
DK3590949T3 (en) 2010-10-01 2022-07-11 Modernatx Inc RIBONUCLEIC ACIDS CONTAINING N1-METHYL-PSEUDOURACIL AND USE THEREOF
CA2831613A1 (en) 2011-03-31 2012-10-04 Moderna Therapeutics, Inc. Delivery and formulation of engineered nucleic acids
US8987354B2 (en) * 2011-04-25 2015-03-24 Jingxu Zhu Biocompatible polymer nanoparticle coating composition and method of production thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
SG10201602654SA (en) 2011-10-03 2016-05-30 Moderna Therapeutics Inc Modified nucleosides,nucleotides,and nucleic acids,and uses thereof
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US10555911B2 (en) * 2012-05-04 2020-02-11 Yale University Highly penetrative nanocarriers for treatment of CNS disease
US9314532B2 (en) 2012-08-10 2016-04-19 University Of North Texas Health Science Center Drug delivery vehicle
US9309114B2 (en) * 2013-01-14 2016-04-12 Xerox Corporation Porous nanoparticles produced by solvent-free emulsification
ES2641209T3 (en) 2013-05-28 2017-11-08 Sintef Tto As Procedure to prepare stealth nanoparticles
US20140378083A1 (en) * 2013-06-25 2014-12-25 Plantronics, Inc. Device Sensor Mode to Identify a User State
CA2916656A1 (en) * 2013-06-28 2014-12-31 Bind Therapeutics, Inc. Docetaxel polymeric nanoparticles for cancer treatment
JP2016531112A (en) * 2013-07-25 2016-10-06 ネムコア メディカル イノベーションズ インコーポレイテッド Nanoemulsions of hydrophobic platinum derivatives
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2015048744A2 (en) 2013-09-30 2015-04-02 Moderna Therapeutics, Inc. Polynucleotides encoding immune modulating polypeptides
AU2014329452B2 (en) 2013-10-03 2019-06-20 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
JP6324405B2 (en) * 2013-11-22 2018-05-16 株式会社サンノハシ Bolt, nut and strain measurement system
WO2015089154A1 (en) * 2013-12-10 2015-06-18 The Regents Of The University Of California Regionally activated drug delivery nanoparticles
WO2016014846A1 (en) 2014-07-23 2016-01-28 Moderna Therapeutics, Inc. Modified polynucleotides for the production of intrabodies
HUE057613T2 (en) 2015-09-17 2022-05-28 Modernatx Inc Compounds and compositions for intracellular delivery of therapeutic agents
CA3002922A1 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Human cytomegalovirus vaccine
AU2016366978B2 (en) 2015-12-10 2022-07-28 Modernatx, Inc. Compositions and methods for delivery of therapeutic agents
DK3394030T3 (en) 2015-12-22 2022-03-28 Modernatx Inc COMPOUNDS AND COMPOSITIONS FOR INTRACELLULAR RELEASE OF FUNDS
EP3400023A1 (en) 2016-01-10 2018-11-14 ModernaTX, Inc. Therapeutic mrnas encoding anti ctla-4 antibodies
WO2017147240A1 (en) 2016-02-23 2017-08-31 Tarveda Therapeutics, Inc. Hsp90 targeted conjugates and particles and formulations thereof
US11583504B2 (en) 2016-11-08 2023-02-21 Modernatx, Inc. Stabilized formulations of lipid nanoparticles
PL3596041T3 (en) 2017-03-15 2023-03-06 Modernatx, Inc. Compound and compositions for intracellular delivery of therapeutic agents
EP3596042B1 (en) 2017-03-15 2022-01-12 Modernatx, Inc. Crystal forms of amino lipids
AU2018289188B2 (en) 2017-06-22 2020-12-24 SNBioScience Inc. Particle and pharmaceutical composition comprising an insoluble camptothecin compound with double core-shell structure and method for manufacturing the same
CN112384204B (en) * 2018-02-26 2023-03-14 安托瑞斯公司 Tolerogenic liposomes and methods of use thereof
RU2681933C1 (en) * 2018-11-28 2019-03-14 Федеральное государственное бюджетное учреждение "Национальный исследовательский центр "Курчатовский институт" Method for obtaining polymeric anti-tumor particles in flow microreactor and lyophilisate based on them
US11179334B1 (en) * 2019-02-04 2021-11-23 Florida A&M University Targeted carriers for tacrolimus for ocular inflammation
CA3154618A1 (en) 2019-09-19 2021-03-25 Modernatx, Inc. Branched tail lipid compounds and compositions for intracellular delivery of therapeutic agents
CN112294974A (en) * 2020-11-18 2021-02-02 西安组织工程与再生医学研究所 Drug-loaded microparticle with multistage sustained and controlled release effects and preparation method thereof
US11524023B2 (en) 2021-02-19 2022-12-13 Modernatx, Inc. Lipid nanoparticle compositions and methods of formulating the same
WO2023055998A2 (en) * 2021-09-30 2023-04-06 The Trustees Of Princeton University Dna valency sorting chromatography

Family Cites Families (157)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989000846A1 (en) 1987-07-29 1989-02-09 The Liposome Company, Inc. Method for size separation of particles
US5542935A (en) 1989-12-22 1996-08-06 Imarx Pharmaceutical Corp. Therapeutic delivery systems related applications
US5766635A (en) * 1991-06-28 1998-06-16 Rhone-Poulenc Rorer S.A. Process for preparing nanoparticles
DE69228828T2 (en) * 1991-12-09 1999-12-16 Asahi Chemical Ind STABILIZED PARATHORMONE COMPOSITION
US5302401A (en) * 1992-12-09 1994-04-12 Sterling Winthrop Inc. Method to reduce particle size growth during lyophilization
US5543158A (en) * 1993-07-23 1996-08-06 Massachusetts Institute Of Technology Biodegradable injectable nanoparticles
US5565215A (en) 1993-07-23 1996-10-15 Massachusettes Institute Of Technology Biodegradable injectable particles for imaging
GB9412273D0 (en) 1994-06-18 1994-08-10 Univ Nottingham Administration means
US6007845A (en) * 1994-07-22 1999-12-28 Massachusetts Institute Of Technology Nanoparticles and microparticles of non-linear hydrophilic-hydrophobic multiblock copolymers
CA2207961A1 (en) 1995-01-05 1996-07-11 Robert J. Levy Surface-modified nanoparticles and method of making and using same
ES2159726T3 (en) * 1995-03-10 2001-10-16 Roche Diagnostics Gmbh FORMS OF PHARMACEUTICAL ADMINISTRATION CONTAINING POLYPEPTIDES, IN THE FORM OF MICROPARTICLES, AND PROCEDURE FOR MANUFACTURING.
US6902743B1 (en) * 1995-05-22 2005-06-07 The United States Of America As Represented By The Secretary Of The Army Therapeutic treatment and prevention of infections with a bioactive material(s) encapuslated within a biodegradable-bio-compatable polymeric matrix
US7422902B1 (en) * 1995-06-07 2008-09-09 The University Of British Columbia Lipid-nucleic acid particles prepared via a hydrophobic lipid-nucleic acid complex intermediate and use for gene transfer
JPH09208494A (en) * 1995-11-30 1997-08-12 Kanebo Ltd Fine particle formulation
FR2742357B1 (en) 1995-12-19 1998-01-09 Rhone Poulenc Rorer Sa STABILIZED AND FILTRABLE NANOPARTICLES UNDER STERILE CONDITIONS
US5792477A (en) 1996-05-07 1998-08-11 Alkermes Controlled Therapeutics, Inc. Ii Preparation of extended shelf-life biodegradable, biocompatible microparticles containing a biologically active agent
US6275809B1 (en) * 1996-05-15 2001-08-14 Hitachi, Ltd. Business processing system employing a notice board business system database and method of processing the same
US8038994B2 (en) * 1996-05-15 2011-10-18 Quest Pharmatech Inc. Combination therapy for treating disease
JP2942508B2 (en) 1997-01-14 1999-08-30 順也 藤森 Temperature sensitive sustained release base and temperature sensitive sustained release system
US6201072B1 (en) * 1997-10-03 2001-03-13 Macromed, Inc. Biodegradable low molecular weight triblock poly(lactide-co- glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
US6254890B1 (en) * 1997-12-12 2001-07-03 Massachusetts Institute Of Technology Sub-100nm biodegradable polymer spheres capable of transporting and releasing nucleic acids
US6541606B2 (en) * 1997-12-31 2003-04-01 Altus Biologics Inc. Stabilized protein crystals formulations containing them and methods of making them
HUP0102803A3 (en) 1998-06-30 2004-06-28 Amgen Inc Thousand Oaks Thermosensitive biodegradable hydrogels for sustained delivery of biologically active agents
US6265609B1 (en) * 1998-07-06 2001-07-24 Guilford Pharmaceuticals Inc. Thio-substituted pentanedioic acid derivatives
US6395718B1 (en) * 1998-07-06 2002-05-28 Guilford Pharmaceuticals Inc. Pharmaceutical compositions and methods of inhibiting angiogenesis using naaladase inhibitors
KR100274842B1 (en) 1998-10-01 2001-03-02 김효근 Sustained-release Drug Release System of Retinoic Acid Using Microspheres
DE19856432A1 (en) 1998-12-08 2000-06-15 Basf Ag Nanoparticulate core-shell systems and their use in pharmaceutical and cosmetic preparations
US6194006B1 (en) 1998-12-30 2001-02-27 Alkermes Controlled Therapeutics Inc. Ii Preparation of microparticles having a selected release profile
US7311924B2 (en) * 1999-04-01 2007-12-25 Hana Biosciences, Inc. Compositions and methods for treating cancer
US6528499B1 (en) * 2000-04-27 2003-03-04 Georgetown University Ligands for metabotropic glutamate receptors and inhibitors of NAALADase
AU6132700A (en) * 1999-09-30 2001-04-05 Chienna B.V. Polymers loaded with bioactive agents
US6136846A (en) * 1999-10-25 2000-10-24 Supergen, Inc. Formulation for paclitaxel
US20050037086A1 (en) * 1999-11-19 2005-02-17 Zycos Inc., A Delaware Corporation Continuous-flow method for preparing microparticles
KR100416242B1 (en) * 1999-12-22 2004-01-31 주식회사 삼양사 Liquid composition of biodegradable block copolymer for drug delivery and process for the preparation thereof
US6890946B2 (en) 1999-12-23 2005-05-10 Indiana University Research And Technology Corporation Use of parthenolide to inhibit cancer
US7217770B2 (en) * 2000-05-17 2007-05-15 Samyang Corporation Stable polymeric micelle-type drug composition and method for the preparation thereof
US6495164B1 (en) 2000-05-25 2002-12-17 Alkermes Controlled Therapeutics, Inc. I Preparation of injectable suspensions having improved injectability
KR100418916B1 (en) * 2000-11-28 2004-02-14 한국과학기술원 Process for Preparing Sustained Release Form of Micelle Employing Conjugate of Anticancer Drug and Biodegradable Polymer
KR100446101B1 (en) 2000-12-07 2004-08-30 주식회사 삼양사 Sustained delivery composition for poorly water soluble drugs
EP1343478B1 (en) 2000-12-21 2007-10-10 Alrise Biosystems GmbH Induced phase transition method for the production of microparticles containing hydrophilic active agents
US6623761B2 (en) * 2000-12-22 2003-09-23 Hassan Emadeldin M. Method of making nanoparticles of substantially water insoluble materials
IL156531A0 (en) * 2000-12-27 2004-01-04 Ares Trading Sa Lipid nanoparticles, their preparation and pharmaceutical compositions containing them
ATE337011T1 (en) 2001-02-07 2006-09-15 Beth Israel Hospital MODIFIED PSMA LIGANDS AND USE THEREOF
WO2002080846A2 (en) 2001-04-03 2002-10-17 Kosan Biosciences, Inc. Epothilone derivatives and methods for making and using the same
JP2005500304A (en) * 2001-06-21 2005-01-06 アルタス バイオロジックス インコーポレイテッド Spherical protein particles and methods for making and using them
WO2003017987A1 (en) 2001-08-31 2003-03-06 Mcgill University Biodegradable polymeric nanocapsules and uses thereof
US6592899B2 (en) * 2001-10-03 2003-07-15 Macromed Incorporated PLA/PLGA oligomers combined with block copolymers for enhancing solubility of a drug in water
AU2002358831B2 (en) * 2001-10-10 2007-09-06 Pierre Fabre Medicament Prolonged release biodegradable microspheres and method for preparing same
WO2003032906A2 (en) 2001-10-15 2003-04-24 Crititech, Inc. Delivery of poorly soluble drugs
AU2002351626A1 (en) 2001-12-21 2003-07-15 Celator Technologies Inc. Improved polymer-lipid delivery vehicles
US20030235619A1 (en) * 2001-12-21 2003-12-25 Christine Allen Polymer-lipid delivery vehicles
WO2003060523A1 (en) * 2002-01-10 2003-07-24 Johns Hopkins University Imaging agents and methods of imaging naaladase of psma
US20060165987A1 (en) * 2002-04-05 2006-07-27 Patrice Hildgen Stealthy polymeric biodegradable nanospheres and uses thereof
US20030232887A1 (en) * 2002-04-10 2003-12-18 Johnson Douglas Giles Preparation and use of a stable formulation of allosteric effector compounds
US6890950B2 (en) * 2002-04-23 2005-05-10 Case Western Reserve University Lapachone delivery systems, compositions and uses related thereto
JP2003342168A (en) * 2002-05-24 2003-12-03 Nano Career Kk Method for producing polymer micelle preparation containing drug for injection
US7767803B2 (en) * 2002-06-18 2010-08-03 Archemix Corp. Stabilized aptamers to PSMA and their use as prostate cancer therapeutics
US7879351B2 (en) * 2002-10-29 2011-02-01 Transave, Inc. High delivery rates for lipid based drug formulations, and methods of treatment thereof
AU2003304108B2 (en) * 2002-10-30 2007-03-22 Spherics, Inc. Nanoparticulate bioactive agents
EP1578193A4 (en) 2002-12-23 2011-06-15 Vical Inc Method for freeze-drying nucleic acid/block copolymer/cationic surfactant complexes
US6841547B2 (en) * 2003-02-28 2005-01-11 Albert Einstein College Of Medicine Of Yeshevia University Method for decreasing low density lipoprotein
US20040185170A1 (en) * 2003-03-21 2004-09-23 Shubha Chungi Method for coating drug-containing particles and formulations and dosage units formed therefrom
ATE454886T1 (en) * 2003-03-26 2010-01-15 Egalet As MATRIX PREPARATIONS FOR THE CONTROLLED PRESENTATION OF MEDICINAL MEDICINAL PRODUCTS
US20060233883A1 (en) * 2003-03-26 2006-10-19 Tsutomu Ishihara Intravenous nanoparticles for targeting drug delivery and sustained drug release
JP4927535B2 (en) 2003-04-03 2012-05-09 ジェシー エル エス オウ Particles encapsulating drugs that target tumors
US20040247624A1 (en) * 2003-06-05 2004-12-09 Unger Evan Charles Methods of making pharmaceutical formulations for the delivery of drugs having low aqueous solubility
US7727969B2 (en) 2003-06-06 2010-06-01 Massachusetts Institute Of Technology Controlled release nanoparticle having bound oligonucleotide for targeted delivery
KR100534033B1 (en) 2003-07-12 2005-12-08 주식회사 피스포스 Pumping means of shoes
US20050256071A1 (en) * 2003-07-15 2005-11-17 California Institute Of Technology Inhibitor nucleic acids
US20050142205A1 (en) * 2003-07-18 2005-06-30 Julia Rashba-Step Methods for encapsulating small spherical particles prepared by controlled phase separation
CA2533592C (en) 2003-07-23 2015-11-10 Pr Pharmaceuticals, Inc. Controlled release compositions
JP2007504259A (en) 2003-09-02 2007-03-01 ノバルティス アクチエンゲゼルシャフト Cancer treatment with epothilone
US7311901B2 (en) * 2003-10-10 2007-12-25 Samyang Corporation Amphiphilic block copolymer and polymeric composition comprising the same for drug delivery
US7846412B2 (en) * 2003-12-22 2010-12-07 Emory University Bioconjugated nanostructures, methods of fabrication thereof, and methods of use thereof
JP2007526322A (en) * 2004-03-02 2007-09-13 マサチューセッツ インスティテュート オブ テクノロジー Nanocell drug delivery system
US20070053845A1 (en) * 2004-03-02 2007-03-08 Shiladitya Sengupta Nanocell drug delivery system
US8043631B2 (en) * 2004-04-02 2011-10-25 Au Jessie L S Tumor targeting drug-loaded particles
WO2006002365A2 (en) * 2004-06-24 2006-01-05 Angiotech International Ag Microparticles with high loadings of a bioactive agent
WO2006080951A2 (en) * 2004-07-01 2006-08-03 Yale University Targeted and high density drug loaded polymeric materials
US7473678B2 (en) * 2004-10-14 2009-01-06 Biomimetic Therapeutics, Inc. Platelet-derived growth factor compositions and methods of use thereof
JP2006131577A (en) 2004-11-09 2006-05-25 Ltt Bio-Pharma Co Ltd Method for preparing nanoparticle having different particle diameters and containing sealed medicine and nanoparticle obtained by the method
BRPI0519739A2 (en) 2004-12-30 2009-03-10 Cinv Ag combination comprising an agent providing a signal, an implant material and a drug
WO2006093991A1 (en) 2005-03-02 2006-09-08 The Cleveland Clinic Foundation Compounds which bind psma and uses thereof
KR20080005939A (en) 2005-04-04 2008-01-15 시네수스 인코포레이티드 Device and methods for treating paranasal sinus conditions
JP2006321763A (en) 2005-05-20 2006-11-30 Hosokawa Funtai Gijutsu Kenkyusho:Kk Biocompatibilie nanoparticle and method for production of the same
EP1904932A4 (en) 2005-06-17 2013-02-27 Univ North Carolina Nanoparticle fabrication methods, systems, and materials
PE20100809A1 (en) * 2005-07-11 2011-01-07 Wyeth Llc GLUTAMATE INHIBITORS OF MATRIX METALOPROTEINASES AND AGRECANASES
KR101566393B1 (en) * 2005-08-03 2015-11-05 이뮤노젠 아이엔씨 Immunoconjugate formulations
BRPI0615292A8 (en) 2005-08-31 2018-03-06 Abraxis Bioscience Llc compositions and methods for preparing poorly soluble water drugs with increased stability
US8765181B2 (en) * 2005-09-09 2014-07-01 Beijing Diacrid Medical Technology Co., Ltd Nano anticancer micelles of vinca alkaloids entrapped in polyethylene glycolylated phospholipids
US20090022806A1 (en) * 2006-12-22 2009-01-22 Mousa Shaker A Nanoparticle and polymer formulations for thyroid hormone analogs, antagonists and formulations and uses thereof
WO2007034479A2 (en) 2005-09-20 2007-03-29 Yissum Research Development Company Nanoparticles for targeted delivery of active agents
WO2008051245A2 (en) 2005-12-02 2008-05-02 Novartis Ag Nanoparticles for use in immunogenic compositions
WO2007074604A1 (en) * 2005-12-26 2007-07-05 Ltt Bio-Pharma Co., Ltd. Nanoparticle containing water-soluble non-peptidic low molecular weight substance
US7842312B2 (en) * 2005-12-29 2010-11-30 Cordis Corporation Polymeric compositions comprising therapeutic agents in crystalline phases, and methods of forming the same
KR20090006062A (en) * 2006-02-23 2009-01-14 에리모스 파마슈티컬스 엘엘씨 Methods of treating influenza viral infections
DE102006013531A1 (en) * 2006-03-24 2007-09-27 Lts Lohmann Therapie-Systeme Ag Drug delivery system, useful for supplying active substance to central nervous system of a mammal over the blood-brain barrier, comprises: nanoparticles of poly(DL-lactide-co-glycolide) and pharmaceutical substance e.g. cytostatic agent
KR20070096729A (en) * 2006-03-24 2007-10-02 고쿠리츠다이가쿠호진 규슈다이가쿠 Organic compounds
EP2007435B1 (en) * 2006-03-31 2019-12-18 Massachusetts Institute Of Technology System for targeted delivery of therapeutic agents
JP5630998B2 (en) 2006-05-15 2014-11-26 マサチューセッツ インスティテュート オブ テクノロジー Polymers for functional particles
KR101536781B1 (en) * 2006-07-26 2015-07-14 산도즈 아게 Caspofungin formulations
US20080145439A1 (en) 2006-07-31 2008-06-19 Neurosystec Corporation Nanoparticle drug formulations
WO2008019142A2 (en) 2006-08-04 2008-02-14 Massachusetts Institute Of Technology Oligonucleotide systems for targeted intracellular delivery
US20080057103A1 (en) * 2006-08-21 2008-03-06 Wouter Roorda Methods of using medical devices for controlled drug release
KR100946275B1 (en) * 2006-09-26 2010-03-08 주식회사 삼양사 Submicron nanoparticle of poorly water soluble camptothecin derivatives and process for preparation thereof
PL3699162T3 (en) 2006-11-08 2022-10-31 Molecular Insight Pharmaceuticals, Inc. Heterodimers of glutamic acid
US20100303723A1 (en) 2006-11-20 2010-12-02 Massachusetts Institute Of Technology Drug delivery systems using fc fragments
CN1957911A (en) 2006-12-01 2007-05-09 济南康泉医药科技有限公司 Controlled release formulation for anti entity tumour
CN101433520A (en) 2006-12-12 2009-05-20 济南帅华医药科技有限公司 Anticancer sustained-release agent containing epothilone
CN1969818A (en) 2006-12-12 2007-05-30 济南帅华医药科技有限公司 Anticancer sustained release injection containing epothilone derivatives
CN101396342A (en) 2006-12-12 2009-04-01 济南帅华医药科技有限公司 Anti-cancer sustained-released injection containing epothilone derivate
CN1961864A (en) 2006-12-12 2007-05-16 济南帅华医药科技有限公司 Anticancer composition
CN1969816A (en) 2006-12-12 2007-05-30 济南帅华医药科技有限公司 Anticancer sustained release agent containing epothilone
CN101396340A (en) 2006-12-12 2009-04-01 济南帅华医药科技有限公司 Anti-cancer sustained-released injection containing epothilone derivate
WO2008091465A2 (en) * 2006-12-21 2008-07-31 Wayne State University Peg and targeting ligands on nanoparticle surface
JP2008162904A (en) * 2006-12-27 2008-07-17 Ltt Bio-Pharma Co Ltd Therapeutic agent for bronchial asthma
SI2131821T1 (en) 2007-03-07 2018-11-30 Abraxis Bioscience, Llc Nanoparticle comprising rapamycin and albumin as anticancer agent
CN101053553B (en) 2007-03-16 2011-04-20 吉林大学 Biodegradable fluorourcacil polyester medicine-carried nanospheres and its preparation method
EP2144600A4 (en) 2007-04-04 2011-03-16 Massachusetts Inst Technology Poly (amino acid) targeting moieties
WO2008124634A1 (en) 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Polymer-encapsulated reverse micelles
JP5248487B2 (en) 2007-05-14 2013-07-31 株式会社Lttバイオファーマ Low-molecular-weight drug-containing nanoparticles with sustained-release negatively charged groups
CA3076115C (en) * 2007-08-21 2022-06-28 Board Of Regents, The University Of Texas System Thermo-kinetic mixing for pharmaceutical applications
US20090061009A1 (en) * 2007-08-29 2009-03-05 Joseph Schwarz Composition and Method of Treatment of Bacterial Infections
CA2891005A1 (en) * 2007-09-28 2008-10-09 Bind Therapeutics, Inc. Cancer cell targeting using nanoparticles
US20090306120A1 (en) * 2007-10-23 2009-12-10 Florencia Lim Terpolymers containing lactide and glycolide
US9422234B2 (en) 2007-11-30 2016-08-23 The Johns Hopkins University Prostate specific membrane antigen (PSMA) targeted nanoparticles for therapy of prostate cancer
DE102007059752A1 (en) 2007-12-10 2009-06-18 Bayer Schering Pharma Aktiengesellschaft Functionalized solid polymer nanoparticles containing epothilones
WO2009084801A1 (en) 2007-12-31 2009-07-09 Samyang Corporation Amphiphilic block copolymer micelle composition containing taxane and manufacturing process of the same
EP2106806A1 (en) 2008-03-31 2009-10-07 Fraunhofer-Gesellschaft zur Förderung der Angewandten Forschung e.V. Nanoparticles for targeted delivery of active agents to the lung
JP2012501966A (en) * 2008-06-16 2012-01-26 バインド バイオサイエンシズ インコーポレイテッド Vinca alkaloid-containing therapeutic polymer nanoparticles and methods for making and using the same
EP2285350B1 (en) 2008-06-16 2017-11-15 Pfizer Inc Methods for the preparation of targeting agent functionalized diblock copolymers for use in fabrication of therapeutic nanoparticles
WO2010005726A2 (en) * 2008-06-16 2010-01-14 Bind Biosciences Inc. Therapeutic polymeric nanoparticles with mtor inhibitors and methods of making and using same
JP2012501965A (en) 2008-06-16 2012-01-26 バインド バイオサイエンシズ インコーポレイテッド Drug-loaded polymer nanoparticles and methods for producing and using the same
EP2310049A4 (en) * 2008-07-08 2013-06-26 Abbvie Inc Prostaglandin e2 binding proteins and uses thereof
US20100087337A1 (en) * 2008-09-10 2010-04-08 Bind Biosciences, Inc. High Throughput Fabrication of Nanoparticles
WO2010068866A2 (en) * 2008-12-12 2010-06-17 Bind Biosciences Therapeutic particles suitable for parenteral administration and methods of making and using same
ES2776126T3 (en) 2008-12-15 2020-07-29 Pfizer Long-circulating nanoparticles for sustained release of therapeutic agents
WO2010114770A1 (en) 2009-03-30 2010-10-07 Cerulean Pharma Inc. Polymer-agent conjugates, particles, compositions, and related methods of use
EA201171195A8 (en) 2009-03-30 2014-08-29 Серулин Фарма Инк. CONJUGATES, PARTICLES, POLYMER-AGENT COMPOSITIONS AND METHODS OF THEIR APPLICATION
WO2010114768A1 (en) 2009-03-30 2010-10-07 Cerulean Pharma Inc. Polymer-epothilone conjugates, particles, compositions, and related methods of use
WO2010138539A2 (en) * 2009-05-27 2010-12-02 Elan Pharma International Ltd. Reduction of flake-like aggregation in nanoparticulate active agent compositions
JP5442746B2 (en) 2009-10-01 2014-03-12 三洋電機株式会社 Image display device
EP2509634B1 (en) 2009-12-11 2019-03-06 Pfizer Inc Stable formulations for lyophilizing therapeutic particles
EP2512487A4 (en) * 2009-12-15 2013-08-07 Therapeutic polymeric nanoparticles comprising corticosteroids and methods of making and using same
ES2780156T3 (en) * 2009-12-15 2020-08-24 Pfizer Therapeutic compositions of polymeric nanoparticles with high glass transition temperature or high molecular weight copolymers
EA201290498A1 (en) * 2009-12-15 2013-01-30 Байнд Байосайенсиз, Инк. THERAPEUTIC POLYMER NANOPARTICLES, INCLUDING EPOTILON, AND METHODS FOR THEIR PREPARATION AND APPLICATION
US9295651B2 (en) * 2009-12-23 2016-03-29 The Board Of Trustees Of The University Of Illinois Nanoconjugates and nanoconjugate formulations
WO2011119995A2 (en) 2010-03-26 2011-09-29 Cerulean Pharma Inc. Formulations and methods of use
EP2395259B1 (en) * 2010-06-11 2012-11-07 iwis motorsysteme GmbH & Co. KG Tensioning device with stop bracket
WO2012040513A1 (en) 2010-09-22 2012-03-29 The Board Of Regents Of The University Of Texas System Compositions and methods for the delivery of beta lapachone
WO2012054923A2 (en) 2010-10-22 2012-04-26 Bind Biosciences, Inc. Therapeutic nanoparticles with high molecular weight copolymers
US20140308363A1 (en) 2011-05-31 2014-10-16 Bind Therapeutics, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
JP2014531456A (en) 2011-09-22 2014-11-27 バインド セラピューティックス インコーポレイテッド Therapeutic nanoparticles and methods of treating cancer
CN104136012B (en) 2012-02-29 2019-07-12 默克专利股份有限公司 The method that preparation is mounted with the nano particle of active material
CA2885022A1 (en) 2012-09-17 2014-03-20 Bind Therapeutics, Inc. Therapeutic nanoparticles comprising a therapeutic agent and methods of making and using same
EA032943B1 (en) 2012-09-17 2019-08-30 Пфайзер Инк. Process for preparing therapeutic nanoparticles (embodiments) and therapeutic nanoparticle (embodiments)
CA2916656A1 (en) 2013-06-28 2014-12-31 Bind Therapeutics, Inc. Docetaxel polymeric nanoparticles for cancer treatment

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None

Cited By (193)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9492400B2 (en) 2004-11-04 2016-11-15 Massachusetts Institute Of Technology Coated controlled release polymer particles as efficient oral delivery vehicles for biopharmaceuticals
US8709483B2 (en) 2006-03-31 2014-04-29 Massachusetts Institute Of Technology System for targeted delivery of therapeutic agents
US8802153B2 (en) 2006-03-31 2014-08-12 Massachusetts Institute Of Technology System for targeted delivery of therapeutic agents
US9381477B2 (en) 2006-06-23 2016-07-05 Massachusetts Institute Of Technology Microfluidic synthesis of organic nanoparticles
US8246968B2 (en) 2007-03-30 2012-08-21 Bind Biosciences, Inc. Cancer cell targeting using nanoparticles
US10071056B2 (en) 2007-09-28 2018-09-11 Pfizer Inc. Cancer cell targeting using nanoparticles
US9295727B2 (en) 2007-09-28 2016-03-29 Bind Therapeutics, Inc. Cancer cell targeting using nanoparticles
US9539210B2 (en) 2007-10-12 2017-01-10 Massachusetts Institute Of Technology Vaccine nanotechnology
US9526702B2 (en) 2007-10-12 2016-12-27 Massachusetts Institute Of Technology Vaccine nanotechnology
US10736848B2 (en) 2007-10-12 2020-08-11 Massachusetts Institute Of Technology Vaccine nanotechnology
US11547667B2 (en) 2007-10-12 2023-01-10 Massachusetts Institute Of Technology Vaccine nanotechnology
US9393310B2 (en) 2008-06-16 2016-07-19 Bind Therapeutics, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US9351933B2 (en) 2008-06-16 2016-05-31 Bind Therapeutics, Inc. Therapeutic polymeric nanoparticles comprising vinca alkaloids and methods of making and using same
US8318211B2 (en) 2008-06-16 2012-11-27 Bind Biosciences, Inc. Therapeutic polymeric nanoparticles comprising vinca alkaloids and methods of making and using same
US8603534B2 (en) 2008-06-16 2013-12-10 Bind Therapeutics, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US8609142B2 (en) 2008-06-16 2013-12-17 Bind Therapeutics, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US9375481B2 (en) 2008-06-16 2016-06-28 Bind Therapeutics, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US8613954B2 (en) 2008-06-16 2013-12-24 Bind Therapeutics, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US8617608B2 (en) 2008-06-16 2013-12-31 Bind Therapeutics, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US8623417B1 (en) 2008-06-16 2014-01-07 Bind Therapeutics, Inc. Therapeutic polymeric nanoparticles with mTOR inhibitors and methods of making and using same
US8293276B2 (en) 2008-06-16 2012-10-23 Bind Biosciences, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US8652528B2 (en) 2008-06-16 2014-02-18 Bind Therapeutics, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US8663700B2 (en) 2008-06-16 2014-03-04 Bind Therapeutics, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US8318208B1 (en) 2008-06-16 2012-11-27 Bind Biosciences, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US9579284B2 (en) 2008-06-16 2017-02-28 Pfizer Inc. Therapeutic polymeric nanoparticles with mTOR inhibitors and methods of making and using same
US9579386B2 (en) 2008-06-16 2017-02-28 Pfizer Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US8906381B2 (en) 2008-10-12 2014-12-09 Massachusetts Institute Of Technology Immunonanotherapeutics that provide IGG humoral response without T-cell antigen
US8932595B2 (en) 2008-10-12 2015-01-13 Massachusetts Institute Of Technology Nicotine immunonanotherapeutics
US8905997B2 (en) 2008-12-12 2014-12-09 Bind Therapeutics, Inc. Therapeutic particles suitable for parenteral administration and methods of making and using same
US9198874B2 (en) 2008-12-15 2015-12-01 Bind Therapeutics, Inc. Long circulating nanoparticles for sustained release of therapeutic agents
US9308179B2 (en) 2008-12-15 2016-04-12 Bind Therapeutics, Inc. Long circulating nanoparticles for sustained release of therapeutic agents
US9498443B2 (en) 2009-12-11 2016-11-22 Pfizer Inc. Stable formulations for lyophilizing therapeutic particles
US8211473B2 (en) 2009-12-11 2012-07-03 Bind Biosciences, Inc. Stable formulations for lyophilizing therapeutic particles
US8956657B2 (en) 2009-12-11 2015-02-17 Bind Therapeutics, Inc. Stable formulations for lyophilizing therapeutic particles
US9872848B2 (en) 2009-12-11 2018-01-23 Pfizer Inc. Stable formulations for lyophilizing therapeutic particles
US8603535B2 (en) 2009-12-11 2013-12-10 Bind Therapeutics, Inc. Stable formulations for lyophilizing therapeutic particles
US8916203B2 (en) 2009-12-11 2014-12-23 Bind Therapeutics, Inc. Stable formulations for lyophilizing therapeutic particles
US8637083B2 (en) 2009-12-11 2014-01-28 Bind Therapeutics, Inc. Stable formulations for lyophilizing therapeutic particles
EP2512487A4 (en) * 2009-12-15 2013-08-07 Therapeutic polymeric nanoparticles comprising corticosteroids and methods of making and using same
EP2512487A2 (en) * 2009-12-15 2012-10-24 Bind Biosciences, Inc. Therapeutic polymeric nanoparticles comprising corticosteroids and methods of making and using same
US9295649B2 (en) 2009-12-15 2016-03-29 Bind Therapeutics, Inc. Therapeutic polymeric nanoparticle compositions with high glass transition temperature or high molecular weight copolymers
EP2512459A4 (en) * 2009-12-15 2013-08-07 Therapeutic polymeric nanoparticles comprising epothilone and methods of making and using same
US8518963B2 (en) 2009-12-15 2013-08-27 Bind Therapeutics, Inc. Therapeutic polymeric nanoparticle compositions with high glass transition temperature or high molecular weight copolymers
EP2512459A2 (en) * 2009-12-15 2012-10-24 Bind Biosciences, Inc. Therapeutic polymeric nanoparticles comprising epothilone and methods of making and using same
US9835572B2 (en) 2009-12-15 2017-12-05 Pfizer Inc. Therapeutic polymeric nanoparticle compositions with high glass transition temperature or high molecular weight copolymers
US8912212B2 (en) 2009-12-15 2014-12-16 Bind Therapeutics, Inc. Therapeutic polymeric nanoparticle compositions with high glass transition temperature or high molecular weight copolymers
WO2012068531A2 (en) 2010-11-18 2012-05-24 The General Hospital Corporation Novel compositions and uses of anti-hypertension agents for cancer therapy
US10729786B2 (en) 2011-02-08 2020-08-04 The Johns Hopkins University Mucus penetrating gene carriers
US11235057B2 (en) 2011-04-29 2022-02-01 Selecta Biosciences, Inc. Methods for providing polymeric synthetic nanocarriers for generating antigen-specific tolerance immune responses
US10004802B2 (en) 2011-04-29 2018-06-26 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for generating CD8+ regulatory T cells
US11779641B2 (en) 2011-04-29 2023-10-10 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for allergy therapy
US9987354B2 (en) 2011-04-29 2018-06-05 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for antigen-specific deletion of T effector cells
US11717569B2 (en) 2011-04-29 2023-08-08 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers
US9993548B2 (en) 2011-04-29 2018-06-12 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for inducing regulatory B cells
US10039822B2 (en) 2011-04-29 2018-08-07 Selecta Biosciences, Inc. Method for providing polymeric synthetic nanocarriers for generating antigen-specific tolerance immune responses
US10420835B2 (en) 2011-04-29 2019-09-24 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for antigen-specific deletion of T effector cells
US10441651B2 (en) 2011-04-29 2019-10-15 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for generating CD8+ regulatory T cells
EP4144378A1 (en) 2011-12-16 2023-03-08 ModernaTX, Inc. Modified nucleoside, nucleotide, and nucleic acid compositions
WO2013151736A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics In vivo production of proteins
WO2013151666A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of biologics and proteins associated with human disease
US9877923B2 (en) 2012-09-17 2018-01-30 Pfizer Inc. Process for preparing therapeutic nanoparticles
EP4074834A1 (en) 2012-11-26 2022-10-19 ModernaTX, Inc. Terminally modified rna
US10568975B2 (en) 2013-02-05 2020-02-25 The Johns Hopkins University Nanoparticles for magnetic resonance imaging tracking and methods of making and using thereof
WO2014124006A1 (en) * 2013-02-05 2014-08-14 The Johns Hopkins University Nanoparticles for magnetic resonance imaging tracking and methods of making and using thereof
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
WO2014152540A1 (en) 2013-03-15 2014-09-25 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US10357482B2 (en) 2013-05-03 2019-07-23 Selecta Biosciences, Inc. Methods providing a therapeutic macromolecule and synthetic nanocarriers comprising immunosuppressant locally and concomitantly to reduce both type I and type IV hypersensitivity
US10357483B2 (en) 2013-05-03 2019-07-23 Selecta Biosciences, Inc. Methods comprising dosing combinations for reducing undesired humoral immune responses
US10668053B2 (en) 2013-05-03 2020-06-02 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers to reduce or prevent anaphylaxis in response to a non-allergenic antigen
US10335395B2 (en) 2013-05-03 2019-07-02 Selecta Biosciences, Inc. Methods of administering immunosuppressants having a specified pharmacodynamic effective life and therapeutic macromolecules for the induction of immune tolerance
US10434088B2 (en) 2013-05-03 2019-10-08 Selecta Biosciences, Inc. Methods related to administering immunosuppressants and therapeutic macromolecules at a reduced pharmacodynamically effective dose
US11298342B2 (en) 2013-05-03 2022-04-12 Selecta Biosciences, Inc. Methods providing a therapeutic macromolecule and synthetic nanocarriers comprising immunosuppressant locally and concomitantly to reduce both type I and type IV hypersensitivity
EP3971287A1 (en) 2013-07-11 2022-03-23 ModernaTX, Inc. Compositions comprising synthetic polynucleotides encoding crispr related proteins and synthetic sgrnas and methods of use
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
US10047072B2 (en) 2013-09-16 2018-08-14 Astrazeneca Ab Therapeutic polymeric nanoparticles and methods of making and using same
US10577351B2 (en) 2013-09-16 2020-03-03 Astrazeneca Ab Therapeutic polymeric nanoparticles and methods of making and using same
CN103446060A (en) * 2013-09-22 2013-12-18 上海市第八人民医院 Docetaxel polydroxybutyrate nanoparticle, manufacturing method and pisum sativum agglutinin modification method and application
CN103446060B (en) * 2013-09-22 2015-09-02 上海市第八人民医院 Carry Docetaxel sativum agglutinin and preparation method, pisum sativum agglutinin method of modifying and application
US9644180B2 (en) 2013-11-18 2017-05-09 Rubius Therapeutics, Inc. Synthetic membrane-receiver complexes
US10344263B2 (en) 2013-11-18 2019-07-09 Rubius Therapeutics, Inc. Synthetic membrane-receiver complexes
US10557119B2 (en) 2013-11-18 2020-02-11 Rubius Therapeutics, Inc. Erythroid cells comprising phenylalanine ammonia lyase
US10253296B2 (en) 2013-11-18 2019-04-09 Rubius Therapeutics, Inc. Synthetic membrane-receiver complexes
US10329531B2 (en) 2013-11-18 2019-06-25 Rubius Therapeutics, Inc. Synthetic membrane-receiver complexes
WO2015073587A3 (en) * 2013-11-18 2015-08-06 Rubius Therapeutics, Inc. Synthetic membrane-receiver complexes
US10301593B2 (en) 2013-11-18 2019-05-28 Rubius Therapeutics, Inc. Synthetic membrane-receiver complexes
US10301594B1 (en) 2013-11-18 2019-05-28 Rubius Therapeutics, Inc Synthetic membrane-receiver complexes
EP3985118A1 (en) 2013-11-22 2022-04-20 MiNA Therapeutics Limited C/ebp alpha short activating rna compositions and methods of use
EP3594348A1 (en) 2013-11-22 2020-01-15 Mina Therapeutics Limited C/ebp alpha short activating rna compositions and methods of use
WO2015075557A2 (en) 2013-11-22 2015-05-28 Mina Alpha Limited C/ebp alpha compositions and methods of use
US11633350B2 (en) 2014-02-23 2023-04-25 The Johns Hopkins University Hypotonic microbicidal formulations and methods of use
US9895378B2 (en) 2014-03-14 2018-02-20 Pfizer Inc. Therapeutic nanoparticles comprising a therapeutic agent and methods of making and using the same
US10071100B2 (en) 2014-03-14 2018-09-11 Pfizer Inc. Therapeutic nanoparticles comprising a therapeutic agent and methods of making and using the same
US10869898B2 (en) 2014-04-01 2020-12-22 Rubius Therapeutics, Inc. Methods and compositions for immunomodulation
US11554141B2 (en) 2014-04-01 2023-01-17 Rubius Therapeutics, Inc. Methods and compositions for immunomodulation
US11576934B2 (en) 2014-04-01 2023-02-14 Rubius Therapeutics, Inc. Methods and compositions for immunomodulation
US11007279B2 (en) 2014-05-12 2021-05-18 The Johns Hopkins University Highly stable biodegradable gene vector platforms for overcoming biological barriers
EP4159741A1 (en) 2014-07-16 2023-04-05 ModernaTX, Inc. Method for producing a chimeric polynucleotide encoding a polypeptide having a triazole-containing internucleotide linkage
WO2017025814A1 (en) 2014-08-07 2017-02-16 Acerta Pharma B.V. Methods of treating cancers, immune and autoimmune diseases, and inflammatory diseases based on btk occupancy and btk resynthesis rate
WO2016020901A1 (en) 2014-08-07 2016-02-11 Acerta Pharma B.V. Methods of treating cancers, immune and autoimmune diseases, and inflammatory diseases based on btk occupancy and btk resynthesis rate
US11633422B2 (en) 2014-09-07 2023-04-25 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector immune responses
US10071114B2 (en) 2014-09-07 2018-09-11 Selecta Biosciences, Inc. Methods and compositions for attenuating gene expression modulating anti-viral transfer vector immune responses
US10046064B2 (en) 2014-09-07 2018-08-14 Selecta Biosciences, Inc. Methods and compositions for attenuating exon skipping anti-viral transfer vector immune responses
EP3834823A1 (en) * 2014-11-05 2021-06-16 Selecta Biosciences, Inc. Methods and compositions related to the use of low hlb surfactants in the production of synthetic nanocarriers comprising a rapalog
CN113244191A (en) * 2014-11-05 2021-08-13 西莱克塔生物科技公司 Methods and compositions related to synthetic nanoparticles with rapamycin in a stable supersaturated state
EP3906918A1 (en) * 2014-11-05 2021-11-10 Selecta Biosciences, Inc. Methods and compositions related to synthetic nanocarriers with rapamycin in a stable, super-saturated state
AU2015342968B2 (en) * 2014-11-05 2021-05-27 Selecta Biosciences, Inc. Methods and compositions related to the use of low HLB surfactants in the production of synthetic nanocarriers comprising a rapalog
EP3215192A4 (en) * 2014-11-05 2018-05-02 Selecta Biosciences, Inc. Methods and compositions related to synthetic nanocarriers with rapamycin in a stable, super-saturated state
KR20170081660A (en) * 2014-11-05 2017-07-12 셀렉타 바이오사이언시즈, 인크. Methods and compositions related to synthetic nanocarriers with rapamycin in a stable, super-saturated state
AU2015342969B2 (en) * 2014-11-05 2021-05-27 Selecta Biosciences, Inc. Methods and compositions related to synthetic nanocarriers with rapamycin in a stable, super-saturated state
CN107072965B (en) * 2014-11-05 2021-05-11 西莱克塔生物科技公司 Methods and compositions related to synthetic nanoparticles with rapamycin in a stable supersaturated state
CN107072965A (en) * 2014-11-05 2017-08-18 西莱克塔生物科技公司 The method and composition related to the synthesis nano particle with the rapamycin in stable over-saturation state
WO2016073798A1 (en) * 2014-11-05 2016-05-12 Selecta Biosciences, Inc. Methods and compositions related to the use of low hlb surfactants in the production of synthetic nanocarriers comprising a rapalog
KR102601922B1 (en) 2014-11-05 2023-11-15 셀렉타 바이오사이언시즈, 인크. Methods and compositions related to synthetic nanocarriers with rapamycin in a stable, super-saturated state
WO2017053346A1 (en) 2015-09-21 2017-03-30 Teva Pharmaceuticals International Gmbh Sustained release olanzapine formulations
EP4331570A1 (en) 2015-09-21 2024-03-06 Teva Pharmaceuticals International GmbH Sustained release olanzapine formulations
WO2017070620A2 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Broad spectrum influenza virus vaccine
WO2017070626A2 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Respiratory virus vaccines
WO2017070623A1 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Herpes simplex virus vaccine
EP4349405A2 (en) 2015-10-22 2024-04-10 ModernaTX, Inc. Respiratory virus vaccines
WO2017070622A1 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Respiratory syncytial virus vaccine
EP4011451A1 (en) 2015-10-22 2022-06-15 ModernaTX, Inc. Metapneumovirus mrna vaccines
EP4349404A2 (en) 2015-10-22 2024-04-10 ModernaTX, Inc. Respiratory virus vaccines
WO2017070601A1 (en) 2015-10-22 2017-04-27 Modernatx, Inc. Nucleic acid vaccines for varicella zoster virus (vzv)
WO2017106630A1 (en) 2015-12-18 2017-06-22 The General Hospital Corporation Polyacetal polymers, conjugates, particles and uses thereof
EP4039699A1 (en) 2015-12-23 2022-08-10 ModernaTX, Inc. Methods of using ox40 ligand encoding polynucleotides
US11426451B2 (en) 2017-03-11 2022-08-30 Selecta Biosciences, Inc. Methods and compositions related to combined treatment with antiinflammatories and synthetic nanocarriers comprising an immunosuppressant
WO2018172850A1 (en) 2017-03-20 2018-09-27 Teva Pharmaceuticals International Gmbh Sustained release olanzapine formulaitons
US10646443B2 (en) 2017-03-20 2020-05-12 Teva Pharmaceuticals International Gmbh Sustained release olanzapine formulations
US11813359B2 (en) 2017-03-20 2023-11-14 Teva Pharmaceuticals International Gmbh Sustained release olanzapine formulations
EP4253544A2 (en) 2017-05-18 2023-10-04 ModernaTX, Inc. Modified messenger rna comprising functional rna elements
WO2018213731A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (il12) polypeptides and uses thereof
WO2018213789A1 (en) 2017-05-18 2018-11-22 Modernatx, Inc. Modified messenger rna comprising functional rna elements
WO2018232006A1 (en) 2017-06-14 2018-12-20 Modernatx, Inc. Polynucleotides encoding coagulation factor viii
EP3641766A4 (en) * 2017-06-20 2021-03-17 Tarveda Therapeutics, Inc. Hsp90 targeted conjugates and particle formulations thereof
EP4233880A2 (en) 2017-09-08 2023-08-30 MiNA Therapeutics Limited Hnf4a sarna compositions and methods of use
EP4219715A2 (en) 2017-09-08 2023-08-02 MiNA Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
EP4183882A1 (en) 2017-09-08 2023-05-24 MiNA Therapeutics Limited Stabilized hnf4a sarna compositions and methods of use
WO2019048645A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Stabilized cebpa sarna compositions and methods of use
WO2019048632A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Stabilized hnf4a sarna compositions and methods of use
WO2019048631A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited Hnf4a sarna compositions and methods of use
WO2019104195A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits for the treatment of propionic acidemia
WO2019104152A1 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding ornithine transcarbamylase for the treatment of urea cycle disorders
WO2019104160A2 (en) 2017-11-22 2019-05-31 Modernatx, Inc. Polynucleotides encoding phenylalanine hydroxylase for the treatment of phenylketonuria
WO2019136241A1 (en) 2018-01-05 2019-07-11 Modernatx, Inc. Polynucleotides encoding anti-chikungunya virus antibodies
WO2019195350A1 (en) 2018-04-03 2019-10-10 Vaxess Technologies, Inc. Microneedle comprising silk fibroin applied to a dissolvable base
WO2019200171A1 (en) 2018-04-11 2019-10-17 Modernatx, Inc. Messenger rna comprising functional rna elements
EP4242307A2 (en) 2018-04-12 2023-09-13 MiNA Therapeutics Limited Sirt1-sarna compositions and methods of use
US11345932B2 (en) 2018-05-16 2022-05-31 Synthego Corporation Methods and systems for guide RNA design and use
US11802296B2 (en) 2018-05-16 2023-10-31 Synthego Corporation Methods and systems for guide RNA design and use
US11697827B2 (en) 2018-05-16 2023-07-11 Synthego Corporation Systems and methods for gene modification
WO2019226650A1 (en) 2018-05-23 2019-11-28 Modernatx, Inc. Delivery of dna
WO2020023390A1 (en) 2018-07-25 2020-01-30 Modernatx, Inc. Mrna based enzyme replacement therapy combined with a pharmacological chaperone for the treatment of lysosomal storage disorders
WO2020047201A1 (en) 2018-09-02 2020-03-05 Modernatx, Inc. Polynucleotides encoding very long-chain acyl-coa dehydrogenase for the treatment of very long-chain acyl-coa dehydrogenase deficiency
WO2020056155A2 (en) 2018-09-13 2020-03-19 Modernatx, Inc. Polynucleotides encoding branched-chain alpha-ketoacid dehydrogenase complex e1-alpha, e1-beta, and e2 subunits for the treatment of maple syrup urine disease
WO2020056147A2 (en) 2018-09-13 2020-03-19 Modernatx, Inc. Polynucleotides encoding glucose-6-phosphatase for the treatment of glycogen storage disease
WO2020056239A1 (en) 2018-09-14 2020-03-19 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
WO2020069169A1 (en) 2018-09-27 2020-04-02 Modernatx, Inc. Polynucleotides encoding arginase 1 for the treatment of arginase deficiency
WO2020097409A2 (en) 2018-11-08 2020-05-14 Modernatx, Inc. Use of mrna encoding ox40l to treat cancer in human patients
US11884918B2 (en) 2019-01-25 2024-01-30 Synthego Corporation Systems and methods for modulating CRISPR activity
WO2020208361A1 (en) 2019-04-12 2020-10-15 Mina Therapeutics Limited Sirt1-sarna compositions and methods of use
WO2020227642A1 (en) 2019-05-08 2020-11-12 Modernatx, Inc. Compositions for skin and wounds and methods of use thereof
WO2020263883A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Endonuclease-resistant messenger rna and uses thereof
WO2020263985A1 (en) 2019-06-24 2020-12-30 Modernatx, Inc. Messenger rna comprising functional rna elements and uses thereof
WO2021061815A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. COMPOSITIONS AND METHODS FOR MODULATING HEPATOCYTE NUCLEAR FACTOR 4-ALPHA (HNF4α) GENE EXPRESSION
WO2021061707A1 (en) 2019-09-23 2021-04-01 Omega Therapeutics, Inc. Compositions and methods for modulating apolipoprotein b (apob) gene expression
WO2021183720A1 (en) 2020-03-11 2021-09-16 Omega Therapeutics, Inc. Compositions and methods for modulating forkhead box p3 (foxp3) gene expression
WO2021247507A1 (en) 2020-06-01 2021-12-09 Modernatx, Inc. Phenylalanine hydroxylase variants and uses thereof
WO2021252354A1 (en) 2020-06-12 2021-12-16 University Of Rochester ENCODING AND EXPRESSION OF ACE-tRNAs
WO2022104131A1 (en) 2020-11-13 2022-05-19 Modernatx, Inc. Polynucleotides encoding cystic fibrosis transmembrane conductance regulator for the treatment of cystic fibrosis
WO2022122872A1 (en) 2020-12-09 2022-06-16 Ucl Business Ltd Therapeutics for the treatment of neurodegenerative disorders
WO2022153262A1 (en) 2021-01-18 2022-07-21 Anton Frenkel Pharmaceutical dosage form
WO2022204390A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding phenylalanine hydroxylase and uses thereof
WO2022204369A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Polynucleotides encoding methylmalonyl-coa mutase for the treatment of methylmalonic acidemia
WO2022204370A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles and polynucleotides encoding ornithine transcarbamylase for the treatment of ornithine transcarbamylase deficiency
WO2022204380A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding propionyl-coa carboxylase alpha and beta subunits and uses thereof
WO2022204371A1 (en) 2021-03-24 2022-09-29 Modernatx, Inc. Lipid nanoparticles containing polynucleotides encoding glucose-6-phosphatase and uses thereof
WO2022200810A1 (en) 2021-03-26 2022-09-29 Mina Therapeutics Limited Tmem173 sarna compositions and methods of use
WO2022240806A1 (en) 2021-05-11 2022-11-17 Modernatx, Inc. Non-viral delivery of dna for prolonged polypeptide expression in vivo
WO2022266083A2 (en) 2021-06-15 2022-12-22 Modernatx, Inc. Engineered polynucleotides for cell-type or microenvironment-specific expression
WO2022271776A1 (en) 2021-06-22 2022-12-29 Modernatx, Inc. Polynucleotides encoding uridine diphosphate glycosyltransferase 1 family, polypeptide a1 for the treatment of crigler-najjar syndrome
WO2023281406A1 (en) 2021-07-06 2023-01-12 Mark Hasleton Treatment of serotonin reuptake inhibitor withdrawal syndrome
WO2023283359A2 (en) 2021-07-07 2023-01-12 Omega Therapeutics, Inc. Compositions and methods for modulating secreted frizzled receptor protein 1 (sfrp1) gene expression
WO2023056044A1 (en) 2021-10-01 2023-04-06 Modernatx, Inc. Polynucleotides encoding relaxin for the treatment of fibrosis and/or cardiovascular disease
WO2023099884A1 (en) 2021-12-01 2023-06-08 Mina Therapeutics Limited Pax6 sarna compositions and methods of use
WO2023104964A1 (en) 2021-12-09 2023-06-15 Ucl Business Ltd Therapeutics for the treatment of neurodegenerative disorders
WO2023150753A1 (en) 2022-02-07 2023-08-10 University Of Rochester Optimized sequences for enhanced trna expression or/and nonsense mutation suppression
WO2023161350A1 (en) 2022-02-24 2023-08-31 Io Biotech Aps Nucleotide delivery of cancer therapy
WO2023170435A1 (en) 2022-03-07 2023-09-14 Mina Therapeutics Limited Il10 sarna compositions and methods of use
WO2023183909A2 (en) 2022-03-25 2023-09-28 Modernatx, Inc. Polynucleotides encoding fanconi anemia, complementation group proteins for the treatment of fanconi anemia
WO2023196399A1 (en) 2022-04-06 2023-10-12 Modernatx, Inc. Lipid nanoparticles and polynucleotides encoding argininosuccinate lyase for the treatment of argininosuccinic aciduria
WO2023215498A2 (en) 2022-05-05 2023-11-09 Modernatx, Inc. Compositions and methods for cd28 antagonism
WO2023250117A2 (en) 2022-06-24 2023-12-28 Vaxess Technologies, Inc. Applicator for medicament patch
WO2024026254A1 (en) 2022-07-26 2024-02-01 Modernatx, Inc. Engineered polynucleotides for temporal control of expression

Also Published As

Publication number Publication date
JP2012512175A (en) 2012-05-31
JP2015143268A (en) 2015-08-06
US20110217377A1 (en) 2011-09-08
US20100216804A1 (en) 2010-08-26
US20140093579A1 (en) 2014-04-03
EP2379064B1 (en) 2020-02-26
EP2379064A4 (en) 2012-06-20
EP2379064A2 (en) 2011-10-26
WO2010075072A3 (en) 2010-10-14
US9308179B2 (en) 2016-04-12
EA201100765A1 (en) 2012-04-30
US9198874B2 (en) 2015-12-01
ES2776126T3 (en) 2020-07-29
JP2019142924A (en) 2019-08-29
US20130034608A1 (en) 2013-02-07
US20160045608A1 (en) 2016-02-18
JP2017165780A (en) 2017-09-21

Similar Documents

Publication Publication Date Title
US9198874B2 (en) Long circulating nanoparticles for sustained release of therapeutic agents
US9872848B2 (en) Stable formulations for lyophilizing therapeutic particles
US20170266293A1 (en) Methods of treating cancers with therapeutic nanoparticles
EP2309989B1 (en) Drug loaded polymeric nanoparticles and methods of making and using same
US20100104655A1 (en) Therapeutic Polymeric Nanoparticles Comprising Vinca Alkaloids and Methods of Making and Using Same

Legal Events

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

Ref document number: 09835578

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 201100765

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 2011540968

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2009835578

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE