US20130028857A1 - Synthetic nanocarriers comprising polymers comprising multiple immunomodulatory agents - Google Patents

Synthetic nanocarriers comprising polymers comprising multiple immunomodulatory agents Download PDF

Info

Publication number
US20130028857A1
US20130028857A1 US13/560,943 US201213560943A US2013028857A1 US 20130028857 A1 US20130028857 A1 US 20130028857A1 US 201213560943 A US201213560943 A US 201213560943A US 2013028857 A1 US2013028857 A1 US 2013028857A1
Authority
US
United States
Prior art keywords
polymer
immunomodulatory agent
synthetic nanocarriers
agent moieties
moieties
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/560,943
Other languages
English (en)
Inventor
Yun Gao
David H. Altreuter
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cartesian Therapeutics Inc
Original Assignee
Selecta Biosciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Selecta Biosciences Inc filed Critical Selecta Biosciences Inc
Priority to US13/560,943 priority Critical patent/US20130028857A1/en
Assigned to SELECTA BIOSCIENCES, INC. reassignment SELECTA BIOSCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ALTREUTER, DAVID H., GAO, YUN
Publication of US20130028857A1 publication Critical patent/US20130028857A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/385Haptens or antigens, bound to carriers
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • 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
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • 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
    • 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
    • A61K47/62Medicinal 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 a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/645Polycationic or polyanionic oligopeptides, polypeptides or polyamino acids, e.g. polylysine, polyarginine, polyglutamic acid or peptide TAT
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55555Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T428/00Stock material or miscellaneous articles
    • Y10T428/29Coated or structually defined flake, particle, cell, strand, strand portion, rod, filament, macroscopic fiber or mass thereof
    • Y10T428/2982Particulate matter [e.g., sphere, flake, etc.]

Definitions

  • This invention relates to compositions, and related methods, of synthetic nanocarriers that comprise polymers that comprise at least two immunomodulatory agent moieties.
  • Immunomodulatory agents such as adjuvants
  • effective ways of delivering high concentrations of immunomodulatory agents to target cells, such as dendritic cells, are needed.
  • a composition comprising synthetic nanocarriers comprising a first type of polymer that comprises at least two immunomodulatory agent moieties.
  • the immunomodulatory agent moieties are between 2 and 100% of the polymer weight.
  • the immunomodulatory agent moieties are at least 4% of the polymer weight.
  • the immunomodulatory agent moieties are at least 8% of the polymer weight.
  • at least a portion of the immunomodulatory agent moieties are not present at the surface of the synthetic nanocarriers.
  • at least a portion of the immunomodulatory agent moieties are not at the terminus of a polymer.
  • the first type of polymer comprises at least three, four, five, six, seven, eight, nine or ten immunomodulatory agent moieties.
  • the at least two immunomodulatory agent moieties are at at least one terminus of the first type of polymer.
  • the at least two immunomodulatory agent moieties are along the backbone of the first type of polymer.
  • the at least two immunomodulatory agent moieties are themselves polymerized and form the backbone or portion of the backbone of the first type of polymer.
  • the synthetic nanocarriers further comprise another material in addition to the immunomodulatory agent moieties.
  • the other material is another type of polymer.
  • the polymer backbone comprises at least one type of monomeric residue that is not the immunomodulatory agent moiety.
  • the synthetic nanocarriers comprise at least a second type of polymer.
  • the first type of polymer and/or the second type of polymer is a linear polymer. In another embodiment, the first type of polymer and/or the second type of polymer is a branched polymer. In yet another embodiment, the first type of polymer and/or the second type of polymer is part of or forms a dendrimer. In still another embodiment, the first type of polymer and/or the second type of polymer is part of or forms a polymeric matrix.
  • the immunomodulatory agent moieties are the same type of immunomodulatory agent moiety.
  • the immunomodulatory agent moieties comprise a toll-like receptor (TLR) agonist.
  • the TLR agonist is a TLR-7 and/or TLR-8 agonist.
  • the TLR agonist comprises an aminodiazepine, adenine derivative or an imidazoquinoline.
  • the imidazoquinoline comprises resiquimod or imiquimod.
  • the adenine derivative comprises PF-4171455 or SM-276001.
  • composition further comprises an antigen.
  • the antigen comprises a B cell or T cell antigen.
  • the T cell antigen is a T helper cell antigen.
  • the first type of polymer and/or the second type of polymer comprises a polyester, polyether, polycarbonate or polyamino acid.
  • the polyester comprises a poly(lactic acid), poly(glycolic acid), poly(lactic-co-glycolic acid) or polycaprolactone.
  • the polyester is coupled to a hydrophilic polymer.
  • the hydrophilic polymer comprises a polyether.
  • the polyether comprises polyethylene glycol.
  • the polyamino acid comprises polyglutamic acid.
  • the first type of polymer and/or the second type of polymer has a weight average or number average molecular weight of at least 2000 Da, at least 2500 Da, at least 3000 Da, at least 3500 Da, at least 4000 Da, at least 4500 Da or at least 5000 Da.
  • the mean of a particle size distribution obtained using dynamic light scattering of the synthetic nanocarriers is a maximum dimension of from 20 nm to 500 nm. In another embodiment, the mean of a particle size distribution obtained using dynamic light scattering of the synthetic nanocarriers is a maximum dimension of from 20 nm to 400 nm. In another embodiment, the mean of a particle size distribution obtained using dynamic light scattering of the synthetic nanocarriers is a maximum dimension of from 20 nm to 300 nm. In another embodiment, the mean of a particle size distribution obtained using dynamic light scattering of the synthetic nanocarriers is a maximum dimension of from 20 nm to 250 nm.
  • composition further comprises a pharmaceutically acceptable excipient.
  • composition is sterile. In another embodiment, the compositions is in lyophilized form.
  • a dosage form comprising any of the compositions provided herein is provided.
  • a vaccine comprising any of the dosage forms provided herein is provided.
  • a method comprising administering any of the compositions, dosage forms or vaccines to a subject.
  • the subject is a human.
  • the method further comprises administering an antigen.
  • the antigen comprises a B cell or T cell antigen.
  • the T cell antigen is a T helper cell antigen.
  • the subject has or is at risk of having cancer.
  • the subject has or is at risk of having an infection or infectious disease.
  • the subject has or is at risk of having an autoimmune disease, an inflammatory disease, an allergy or graft versus host disease.
  • the subject has undergone or will undergo transplantation.
  • a method of producing a polymer comprising at least two immunomodulatory agent moieties at a terminus of a polymer comprising preparing a ring-opened polyester polymer with polyalcohol, contacting the ring-opened polyester polymer with succinic anhydride, and reacting the polyester polymer with immunomodulatory agent moieties in the presence of a coupling agent and a base
  • the polyester comprises a poly(lactic acid), poly(glycolic acid), poly(lactic-co-glycolic acid) or polycaprolactone.
  • the coupling agent comprises TBTU, HBTU, EDC, DCC or PyBop.
  • the base comprises DIPEA, DMAP or Et3N.
  • a method of producing a polymer comprising at least two immunomodulatory agent moieties along the polymer backbone comprising preparing a polyamino acid polymer with a free side chain acid group, and coupling the polymer with immunomodulatory agent moieties in the presence of a coupling agent and a base.
  • the polyamino acid polymer comprises polyglutamic acid.
  • the coupling agent comprises TBTU, HBTU, EDC, DCC or PyBop.
  • the base comprises DIPEA, DMAP or Et3N.
  • a method of producing a polymer comprising at least two immunomodulatory agent moieties along the polymer backbone comprising polymerizing a monomer in the presence of a polyol to provide a multi-armed polymer, functionalizing the multi-armed polymer with one or more carboxylic acid groups, and coupling the immunomodulatory agent moieties comprising an amino group with the multi-armed polymer in the presence of a coupling agent.
  • the multi-armed polymer comprises a poly(lactic acid), poly(glycolic acid), poly(lactic-co-glycolic acid) or polycaprolactone.
  • the coupling agent comprises TBTU, HBTU, EDC, DCC or PyBop.
  • the coupling is performed also in the presence of a base.
  • the base is DIPEA, DMAP or Et3N.
  • a method of producing a polymer comprising at least two immunomodulatory agent moieties comprising providing a linear polymer comprising two or more side chain groups comprising an electrophilic or nucleophilic chemical moiety attached thereto, and coupling the immunomodulatory agent moieties to the side chain group.
  • the side chain chemical moiety comprises a carboxylic acid and the immunomodulatory agent moieties, comprising an amino group, are coupled to carboxylic acid group in the presence of a coupling agent.
  • the coupling agent is TBTU, HBTU, EDC, DCC or PyBop.
  • the coupling is performed also in the presence of a base.
  • the base is DIPEA, DMAP or Et3N.
  • a method of producing a polymer comprising at least two immunomodulatory agent moieties along the polymer backbone comprising functionalizing monomers of a polymer, coupling the functionalized monomers with immunomodulatory agent moieties, and polymerizing the coupled monomers is provided.
  • a method of producing a polymer comprising at least two immunomodulatory agent moieties along the polymer backbone comprising providing a monomer functionalized with immunomodulatory agent moieties, and polymerizing the monomer.
  • the monomer comprises lactide, glycolide or caprolactone monomer.
  • a method of producing a polymer comprising at least two immunomodulatory agent moieties along the polymer backbone, comprising producing or obtaining reactive bifunctional immunomodulatory agent moieties, and reacting the bifunctional immunomodulatory agent moieties such that a polymer is formed is provided.
  • the immunomodulatory agent moieties comprise a TLR agonist.
  • the TLR agonist comprises a TLR-7 and/or TLR-8 agonist.
  • the TLR agonist comprises an aminodiazepine, adenine derivative or an imidazoquinoline.
  • the imidazoquinoline comprises resiquimod or imiquimod.
  • the adenine derivative comprises PF-4171455 or SM-276001.
  • any of the methods provided further comprise producing a synthetic nanocarrier with the polymer.
  • a polymer, synthetic nanocarrier or vaccine obtainable by a process comprising the steps of any method provided herein is provided.
  • any of the compositions, dosage forms, vaccines, polymers or synthetic nanocarriers can be for use in therapy or prophylaxis.
  • compositions, dosage forms, vaccines, polymers or synthetic nanocarriers can be for use in any of the methods provided herein.
  • any of the compositions, dosage forms, vaccines, polymers or synthetic nanocarriers can be for use in a method of treating or preventing cancer.
  • any of the compositions, dosage forms, vaccines, polymers or synthetic nanocarriers can be for use in a method of treating or preventing infection or infectious disease.
  • any of the compositions, dosage forms, vaccines, polymers or synthetic nanocarriers can be for use in a method of treating or preventing an autoimmune disease, an inflammatory disease, an allergy or graft versus host disease.
  • any of the compositions, dosage forms, vaccines, polymers or synthetic nanocarriers can be for use in a method of treating a subject that has undergone or will undergo transplantation.
  • any of the compositions, dosage forms, vaccines, polymers or synthetic nanocarriers can be for the manufacture of a medicament for use in any of the methods provided herein.
  • a polymer includes a mixture of two or more such molecules or a mixture of differing molecular weights of a single polymer species
  • a synthetic nanocarrier includes a mixture of two or more such synthetic nanocarriers or a plurality of such synthetic nanocarriers
  • reference to “a DNA molecule” includes a mixture of two or more such DNA molecules or a plurality of such DNA molecules
  • reference to “an adjuvant” includes mixture of two or more such adjuvant molecules or a plurality of such adjuvant molecules, and the like.
  • the term “comprise” or variations thereof such as “comprises” or “comprising” are to be read to indicate the inclusion of any recited integer (e.g. a feature, element, characteristic, property, method/process step or limitation) or group of integers (e.g. features, elements, characteristics, properties, method/process steps or limitations) but not the exclusion of any other integer or group of integers.
  • the term “comprising” is inclusive and does not exclude additional, unrecited integers or method/process steps.
  • compositions and methods comprising or may be replaced with “consisting essentially of” or “consisting of”.
  • the phrase “consisting essentially of” is used herein to require the specified integer(s) or steps as well as those which do not materially affect the character or function of the claimed invention.
  • the term “consisting” is used to indicate the presence of the recited integer (e.g. a feature, element, characteristic, property, method/process step or limitation) or group of integers (e.g. features, elements, characteristics, properties, method/process steps or limitations) alone.
  • the inventors have unexpectedly discovered that it is possible to produce polymers comprising multiple immunomodulatory agent moietes (e.g., at least two, three, four, five, six, seven, eight, nine, ten, or more moieties of immunomodulatory agent per polymer) and use such polymers to form synthetic nanocarriers.
  • These polymers can be useful for targeted delivery of high concentrations of immunomodulatory agents to cells, such as dendritic cells, and can be useful in the treatment and prevention of diseases and conditions, such as cancer, infection or infectious disease, addiction, allergy, autoimmune disease, inflammatory disease, etc.
  • multiply-loaded polymers e.g., moles of immunomodulatory agent per weight polymer
  • a composition comprising synthetic nanocarriers comprising a first type of polymer that comprises at least two immunomodulatory agent moieties.
  • at least a portion of the immunomodulatory agent moieties are not present at the surface of the synthetic nanocarriers.
  • at least one of the immunomodulatory agent moieties of a polymer is not at the terminus (or end) of the polymer.
  • the synthetic nanocarrier comprises another material in addition to the immunomodulatory agent moieties or the polymer comprises at least one type of monomeric residue that is not the immunomodulatory agent moiety.
  • compositions are further provided.
  • compositions provided herein are also provided.
  • the subject is a human.
  • adjuvant means an agent that does not constitute a specific antigen, but boosts the strength and longevity of immune response to a concomitantly administered antigen.
  • adjuvants may include, but are not limited to stimulators of pattern recognition receptors, such as Toll-like receptors, RIG-1 and NOD-like receptors (NLR), etc.
  • adjuvants comprise agonists for pattern recognition receptors (PRR), including, but not limited to Toll-Like Receptors (TLRs), specifically TLRs 2, 3, 4, 5, 7, 8, 9 and/or combinations thereof.
  • PRR pattern recognition receptors
  • TLRs Toll-Like Receptors
  • adjuvants comprise agonists for Toll-Like Receptors 3, agonists for Toll-Like Receptors 7 and 8, or agonists for Toll-Like Receptor 9; preferably the recited adjuvants comprise aminodiazepine, such as those disldosed in WO 2010/054215, WO 2007/040840, US 2008/0306050, US 2008/0234251; imidazoquinolines; such as R848; adenine derivatives, such as those disclosed in U.S. Pat. No.
  • the adenine derivative comprises SM-276001(9-benzyl-2-butoxy-8-hydroxyadenine) and its analogs with different substituents at C-9 and C-2 positions and 8-oxo-purine derivatives such as PF-4171455 (4-Amino-1-benzyl-6-trifluoromethyl-1,3-dihydroimidazol[4,5-c]pyridin-2-one) and its analogs with different substituents at C-6 position.
  • PF-4171455 4-Amino-1-benzyl-6-trifluoromethyl-1,3-dihydroimidazol[4,5-c]pyridin-2-one
  • synthetic nanocarriers incorporate as adjuvants compounds that are agonists for toll-like receptors (TLRs) 7 & 8 (“TLR 7/8 agonists”).
  • TLR 7/8 agonists include the TLR 7/8 agonist compounds disclosed in U.S. Pat. No. 6,696,076 to Tomai et al., including but not limited to imidazoquinoline amines, imidazopyridine amines, 6,7-fused cycloalkylimidazopyridine amines, and 1,2-bridged imidazoquinoline amines.
  • Preferred adjuvants comprise imiquimod and resiquimod (also known as R848).
  • an adjuvant may be an agonist for the DC surface molecule CD40.
  • a synthetic nanocarrier incorporates an adjuvant that promotes DC maturation (needed for priming of naive T cells) and the production of cytokines, such as type I interferons, which promote antibody immune responses.
  • an adjuvant may be a TLR-4 agonist.
  • adjuvants may comprise TLR-5 agonists.
  • synthetic nanocarriers incorporate a ligand for Toll-like receptor (TLR)-9. Examples of TLR9 antagonists include hydroxychloroquine and its analogs as well as adenine derivatives.
  • administering means providing a material, such as a drug to a subject in a manner that is pharmacologically useful.
  • an “allergy” also referred to herein as an “allergic condition,” is any condition where there is an undesired (e.g., a Type 1 hypersensitive) immune response (i.e., allergic response or reaction) to a substance.
  • allergens include, but are not limited to, allergic asthma, hay fever, hives, eczema, plant allergies, bee sting allergies, pet allergies, latex allergies, mold allergies, cosmetic allergies, food allergies, allergic rhinitis or coryza, topic allergic reactions, anaphylaxis, atopic dermatitis, hypersensitivity reactions and other allergic conditions.
  • the allergic reaction may be the result of an immune reaction to any allergen.
  • the allergy is a food allergy.
  • Food allergies include, but are not limited to, milk allergies, egg allergies, nut allergies, fish allergies, shellfish allergies, soy allergies or wheat allergies.
  • “Amount effective” is any amount of a composition provided herein that produces one or more desired responses, such as one or more desired immune responses. This amount can be for in vitro or in vivo purposes. For in vivo purposes, the amount can be one that a clinician would believe may have a clinical benefit for a subject in need thereof. In embodiments, clinically effective amounts are effective amounts that can be helpful in the treatment of a subject with a disease or condition. Such subjects include, in some embodiments, those that have or are at risk of having cancer, an infection or infectious disease, a non-autoimmune or degenerative disease or an addiction. In other embodiments, the subjects include those that have or are at risk of having an autoimmune disease, an inflammatory disease, an allergy or graft versus host disease or has undergone or will undergo transplantation.
  • a subject's immune response can be monitored by routine methods.
  • An amount that is effective to produce the desired immune responses as provided herein can also be an amount of a composition provided herein that produces a desired therapeutic endpoint or a desired therapeutic result.
  • Amounts effective will depend, of course, on the particular subject being treated; the severity of a condition, disease or disorder; the individual patient parameters including age, physical condition, size and weight; the duration of the treatment; the nature of concurrent therapy (if any); the specific route of administration and like factors within the knowledge and expertise of the health practitioner. These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is generally preferred that a maximum dose be used, that is, the highest safe dose according to sound medical judgment. It will be understood by those of ordinary skill in the art, however, that a patient may insist upon a lower dose or tolerable dose for medical reasons, psychological reasons or for virtually any other reason.
  • doses of the compositions of the invention can range from about 10 ⁇ g/kg to about 100,000 ⁇ g/kg. In some embodiments, the doses can range from about 0.1 mg/kg to about 100 mg/kg. In still other embodiments, the doses can range from about 0.1 mg/kg to about 25 mg/kg, about 25 mg/kg to about 50 mg/kg, about 50 mg/kg to about 75 mg/kg or about 75 mg/kg to about 100 mg/kg. Alternatively, the dose can be administered based on the number of synthetic nanocarriers. For example, useful doses include greater than 10 6 , 10 7 , 10 8 , 10 9 or 10 10 synthetic nanocarriers per dose. Other examples of useful doses include from about 1 ⁇ 10 6 to about 1 ⁇ 10 10 , about 1 ⁇ 10 7 to about 1 ⁇ 10 9 or about 1 ⁇ 10 8 to about 1 ⁇ 10 9 synthetic nanocarriers per dose.
  • Antigen means a B cell antigen or T cell antigen. In embodiments, antigens are coupled to the synthetic nanocarriers. In other embodiments, antigens are not coupled to the synthetic nanocarriers. “Type(s) of antigens” means molecules that share the same, or substantially the same, antigenic characteristics.
  • An “at risk” subject is one in which a health practitioner believes has a chance of having a disease or condition as provided herein.
  • an “autoimmune disease” is any disease where the immune system mounts an undesired immune response against self (e.g., one or more autoantigens).
  • an autoimmune disease comprises an aberrant destruction of cells of the body as part of the self-targeted immune response.
  • the destruction of self manifests in the malfunction of an organ, for example, the colon or pancreas. Examples of autoimmune diseases are described elsewhere herein. Additional autoimmune diseases will be known to those of skill in the art and the invention is not limited in this respect.
  • Average refers to the arithmetic mean unless otherwise noted.
  • B cell antigen means any antigen that is recognized by or triggers an immune response in a B cell (e.g., an antigen that is specifically recognized by a B cell or a receptor thereon).
  • an antigen that is a T cell antigen is also a B cell antigen.
  • the T cell antigen is not also a B cell antigen.
  • B cell antigens include, but are not limited to proteins, peptides, small molecules, oligosaccharides and carbohydrates.
  • the B cell antigen comprises a non-protein antigen (i.e., not a protein or peptide antigen).
  • the B cell antigen comprises a carbohydrate associated with an infectious agent.
  • the B cell antigen comprises a glycoprotein or glycopeptide associated with an infectious agent.
  • the infectious agent can be a bacterium, virus, fungus, protozoan, or parasite.
  • the B cell antigen comprises a poorly immunogenic antigen.
  • the B cell antigen comprises an abused substance or a portion thereof.
  • the B cell antigen comprises an addictive substance or a portion thereof.
  • Addictive substances include, but are not limited to, nicotine, a narcotic, a cough suppressant, a tranquilizer, and a sedative.
  • the B cell antigen comprises a toxin, such as a toxin from a chemical weapon or natural sources.
  • the B cell antigen may also comprise a hazardous environmental agent.
  • the B cell antigen comprises a self antigen.
  • the B cell antigen comprises an alloantigen, an allergen, a contact sensitizer, a degenerative disease antigen, a hapten, an infectious disease antigen, a cancer antigen, an atopic disease antigen, an autoimmune disease antigen, a non-autoimmune disease antigen, an addictive substance, a xenoantigen, or a metabolic disease enzyme or enzymatic product thereof.
  • Couple or “Coupled” or “Couples” (and the like) means to chemically associate one entity (for example a moiety) with another.
  • the coupling is covalent, meaning that the coupling occurs in the context of the presence of a covalent bond between the two entities.
  • the non-covalent coupling is mediated by non-covalent interactions including but not limited to charge interactions, affinity interactions, metal coordination, physical adsorption, host-guest interactions, hydrophobic interactions, TT stacking interactions, hydrogen bonding interactions, van der Waals interactions, magnetic interactions, electrostatic interactions, dipole-dipole interactions, and/or combinations thereof.
  • encapsulation is a form of coupling.
  • Dosage form means a pharmacologically and/or immunologically active material in a medium, carrier, vehicle, or device suitable for administration to a subject.
  • Encapsulate means to enclose at least a portion of a substance within a synthetic nanocarrier. In some embodiments, a substance is enclosed completely within a synthetic nanocarrier. In other embodiments, most or all of a substance that is encapsulated is not exposed to the local environment external to the synthetic nanocarrier. In other embodiments, no more than 50%, 40%, 30%, 20%, 10% or 5% (weight/weight) is exposed to the local environment. Encapsulation is distinct from absorption, which places most or all of a substance on a surface of a synthetic nanocarrier, and leaves the substance exposed to the local environment external to the synthetic nanocarrier.
  • Immunomodulatory agent means an agent that modulates an immune response to an antigen but is not the antigen or derived from the antigen. “Modulate”, as used herein, refers to inducing, enhancing, suppressing, directing, or redirecting an immune response. Such agents include immunostimulatory agents, such as adjuvants, that stimulate (or boost) an immune response to an antigen but is not an antigen or derived from an antigen. There are several distinct types of immunomodulatory agents, which include, but are not limited to, Toll-like Receptor (TLR) agonists and Toll-like Receptor (TLR) antagonists. Such agents also include immunosuppressants.
  • TLR Toll-like Receptor
  • TLR Toll-like Receptor
  • TLR Toll-like Receptor
  • “Moieties” are the active portions of a molecule of the immunomodulatory agents and are useful in the practice of the invention. Such moieties can be chemically modified, e.g., for coupling to the synthetic nanocarrier, and still remain immunologically active. In certain embodiments, multiple moieties of the immunomodulatory agent (e.g., two, three, four, five, six, seven, eight, nine, ten, or more moieties) are coupled to a polymer, e.g., at an end (or terminus) of a polymer, to a polymer backbone, or form at least part of the polymer backbone.
  • a polymer e.g., at an end (or terminus) of a polymer, to a polymer backbone, or form at least part of the polymer backbone.
  • the immunomodulatory agent moieties are incorporated within the synthetic nanocarriers. Some of the immunomodulatory agent moieties may be present at the surface of the synthetic nanocarriers. In some embodiments, not all of the immunomodulatory agent moieties are present at the surface of the synthetic nanocarriers. In some embodiments, all of the immunomodulatory moieties that are attached to or form part of the polymer, or synthetic nanocarrier that comprises the polymer, are the same type of immunomodulatory agent moiety (i.e., are identical to one another in chemical structure).
  • a polymer, or synthetic nanocarrier that comprises the polymer, as provided herein comprises one or more moieties of a number of different types of immunomodulatory agents (e.g., two, three, four, five, six, seven, eight, nine, or ten different types of immunomodulatory agent moieties).
  • a polymer, or synthetic nanocarrier that comprises the polymer, as provided herein comprises exactly one type of immunomodulatory agent moiety.
  • a polymer, or synthetic nanocarrier that comprises the polymer, as provided herein comprises exactly two distinct types of immunomodulatory agent moieties.
  • a polymer, or synthetic nanocarrier that comprises the polymer comprises three or more distinct types of immunomodulatory agent moieties (e.g., three, four, five, six, seven, eight, nine, or ten distinct types of immunomodulatory agent moieties).
  • Immunosuppressant means a compound that causes an immunosuppressive (e.g., tolerogenic) effect.
  • An immunosuppressive effect generally refers to the production or expression of cytokines or other factors by immune cells, such as antigen-presenting cells, that reduce, inhibit or prevent an undesired immune response
  • Immunosuppressants include, but are not limited to, statins; mTOR inhibitors, such as rapamycin or a rapamycin analog; TGF- ⁇ signaling agents; TGF- ⁇ receptor agonists; histone deacetylase inhibitors, such as Trichostatin A; corticosteroids; inhibitors of mitochondrial function, such as rotenone; P38 inhibitors; NF- ⁇ inhibitors, such as 6Bio, Dexamethasone, TCPA-1, IKK VII; adenosine receptor agonists; prostaglandin E2 agonists (PGE2), such as Misoprostol; phosphodiesterase inhibitors, such as phosphodie
  • Immunosuppressants also include IDO, vitamin D3, cyclosporins, such as cyclosporine A, aryl hydrocarbon receptor inhibitors, resveratrol, azathiopurine (Aza), 6-mercaptopurine (6-MP), 6-thioguanine (6-TG), FK506, sanglifehrin A, salmeterol, mycophenolate mofetil (MMF), aspirin and other COX inhibitors, niflumic acid, estriol and triptolide.
  • cyclosporins such as cyclosporine A, aryl hydrocarbon receptor inhibitors, resveratrol, azathiopurine (Aza), 6-mercaptopurine (6-MP), 6-thioguanine (6-TG), FK506, sanglifehrin A, salmeterol, mycophenolate mofetil (MMF), aspirin and other COX inhibitors, niflumic acid, estriol and triptolide.
  • immunosuppressants include, but are not limited, small molecule drugs, natural products, antibodies (e.g., antibodies against CD20, CD3, CD4), biologics-based drugs, carbohydrate-based drugs, nanoparticles, liposomes, RNAi, antisense nucleic acids, aptamers, methotrexate, NSAIDs; fingolimod; natalizumab; alemtuzumab; anti-CD3; tacrolimus (FK506), etc. Further immunosuppressants, are known to those of skill in the art, and the invention is not limited in this respect.
  • infectious disease is any condition or disease caused by a microorganism, pathogen or other agent, such as a bacterium, fungus, prion or virus.
  • infectious disease antigen is an antigen associated with an infection or infectious disease. Such antigens include antigens that can be used to generate an immune response against a pathogen or other infectious agent, or component thereof, or that can generate an immune response against infected cells.
  • Inflammatory disease means any disease, disorder or condition in which undesired inflammation occurs.
  • Load is the amount of a component (e.g., immunomodulatory agent) of a synthetic nanocarrier based on the total weight of materials in an entire synthetic nanocarrier (weight/weight). Generally, the load is calculated as an average across a population of synthetic nanocarriers. In one embodiment, the load of the immunomodulatory agent on average across the synthetic nanocarriers is between 0.0001% and 50%. In another embodiment, the load of the immunomodulatory agent on average across the synthetic nanocarriers is between 0.001% and 50%. In yet another embodiment, the load of the immunomodulatory agent is between 0.01% and 20%. In a further embodiment, the load of the immunomodulatory agent is between 0.1% and 10%. In still a further embodiment, the load of the immunomodulatory agent is between 1% and 10%.
  • the load of the immunomodulatory agent is between 1% and 10%.
  • the load is calculated as follows: Approximately 3 mg of synthetic nanocarriers are collected and centrifuged to separate supernatant from synthetic nanocarrier pellet. Acetonitrile is added to the pellet, and the sample is sonicated and centrifuged to remove any insoluble material. The supernatant and pellet are injected on RP-HPLC and absorbance is read at 278 nm. The ⁇ g found in the pellet is used to calculate % entrapped (load), ⁇ g in supernatant and pellet are used to calculate total ⁇ g recovered.
  • “Maximum dimension of a synthetic nanocarrier” means the largest dimension of a nanocarrier measured along any axis of the synthetic nanocarrier. “Minimum dimension of a synthetic nanocarrier” means the smallest dimension of a synthetic nanocarrier measured along any axis of the synthetic nanocarrier. For example, for a spheroidal synthetic nanocarrier, the maximum and minimum dimension of a synthetic nanocarrier would be substantially identical, and would be the size of its diameter. Similarly, for a cuboidal synthetic nanocarrier, the minimum dimension of a synthetic nanocarrier would be the smallest of its height, width or length, while the maximum dimension of a synthetic nanocarrier would be the largest of its height, width or length.
  • a minimum dimension of at least 75%, preferably at least 80%, more preferably at least 90%, of the synthetic nanocarriers in a sample, based on the total number of synthetic nanocarriers in the sample is equal to or greater than 100 nm.
  • a maximum dimension of at least 75%, preferably at least 80%, more preferably at least 90%, of the synthetic nanocarriers in a sample, based on the total number of synthetic nanocarriers in the sample is equal to or less than 5 ⁇ m.
  • a minimum dimension of at least 75%, preferably at least 80%, more preferably at least 90%, of the synthetic nanocarriers in a sample, based on the total number of synthetic nanocarriers in the sample is greater than 110 nm, more preferably greater than 120 nm, more preferably greater than 130 nm, and more preferably still greater than 150 nm.
  • Aspects ratios of the maximum and minimum dimensions of synthetic nanocarriers may vary depending on the embodiment.
  • aspect ratios of the maximum to minimum dimensions of the synthetic nanocarriers may vary from 1:1 to 1,000,000:1, preferably from 1:1 to 100,000:1, more preferably from 1:1 to 10,000:1, more preferably from 1:1 to 1000:1, still more preferably from 1:1 to 100:1, and yet more preferably from 1:1 to 10:1.
  • a maximum dimension of at least 75%, preferably at least 80%, more preferably at least 90%, of the synthetic nanocarriers in a sample, based on the total number of synthetic nanocarriers in the sample is equal to or less than 3 ⁇ m, more preferably equal to or less than 2 ⁇ m, more preferably equal to or less than 1 ⁇ m, more preferably equal to or less than 800 nm, more preferably equal to or less than 600 nm, and more preferably still equal to or less than 500 nm.
  • a minimum dimension of at least 75%, preferably at least 80%, more preferably at least 90%, of the synthetic nanocarriers in a sample, based on the total number of synthetic nanocarriers in the sample is equal to or greater than 100 nm, more preferably equal to or greater than 120 nm, more preferably equal to or greater than 130 nm, more preferably equal to or greater than 140 nm, and more preferably still equal to or greater than 150 nm.
  • Measurement of synthetic nanocarrier dimensions e.g., diameter
  • DLS dynamic light scattering
  • a suspension of synthetic nanocarriers can be diluted from an aqueous buffer into purified water to achieve a final synthetic nanocarrier suspension concentration of approximately 0.01 to 0.1 mg/mL.
  • the diluted suspension may be prepared directly inside, or transferred to, a suitable cuvette for DLS analysis.
  • the cuvette may then be placed in the DLS, allowed to equilibrate to the controlled temperature, and then scanned for sufficient time to acquire a stable and reproducible distribution based on appropriate inputs for viscosity of the medium and refractive indicies of the sample.
  • the effective diameter, or mean of the distribution is then reported.
  • “Dimension” or “size” or “diameter” of synthetic nanocarriers means the mean of a particle size distribution obtained using dynamic light scattering.
  • “Not present at the surface of synthetic nanocarriers” refers to an entity that is not exposed to the environment that is external to the synthetic nanocarrier.
  • “Pharmaceutically acceptable excipient” means a pharmacologically inactive material used together with the recited synthetic nanocarriers to formulate the compositions.
  • Pharmaceutically acceptable excipients comprise a variety of materials known in the art, including but not limited to saccharides (such as glucose, lactose, and the like), preservatives such as antimicrobial agents, reconstitution aids, colorants, saline (such as phosphate buffered saline), and buffers.
  • Polymeric monomer refers to a monomeric unit of a polymer, the polymer generally being made up of a series of linked monomeric residues.
  • Subject means animals, including warm blooded mammals such as humans and primates; avians; domestic household or farm animals such as cats, dogs, sheep, goats, cattle, horses and pigs; laboratory animals such as mice, rats and guinea pigs; fish; reptiles; zoo and wild animals; and the like.
  • “Synthetic nanocarrier(s)” means a discrete object that is not found in nature, and that possesses at least one dimension that is less than or equal to 5 microns in size.
  • Albumin nanoparticles are generally included as synthetic nanocarriers, however in certain embodiments the synthetic nanocarriers do not comprise albumin nanoparticles.
  • synthetic nanocarriers do not comprise chitosan.
  • the synthetic nanocarriers do not comprise chitosan.
  • synthetic nanocarriers are not lipid-based nanoparticles.
  • synthetic nanocarriers do not comprise a phospholipid.
  • a synthetic nanocarrier can be, but is not limited to, one or a plurality of lipid-based nanoparticles (also referred to herein as lipid nanoparticles, i.e., nanoparticles where the majority of the material that makes up their structure are lipids), polymeric nanoparticles, metallic nanoparticles, surfactant-based emulsions, dendrimers, buckyballs, nanowires, virus-like particles (i.e., particles that are primarily made up of viral structural proteins but that are not infectious or have low infectivity), peptide or protein-based particles (also referred to herein as protein particles, i.e., particles where the majority of the material that makes up their structure are peptides or proteins) (such as albumin nanoparticles) and/or nanoparticles that are developed using a combination of nanomaterials such as lipid-polymer nanoparticles.
  • lipid-based nanoparticles also referred to herein as lipid nanoparticles, i
  • Synthetic nanocarriers may be a variety of different shapes, including but not limited to spheroidal, cuboidal, pyramidal, oblong, cylindrical, toroidal, and the like. Synthetic nanocarriers according to the invention comprise one or more surfaces. Exemplary synthetic nanocarriers that can be adapted for use in the practice of the present invention comprise: (1) the biodegradable nanoparticles disclosed in U.S. Pat. No.
  • synthetic nanocarriers may possess an aspect ratio greater than 1:1, 1:1.2, 1:1.5, 1:2, 1:3, 1:5, 1:7, or greater than 1:10.
  • Synthetic nanocarriers according to the invention that have a minimum dimension of equal to or less than about 100 nm, preferably equal to or less than 100 nm, do not comprise a surface with hydroxyl groups that activate complement or alternatively comprise a surface that consists essentially of moieties that are not hydroxyl groups that activate complement.
  • synthetic nanocarriers according to the invention that have a minimum dimension of equal to or less than about 100 nm, preferably equal to or less than 100 nm, do not comprise a surface that substantially activates complement or alternatively comprise a surface that consists essentially of moieties that do not substantially activate complement.
  • synthetic nanocarriers according to the invention that have a minimum dimension of equal to or less than about 100 nm, preferably equal to or less than 100 nm, do not comprise a surface that activates complement or alternatively comprise a surface that consists essentially of moieties that do not activate complement.
  • synthetic nanocarriers exclude virus-like particles.
  • the virus-like particles comprise non-natural adjuvant (meaning that the VLPs comprise an adjuvant other than naturally occurring RNA generated during the production of the VLPs).
  • synthetic nanocarriers may possess an aspect ratio greater than 1:1, 1:1.2, 1:1.5, 1:2, 1:3, 1:5, 1:7, or greater than 1:10.
  • T cell antigen means any antigen that is recognized by and triggers an immune response in a T cell (e.g., an antigen that is specifically recognized by a T cell receptor on a T cell or an NKT cell via presentation of the antigen or portion thereof bound to a Class I or Class II major histocompatability complex molecule (MHC), or bound to a CD1 complex).
  • an antigen that is a T cell antigen is also a B cell antigen.
  • the T cell antigen is not also a B cell antigen.
  • T cell antigens generally are proteins or peptides.
  • T cell antigens may be an antigen that stimulates a CD8+ T cell response, a CD4+ T cell response, or both. The nanocarriers, therefore, in some embodiments can effectively stimulate both types of responses.
  • the T cell antigen is a T helper cell antigen (i.e. one that can generate an enhanced response to a B cell antigen, preferably an unrelated B cell antigen, through stimulation of T cell help).
  • a T helper cell antigen may comprise one or more peptides obtained or derived from tetanus toxoid, Epstein-Barr virus, influenza virus, respiratory syncytial virus, measles virus, mumps virus, rubella virus, cytomegalovirus, adenovirus, diphtheria toxoid, or a PADRE peptide (known from the work of Sette et al. U.S. Pat. No. 7,202,351).
  • a T helper cell antigen may comprise one or more lipids, or glycolipids, including but not limited to: ⁇ -galactosylceramide ( ⁇ -GalCer), ⁇ -linked glycosphingolipids (from Sphingomonas spp.), galactosyl diacylglycerols (from Borrelia burgdorferi ), lypophosphoglycan (from Leishmania donovani ), and phosphatidylinositol tetramannoside (PIM4) (from Mycobacterium leprae ).
  • ⁇ -galactosylceramide ⁇ -GalCer
  • ⁇ -linked glycosphingolipids from Sphingomonas spp.
  • galactosyl diacylglycerols from Borrelia burgdorferi
  • lypophosphoglycan from Leishmania donovani
  • PIM4 phosphatidylinositol tetramann
  • CD4+ T-cell antigens may be derivatives of a CD4+ T-cell antigen that is obtained from a source, such as a natural source.
  • CD4+ T-cell antigen sequences such as those peptides that bind to MHC II, may have at least 70%, 80%, 90%, or 95% identity to the antigen obtained from the source.
  • the T cell antigen preferably a T helper cell antigen, may be coupled to, or uncoupled from, a synthetic nanocarrier.
  • the T cell antigen is encapsulated in the synthetic nanocarriers of the compositions.
  • a “terminus of a polymer” is an end of a polymer chain or branch.
  • Vaccine means a composition of matter that improves the immune response to a particular pathogen or disease.
  • a vaccine typically contains factors that stimulate a subject's immune system to recognize a specific antigen as foreign and eliminate it from the subject's body.
  • a vaccine also establishes an immunologic ‘memory’ so the antigen will be quickly recognized and responded to if a person is re-challenged.
  • Vaccines can be prophylactic (for example to prevent future infection by any pathogen), or therapeutic (for example a vaccine against a tumor specific antigen for the treatment of cancer).
  • a vaccine may comprise dosage forms according to the invention.
  • Weight refers to mass unless otherwise noted. When a molecular weight of a polymer is measured, it can be measured as the weight average molecular weight or a number average molecular weight. “Weight average molecular weight” for the polymers of the compositions provided herein is calculated by the following formula:
  • M _ w ⁇ i ⁇ N i ⁇ ⁇ M i 2 ⁇ i ⁇ N i ⁇ M i
  • Weight average molecular weight can be determined by a variety of methods including light scattering, small angle neutron scattering (SANS), X-ray scattering, Nuclear Magnetic Resonance (NMR) and sedimentation velocity.
  • SANS small angle neutron scattering
  • NMR Nuclear Magnetic Resonance
  • An example of an alternative for weight average molecular weight is to perform gel permeation chromatography using suitable traceable-weight standards to establish a retention-time versus weight curve, and calculating the mean weight-averaged molecular weight of a sample polymer from the mean of the integrated sample peak as compared to the calibration curve.
  • the “number average molecular weight” can be determined by NMR.
  • number average molecular weight can be determined by proton NMR wherein the ratio of the polymer repeating units to the end group is established and then multiplied by theoretical repeating unit molecular weight.
  • a known weight concentration may be established and then titrated in the presense of an indicator dye with an appropriate neutralizing agent of known molar concentration to provide moles of end group per mass of polymer.
  • Any of the weights of a polymer as provided herein can be a weight average molecular weight or a number average molecular weight.
  • the inventors have discovered that it is possible to couple multiple molecules of immunomodulatory agent to a polymer backbone, e.g., coupling two, three, four, five, six, seven, eight, nine, ten, or more moieties of immunomodulatory agent to the polymer.
  • Such immunomodulatory agent moieties are, for example, coupled to the terminus (or end) of a polymer and/or or to its backbone and/or are coupled to monomers and/or themselves are monomers used in the preparation of a polymer.
  • the present invention therefore, also provides synthetic nanocarriers comprising such polymers. Further provided are synthetic nanocarriers that further comprise other components coupled thereto, such as additional immunomodulatory agents, antigens, etc.
  • the immunomodulatory agent moieties can be coupled to or form the polymers by a variety of methods.
  • the immunomodulatory agent moieties e.g., adjuvant moieties
  • the polymer is a branched polymer prepared from the reaction of a polyol with one or more different types of monomers to provide a multi-armed polymer (see, e.g., Scheme 3).
  • the polyol wherein n is an integer between 2 and 100, is reacted with lactide, glycolide, or caprolactam monomers, any of which may be functionalized with other groups, such as optionally substituted alkyl groups, to provide a multi-armed polymer.
  • R 1 is selected from hydrogen and a polymer, wherein at least two R 1 groups comprise a polymer arm.
  • the polymer arm of the multi-armed polymer may be further functionalized with other monomers, e.g., lactide, glycolide, or caprolactam monomers, to provide a block-co-polymer arm.
  • the polymer arm may then be further functionalized with one or more groups useful for conjugation with an appropriately functionalized immunomodulatory agent moiety.
  • a polymer is designed such that multiple immunomodulatory agent moieties may be installed along the branch points of the polymer.
  • the polymer is treated with succinic anhydride to provide a polyacid, e.g., a polymer comprising two or more carboxylic acid groups.
  • Scheme 3 depicts only one exemplary way of installing a carboxylic acid via a linking group L 1 , and many other ways are contemplated, e.g., wherein L 1 may be any group linking the polymer to one or more carboxylic acids.
  • Exemplary L 1 groups include, but are not limited to, optionally substituted alkylene, optionally substituted alkenylene, optionally substituted alkynylene, optionally substituted carbocyclene, optionally substituted heterocyclene, optionally substituted arylene, and optionally substituted heteroarylene.
  • L 1 is a substituted alkylene group, i.e., —C( ⁇ O)CH 2 CH 2 —, linking the polymer arm to a terminal carboxylic acid —CO 2 H to provide a polyacid.
  • the polyacid may then be conjugated to an appropriately functionalized immunomodulatory agent moiety (e.g., comprising an amino group —NH 2 ), or the polyacid may be used as a handle to further functionalize the polymer arm prior to conjugation with an immunomodulatory agent moiety.
  • an appropriately functionalized immunomodulatory agent moiety e.g., comprising an amino group —NH 2
  • the polyacid may be used as a handle to further functionalize the polymer arm prior to conjugation with an immunomodulatory agent moiety.
  • Scheme 4 depicts these two possible situations.
  • the polyacid is eventually conjugated via an amide linker to a group R5, wherein R5 comprises an immunomodulatory agent moiety, e.g., an adjuvant moiety such as a TLR agonist moiety.
  • Exemplary polyols for use in the compositions and methods provided herein include the following:
  • polyols can be found in the literature (see, e.g., Chem Soc Rev 2011, 40, 1761-1776) and include the following: trimethylolpropane (3-arm), glycerol (3-arm), pentaerythritol (4-arm), erythritol (4-arm), xylitol (5-arm), di(trimethylolpropane) (4-arm), sorbitol (6-arm), inositol (6-arm) and tripentaerythritol (8-arm).
  • R 5 may also be coupled to NH or OH as an alternative to NH2 in the above scheme.
  • the polymer is a linear polymer functionalized along the polymeric backbone with carboxylic acid groups.
  • the polyacid may be conjugated to an appropriately functionalized immunomodulatory agent moiety (e.g., comprising an amino group —NH 2 ), or the polyacid may be used as a handle to further functionalize the polymer arm prior to conjugation with an immunomodulatory agent moiety.
  • the polyacid is eventually conjugated via an amide linker to a group R 5 , wherein R 5 is an immunomodulatory agent moiety, e.g., a TLR agonist moiety of formulae (i), (ii), or (iii).
  • R 5 may also be coupled to NH or OH as an alternative to NH2 in the below scheme.
  • Acid functionalized polymer Coupled polymer R 7 —OH and —NHR 5 , wherein at least two R 7 is —NHR 5
  • a polycaprolactone polymer is alkylated at the ⁇ -position via an enolate intermediate with various groups, e.g., —CH 2 OH, —CO 2 H, and/or other groups comprising nucleophilic or electrophilic moieties, which ultimately are used in the conjugation to an appropriately functionalized immunomodulatory agent moiety, e.g., NH 2 R 5 , wherein R 5 is a group of the formulae (I), (ii), or (iii).
  • R 5 may also be coupled to NH or OH as an alternative to NH2 in the above scheme.
  • the side chain chemical moiety is a carboxylic acid and the immunomodulatory agent moiety, comprising an amino group, is coupled to carboxylic acid group in the presence of a peptide coupling agent.
  • a peptide coupling agent used for peptide synthesis may be used. These include, for example, EDC/NHS or DCC/NHS; TBTU/base (DIPEA or Et3N), HBTU/base; PyBop/base, etc. These may also be used for making ester bond if excess base is present.
  • Scheme 7 depicts a particular embodiment wherein the polymer arm of the multi-armed polymer functionalized with a terminal hydroxyl group is conjugated to the immunomodulatory agent moiety via an amide linker through nucleophilic attack of a morpholine-3,5-dione present thereon.
  • Other methods of coupling a terminal hydroxyl group present on the polymer to an electrophilic group present on the immunomodulatory agent moiety are further contemplated herein, e.g., nucleophilic attack of an ⁇ , ⁇ -unsaturated group or vinyl sulfone via Michael addition.
  • Other electrophilic groups for functionalizing the immunomodulatory agent moiety contemplated herein include ⁇ -halo acetic acid ester or an amide derivative of the immunomodulatory agent moiety.
  • the polymer is prepared from a monomer which is functionalized with the immunomodulatory agent moiety, or is functionalized with a group suitable for conjugation to the immunomodulatory agent moiety after polymerization.
  • Schemes 8 and 9 depict construction of a functionalized polycaprolactone and functionalized polylactide via such a monomer.
  • a caprolactone monomer functionalized with an acrylate group is polymerized to provide a polycaprolactone polymer comprising an acrylate group, wherein A and Z are terminal groups, A is the initiator for the ring-opening polymerization (ROP) of the lactone, such as simple alcohol, MeO-PEG-OH; Z is Hydrogen in such case.
  • ROP ring-opening polymerization
  • the functionalized polycaprolactone polymer is then conjugated to the immunomodulatory agent via Michael addition.
  • the immunomodulatory agent moiety may comprise a nucleophilic group, e.g., an —OH, —SH, or —NH 2 group, optionally tethered to the amino group, wherein L 3 is optionally substituted alkylene, optionally substituted alkenylene, optionally substituted alkynylene, optionally substituted carbocyclene, optionally substituted heterocyclene, optionally substituted arylene, or optionally substituted heteroarylene, to provide the conjugated product.
  • a nucleophilic group e.g., an —OH, —SH, or —NH 2 group
  • L 3 is optionally substituted alkylene, optionally substituted alkenylene, optionally substituted alkynylene, optionally substituted carbocyclene, optionally substituted heterocyclene, optionally substituted arylene, or optionally substituted heteroarylene, to provide the conjugated product.
  • the polylactide is constructed from a functionalized lactide monomer wherein R 9 is an oxygen protecting group. Subsequent deprotection and treatment with succinic anhydride provides a polymer suitable for conjugation to an immunomodulatory agent moiety, e.g., NH 2 R 5 , wherein R 5 is a group of the formulae (i), (ii), or (iii). In some embodiment, R 5 may also be coupled to NH or OH as an alternative to NH2 as described above.
  • the synthetic nanocarriers provided herein comprise polymers to which immunomodulatory agent moieties are coupled. Such polymers can form part of or all of a synthetic nanocarrier. In some embodiments, such polymers form part of or all of a synthetic nanocarrier that is completely polymeric. In such embodiments, the polymeric synthetic nanocarriers may comprise other polymers to which immunomodulatory agents are not attached. These other polymers may be the same type or a different type of polymer as those that are coupled to the immunmodulatory agent moieties. In embodiments where the synthetic nanocarriers are not completely polymeric, the polymers that are coupled to the immunomodulatory agent moieties form part of the synthetic nanocarriers, and the synthetic nanocarriers are made up one or more different materials.
  • the polymers provided herein may be coupled to the immunomodulatory agent moieties and form at least part of the synthetic nanocarriers as provided or are not coupled to the immunomodulatory agent moieties but still form part of the synthetic nanocarriers.
  • Such polymers preferably have a molecular weight of at least 2000 Da (as weight average or number average molecular weight).
  • the polymers have a molecular weight of at least 2500 Da, 3000 Da, 3500 Da, 4000 Da, 4500 Da, 5000 Da, 5500 Da, 6000 Da, 6500 Da, 7000 Da, 7500 Da, etc.
  • the polymers have a molecular weight of 2000 Da, 2500 Da, 3000 Da, 3500 Da, 4000 Da, 4500 Da, 5000 Da, 5500 Da, 6000 Da, 6500 Da, 7000 Da, 7500 Da, etc.
  • the molecular weights are weight average molecular weights or number average molecular weights.
  • the polymer comprises polyethylene glycol the molecular weight is a number average molecular weight.
  • the polymer does not comprise polyethylene glycol the molecular weight is the weight average molecular weight.
  • the polymers provided herein can comprise one or more types of polymers (e.g., a co-polymer and/or a block co-polymer).
  • polymers suitable for coupling to the immunomodulatory agent moieties or that are otherwise used to produce the synthetic nanocarriers of the present invention include, but are not limited to polyethylenes, polycarbonates (e.g. poly(1,3-dioxan-2one)), polyanhydrides (e.g. poly(sebacic anhydride)), polypropylfumerates, polyamides (e.g. polycaprolactam), polyacetals, polyethers, polyesters (e.g., polylactide, polyglycolide, polylactide-co-glycolide, polycaprolactone, polyhydroxyacid (e.g.
  • polymers in accordance with the present invention include polymers which have been approved for use in humans by the U.S. Food and Drug Administration (FDA) under 21 C.F.R. ⁇ 177.2600, including but not limited to polyesters (e.g., polylactic acid, poly(lactic-co-glycolic acid), polycaprolactone, polyvalerolactone, poly(1,3-dioxan-2one)); polyanhydrides (e.g., poly(sebacic anhydride)); polyethers (e.g., polyethylene glycol); polyurethanes; polymethacrylates; polyacrylates; and polycyanoacrylates.
  • FDA U.S. Food and Drug Administration
  • polymers can be hydrophilic.
  • polymers may comprise anionic groups (e.g., phosphate group, sulphate group, carboxylate group); cationic groups (e.g., quaternary amine group); or polar groups (e.g., hydroxyl group, thiol group, amine group).
  • a synthetic nanocarrier comprising a hydrophilic polymeric matrix generates a hydrophilic environment within the synthetic nanocarrier.
  • polymers can be hydrophobic.
  • a synthetic nanocarrier comprising a hydrophobic polymeric matrix generates a hydrophobic environment within the synthetic nanocarrier. Selection of the hydrophilicity or hydrophobicity of the polymer may have an impact on the nature of materials that are incorporated (e.g., coupled) within the synthetic nanocarrier.
  • polymers may be modified with one or more moieties and/or functional groups.
  • moieties or functional groups can be used in accordance with the present invention.
  • polymers may be modified with polyethylene glycol (PEG), with a carbohydrate, and/or with acyclic polyacetals derived from polysaccharides (Papisov, 2001, ACS Symposium Series, 786:301). Certain embodiments may be made using the general teachings of U.S. Pat. No. 5,543,158 to Gref et al., or WO publication WO2009/051837 by Von Andrian et al.
  • polymers may be modified with a lipid or fatty acid group.
  • a fatty acid group may be one or more of butyric, caproic, caprylic, capric, lauric, myristic, palmitic, stearic, arachidic, behenic, or lignoceric acid.
  • a fatty acid group may be one or more of palmitoleic, oleic, vaccenic, linoleic, alpha-linoleic, gamma-linoleic, arachidonic, gadoleic, arachidonic, eicosapentaenoic, docosahexaenoic, or erucic acid.
  • polymers may be polyesters, including copolymers comprising lactic acid and glycolic acid units, such as poly(lactic acid-co-glycolic acid) and poly(lactide-co-glycolide), collectively referred to herein as “PLGA”; and homopolymers comprising glycolic acid units, referred to herein as “PGA,” and lactic acid units, such as poly-L-lactic acid, poly-D-lactic acid, poly-D,L-lactic acid, poly-L-lactide, poly-D-lactide, and poly-D,L-lactide, collectively referred to herein as “PLA.”
  • exemplary polyesters include, for example, polyhydroxyacids; PEG copolymers and copolymers of lactide and glycolide (e.g., PLA-PEG copolymers, PGA-PEG copolymers, PLGA-PEG copolymers, and derivatives thereof.
  • polyesters include, for example, poly(caprolactone), poly(caprolactone)-PEG copolymers, poly(L-lactide-co-L-lysine), poly(serine ester), poly(4-hydroxy-L-proline ester), poly[ ⁇ -(4-aminobutyl)-L-glycolic acid], and derivatives thereof.
  • a polymer may be PLGA.
  • PLGA is a biocompatible and biodegradable co-polymer of lactic acid and glycolic acid, and various forms of PLGA are characterized by the ratio of lactic acid:glycolic acid.
  • Lactic acid can be L-lactic acid, D-lactic acid, or D,L-lactic acid.
  • the degradation rate of PLGA can be adjusted by altering the lactic acid:glycolic acid ratio.
  • PLGA to be used in accordance with the present invention is characterized by a lactic acid:glycolic acid ratio of approximately 85:15, approximately 75:25, approximately 60:40, approximately 50:50, approximately 40:60, approximately 25:75, or approximately 15:85.
  • polymers may be 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 anhydride), methyl methacrylate, polymethacrylate, poly(methyl methacrylate) copolymer, polyacrylamide, aminoalkyl methacrylate copolymer, glycidyl methacrylate copolymers, polycyanoacrylates, and combinations comprising one or more of the foregoing polymers.
  • the acrylic polymer may comprise fully-polymerized copolymers of acrylic and methacrylic acid esters with a low content of quaternary ammoni
  • polymers can be cationic polymers.
  • cationic polymers are able to condense and/or protect negatively charged strands of nucleic acids (e.g. DNA, or derivatives thereof).
  • Amine-containing polymers such as poly(lysine) (Zauner et al., 1998, Adv. Drug Del. Rev., 30:97; and Kabanov et al., 1995, Bioconjugate Chem., 6:7), poly(ethylene imine) (PEI; Boussif et al., 1995, Proc. Natl. Acad.
  • the synthetic nanocarriers may not comprise (or may exclude) cationic polymers.
  • polymers can be degradable polyesters bearing cationic side chains (Putnam et al., 1999, Macromolecules, 32:3658; Barrera et al., 1993, J. Am. Chem. Soc., 115:11010; Kwon et al., 1989, Macromolecules, 22:3250; Lim et al., 1999, J. Am. Chem. Soc., 121:5633; and Zhou et al., 1990, Macromolecules, 23:3399).
  • polyesters include poly(L-lactide-co-L-lysine) (Barrera et al., 1993, J. Am. Chem.
  • polymers can be linear or branched polymers. In some embodiments, polymers can be dendrimers. In some embodiments, polymers can be substantially cross-linked to one another. In some embodiments, polymers can be substantially free of cross-links. In some embodiments, polymers can be used in accordance with the present invention without undergoing a cross-linking step. It is further to be understood that synthetic nanocarriers may comprise block copolymers, graft copolymers, blends, mixtures, and/or adducts of any of the foregoing and other polymers. Those skilled in the art will recognize that the polymers listed herein represent an exemplary, not comprehensive, list of polymers that can be of use in accordance with the present invention.
  • the polymers can form a polymerix matrix.
  • a polymeric matrix comprises one or more polymers.
  • Polymers may be natural or unnatural (synthetic) polymers.
  • Polymers may be homopolymers or copolymers comprising two or more monomers. In terms of sequence, copolymers may be random, block, or comprise a combination of random and block sequences.
  • polymers in accordance with the present invention are organic polymers.
  • synthetic nanocarriers are spheres or spheroids.
  • synthetic nanocarriers are flat or plate-shaped.
  • synthetic nanocarriers are cubes or cubic.
  • synthetic nanocarriers are ovals or ellipses.
  • synthetic nanocarriers are cylinders, cones, or pyramids.
  • a population of synthetic nanocarriers that is relatively uniform in terms of size, shape, and/or composition so that each synthetic nanocarrier has similar properties. For example, at least 80%, at least 90%, or at least 95% of the synthetic nanocarriers, based on the total number of synthetic nanocarriers, may have a minimum dimension or maximum dimension that falls within 5%, 10%, or 20% of the average diameter or average dimension of the synthetic nanocarriers. In some embodiments, a population of synthetic nanocarriers may be heterogeneous with respect to size, shape, and/or composition.
  • Synthetic nanocarriers can be solid or hollow and can comprise one or more layers. In some embodiments, each layer has a unique composition and unique properties relative to the other layer(s).
  • synthetic nanocarriers may have a core/shell structure, wherein the core is one layer (e.g. a polymeric core) and the shell is a second layer (e.g. a lipid bilayer or monolayer). Synthetic nanocarriers may comprise a plurality of different layers.
  • a synthetic nanocarrier may, therefore, comprise a liposome.
  • a synthetic nanocarrier may comprise a lipid bilayer.
  • a synthetic nanocarrier may comprise a lipid monolayer.
  • a synthetic nanocarrier may comprise a micelle.
  • a synthetic nanocarrier may comprise a non-polymeric core (e.g., metal particle, quantum dot, ceramic particle, bone particle, viral particle, proteins, nucleic acids, carbohydrates, etc.) surrounded by a polymeric layer.
  • the polymers coupled to immunomodulatory agents can be combined with metal particles, quantum dots, ceramic particles, etc.
  • the polymers coupled to immunomodulatory agents can be combined with non-polymeric synthetic nanocarrier aggregates of non-polymeric components, such as an aggregate of metal atoms (e.g., gold atoms).
  • the synthetic nanocarriers comprising the polymers coupled to immunomodulatory agents may optionally comprise one or more amphiphilic entities.
  • an amphiphilic entity can promote the production of synthetic nanocarriers with increased stability, improved uniformity, or increased viscosity.
  • amphiphilic entities can be associated with the interior surface of a lipid membrane (e.g., lipid bilayer, lipid monolayer, etc.). Many amphiphilic entities known in the art are suitable for use in making synthetic nanocarriers in accordance with the present invention.
  • amphiphilic entities include, but are not limited to, phosphoglycerides; phosphatidylcholines; dipalmitoyl phosphatidylcholine (DPPC); dioleylphosphatidyl ethanolamine (DOPE); dioleyloxypropyltriethylammonium (DOTMA); dioleoylphosphatidylcholine; cholesterol; cholesterol ester; diacylglycerol; diacylglycerolsuccinate; diphosphatidyl glycerol (DPPG); hexanedecanol; fatty alcohols such as polyethylene glycol (PEG); polyoxyethylene-9-lauryl ether; a surface active fatty acid, such as palmitic acid or oleic acid; fatty acids; fatty acid monoglycerides; fatty acid diglycerides; fatty acid amides; sorbitan trioleate (Span®85) glycocholate; sorbitan monolaurate (Span®20); polysorbate 20
  • amphiphilic entity component may be a mixture of different amphiphilic entities. Those skilled in the art will recognize that this is an exemplary, not comprehensive, list of substances with surfactant activity. Any amphiphilic entity may be used in the production of synthetic nanocarriers to be used in accordance with the present invention.
  • the synthetic nanocarriers comprising the polymers coupled to immunomodulatory agents may optionally comprise one or more carbohydrates.
  • Carbohydrates may be natural or synthetic.
  • a carbohydrate may be a derivatized natural carbohydrate.
  • a carbohydrate comprises monosaccharide or disaccharide, including but not limited to glucose, fructose, galactose, ribose, lactose, sucrose, maltose, trehalose, cellbiose, mannose, xylose, arabinose, glucoronic acid, galactoronic acid, mannuronic acid, glucosamine, galatosamine, and neuramic acid.
  • a carbohydrate is a polysaccharide, including but not limited to pullulan, cellulose, microcrystalline cellulose, hydroxypropyl methylcellulose (HPMC), hydroxycellulose (HC), methylcellulose (MC), dextran, cyclodextran, glycogen, hydroxyethylstarch, carageenan, glycon, amylose, chitosan, N,O-carboxylmethylchitosan, algin and alginic acid, starch, chitin, inulin, konjac, glucommannan, pustulan, heparin, hyaluronic acid, curdlan, and xanthan.
  • the synthetic nanocarriers do not comprise (or specifically exclude) carbohydrates, such as a polysaccharide.
  • the carbohydrate may comprise a carbohydrate derivative such as a sugar alcohol, including but not limited to mannitol, sorbitol, xylitol, erythritol, maltitol, and lactitol.
  • elements can also be coupled to the synthetic nanocarriers, such as to polymers (that are coupled and/or not coupled to immunomodulatory agents) provided herein. Accordingly, the elements can be covalently associated with, for example, a polymeric matrix. In some embodiments, covalent association is mediated by a linker.
  • a component can be noncovalently associated with a polymeric matrix. For example, in some embodiments, a component can be encapsulated within, surrounded by, and/or dispersed throughout a polymeric matrix. Alternatively or additionally, a component can be associated with a polymeric matrix by hydrophobic interactions, charge interactions, van der Waals forces, etc.
  • the coupling can be a covalent linker.
  • components according to the invention can be covalently coupled to the external surface via a 1,2,3-triazole linker formed by the 1,3-dipolar cycloaddition reaction of azido groups on the surface of the nanocarrier with a component containing an alkyne group or by the 1,3-dipolar cycloaddition reaction of alkynes on the surface of the nanocarrier with components containing an azido group.
  • Such cycloaddition reactions are preferably performed in the presence of a Cu(I) catalyst along with a suitable Cu(I)-ligand and a reducing agent to reduce Cu(II) compound to catalytic active Cu(I) compound.
  • This Cu(I)-catalyzed azide-alkyne cycloaddition (CuAAC) can also be referred as the click reaction.
  • the covalent coupling may comprise a covalent linker that comprises an amide linker, a disulfide linker, a thioether linker, a hydrazone linker, a hydrazide linker, an imine or oxime linker, an urea or thiourea linker, an amidine linker, an amine linker, and a sulfonamide linker.
  • a covalent linker that comprises an amide linker, a disulfide linker, a thioether linker, a hydrazone linker, a hydrazide linker, an imine or oxime linker, an urea or thiourea linker, an amidine linker, an amine linker, and a sulfonamide linker.
  • An amide linker is formed via an amide bond between an amine on one component with the carboxylic acid group of a second component such as the nanocarrier.
  • the amide bond in the linker can be made using any of the conventional amide bond forming reactions with suitably protected amino acids and activated carboxylic acid such N-hydroxysuccinimide-activated ester.
  • a disulfide linker is made via the formation of a disulfide (S—S) bond between two sulfur atoms of the form, for instance, of R1-S—S—R2.
  • a disulfide bond can be formed by thiol exchange of a component containing thiol/mercaptan group (—SH) with another activated thiol group on a polymer or nanocarrier or a nanocarrier containing thiol/mercaptan groups with a component containing activated thiol group.
  • a triazole linker specifically a 1,2,3-triazole of the form
  • R1 and R2 may be any chemical entities, is made by the 1,3-dipolar cycloaddition reaction of an azide attached to a first component such as the nanocarrier with a terminal alkyne attached to a second component.
  • the 1,3-dipolar cycloaddition reaction is performed with or without a catalyst, preferably with Cu(I)-catalyst, which links the two components through a 1,2,3-triazole function.
  • This chemistry is described in detail by Sharpless et al., Angew. Chem. Int. Ed. 41(14), 2596, (2002) and Meldal, et al, Chem. Rev., 2008, 108(8), 2952-3015 and is often referred to as a “click” reaction or CuAAC.
  • a polymer containing an azide or alkyne group, terminal to the polymer chain is prepared.
  • This polymer is then used to prepare a synthetic nanocarrier in such a manner that a plurality of the alkyne or azide groups are positioned on the surface of that nanocarrier.
  • the synthetic nanocarrier can be prepared by another route, and subsequently functionalized with alkyne or azide groups.
  • the component is prepared with the presence of either an alkyne (if the polymer contains an azide) or an azide (if the polymer contains an alkyne) group.
  • the component is then allowed to react with the nanocarrier via the 1,3-dipolar cycloaddition reaction with or without a catalyst which covalently couples the component to the particle through the 1,4-disubstituted 1,2,3-triazole linker.
  • a thioether linker is made by the formation of a sulfur-carbon (thioether) bond in the form, for instance, of R1-S—R2.
  • Thioether can be made by either alkylation of a thiol/mercaptan (—SH) group on one component with an alkylating group such as halide or epoxide on a second component such as the nanocarrier.
  • Thioether linkers can also be formed by Michael addition of a thiol/mercaptan group on one component to an electron-deficient alkene group on a second component such as a polymer containing a maleimide group or vinyl sulfone group as the Michael acceptor.
  • thioether linkers can be prepared by the radical thiol-ene reaction of a thiol/mercaptan group on one component with an alkene group on a second component such as a polymer or nanocarrier.
  • a hydrazone linker is made by the reaction of a hydrazide group on one component with an aldehyde/ketone group on the second component such as the nanocarrier.
  • a hydrazide linker is formed by the reaction of a hydrazine group on one component with a carboxylic acid group on the second component such as the nanocarrier. Such reaction is generally performed using chemistry similar to the formation of amide bond where the carboxylic acid is activated with an activating reagent.
  • An imine or oxime linker is formed by the reaction of an amine or N-alkoxyamine (or aminooxy) group on one component an aldehyde or ketone group on the second component such as the nanocarrier.
  • An urea or thiourea linker is prepared by the reaction of an amine group on one component with an isocyanate or thioisocyanate group on the second component such as the nanocarrier.
  • An amidine linker is prepared by the reaction of an amine group on one component with an imidoester group on the second component such as the nanocarrier.
  • An amine linker is made by the alkylation reaction of an amine group on one component with an alkylating group such as halide, epoxide, or sulfonate ester group on the second component such as the nanocarrier.
  • an amine linker can also be made by reductive amination of an amine group on one component with an aldehyde or ketone group on the second component such as the nanocarrier with a suitable reducing reagent such as sodium cyanoborohydride or sodium triacetoxyborohydride.
  • a sulfonamide linker is made by the reaction of an amine group on one component with a sulfonyl halide (such as sulfonyl chloride) group on the second component such as the nanocarrier.
  • a sulfonyl halide such as sulfonyl chloride
  • a sulfone linker is made by Michael addition of a nucleophile to a vinyl sulfone.
  • Either the vinyl sulfone or the nucleophile may be on the surface of the nanocarrier or attached to the component.
  • the components can also be conjugated to the nanocarrier via non-covalent conjugation methods.
  • a negative charged component can be conjugated to a positive charged nanocarrier through electrostatic adsorption.
  • a component containing a metal ligand can also be conjugated to a nanocarrier containing a metal complex via a metal-ligand complex.
  • the component can be attached to a polymer, for example polylactic acid-block-polyethylene glycol, prior to the assembly of the synthetic nanocarrier or the synthetic nanocarrier can be formed with reactive or activatable groups on its surface.
  • the component may be prepared with a group which is compatible with the attachment chemistry that is presented by the synthetic nanocarriers' surface.
  • a component can be attached to VLPs or liposomes using a suitable linker.
  • a linker is a compound or reagent that capable of coupling two molecules together.
  • the linker can be a homobifuntional or heterobifunctional reagent as described in Hermanson 2008.
  • an VLP or liposome synthetic nanocarrier containing a carboxylic group on the surface can be treated with a homobifunctional linker, adipic dihydrazide (ADH), in the presence of EDC to form the corresponding synthetic nanocarrier with the ADH linker.
  • ADH adipic dihydrazide
  • the resulting ADH linked synthetic nanocarrier is then conjugated with a peptide containing an acid group via the other end of the ADH linker on NC to produce the corresponding VLP or liposome peptide conjugate.
  • the component can be coupled by adsorption to a pre-formed synthetic nanocarrier or it can be coupled by encapsulation during the formation of the synthetic nanocarrier.
  • a component such as an antigen or immunomodulatory agent
  • Isolated refers to the element being separated from its native environment and present in sufficient quantities to permit its identification or use. This means, for example, the element may be (i) selectively produced by expression cloning or (ii) purified as by chromatography or electrophoresis. Isolated elements may be, but need not be, substantially pure. Because an isolated element may be admixed with a pharmaceutically acceptable excipient in a pharmaceutical preparation, the element may comprise only a small percentage by weight of the preparation. The element is nonetheless isolated in that it has been separated from the substances with which it may be associated in living systems, i.e., isolated from other lipids or proteins. Any of the elements provided herein may be isolated. Any of the antigens provided herein can be included in the compositions in isolated form.
  • Synthetic nanocarriers may be prepared using a wide variety of methods known in the art.
  • synthetic nanocarriers can be formed by methods as nanoprecipitation, flow focusing fluidic channels, spray drying, single and double emulsion solvent evaporation, solvent extraction, phase separation, milling, microemulsion procedures, microfabrication, nanofabrication, sacrificial layers, simple and complex coacervation, and other methods well known to those of ordinary skill in the art.
  • aqueous and organic solvent syntheses for monodisperse semiconductor, conductive, magnetic, organic, and other nanomaterials have been described (Pellegrino et al., 2005, Small, 1:48; Murray et al., 2000, Ann Rev. Mat.
  • Various materials may be encapsulated into synthetic nanocarriers as desirable using a variety of methods including but not limited to C. Astete et al., “Synthesis and characterization of PLGA nanoparticles” J. Biomater. Sci. Polymer Edn, Vol. 17, No. 3, pp. 247-289 (2006); K. Avgoustakis “Pegylated Poly(Lactide) and Poly(Lactide-Co-Glycolide) Nanoparticles: Preparation, Properties and Possible Applications in Drug Delivery” Current Drug Delivery 1:321-333 (2004); C. Reis et al., “Nanoencapsulation I. Methods for preparation of drug-loaded polymeric nanoparticles” Nanomedicine 2:8-21 (2006); P.
  • synthetic nanocarriers are prepared by a nanoprecipitation process or spray drying. Conditions used in preparing synthetic nanocarriers may be altered to yield particles of a desired size or property (e.g., hydrophobicity, hydrophilicity, external morphology, “stickiness,” shape, etc.). The method of preparing the synthetic nanocarriers and the conditions (e.g., solvent, temperature, concentration, air flow rate, etc.) used may depend on the materials to be coupled to the synthetic nanocarriers and/or the composition of the polymer matrix.
  • Conditions used in preparing synthetic nanocarriers may be altered to yield particles of a desired size or property (e.g., hydrophobicity, hydrophilicity, external morphology, “stickiness,” shape, etc.).
  • the method of preparing the synthetic nanocarriers and the conditions (e.g., solvent, temperature, concentration, air flow rate, etc.) used may depend on the materials to be coupled to the synthetic nanocarriers and/or the composition of the polymer matrix.
  • particles prepared by any of the above methods have a size range outside of the desired range, particles can be sized, for example, using a sieve.
  • Elements (components) of the synthetic nanocarriers may be coupled to the overall synthetic nanocarrier, e.g., by one or more covalent bonds, or may be coupled by means of one or more linkers. Additional methods of functionalizing synthetic nanocarriers may be adapted from Published US Patent Application 2006/0002852 to Saltzman et al., Published US Patent Application 2009/0028910 to DeSimone et al., or Published International Patent Application WO/2008/127532 A1 to Murthy et al.
  • synthetic nanocarriers can be coupled to other elements directly or indirectly via non-covalent interactions.
  • the non-covalent coupling is mediated by non-covalent interactions including but not limited to charge interactions, affinity interactions, metal coordination, physical adsorption, host-guest interactions, hydrophobic interactions, TT stacking interactions, hydrogen bonding interactions, van der Waals interactions, magnetic interactions, electrostatic interactions, dipole-dipole interactions, and/or combinations thereof.
  • Such couplings may be arranged to be on an external surface or an internal surface of an synthetic nanocarrier.
  • encapsulation and/or absorption is a form of coupling.
  • the synthetic nanocarriers can be combined with other immunomodulatory agents or moieties thereof and/or one or more antigens by admixing in the same vehicle or delivery system.
  • immunomodulatory agents or moieties thereof may include adjuvants that include, but are not limited to mineral salts, such as alum, alum combined with monphosphoryl lipid (MPL) A of Enterobacteria, such as Escherihia coli, Salmonella minnesota, Salmonella typhimurium , or Shigella flexneri or specifically with MPL® (AS04), MPL A of above-mentioned bacteria separately, saponins, such as QS-21, Quil-A, ISCOMs, ISCOMATRIXTM, emulsions such as MF59TM, Montanide® ISA 51 and ISA 720, AS02 (QS21+squalene+MPL®), liposomes and liposomal formulations such as AS01, synthesized or specifically prepared microparticles
  • gonorrheae Chlamydia trachomatis and others, or chitosan particles
  • depot-forming agents such as Pluronic® block co-polymers, specifically modified or prepared peptides, such as muramyl dipeptide, aminoalkyl glucosaminide 4-phosphates, such as RC529, or proteins, such as bacterial toxoids or toxin fragments.
  • the doses of such other immunomodulatory agents or moieies thereof can be determined using conventional dose ranging studies.
  • the synthetic nanocarriers can be combined with an antigen different, similar or identical to any coupled to a nanocarrier (with or without immunomodulatory agent, utilizing or not utilizing another delivery vehicle) administered separately at a different time-point and/or at a different body location and/or by a different immunization route or with another antigen and/or immunomodulatory agent-carrying synthetic nanocarrier administered separately at a different time-point and/or at a different body location and/or by a different immunization route.
  • compositions for use in the methods according to the invention comprise synthetic nanocarriers in combination with pharmaceutically acceptable excipients, such as preservatives, buffers, saline, or phosphate buffered saline.
  • pharmaceutically acceptable excipients such as preservatives, buffers, saline, or phosphate buffered saline.
  • the compositions may be made using conventional pharmaceutical manufacturing and compounding techniques to arrive at useful dosage forms.
  • synthetic nanocarriers are suspended in sterile saline solution for injection together with a preservative.
  • the component when preparing synthetic nanocarriers as carriers for use in vaccines, methods for coupling to the synthetic nanocarriers may be useful. If the component is a small molecule it may be of advantage to attach the component to a polymer prior to the assembly of the synthetic nanocarriers. In embodiments, it may also be an advantage to prepare the synthetic nanocarriers with surface groups that are used to couple the component to the synthetic nanocarrier through the use of these surface groups rather than attaching the component to a polymer and then using this polymer conjugate in the construction of synthetic nanocarriers.
  • Synthetic nanocarriers may be combined to form pharmaceutical dosage forms according to the present invention using traditional pharmaceutical mixing methods. These include liquid-liquid mixing in which two or more suspensions, each containing one or more subsets of nanocarriers, are directly combined or are brought together via one or more vessels containing diluent. As synthetic nanocarriers may also be produced or stored in a powder form, dry powder-powder mixing could be performed as could the re-suspension of two or more powders in a common media. Depending on the properties of the nanocarriers and their interaction potentials, there may be advantages conferred to one or another route of mixing.
  • compositions that comprise synthetic nanocarriers may comprise inorganic or organic buffers (e.g., sodium or potassium salts of phosphate, carbonate, acetate, or citrate) and pH adjustment agents (e.g., hydrochloric acid, sodium or potassium hydroxide, salts of citrate or acetate, amino acids and their salts) antioxidants (e.g., ascorbic acid, alpha-tocopherol), surfactants (e.g., polysorbate 20, polysorbate 80, polyoxyethylene9-10 nonyl phenol, sodium desoxycholate), solution and/or cryo/lyo stabilizers (e.g., sucrose, lactose, mannitol, trehalose), osmotic adjustment agents (e.g., salts or sugars), antibacterial agents (e.g., benzoic acid, phenol, gentamicin), antifoaming agents (e.g., polydimethylsilozone), preservatives (e.g., thoxy
  • compositions according to the invention comprise synthetic nanocarriers in combination with pharmaceutically acceptable excipients.
  • the compositions may be made using conventional pharmaceutical manufacturing and compounding techniques to arrive at useful dosage forms. Techniques suitable for use in practicing the present invention may be found in Handbook of Industrial Mixing: Science and Practice, Edited by Edward L. Paul, Victor A. Atiemo-Obeng, and Suzanne M. Kresta, 2004 John Wiley & Sons, Inc.; and Pharmaceutics: The Science of Dosage Form Design, 2nd Ed. Edited by M. E. Auten, 2001, Churchill Livingstone.
  • synthetic nanocarriers are suspended in sterile saline solution for injection together with a preservative.
  • compositions of synthetic nanocarriers can be made in any suitable manner, and the invention is in no way limited to the use of compositions that can be produced using the methods described herein. Selection of an appropriate method may require attention to the properties of the particular moieties being associated.
  • synthetic nanocarriers are manufactured under sterile conditions or are terminally sterilized. This can ensure that resulting compositions are sterile and non-infectious, thus improving safety when compared to non-sterile compositions. This provides a valuable safety measure, especially when subjects receiving synthetic nanocarriers have immune defects, are suffering from infection, and/or are susceptible to infection.
  • synthetic nanocarriers may be lyophilized and stored in suspension or as lyophilized powder depending on the formulation strategy for extended periods without losing activity.
  • compositions of the invention can be administered by a variety of routes, including or not limited to subcutaneous, intranasal, oral, intravenous, intraperitoneal, intramuscular, transmucosal, transmucosal, sublingual, rectal, ophthalmic, pulmonary, intradermal, transdermal, transcutaneous or intradermal or by a combination of these routes.
  • Routes of administration also include administration by inhalation or pulmonary aerosol. Techniques for preparing aerosol delivery systems are well known to those of skill in the art (see, for example, Sciarra and Cutie, “Aerosols,” in Remington's Pharmaceutical Sciences, 18th edition, 1990, pp. 1694-1712; incorporated by reference).
  • Doses of dosage forms contain varying amounts of populations of synthetic nanocarriers and varying amounts of components, such as immunomodulatory agents and/or antigens, according to the invention.
  • the amount of synthetic nanocarriers, immunomodulatory agents and/or antigens present in the dosage forms can be varied according to the nature of the immunomodulatory agents and/or antigens, the therapeutic benefit to be accomplished, and other such parameters.
  • dose ranging studies can be conducted to establish optimal therapeutic amount of the population of synthetic nanocarriers and the amount of immunomodulatory agents and/or antigens to be present in the dosage form.
  • the synthetic nanocarriers and the immunomodulatory agents and/or antigens are present in the dosage form in an amount effective to generate an immune response to the antigens upon administration to a subject. It may be possible to determine amounts of the immunomodulatory agents and/or antigens effective to generate an immune response using conventional dose ranging studies and techniques in subjects. Dosage forms may be administered at a variety of frequencies. In a preferred embodiment, at least one administration of the dosage form is sufficient to generate a pharmacologically relevant response. In more preferred embodiment, at least two administrations, at least three administrations, or at least four administrations, of the dosage form are utilized to ensure a pharmacologically relevant response.
  • compositions and methods described herein can be used to induce, enhance, suppress, modulate, direct, or redirect an immune response.
  • the compositions and methods described herein can be used in the diagnosis, prophylaxis and/or treatment of conditions such as cancers, infectious diseases, metabolic diseases, degenerative diseases, non-autoimmune diseases or other disorders and/or conditions.
  • the compositions and methods described herein can also be used for the prophylaxis or treatment of an addiction, such as an addiction to an illegal drug, an over-the-counter drug, a prescription drug.
  • the addiction is to cocaine, heroin, marijuana, methamphetamines, nicotine or a narcotic.
  • the compositions and methods described herein can also be used for the prophylaxis and/or treatment of a condition resulting from the exposure to a toxin, hazardous substance, environmental toxin, or other harmful agent.
  • infectious disease examples include, but are not limited to, viral infectious diseases, such as AIDS, Chickenpox (Varicella), Common cold, Cytomegalovirus Infection, Colorado tick fever, Dengue fever, Ebola hemorrhagic fever, Hand, foot and mouth disease, Hepatitis, Herpes simplex, Herpes zoster, HPV, Influenza (Flu), Lassa fever, Measles, Marburg hemorrhagic fever, Infectious mononucleosis, Mumps, Norovirus, Poliomyelitis, Progressive multifocal leukencephalopathy, Rabies, Rubella, SARS, Smallpox (Variola), Viral encephalitis, Viral gastroenteritis, Viral meningitis, Viral pneumonia, West Nile disease and Yellow fever; bacterial infectious diseases, such as Anthrax, Bacterial Meningitis, Botulism, Brucellosis, Campylobacteriosis, Cat Scratch Disease, Cholera, Diphth
  • cancers include, but are not limited to breast cancer; biliary tract cancer; bladder cancer; brain cancer including glioblastomas and medulloblastomas; cervical cancer; choriocarcinoma; colon cancer; endometrial cancer; esophageal cancer; gastric cancer; hematological neoplasms including acute lymphocytic and myelogenous leukemia, e.g., B Cell CLL; T-cell acute lymphoblastic leukemia/lymphoma; hairy cell leukemia; chronic myelogenous leukemia, multiple myeloma; AIDS-associated leukemias and adult T-cell leukemia/lymphoma; intraepithelial neoplasms including Bowen's disease and Paget's disease; liver cancer; lung cancer; lymphomas including Hodgkin's disease and lymphocytic lymphomas; neuroblastomas; oral cancer including squamous cell carcinoma; ovarian cancer including those arising from epit
  • metabolic diseases include, but are not limited to, disorders of carbohydrate metabolism, amino acid metabolism, organic acid metabolism, fatty acid oxidation and mitochondrial metabolism, prophyrin metabolism, purine or pyrimidine metabolism, steroid metabolism, lysosomal mitochondrial function, peroxisomal function, lysosomal storage, urea cycle disorders (e.g., N-acetyl glutamate synthetase deficiency, carbamylphosphate synthase deficiency, ornithine carbamyl transferase deficiency, crginosuccinic aciduria, citrullinaemia, arginase deficiency), amino acid disorders (e.g., Non-ketotic hyperglycinaemia, tyrosinaemia (Type I), Maple syrup urine disease), organic acidemias (e.g, isovaleric acidemia, methylmalonic acidemia, propionic acidemia, glutaric aciduria type I, glutaric acidemia type I & II),
  • degenerative diseases include, but are not limited to, mesenchyme/mesoderm degenerative disease, muscle degenerative disease, endothelial degenerative disease, neurodegenerative disease, degenerative joint disease (e.g., osteoarthritis), major types of degenerative heart disease (e.g., coronary heart disease, congenital heart disease, rheumatic heart disease, angina pectoris), neurodegenerative disease (e.g., Alzheimer's disease, amyotrophic lateral sclerosis, Friedreich's ataxia, Huntington's disease, Lewy body disease, Parkinson's disease, spinal muscular atrophy), neuromuscular disorders (e.g., muscular dystrophy, duchenne muscular dystrophy, facioscapulohumeral muscular dystrophy, myotonic muscular dystrophy, congenital myopathy, familial cardiomyopathy, dilated cardiomyopathy, hypertrophic cardiomyopathy, restrictive cardiomyopathy, or coronary artery disease).
  • degenerative heart disease e.g., coronary heart disease,
  • compositions and methods described herein can be used in the diagnosis, prophylaxis and/or treatment of diseases, disorders or conditions in which immune suppression or tolerance would confer a treatment benefit.
  • diseases, disorders or conditions include autoimmune diseases, inflammatory diseases, allergies or graft versus host disease.
  • the compositions and methods described herein can also be used in subjects who have undergone or will undergo transplantation.
  • the compositions and methods described herein can also be used in subject who have undergone, are undergoing or will undergo treatment with a therapeutic agent against which agent an undesired immune response occurs or may occur.
  • Autoimmune disease include, but are not limited to, rheumatoid arthritis, multiple sclerosis, immune-mediated or Type I diabetes mellitus, inflammatory bowel disease (e.g., Crohn's disease or ulcerative colitis), systemic lupus erythematosus, psoriasis, scleroderma, autoimmune thyroid disease, alopecia greata, Grave's disease, Guillain-Barré syndrome, celiac disease, Sjögren's syndrome, rheumatic fever, gastritis, autoimmune atrophic gastritis, autoimmune hepatitis, insulitis, oophoritis, orchitis, uveitis, phacogenic uveitis, myasthenia gravis, primary myxoedema, pernicious anemia, autoimmune haemolytic anemia, Addison's disease, scleroderma, Goodpasture's syndrome, neph
  • Inflammatory diseases include, but are not limited to, Alzheimer's, Ankylosing spondylitis, arthritis, asthma, atherosclerosis, Behcet's disease, chronic inflammatory demyelinating polyradiculoneuropathy, Crohn's disease, colitis, cystic fibrosis, dermatitis, diverticulitis, hepatitis, irritable bowel syndrome (IBS), lupus erythematous, muscular dystrophy, nephritis, Parkinson's, shingles and ulcerative colitis.
  • IBS irritable bowel syndrome
  • Inflammatory diseases also include, for example, cardiovascular disease, chronic obstructive pulmonary disease (COPD), bronchiectasis, chronic cholecystitis, tuberculosis, Hashimoto's thyroiditis, sepsis, sarcoidosis, silicosis and other pneumoconioses, and an implanted foreign body in a wound, but are not so limited.
  • COPD chronic obstructive pulmonary disease
  • bronchiectasis chronic cholecystitis
  • tuberculosis Hashimoto's thyroiditis
  • sepsis sepsis
  • sarcoidosis silicosis and other pneumoconioses
  • an implanted foreign body in a wound but are not so limited.
  • the term “sepsis” refers to a well-recognized clinical syndrome associated with a host's systemic inflammatory response to microbial invasion.
  • fever refers to a condition that is typically signaled by fever or hypothermia, tachycardia, and tachypnea, and in severe instances can progress to hypotension, organ dysfunction, and even death.
  • the inflammatory disease is non-autoimmune inflammatory bowel disease, post-surgical adhesions, coronary artery disease, hepatic fibrosis, acute respiratory distress syndrome, acute inflammatory pancreatitis, endoscopic retrograde cholangiopancreatography-induced pancreatitis, burns, atherogenesis of coronary, cerebral and peripheral arteries, appendicitis, cholecystitis, diverticulitis, visceral fibrotic disorders, wound healing, skin scarring disorders (keloids, hidradenitis suppurativa), granulomatous disorders (sarcoidosis, primary biliary cirrhosis), asthma, pyoderma gandrenosum, Sweet's syndrome, Behcet's disease, primary sclerosing cholangitis or an abscess.
  • the inflammatory disease is inflammatory bowel disease (e.g., Crohn's disease or ulcerative colitis).
  • the inflammatory disease is an autoimmune disease.
  • the autoimmune disease in some embodiments is rheumatoid arthritis, rheumatic fever, ulcerative colitis, Crohn's disease, autoimmune inflammatory bowel disease, insulin-dependent diabetes mellitus, diabetes mellitus, juvenile diabetes, spontaneous autoimmune diabetes, gastritis, autoimmune atrophic gastritis, autoimmune hepatitis, thyroiditis, Hashimoto's thyroiditis, insulitis, oophoritis, orchitis, uveitis, phacogenic uveitis, multiple sclerosis, myasthenia gravis, primary myxoedema, thyrotoxicosis, pernicious anemia, autoimmune haemolytic anemia, Addison's disease, Anklosing spondylitis, sarcoidosis, scleroderma, Goodpasture's syndrome, Guillain-Barre syndrome, Graves' disease, glomerular
  • the inflammatory disease is a condition in which a subject experiences pain due to inflammation.
  • the pain is injury-induced pain.
  • the pain is cancer-induced.
  • a subject that is treated with a composition of the invention is one that has or has had an injury or cancer and has experienced, is experiencing or will experience pain that is or is thought to be associated with the injury or cancer.
  • GVHD graft versus host disease
  • aGVHD The acute or fulminant form of the disease
  • cGVHD The chronic form of graft-versus-host-disease (cGVHD) normally occurs after 100 days. The appearance of moderate to severe cases of cGVHD adversely influences long-term survival.
  • Step-2 The polymer from Step-1 (9 g) was combined with glycolide (2.43 g, 0.021 mol) and anhydrous sodium sulfate (10 g) in 200 mL of dry toluene. The mixture was heated to reflux while 30 mL of toluene was distilled out. Sn(Oct) 2 (0.20 mL) was then added and the resulting mixture was heated at 120° C. under argon overnight. After cooling, the toluene solution was decanted from solid sodium sulfate and concentrated to dryness. The residue was then dissolved in 200 mL of dichloromethane (DCM) and the resulting solution was washed with 100 mL of water.
  • DCM dichloromethane
  • TBD 1,5,7-triazabicyclo[4.4.0]dec-5-ene
  • the solution was diluted with 100 mL of CHCl 3 and washed with 10% tartaric acid (100 mL). After drying, the organic solution was filtered and concentrated under vacuum to ca. 25 mL. This solution was then added to 300 mL of diethyl ether to precipitate out the polymer. The polymer was then dried under high vacuum give the dendrimeric polymer-R848 conjugate as a white solid (4.2 g, 64%, 1 H NMR showed the R848 content at 18% wt).
  • Step-1 A solution of dipentaerythritol (1.27 g, 5.0 mmol) and 6-caprolactone (25 g, 219 mmol) in 250 mL of dry toluene is heated to reflux while ca 50 mL of toluene is removed. Sn(Oct) 2 (0.25 mL) is then added. The resulting solution is refluxed under argon overnight. After cooling, the toluene solution is concentrated to ca. 50 mL in volume and added to 500 mL of 2-propanol (IPA) to precipitate out the polymer. The polymer is then washed with 100 mL of IPA followed by 100 mL of t-butylmethyl ether (MTBE). The polymer is then dried under high vacuum to give 6-arm dendrimeric polycaprolactone (PCL) (ca. 25 g as white solid).
  • PCL 6-arm dendrimeric polycaprolactone
  • Step-2 The 6-arm PCL from Step-1 (25 g, ca. 5 mmol), succinic anhydride (12.0 g, 120 mmol) and 9.6 mL of pyridine (120 mmol) in 200 mL of chloroform are refluxed with argon overnight. After adding 100 mL of chloroform, the mixture is washed successively with 100 mL each of dilute HCl, saturated NaCl, and water. The organic layer is dried over magnesium sulfate, and then the volume of the mixture is reduced by half using rotary evaporation. After pouring the mixture into 800 mL of a 1:1 mixture of hexane and diethyl ether, the polymer is precipitated overnight at 4° C. The polymer is collected and dried under vacuum to yield ca. 26 g of 6-arm PCL-succinic acid monoester.
  • Step-3 A mixture of the 6-arm PLC-succinic acid monoester from Step-2 (5.5 g, ca. 1.0 mmol), TBTU coupling agent (2.9 g, 9 mmol) in dry THF (100 mL) is stirred at rt under for 30 min. R848 (2.82 g, 9 mmol) is then added, followed by DIPEA (3.2 mL, 18 mmol). The resulting mixture is heated at ca. 50-60 C under argon overnight. The mixture is then diluted with EtOAc (300 mL) and washed with water, saturated NH 4 Cl and NaCl solution. After drying over Na 2 SO 4 , the solution is filtered and concentrated to ca 30 mL in volume.
  • Step-1 PLGA-COOH (3.0 g, 0.17 mmol) in anhydrous methylene chloride (15 mL) is converted to PLGA-NHS with excess N-hydroxysuccinimide (NHS, 76 mg, 0.66 mmol, 4 equiv) in the presence of 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide (EDC, 140 mg, 0.72 mmol, 4.3 equiv) by magnetically stirring at room temperature for 12 h under nitrogen atmosphere.
  • NHS N-hydroxysuccinimide
  • EDC 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide
  • PLGA NHS ester (“PLGA-NHS”) is precipitated with cold diethyl ether (20 mL), filtered, repeatedly washed in a cold mixture of diethyl ether and methanol (few drops), and dried with nitrogen and under vacuum to remove solvent (yield: 97%).
  • Step-2 PLGA-NHS (3.0 g, 0.17 mmol) is dissolved in anhydrous DMSO (10 mL) followed by addition of N,N-bis(carboxymethyl)-L-lysine hydrate (NTA-L-lysine) (60 mg, 0.20 mmol, 1.2 equiv) and N,N-diisopropylethylamine (DIPEA) (42 mg, 0.33 mmol, 3.8 equiv), and the reaction mixture is stirred at room temperature for 24 h.
  • the PLGA-NTA-L-lysine is precipitated with cold diethyl ether and dried under vacuum as white powder (2.7 g, 91%).
  • Step-3 PLGA-NTA (2.5 g, 0.14 mmol) and TBTU coupling agent (0.21 g, 0.63 mmol) in dry THF (50 mL) is stirred at rt under for 30 min.
  • the TLR7 agonist, 9-benzyl-2-butoxy-8-hydroxyadenine (0.20 g, 0.63 mmol) is then added, followed by DIPEA (0.21 mL, 1.2 mmol).
  • the resulting mixture is heated at ca. 50-60° C. under argon overnight.
  • the mixture is then diluted with EtOAc (200 mL) and washed with water, saturated NH 4 Cl and NaCl solution.
  • the solution is filtered and concentrated to ca 10 mL in volume.
  • the solution is then added to 200 mL of IPA to precipitate out the polymer which is then washed with IPA and MTBE (100 mL each).
  • the conjugated polymer is then dried under high vacuum to give a light brown solid (2.5 g).
  • Polylactide with amino end group (PLA-NH 2 ) is prepared by DBU (1,8-diazabicyclo[5.4.0]undec-7-ene) catalyzed ring opening polymerization of d1-lactide with 2-BocNH-ethanol followed by deprotection with TFA in DCM.
  • Bz-Glu-NCA Polymerization of Bz-Glu-NCA is performed by mixing a solution of 0.4 g of NH2-PLA (0.0713 mmol) in 2 mL anhydrous dimethylformamide (DMF) with a solution of 0.66 g (0.0025 mol) freshly prepared Bz-Glu-NCA in 7.5 mL anhydrous DMF. The reaction mixture is stirred under argon at 40° C. for 48 h. The reaction mixture is then poured into a large excess of diethyl ether. The precipitate is collected by centrifugation, washed with diethyl ether, and dried in a vacuum to yield poly(benzyl-L-glutamate)-block-PLA (ca. 0.8 g).
  • DMF dimethylformamide
  • poly(benzyl-L-glutamate)-block-PLA (0.5 g) is dissolved in 6 mL trifluoroacetic acid (TFA) under argon at 0° C., followed by the addition of 0.6 mL of trifluoromethanesulfonic acid (TFMSA) and 0.7 mL of thioanisole.
  • TFA trifluoroacetic acid
  • TFMSA trifluoromethanesulfonic acid
  • thioanisole 0.6 mL of trifluoromethanesulfonic acid
  • the reaction mixture is gently stirred under argon at 0° C. for 1 h and then at room temperature for 30 min.
  • the reaction mixture is poured into cooled diethyl ether.
  • the resulting white precipitate is collected by filtration and dried in a vacuum to yield 0.4 g of Poly(L-glutamic acid)-block-PLA.
  • Poly(L-glutamic acid)-block-PLA is then conjugated with R848 in the presence of TBTU/DIPEA as described above to give Poly(L-glutamic R848 amide)-block-PLA after workup and precipitation from ether and drying under vacuum.
  • Poly-L-malic acid is prepared by lipase catalyzed ring opening polymerization (ROP) of benzyl beta-malolactonate, followed by Pd-catalyzed debenzylation as described in Cameron et al., Chem. Soc. Rev. (2011) 40:1761-1776. See also Patil et al., Pharm Res (2010) 27:2317-2329.
  • ROP ring opening polymerization
  • N-Hydroxysuccinimide (NHS) (1 mmol) and N,N′-dicyclohexylcarbodiimide DCC (1 mmol) dissolved in 2 ml of DMF are added consecutively to the solution of Poly-L-malic acid (1 mmol with regard to malyl units) dissolved in 1 ml of anhydrous acetone under vigorous stirring at room temperature (RT).
  • RT room temperature
  • the TLR7 agonist 1.2 mmol of 9-aminobutyl-2-butoxy-8-hydroxyadenine in 0.5 ml of DMF is added followed by 2.4 mmol of triethylamine (TEA).
  • TAA triethylamine
  • Step-2 Preparation of polycarbonate-R848 conjugate by ROP of R848 containing cyclic carbonate monomer
  • ROP of cyclic carbonate can be performed in the presence of organic catalyst such as guanidines (TBD or MTBD), amidines (DBU), N-heterocyclic carbenes (NHCs), and bifunctional amino-thioureas.
  • organic catalyst such as guanidines (TBD or MTBD), amidines (DBU), N-heterocyclic carbenes (NHCs), and bifunctional amino-thioureas.
  • TBD guanidines
  • DBU amidines
  • NHS N-heterocyclic carbenes
  • bifunctional amino-thioureas bifunctional amino-thioureas.
  • the cyclic carbonate from Step-1 is treated with an initiator such as an organic alcohol (e.g., wherein R is substituted or unsubstituted alkyl, such as benzyl alcohol) in the presence of TBD to give the polycarbonate-R848 conjugate.
  • an organic alcohol e.g., wherein R is substituted or
  • immunomodulatory agents comprising a free primary amine may be used or synthetically modified following any of the aforementioned Examples to provide compositions of the present invention.
  • immunomodulatory agent moieties are provided below. Further examples of immunomodulatory agent moieties, such as adenine analogs, can also be found in the literature (see, e.g., J Med. Chem. 2008 Nov. 13; 51(21):6621-6.)
  • polymers can be prepared and conjugated with the multiple immunomodulatory agents (e.g., adjuvants) to give the corresponding polymer-immunomodulatory agent conjugates.
  • polymers can be functionalized to provide reactive groups or linkers on the polymers for conjugation with suitably derivatized immunomodulatory agents.
  • the reactive groups or linkers are carboxylic acid, aldehyde, ketone, alcohol, amine, alkene, azide and thiol groups.
  • functionalized monomers can be polymerized and then converted to polymers with reactive groups or linkers for conjugation with suitably derivatized immunomodulatory agents.
  • Adjuvant containing monomers such as lactide, cyclic lactone and cyclic carbonate can be prepared by reaction of suitable immunomodulatory agents (e.g., adjuvants) such as imidazoquinolines or adenine derivatives with functionalized monomers.
  • suitable immunomodulatory agents e.g., adjuvants
  • the resulting monomers can then be polymerized under standard ring-opening polymerization (ROP) conditions to give polymer-multi-immunomodulatory agent conjugates.
  • Step-1 PLGA-COOH (3.0 g, 0.17 mmol) in anhydrous methylene chloride (15 mL) is converted to PLGA-NHS with excess N-hydroxysuccinimide (NHS, 76 mg, 0.66 mmol, 4 equiv) in the presence of 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide (EDC, 140 mg, 0.72 mmol, 4.3 equiv) by magnetically stirring at room temperature for 12 h under nitrogen atmosphere.
  • NHS N-hydroxysuccinimide
  • EDC 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide
  • PLGA NHS ester (“PLGA-NHS”) is precipitated with cold diethyl ether (20 mL), filtered, repeatedly washed in a cold mixture of diethyl ether and methanol (few drops), and dried with nitrogen and under vacuum to remove solvent (yield: 97%).
  • Step 2 PLGA-NHS (3.0 g, 0.17 mmol) is dissolved in anhydrous DMSO (10 mL) followed by addition of N,N-bis(carboxymethyl)-L-lysine hydrate (NTA-L-lysine) (60 mg, 0.20 mmol, 1.2 equiv) and N,N-diisopropylethylamine (DIPEA) (42 mg, 0.33 mmol, 3.8 equiv), and the reaction mixture is stirred at room temperature for 24 h.
  • the PLGA-NTA-L-lysine is precipitated with cold diethyl ether and dried under vacuum as white powder (2.7 g, 91%).
  • Step 3 PLGA-NTA (2.5 g, 0.14 mmol) and DCC coupling agent (0.13 g, 0.63 mmol) in dry DMF (10 mL) is stirred at rt under for 30 min Rapamycin (0.58 g, 0.63 mmol) is then added, followed by 4-dimethylaminopyridine (DMAP) (0.08 g, 0.63 mmol). The mixture is stirred at rt for 2 days. The mixture is then filtered to remove insoluble dicyclohexylurea. The filtrate is then added to 100 mL of isopropyl alcohol (IPA) to precipitate out the PLGA-(rapamycin) 3 conjugate.
  • IPA isopropyl alcohol
  • IPA layer is removed and the polymer is then washed with 50 mL of IPA and 50 mL of methyl t-butyl ether (MTBE).
  • MTBE methyl t-butyl ether
  • the polymer is then dried under vacuum at 35 C for 2 days to give PLGA-(rapamycin) 3 conjugate (ca. 2.5 g).
  • Ovalbumin peptide 323-339 amide acetate salt is purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance Calif. 90505. Product code 4065609.) PLGA with 76% lactide and 24% glycolide content and an inherent viscosity of 0.49 dL/g is purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211.
  • PLA-PEG-Nicotine, poly-D/L lactide-block-poly(ethylene glycol)-( ⁇ )-trans-3′-hydroxymethylnicotine ether with PEG block of approximately 5,000 Da and PLA block of approximately 21,000 Da is custom manufactured at Princeton Global Synthesis (300 George Patterson Drive #206, Bristol, Pa. 19007.)
  • Dendrimeric (4-arm) PLGA-R848 conjugate of Example 1 is made having approximate molecular weight of 7,000 g/mol and 18% R848 loading on a weight/weight basis.
  • Polyvinyl alcohol PhEur, USP (85-89% hydrolyzed, viscosity of 3.4-4.6 mPa ⁇ s) is purchased from EMD Chemicals Inc. (480 South Democrat Road Gibbstown, N.J. 08027. Part Number 4-88).
  • Ovalbumin peptide 323-339 amide acetate salt at 20 mg/mL is prepared by dissolution in 0.13N hydrochloric acid at room temperature.
  • Solution 2 PLGA-R848 at 50 mg/mL, PLGA at 25 mg/mL, and PLA-PEG-Nicotine at 25 mg/mL in dichloromethane is prepared by creating individual solutions of each polymer in dichloromethane at 100 mg/mL, and then combining portions of those solutions in a 2:1:1 volume ratio of the PLGA-R848:PLGA:PLA-PEG-Nicotine.
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM in 100 mM phosphate buffer, pH 8.
  • Solution 4 70 mM phosphate buffer, pH 8.
  • a primary (W1/O) emulsion is first created using Solution 1 & Solution 2.
  • Solution 1 (0.2 mL) and Solution 2 (1.0 mL) are combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/O/W2) emulsion is then formed by adding Solution 3 (3.0 mL) to the primary emulsion, vortexing to create a coarse dispersion, and then sonicating at 30% amplitude for 60 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion is added to an open 50 mL beaker containing 70 mM phosphate buffer solution (30 mL) and stirred at room temperature for 2 to 3 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension.
  • a portion of the suspended nanocarriers is washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 75,600 rcf for 60 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure is repeated and then the pellet is re-suspended in phosphate buffered saline to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis.
  • Nanocarrier size is determined by dynamic light scattering.
  • the amounts of peptide and R848 in the nanocarrier are determined by HPLC analysis.
  • the total dry-nanocarrier mass per mL of suspension is determined by a gravimetric method.
  • Ovalbumin protein is purchased from Worthington Biochemical Corporation (730 Vassar Avenue, Lakewood, N.J. 08701. Product Code 3048.) PLA with an inherent viscosity of 0.22 dL/g is purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211. Product Code 100 DL 2A.) PLA-PEG-OMe block co-polymer with a methyl ether terminated PEG block of approximately 5,000 Da and PLA block of approximately 19,000 Da is synthesized.
  • Poly(L-glutamic R848 amide)-block-Polylactide of Example 4 is made having approximate PLA block molecular weight of 40,000 g/mol, and poly(L-glutamic R848 amide) block of approximately 18,000 g/mL, providing R848 loading of 22.5% on a weight/weight basis.
  • Polyvinyl alcohol PhEur, USP (85-89% hydrolyzed, viscosity of 3.4-4.6 mPa ⁇ s) is purchased from EMD Chemicals Inc. (480 South Democrat Road Gibbstown, N.J. 08027. Part Number 4-88). Phosphate-buffered saline 1 ⁇ (PBS1 ⁇ ). From Mediatech Inc. (9345 Discovery Boulevard. Manassas, Va. 20109.) Product Code 21-040-CV.
  • Solution 1 Ovalbumin protein @ 40 mg/mL is prepared in PBS 1 ⁇ at room temperature.
  • Solution 2 Poly(L-glutamic R848-amide)-block-polylactide at 20 mg/mL, PLA at 55 mg/mL, and PLA-PEG-OMe at 25 mg/mL in dichloromethane is prepared by creating individual solutions of each polymer in dichloromethane at 100 mg/mL, and then combining portions of those solutions in a 0.2:0.55:0.25 volume ratio, respectively.
  • Solution 3 Polyvinyl alcohol @ 100 mg/mL in 100 mM in 100 mM phosphate buffer, pH 8.
  • Solution 4 70 mM phosphate buffer, pH 8.
  • a primary (W1/O) emulsion is first created using Solution 1 & Solution 2.
  • Solution 1 (0.2 mL) and Solution 2 (1.0 mL) are combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/O/W2) emulsion is then formed by adding Solution 3 (3.0 mL) to the primary emulsion, vortexing to create a coarse dispersion, and then sonicating at 30% amplitude for 60 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion is added to an open 50 mL beaker containing 70 mM phosphate buffer solution (30 mL) and stirred at room temperature for 2 to 3 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension.
  • a portion of the suspended nanocarriers is washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 75,600 rcf for 60 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure is repeated and then the pellet is re-suspended in phosphate buffered saline to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis.
  • Nanocarrier size is determined by dynamic light scattering.
  • the amount of R848 in the nanocarrier are determined by HPLC analysis.
  • Ovalbumin protein content is established by PAGE.
  • the total dry-nanocarrier mass per mL of suspension is determined by a gravimetric method.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Virology (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Molecular Biology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pulmonology (AREA)
  • Biotechnology (AREA)
  • AIDS & HIV (AREA)
  • Inorganic Chemistry (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Compositions Of Macromolecular Compounds (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)
US13/560,943 2011-07-29 2012-07-27 Synthetic nanocarriers comprising polymers comprising multiple immunomodulatory agents Abandoned US20130028857A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/560,943 US20130028857A1 (en) 2011-07-29 2012-07-27 Synthetic nanocarriers comprising polymers comprising multiple immunomodulatory agents

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161513496P 2011-07-29 2011-07-29
US201161513526P 2011-07-29 2011-07-29
US201161513527P 2011-07-29 2011-07-29
US13/560,943 US20130028857A1 (en) 2011-07-29 2012-07-27 Synthetic nanocarriers comprising polymers comprising multiple immunomodulatory agents

Publications (1)

Publication Number Publication Date
US20130028857A1 true US20130028857A1 (en) 2013-01-31

Family

ID=47597380

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/560,955 Active US10933129B2 (en) 2011-07-29 2012-07-27 Methods for administering synthetic nanocarriers that generate humoral and cytotoxic T lymphocyte responses
US13/560,943 Abandoned US20130028857A1 (en) 2011-07-29 2012-07-27 Synthetic nanocarriers comprising polymers comprising multiple immunomodulatory agents
US13/560,925 Abandoned US20130039954A1 (en) 2011-07-29 2012-07-27 Control of antibody responses to synthetic nanocarriers

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US13/560,955 Active US10933129B2 (en) 2011-07-29 2012-07-27 Methods for administering synthetic nanocarriers that generate humoral and cytotoxic T lymphocyte responses

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/560,925 Abandoned US20130039954A1 (en) 2011-07-29 2012-07-27 Control of antibody responses to synthetic nanocarriers

Country Status (12)

Country Link
US (3) US10933129B2 (ru)
EP (1) EP2736537A4 (ru)
JP (2) JP2014521687A (ru)
KR (1) KR20140050698A (ru)
CN (3) CN109172819A (ru)
AU (3) AU2012290306B2 (ru)
BR (1) BR112014002139A2 (ru)
CA (1) CA2843274A1 (ru)
EA (1) EA201490381A1 (ru)
IL (2) IL230269B (ru)
MX (1) MX2014001142A (ru)
WO (3) WO2013019648A1 (ru)

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110020388A1 (en) * 2009-05-27 2011-01-27 Selecta Biosciences, Inc. Targeted synthetic nanocarriers with ph sensitive release of immunomodulatory agents
US20110110965A1 (en) * 2009-08-26 2011-05-12 Selecta Biosciences, Inc. Compositions that induce t cell help
US20110223201A1 (en) * 2009-04-21 2011-09-15 Selecta Biosciences, Inc. Immunonanotherapeutics Providing a Th1-Biased Response
US8652487B2 (en) 2011-04-29 2014-02-18 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for inducing regulatory B cells
US9066978B2 (en) 2010-05-26 2015-06-30 Selecta Biosciences, Inc. Dose selection of adjuvanted synthetic nanocarriers
US20150359865A1 (en) * 2014-06-17 2015-12-17 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for t-cell-mediated autoimmune disease
US20160022835A1 (en) * 2013-03-15 2016-01-28 The Brigham And Women's Hospital, Inc. Targeted Polymeric Inflammation-Resolving Nanoparticles
EP2991629A4 (en) * 2013-05-03 2016-11-16 Selecta Biosciences Inc ADMINISTRATION OF IMMUNOSUPPRESSIVA WITH SPECIFIC PHARMACODYNAMIC EFFECTIVE LIFE AND ANTIGEN FOR INDICATING IMMUNOTOLERANCE
WO2017205338A1 (en) * 2016-05-23 2017-11-30 University Of Miami Compositions for selective humoral responses and methods of use thereof
US9994443B2 (en) 2010-11-05 2018-06-12 Selecta Biosciences, Inc. Modified nicotinic compounds and related methods
US10046064B2 (en) 2014-09-07 2018-08-14 Selecta Biosciences, Inc. Methods and compositions for attenuating exon skipping anti-viral transfer vector immune responses
WO2019035971A1 (en) 2017-08-16 2019-02-21 Bristol-Myers Squibb Company 6-AMINO-7,9-DIHYDRO-8H-PURIN-8-ONE DERIVATIVES AS TOLL 7 RECEPTOR IMMUNOSTIMULATING AGONISTS (TLR7)
WO2019035970A1 (en) 2017-08-16 2019-02-21 Bristol-Myers Squibb Company 6-AMINO-7,9-DIHYDRO-8H-PURIN-8-ONE DERIVATIVES FOR USE AS TOLL 7 RECEPTOR IMMUNOSTIMULATING AGONISTS (TLR7)
WO2019035968A1 (en) 2017-08-16 2019-02-21 Bristol-Myers Squibb Company 6-AMINO-7,9-DIHYDRO-8H-PURIN-8-ONE DERIVATIVES AS TOLL RECEPTOR 7 (TLR7) AGONISTS AS IMMUNOSTIMULANTS
WO2019035969A1 (en) 2017-08-16 2019-02-21 Bristol-Myers Squibb Company TOLL RECEPTOR 7 (TLR7) AGONISTS COMPRISING A TRICYCLIC FRAGMENT, CONJUGATES CONTAINING SAME, RELATED METHODS AND USES THEREOF
WO2019036023A1 (en) 2017-08-16 2019-02-21 Bristol-Myers Squibb Company 6-AMINO-7,9-DIHYDRO-8H-PURIN-8-ONE DERIVATIVES AS IMMUNOSTIMULANT TOLL (TLR7) RECEPTOR 7 AGONISTS
US10273333B2 (en) * 2014-08-13 2019-04-30 The Regents Of The University Of California Substituted polyesters by thiol-ene modification: rapid diversification for therapeutic protein stabilization
WO2020028608A1 (en) 2018-08-03 2020-02-06 Bristol-Myers Squibb Company 1H-PYRAZOLO[4,3-d]PYRIMIDINE COMPOUNDS AS TOLL-LIKE RECEPTOR 7 (TLR7) AGONISTS AND METHODS AND USES THEREFOR
AU2014262164B2 (en) * 2013-05-03 2020-02-27 Selecta Biosciences, Inc. Dosing combinations for reducing undesired humoral immune responses
CN111122429A (zh) * 2019-12-31 2020-05-08 华南理工大学 纳米纤维素纤化程度的快速检测方法
US10837018B2 (en) 2013-07-25 2020-11-17 Exicure, Inc. Spherical nucleic acid-based constructs as immunostimulatory agents for prophylactic and therapeutic use
US10933129B2 (en) 2011-07-29 2021-03-02 Selecta Biosciences, Inc. Methods for administering synthetic nanocarriers that generate humoral and cytotoxic T lymphocyte responses
US11123294B2 (en) 2014-06-04 2021-09-21 Exicure Operating Company Multivalent delivery of immune modulators by liposomal spherical nucleic acids for prophylactic or therapeutic applications
US11213593B2 (en) 2014-11-21 2022-01-04 Northwestern University Sequence-specific cellular uptake of spherical nucleic acid nanoparticle conjugates
US11364304B2 (en) 2016-08-25 2022-06-21 Northwestern University Crosslinked micellar spherical nucleic acids
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
US11690920B2 (en) 2017-07-13 2023-07-04 Northwestern University General and direct method for preparing oligonucleotide-functionalized metal-organic framework nanoparticles
US11696954B2 (en) 2017-04-28 2023-07-11 Exicure Operating Company Synthesis of spherical nucleic acids using lipophilic moieties
US11883535B2 (en) 2013-12-03 2024-01-30 Northwestern University Liposomal particles, methods of making same and uses thereof

Families Citing this family (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
HUE058896T2 (hu) 2010-10-01 2022-09-28 Modernatx Inc N1-metil-pszeudo-uracilt tartalmazó ribonukleinsavak és azok felhasználásai
US20130323319A1 (en) 2010-11-12 2013-12-05 Getts Consulting And Project Management Modified immune-modulating particles
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
RS62993B1 (sr) 2011-10-03 2022-03-31 Modernatx Inc Modifikovani nukleozidi, nukleotidi, i nukleinske kiseline, i njihove upotrebe
CN104114572A (zh) 2011-12-16 2014-10-22 现代治疗公司 经修饰的核苷、核苷酸和核酸组合物
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
JP2015518705A (ja) 2012-04-02 2015-07-06 モデルナ セラピューティクス インコーポレイテッドModerna Therapeutics,Inc. ヒト疾患に関連する生物製剤およびタンパク質の産生のための修飾ポリヌクレオチド
DE18200782T1 (de) 2012-04-02 2021-10-21 Modernatx, Inc. Modifizierte polynukleotide zur herstellung von proteinen im zusammenhang mit erkrankungen beim menschen
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
ES2738481T3 (es) 2012-06-21 2020-01-23 Univ Northwestern Partículas peptídicas conjugadas
EP4074834A1 (en) 2012-11-26 2022-10-19 ModernaTX, Inc. Terminally modified rna
IL292823B2 (en) 2013-03-13 2023-11-01 Cour Pharmaceuticals Dev Company Secondary vaccine particles for the treatment of inflammation
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
KR20230008909A (ko) 2013-08-13 2023-01-16 노쓰웨스턴유니버시티 펩티드-접합된 입자
AU2014315287A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
EP3041938A1 (en) 2013-09-03 2016-07-13 Moderna Therapeutics, Inc. Circular polynucleotides
EP3052106A4 (en) 2013-09-30 2017-07-19 ModernaTX, Inc. Polynucleotides encoding immune modulating polypeptides
MX2016004249A (es) 2013-10-03 2016-11-08 Moderna Therapeutics Inc Polinulcleotidos que codifican el receptor de lipoproteina de baja densidad.
JP6621409B2 (ja) 2013-11-22 2019-12-18 ミナ セラピューティクス リミテッド C/EBPα小分子活性化RNA組成物
EP2966160A1 (en) 2014-07-09 2016-01-13 Clariant International Ltd. Storage-stable compositions comprising soil release polymers
EP3167033B1 (en) 2014-07-09 2020-04-29 Unilever Plc. Process for making a laundry liquid composition
US9919058B2 (en) 2014-07-15 2018-03-20 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Polyketal particles including a CpG oligodeoxynucleotide for the treatment of lung cancer
US20170210788A1 (en) 2014-07-23 2017-07-27 Modernatx, Inc. Modified polynucleotides for the production of intrabodies
KR101892689B1 (ko) * 2014-10-14 2018-08-28 삼성전기주식회사 칩 전자부품 및 칩 전자부품의 실장 기판
US20170025028A1 (en) * 2015-07-23 2017-01-26 Rhythmalytics LLC Actigraphy based biological rhythm modification methods and systems that result in a greater efficacy of applied medical treatment to a patient
MD3718565T2 (ro) 2015-10-22 2022-09-30 Modernatx Inc Vaccinuri împotriva virusului respirator
WO2017106630A1 (en) 2015-12-18 2017-06-22 The General Hospital Corporation Polyacetal polymers, conjugates, particles and uses thereof
PT3394093T (pt) 2015-12-23 2022-05-30 Modernatx Inc Métodos de utilização de polinucleotídeos que codificam ligandos ox40
EP3400023A1 (en) 2016-01-10 2018-11-14 ModernaTX, Inc. Therapeutic mrnas encoding anti ctla-4 antibodies
CN106065047B (zh) * 2016-07-14 2019-02-26 山东省肿瘤防治研究院 一种肝靶向阳离子聚合物及其制备方法与应用
US11542490B2 (en) 2016-12-08 2023-01-03 CureVac SE RNAs for wound healing
EP3808380A1 (en) 2016-12-08 2021-04-21 CureVac AG Rna for treatment or prophylaxis of a liver disease
US11285203B2 (en) 2017-06-23 2022-03-29 Verimmune Inc. Chimeric virus-like particles and uses thereof as antigen-specific redirectors of immune responses
WO2019048632A1 (en) 2017-09-08 2019-03-14 Mina Therapeutics Limited STABILIZED COMPOSITIONS OF SMALL ACTIVATORY RNA (PARNA) OF HNF4A AND METHODS OF USE
US20200208152A1 (en) 2017-09-08 2020-07-02 Mina Therapeutics Limited Stabilized sarna compositions and methods of use
SG11202002044WA (en) 2017-09-21 2020-04-29 Emergex Vaccines Holding Ltd MHC Class I Associated Peptides for Prevention and Treatment of Zika Virus
EP4242307A3 (en) 2018-04-12 2023-12-27 MiNA Therapeutics Limited Sirt1-sarna compositions and methods of use
US11560408B2 (en) 2018-12-27 2023-01-24 Verimmune Inc. Conjugated virus-like particles and uses thereof as anti-tumor immune redirectors
EP3953473A1 (en) 2019-04-12 2022-02-16 MiNA Therapeutics Limited Sirt1-sarna compositions and methods of use
CN110938200B (zh) * 2019-12-05 2021-04-20 大连理工大学 一种侧链含二甲基吡啶胺类聚酯的制备方法
MX2019015508A (es) 2019-12-18 2021-06-21 Univ Guadalajara Nanopartículas para el tratamiento del cáncer.
US20220193225A1 (en) * 2020-08-31 2022-06-23 Bruce Lyday Compositions and methods for sars-2 vaccine with virus replicative particles and recombinant glycoproteins
CN114177282B (zh) * 2020-09-14 2024-03-15 苏州大学 氟化聚乙烯亚胺在制备疫苗或预防/治疗病毒/细菌引起的疾病的制剂中的应用
JP2023552040A (ja) 2020-10-19 2023-12-14 ヴェリミューン インコーポレイテッド ウイルスにインスパイアされた組成物及びがんの治療のために同じものを使用して、既存の免疫応答をリダイレクトする方法
GB2603454A (en) 2020-12-09 2022-08-10 Ucl Business Ltd Novel therapeutics for the treatment of neurodegenerative disorders
AU2022246144A1 (en) 2021-03-26 2023-09-21 Mina Therapeutics Limited Tmem173 sarna compositions and methods of use
EP4377331A2 (en) 2021-07-30 2024-06-05 CureVac SE Mrnas for treatment or prophylaxis of liver diseases
WO2023099884A1 (en) 2021-12-01 2023-06-08 Mina Therapeutics Limited Pax6 sarna compositions and methods of use
GB202117758D0 (en) 2021-12-09 2022-01-26 Ucl Business Ltd Therapeutics for the treatment of neurodegenerative disorders
WO2023144193A1 (en) 2022-01-25 2023-08-03 CureVac SE Mrnas for treatment of hereditary tyrosinemia type i
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
WO2024134199A1 (en) 2022-12-22 2024-06-27 Mina Therapeutics Limited Chemically modified sarna compositions and methods of use

Family Cites Families (332)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR7461M (ru) 1968-06-19 1970-01-05
GB1355961A (en) 1970-02-27 1974-06-12 Wellcome Found Preparation of immunosuppressive antilymphocytic serum
CH594444A5 (ru) 1972-12-04 1978-01-13 Gerd Birrenbach
DK143689C (da) 1975-03-20 1982-03-15 J Kreuter Fremgangsmaade til fremstilling af en adsorberet vaccine
US4756907A (en) 1978-10-17 1988-07-12 Stolle Research & Development Corp. Active/passive immunization of the internal female reproductive organs
US4946929A (en) 1983-03-22 1990-08-07 Massachusetts Institute Of Technology Bioerodible articles useful as implants and prostheses having predictable degradation rates
US6309669B1 (en) 1984-03-16 2001-10-30 The United States Of America As Represented By The Secretary Of The Army Therapeutic treatment and prevention of infections with a bioactive materials encapsulated within a biodegradable-biocompatible polymeric matrix
US4638045A (en) 1985-02-19 1987-01-20 Massachusetts Institute Of Technology Non-peptide polyamino acid bioerodible polymers
US4631211A (en) 1985-03-25 1986-12-23 Scripps Clinic & Research Foundation Means for sequential solid phase organic synthesis and methods using the same
US4806621A (en) 1986-01-21 1989-02-21 Massachusetts Institute Of Technology Biocompatible, bioerodible, hydrophobic, implantable polyimino carbonate article
JPS63122620A (ja) 1986-11-12 1988-05-26 Sanraku Inc ポリ乳酸マイクロスフエア及びその製造方法
CA1340581C (en) 1986-11-20 1999-06-08 Joseph P. Vacanti Chimeric neomorphogenesis of organs by controlled cellular implantation using artificial matrices
US5736372A (en) 1986-11-20 1998-04-07 Massachusetts Institute Of Technology Biodegradable synthetic polymeric fibrous matrix containing chondrocyte for in vivo production of a cartilaginous structure
US5804178A (en) 1986-11-20 1998-09-08 Massachusetts Institute Of Technology Implantation of cell-matrix structure adjacent mesentery, omentum or peritoneum tissue
FR2608988B1 (fr) 1986-12-31 1991-01-11 Centre Nat Rech Scient Procede de preparation de systemes colloidaux dispersibles d'une substance, sous forme de nanoparticules
US5912017A (en) 1987-05-01 1999-06-15 Massachusetts Institute Of Technology Multiwall polymeric microspheres
US5229490A (en) 1987-05-06 1993-07-20 The Rockefeller University Multiple antigen peptide system
US5019379A (en) 1987-07-31 1991-05-28 Massachusetts Institute Of Technology Unsaturated polyanhydrides
US6130082A (en) 1988-05-05 2000-10-10 American Cyanamid Company Recombinant flagellin vaccines
US4929624A (en) 1989-03-23 1990-05-29 Minnesota Mining And Manufacturing Company Olefinic 1H-imidazo(4,5-c)quinolin-4-amines
US5010167A (en) 1989-03-31 1991-04-23 Massachusetts Institute Of Technology Poly(amide-and imide-co-anhydride) for biological application
US5114703A (en) 1989-05-30 1992-05-19 Alliance Pharmaceutical Corp. Percutaneous lymphography using particulate fluorocarbon emulsions
US5733572A (en) 1989-12-22 1998-03-31 Imarx Pharmaceutical Corp. Gas and gaseous precursor filled microspheres as topical and subcutaneous delivery vehicles
GB9016885D0 (en) 1990-08-01 1990-09-12 Scras Sustained release pharmaceutical compositions
US6699474B1 (en) 1990-08-20 2004-03-02 Erich Hugo Cerny Vaccine and immunserum against drugs of abuse
US5389640A (en) 1991-03-01 1995-02-14 Minnesota Mining And Manufacturing Company 1-substituted, 2-substituted 1H-imidazo[4,5-c]quinolin-4-amines
US5175296A (en) 1991-03-01 1992-12-29 Minnesota Mining And Manufacturing Company Imidazo[4,5-c]quinolin-4-amines and processes for their preparation
US6159502A (en) 1991-04-02 2000-12-12 Biotech Australia Pty Ltd Oral delivery systems for microparticles
US5811447A (en) 1993-01-28 1998-09-22 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
IL105325A (en) 1992-04-16 1996-11-14 Minnesota Mining & Mfg Immunogen/vaccine adjuvant composition
US6235313B1 (en) 1992-04-24 2001-05-22 Brown University Research Foundation Bioadhesive microspheres and their use as drug delivery and imaging systems
DK0671948T3 (da) 1992-06-25 1997-09-01 Smithkline Beecham Biolog Vaccinepræparat indeholdende adjuvanser
AU4778893A (en) 1992-07-21 1994-02-14 General Hospital Corporation, The System of drug delivery to the lymphatic tissues
GB9216082D0 (en) 1992-07-28 1992-09-09 Univ Nottingham Lymphatic delivery composition
US6608201B2 (en) 1992-08-28 2003-08-19 3M Innovative Properties Company Process for preparing 1-substituted, 2-substituted 1H-imidazo[4,5-c]quinolin-4-amines
FR2695563B1 (fr) 1992-09-11 1994-12-02 Pasteur Institut Microparticules portant des antigènes et leur utilisation pour l'induction de réponses humorales ou cellulaires.
AU4932493A (en) 1992-09-25 1994-04-26 Dynagen, Inc. An immunobooster for delayed release of immunogen
US5399665A (en) 1992-11-05 1995-03-21 Massachusetts Institute Of Technology Biodegradable polymers for cell transplantation
ES2131676T3 (es) 1993-01-11 1999-08-01 Dana Farber Cancer Inst Inc Induccion de respuestas con linfocitos t citotoxicos.
US5512600A (en) 1993-01-15 1996-04-30 Massachusetts Institute Of Technology Preparation of bonded fiber structures for cell implantation
US5514378A (en) 1993-02-01 1996-05-07 Massachusetts Institute Of Technology Biocompatible polymer membranes and methods of preparation of three dimensional membrane structures
EP0684814B1 (en) 1993-02-22 1998-06-17 Alza Corporation Compositions for oral delivery of active agents
ATE156706T1 (de) 1993-03-17 1997-08-15 Silica Gel Gmbh Superparamagnetische teilchen, verfahren zu ihrer herstellung und verwendung derselben
US5565215A (en) 1993-07-23 1996-10-15 Massachusettes Institute Of Technology Biodegradable injectable particles for imaging
US5543158A (en) 1993-07-23 1996-08-06 Massachusetts Institute Of Technology Biodegradable injectable nanoparticles
WO1995003035A1 (en) 1993-07-23 1995-02-02 Massachusetts Institute Of Technology Polymerized liposomes with enhanced stability for oral delivery
JP3926839B2 (ja) 1993-09-14 2007-06-06 エピミューン,インコーポレイティド 万能dr−結合性ペプチドを用いる免疫応答の改変
US5798340A (en) 1993-09-17 1998-08-25 Gilead Sciences, Inc. Nucleotide analogs
US5500161A (en) 1993-09-21 1996-03-19 Massachusetts Institute Of Technology And Virus Research Institute Method for making hydrophobic polymeric microparticles
CA2184242C (en) 1994-02-28 2000-05-02 Jorg Kreuter Drug targeting system, method for preparing same and its use
WO1995026204A1 (en) 1994-03-25 1995-10-05 Isis Pharmaceuticals, Inc. Immune stimulation by phosphorothioate oligonucleotide analogs
GB9412273D0 (en) 1994-06-18 1994-08-10 Univ Nottingham Administration means
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
WO1996002555A1 (en) 1994-07-15 1996-02-01 The University Of Iowa Research Foundation Immunomodulatory oligonucleotides
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6007845A (en) 1994-07-22 1999-12-28 Massachusetts Institute Of Technology Nanoparticles and microparticles of non-linear hydrophilic-hydrophobic multiblock copolymers
US5716404A (en) 1994-12-16 1998-02-10 Massachusetts Institute Of Technology Breast tissue engineering
ATE252894T1 (de) 1995-01-05 2003-11-15 Univ Michigan Oberflächen-modifizierte nanopartikel und verfahren für ihre herstellung und verwendung
US5876727A (en) 1995-03-31 1999-03-02 Immulogic Pharmaceutical Corporation Hapten-carrier conjugates for use in drug-abuse therapy and methods for preparation of same
US6123727A (en) 1995-05-01 2000-09-26 Massachusetts Institute Of Technology Tissue engineered tendons and ligaments
US5866132A (en) 1995-06-07 1999-02-02 Alberta Research Council Immunogenic oligosaccharide compositions
WO1997004747A1 (en) 1995-07-27 1997-02-13 Dunn James M Drug delivery systems for macromolecular drugs
EP0850051A2 (en) 1995-08-31 1998-07-01 Alkermes Controlled Therapeutics, Inc. Composition for sustained release of an agent
US6095148A (en) 1995-11-03 2000-08-01 Children's Medical Center Corporation Neuronal stimulation using electrically conducting polymers
US5902599A (en) 1996-02-20 1999-05-11 Massachusetts Institute Of Technology Biodegradable polymer networks for use in orthopedic and dental applications
US6946133B1 (en) 1996-03-20 2005-09-20 The United States Of America As Represented By The Department Of Health And Human Services Prostate specific antigen oligo-epitope peptide
DE69723434T2 (de) 1996-04-26 2004-05-19 Rijksuniversiteit Te Leiden Verfahren zur selektion und produktion von t-zell-peptide epitope und vakzine mit diese epitope
US5874064A (en) 1996-05-24 1999-02-23 Massachusetts Institute Of Technology Aerodynamically light particles for pulmonary drug delivery
US5922695A (en) 1996-07-26 1999-07-13 Gilead Sciences, Inc. Antiviral phosphonomethyoxy nucleotide analogs having increased oral bioavarilability
CA2279651A1 (en) 1996-09-05 1998-03-12 Massachusetts Institute Of Technology Compositions and methods for treatment of neurological disorders and neurodegenerative diseases
HUP9904665A3 (en) 1996-10-25 2000-11-28 Minnesota Mining And Mfg Co Sa Immune response modifier compounds for treatment of th2 mediated and related diseases
US6042820A (en) 1996-12-20 2000-03-28 Connaught Laboratories Limited Biodegradable copolymer containing α-hydroxy acid and α-amino acid units
JP2001513776A (ja) 1997-02-28 2001-09-04 ユニバーシティ オブ アイオワ リサーチ ファウンデーション LPS関連障害の処置における非メチル化CpGジヌクレオチドを含む核酸の使用
DK1005368T3 (da) 1997-03-10 2010-01-04 Ottawa Hospital Res Inst Anvendelse af nukleinsyrer, der indeholder ikke-metyleret CpG dinukleotid i kombination med alun som hjælpestoffer
US6211159B1 (en) 1997-04-11 2001-04-03 University Of Toronto Flagellin gene, FlaC of campylobacter
US6060082A (en) 1997-04-18 2000-05-09 Massachusetts Institute Of Technology Polymerized liposomes targeted to M cells and useful for oral or mucosal drug delivery
ATE370740T1 (de) 1997-05-20 2007-09-15 Ottawa Health Research Inst Verfahren zur herstellung von nukleinsäurekonstrukten
US5837752A (en) 1997-07-17 1998-11-17 Massachusetts Institute Of Technology Semi-interpenetrating polymer networks
US6989435B2 (en) 1997-09-11 2006-01-24 Cambridge University Technical Services Ltd. Compounds and methods to inhibit or augment an inflammatory response
AU741130B2 (en) 1997-09-16 2001-11-22 Oregon Health Sciences University Recombinant MHC molecules useful for manipulation of antigen-specific T-cells
DE19745950A1 (de) 1997-10-17 1999-04-22 Dds Drug Delivery Service Ges Arzneistoffträgerpartikel für die gewebespezifische Arzneistoffapplikation
WO1999028321A1 (fr) 1997-11-28 1999-06-10 Sumitomo Pharmaceuticals Company, Limited Nouveaux composes heterocycliques
US6254890B1 (en) 1997-12-12 2001-07-03 Massachusetts Institute Of Technology Sub-100nm biodegradable polymer spheres capable of transporting and releasing nucleic acids
US6197229B1 (en) 1997-12-12 2001-03-06 Massachusetts Institute Of Technology Method for high supercoiled DNA content microspheres
FR2775435B1 (fr) 1998-02-27 2000-05-26 Bioalliance Pharma Nanoparticules comprenant au moins un polymere et au moins un compose apte a complexer un ou plusieurs principes actifs
US6232287B1 (en) 1998-03-13 2001-05-15 The Burnham Institute Molecules that home to various selected organs or tissues
US6506577B1 (en) 1998-03-19 2003-01-14 The Regents Of The University Of California Synthesis and crosslinking of catechol containing copolypeptides
US6632922B1 (en) 1998-03-19 2003-10-14 The Regents Of The University Of California Methods and compositions for controlled polypeptide synthesis
US6686446B2 (en) 1998-03-19 2004-02-03 The Regents Of The University Of California Methods and compositions for controlled polypeptide synthesis
ES2284247T3 (es) 1998-04-03 2007-11-01 University Of Iowa Research Foundation Metodos y productos para estimular el sistema inmunitario usando oligonucleotidos y citoquinas inmunoterapeuticos.
AU3884199A (en) 1998-05-06 1999-11-23 Ottawa Health Research Institute Methods for the prevention and treatment of parasitic infections and related diseases using cpg oligonucleotides
SE9801923D0 (sv) 1998-05-29 1998-05-29 Independent Pharmaceutical Ab Nicotine vaccine
US6693086B1 (en) 1998-06-25 2004-02-17 National Jewish Medical And Research Center Systemic immune activation method using nucleic acid-lipid complexes
US6242589B1 (en) 1998-07-14 2001-06-05 Isis Pharmaceuticals, Inc. Phosphorothioate oligonucleotides having modified internucleoside linkages
ATE321535T1 (de) 1998-07-29 2006-04-15 Chiron Corp Mikropartikel mit adsorbenten oberflächen, verfahren zu ihrer herstellung und ihrer verwendung
DE19839214C1 (de) 1998-08-28 2000-05-25 Aventis Res & Tech Gmbh & Co Verfahren zur Herstellung von sphärischen Mikropartikeln mit glatter Oberfläche, die ganz oder teilweise aus mindestens einem wasserunlöslichen linearen Polysaccharid bestehen, sowie mit diesem Verfahren erhältliche Mikropartikel und deren Verwendung
US6306640B1 (en) 1998-10-05 2001-10-23 Genzyme Corporation Melanoma antigenic peptides
HUP0103976A3 (en) 1998-10-05 2008-04-28 Pharmexa As Novel methods for therapeutic vaccination
PT1126876E (pt) 1998-10-16 2007-04-30 Glaxosmithkline Biolog Sa Sistemas adjuvantes e vacinas
ID29852A (id) 1998-11-02 2001-10-18 Elan Corp Plc Cs Komposisi pelepasan modifikasi multi partikulasi
US7521068B2 (en) 1998-11-12 2009-04-21 Elan Pharma International Ltd. Dry powder aerosols of nanoparticulate drugs
WO2000032626A1 (en) 1998-11-25 2000-06-08 Regents Of The University Of Minnesota Methods of using epitope peptides of human pathogens
BR9915771A (pt) 1998-11-30 2001-12-26 Cytos Biotechnology Ag Apresentação molecular ordenada de antìgenos,processo de preparação e utilização
US6232082B1 (en) 1998-12-01 2001-05-15 Nabi Hapten-carrier conjugates for treating and preventing nicotine addiction
BR0007435A (pt) 1999-01-08 2001-12-04 3M Innovative Properties Co Formulações e métodos para tratamento decondições associadas com mucosa com ummodificador de resposta imunológica
US7238711B1 (en) 1999-03-17 2007-07-03 Cambridge University Technical Services Ltd. Compounds and methods to inhibit or augment an inflammatory response
US6444192B1 (en) 1999-02-05 2002-09-03 The Regents Of The University Of California Diagnostic imaging of lymph structures
US6558951B1 (en) 1999-02-11 2003-05-06 3M Innovative Properties Company Maturation of dendritic cells with immune response modifying compounds
ES2228454T3 (es) 1999-02-26 2005-04-16 Chiron S.R.L. Mejora de la actividad bacteriana de antigenos de neisseria con oligonucleotidos que contienen motivos cg.
AU764969B2 (en) 1999-04-19 2003-09-04 Smithkline Beecham Biologicals (Sa) Vaccines
AU4825700A (en) 1999-05-06 2000-11-21 Genetics Institute Inc. Use of soluble costimulatory molecules to enhance immune responses
US6800296B1 (en) 1999-05-19 2004-10-05 Massachusetts Institute Of Technology Modification of surfaces using biological recognition events
US6815170B1 (en) 1999-06-30 2004-11-09 John Wayne Cancer Institute Methods for lymph node identification
WO2001012071A1 (en) 1999-08-13 2001-02-22 Point Biomedical Corporation Microparticles useful as ultrasonic contrast agents and for lymphatic system
MXPA02003108A (es) 1999-09-25 2003-10-14 Univ Iowa Res Found Acidos nucleicos inmunoestimuladores.
US7223398B1 (en) 1999-11-15 2007-05-29 Dynavax Technologies Corporation Immunomodulatory compositions containing an immunostimulatory sequence linked to antigen and methods of use thereof
CA2391534A1 (en) 1999-11-15 2001-05-25 Drug Innovation & Design, Inc. Selective cellular targeting: multifunctional delivery vehicles
US7462354B2 (en) 1999-12-28 2008-12-09 Pharmexa Inc. Method and system for optimizing minigenes and peptides encoded thereby
CA2396113C (en) 2000-01-13 2009-04-07 Nanosphere, Inc. Nanoparticles having oligonucleotides attached thereto and uses therefor
AT409085B (de) * 2000-01-28 2002-05-27 Cistem Biotechnologies Gmbh Pharmazeutische zusammensetzung zur immunmodulation und herstellung von vakzinen
US8202979B2 (en) 2002-02-20 2012-06-19 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid
US20050020525A1 (en) 2002-02-20 2005-01-27 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (siNA)
US20050032733A1 (en) 2001-05-18 2005-02-10 Sirna Therapeutics, Inc. RNA interference mediated inhibition of gene expression using chemically modified short interfering nucleic acid (SiNA)
JP4751556B2 (ja) 2000-02-28 2011-08-17 ジーンシーグス, インコーポレイテッド ナノカプセルカプセル化システムおよび方法
US7157437B2 (en) 2000-03-10 2007-01-02 Dynavax Technologies Corporation Methods of ameliorating symptoms of herpes infection using immunomodulatory polynucleotide sequences
US7129222B2 (en) 2000-03-10 2006-10-31 Dynavax Technologies Corporation Immunomodulatory formulations and methods for use thereof
US20030129251A1 (en) 2000-03-10 2003-07-10 Gary Van Nest Biodegradable immunomodulatory formulations and methods for use thereof
SE0000933D0 (sv) 2000-03-21 2000-03-21 Independent Pharmaceutica Ab Method of producing 6-substituted (S)-nicotine derivatives and intermediate compounds
AU2001252458A1 (en) 2000-05-05 2001-11-20 Martin Bachmann Molecular antigen arrays and vaccines
US7192725B2 (en) 2000-05-19 2007-03-20 University Of Toronto Flagellin gene, flaC of Campylobacter
US6610713B2 (en) 2000-05-23 2003-08-26 North Shore - Long Island Jewish Research Institute Inhibition of inflammatory cytokine production by cholinergic agonists and vagus nerve stimulation
US20020099013A1 (en) * 2000-11-14 2002-07-25 Thomas Piccariello Active agent delivery systems and methods for protecting and administering active agents
CA2319928A1 (en) 2000-09-18 2002-03-18 Vasogen Ireland Limited Apoptosis-mimicking synthetic entities and use thereof in medical treatments
JP2004509970A (ja) 2000-09-26 2004-04-02 ハイブリドン・インコーポレイテッド 位置的化学変化による免疫刺激オリゴヌクレオチドアナログの免疫刺激活性の調節
AU2001297913A1 (en) 2000-10-13 2002-12-23 Ligocyte Pharmaceuticals, Inc. Polyvalent nanoparticles
GB0025414D0 (en) 2000-10-16 2000-11-29 Consejo Superior Investigacion Nanoparticles
WO2002032450A2 (en) 2000-10-18 2002-04-25 Glaxosmithkline Biologicals S.A. Vaccines
EP1355965B1 (en) 2000-10-19 2012-09-19 Ecole Polytechnique Fédérale de Lausanne (EPFL) Method of synthesizing block copolymers for multifunctional self-assembled systems
US7592008B2 (en) 2000-11-20 2009-09-22 The Board Of Trustees Of The University Of Illinois, A Body Corporate And Politic Of The State Of Illinois Membrane scaffold proteins
CA2598144A1 (en) 2000-12-08 2006-08-31 3M Innovative Properties Company Compositions and methods for targeted delivery of immune response modifiers
AU2002231336B2 (en) 2000-12-27 2007-06-28 Surefire Medical, Inc. D/B/A Trisalus Life Sciences Immunomodulatory polynucleotides and methods of using the same
US7320793B2 (en) 2001-01-19 2008-01-22 Cytos Biotechnology Ag Molecular antigen array
US7097837B2 (en) 2001-02-19 2006-08-29 Pharmexa A/S Synthetic vaccine agents
US20030175950A1 (en) 2001-05-29 2003-09-18 Mcswiggen James A. RNA interference mediated inhibition of HIV gene expression using short interfering RNA
AU2002367976B2 (en) 2001-06-05 2007-06-21 The Regents Of The University Of Michigan Nanoemulsion vaccines
US20030108565A1 (en) 2001-07-10 2003-06-12 Johnson Mark E. Compositions and methods for delivery of proteins and adjuvants encapsulated in microspheres
US20030133988A1 (en) 2001-08-07 2003-07-17 Fearon Karen L. Immunomodulatory compositions, formulations, and methods for use thereof
US6818732B2 (en) 2001-08-30 2004-11-16 The Regents Of The University Of California Transition metal initiators for controlled poly (beta-peptide) synthesis from beta-lactam monomers
US7276489B2 (en) 2002-10-24 2007-10-02 Idera Pharmaceuticals, Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by optimal presentation of 5′ ends
CN100509057C (zh) 2001-11-07 2009-07-08 赛托斯生物技术公司 用于治疗骨病的抗原阵列
DK1443960T3 (da) 2001-11-07 2009-03-23 Cytos Biotechnology Ag Antigen-arrays der betegner IL-5, IL-13 eller eotaxin til behandling af allergiske eosinofile sygdomme
US8088388B2 (en) 2002-02-14 2012-01-03 United Biomedical, Inc. Stabilized synthetic immunogen delivery system
AU2003215316A1 (en) 2002-02-20 2003-09-09 Chiron Corporation Microparticles with adsorbed polypeptide-containing molecules
US20030232013A1 (en) 2002-02-22 2003-12-18 Gary Sieckman Therapeutic and diagnostic targeting of cancers cells with tumor homing peptides
EP2368431A1 (en) 2002-04-04 2011-09-28 Coley Pharmaceutical GmbH Immunostimulatory G,U-containing oligoribonucleotides
US20040038303A1 (en) 2002-04-08 2004-02-26 Unger Gretchen M. Biologic modulations with nanoparticles
US20080233181A1 (en) 2002-04-12 2008-09-25 Nagy Jon O Nanoparticle adjuvants for sub-unit vaccines
US7285289B2 (en) 2002-04-12 2007-10-23 Nagy Jon O Nanoparticle vaccines
US7524630B2 (en) 2002-04-22 2009-04-28 University Of Florida Research Foundation, Inc. Functionalized nanoparticles and methods of use
AU2003276131A1 (en) 2002-06-18 2003-12-31 Epigenesis Pharmaceuticals, Inc. A dry powder oligonucleotide formulation, preparation and its uses
WO2004005325A2 (en) * 2002-07-10 2004-01-15 The Ohio State University Research Foundation Antigen-polymer compositions
CA2489410C (en) 2002-07-17 2015-01-13 Cytos Biotechnology Ag Molecular antigen arrays
IN2008CN05409A (ru) 2002-07-18 2015-10-02 Cytos Biotechnology Ag
WO2004016282A1 (en) 2002-07-19 2004-02-26 Cytos Biotechnology Ag Vaccine compositions containing amyloid beta1-6 antigen arrays
DK1545597T3 (da) 2002-08-15 2011-01-31 3M Innovative Properties Co Immunstimulerende sammensætninger og fremgangsmåde til stimulering af en immunrespons
US7488792B2 (en) 2002-08-28 2009-02-10 Burnham Institute For Medical Research Collagen-binding molecules that selectively home to tumor vasculature and methods of using same
US20040152105A1 (en) 2002-09-06 2004-08-05 Cytos Biotechnology Ag. Immune modulatory compounds and methods
US20060189554A1 (en) 2002-09-24 2006-08-24 Russell Mumper Nanoparticle-Based vaccine delivery system containing adjuvant
US7008411B1 (en) 2002-09-30 2006-03-07 Advanced Cardiovascular Systems, Inc. Method and apparatus for treating vulnerable plaque
NO20024755D0 (no) 2002-10-03 2002-10-03 Amersham Health As Metode
AU2003300919A1 (en) 2002-12-11 2004-06-30 Coley Pharmaceutical Gmbh 5' cpg nucleic acids and methods of use
SE0203687D0 (sv) 2002-12-13 2002-12-13 Ian Harwigsson Med Adagit Fa Pharmaceutical Porous Particles
SI1575977T1 (sl) 2002-12-23 2010-01-29 Dynavax Tech Corp Imunostimulirni sekvenčni polinukleotidi in postopki za njihovo uporabo
AU2003300184B8 (en) 2002-12-30 2009-12-03 3M Innovative Properties Company Immunostimulatory combinations
CA2512693A1 (en) 2003-01-08 2004-07-29 Xencor, Inc. Novel proteins with altered immunogenicity
US20040156846A1 (en) 2003-02-06 2004-08-12 Triton Biosystems, Inc. Therapy via targeted delivery of nanoscale particles using L6 antibodies
WO2004071459A2 (en) 2003-02-13 2004-08-26 3M Innovative Properties Company Methods and compositions related to irm compounds and toll-like receptor 8
DE602004008582T2 (de) * 2003-02-17 2008-05-21 Peter Burkhard Peptidische nanoteilchen als arzneimittelabgabe- und antigen-display-systeme
US20040191215A1 (en) 2003-03-25 2004-09-30 Michael Froix Compositions for induction of a therapeutic response
JP2006521367A (ja) 2003-03-26 2006-09-21 株式会社Lttバイオファーマ ターゲッティングと徐放を目的とした静脈注射用ナノ粒子
EP1606398A1 (en) 2003-03-26 2005-12-21 Cytos Biotechnology AG Melan-a peptide analogue-virus-like-particle conjugates
US7923560B2 (en) 2003-04-10 2011-04-12 3M Innovative Properties Company Delivery of immune response modifier compounds
US7731967B2 (en) 2003-04-30 2010-06-08 Novartis Vaccines And Diagnostics, Inc. Compositions for inducing immune responses
US7727969B2 (en) 2003-06-06 2010-06-01 Massachusetts Institute Of Technology Controlled release nanoparticle having bound oligonucleotide for targeted delivery
US7149574B2 (en) 2003-06-09 2006-12-12 Palo Alto Investors Treatment of conditions through electrical modulation of the autonomic nervous system
US20060182793A1 (en) 2003-07-22 2006-08-17 Cytos Biotechnology Ag Cpg-packaged liposomes
US20050042298A1 (en) 2003-08-20 2005-02-24 Pardridge William M. Immunonanoparticles
US20070014807A1 (en) 2003-09-03 2007-01-18 Maida Anthony E Iii Multiplex vaccine
US7943179B2 (en) 2003-09-23 2011-05-17 Massachusetts Institute Of Technology pH triggerable polymeric particles
US7771726B2 (en) 2003-10-08 2010-08-10 New York University Use of synthetic glycolipids as universal adjuvants for vaccines against cancer and infectious diseases
US20080160089A1 (en) 2003-10-14 2008-07-03 Medivas, Llc Vaccine delivery compositions and methods of use
DE10347710B4 (de) 2003-10-14 2006-03-30 Johannes-Gutenberg-Universität Mainz Rekombinante Impfstoffe und deren Verwendung
JP2007514519A (ja) 2003-10-20 2007-06-07 ウィリアム・マーシュ・ライス・ユニバーシティ ポリマー及び帯電ナノ粒子からなるマイクロカプセルを製造する方法
UA88457C2 (ru) 2003-10-30 2009-10-26 Коли Фармасьютикал Гмбх Иммуностимулирующая нуклеиновая кислота с улучшенной иммуностимулирующей эффективностью
CA2546616A1 (en) 2003-11-21 2005-06-09 Alza Corporation Gene delivery mediated by liposome-dna complex with cleavable peg surface modification
WO2005055949A2 (en) 2003-12-09 2005-06-23 The Children's Hospital Of Philadelphia Sustained release preparations composed of biocompatible complex microparticles
KR20120105062A (ko) 2003-12-19 2012-09-24 더 유니버시티 오브 노쓰 캐롤라이나 엣 채플 힐 소프트 또는 임프린트 리소그래피를 이용하여 분리된 마이크로- 및 나노- 구조를 제작하는 방법
US20050191294A1 (en) 2003-12-31 2005-09-01 Board Of Regents, The University Of Texas System Compositions and methods of use of targeting peptides for diagnosis and therapy
US20070087986A1 (en) 2004-01-26 2007-04-19 Brett Premack Compositions and methods for enhancing immunity by chemoattractant adjuvants
AU2005230938A1 (en) 2004-02-19 2005-10-20 Coley Pharmaceutical Gmbh Immunostimulatory viral RNA oligonucleotides
US20070166384A1 (en) 2004-04-09 2007-07-19 Zarraga Isidro Angelo E Methods , composition and preparations for delivery of immune response modifiers
ES2246695B1 (es) 2004-04-29 2007-05-01 Instituto Cientifico Y Tecnologico De Navarra, S.A. Composicion estimuladora de la respuesta inmunitaria que comprende nanoparticulas a base de un copolimero de metil vinil eter y anhidrido maleico.
WO2005108425A1 (en) 2004-05-10 2005-11-17 Cytos Biotechnology Ag Il-23 p19 antigen array and uses thereof
JP2008509654A (ja) 2004-06-01 2008-04-03 イノジェネティックス・ナムローゼ・フェンノートシャップ C型肝炎ウイルスに対するctlおよび/またはhtl応答を誘導するためのペプチド
PT1767216E (pt) 2004-06-11 2012-08-27 Riken Fármaco tendo um ligando de células reguladoras contido num lipossoma
WO2006080951A2 (en) 2004-07-01 2006-08-03 Yale University Targeted and high density drug loaded polymeric materials
US20080206270A1 (en) 2004-07-08 2008-08-28 Minev Boris R Enhancing Class I Antigen Presentation With Synthetic Sequences
US8017151B2 (en) 2004-09-07 2011-09-13 Board Of Regents Of The University Of Nebraska By And Behalf Of The University Of Nebraska Medical Center Amphiphilic polymer-protein conjugates and methods of use thereof
WO2006031878A2 (en) 2004-09-14 2006-03-23 Novartis Vaccines And Diagnostics Inc. Imidazoquinoline compounds
CN1692943A (zh) 2004-09-17 2005-11-09 四川大学 CpG DNA分子抗感染免疫制剂的制备和应用
US9079765B2 (en) 2004-10-01 2015-07-14 Midatech Ltd. Nanoparticles comprising antigens and adjuvants, and immunogenic structures
EP1812864A2 (en) 2004-10-07 2007-08-01 Emory University Multifunctional nanoparticles conjugates and their use
MY159370A (en) 2004-10-20 2016-12-30 Coley Pharm Group Inc Semi-soft-class immunostimulatory oligonucleotides
WO2006045796A2 (en) 2004-10-25 2006-05-04 Cytos Biotechnology Ag Gastric inhibitory polypeptide (gip) antigen arrays and uses thereof
WO2006045849A1 (en) 2004-10-29 2006-05-04 Cytos Biotechnology Ag T-cadherin antigen arrays and uses thereof
US9492400B2 (en) 2004-11-04 2016-11-15 Massachusetts Institute Of Technology Coated controlled release polymer particles as efficient oral delivery vehicles for biopharmaceuticals
US9789171B2 (en) 2004-11-05 2017-10-17 The General Hospital Corporation Anti-fugetactic agents for the treatment of ovarian cancer
WO2007013893A2 (en) 2004-11-15 2007-02-01 Novartis Vaccines And Diagnostics Inc. Immunogenic compositions containing anthrax antigen, biodegradable polymer microparticles, and polynucleotide-containing immunological adjuvant
US20060111271A1 (en) 2004-11-24 2006-05-25 Cerny Erich H Active and passive immunization against pharmacologically active hapten molecules using a synthetic carrier compound composed of similar elements
US20070292386A9 (en) 2004-12-02 2007-12-20 Campbell Robert L Vaccine formulations for intradermal delivery comprising adjuvants and antigenic agents
AU2005315658A1 (en) 2004-12-13 2006-06-22 Cytos Biotechnology Ag IL-15 antigen arrays and uses thereof
JP2008523157A (ja) 2004-12-14 2008-07-03 アルナイラム ファーマシューティカルズ インコーポレイテッド MLL−AF4のRNAi調節およびその使用方法
US20060257359A1 (en) 2005-02-28 2006-11-16 Cedric Francois Modifying macrophage phenotype for treatment of disease
CN101189339A (zh) 2005-03-22 2008-05-28 美得思达健康有限公司 诊断和治疗心血管疾病的递送系统和方法
US7709001B2 (en) 2005-04-08 2010-05-04 Wyeth Llc Multivalent pneumococcal polysaccharide-protein conjugate composition
US20080305161A1 (en) 2005-04-13 2008-12-11 Pfizer Inc Injectable depot formulations and methods for providing sustained release of nanoparticle compositions
US20060241076A1 (en) 2005-04-26 2006-10-26 Coley Pharmaceutical Gmbh Modified oligoribonucleotide analogs with enhanced immunostimulatory activity
WO2006117217A2 (de) 2005-05-04 2006-11-09 Noxxon Pharma Ag Neue verwendung von spiegelmeren
JP2008540558A (ja) 2005-05-10 2008-11-20 エモリー・ユニバーシティ ミセルおよび粒子を用いた、活性物質の送達のための新規の戦略
CA2614049C (en) 2005-07-06 2014-12-02 Molly S. Shoichet Method of biomolecule immobilization on polymers using click-type chemistry
CN101287507B (zh) 2005-08-12 2012-12-05 刘江 用于淋巴靶向的方法和装置
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
CA2630738C (en) 2005-11-25 2013-09-03 Coley Pharmaceutical Gmbh Immunostimulatory oligoribonucleotides
EP1954252B1 (en) 2005-12-02 2016-02-03 GlaxoSmithKline Biologicals SA Nanoparticles for use in immunogenic compositions
EP1973608A1 (en) 2005-12-14 2008-10-01 Cytos Biotechnology AG Immunostimulatory nucleic acid packaged particles for the treatment of hypersensitivity
US9267937B2 (en) 2005-12-15 2016-02-23 Massachusetts Institute Of Technology System for screening particles
US7842312B2 (en) 2005-12-29 2010-11-30 Cordis Corporation Polymeric compositions comprising therapeutic agents in crystalline phases, and methods of forming the same
EP1986685A4 (en) 2006-01-31 2009-02-25 Medivas Llc COMPOSITIONS FOR DISPENSING A VACCINATE AND METHOD FOR THE APPLICATION THEREOF
US8021689B2 (en) 2006-02-21 2011-09-20 Ecole Polytechnique Federale de Lausanne (“EPFL”) Nanoparticles for immunotherapy
EP1991204A2 (en) 2006-02-24 2008-11-19 Novartis AG Microparticles containing biodegradable polymer and cationic polysaccharide for use in immunogenic compositions
EP2010530A2 (en) 2006-03-23 2009-01-07 Novartis AG Methods for the preparation of imidazole-containing compounds
EP2007435B1 (en) 2006-03-31 2019-12-18 Massachusetts Institute Of Technology System for targeted delivery of therapeutic agents
US20100247653A1 (en) 2006-04-11 2010-09-30 Hans Lautenschlager Nanoparticles containing nicotine and/or cotinine, dispersions, and use thereof
CA2652280C (en) 2006-05-15 2014-01-28 Massachusetts Institute Of Technology Polymers for functional particles
WO2007137117A2 (en) 2006-05-17 2007-11-29 Massachusetts Institute Of Technology Aptamer-directed drug delivery
CA2655108C (en) 2006-06-12 2019-05-07 Cytos Biotechnology Ag Processes for packaging oligonucleotides into virus-like particles of rna bacteriophages
US20080014281A1 (en) 2006-06-16 2008-01-17 Florida Atlantic University Chitin Micro-Particles As An Adjuvant
US20100028381A1 (en) 2006-06-19 2010-02-04 3M Innovative Properties Company Formulation for delivery of immune response modifiers
WO2007150030A2 (en) 2006-06-23 2007-12-27 Massachusetts Institute Of Technology Microfluidic synthesis of organic nanoparticles
US20100144845A1 (en) 2006-08-04 2010-06-10 Massachusetts Institute Of Technology Oligonucleotide systems for targeted intracellular delivery
WO2008019366A2 (en) 2006-08-07 2008-02-14 Ludwig Institute For Cancer Research Methods and compositions for increased priming of t-cells through cross-presentation of exogenous antigens
CA2657829A1 (en) 2006-08-11 2008-05-29 Panacea Biotec Limited Particles for delivery of active ingredients, process of making and compositions thereof
WO2008033432A2 (en) 2006-09-12 2008-03-20 Coley Pharmaceutical Group, Inc. Immune modulation by chemically modified ribonucleosides and oligoribonucleotides
US20100111973A1 (en) 2006-09-22 2010-05-06 Glenn Dranoff Methods for treating mica-related disorders
EP2077821B1 (en) 2006-10-12 2019-08-14 The University Of Queensland Compositions and methods for modulating immune responses
US20100303723A1 (en) 2006-11-20 2010-12-02 Massachusetts Institute Of Technology Drug delivery systems using fc fragments
CA2671850A1 (en) 2006-12-08 2008-06-19 Massachusetts Institute Of Technology Delivery of nanoparticles and/or agents to cells
WO2008071774A1 (en) 2006-12-14 2008-06-19 Cytos Biotechnology Ag Purification process for coat protein of rna bacteriophages
US20080149123A1 (en) 2006-12-22 2008-06-26 Mckay William D Particulate material dispensing hairbrush with combination bristles
PL2115140T3 (pl) 2007-01-31 2017-03-31 Yu Dodatnio naładowane rozpuszczalne w wodzie proleki 1H-imidazo[4,5-c]chinolino-4-amin i powiązane związki o bardzo wysokich wskaźnikach penetracji przez skórę
CN101790380B (zh) 2007-02-07 2013-07-10 加利福尼亚大学董事会 合成tlr激动剂的缀合物及其应用
US9217129B2 (en) 2007-02-09 2015-12-22 Massachusetts Institute Of Technology Oscillating cell culture bioreactor
WO2008115641A2 (en) 2007-02-15 2008-09-25 Yale University Modular nanoparticles for adaptable vaccines
EP2842570B1 (en) 2007-03-07 2020-05-06 UTI Limited Partnership Compositions and methods for the prevention and treatment of autoimmune conditions
US20100151031A1 (en) 2007-03-23 2010-06-17 Desimone Joseph M Discrete size and shape specific organic nanoparticles designed to elicit an immune response
ES2542058T3 (es) 2007-03-30 2015-07-30 Particle Sciences, Inc. Formulaciones en forma de partículas y usos de las mismas
US20090074828A1 (en) 2007-04-04 2009-03-19 Massachusetts Institute Of Technology Poly(amino acid) targeting moieties
WO2008124634A1 (en) 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Polymer-encapsulated reverse micelles
WO2008124165A2 (en) 2007-04-09 2008-10-16 Chimeros, Inc. Self-assembling nanoparticle drug delivery system
JP2010523656A (ja) 2007-04-12 2010-07-15 エモリー・ユニバーシティ ミセルおよび粒子を用いた、活性物質の送達のための新規の戦略
EP1982729A1 (en) 2007-04-20 2008-10-22 Cytos Biotechnology AG Vaccination Regimen for B-Cell Vaccines
JP6035503B2 (ja) 2007-05-31 2016-11-30 アカデミシュ ジーケンハウス ライデン ハー.オー.デー.エン. ルムク ワクチンで使用するための、子宮頸部悪性腫瘍に浸潤するt細胞によって標的とされるhpvエピトープ
US8778847B2 (en) 2007-06-13 2014-07-15 The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services Immunogenic peptides of influenza virus
KR101699554B1 (ko) 2007-08-15 2017-01-25 서카시아 리미티드 백신용 펩티드
US8394914B2 (en) 2007-08-24 2013-03-12 Board Of Trustees Of Michigan State University Functional polyglycolide nanoparticles derived from unimolecular micelles
CN105770878A (zh) 2007-10-12 2016-07-20 麻省理工学院 疫苗纳米技术
US8815253B2 (en) 2007-12-07 2014-08-26 Novartis Ag Compositions for inducing immune responses
EP2235043A2 (en) 2008-02-01 2010-10-06 Alpha-O Peptides AG Self-assembling peptide nanoparticles useful as vaccines
KR101617790B1 (ko) * 2008-02-26 2016-05-03 아파르나 바이오사이언시스 치료, 진단과 실험적 혼합물들의 운반을 위한 공학적으로 가변된 나노입자 및 치료용 관련 조성물
WO2009108807A1 (en) 2008-02-26 2009-09-03 The Regents Of The University Of California Glycopeptides and methods of making and using them
EP2262489A2 (en) 2008-02-28 2010-12-22 Deutsches Krebsforschungszentrum, Stiftung des öffentlichen Rechts Hollow nanoparticles and uses thereof
WO2009111588A1 (en) 2008-03-04 2009-09-11 Liquidia Technologies, Inc. Immunomodulator particles and methods of treating
WO2010005721A2 (en) 2008-06-16 2010-01-14 Bind Biosciences, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US20090297621A1 (en) 2008-06-03 2009-12-03 Abbott Cardiovascular Systems Inc. Microparticles For The Treatment Of Disease
US8613951B2 (en) 2008-06-16 2013-12-24 Bind Therapeutics, Inc. Therapeutic polymeric nanoparticles with mTor inhibitors and methods of making and using same
WO2010003009A2 (en) 2008-07-01 2010-01-07 Emory University Synergistic induction of humoral and cellular immunity by combinatorial activation of toll-like receptors
US20110280949A1 (en) 2008-08-06 2011-11-17 Padma Malyala Microparticles for use in immunogenic compositions
WO2010018132A1 (en) 2008-08-11 2010-02-18 Smithkline Beecham Corporation Compounds
EA201100114A1 (ru) 2008-08-11 2011-10-31 ГЛАКСОСМИТКЛАЙН ЭлЭлСи Производные пурина для применения в лечении аллергических, воспалительных и инфекционных заболеваний
UA103195C2 (ru) 2008-08-11 2013-09-25 Глаксосмитклайн Ллк Производные пурина для применения в лечении аллергий, воспалительных и инфекционных заболеваний
WO2010018130A1 (en) 2008-08-11 2010-02-18 Smithkline Beecham Corporation Purine derivatives for use in the treatment of allergic, inflammatory and infectious diseases
US20120034155A1 (en) * 2010-08-03 2012-02-09 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Artificial cells
EP2331124A1 (en) 2008-08-15 2011-06-15 Circassia Limited T-cell antigen peptide from allergen for stimulation of il-10 production
US8323696B2 (en) 2008-08-29 2012-12-04 Ecole Polytechnique Federale De Lausanne Nanoparticles for immunotherapy
WO2010039861A2 (en) 2008-09-30 2010-04-08 The Regents Of The University Of Michigan Dendrimer conjugates
WO2010037402A1 (en) 2008-10-02 2010-04-08 Dako Denmark A/S Molecular vaccines for infectious disease
US8343498B2 (en) 2008-10-12 2013-01-01 Massachusetts Institute Of Technology Adjuvant incorporation in immunonanotherapeutics
US8277812B2 (en) 2008-10-12 2012-10-02 Massachusetts Institute Of Technology Immunonanotherapeutics that provide IgG humoral response without T-cell antigen
US8343497B2 (en) 2008-10-12 2013-01-01 The Brigham And Women's Hospital, Inc. Targeting of antigen presenting cells with immunonanotherapeutics
US8591905B2 (en) 2008-10-12 2013-11-26 The Brigham And Women's Hospital, Inc. Nicotine immunonanotherapeutics
WO2010047839A1 (en) 2008-10-25 2010-04-29 Aura Biosciences Modified plant virus particles and uses therefor
EP2413964B1 (en) 2009-04-01 2019-11-13 University of Miami Vaccine compositions and methods of use thereof
EA201101530A1 (ru) 2009-04-21 2012-03-30 Селекта Байосайенсиз, Инк. ИММУНОНАНОТЕРАПИЯ, ОБЕСПЕЧИВАЮЩАЯ Th1-СМЕЩЕННЫЙ ОТВЕТ
GB0907989D0 (en) * 2009-05-08 2009-06-24 Hybrid Systems Ltd Multivalent adjuvant display
EP2435092A2 (en) 2009-05-27 2012-04-04 Selecta Biosciences, Inc. Nanocarriers possessing components with different rates of release
EP2451485B1 (en) 2009-07-07 2016-03-02 The Research Foundation Of State University Of New York Lipidic compositions for induction of immune tolerance
US8653155B2 (en) * 2009-08-13 2014-02-18 Boston Scientific Scimed, Inc. Polymers having lipophilic hydrocarbon and biodegradable polymeric segments
EA201592264A1 (ru) 2009-08-26 2016-08-31 Селекта Байосайенсиз, Инк. Композиции, которые индуцируют хелперное действие т-клеток
ES2780156T3 (es) 2009-12-15 2020-08-24 Pfizer Composiciones terapéuticas de nanopartículas poliméricas con alta temperatura de transición vítrea o copolímeros de alto peso molecular
US20110171248A1 (en) 2010-01-08 2011-07-14 Selecta Biosciences, Inc. Synthetic virus-like particles conjugated to human papillomavirus capsid peptides for use as vaccines
US20110229556A1 (en) 2010-03-19 2011-09-22 Massachusetts Institute Of Technology Lipid-coated polymer particles for immune stimulation
US20110272836A1 (en) 2010-04-12 2011-11-10 Selecta Biosciences, Inc. Eccentric vessels
US20110262491A1 (en) 2010-04-12 2011-10-27 Selecta Biosciences, Inc. Emulsions and methods of making nanocarriers
ES2661978T3 (es) 2010-05-26 2018-04-04 Selecta Biosciences, Inc. Vacunas multivalentes de nanovehículos sintéticos
AU2011291519A1 (en) 2010-08-20 2013-01-24 Selecta Biosciences, Inc. Synthetic nanocarrier vaccines comprising proteins obtained or derived from human influenza A virus hemagglutinin
US20120070493A1 (en) 2010-08-23 2012-03-22 Selecta Biosciences, Inc. Targeted multi-epitope dosage forms for induction of an immune response to antigens
EA201390660A1 (ru) 2010-11-05 2013-11-29 Селекта Байосайенсиз, Инк. Модифицированные никотиновые соединения и связанные способы
WO2012092552A1 (en) 2010-12-30 2012-07-05 Selecta Biosciences, Inc. Synthetic nanocarriers with reactive groups that release biologically active agents
AU2012236937B2 (en) 2011-03-25 2017-06-08 Selecta Biosciences, Inc. Osmotic mediated release synthetic nanocarriers
CN107261154A (zh) 2011-04-29 2017-10-20 西莱克塔生物科技公司 致耐受性合成纳米载体
CN109172819A (zh) 2011-07-29 2019-01-11 西莱克塔生物科技公司 产生体液和细胞毒性t淋巴细胞(ctl)免疫应答的合成纳米载体
US20130058963A1 (en) 2011-09-06 2013-03-07 Selecta Biosciences, Inc. Induced tolerogenic dendritic cells for generating cd8+ regulatory t cells
AU2014262163A1 (en) 2013-05-03 2015-11-19 Selecta Biosciences, Inc. Delivery of immunosuppressants having a specified pharmacodynamic effective-life and antigen for the inducation of immune tolerance
CN112933218A (zh) 2013-06-04 2021-06-11 西莱克塔生物科技公司 非免疫抑制性抗原特异性免疫治疗剂的重复施用
US20150359865A1 (en) 2014-06-17 2015-12-17 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for t-cell-mediated autoimmune disease
US20160220501A1 (en) 2015-02-03 2016-08-04 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers to reduce immune responses to therapeutic proteins
EA201692512A1 (ru) 2014-06-25 2017-07-31 Селекта Байосайенсиз, Инк. Способы и композиции для лечения синтетическими наноносителями и ингибиторами иммунной контрольной точки
EP3189138A4 (en) 2014-09-07 2018-04-18 Selecta Biosciences, Inc. Methods and compositions for attenuating gene editing anti-viral transfer vector immune responses
EP3215192B1 (en) 2014-11-05 2021-02-17 Selecta Biosciences, Inc. Methods and compositions related to synthetic nanocarriers with rapamycin in a stable, super-saturated state
AU2017230891B2 (en) 2016-03-11 2024-04-18 Selecta Biosciences, Inc. Formulations and doses of pegylated uricase
WO2018039465A1 (en) 2016-08-25 2018-03-01 Selecta Biosciences, Inc. Polyester polymer matrices for the delivery of allergens
EP3518956A1 (en) 2016-09-27 2019-08-07 Selecta Biosciences, Inc. Recombinant immunotoxins for use in the treatment of cancer

Cited By (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110223201A1 (en) * 2009-04-21 2011-09-15 Selecta Biosciences, Inc. Immunonanotherapeutics Providing a Th1-Biased Response
US20110020388A1 (en) * 2009-05-27 2011-01-27 Selecta Biosciences, Inc. Targeted synthetic nanocarriers with ph sensitive release of immunomodulatory agents
US8629151B2 (en) 2009-05-27 2014-01-14 Selecta Biosciences, Inc. Immunomodulatory agent-polymeric compounds
US9884112B2 (en) 2009-05-27 2018-02-06 Selecta Biosciences, Inc. Immunomodulatory agent-polymeric compounds
US9006254B2 (en) 2009-05-27 2015-04-14 Selecta Biosciences, Inc. Immunomodulatory agent-polymeric compounds
US20110110965A1 (en) * 2009-08-26 2011-05-12 Selecta Biosciences, Inc. Compositions that induce t cell help
US9764031B2 (en) 2010-05-26 2017-09-19 Selecta Biosciences, Inc. Dose selection of adjuvanted synthetic nanocarriers
US9066978B2 (en) 2010-05-26 2015-06-30 Selecta Biosciences, Inc. Dose selection of adjuvanted synthetic nanocarriers
US9994443B2 (en) 2010-11-05 2018-06-12 Selecta Biosciences, Inc. Modified nicotinic compounds and related methods
US9289477B2 (en) 2011-04-29 2016-03-22 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers to reduce cytotoxic T lymphocyte responses
US10441651B2 (en) 2011-04-29 2019-10-15 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
US9295718B2 (en) 2011-04-29 2016-03-29 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers to reduce immune responses to therapeutic proteins
US11717569B2 (en) 2011-04-29 2023-08-08 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers
US11235057B2 (en) 2011-04-29 2022-02-01 Selecta Biosciences, Inc. Methods for providing polymeric synthetic nanocarriers for generating antigen-specific tolerance immune responses
US9993548B2 (en) 2011-04-29 2018-06-12 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for inducing regulatory B cells
US10004802B2 (en) 2011-04-29 2018-06-26 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for generating CD8+ regulatory T cells
US10420835B2 (en) 2011-04-29 2019-09-24 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for antigen-specific deletion of T effector cells
US9289476B2 (en) 2011-04-29 2016-03-22 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for allergy therapy
US9265815B2 (en) 2011-04-29 2016-02-23 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers
US8652487B2 (en) 2011-04-29 2014-02-18 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for inducing regulatory B cells
US9987354B2 (en) 2011-04-29 2018-06-05 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for antigen-specific deletion of T effector 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
US10933129B2 (en) 2011-07-29 2021-03-02 Selecta Biosciences, Inc. Methods for administering synthetic nanocarriers that generate humoral and cytotoxic T lymphocyte responses
US10314917B2 (en) * 2013-03-15 2019-06-11 The Brigham And Women's Hospital, Inc. Targeted polymeric inflammation-resolving nanoparticles
US20160022835A1 (en) * 2013-03-15 2016-01-28 The Brigham And Women's Hospital, Inc. Targeted Polymeric Inflammation-Resolving Nanoparticles
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
AU2020200065B2 (en) * 2013-05-03 2022-02-17 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers and therapeutic macromolecules for reduced or enhanced pharmacodynamic effects
EP2991629A4 (en) * 2013-05-03 2016-11-16 Selecta Biosciences Inc ADMINISTRATION OF IMMUNOSUPPRESSIVA WITH SPECIFIC PHARMACODYNAMIC EFFECTIVE LIFE AND ANTIGEN FOR INDICATING IMMUNOTOLERANCE
AU2020203312B2 (en) * 2013-05-03 2022-06-16 Selecta Biosciences, Inc. Dosing combinations for reducing undesired humoral immune responses
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
EP2991628A4 (en) * 2013-05-03 2016-11-16 Selecta Biosciences Inc ASSAY ASSOCIATIONS TO REDUCE UNDESIRED HUMOR MEDIATION IMMUNE RESPONSES
AU2019250117B2 (en) * 2013-05-03 2021-10-14 Selecta Biosciences, Inc Local, concomitant administration of tolerogenic synthetic nanocarriers to reduce type I and type IV hypersensitivity
EP2991685A4 (en) * 2013-05-03 2016-11-16 Selecta Biosciences Inc LOCAL ADMINISTRATION, CONCOMITANT OF SYNTHETIC NANOSUPPORTS TOLEROGENIC TO REDUCE TYPE I AND TYPE IV HYPERSENSITIVITY
EP3763359A1 (en) * 2013-05-03 2021-01-13 Selecta Biosciences, Inc. Method and compositions for enhancing cd4+ regulatory t cells
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
US10357483B2 (en) 2013-05-03 2019-07-23 Selecta Biosciences, Inc. Methods comprising dosing combinations for reducing undesired humoral immune responses
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
EP2991630A4 (en) * 2013-05-03 2016-11-23 Selecta Biosciences Inc SYNTHETIC TOLEROGENIC NANOVECTORS TO REDUCE OR PREVENT ANAPHYLAXIS IN RESPONSE TO NON-ALLERGENIC ANTIGEN
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
EP2991631A4 (en) * 2013-05-03 2016-11-16 Selecta Biosciences Inc SYNTHETIC TOLEROGENOUS NANOSUPPORTS AND THERAPEUTIC MACROMOLECULES FOR REDUCED OR ENHANCED PHARMACODYNAMIC EFFECTS
AU2014262165B2 (en) * 2013-05-03 2019-10-31 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers to reduce or prevent anaphylaxis in response to a non-allergenic antigen
AU2014262162B2 (en) * 2013-05-03 2019-11-07 Selecta Biosciences, Inc. Local, concomitant administration of tolerogenic synthetic nanocarriers to reduce type I and type IV hypersensitivity
CN110694077A (zh) * 2013-05-03 2020-01-17 西莱克塔生物科技公司 用于增强cd4+调节性t细胞的方法和组合物
AU2014262164B2 (en) * 2013-05-03 2020-02-27 Selecta Biosciences, Inc. Dosing combinations for reducing undesired humoral immune responses
US10837018B2 (en) 2013-07-25 2020-11-17 Exicure, Inc. Spherical nucleic acid-based constructs as immunostimulatory agents for prophylactic and therapeutic use
US10894963B2 (en) 2013-07-25 2021-01-19 Exicure, Inc. Spherical nucleic acid-based constructs as immunostimulatory agents for prophylactic and therapeutic use
US11883535B2 (en) 2013-12-03 2024-01-30 Northwestern University Liposomal particles, methods of making same and uses thereof
US11957788B2 (en) 2014-06-04 2024-04-16 Exicure Operating Company Multivalent delivery of immune modulators by liposomal spherical nucleic acids for prophylactic or therapeutic applications
US11123294B2 (en) 2014-06-04 2021-09-21 Exicure Operating Company Multivalent delivery of immune modulators by liposomal spherical nucleic acids for prophylactic or therapeutic applications
US20150359865A1 (en) * 2014-06-17 2015-12-17 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for t-cell-mediated autoimmune disease
US10273333B2 (en) * 2014-08-13 2019-04-30 The Regents Of The University Of California Substituted polyesters by thiol-ene modification: rapid diversification for therapeutic protein stabilization
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
US11633422B2 (en) 2014-09-07 2023-04-25 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector immune responses
US11213593B2 (en) 2014-11-21 2022-01-04 Northwestern University Sequence-specific cellular uptake of spherical nucleic acid nanoparticle conjugates
WO2017205338A1 (en) * 2016-05-23 2017-11-30 University Of Miami Compositions for selective humoral responses and methods of use thereof
US11364304B2 (en) 2016-08-25 2022-06-21 Northwestern University Crosslinked micellar spherical nucleic acids
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
US11696954B2 (en) 2017-04-28 2023-07-11 Exicure Operating Company Synthesis of spherical nucleic acids using lipophilic moieties
US11690920B2 (en) 2017-07-13 2023-07-04 Northwestern University General and direct method for preparing oligonucleotide-functionalized metal-organic framework nanoparticles
WO2019035971A1 (en) 2017-08-16 2019-02-21 Bristol-Myers Squibb Company 6-AMINO-7,9-DIHYDRO-8H-PURIN-8-ONE DERIVATIVES AS TOLL 7 RECEPTOR IMMUNOSTIMULATING AGONISTS (TLR7)
WO2019035970A1 (en) 2017-08-16 2019-02-21 Bristol-Myers Squibb Company 6-AMINO-7,9-DIHYDRO-8H-PURIN-8-ONE DERIVATIVES FOR USE AS TOLL 7 RECEPTOR IMMUNOSTIMULATING AGONISTS (TLR7)
WO2019035968A1 (en) 2017-08-16 2019-02-21 Bristol-Myers Squibb Company 6-AMINO-7,9-DIHYDRO-8H-PURIN-8-ONE DERIVATIVES AS TOLL RECEPTOR 7 (TLR7) AGONISTS AS IMMUNOSTIMULANTS
WO2019035969A1 (en) 2017-08-16 2019-02-21 Bristol-Myers Squibb Company TOLL RECEPTOR 7 (TLR7) AGONISTS COMPRISING A TRICYCLIC FRAGMENT, CONJUGATES CONTAINING SAME, RELATED METHODS AND USES THEREOF
WO2019036023A1 (en) 2017-08-16 2019-02-21 Bristol-Myers Squibb Company 6-AMINO-7,9-DIHYDRO-8H-PURIN-8-ONE DERIVATIVES AS IMMUNOSTIMULANT TOLL (TLR7) RECEPTOR 7 AGONISTS
WO2020028610A1 (en) 2018-08-03 2020-02-06 Bristol-Myers Squibb Company 2H-PYRAZOLO[4,3-d]PYRIMIDINE COMPOUNDS AS TOLL-LIKE RECEPTOR 7 (TLR7) AGONISTS AND METHODS AND USES THEREFOR
WO2020028608A1 (en) 2018-08-03 2020-02-06 Bristol-Myers Squibb Company 1H-PYRAZOLO[4,3-d]PYRIMIDINE COMPOUNDS AS TOLL-LIKE RECEPTOR 7 (TLR7) AGONISTS AND METHODS AND USES THEREFOR
CN111122429A (zh) * 2019-12-31 2020-05-08 华南理工大学 纳米纤维素纤化程度的快速检测方法

Also Published As

Publication number Publication date
US10933129B2 (en) 2021-03-02
AU2012290306B2 (en) 2017-08-17
JP2014521687A (ja) 2014-08-28
EP2736537A4 (en) 2015-04-15
KR20140050698A (ko) 2014-04-29
WO2013019658A2 (en) 2013-02-07
US20130039954A1 (en) 2013-02-14
EP2736537A2 (en) 2014-06-04
BR112014002139A2 (pt) 2017-02-21
EA201490381A1 (ru) 2014-06-30
WO2013019669A2 (en) 2013-02-07
AU2019236653A1 (en) 2019-10-17
MX2014001142A (es) 2014-02-27
WO2013019658A3 (en) 2013-04-25
JP2018030837A (ja) 2018-03-01
CN103702687A (zh) 2014-04-02
WO2013019669A3 (en) 2013-07-04
IL273674A (en) 2020-05-31
IL230269B (en) 2020-04-30
WO2013019648A1 (en) 2013-02-07
AU2012290306A1 (en) 2014-01-23
CN109125722A (zh) 2019-01-04
AU2017261562A1 (en) 2017-12-07
US20130028941A1 (en) 2013-01-31
CN109172819A (zh) 2019-01-11
CA2843274A1 (en) 2013-02-07

Similar Documents

Publication Publication Date Title
US20130028857A1 (en) Synthetic nanocarriers comprising polymers comprising multiple immunomodulatory agents
US9764031B2 (en) Dose selection of adjuvanted synthetic nanocarriers
US20120171229A1 (en) Synthetic nanocarriers with reactive groups that release biologically active agents

Legal Events

Date Code Title Description
AS Assignment

Owner name: SELECTA BIOSCIENCES, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GAO, YUN;ALTREUTER, DAVID H.;REEL/FRAME:028994/0132

Effective date: 20120803

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION