WO2011150240A1 - Nanocarrier compositions with uncoupled adjuvant - Google Patents

Nanocarrier compositions with uncoupled adjuvant Download PDF

Info

Publication number
WO2011150240A1
WO2011150240A1 PCT/US2011/038190 US2011038190W WO2011150240A1 WO 2011150240 A1 WO2011150240 A1 WO 2011150240A1 US 2011038190 W US2011038190 W US 2011038190W WO 2011150240 A1 WO2011150240 A1 WO 2011150240A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
adjuvant
synthetic nanocarriers
coupled
tlr
Prior art date
Application number
PCT/US2011/038190
Other languages
French (fr)
Inventor
Robert L. Bratzler
Lloyd Johnston
Grayson B. Lipford
Charles Zepp
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
Priority to CN201180025504.3A priority Critical patent/CN102905728B/en
Priority to BR112012029917A priority patent/BR112012029917A2/en
Priority to EP11787434.7A priority patent/EP2575886A4/en
Priority to KR1020127030841A priority patent/KR20130108983A/en
Priority to EA201291156A priority patent/EA030863B1/en
Priority to JP2013512250A priority patent/JP6371058B2/en
Application filed by Selecta Biosciences, Inc. filed Critical Selecta Biosciences, Inc.
Priority to AU2011258147A priority patent/AU2011258147B2/en
Priority to CA2798739A priority patent/CA2798739A1/en
Priority to MX2012013713A priority patent/MX2012013713A/en
Publication of WO2011150240A1 publication Critical patent/WO2011150240A1/en
Priority to IL222722A priority patent/IL222722B/en
Priority to AU2017201082A priority patent/AU2017201082A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/7115Nucleic acids or oligonucleotides having modified bases, i.e. other than adenine, guanine, cytosine, uracil or thymine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/06Aluminium, calcium or magnesium; Compounds thereof, e.g. clay
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0013Therapeutic immunisation against small organic molecules, e.g. cocaine, nicotine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/35Allergens
    • 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
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • 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
    • A61K47/02Inorganic compounds
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/22Heterocyclic compounds, e.g. ascorbic acid, tocopherol or pyrrolidones
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/24Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing atoms other than carbon, hydrogen, oxygen, halogen, nitrogen or sulfur, e.g. cyclomethicone or phospholipids
    • 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
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • 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
    • A61K47/58Medicinal 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 by reactions only involving carbon-to-carbon unsaturated bonds, e.g. poly[meth]acrylate, polyacrylamide, polystyrene, polyvinylpyrrolidone, polyvinylalcohol or polystyrene sulfonic acid resin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/593Polyesters, e.g. PLGA or polylactide-co-glycolide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal 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
    • A61K47/646Drug-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 the entire peptide or protein drug conjugate elicits an immune response, e.g. conjugate vaccines
    • 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
    • A61K47/68Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • A61K47/6931Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5146Organic macromolecular compounds; Dendrimers obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyamines, polyanhydrides
    • A61K9/5153Polyesters, e.g. poly(lactide-co-glycolide)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/34Tobacco-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/36Opioid-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • 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
    • A61P31/20Antivirals for DNA 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
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • 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
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • A61K2039/541Mucosal route
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • A61K2039/541Mucosal route
    • A61K2039/542Mucosal route oral/gastrointestinal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • A61K2039/541Mucosal route
    • A61K2039/543Mucosal route intranasal
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • 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
    • 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
    • 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
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • 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
    • 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
    • A61K2039/55561CpG containing adjuvants; Oligonucleotide containing adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6087Polysaccharides; Lipopolysaccharides [LPS]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6093Synthetic polymers, e.g. polyethyleneglycol [PEG], Polymers or copolymers of (D) glutamate and (D) lysine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/62Medicinal preparations containing antigens or antibodies characterised by the link between antigen and carrier
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/70Multivalent vaccine
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2730/00Reverse transcribing DNA viruses
    • C12N2730/00011Details
    • C12N2730/10011Hepadnaviridae
    • C12N2730/10111Orthohepadnavirus, e.g. hepatitis B virus
    • C12N2730/10134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein
    • 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

Definitions

  • This invention relates to synthetic nanocarrier and separate adjuvant compositions, and related methods, such as for treating diseases in which generating an immune response is desirable.
  • Adjuvants are generally important components for the majority of currently used vaccination regimens.
  • the optimal approach for augmenting the immune response with adjuvant, however, in a number of cases, is not yet known. Therefore, improved
  • compositions and therapeutic methods are needed to provide improved therapies for diseases in which generating an immune response and/or augmenting it is desirable.
  • composition comprising a dosage form that comprises (1) a population of synthetic nanocarriers, (2) a first adjuvant that is not coupled to any synthetic nanocarriers, and (3) a pharmaceutically acceptable excipient is provided.
  • a method comprising administering the composition to a subject is provided.
  • compositions provided herein, including those of the methods provided comprise a systemic dose of the first adjuvant.
  • said compositions further comprise a second adjuvant.
  • the first adjuvant and second adjuvant are different.
  • the first adjuvant and the second adjuvant are the same.
  • the compositions provided herein, including those of the methods provided comprise a systemic dose of the first adjuvant and/or second adjuvant.
  • the systemic dose results in the systemic release of TNF-oc, IL-6 and/or IL-12.
  • the systemic dose results in the systemic release of IFN- ⁇ , IL-12 and/or IL-18.
  • the second adjuvant of the compositions provided is coupled to the synthetic nanocarriers.
  • the second adjuvant is not coupled to any synthetic nanocarriers.
  • the second adjuvant is coupled to another population of synthetic nanocarriers.
  • compositions provided herein, including those of the methods provided comprise one or more antigens.
  • the one or more antigens are coupled to the synthetic nanocarriers.
  • the one or more antigens are coupled to another population of synthetic nanocarriers.
  • the one or more antigens are not coupled to any synthetic nanocarriers.
  • the one or more antigens of the compositions provided, including those of the methods provided comprise a B cell antigen and/or a T cell antigen.
  • the T cell antigen is a universal T cell antigen or T helper cell antigen.
  • the one or more antigens comprise a B cell antigen and/or a T cell antigen and a universal T cell antigen or T helper cell antigen.
  • the B cell antigen is nicotine.
  • the compositions provided, including those of the methods provided do not comprise an antigen.
  • the first adjuvant and/or second adjuvant comprises a mineral salt, gel-type adjuvant, a microbial adjuvant, an oil-emulsion or emulsifier-based adjuvant, a particulate adjuvant, a synthetic adjuvant, a phosphate adjuvant, a polymer, a liposome, a microcarrier, an immunostimulatory nucleic acid, alum, a saponin, an interleukin, an interferon, a cytokine, a toll-like receptor (TLR) agonist, an imidazoquinoline, a cytokine receptor agonist, a CD40 agonist, an Fc receptor agonist, a complement receptor agonist, QS21, vitamin E, squalene, tocopherol, Quil A, ISCOMs, ISCOMATRIX, Ribi Detox, CRL-1005, L-121,
  • the immunostimulatory nucleic acid comprises a CpG-containing nucleic acid.
  • the imidazoquinoline comprises resiquimod or imiquimod.
  • the first and/or second adjuvant comprises alum.
  • the second adjuvant when the first adjuvant comprises a CpG-containing nucleic acid, the second adjuvant comprises an imidazoquinoline or alum. In another embodiment, the imidazoquinoline is resiquimod. In still another embodiment, when the first adjuvant comprises an imidazoquinoline, the second adjuvant comprises a CpG-containing nucleic acid or alum. In a further embodiment, the imidazoquinoline is resiquimod. In one embodiment, when the first adjuvant comprises alum, the second adjuvant comprises an imidazoquinoline or a CpG-containing nucleic acid. In another embodiment, the
  • the TLR agonist comprises a TLR-1, TLR-2, TLR-3, TLR-4, TLR- 5, TLR-6, TLR-7, TLR-8, TLR-9, TLR-10, TLR-11 agonist or a combination thereof.
  • the first adjuvant and/or second adjuvant does not comprise a TLR agonist.
  • the first adjuvant and/or second adjuvant does not comprise a TLR-3, TLR-7, TLR-8 or TLR-9 agonist.
  • the second adjuvant is coupled to the synthetic nanocarriers and comprises resiquimod.
  • the synthetic nanocarriers of the compositions provided herein, including those of the methods provided comprise lipid nanoparticles, polymeric
  • nanoparticles metallic nanoparticles, surfactant-based emulsions, dendrimers, buckyballs, nanowires, virus-like particles, peptide or protein particles, nanoparticles that comprise a combination of nanomaterials, spheroidal nanoparticles, cuboidal nanoparticles, pyramidal nanoparticles, oblong nanoparticles, cylindrical nanoparticles or toroidal nanoparticles.
  • the synthetic nanocarriers comprise one or more polymers.
  • the one or more polymers comprise a polyester.
  • the one or more polymers comprise or further comprise a polyester coupled to a hydrophilic polymer.
  • the polyester comprises a poly(lactic acid), poly(glycolic acid), poly(lactic-co-glycolic acid), or polycaprolactone.
  • the hydrophilic polymer comprises a polyether.
  • the polyether comprises polyethylene glycol.
  • the one or more antigens of the compositions provided herein, including those of the methods provided comprise nicotine and a universal T cell antigen or T-helper cell antigen, each of which are coupled to the synthetic nanocarriers.
  • the universal T cell antigen or T helper cell antigen of the compositions provided herein, including those of the methods provided, is coupled by encapsulation.
  • the T-helper cell antigen comprises a peptide obtained or derived from ovalbumin.
  • the peptide obtained or derived from ovalbumin comprises the sequence as set forth in SEQ ID NO: 1.
  • compositions provided herein comprising administering any of the compositions provided herein to a subject.
  • the subject is a human.
  • a method comprising administering any of the compositions provided and a second adjuvant to a subject, wherein the second adjuvant is administered at a time different from the administration of the composition.
  • the subject is a human.
  • the composition and second adjuvant are coadministered.
  • the composition and second adjuvant are not coadministered.
  • the second adjuvant is administered prior to the composition.
  • any of the methods provided further comprises administering one or more antigens.
  • any of the compositions provided, including those of the methods provided further comprises one or more antigens.
  • the one or more antigens are coadministered.
  • the subject of any of the methods provided or to which any of the compositions provided is administered is in need of an inflammatory response.
  • the subject is in need of a Thl immune response.
  • the subject is in need of a humoral immune response.
  • the subject is in need of a specific local cytotoxic T lymphocyte response.
  • 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 atopic condition, asthma, COPD or a chronic infection.
  • any of the compositions can be for use in therapy or prophylaxis. In another embodiment, any of the compositions can be for use in any of the methods provided. In yet another embodiment, any of the compositions can be for use in a method of inducing an inflammatory response in a subject. In still another embodiment, any of the compositions can be for use in a method of inducing a Thl immune response in a subject. In yet another embodiment, any of the compositions can be for use in a method of inducing a humoral immune response in a subject. In a further embodiment, any of the compositions can be for use in a method of inducing a specific local cytotoxic T lymphocyte response in a subject.
  • any of the compositions can be for use in a method of treating or preventing cancer. In yet a further embodiment, any of the compositions can be for use in a method of treating or preventing infection or infectious disease. In another embodiment, any of the compositions can be for use in a method of treating or preventing an atopic condition, asthma, COPD or a chronic infection.
  • compositions provided for the manufacture of a medicament for use in any of the methods provided is provided herein.
  • Fig. 1 shows the systemic cytokine production in mice after nanocarrier (NC) inoculation.
  • Sera from groups of three mice were pooled and analyzed by ELISA.
  • Fig. 2 demonstrates the systemic IFN- ⁇ production in mice after NC inoculation. Sera from groups of three mice were pooled and analyzed by ELISA.
  • Fig. 3 demonstrates that utilization of entrapped R848 within NCs generates an immune response, which is superior to one induced by NC without R848.
  • Fig. 4 shows anti-nicotine antibody titers in mice immunized with NC containing surface nicotine and T-helper ovalbumin-derived peptide OP-II (NC-Nic) without adjuvant or with the same NC-Nic admixed with 20 ⁇ g of free R848 (5 animals/group; s.c, 100 ⁇ g of NC per injection, 3 times with 4-wk intervals).
  • Fig. 5 shows anti-nicotine antibody titers in mice immunized with NC containing surface nicotine and T-helper ovalbumin-derived peptide OP-II (NC-Nic) with R848 adjuvant or with the same NC-Nic admixed with 80 ⁇ g of free alum or 25 ⁇ g of free CpG-1826 (5 animals/group; s.c, 100 ⁇ g of NC per injection, 3 times with 4-wk intervals).
  • Fig. 6 shows anti-nicotine antibody titers in mice immunized with NC containing surface nicotine, R848 and T-helper ovalbumin-derived peptide OP-II NC[Nic,R848, OP-II] or with the same NC-Nic admixed with 80 ⁇ g of free alum (5 animals/group; s.c, 100 ⁇ g of NC per injection, 3 times with 4-wk intervals). Titers for days 40 and 70 after the first immunization are shown (ELIS A against polylysine-nicotine) (group 1 : immunized with NC[Nic,R848,OP-II] (3.1% of R848, 1.5% of OP-II); group 2: immunized with
  • Fig. 7 shows specific local CTL response in mice immunized with NC containing ovalbumin or free ovalbumin. Mice were immunized once (s.c, 100 ⁇ g of NC, containing 2.8% of OVA, or with 2.5 ⁇ g of OVA; both immunogens admixed with 10 ⁇ g of free 1826- CpG).
  • Fig. 8 shows anti-nicotine antibody titers in mice immunized with NC containing surface nicotine and T-helper ovalbumin-derived peptide OP-II (NC-Nic) (no adjuvant within NC) admixed with 20 ⁇ g of free CpG (PS) or 20 ⁇ g of free CpG (PO) (5 animals/group; s.c, 100 ⁇ g of NC per injection, 3 times with 2-wk intervals). Control mice received PBS alone. Titers for days 26 and 40 are shown (ELIS A against polylysine-nicotine) (group 1 :
  • Fig. 9 shows anti-ovalbumin (OVA) antibody titers in mice immunized with NC containing surface OVA (NC-OVA) (no adjuvant within NC) admixed with 20 ⁇ g of free R848 or CpG (PS) (5 animals/group; s.c, 100 ⁇ g of NC per injection, 3 times with 2-wk intervals).
  • NC-OVA NC containing surface OVA
  • PS CpG
  • Fig. 10 shows anti-nicotine antibody titers in mice injected with CpG (20 ⁇ g per injection, 2 times with 2-wk intervals) followed by immunization at day 35 with NC containing surface nicotine and T-helper ovalbumin-derived peptide OP-II (NC-Nic) either with or without NC-contained R848 (5 animals/group; s.c, 100 ⁇ g of NC per injection, 2 times with 2-wk intervals).
  • compositions and methods that relate to a composition comprising a dosage form that comprises (1) a population of synthetic nanocarriers, (2) a first adjuvant that is not coupled to any synthetic nanocarriers, and (3) a pharmaceutically acceptable excipient.
  • the administration of adjuvant separate from synthetic nanocarriers leads to a rapid and strong systemic induction of pro-inflammatory cytokines, such as TNF-a, IL-6 and/or IL-12.
  • the dose of the adjuvant or adjuvants in the compositions in some embodiments, therefore, is a systemic dose.
  • the systemic dose results in the release of TNF-a, IL-6 or IL-12.
  • the systemic dose results in the systemic release of TNF-a, IL-6 and IL-12.
  • the administration of compositions provided herein can be beneficial to subjects where an inflammatory response is desired.
  • the compositions provided are administered to such subjects.
  • such subjects have or are at risk of having cancer.
  • such subjects have or are at risk of having an infection or an infectious disease. Methods for the administration of the compositions to such subjects are also provided.
  • the administration of adjuvant separate from synthetic nanocarriers leads to a rapid and strong systemic induction of cytokines that are important for a Thl immune response, such as IFN- ⁇ , IL-12 and/or IL-18. Therefore, the dose of the adjuvant or adjuvants in the compositions in some embodiments is a systemic dose that results in the systemic release of IFN- ⁇ , IL-12 and/or IL-18. As such cytokines are important for a Thl immune response, the administration of compositions provided herein can be beneficial to subjects where a Thl immune response is desired. In some embodiments, the compositions provided are administered to such subjects. In embodiments, such subjects have or are at risk of having an atopic condition, asthma, chronic obstructive pulmonary disease (COPD) or a chronic infection. Methods for the administration of the compositions to such subjects are also provided.
  • COPD chronic obstructive pulmonary disease
  • the inventors have also unexpectedly discovered that it is possible to administer a second adjuvant with the aforementioned compositions to provide a strong humoral response.
  • the aforementioned compositions can further comprise a second adjuvant.
  • the second adjuvant is coupled to the synthetic nanocarriers.
  • the second adjuvant is not coupled to any synthetic nanocarriers.
  • the second adjuvant is coupled to another population of synthetic nanocarriers.
  • the second adjuvant is administered to a subject at a time different from when the composition that comprises a population of synthetic nanocarriers and a first adjuvant that is not coupled to any synthetic nanocarriers is administered.
  • the second adjuvant is administered at a different time but is coadministered. In other embodiments, the second adjuvant is not coadministered. In still other embodiments, the second adjuvant is administered prior to or after the administration of the composition that comprises a population of synthetic nanocarriers and a first adjuvant that is not coupled to any synthetic nanocarriers. In some embodiments, the second adjuvant is also not coupled to any synthetic nanocarriers. In other embodiments, the second adjuvant is coupled to another population of synthetic nanocarriers.
  • the administration of one or more antigens with the compositions provided above provides a strong specific local cytotoxic T lymphocyte (CTL) response.
  • the antigen(s) are coadministered with the compositions provided.
  • the antigen(s) are coupled to the synthetic nanocarriers.
  • the antigen(s) are not coupled to the synthetic nanocarriers but to another population of synthetic nanocarriers.
  • the antigen(s) can comprise a B cell or T cell antigen.
  • the T cell antigen is a T helper cell antigen.
  • the antigen(s) comprise a B cell or T cell antigen as well as a T helper cell antigen. Therefore, the compositions provided can be beneficial to subjects where a specific local CTL response is desired.
  • the compositions provided are administered to such subjects. Methods for the administration of the compositions to such subjects are also provided.
  • 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), 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®), AS15, liposomes and liposomal formulations such as AS01, synthesized or specifically prepared microparticles and microcarriers such as bacteria-derived outer membrane vesicles (OMV) of N.
  • 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+squalen
  • 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.
  • 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 imidazoquinolines; such as R848; adenine derivatives, such as those disclosed in US patent 6,329,381 (Sumitomo
  • synthetic nanocarriers incorporate as adjuvants compounds that are agonists for toll-like receptors (TLRs) 7 & 8 ("TLR 7/8 agonists").
  • TLR 7/8 agonists are agonists for toll-like receptors (TLRs) 7 & 8
  • TLR 7/8 agonists are the TLR 7/8 agonist compounds disclosed in US Patent 6,696,076 to Tomai et al., including but not limited to imidazoquinoline amines, imidazopyridine amines, 6,7-fused
  • 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 to stimulate immunity rather than tolerance, a synthetic nanocarrier
  • adjuvants 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.
  • adjuvants also may comprise immuno stimulatory RNA molecules, such as but not limited to dsRNA, poly I:C or poly Lpoly C12U (available as
  • an adjuvant may be a TLR-4 agonist, such as bacterial lipopolysacccharide (LPS), VSV-G, and/or HMGB-1.
  • adjuvants may comprise TLR-5 agonists, such as flagellin, or portions or derivatives thereof, including but not limited to those disclosed in US Patents 6,130,082, 6,585,980, and 7,192,725.
  • synthetic nanocarriers incorporate a ligand for Toll-like receptor (TLR)-9, such as
  • immuno stimulatory DNA molecules comprising CpGs, which induce type I interferon secretion, and stimulate T and B cell activation leading to increased antibody production and cytotoxic T cell responses
  • adjuvants may be proinflammatory stimuli released from necrotic cells (e.g., urate crystals).
  • adjuvants may be activated components of the complement cascade (e.g., CD21, CD35, etc.).
  • adjuvants may be activated components of immune complexes.
  • the adjuvants also include complement receptor agonists, such as a molecule that binds to CD21 or CD35.
  • the complement receptor agonist induces endogenous complement
  • adjuvants are cytokines, which are small proteins or biological factors (in the range of 5 kD - 20 kD) that are released by cells and have specific effects on cell-cell interaction, communication and behavior of other cells.
  • the cytokine receptor agonist is a small molecule, antibody, fusion protein, or aptamer.
  • the dose of adjuvant is not coupled to any synthetic nanocarriers, preferably, all of the dose of adjuvant is not coupled to any synthetic nanocarriers.
  • the dose of adjuvant comprises two or more types of adjuvants, and at least a portion of at least one of the types of adjuvant is not coupled to any synthetic nanocarriers.
  • adjuvants that act on different receptors such as different TLR receptors, may be combined.
  • a TLR 7/8 agonist may be combined with a TLR 9 agonist.
  • a TLR 7/8 agonist may be combined with a TLR 4 agonist.
  • a TLR 9 agonist may be combined with a TLR 3 agonist.
  • administering means providing a substance (e.g., 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 immune response to an allergen (i.e., allergic reaction).
  • allergen i.e., allergic reaction
  • Allergies or allergic conditions 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.
  • Amount effective is any amount of a composition provided herein that produces 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 of an inflammatory, a Thl, a humoral or specific local CTL immune response. Such subjects include those that have or are at risk of having cancer, an infection or infectious disease, an atopic condition, asthma, chronic obstructive pulmonary disease (COPD) or a chronic infection.
  • COPD chronic obstructive pulmonary disease
  • Amounts effective include those that involve the systemic release of one or more cytokines. In embodiments, the amounts effective include those that involve the production of a systemic cytokine release profile. In some embodiments, the one or more cytokines or cytokine release profile comprises the systemic release of TNF-oc, IL-6 and/or IL-12. In other embodiments, the one or more cytokines or cytokine release profile comprises the systemic release of IFN- ⁇ , IL-12 and/or IL-18. This can be monitored by routine methods. An amount that is effective to produce one or more desired immune responses 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,
  • 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 ,
  • the doses of the compositions provided are systemic doses.
  • 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. In embodiments, antigens are coadministered with the synthetic nanocarriers. In other embodiments, antigens are not coadministered with the synthetic nanocarriers. "Type(s) of antigens” means molecules that share the same, or substantially the same, antigenic characteristics. In embodiments, antigens of the compositions provided are associated with the disease or condition that is being treated.
  • the antigen can be an allergen (for the treatment of an allergy or allergic condition), a cancer-associated antigen (for the treatment of cancer or a tumor), an infectious agent antigen (for the treatment of an infection, an infectious disease or a chronic infectious disease), etc.
  • At least a portion of the dose means at least some part of the dose, ranging up to including all of the dose.
  • B cell antigen means any antigen that is recognized by a B cell, and triggers an immune response in a B cell (e.g., an antigen that is specifically recognized by a B cell receptor on a B cell).
  • 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, 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. In some embodiments, the B cell antigen comprises a glycoprotein or glycopeptide associated with an infectious agent.
  • the infectious agent can be a bacterium, virus, fungus, protozoan, parasite or prion.
  • the B cell antigen comprises a poorly immunogenic antigen. In some embodiments, the B cell antigen comprises an abused substance or a portion thereof. In some embodiments, 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 source, or a pollutant.
  • the B cell antigen may also comprise a hazardous environmental agent.
  • 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, an addictive substance, a xenoantigen, or a metabolic disease enzyme or enzymatic product thereof.
  • Coadministered means administering two or more substances to a subject in a manner that is correlated in time, preferably sufficiently correlated in time so as to provide a modulation in an immune response.
  • coadministration may occur through administration of two or more substances in the same dosage form.
  • coadministration may encompass administration of two or more substances in different dosage forms, but within a specified period of time, preferably within 1 month, more preferably within 1 week, still more preferably within 1 day, and even more preferably within 1 hour.
  • 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.
  • at least a portion of a dose of adjuvant(s) is not coupled to any synthetic nanocarriers, preferably all of a dose of adjuvant(s) is not coupled to any synthetic
  • Derived means taken from a source and subjected to substantial modification. For instance, a peptide or nucleic acid with a sequence with only 50% identity to a natural peptide or nucleic acid, preferably a natural consensus peptide or nucleic acid, would be said to be derived from the natural peptide or nucleic acid. Substantial modification is modification that significantly affects the chemical or immunological properties of the material in question. Derived peptides and nucleic acids can also include those with a sequence with greater than 50% identity to a natural peptide or nucleic acid sequence if said derived peptides and nucleic acids have altered chemical or immunological properties as compared to the natural peptide or nucleic acid. These chemical or immunological properties comprise hydrophilicity, stability, affinity, and ability to couple with a carrier such as a synthetic nanocarrier.
  • 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 within a synthetic nanocarrier, preferably enclose completely within a synthetic nanocarrier. Most or all of a substance that is encapsulated is not exposed to the local environment external to the synthetic nanocarrier. 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.
  • Human response means any immune response that results in the production or stimulation of B cells and/or the production of antibodies.
  • the humoral immune response is specific to an antigen comprised within an inventive composition or administered during the practice of an inventive method.
  • Methods for assessing whether a humoral response is induced are known to those of ordinary skill in the art. Examples of such methods are provided below in the Examples.
  • An "infection” or “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 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
  • Fasciolopsiasis Fasciolopsiasis, Filariasis, Free-living amebic infection, Giardiasis, Gnathostomiasis,
  • Hymenolepiasis Isosporiasis, Kala-azar, Leishmaniasis, Malaria, Metagonimiasis, Myiasis, Onchocerciasis, Pediculosis, Pinworm Infection, Scabies, Schistosomiasis, Taeniasis, Toxocariasis, Toxoplasmosis, Trichinellosis, Trichinosis, Trichuriasis, Trichomoniasis and Trypanosomiasis; fungal infectious disease, such as Aspergillosis, Blastomycosis,
  • “Inflammatory response” means any immune response involved in the body's innate immune defense system that operates in response to, for example, exposure to an infectious agent, cell injury, etc.
  • the inflammatory response includes the systemic release of cytokines, such as TNF-oc, IL-6 and/or IL-12.
  • Methods for assessing whether an inflammatory response is induced, such as an assessment of the production of pro- inflammatory cytokines, are known to those of ordinary skill in the art. Examples of such methods are provided below in the Examples.
  • isolated nucleic acid means a nucleic acid that is separated from its native environment and present in sufficient quantity to permit its identification or use.
  • An isolated nucleic acid may be one that is (i) amplified in vitro by, for example, polymerase chain reaction (PCR); (ii) recombinantly produced by cloning; (iii) purified, as by cleavage and gel separation; or (iv) synthesized by, for example, chemical synthesis.
  • PCR polymerase chain reaction
  • recombinantly produced by cloning recombinantly produced by cloning
  • purified as by cleavage and gel separation
  • synthesized by, for example, chemical synthesis synthesized by, for example, chemical synthesis.
  • An isolated nucleic acid is one which is readily manipulable by recombinant DNA techniques well known in the art.
  • nucleotide sequence contained in a vector in which 5' and 3' restriction sites are known or for which polymerase chain reaction (PCR) primer sequences have been disclosed is considered isolated but a nucleic acid sequence existing in its native state in its natural host is not.
  • An isolated nucleic acid may be substantially purified, but need not be.
  • a nucleic acid that is isolated within a cloning or expression vector is not pure in that it may comprise only a tiny percentage of the material in the cell in which it resides.
  • Such a nucleic acid is isolated, however, as the term is used herein because it is readily manipulable by standard techniques known to those of ordinary skill in the art. Any of the nucleic acids provided herein may be isolated.
  • the antigens in the compositions provided herein are present in the form of an isolated nucleic acid, such as an isolated nucleic acid that encodes an antigenic peptide, polypeptide or protein.
  • isolated peptide, polypeptide or protein means the polypeptide (or peptide or protein) is separated from its native environment and present in sufficient quantity to permit its identification or use. This means, for example, the polypeptide (or peptide or protein) may be (i) selectively produced by expression cloning or (ii) purified as by chromatography or electrophoresis. Isolated peptides, proteins or polypeptides may be, but need not be, substantially pure. Because an isolated peptide, polypeptide or protein may be admixed with a pharmaceutically acceptable carrier in a pharmaceutical preparation, the polypeptide (or peptide or protein) may comprise only a small percentage by weight of the preparation.
  • polypeptide or peptide or protein
  • the polypeptide 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 proteins (or peptides or polypeptides). Any of the peptides, polypeptides or proteins provided herein may be isolated.
  • the antigens in the compositions provided herein are in the form of isolated peptides, polypeptides or proteins.
  • “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.
  • a minimum dimension of a synthetic nanocarrier would be substantially identical, and would be the size of its diameter.
  • 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 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 ⁇ .
  • 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 inventive 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 1000:1, still 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 ⁇ , more preferably equal to or less than 2 ⁇ , more preferably equal to or less than 1 ⁇ , 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 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 greater than lOOnm, more preferably equal to or greater than 120nm, 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 sizes is obtained by suspending the synthetic nanocarriers in a liquid (usually aqueous) media and using dynamic light scattering (e.g. using a Brookhaven ZetaPALS instrument).
  • Obtained means taken from a source without substantial modification.
  • Substantial modification is modification that significantly affects the chemical or immunological properties of the material in question.
  • a peptide or nucleic acid with a sequence with greater than 90%, preferably greater than 95%, preferably greater than 97%, preferably greater than 98%, preferably greater than 99%, preferably 100%, identity to a natural peptide or nucleotide sequence, preferably a natural consensus peptide or nucleotide sequence, and chemical and/or immunological properties that are not significantly different from the natural peptide or nucleic acid would be said to be obtained from the natural peptide or nucleotide sequence.
  • These chemical or immunological properties comprise hydrophilicity, stability, affinity, and ability to couple with a carrier such as a synthetic nanocarrier.
  • “Pharmaceutically acceptable carrier or excipient” means a pharmacologically inactive material used together with the recited synthetic nanocarriers to formulate the inventive compositions.
  • Pharmaceutically acceptable carriers or 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.
  • pharmaceutically acceptable carriers or excipients comprise calcium carbonate, calcium phosphate, various diluents, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
  • Specific local cytotoxic T lymphocyte (CTL) response means any stimulation, induction or proliferation of cytotoxic T cells, preferably cytotoxic T cells that are specific to an antigen.
  • the antigen is associated with any of the diseases or conditions provided herein.
  • the antigen is comprised within an inventive composition or is administered in an inventive method provided herein. Methods for assessing CTL response are known to those of skill in the art. An examples of such a method is provided in the Examples.
  • 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
  • the synthetic nanocarriers do not comprise albumin nanoparticles. In embodiments, inventive synthetic nanocarriers do not comprise chitosan.
  • a synthetic nanocarrier can be, but is not limited to, one or a plurality of lipid-based nanoparticles(e.g. liposomes) (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.
  • Synthetic nanocarriers may be a variety of different shapes
  • Synthetic nanocarriers according to the invention comprise one or more surfaces, including but not limited to internal surfaces (surfaces generally facing an interior portion of the synthetic nanocarrier) and external surfaces (surfaces generally facing an external environment of the synthetic nanocarrier).
  • Exemplary synthetic nanocarriers that can be adapted for use in the practice of the present invention comprise: (1) the biodegradable nanoparticles disclosed in US Patent 5,543,158 to Gref et al., (2) the polymeric nanoparticles of Published US Patent Application 20060002852 to Saltzman et al., (3) the lithographically constructed nanoparticles of Published US Patent Application 20090028910 to DeSimone et al., (4) the disclosure of WO 2009/051837 to von Andrian et al., (5) the nanoparticles disclosed in Published US Patent Application 2008/0145441 to Penades et al., (6) the protein nanoparticles disclosed in Published US Patent Application 20090226525 to de los Rios et al., (7) the virus-like
  • 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 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.
  • Systemic dose means a dose of an adjuvant that provides a particular systemic cytokine release, preferably a particular systemic cytokine release profile.
  • the particular systemic cytokine release preferably a particular systemic cytokine release profile, is in a human.
  • the compositions and methods provided herein result in a particular systemic cytokine release profile in a subject.
  • the term “separately” is also used to mean adjuvant that is not coupled to any synthetic nanocarriers.
  • systemic cytokine release profile means a pattern of systemic cytokine release, wherein the pattern comprises cytokine levels measured for several different systemic cytokines.
  • the particular systemic cytokine release profile comprises the systemic release of TNF-oc, IL-6 and/or IL-12.
  • the particular systemic cytokine release profile comprises the systemic release of IFN- ⁇ , IL12 and/or IL-18.
  • 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, polypeptides 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 'universal' T cell antigen, or T cell memory antigen, (i.e., one to which a subject has a pre-existing memory and that can be used to boost T cell help to an unrelated antigen, for example an unrelated B cell antigen).
  • Universal T cell antigens include tetanus toxoid, as well as one or more peptides derived from tetanus toxoid, Epstein-Barr virus, or influenza virus. Universal T cell antigens also include a components of influenza virus, such as hemagglutinin, neuraminidase, or nuclear protein, or one or more peptides derived therefrom.
  • the universal T cell antigen is not one that is presented in a complex with a MHC molecule.
  • the universal T cell antigen is not complexed with a MHC molecule for presentation to a T helper cell. Accordingly, in some embodiments, the universal T cell antigen is not a T helper cell antigen. However, in other embodiments, the universal T cell antigen is a T helper cell antigen.
  • 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. US Patent 7,202,351).
  • a T-helper cell antigen may comprise ovalbumin or a peptide obtained or derived therefrom.
  • the ovalbumin comprises the amino acid sequence as set forth in Accession No. AAB59956, NP . 990483.1, AAA48998, or CAA2371.
  • the peptide obtained or derived from ovalbumin comprises the following amino acid sequence: H-Ile-Ser-Gln-Ala-Val-His-Ala-Ala-His-Ala-Glu-Ile-Asn-Glu-Ala- Gly-Arg-OH (SEQ ID NO: 1).
  • a T-helper cell antigen may comprise one or more lipids, or glycolipids, including but not limited to: a-galactosylceramide (a -
  • GalCer a -linked glycosphingolipids (from Sphingomonas spp.), galactosyl diacylglycerols (from Borrelia burgdorferi), lypophosphoglycan (from Leishmania donovani), and phosphatidylinositol tetramannoside (PEVI4) (from Mycobacterium leprae).
  • PVI4 phosphatidylinositol tetramannoside
  • 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 universal T cell antigen or T-helper cell antigen, may be coupled to, or uncoupled from, a synthetic nanocarrier.
  • the universal T cell antigen or T-helper cell antigen is encapsulated in the synthetic nanocarriers of the inventive compositions.
  • Thl immune response means any immune response that results in the production of Thl cells and Thl-associated cytokines, IFN- ⁇ , IL-12 and/or IL-18, or that counteracts the differentiation of Th2 cells and the action of Th2 cytokines. Methods for assessing whether a Thl immune response is induced are known to those of ordinary skill in the art. Examples of such methods are provided below in the Examples.
  • Time different from administration or "a time different from a time when the composition is administered” means a time more than about 30 seconds either before or after administration, preferably more than about 1 minute either before or after administration, more preferably more than 5 minutes either before or after administration, still more preferably more than 1 day either before or after administration, still more preferably more than 2 days either before or after administration, still more preferably more than 1 week either before or after administration, still more preferably more than 2 weeks either before or after administration, still more preferably more than 3 weeks either before or after administration, still more preferably more than 1 month either before or after administration, and still more preferably more than 2 months either before or after administration.
  • 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 or against an antigen derived from an infectious agent for the treatment of an infection or infectious disease).
  • a vaccine may comprise dosage forms according to the invention.
  • these vaccines comprise an adjuvant not coupled to any synthetic nanocarriers.
  • the inventive compositions incorporate adjuvants that comprise agonists for toll-like receptors (TLRs) 7 & 8 ("TLR 7/8 agonists").
  • TLR 7/8 agonists are the TLR 7/8 agonist compounds disclosed in U.S. Patent 6,696,076 to Tomai et al., including but not limited to imidazoquinoline amines, imidazopyridine amines, 6,7-fused
  • Preferred adjuvants comprise imiquimod and R848.
  • the inventive compositons incorporate adjuvants that comprise a ligand for Toll-like receptor (TLR)-9, such as immunostimulatory DNA molecules comprising CpGs, which induce type I interferon secretion, and stimulate T and B cell activation leading to increased antibody production and cytotoxic T cell responses (Krieg et al., CpG motifs in bacterial DNA trigger direct B cell activation. Nature. 1995. 374:546- 549; Chu et al. CpG oligodeoxynucleotides act as adjuvants that switch on T helper 1 (Thl) immunity. J. Exp. Med. 1997. 186:1623-1631; Lipford et al.
  • TLR Toll-like receptor
  • CpG-containing synthetic oligonucleotides promote B and cytotoxic T cell responses to protein antigen: a new class of vaccine adjuvants.
  • CpGs may comprise modifications intended to enhance stability, such as phosphorothioate linkages, or other modifications, such as modified bases. See, for example, U.S. Patents, 5,663,153, 6,194,388, 7,262,286, or 7,276,489.
  • a composition provided herein 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 responses and anti-viral immunity.
  • the adjuvant comprises a TLR-4 agonist, such as bacterial lipopolysacharide (LPS), VSV-G, and/or HMGB-1.
  • adjuvants comprise cytokines, which are small proteins or biological factors (in the range of 5 kD - 20 kD) that are released by cells and have specific effects on cell-cell interaction, communication and behavior of other cells.
  • adjuvants comprise proinflammatory stimuli released from necrotic cells (e.g., urate crystals).
  • adjuvants comprise activated components of the complement cascade (e.g., CD21, CD35, etc.).
  • adjuvants comprise activated components of immune complexes.
  • the adjuvants also include those that comprise complement receptor agonists, such as a molecule that binds to CD21 or CD35.
  • the complement receptor agonist induces endogenous complement opsonization of the nanocarrier.
  • Adjuvants also include those that comprise cytokine receptor agonists, such as a cytokine.
  • the cytokine receptor agonist is a small molecule, antibody, fusion protein, or aptamer.
  • adjuvants also may comprise
  • immunostimulatory RNA molecules such as but not limited to dsRNA or poly I:C (a TLR3 stimulant), and/or those disclosed in F. Heil et al., "Species-Specific Recognition of Single- Stranded RNA via Toll-like Receptor 7 and 8" Science 303(5663), 1526-1529 (2004); J. Vollmer et al., "Immune modulation by chemically modified ribonucleosides and
  • the adjuvants comprise gel-type adjuvants (e.g., aluminum hydroxide, aluminum phosphate, calcium phosphate, etc.), microbial adjuvants (e.g., immunomodulatory DNA sequences that include CpG motifs; immunostimulatory RNA molecules; endotoxins such as monophosphoryl lipid A; exotoxins such as cholera toxin, E.
  • gel-type adjuvants e.g., aluminum hydroxide, aluminum phosphate, calcium phosphate, etc.
  • microbial adjuvants e.g., immunomodulatory DNA sequences that include CpG motifs; immunostimulatory RNA molecules; endotoxins such as monophosphoryl lipid A; exotoxins such as cholera toxin, E.
  • oil-emulsion and emulsifier-based adjuvants e.g., Freund's Adjuvant, MF59 [Novartis], SAF, etc.
  • particulate adjuvants e.g., liposomes, biodegradable microspheres, saponins, etc.
  • synthetic adjuvants
  • synthetic nanocarriers are spheres or spheroids. In some embodiments, synthetic nanocarriers are flat or plate-shaped. In some embodiments, synthetic nanocarriers are cubes, cuboidal or cubic. In some embodiments, synthetic nanocarriers are ovals or ellipses. In some embodiments, 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.
  • synthetic nanocarriers may optionally comprise one or more lipids.
  • a synthetic nanocarrier may 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 core comprising a polymeric matrix surrounded by a lipid layer (e.g., lipid bilayer, lipid monolayer, etc.).
  • 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 lipid layer (e.g., lipid bilayer, lipid monolayer, etc.).
  • a non- polymeric core e.g., metal particle, quantum dot, ceramic particle, bone particle, viral particle, proteins, nucleic acids, carbohydrates, etc.
  • lipid layer e.g., lipid bilayer, lipid monolayer, etc.
  • synthetic nanocarriers can comprise one or more polymers or polymeric matrices.
  • such a polymer or polymeric matrix can be surrounded by a coating layer (e.g., liposome, lipid monolayer, micelle, etc.).
  • various elements of the synthetic nanocarriers can be coupled with the polymer or polymeric matrix.
  • an element such as a targeting moiety, oligonucleotide, antigen, adjuvant, etc. can be covalently associated with a polymeric matrix.
  • covalent association is mediated by a linker.
  • an element can be noncovalently associated with a polymeric matrix.
  • an element can be encapsulated within, surrounded by, and/or dispersed throughout a polymeric matrix.
  • an element can be associated with a polymeric matrix by hydrophobic interactions, charge interactions, van der Waals forces, etc.
  • 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.
  • polymers suitable for use in the present invention include, but are not limited to polyethylenes, polycarbonates (e.g. poly(l,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., poly(P-hydroxyalkanoate)), poly(orthoesters), polycyanoacrylates, polyvinyl alcohols, polyurethanes, polyphosphazenes, polyacrylates, polymethacrylates, polyureas, polystyrenes, polyamines, polylysine, polylysine-PEG copolymers, and poly(ethyleneimine), poly(ethylene imine)-PEG copolymers.
  • polyesters e.g., polylactide, polyglycolide, polylactide-co-glycolide, polycaprolactone, polyhydroxyacid (e.g., poly(P-hydroxyalkanoate)), poly(orthoesters), polycyanoacrylates, polyvinyl alcohols, polyure
  • polymers in accordance with the present invention include polymers which have been approved for use in humans by the U.S. Food and Drug
  • polyesters e.g., polylactic acid, poly(lactic-co-glycolic acid), polycaprolactone, polyvalerolactone, poly(l,3-dioxan-2one)
  • polyanhydrides e.g., poly(sebacic anhydride)
  • polyethers e.g., polyethylene glycol
  • polyurethanes polymethacrylates; polyacrylates; and
  • 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
  • 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
  • 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[a-(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 ammonium
  • 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), polyethylene imine) (PEI; Boussif et al., 1995, Proc. Natl. Acad.
  • inventive 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
  • polymers can be used in accordance with the present invention without undergoing a cross-linking step.
  • inventive synthetic nanocarriers may comprise block copolymers, graft copolymers, blends, mixtures, and/or adducts of any of the foregoing and other polymers.
  • polymers listed herein represent an exemplary, not comprehensive, list of polymers that can be of use in accordance with the present invention.
  • the synthetic nanocarriers comprise one or more polymers.
  • the polymeric synthetic nanocarriers can also include those described in WO publication WO2009/051837 by Von Andrian et al., including, but not limited to those, with one or more hydrophilic components.
  • the one or more polymers comprise a polyester, such as a poly(lactic acid), poly(glycolic acid), poly(lactic-co-glycolic acid), or polycaprolactone. More preferably, the one or more polymers comprise or further comprise a polyester coupled to a hydrophilic polymer, such as a polyether.
  • the polyether comprises polyethylene glycol. Still more preferably, the one or more polymers comprise a polyester and a polyester coupled to a hydrophilic polymer, such as a polyether.
  • the one or more polymers are coupled to one or more antigens and/or one or more adjuvants. In embodiments, at least some of the polymers are coupled to the antigen(s) and/or at least some of the polymers are coupled to the adjuvant(s). Preferably, when there are more than one type of polymer, one of the types of polymer is coupled to the antigen(s). In embodiments, one of the other types of polymer is coupled to the adjuvant(s).
  • the nanocarriers when the nanocarriers comprise a polyester and a polyester coupled to a hydrophilic polymer, such as a polyether, the polyester is coupled to the adjuvant, while the polyester coupled to the hydrophilic polymer, such as a polyether, is coupled to the antigen(s).
  • the T helper cell antigen can be encapsulated in the nanocarrier.
  • synthetic nanocarriers may not comprise a polymeric component.
  • synthetic nanocarriers may comprise metal particles, quantum dots, ceramic particles, etc.
  • a non-polymeric synthetic nanocarrier is an aggregate of non-polymeric components, such as an aggregate of metal atoms (e.g., gold atoms).
  • synthetic nanocarriers 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;
  • DPPC dipalmitoyl phosphatidylcholine
  • DOPE dioleylphosphatidyl ethanolamine
  • DOTMA dioleyloxypropyltriethylammonium
  • 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 (Tween®20); polysorbate 60 (Tween®60);
  • polysorbate 65 (Tween®65); polysorbate 80 (Tween®80); polysorbate 85 (Tween®85); polyoxyethylene monostearate; surfactin; a poloxomer; a sorbitan fatty acid ester such as sorbitan trioleate; lecithin; lysolecithin; phosphatidylserine; phosphatidylinositol;sphingomyelin; phosphatidylethanolamine (cephalin); cardiolipin; phosphatidic acid; cerebrosides; dicetylphosphate; dipalmitoylphosphatidylglycerol;
  • 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.
  • synthetic nanocarriers 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, starch, hydroxyethylstarch, carageenan, glycon, amylose, chitosan, N,0-carboxylmethylchitosan, algin and alginic acid, starch, chitin, heparin, inulin, konjac, glucommannan, pustulan, heparin, hyaluronic acid, curdlan, and xanthan.
  • the inventive synthetic nanocarriers do not comprise (or specifically exclude) carbohydrates, such as a
  • the carbohydrate may comprise a carbohydrate derivative such as a sugar alcohol, including but not limited to mannitol, sorbitol, xylitol, erythritol, maltitol, and lactitol.
  • a sugar alcohol including but not limited to mannitol, sorbitol, xylitol, erythritol, maltitol, and lactitol.
  • compositions according to the invention comprise inventive synthetic nanocarriers in combination with pharmaceutically acceptable excipients, such as preservatives, buffers, saline, or phosphate buffered saline.
  • inventive synthetic nanocarriers are suspended in sterile saline solution for injection together with a preservative.
  • agents e.g., antigen or adjuvant
  • methods for coupling the agents to the synthetic nanocarriers may be useful. If the agent is a small molecule it may be of advantage to attach the agent to a polymer prior to the assembly of the synthetic nanocarriers.
  • a variety of reactions can be used for the purpose of attaching agents to synthetic nanocarriers.
  • the coupling can be a covalent linker.
  • peptides 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 antigen or adjuvant containing an alkyne group or by the 1,3- dipolar cycloaddition reaction of alkynes on the surface of the nanocarrier with antigens or adjuvants 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 such as the antigen or adjuvant 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 or antigens or adjuvants 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 Ri-S-S-R 2 .
  • a disulfide bond can be formed by thiol exchange of an antigen or adjuvant containing thiol/mercaptan group(-SH) with another activated thiol group on a polymer or nanocarrier or a nanocarrier containing thiol/mercaptan groups with a antigen or adjuvant containing activated thiol group.
  • a triazole linker specifically a 1,2,3-triazole of the form , wherein Ri and R 2 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 such as the peptide.
  • 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 antigen or adjuvant 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 antigen or adjuvant is then allowed to react with the nanocarrier via the 1,3-dipolar cycloaddition reaction with or without a catalyst which covalently couples the antigen or adjuvant 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 R S-R 2 .
  • Thioether can be made by either alkylation of a
  • thiol/mercaptan (-SH) group on one component such as the antigen or adjuvant 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 such as a antigen or adjuvant 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 such as a antigen or adjuvant 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 such as the antigen or adjuvant 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 such as the antigen or adjuvant 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 such as the antigen or adjuvant with 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 such as the antigen or adjuvant 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 such as the antigen or adjuvant 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 such as the antigen or adjuvant 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 such as the antigen or adjuvant 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 such as the antigen or adjuvant 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 nanoparticle or attached to the antigen or adjuvant.
  • the antigen or adjuvant can also be conjugated to the nanocarrier via non-covalent conjugation methods.
  • a negative charged antigen or adjuvant can be conjugated to a positive charged nanocarrier through electrostatic adsorption.
  • An antigen or adjuvant containing a metal ligand can also be conjugated to a nanocarner containing a metal complex via a metal-ligand complex.
  • the antigen or adjuvant 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 activatible groups on its surface.
  • the antigen or adjuvant is prepared with a group which is compatible with the attachment chemistry that is presented by the synthetic nanocarriers' surface.
  • agents such as a peptide antigen, 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
  • ADH adipic dihydrazide
  • the antigen or adjuvant can be coupled by adsorbtion to a preformed synthetic nanocarrier or it can be coupled by encapsulation during the formation of the synthetic nanocarrier.
  • 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 using 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.
  • Nanoparticles that can Efficiently Associate and Deliver Virus-like Particles Nanomedicine. 5(6):843-853 (2010)).
  • 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 of the inventive synthetic nanocarriers may be coupled to the 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 U.S. Patent Application 2006/0002852 to Saltzman et al., Published U.S. Patent Application 2009/0028910 to DeSimone et al., or Published International Patent Application WO/2008/127532 Al to Murthy et al.
  • synthetic nanocarriers can be coupled to an element, such as targeting moieties, adjuvants, various antigens, etc., 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 inventive synthetic nanocarrier.
  • encapsulation and/or absorption is a form of coupling.
  • the inventive synthetic nanocarriers can be combined with other adjuvants by admixing in the same vehicle or delivery system.
  • adjuvants may 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®), AS15, liposomes and liposomal formulations such as AS01, synthesized or specifically prepared microparticles and microcarriers such as bacteria-derived outer membrane vesicles (OMV) of N.
  • 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.
  • 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.
  • Additional useful adjuvants may be found in WO 2002/032450; US 7,357,936 "Adjuvant Systems and
  • Vaccines Vaccines
  • US 7,147,862 Vaccine composition containing adjuvants
  • US 6,544,518 Vaccines
  • US 5,750,110 Vaccine composition containing adjuvants.
  • the doses of such other adjuvants can be determined using conventional dose ranging studies.
  • adjuvant that is not coupled to the recited synthetic nanocarriers may be the same or different from adjuvant that is coupled to the synthetic nanocarriers, if any.
  • the doses of such adjuvants may also be the same or different.
  • any adjuvant coupled to the inventive synthetic nanocarriers can be different, similar or identical to those not coupled to any nanocarriers.
  • the adjuvants can be administered separately at a different time-point and/or at a different body location and/or by a different immunization route. Additionally, the separate adjuvant and population of nanocarriers can be administered separately at a different time- point and/or at a different body location and/or by a different immunization route.
  • 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 subset 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.
  • Typical inventive 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.,
  • carboxymethylcellulose carboxymethylcellulose
  • co-solvents e.g., glycerol, polyethylene glycol, ethanol
  • compositions according to the invention comprise inventive 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.
  • inventive synthetic nanocarriers are suspended in sterile saline solution for injection together with a preservative.
  • compositions of the invention can be made in any suitable manner, and the invention is in no way limited to 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.
  • inventive synthetic nanocarriers are manufactured under sterile conditions or are terminally sterilized. This can ensure that resulting composition 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.
  • inventive 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 may be administered by a variety of routes of
  • administration including but not limited to subcutaneous, intramuscular, intradermal, oral, intranasal, transmucosal, sublingual, rectal, ophthalmic, transdermal, transcutaneous or by a combination of these routes.
  • Doses of dosage forms contain varying amounts of populations of synthetic nanocarriers and/or varying amounts of adjuvants and/or antigens, according to the invention.
  • the amount of synthetic nanocarriers and/or adjuvants and/or antigens present in the inventive dosage forms can be varied according to the nature of the adjuvants and/or antigens, the therapeutic benefit to be accomplished, and other such parameters.
  • the doses of the dosage forms are systemic doses.
  • dose ranging studies can be conducted to establish optimal therapeutic amount of the population of synthetic nanocarriers and/or the amount of adjuvants and/or antigens to be present in the dosage form.
  • the synthetic nanocarriers and/or the adjuvants and/or antigens are present in the dosage form in an amount effective to generate an immune response upon administration to a subject. It may be possible to determine amounts of the adjuvants and/or antigens effective to generate an immune response using conventional dose ranging studies and techniques in subjects.
  • Inventive 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, stimulate, 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, autoimmune diseases, inflammatory diseases, immunological 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 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.
  • the compositions provided can be used to induce a rapid and strong systemic induction of pro-inflammatory cytokines, such as TNF-oc, IL-6 and/or IL-12.
  • the compositions provided therefore, can be administered to subjects in need of an inflammatory response, preferably a systemic inflammatory response.
  • the compositions provided can be used for the rapid and strong systemic induction of cytokines that are important for a Thl immune response, such as IFN- ⁇ , IL-12 and/or IL-18.
  • the compositions provided therefore, can be administered to subjects in need of a Thl response, preferably a systemic Thl response.
  • the compositions provided can be used to induce a strong humoral response.
  • compositions provided can be administered to subjects in need of a humoral response.
  • the compositions provided can be used to induce a strong specific local CTL response.
  • the compositions provided therefore, can be administered to subjects in need of a specific local CTL response.
  • Such a response can be specific to any of the antigens provided herein, preferably to one or more antigens in an inventive composition or that is administered according to an inventive method provided herein.
  • 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 epithelial cells, stromal cells, germ cells and mesenchymal cells;
  • pancreatic cancer pancreatic cancer
  • prostate cancer rectal cancer
  • sarcomas including leiomyosarcoma, rhabdomyosarcoma, liposarcoma, fibrosarcoma, and osteosarcoma
  • skin cancer including melanoma, Merkel cell carcinoma, Kaposi's sarcoma, basal cell carcinoma, and squamous cell cancer
  • testicular cancer including germinal tumors such as seminoma, non-seminoma (teratomas, choriocarcinomas), stromal tumors, and germ cell tumors
  • thyroid cancer including thyroid adenocarcinoma and medullar carcinoma
  • renal cancer including adenocarcinoma and Wilms tumor.
  • Infections or infectious diseases 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,
  • 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 mononucleos
  • 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, Diphtheria, Epidemic Typhus, Gonorrhea, Impetigo, Legionellosis, Leprosy (Hansen's
  • Subject provided here also include those that have or are at risk of having an atopic condition, such as but not limited to allergy, allergic asthma, or atopic dermatitis; asthma; chronic obstructive pulmonary disease (COPD, e.g. emphysema or chronic bronchitis); and chronic infections due to chronic infectious agents such as chronic leishmaniasis, candidiasis or schistosomiasis and infections caused by plasmodia, Toxoplasma gondii, mycobacteria, HIV, HBV, HCV EBV or CMV, or any one of the above, or any subset of the above.
  • COPD chronic obstructive pulmonary disease
  • chronic infections due to chronic infectious agents such as chronic leishmaniasis, candidiasis or schistosomiasis and infections caused by plasmodia, Toxoplasma gondii, mycobacteria, HIV, HBV, HCV EBV or CMV, or any one of the above, or any subset of the
  • indications treatable using the inventive compositions include but are not limited to indications in which a subject's Thl response is suboptimal and/or ineffective.
  • Use of the present invention can enhance a subject's Thl immune response with an adjuvant that can stimulate a Thl immune response.
  • the subjects therefore, also include those that have or are at risk of having cancer, subjects with compromised or suboptimal immunity, such as infants, the elderly, cancer patients, individuals receiving immunosuppressive drugs or irradiation, hemodialysis patients and those with genetic or idiopathic immune dysfunction.
  • Ovalbumin peptide 323-339 amide acetate salt was purchased from Bachem
  • PLGA-R848 conjugate of 75/25 lactide/glycolide monomer composition and of approximately 4100 Da molecular weight having 5.2% w/w R848 content was synthesized.
  • PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized
  • Ovalbumin peptide 323 - 339 @ 70mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
  • PLGA-R848 @ 75 mg/mL and PLA-PEG-Nicotine @ 25 mg/mL in dichloromethane was prepared by dissolving PLGA-R848 at 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at 100 mg/mL in dichloromethane, then combining 3 parts of the PLGA-R848 solution to 1 part of the PLA-PEG-Nicotine solution.
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
  • Solution 4 70 mM phosphate buffer, pH 8.
  • Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension.
  • Nanocarrier ID Effective Diameter TLR Agonist % w/w T-cell helper peptide, % w/w
  • Ovalbumin peptide 323-339 amide acetate salt was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4065609.)
  • PLGA-R848 conjugate of 75/25 lactide/glycolide monomer composition and of approximately 4100 Da molecular weight having 5.2% w/w R848 content was synthesized.
  • PLA-PEG-Nicotine with a nicotine- terminated PEG block of approximately 5,000 Da and DL-PLA block of approximately 17,000 Da was synthesized.
  • Ovalbumin peptide 323 - 339 @ 70mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
  • PLGA-R848 @ 75 mg/mL and PLA-PEG-Nicotine @ 25 mg/mL in dichloromethane was prepared by dissolving PLGA-R848 at 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at 100 mg/mL in dichloromethane, then combining 3 parts of the PLGA-R848 solution to 1 part of the PLA-PEG-Nicotine solution.
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
  • Solution 4 70 mM phosphate buffer, pH 8.
  • a primary (Wl/O) emulsion was first created using Solution 1 & Solution 2.
  • Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension.
  • a portion of the suspended nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 13800 rcf for 60 minutes, removing the supernatant, and re- suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use.
  • Ovalbumin peptide 323 - 339 @ 70mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
  • Solution 2 PLGA @ 75 mg/mL and PLA-PEG-Nicotine @ 25 mg/mL in
  • dichloromethane was prepared by dissolving PLGA at 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at 100 mg/mL in dichloromethane, then combining 3 parts of the PLGA solution to 1 part of the PLA-PEG-Nicotine solution.
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
  • Solution 4 70 mM phosphate buffer, pH 8.
  • a primary (Wl/O) emulsion was first created using Solution 1 & Solution 2.
  • Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution
  • nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 13800 rcf for
  • mice were injected subcutaneously into hind limbs with 100 ⁇ g of nanocarriers (NC) coupled, non-coupled or admixed with small molecule nucleoside analogue and known TLR7/8 agonist and adjuvant, R848.
  • R848 amounts in nanocarrier were 2-3% resulting in
  • NC not coupled to R848 did not induce any systemic cytokines when used without admixed R848.
  • Ovalbumin peptide 323-339 amide acetate salt was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4065609.)
  • PLGA-R848 conjugate of 75/25 lactide/glycolide monomer composition and of approximately 4100 Da molecular weight having 5.2% w/w R848 content was synthesized.
  • PLA-PEG-Nicotine with a nicotine- terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized.
  • Ovalbumin peptide 323 - 339 @ 70mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
  • PLGA-R848 @ 75 mg/mL and PLA-PEG-Nicotine @ 25 mg/mL in dichloromethane was prepared by dissolving PLGA-R848 at 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at 100 mg/mL in dichloromethane, then combining 3 parts of the PLGA-R848 solution to 1 part of the PLA-PEG-Nicotine solution.
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
  • Solution 4 70 mM phosphate buffer, pH 8.
  • Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution
  • Nanocarrier ID Effective Diameter TLR Agonist % w/w T-cell helper peptide, % w/w (nm)
  • Ovalbumin peptide 323-339 amide acetate salt was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4065609.)
  • PLGA with 73% lactide and 27% glycolide content and an inherent viscosity of 0.12 dL/g was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4065609.)
  • PLGA with 73% lactide and 27% glycolide content and an inherent viscosity of 0.12 dL/g was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4065609.)
  • PLGA with 73% lactide and 27% glycolide content and an inherent viscosity of 0.12 dL/g was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4065609.)
  • PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized.
  • Polyvinyl alcohol (Mw 11,000 - 31,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232-08).
  • Ovalbumin peptide 323 - 339 @ 70mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
  • Solution 2 PLGA @ 75 mg/mL and PLA-PEG-Nicotine @ 25 mg/mL in
  • dichloromethane was prepared by dissolving PLGA at 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at 100 mg/mL in dichloromethane, then combining 3 parts of the PLGA solution to 1 part of the PLA-PEG-Nicotine solution.
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
  • Solution 4 70 mM phosphate buffer, pH 8.
  • a primary (Wl/O) emulsion was first created using Solution 1 & Solution 2.
  • Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution
  • Ovalbumin peptide 323-339 amide TFA salt was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4064565.) PLA with an inherent viscosity of 0.19 dL/g was purchased from Boehringer Ingelheim (Ingelheim Germany.
  • PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized.
  • Polyvinyl alcohol (Mw 11,000 - 31,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232-08). Methods for NC-Nic w/o R848 Nanocarrier Production
  • Ovalbumin peptide 323 - 339 @ 69mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
  • Solution 2 PLA @ 75 mg/mL and PLA-PEG-Nicotine @ 25mg/mL in
  • dichloromethane was prepared by dissolving PLA @ 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at lOOmg/mL in dichloromethane, then combining 3 parts of the PLA solution to 1 part of the PLA-PEG-Nicotine solution.
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM in deionized water.
  • Solution 4 70 mM phosphate buffer, pH 8.
  • Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 35% amplitude for 40 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to a beaker containing 70 mM phosphate buffer solution (30 mL) in an open 50ml beaker and stirred at room temperature for 2 hours to allow for the dichloromethane to evaporate and for the nanocarriers to form in suspension.
  • Nanocarrier Effective Diameter TLR Agonist % w/w T-cell helper peptide, % w/w
  • Ovalbumin peptide 323-339 amide TFA salt was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4064565.) R848 (Resiquimod) of approximately 98-99% purity was synthesized and purified.
  • PLA with an inherent viscosity of 0.19 dL/g was purchased from Boehringer Ingelheim (Ingelheim Germany. Product Code R202H).
  • PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized.
  • Polyvinyl alcohol (Mw 11,000 - 31,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232- 08).
  • Ovalbumin peptide 323 - 339 @ 69mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
  • Solution 2 PLA @ 75 mg/mL, R848 @ 7.5 mg/mL, and PLA-PEG-Nicotine @ 25mg/mL in dichloromethane was prepared by dissolving PLA @ 100 mg/mL in
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM in deionized water.
  • Solution 4 70 mM phosphate buffer, pH 8.
  • a primary (Wl/O) emulsion was first created using Solution 1 & Solution 2.
  • Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 35% amplitude for 40 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to a beaker containing 70 mM phosphate buffer solution (30 mL) in an open 50ml beaker and stirred at room temperature for 2 hours to allow for the dichloromethane to evaporate and for the nanocarriers to form in suspension.
  • a portion of the suspended nanocarriers were washed by transferring the nanocarrier suspension to centrifuge tubes, spinning at 5300 rcf for 60 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis.
  • the suspension was stored frozen at -20°C until use.
  • mice were immunized with NC-Nic (nanocarrier exhibiting nicotine on the outer surface) carrying entrapped (non-conjugated) R848 with or without second adjuvant.
  • Groups of five mice were immunized three times (subcutaneously, hind limbs) at 2-week intervals (days 0, 14 and 28) with 100 ⁇ g of NC-Nic.
  • Serum anti-nicotine antibodies were then measured on days 26 and 40.
  • EC 50 for anti-nicotine antibodies were measured by standard ELISA against polylysine-nicotine (Fig. 3) (Group 1: NC-Nic w/o entrapped R848; group 2: NC-Nic w.
  • Ovalbumin peptide 323-339 amide TFA salt was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4064565.) PLA with an inherent viscosity of 0.19 dL/g was purchased from Boehringer Ingelheim (Ingelheim Germany.
  • PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized.
  • Polyvinyl alcohol (Mw 11,000 - 31,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232-08).
  • Ovalbumin peptide 323 - 339 @ 69mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
  • Solution 2 PLA @ 75 mg/mL and PLA-PEG-Nicotine @ 25mg/mL in
  • dichloromethane was prepared by dissolving PLA @ 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at lOOmg/mL in dichloromethane, then combining 3 parts of the PLA solution to 1 part of the PLA-PEG-Nicotine solution.
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM in deionized water.
  • Solution 4 70 mM phosphate buffer, pH 8.
  • a primary (Wl/O) emulsion was first created using Solution 1 & Solution 2.
  • Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 35% amplitude for 40 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to a beaker containing 70 mM phosphate buffer solution (30 mL) in an open 50ml beaker and stirred at room temperature for 2 hours to allow for the dichloromethane to evaporate and for the nanocarriers to form in suspension.
  • a portion of the suspended nanocarriers were washed by transferring the nanocarrier suspension to centrifuge tubes, spinning at 5300 rcf for 60 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use. Table 8: Characterization of the Nanocarriers Produced According to the Above
  • Ovalbumin peptide 323-339 amide TFA salt was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4064565.) R848 (Resiquimod) of approximately 98-99% purity was synthesized and purified.
  • PLA with an inherent viscosity of 0.19 dL/g was purchased from Boehringer Ingelheim (Ingelheim Germany. Product Code R202H).
  • PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized.
  • Polyvinyl alcohol (Mw 11 ,000 - 31 ,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232- 08).
  • Ovalbumin peptide 323 - 339 @ 69mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
  • Solution 2 PLA @ 75 mg/mL, R848 @ 7.5 mg/mL, and PLA-PEG-Nicotine @ 25mg/mL in dichloromethane was prepared by dissolving PLA @ 100 mg/mL in
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM in deionized water.
  • a primary (Wl/O) emulsion was first created using Solution 1 & Solution 2.
  • Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 35% amplitude for 40 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to a beaker containing 70 mM phosphate buffer solution (30 mL) in an open 50ml beaker and stirred at room temperature for 2 hours to allow for the dichloromethane to evaporate and for the nanocarriers to form in suspension.
  • a portion of the suspended nanocarriers were washed by transferring the nanocarrier suspension to centrifuge tubes, spinning at 5300 rcf for 60 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use.
  • mice were immunized with NC-Nic (nanocarrier exhibiting nicotine on the outer surface) that did not have adjuvant in the NC with or without admixed R848.
  • Groups of five mice were immunized three times (subcutaneously, hind limbs) at 2-week intervals (days 0, 14 and 28) with 100 ⁇ g of NC-Nic.
  • Serum anti-nicotine antibodies were then measured on days 26 and 40.
  • EC 50 for anti-nicotine antibodies were measured by standard ELISA against polylysine-nicotine (Fig. 4) (group 1: NC-Nic w/o entrapped R848; group 2: NC-Nic w/o entrapped R848 + 20 ⁇ g of free R848).
  • NC-Nic-R84848 adjuvant encapsulated R848 adjuvant
  • Enzo free Thl adjuvant CpG-1826
  • Th2 adjuvant alum Pieris
  • Ovalbumin peptide 323-339 amide acetate salt was purchased from Bachem
  • PLA-R848 conjugate having molecular weight of approximately 1300 Da and R848 content of approximately 9% by weight was synthesized at by a ring-opening process.
  • PLA-PEG- Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized.
  • Ovalbumin peptide 323 - 339 @ 70 mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
  • PLA-R848 @ 75 mg/mL, PLA-PEG-Nicotine @ 25 mg/mL, and R848 @ 1.9 mg/mL in dichloromethane was prepared by dissolving the polymers at 100 mg/mL, adding the R848 to the PLA-PEG-Nicotine solution, and then combining 3 parts of the PLA- R848 solution to 1 part of the PLA-PEG-Nicotine/R848 solution.
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM in deionized water.
  • Solution 4 70 mM phosphate buffer, pH 8.
  • Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 35% amplitude for 40 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to a beaker containing 70 mM phosphate buffer solution (30 mL) in an open 50ml beaker and stirred at room temperature for 2 hours to allow for the dichloromethane to evaporate and for the nanocarriers to form in suspension.
  • a portion of the suspended nanocarriers were washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 13,800 rcf for 60 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use.
  • mice were immunized with the nanocarriers, NC-Nic (nanocarrier exhibiting nicotine on the outer surface) which carried R848 and OP-II helper peptide, with or without admixed alum.
  • Groups of five mice were immunized three times (subcutaneously, hind limbs) at 2- week intervals (days 0, 14 and 28) with 100 ⁇ g of NC[Nic,R848,OP-II] +/- 80 ⁇ g of admixed alum (Pierce).
  • Serum anti-nicotine antibodies were then measured on days 40 and 70.
  • EC 50 for anti-nicotine antibodies were measured by standard ELISA against polylysine-nicotine (Fig.
  • NC-encapsulated Antigen Generates a Stronger Cellular Immune Response than Free Antigen (Free Adjuvant Admixed)
  • Ovalbumin protein was purchased from Worthington Biochemical Corporation (730
  • Solution 1 Ovalbumin protein @ 20mg/mL was prepared in phosphate buffered saline at room temperature.
  • PLA @ 75 mg/mL and PLA-PEG-OMe @ 25 mg/mL in dichloromethane was prepared by dissolving PLA at 100 mg/mL in dichloromethane and PLA-PEG-OMe at 100 mg/mL in dichloromethane, then combining 3 parts of the PLA solution to 1 part of the PLA-PEG-OMe solution.
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
  • Solution 4 70 mM phosphate buffer, pH 8.
  • a primary (Wl/O) emulsion was first created using Solution 1 & Solution 2.
  • Solution 1 (0.2 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (3.0 mL) to the primary emulsion, vortexing to create a course dispersion, and then sonicating at 30% amplitude for 60 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension.
  • a portion of the suspended nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 21,000 rcf for 45 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use.
  • mice were immunized either with the nanocarriers, NC-OVA (nanocarrier carrying encapsulated ovalbumin protein), or with free ovalbumin (OVA) with a free adjuvant admixed.
  • Groups of 3 mice were immunized once (s.c, hind limbs) with 100 ⁇ g of NC-OVA (2.8% OVA) or with 2.5 ⁇ g of free OVA admixed with 10 ⁇ g of free 1826-CpG (TLR9 agonist).
  • Ovalbumin peptide 323-339 amide acetate salt was purchased from Bachem Americas
  • PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 5,000 Da and DL-PLA block of approximately 17,000 Da was synthesized.
  • Polyvinyl alcohol (Mw 11 ,000 - 31 ,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232-08).
  • Solution 1 Ovalbumin peptide 323 - 339 @ 17.5 mg/mL in dilute hydrochloric acid aqueous solution.
  • the solution was prepared by dissolving ovalbumin peptide in 0.13N hydrochloric acid solution at room temperature.
  • Solution 2 0.19-IV PLA @ 75 mg/mL and PLA-PEG-nicotine @ 25 mg/ml in dichloromethane.
  • the solution was prepared by separately dissolving PLA @ 100 mg/mL in dichloromethane and PLA-PEG-nicotine @ 100 mg/mL in dichloromethane, then mixing the solutions by adding 3 parts PLA solution for each part of PLA-PEG-nicotine solution.
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • a primary (Wl/O) emulsion was first created using Solution 1 & Solution 2.
  • Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and then sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was then added to an open 50 mL beaker containing 70mM pH 8 phosphate buffer solution (30 mL) and stirred at room temperature for 2 hours to evaporate dichloromethane and to form nanocarriers in aqueous suspension.
  • a portion of the nanocarriers was washed by transferring the suspension to a centrifuge tube and spinning at 13,800g for one hour, removing the supernatant, and re- suspending the pellet in phosphate buffered saline. The washing procedure was repeated, and the pellet was re- suspended in phosphate buffered saline for a final nanocarrier dispersion of about 10 mg/mL.
  • the amounts of oligonucleotide and peptide in the nanocarrier were determined by HPLC analysis.
  • the total dry-nanocarrier mass per mL of suspension was determined by a gravimetric method and was adjusted to 5 mg/mL.
  • mice were immunized with NC-Nic (nanocarrier exhibiting nicotine on the outer surface and containing OP-II helper peptide, no adjuvant in the NC) admixed with CpG in either the phosphodiester (PO) or phosphorothioate (PS) form.
  • the PO form is degraded by nucleases and, therefore, is not stable once injected into mice.
  • the PS form is nuclease- resistant and, therefore, stable once injected into mice.
  • mice were immunized with PBS only. Groups of five mice were immunized three times
  • NC-Nic + 20 ⁇ g of CpG (PS or PO) or PBS Serum anti-nicotine antibody titers were measured on days 26 and 40.
  • Anti-nicotine antibody titers (EC 50 ) were measured by ELISA against polylysine-nicotine (Fig. 8) (group 1: NC-Nic (no adjuvant) + free CpG (PS); group 2: NC- Nic (no adjuvant) + free CpG (PO); group 3: PBS only).
  • Solution 1 Ovalbumin protein @ 20mg/mL was prepared in phosphate buffered saline at room temperature.
  • PLA @ 75 mg/mL and PLA-PEG-OMe @ 25 mg/mL in dichloromethane was prepared by dissolving PLA at 100 mg/mL in dichloromethane and PLA-PEG-OMe at 100 mg/mL in dichloromethane, then combining 3 parts of the PLA solution to 1 part of the PLA-PEG-OMe solution.
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
  • Solution 4 70 mM phosphate buffer, pH 8.
  • a primary (Wl/O) emulsion was first created using Solution 1 & Solution 2.
  • Solution 1 (0.2 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (3.0 mL) to the primary emulsion, vortexing to create a course dispersion, and then sonicating at 30% amplitude for 60 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension.
  • a portion of the suspended nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 21,000 rcf for 45 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use.
  • Nanocarrier Effective Diameter TLR Agonist % w/w Antigen, % w/w
  • mice were immunized with NC-OVA (nanocarrier exhibiting ovalbumin (OVA) on the outer surface, no adjuvant in the NC) admixed with either 20 ⁇ g of R848 or CpG (PS; nuclease-resistant).
  • Control mice received 2.5 ⁇ g of soluble antigen (OVA) admixed with 20 ⁇ g of CpG (PS).
  • Groups of five mice were immunized three times (subcutaneously, hind limbs) at 2-week intervals (days 0, 14 and 28) with 100 ⁇ g of NC-OVA + 20 ⁇ g of R848 or CpG (PS) or 2.5 ⁇ g of soluble OVA + 20 ⁇ g of CpG (PS).
  • Serum anti-OVA antibody titers were measured on days 26 and 44.
  • Anti-OVA antibody titers (EC 50 ) were measured by ELISA against OVA protein (Fig. 9) (group 1: NC-OVA (no adjuvant) + free R848; group 2: NC-OVA (no adjuvant) + free CpG (PS); group 3: soluble OVA + CpG (PS)).
  • Ovalbumin peptide 323-339 amide acetate salt was purchased from Bachem Americas
  • PLGA-R848 conjugate of 75/25 lactide/glycolide monomer composition and of approximately 4100 Da molecular weight having 5.2% w/w R848 content was synthesized.
  • PLA-PEG-Nicotine with a nicotine- terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized.
  • Ovalbumin peptide 323 - 339 @ 70mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
  • PLGA-R848 @ 75 mg/mL and PLA-PEG-Nicotine @ 25 mg/mL in dichloromethane was prepared by dissolving PLGA-R848 at 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at 100 mg/mL in dichloromethane, then combining 3 parts of the PLGA-R848 solution to 1 part of the PLA-PEG-Nicotine solution.
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
  • Solution 4 70 mM phosphate buffer, pH 8.
  • a primary (Wl/O) emulsion was first created using Solution 1 & Solution 2.
  • Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution (30 mL) and stirred at room temperature for 2 hours to allow for the dichloromethane to evaporate and for the nanocarriers to form in suspension.
  • a portion of the suspended nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 13800 rcf for 60 minutes, removing the supernatant, and re- suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use.
  • Ovalbumin peptide 323-339 amide acetate salt was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4065609.)
  • PLGA with 73% lactide and 27% glycolide content and an inherent viscosity of 0.12 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, AL 35211. Product Code 7525 DLG 1A.
  • PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized.
  • Ovalbumin peptide 323 - 339 @ 70mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
  • Solution 2 PLGA @ 75 mg/mL and PLA-PEG-Nicotine @ 25 mg/mL in
  • dichloromethane was prepared by dissolving PLGA at 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at 100 mg/mL in dichloromethane, then combining 3 parts of the PLGA solution to 1 part of the PLA-PEG-Nicotine solution.
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
  • Solution 4 70 mM phosphate buffer, pH 8.
  • Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension.
  • Nanocarrier Effective Diameter TLR Agonist % w/w T-cell helper peptide, % w/w
  • mice were injected with 20 ⁇ g of CpG twice (subcutaneously, hind limbs) at 2-week intervals (days 0 and 14). At days 35 and 49, mice were immunized with 100 ⁇ g of NC-Nic (containing 2.6% R848 and 0.9% OP-II peptide) or 100 ⁇ g of NC-Nic (containing 1.1% OP- II peptide only). Serum anti-nicotine antibody titers were measured at days 12, 26, and 40 after immunization with NC. Anti-nicotine antibody titers (EC 50 ) were measured by ELISA against polylysine-nicotine (Fig.
  • mice immunized with a combination of CpG followed at a later date by NC-Nic that contain R848 generate higher antibody titers to nicotine than mice immunized with CpG followed at a later date by NC-Nic that do not contain R848 (group 1 > group 2).
  • Ovalbumin peptide 323-339 amide acetate salt is purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4064565.)
  • PLA with an inherent viscosity of 0.19 dL/g is purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, AL 35211 (Product Code 100 DL 2A).
  • PLA-PEG-Nicotine with a nicotine- terminated PEG block of approximately 5,000 Da and DL-PLA block of approximately 20,000 Da is synthesized.
  • Ovalbumin peptide 323 - 339 @ 20mg/mL is prepared in 0.13N hydrochloric acid at room temperature.
  • Solution 2 PLA @ 75 mg/mL and PLA-PEG-Nicotine @ 25mg/mL in
  • dichloromethane is prepared by dissolving PLA @ 100 mg/mL in dichloromethane and PLA- PEG-Nicotine at lOOmg/mL in dichloromethane, then combining 3 parts of the PLA solution to 1 part of the PLA-PEG-Nicotine solution.
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM in deionized water.
  • Solution 4 70 mM phosphate buffer, pH 8.
  • a primary (Wl/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.
  • (W1/0/W2) emulsion is then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion is added to a beaker containing 70 mM phosphate buffer solution (30 mL) in an open 50ml beaker and stirred at room temperature for 2 hours to allow for the dichloromethane to evaporate and for the nanocarriers to form in suspension.
  • a portion of the suspended nanocarriers are washed by transferring the nanocarrier suspension to centrifuge tubes, spinning at 21,000 rcf for 45 minutes, removing the supernatant, and re- suspending the pellet in phosphate buffered saline.
  • mice are immunized with NC-Nic (nanocarrier exhibiting nicotine on the outer surface) admixed with a first (R848) and second adjuvant (alum). Groups of five mice are immunized three times (subcutaneously, hind limbs) at 2-week intervals (days 0, 14 and 28) with 100 ⁇ g of NC-Nic. Serum anti-nicotine antibodies are then measured on days 26 and 40. EC 50 for anti-nicotine antibodies are measured by standard ELISA against polylysine- nicotine.

Abstract

Disclosed are synthetic nanocarrier compositions with separate adjuvant compositions as well as related methods.

Description

NANOCARRIER COMPOSITIONS WITH UNCOUPLED ADJUVANT
RELATED APPLICATIONS
This application claims the benefit under 35 U.S.C. §119 of United States provisional applications 61/348713, filed May 26, 2010, 61/348717, filed May 26, 2010, 61/348728, filed May 26, 2010, and 61/358635, filed June 25, 2010, the entire contents of each of which are incorporated herein by reference.
FIELD OF THE INVENTION
This invention relates to synthetic nanocarrier and separate adjuvant compositions, and related methods, such as for treating diseases in which generating an immune response is desirable.
BACKGROUND OF THE INVENTION
Adjuvants are generally important components for the majority of currently used vaccination regimens. The optimal approach for augmenting the immune response with adjuvant, however, in a number of cases, is not yet known. Therefore, improved
compositions and therapeutic methods are needed to provide improved therapies for diseases in which generating an immune response and/or augmenting it is desirable.
SUMMARY OF THE INVENTION
In one aspect, a composition comprising a dosage form that comprises (1) a population of synthetic nanocarriers, (2) a first adjuvant that is not coupled to any synthetic nanocarriers, and (3) a pharmaceutically acceptable excipient is provided. In another aspect, a method comprising administering the composition to a subject is provided.
In one embodiment, the compositions provided herein, including those of the methods provided, comprise a systemic dose of the first adjuvant. In another embodiment, said compositions further comprise a second adjuvant. In still another embodiment, the first adjuvant and second adjuvant are different. In yet another embodiment, the first adjuvant and the second adjuvant are the same. In still another embodiment, the compositions provided herein, including those of the methods provided, comprise a systemic dose of the first adjuvant and/or second adjuvant. In one embodiment, the systemic dose results in the systemic release of TNF-oc, IL-6 and/or IL-12. In another embodiment, the systemic dose results in the systemic release of IFN-γ, IL-12 and/or IL-18.
In one embodiment, the second adjuvant of the compositions provided, including those of the methods provided, is coupled to the synthetic nanocarriers. In another embodiment, the second adjuvant is not coupled to any synthetic nanocarriers. In yet another embodiment, the second adjuvant is coupled to another population of synthetic nanocarriers.
In another embodiment, the compositions provided herein, including those of the methods provided, comprise one or more antigens. In a further embodiment, the one or more antigens are coupled to the synthetic nanocarriers. In yet a further embodiment, the one or more antigens are coupled to another population of synthetic nanocarriers. In another embodiment, the one or more antigens are not coupled to any synthetic nanocarriers.
In one embodiment, the one or more antigens of the compositions provided, including those of the methods provided, comprise a B cell antigen and/or a T cell antigen. In another embodiment, the T cell antigen is a universal T cell antigen or T helper cell antigen. In a further embodiment, the one or more antigens comprise a B cell antigen and/or a T cell antigen and a universal T cell antigen or T helper cell antigen. In one embodiment, the B cell antigen is nicotine. In yet another embodiment, the compositions provided, including those of the methods provided, do not comprise an antigen.
In one embodiment, of the compositions provided, including those of the methods provided, the first adjuvant and/or second adjuvant comprises a mineral salt, gel-type adjuvant, a microbial adjuvant, an oil-emulsion or emulsifier-based adjuvant, a particulate adjuvant, a synthetic adjuvant, a phosphate adjuvant, a polymer, a liposome, a microcarrier, an immunostimulatory nucleic acid, alum, a saponin, an interleukin, an interferon, a cytokine, a toll-like receptor (TLR) agonist, an imidazoquinoline, a cytokine receptor agonist, a CD40 agonist, an Fc receptor agonist, a complement receptor agonist, QS21, vitamin E, squalene, tocopherol, Quil A, ISCOMs, ISCOMATRIX, Ribi Detox, CRL-1005, L-121,
tetrachlorodecaoxide, alum, MF59, AS02, AS15, cholera toxin, monophosphoryl lipid A, incomplete Freund's adjuvant, complete Freund's adjuvant, muramyl dipeptide or montanide. In one embodiment, the immunostimulatory nucleic acid comprises a CpG-containing nucleic acid. In another embodiment, the imidazoquinoline comprises resiquimod or imiquimod. In still another embodiment, the first and/or second adjuvant comprises alum. In one
embodiment, when the first adjuvant comprises a CpG-containing nucleic acid, the second adjuvant comprises an imidazoquinoline or alum. In another embodiment, the imidazoquinoline is resiquimod. In still another embodiment, when the first adjuvant comprises an imidazoquinoline, the second adjuvant comprises a CpG-containing nucleic acid or alum. In a further embodiment, the imidazoquinoline is resiquimod. In one embodiment, when the first adjuvant comprises alum, the second adjuvant comprises an imidazoquinoline or a CpG-containing nucleic acid. In another embodiment, the
imidazoquinoline is resiquimod. In a further embodiment, the TLR agonist comprises a TLR-1, TLR-2, TLR-3, TLR-4, TLR- 5, TLR-6, TLR-7, TLR-8, TLR-9, TLR-10, TLR-11 agonist or a combination thereof. In another embodiment, the first adjuvant and/or second adjuvant does not comprise a TLR agonist. In yet another embodiment, the first adjuvant and/or second adjuvant does not comprise a TLR-3, TLR-7, TLR-8 or TLR-9 agonist. In one embodiment, the second adjuvant is coupled to the synthetic nanocarriers and comprises resiquimod.
In one embodiment, the synthetic nanocarriers of the compositions provided herein, including those of the methods provided, comprise lipid nanoparticles, polymeric
nanoparticles, metallic nanoparticles, surfactant-based emulsions, dendrimers, buckyballs, nanowires, virus-like particles, peptide or protein particles, nanoparticles that comprise a combination of nanomaterials, spheroidal nanoparticles, cuboidal nanoparticles, pyramidal nanoparticles, oblong nanoparticles, cylindrical nanoparticles or toroidal nanoparticles.
In one embodiment, the synthetic nanocarriers comprise one or more polymers. In another embodiment, the one or more polymers comprise a polyester. In yet another embodiment, the one or more polymers comprise or further comprise a polyester coupled to a hydrophilic polymer. In a further embodiment, the polyester comprises a poly(lactic acid), poly(glycolic acid), poly(lactic-co-glycolic acid), or polycaprolactone. In still a further embodiment, the hydrophilic polymer comprises a polyether. In another embodiment, the polyether comprises polyethylene glycol.
In another embodiment, the one or more antigens of the compositions provided herein, including those of the methods provided, comprise nicotine and a universal T cell antigen or T-helper cell antigen, each of which are coupled to the synthetic nanocarriers.
In a further embodiment, the universal T cell antigen or T helper cell antigen of the compositions provided herein, including those of the methods provided, is coupled by encapsulation. In still another embodiment, the T-helper cell antigen comprises a peptide obtained or derived from ovalbumin. In a further embodiment, the peptide obtained or derived from ovalbumin comprises the sequence as set forth in SEQ ID NO: 1.
In another aspect, a method comprising administering any of the compositions provided herein to a subject is provided. In one embodiment, the subject is a human.
In a further aspect, a method comprising administering any of the compositions provided and a second adjuvant to a subject, wherein the second adjuvant is administered at a time different from the administration of the composition, is provided. In one embodiment, the subject is a human. In another embodiment, the composition and second adjuvant are coadministered. In yet another embodiment, the composition and second adjuvant are not coadministered. In still another embodiment, the second adjuvant is administered prior to the composition.
In one embodiment, any of the methods provided further comprises administering one or more antigens. In another embodiment, any of the the compositions provided, including those of the methods provided, further comprises one or more antigens. In one embodiment, the one or more antigens are coadministered.
In one embodiment, the subject of any of the methods provided or to which any of the compositions provided is administered is in need of an inflammatory response. In another embodiment, the subject is in need of a Thl immune response. In yet another embodiment, the subject is in need of a humoral immune response. In still another embodiment, the subject is in need of a specific local cytotoxic T lymphocyte response. In a further embodiment, the subject has or is at risk of having cancer. In still a further embodiment, the subject has or is at risk of having an infection or infectious disease. In yet a further embodiment, the subject has or is at risk of having an atopic condition, asthma, COPD or a chronic infection.
In one embodiment, any of the compositions can be for use in therapy or prophylaxis. In another embodiment, any of the compositions can be for use in any of the methods provided. In yet another embodiment, any of the compositions can be for use in a method of inducing an inflammatory response in a subject. In still another embodiment, any of the compositions can be for use in a method of inducing a Thl immune response in a subject. In yet another embodiment, any of the compositions can be for use in a method of inducing a humoral immune response in a subject. In a further embodiment, any of the compositions can be for use in a method of inducing a specific local cytotoxic T lymphocyte response in a subject. In still a further embodiment, any of the compositions can be for use in a method of treating or preventing cancer. In yet a further embodiment, any of the compositions can be for use in a method of treating or preventing infection or infectious disease. In another embodiment, any of the compositions can be for use in a method of treating or preventing an atopic condition, asthma, COPD or a chronic infection.
In another aspect, a use of any of the compositions provided for the manufacture of a medicament for use in any of the methods provided is provided herein.
BRIEF DESCRIPTION OF FIGURES
Fig. 1 shows the systemic cytokine production in mice after nanocarrier (NC) inoculation. Fig. 1A, Fig. IB and Fig. 1C - TNF-a, IL-6, and IL-12 production in experimental groups shown, respectively. Sera from groups of three mice were pooled and analyzed by ELISA.
Fig. 2 demonstrates the systemic IFN-γ production in mice after NC inoculation. Sera from groups of three mice were pooled and analyzed by ELISA.
Fig. 3 demonstrates that utilization of entrapped R848 within NCs generates an immune response, which is superior to one induced by NC without R848.
Fig. 4 shows anti-nicotine antibody titers in mice immunized with NC containing surface nicotine and T-helper ovalbumin-derived peptide OP-II (NC-Nic) without adjuvant or with the same NC-Nic admixed with 20 μg of free R848 (5 animals/group; s.c, 100 μg of NC per injection, 3 times with 4-wk intervals). Titers for days 26 and 40 after the first immunization are shown (ELISA against polylysine-nicotine) (group 1: immunized with NC[Nic,0(i.e., no adjuvant), OP-II] (2.2% of OP-II); group 2: immunized with NC[Nic,0,OP- II] admixed with 20 μg of free R848).
Fig. 5 shows anti-nicotine antibody titers in mice immunized with NC containing surface nicotine and T-helper ovalbumin-derived peptide OP-II (NC-Nic) with R848 adjuvant or with the same NC-Nic admixed with 80 μg of free alum or 25 μg of free CpG-1826 (5 animals/group; s.c, 100 μg of NC per injection, 3 times with 4-wk intervals). Titers for days 26 and 40 after the first immunization are shown (ELISA against polylysine-nicotine) (all groups: immunized with NC[Nic,R848,OP-II]; group 2: NC admixed with 80 μg of free alum; group 3: admixed with 25 μg of free CpG-1826).
Fig. 6 shows anti-nicotine antibody titers in mice immunized with NC containing surface nicotine, R848 and T-helper ovalbumin-derived peptide OP-II NC[Nic,R848, OP-II] or with the same NC-Nic admixed with 80 μg of free alum (5 animals/group; s.c, 100 μg of NC per injection, 3 times with 4-wk intervals). Titers for days 40 and 70 after the first immunization are shown (ELIS A against polylysine-nicotine) (group 1 : immunized with NC[Nic,R848,OP-II] (3.1% of R848, 1.5% of OP-II); group 2: immunized with
NC[Nic,R848,OP-II] admixed with 80 μg of free alum).
Fig. 7 shows specific local CTL response in mice immunized with NC containing ovalbumin or free ovalbumin. Mice were immunized once (s.c, 100 μg of NC, containing 2.8% of OVA, or with 2.5 μg of OVA; both immunogens admixed with 10 μg of free 1826- CpG).
Fig. 8 shows anti-nicotine antibody titers in mice immunized with NC containing surface nicotine and T-helper ovalbumin-derived peptide OP-II (NC-Nic) (no adjuvant within NC) admixed with 20 μg of free CpG (PS) or 20 μg of free CpG (PO) (5 animals/group; s.c, 100 μg of NC per injection, 3 times with 2-wk intervals). Control mice received PBS alone. Titers for days 26 and 40 are shown (ELIS A against polylysine-nicotine) (group 1 :
immunized with NC-Nic (no adjuvant) + free CpG (PS); group 2: immunized with NC-Nic (no adjuvant) + free CpG (PO); group 3: immunized with PBS only).
Fig. 9 shows anti-ovalbumin (OVA) antibody titers in mice immunized with NC containing surface OVA (NC-OVA) (no adjuvant within NC) admixed with 20 μg of free R848 or CpG (PS) (5 animals/group; s.c, 100 μg of NC per injection, 3 times with 2-wk intervals). Control mice were immunized with 2.5 μg of soluble OVA admixed with 20 μg of CpG (PS). Titers for days 26 and 44 are shown (ELIS A against OVA protein) (group 1: immunized with NC-OVA (no adjuvant) + free R848; group 2: immunized with NC-OVA (no adjuvant) + free CpG (PS); group 3: immunized with soluble OVA + CpG (PS)).
Fig. 10 shows anti-nicotine antibody titers in mice injected with CpG (20 μg per injection, 2 times with 2-wk intervals) followed by immunization at day 35 with NC containing surface nicotine and T-helper ovalbumin-derived peptide OP-II (NC-Nic) either with or without NC-contained R848 (5 animals/group; s.c, 100 μg of NC per injection, 2 times with 2-wk intervals). Titers for days 12, 26, and 40 after immunization with NC are shown (ELIS A against polylysine-nicotine) (group 1 : immunized with CpG followed by NC- Nic (R848 + OP-II); group 2: immunized with CpG followed by NC-Nic (OP-II only)).
DETAILED DESCRIPTION OF THE INVENTION
Before describing the present invention in detail, it is to be understood that this invention is not limited to particularly exemplified materials or process parameters as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments of the invention only, and is not intended to be limiting of the use of alternative terminology to describe the present invention.
All publications, patents and patent applications cited herein, whether supra or infra, are hereby incorporated by reference in their entirety for all purposes.
As used in this specification and the appended claims, the singular forms "a," "an" and "the" include plural referents unless the content clearly dictates otherwise. For example, reference to "a polymer" includes a mixture of two or more such molecules, reference to "a solvent" includes a mixture of two or more such solvents, reference to "an adhesive" includes mixtures of two or more such materials, and the like.
INTRODUCTION
The inventors have unexpectedly and surprisingly discovered that the problems and limitations noted above can be overcome by practicing the invention disclosed herein. The inventors have unexpectedly and surprisingly discovered that the administration of a population of synthetic nanocarriers and an adjuvant that is not coupled to any of the synthetic nanocarriers provides stronger and more rapid immune responses. In particular, the inventors have unexpectedly discovered that it is possible to provide compositions and methods that relate to a composition comprising a dosage form that comprises (1) a population of synthetic nanocarriers, (2) a first adjuvant that is not coupled to any synthetic nanocarriers, and (3) a pharmaceutically acceptable excipient.
In embodiments, the administration of adjuvant separate from synthetic nanocarriers leads to a rapid and strong systemic induction of pro-inflammatory cytokines, such as TNF-a, IL-6 and/or IL-12. The dose of the adjuvant or adjuvants in the compositions in some embodiments, therefore, is a systemic dose. In embodiments, the systemic dose results in the release of TNF-a, IL-6 or IL-12. In other embodiments, the systemic dose results in the systemic release of TNF-a, IL-6 and IL-12. As such cytokines are pro-inflammatory, the administration of compositions provided herein can be beneficial to subjects where an inflammatory response is desired. In some embodiments, therefore, the compositions provided are administered to such subjects. In embodiments, such subjects have or are at risk of having cancer. In other embodiments, such subjects have or are at risk of having an infection or an infectious disease. Methods for the administration of the compositions to such subjects are also provided.
In other embodiments, the administration of adjuvant separate from synthetic nanocarriers leads to a rapid and strong systemic induction of cytokines that are important for a Thl immune response, such as IFN-γ, IL-12 and/or IL-18. Therefore, the dose of the adjuvant or adjuvants in the compositions in some embodiments is a systemic dose that results in the systemic release of IFN-γ, IL-12 and/or IL-18. As such cytokines are important for a Thl immune response, the administration of compositions provided herein can be beneficial to subjects where a Thl immune response is desired. In some embodiments, the compositions provided are administered to such subjects. In embodiments, such subjects have or are at risk of having an atopic condition, asthma, chronic obstructive pulmonary disease (COPD) or a chronic infection. Methods for the administration of the compositions to such subjects are also provided.
The inventors have also unexpectedly discovered that it is possible to administer a second adjuvant with the aforementioned compositions to provide a strong humoral response. The aforementioned compositions, therefore, can further comprise a second adjuvant. In some embodiments, the second adjuvant is coupled to the synthetic nanocarriers. In other embodiments, the second adjuvant is not coupled to any synthetic nanocarriers. In still other embodiments, the second adjuvant is coupled to another population of synthetic nanocarriers. In some embodiments, however, the second adjuvant is administered to a subject at a time different from when the composition that comprises a population of synthetic nanocarriers and a first adjuvant that is not coupled to any synthetic nanocarriers is administered. In some embodiments, the second adjuvant is administered at a different time but is coadministered. In other embodiments, the second adjuvant is not coadministered. In still other embodiments, the second adjuvant is administered prior to or after the administration of the composition that comprises a population of synthetic nanocarriers and a first adjuvant that is not coupled to any synthetic nanocarriers. In some embodiments, the second adjuvant is also not coupled to any synthetic nanocarriers. In other embodiments, the second adjuvant is coupled to another population of synthetic nanocarriers. The compositions provided herein, therefore, can be beneficial to subjects where a humoral immune response is desired. In some embodiments, the compositions provided are administered to such subjects. In embodiments, such subjects have or are at risk of having cancer, an infection or infectious disease. Methods for the administration of the compositions to such subjects are also provided.
In further embodiments, it is demonstrated that the administration of one or more antigens with the compositions provided above provides a strong specific local cytotoxic T lymphocyte (CTL) response. In embodiments, the antigen(s) are coadministered with the compositions provided. In some embodiments, the antigen(s) are coupled to the synthetic nanocarriers. In other embodiments, the antigen(s) are not coupled to the synthetic nanocarriers but to another population of synthetic nanocarriers. The antigen(s) can comprise a B cell or T cell antigen. In some embodiments, the T cell antigen is a T helper cell antigen. In other embodiments, the antigen(s) comprise a B cell or T cell antigen as well as a T helper cell antigen. Therefore, the compositions provided can be beneficial to subjects where a specific local CTL response is desired. In some embodiments, the compositions provided are administered to such subjects. Methods for the administration of the compositions to such subjects are also provided.
The present invention will now be described in more detail.
DEFINITIONS
"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. Such 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), 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, ISCOMATRIX™, emulsions such as MF59™, Montanide® ISA 51 and ISA 720, AS02 (QS21+squalene+ MPL®), AS15, liposomes and liposomal formulations such as AS01, synthesized or specifically prepared microparticles and microcarriers such as bacteria-derived outer membrane vesicles (OMV) of N. 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. In embodiments, 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. In other embodiments, 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 imidazoquinolines; such as R848; adenine derivatives, such as those disclosed in US patent 6,329,381 (Sumitomo
Pharmaceutical Company), US Published Patent Application 2010/0075995 to Biggadike et al., or WO 2010/018132 to Campos et al.; immuno stimulatory DNA; or immuno stimulatory RNA. In specific embodiments, synthetic nanocarriers incorporate as adjuvants compounds that are agonists for toll-like receptors (TLRs) 7 & 8 ("TLR 7/8 agonists"). Of utility are the TLR 7/8 agonist compounds disclosed in US Patent 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). In specific
embodiments, an adjuvant may be an agonist for the DC surface molecule CD40. In certain embodiments, to stimulate immunity rather than tolerance, 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. In embodiments, adjuvants also may comprise immuno stimulatory RNA molecules, such as but not limited to dsRNA, poly I:C or poly Lpoly C12U (available as
Ampligen ®, both poly I:C and poly I:polyC12U being known as TLR3 stimulants), and/or those disclosed in F. Heil et al., "Species-Specific Recognition of Single-Stranded RNA via Toll-like Receptor 7 and 8" Science 303(5663), 1526-1529 (2004); J. Vollmer et al.,
"Immune modulation by chemically modified ribonucleosides and oligoribonucleotides" WO 2008033432 A2; A. Forsbach et al., "Immunostimulatory oligoribonucleotides containing specific sequence motif(s) and targeting the Toll-like receptor 8 pathway" WO 2007062107 A2; E. Uhlmann et al., "Modified oligoribonucleotide analogs with enhanced
immunostimulatory activity" U.S. Pat. Appl. Publ. US 2006241076; G. Lipford et al., "Immunostimulatory viral RNA oligonucleotides and use for treating cancer and infections" WO 2005097993 A2; G. Lipford et al., "Immunostimulatory G,U-containing
oligoribonucleotides, compositions, and screening methods" WO 2003086280 A2. In some embodiments, an adjuvant may be a TLR-4 agonist, such as bacterial lipopolysacccharide (LPS), VSV-G, and/or HMGB-1. In some embodiments, adjuvants may comprise TLR-5 agonists, such as flagellin, or portions or derivatives thereof, including but not limited to those disclosed in US Patents 6,130,082, 6,585,980, and 7,192,725. In specific embodiments, synthetic nanocarriers incorporate a ligand for Toll-like receptor (TLR)-9, such as
immuno stimulatory DNA molecules comprising CpGs, which induce type I interferon secretion, and stimulate T and B cell activation leading to increased antibody production and cytotoxic T cell responses (Krieg et al., CpG motifs in bacterial DNA trigger direct B cell activation. Nature. 1995. 374:546-549; Chu et al. CpG oligodeoxynucleotides act as adjuvants that switch on T helper 1 (Thl) immunity. J. Exp. Med. 1997. 186:1623-1631; Lipford et al. CpG-containing synthetic oligonucleotides promote B and cytotoxic T cell responses to protein antigen: a new class of vaccine adjuvants. Eur. J. Immunol. 1997.
27:2340-2344; Roman et al. Immunostimulatory DNA sequences function as T helper- 1- promoting adjuvants. Nat. Med. 1997. 3:849-854; Davis et al. CpG DNA is a potent enhancer of specific immunity in mice immunized with recombinant hepatitis B surface antigen. J. Immunol. 1998. 160:870-876; Lipford et al., Bacterial DNA as immune cell activator. Trends Microbiol. 1998. 6:496-500; US Patent 6,207,646 to Krieg et al.; US Patent 7,223,398 to Tuck et al.; US Patent 7,250,403 to Van Nest et al.; or US Patent 7,566,703 to Krieg et al.
In some embodiments, adjuvants may be proinflammatory stimuli released from necrotic cells (e.g., urate crystals). In some embodiments, adjuvants may be activated components of the complement cascade (e.g., CD21, CD35, etc.). In some embodiments, adjuvants may be activated components of immune complexes. The adjuvants also include complement receptor agonists, such as a molecule that binds to CD21 or CD35. In some embodiments, the complement receptor agonist induces endogenous complement
opsonization of the synthetic nanocarrier. In some embodiments, adjuvants are cytokines, which are small proteins or biological factors (in the range of 5 kD - 20 kD) that are released by cells and have specific effects on cell-cell interaction, communication and behavior of other cells. In some embodiments, the cytokine receptor agonist is a small molecule, antibody, fusion protein, or aptamer.
In embodiments, at least a portion of the dose of adjuvant is not coupled to any synthetic nanocarriers, preferably, all of the dose of adjuvant is not coupled to any synthetic nanocarriers. In embodiments, the dose of adjuvant comprises two or more types of adjuvants, and at least a portion of at least one of the types of adjuvant is not coupled to any synthetic nanocarriers. For instance, and without limitation, adjuvants that act on different receptors, such as different TLR receptors, may be combined. As an example, in an embodiment a TLR 7/8 agonist may be combined with a TLR 9 agonist. In another embodiment, a TLR 7/8 agonist may be combined with a TLR 4 agonist. In yet another embodiment, a TLR 9 agonist may be combined with a TLR 3 agonist.
"Administering" or "administration" means providing a substance (e.g., 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 immune response to an allergen (i.e., allergic reaction). Allergies or allergic conditions 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.
"Amount effective" is any amount of a composition provided herein that produces 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 of an inflammatory, a Thl, a humoral or specific local CTL immune response. Such subjects include those that have or are at risk of having cancer, an infection or infectious disease, an atopic condition, asthma, chronic obstructive pulmonary disease (COPD) or a chronic infection.
Amounts effective include those that involve the systemic release of one or more cytokines. In embodiments, the amounts effective include those that involve the production of a systemic cytokine release profile. In some embodiments, the one or more cytokines or cytokine release profile comprises the systemic release of TNF-oc, IL-6 and/or IL-12. In other embodiments, the one or more cytokines or cytokine release profile comprises the systemic release of IFN-γ, IL-12 and/or IL-18. This can be monitored by routine methods. An amount that is effective to produce one or more desired immune responses 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 reasons.
In general, doses of the compositions of the invention can range from about 10 g/kg to about 100,000 μg/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 106,
10 7, 108, 109 or 1010 synthetic nanocarriers per dose. Other examples of useful doses include from about lxlO6 to about lxlO10, about lxlO7 to about lxlO9 or about lxlO8 to about lxlO9 synthetic nanocarriers per dose. In some embodiments, the doses of the compositions provided are systemic doses.
"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. In embodiments, antigens are coadministered with the synthetic nanocarriers. In other embodiments, antigens are not coadministered with the synthetic nanocarriers. "Type(s) of antigens" means molecules that share the same, or substantially the same, antigenic characteristics. In embodiments, antigens of the compositions provided are associated with the disease or condition that is being treated. For examples, the antigen can be an allergen (for the treatment of an allergy or allergic condition), a cancer-associated antigen (for the treatment of cancer or a tumor), an infectious agent antigen (for the treatment of an infection, an infectious disease or a chronic infectious disease), etc.
"At least a portion of the dose" means at least some part of the dose, ranging up to including all of the dose.
An "at risk" subject is one in which a health practitioner believes has a chance of having as disease or condition as provided herein. "B cell antigen" means any antigen that is recognized by a B cell, and triggers an immune response in a B cell (e.g., an antigen that is specifically recognized by a B cell receptor on a B cell). In some embodiments, an antigen that is a T cell antigen is also a B cell antigen. In other embodiments, the T cell antigen is not also a B cell antigen. B cell antigens include, but are not limited to proteins, peptides, small molecules, and carbohydrates. In some embodiments, the B cell antigen comprises a non-protein antigen (i.e., not a protein or peptide antigen). In some embodiments, the B cell antigen comprises a carbohydrate associated with an infectious agent. In some embodiments, the B cell antigen comprises a glycoprotein or glycopeptide associated with an infectious agent. The infectious agent can be a bacterium, virus, fungus, protozoan, parasite or prion. In some embodiments, the B cell antigen comprises a poorly immunogenic antigen. In some embodiments, the B cell antigen comprises an abused substance or a portion thereof. In some embodiments, 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. In some embodiments, the B cell antigen comprises a toxin, such as a toxin from a chemical weapon or natural source, or a pollutant. The B cell antigen may also comprise a hazardous environmental agent. In other embodiments, 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, an addictive substance, a xenoantigen, or a metabolic disease enzyme or enzymatic product thereof.
"Coadministered" means administering two or more substances to a subject in a manner that is correlated in time, preferably sufficiently correlated in time so as to provide a modulation in an immune response. In embodiments, coadministration may occur through administration of two or more substances in the same dosage form. In other embodiments, coadministration may encompass administration of two or more substances in different dosage forms, but within a specified period of time, preferably within 1 month, more preferably within 1 week, still more preferably within 1 day, and even more preferably within 1 hour.
"Couple" or "Coupled" or "Couples" (and the like) means to chemically associate one entity (for example a moiety) with another. In some embodiments, the coupling is covalent, meaning that the coupling occurs in the context of the presence of a covalent bond between the two entities. In non-covalent embodiments, 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. In embodiments, encapsulation is a form of coupling. In embodiments, at least a portion of a dose of adjuvant(s) is not coupled to any synthetic nanocarriers, preferably all of a dose of adjuvant(s) is not coupled to any synthetic
nanocarriers.
"Derived" means taken from a source and subjected to substantial modification. For instance, a peptide or nucleic acid with a sequence with only 50% identity to a natural peptide or nucleic acid, preferably a natural consensus peptide or nucleic acid, would be said to be derived from the natural peptide or nucleic acid. Substantial modification is modification that significantly affects the chemical or immunological properties of the material in question. Derived peptides and nucleic acids can also include those with a sequence with greater than 50% identity to a natural peptide or nucleic acid sequence if said derived peptides and nucleic acids have altered chemical or immunological properties as compared to the natural peptide or nucleic acid. These chemical or immunological properties comprise hydrophilicity, stability, affinity, and ability to couple with a carrier such as a synthetic nanocarrier.
"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 within a synthetic nanocarrier, preferably enclose completely within a synthetic nanocarrier. Most or all of a substance that is encapsulated is not exposed to the local environment external to the synthetic nanocarrier. 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.
"Humoral response" means any immune response that results in the production or stimulation of B cells and/or the production of antibodies. Preferably, the humoral immune response is specific to an antigen comprised within an inventive composition or administered during the practice of an inventive method. Methods for assessing whether a humoral response is induced are known to those of ordinary skill in the art. Examples of such methods are provided below in the Examples. An "infection" or "infectious disease" is any condition or disease caused by a microorganism, pathogen or other agent, such as a bacterium, fungus, prion or virus.
Examples of infectious disease 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, Diphtheria, Epidemic Typhus, Gonorrhea, Impetigo, Legionellosis, Leprosy (Hansen's Disease), Leptospirosis, Listeriosis, Lyme disease, Melioidosis, Rheumatic Fever, MRSA infection, Nocardiosis, Pertussis (Whooping Cough), Plague, Pneumococcal pneumonia, Psittacosis, Q fever, Rocky Mountain Spotted Fever (RMSF), Salmonellosis, Scarlet Fever, Shigellosis, Syphilis, Tetanus, Trachoma, Tuberculosis, Tularemia, Typhoid Fever, Typhus and Urinary Tract Infections; parasitic infectious diseases, such as African trypanosomiasis, Amebiasis, Ascariasis, Babesiosis, Chagas Disease, Clonorchiasis, Cryptosporidiosis, Cysticercosis, Diphyllobothriasis, Dracunculiasis, Echinococcosis, Enterobiasis, Fascioliasis,
Fasciolopsiasis, Filariasis, Free-living amebic infection, Giardiasis, Gnathostomiasis,
Hymenolepiasis, Isosporiasis, Kala-azar, Leishmaniasis, Malaria, Metagonimiasis, Myiasis, Onchocerciasis, Pediculosis, Pinworm Infection, Scabies, Schistosomiasis, Taeniasis, Toxocariasis, Toxoplasmosis, Trichinellosis, Trichinosis, Trichuriasis, Trichomoniasis and Trypanosomiasis; fungal infectious disease, such as Aspergillosis, Blastomycosis,
Candidiasis, Coccidioidomycosis, Cryptococcosis, Histoplasmosis, Tinea pedis (Athlete's Foot) and Tinea cruris; prion infectious diseases, such as Alpers' disease, Fatal Familial Insomnia, Gerstmann-Straussler-Scheinker syndrome, Kuru and Variant Creutzfeldt- Jakob disease.
"Inflammatory response" means any immune response involved in the body's innate immune defense system that operates in response to, for example, exposure to an infectious agent, cell injury, etc. In embodiments, the inflammatory response includes the systemic release of cytokines, such as TNF-oc, IL-6 and/or IL-12. Methods for assessing whether an inflammatory response is induced, such as an assessment of the production of pro- inflammatory cytokines, are known to those of ordinary skill in the art. Examples of such methods are provided below in the Examples.
"Isolated nucleic acid" means a nucleic acid that is separated from its native environment and present in sufficient quantity to permit its identification or use. An isolated nucleic acid may be one that is (i) amplified in vitro by, for example, polymerase chain reaction (PCR); (ii) recombinantly produced by cloning; (iii) purified, as by cleavage and gel separation; or (iv) synthesized by, for example, chemical synthesis. An isolated nucleic acid is one which is readily manipulable by recombinant DNA techniques well known in the art. Thus, a nucleotide sequence contained in a vector in which 5' and 3' restriction sites are known or for which polymerase chain reaction (PCR) primer sequences have been disclosed is considered isolated but a nucleic acid sequence existing in its native state in its natural host is not. An isolated nucleic acid may be substantially purified, but need not be. For example, a nucleic acid that is isolated within a cloning or expression vector is not pure in that it may comprise only a tiny percentage of the material in the cell in which it resides. Such a nucleic acid is isolated, however, as the term is used herein because it is readily manipulable by standard techniques known to those of ordinary skill in the art. Any of the nucleic acids provided herein may be isolated. In some embodiments, the antigens in the compositions provided herein are present in the form of an isolated nucleic acid, such as an isolated nucleic acid that encodes an antigenic peptide, polypeptide or protein.
"Isolated peptide, polypeptide or protein" means the polypeptide (or peptide or protein) is separated from its native environment and present in sufficient quantity to permit its identification or use. This means, for example, the polypeptide (or peptide or protein) may be (i) selectively produced by expression cloning or (ii) purified as by chromatography or electrophoresis. Isolated peptides, proteins or polypeptides may be, but need not be, substantially pure. Because an isolated peptide, polypeptide or protein may be admixed with a pharmaceutically acceptable carrier in a pharmaceutical preparation, the polypeptide (or peptide or protein) may comprise only a small percentage by weight of the preparation. The polypeptide (or peptide or protein) 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 proteins (or peptides or polypeptides). Any of the peptides, polypeptides or proteins provided herein may be isolated. In some embodiments, the antigens in the compositions provided herein are in the form of isolated peptides, polypeptides or proteins. "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 spheriodal 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. In an embodiment, 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 100 nm. In an embodiment, 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 μπι. Preferably, 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 inventive synthetic nanocarriers may vary depending on the embodiment. For instance, 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 1000:1, still preferably from 1:1 to 100:1, and yet more preferably from 1:1 to 10:1. Preferably, 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 μπι, more preferably equal to or less than 2 μπι, more preferably equal to or less than 1 μπι, 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. In preferred embodiments, 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 greater than lOOnm, more preferably equal to or greater than 120nm, 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 sizes is obtained by suspending the synthetic nanocarriers in a liquid (usually aqueous) media and using dynamic light scattering (e.g. using a Brookhaven ZetaPALS instrument).
"Obtained" means taken from a source without substantial modification. Substantial modification is modification that significantly affects the chemical or immunological properties of the material in question. For example, as a non-limiting example, a peptide or nucleic acid with a sequence with greater than 90%, preferably greater than 95%, preferably greater than 97%, preferably greater than 98%, preferably greater than 99%, preferably 100%, identity to a natural peptide or nucleotide sequence, preferably a natural consensus peptide or nucleotide sequence, and chemical and/or immunological properties that are not significantly different from the natural peptide or nucleic acid would be said to be obtained from the natural peptide or nucleotide sequence. These chemical or immunological properties comprise hydrophilicity, stability, affinity, and ability to couple with a carrier such as a synthetic nanocarrier.
"Pharmaceutically acceptable carrier or excipient" means a pharmacologically inactive material used together with the recited synthetic nanocarriers to formulate the inventive compositions. Pharmaceutically acceptable carriers or 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. In some embodiments, pharmaceutically acceptable carriers or excipients comprise calcium carbonate, calcium phosphate, various diluents, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
"Specific local cytotoxic T lymphocyte (CTL) response" means any stimulation, induction or proliferation of cytotoxic T cells, preferably cytotoxic T cells that are specific to an antigen. In embodiments, the antigen is associated with any of the diseases or conditions provided herein. In some embodiments, the antigen is comprised within an inventive composition or is administered in an inventive method provided herein. Methods for assessing CTL response are known to those of skill in the art. An examples of such a method is provided in the Examples.
"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. In embodiments, inventive synthetic nanocarriers do not comprise chitosan.
A synthetic nanocarrier can be, but is not limited to, one or a plurality of lipid-based nanoparticles(e.g. liposomes) (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. 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, including but not limited to internal surfaces (surfaces generally facing an interior portion of the synthetic nanocarrier) and external surfaces (surfaces generally facing an external environment of the synthetic nanocarrier). Exemplary synthetic nanocarriers that can be adapted for use in the practice of the present invention comprise: (1) the biodegradable nanoparticles disclosed in US Patent 5,543,158 to Gref et al., (2) the polymeric nanoparticles of Published US Patent Application 20060002852 to Saltzman et al., (3) the lithographically constructed nanoparticles of Published US Patent Application 20090028910 to DeSimone et al., (4) the disclosure of WO 2009/051837 to von Andrian et al., (5) the nanoparticles disclosed in Published US Patent Application 2008/0145441 to Penades et al., (6) the protein nanoparticles disclosed in Published US Patent Application 20090226525 to de los Rios et al., (7) the virus-like particles disclosed in published US Patent Application 20060222652 to Sebbel et al., (8) the nucleic acid coupled virus-like particles disclosed in published US Patent Application 20060251677 to Bachmann et al., (9) the virus-like particles disclosed in WO2010047839A1 or WO2009106999 A2, or (10) the nanoprecipitated nanoparticles disclosed in P. Paolicelli et al., "Surface-modified PLGA-based Nanoparticles that can Efficiently Associate and Deliver Virus-like Particles" Nanomedicine. 5(6):843-853 (2010). In embodiments, 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. In a preferred embodiment, 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. In a more preferred embodiment, 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. In embodiments, 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.
"Systemic dose" means a dose of an adjuvant that provides a particular systemic cytokine release, preferably a particular systemic cytokine release profile. In some embodiments, the particular systemic cytokine release, preferably a particular systemic cytokine release profile, is in a human. In embodiments, the compositions and methods provided herein (where at least a portion of a dose of adjuvant is not coupled to any nanocarriers) result in a particular systemic cytokine release profile in a subject. The term "separately" is also used to mean adjuvant that is not coupled to any synthetic nanocarriers. Additionally, "systemic cytokine release profile" means a pattern of systemic cytokine release, wherein the pattern comprises cytokine levels measured for several different systemic cytokines. In some embodiments, the particular systemic cytokine release profile comprises the systemic release of TNF-oc, IL-6 and/or IL-12. In other embodiments, the particular systemic cytokine release profile comprises the systemic release of IFN-γ, IL12 and/or IL-18.
"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.) In some embodiments, an antigen that is a T cell antigen is also a B cell antigen. In other embodiments, the T cell antigen is not also a B cell antigen. T cell antigens generally are proteins, polypeptides 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.
In some embodiments the T cell antigen is a 'universal' T cell antigen, or T cell memory antigen, (i.e., one to which a subject has a pre-existing memory and that can be used to boost T cell help to an unrelated antigen, for example an unrelated B cell antigen).
Universal T cell antigens include tetanus toxoid, as well as one or more peptides derived from tetanus toxoid, Epstein-Barr virus, or influenza virus. Universal T cell antigens also include a components of influenza virus, such as hemagglutinin, neuraminidase, or nuclear protein, or one or more peptides derived therefrom. In some embodiments, the universal T cell antigen is not one that is presented in a complex with a MHC molecule. In some embodiments, the universal T cell antigen is not complexed with a MHC molecule for presentation to a T helper cell. Accordingly, in some embodiments, the universal T cell antigen is not a T helper cell antigen. However, in other embodiments, the universal T cell antigen is a T helper cell antigen.
In embodiments, 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. US Patent 7,202,351). In other embodiments, a T-helper cell antigen may comprise ovalbumin or a peptide obtained or derived therefrom. Preferably, the ovalbumin comprises the amino acid sequence as set forth in Accession No. AAB59956, NP .990483.1, AAA48998, or CAA2371. In other
embodiments, the peptide obtained or derived from ovalbumin comprises the following amino acid sequence: H-Ile-Ser-Gln-Ala-Val-His-Ala-Ala-His-Ala-Glu-Ile-Asn-Glu-Ala- Gly-Arg-OH (SEQ ID NO: 1). In other embodiments, a T-helper cell antigen may comprise one or more lipids, or glycolipids, including but not limited to: a-galactosylceramide (a -
GalCer), a -linked glycosphingolipids (from Sphingomonas spp.), galactosyl diacylglycerols (from Borrelia burgdorferi), lypophosphoglycan (from Leishmania donovani), and phosphatidylinositol tetramannoside (PEVI4) (from Mycobacterium leprae). For additional lipids and/or glycolipids useful as T-helper cell antigen, see V. Cerundolo et al., "Harnessing invariant NKT cells in vaccination strategies." Nature Rev Immun, 9:28-38 (2009).
In embodiments, CD4+ T-cell antigens may be derivatives of a CD4+ T-cell antigen that is obtained from a source, such as a natural source. In such embodiments, 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. In embodiments, the T cell antigen, preferably a universal T cell antigen or T-helper cell antigen, may be coupled to, or uncoupled from, a synthetic nanocarrier. In some embodiments, the universal T cell antigen or T-helper cell antigen is encapsulated in the synthetic nanocarriers of the inventive compositions.
"Thl immune response" means any immune response that results in the production of Thl cells and Thl-associated cytokines, IFN-γ, IL-12 and/or IL-18, or that counteracts the differentiation of Th2 cells and the action of Th2 cytokines. Methods for assessing whether a Thl immune response is induced are known to those of ordinary skill in the art. Examples of such methods are provided below in the Examples.
"Time different from administration" or "a time different from a time when the composition is administered" means a time more than about 30 seconds either before or after administration, preferably more than about 1 minute either before or after administration, more preferably more than 5 minutes either before or after administration, still more preferably more than 1 day either before or after administration, still more preferably more than 2 days either before or after administration, still more preferably more than 1 week either before or after administration, still more preferably more than 2 weeks either before or after administration, still more preferably more than 3 weeks either before or after administration, still more preferably more than 1 month either before or after administration, and still more preferably more than 2 months either before or after administration.
"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 or against an antigen derived from an infectious agent for the treatment of an infection or infectious disease). In embodiments, a vaccine may comprise dosage forms according to the invention. Preferably, in some embodiments, these vaccines comprise an adjuvant not coupled to any synthetic nanocarriers.
In specific embodiments, the inventive compositions incorporate adjuvants that comprise agonists for toll-like receptors (TLRs) 7 & 8 ("TLR 7/8 agonists"). Of utility are the TLR 7/8 agonist compounds disclosed in U.S. Patent 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 R848.
In specific embodiments, the inventive compositons incorporate adjuvants that comprise a ligand for Toll-like receptor (TLR)-9, such as immunostimulatory DNA molecules comprising CpGs, which induce type I interferon secretion, and stimulate T and B cell activation leading to increased antibody production and cytotoxic T cell responses (Krieg et al., CpG motifs in bacterial DNA trigger direct B cell activation. Nature. 1995. 374:546- 549; Chu et al. CpG oligodeoxynucleotides act as adjuvants that switch on T helper 1 (Thl) immunity. J. Exp. Med. 1997. 186:1623-1631; Lipford et al. CpG-containing synthetic oligonucleotides promote B and cytotoxic T cell responses to protein antigen: a new class of vaccine adjuvants. Eur. J. Immunol. 1997. 27:2340-2344; Roman et al. Immunostimulatory DNA sequences function as T helper- 1 -promoting adjuvants. Nat. Med. 1997. 3:849-854; Davis et al. CpG DNA is a potent enhancer of specific immunity in mice immunized with recombinant hepatitis B surface antigen. J. Immunol. 1998. 160:870-876; Lipford et al., Bacterial DNA as immune cell activator. Trends Microbiol. 1998. 6:496-500. In
embodiments, CpGs may comprise modifications intended to enhance stability, such as phosphorothioate linkages, or other modifications, such as modified bases. See, for example, U.S. Patents, 5,663,153, 6,194,388, 7,262,286, or 7,276,489. In certain embodiments, to stimulate immunity rather than tolerance, a composition provided herein 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 responses and anti-viral immunity. In some embodiments, the adjuvant comprises a TLR-4 agonist, such as bacterial lipopolysacharide (LPS), VSV-G, and/or HMGB-1. In some embodiments, adjuvants comprise cytokines, which are small proteins or biological factors (in the range of 5 kD - 20 kD) that are released by cells and have specific effects on cell-cell interaction, communication and behavior of other cells. In some embodiments, adjuvants comprise proinflammatory stimuli released from necrotic cells (e.g., urate crystals). In some embodiments, adjuvants comprise activated components of the complement cascade (e.g., CD21, CD35, etc.). In some embodiments, adjuvants comprise activated components of immune complexes. The adjuvants also include those that comprise complement receptor agonists, such as a molecule that binds to CD21 or CD35. In some embodiments, the complement receptor agonist induces endogenous complement opsonization of the nanocarrier. Adjuvants also include those that comprise cytokine receptor agonists, such as a cytokine.
In some embodiments, the cytokine receptor agonist is a small molecule, antibody, fusion protein, or aptamer. In embodiments, adjuvants also may comprise
immunostimulatory RNA molecules, such as but not limited to dsRNA or poly I:C (a TLR3 stimulant), and/or those disclosed in F. Heil et al., "Species-Specific Recognition of Single- Stranded RNA via Toll-like Receptor 7 and 8" Science 303(5663), 1526-1529 (2004); J. Vollmer et al., "Immune modulation by chemically modified ribonucleosides and
oligoribonucleotides" WO 2008033432 A2; A. Forsbach et al., "Immunostimulatory oligoribonucleotides containing specific sequence motif(s) and targeting the Toll-like receptor 8 pathway" WO 2007062107 A2; E. Uhlmann et al., "Modified oligoribonucleotide analogs with enhanced immunostimulatory activity" U.S. Pat. Appl. Publ. US 2006241076; G. Lipford et al., "Immunostimulatory viral RNA oligonucleotides and use for treating cancer and infections" WO 2005097993 A2; G. Lipford et al., "Immunostimulatory G,U-containing oligoribonucleotides, compositions, and screening methods" WO 2003086280 A2.
In some embodiments, the adjuvants comprise gel-type adjuvants (e.g., aluminum hydroxide, aluminum phosphate, calcium phosphate, etc.), microbial adjuvants (e.g., immunomodulatory DNA sequences that include CpG motifs; immunostimulatory RNA molecules; endotoxins such as monophosphoryl lipid A; exotoxins such as cholera toxin, E. coli heat labile toxin, and pertussis toxin; muramyl dipeptide, etc.); oil-emulsion and emulsifier-based adjuvants (e.g., Freund's Adjuvant, MF59 [Novartis], SAF, etc.); particulate adjuvants (e.g., liposomes, biodegradable microspheres, saponins, etc.); synthetic adjuvants (e.g., nonionic block copolymers, muramyl peptide analogues, polyphosphazene, synthetic polynucleotides, etc.), and/or combinations thereof.
SYNTHETIC NANOCARRIER COMPOSITIONS
A wide variety of synthetic nanocarriers can be used according to the invention. In some embodiments, synthetic nanocarriers are spheres or spheroids. In some embodiments, synthetic nanocarriers are flat or plate-shaped. In some embodiments, synthetic nanocarriers are cubes, cuboidal or cubic. In some embodiments, synthetic nanocarriers are ovals or ellipses. In some embodiments, synthetic nanocarriers are cylinders, cones, or pyramids.
In some embodiments, it is desirable to use 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). To give but one example, 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.
In some embodiments, synthetic nanocarriers may optionally comprise one or more lipids. In some embodiments, a synthetic nanocarrier may comprise a liposome. In some embodiments, a synthetic nanocarrier may comprise a lipid bilayer. In some embodiments, a synthetic nanocarrier may comprise a lipid monolayer. In some embodiments, a synthetic nanocarrier may comprise a micelle. In some embodiments, a synthetic nanocarrier may comprise a core comprising a polymeric matrix surrounded by a lipid layer (e.g., lipid bilayer, lipid monolayer, etc.). In some embodiments, 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 lipid layer (e.g., lipid bilayer, lipid monolayer, etc.).
In some embodiments, synthetic nanocarriers can comprise one or more polymers or polymeric matrices. In some embodiments, such a polymer or polymeric matrix can be surrounded by a coating layer (e.g., liposome, lipid monolayer, micelle, etc.). In some embodiments, various elements of the synthetic nanocarriers can be coupled with the polymer or polymeric matrix. In some embodiments, an element, such as a targeting moiety, oligonucleotide, antigen, adjuvant, etc. can be covalently associated with a polymeric matrix. In some embodiments, covalent association is mediated by a linker. In some embodiments, an element can be noncovalently associated with a polymeric matrix. For example, in some embodiments, an element can be encapsulated within, surrounded by, and/or dispersed throughout a polymeric matrix. Alternatively or additionally, an element can be associated with a polymeric matrix by hydrophobic interactions, charge interactions, van der Waals forces, etc.
A wide variety of polymers and methods for forming polymeric matrices therefrom are known conventionally. In general, 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. Typically, polymers in accordance with the present invention are organic polymers.
Examples of polymers suitable for use in the present invention include, but are not limited to polyethylenes, polycarbonates (e.g. poly(l,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., poly(P-hydroxyalkanoate)), poly(orthoesters), polycyanoacrylates, polyvinyl alcohols, polyurethanes, polyphosphazenes, polyacrylates, polymethacrylates, polyureas, polystyrenes, polyamines, polylysine, polylysine-PEG copolymers, and poly(ethyleneimine), poly(ethylene imine)-PEG copolymers.
In some embodiments, 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(l,3-dioxan-2one)); polyanhydrides (e.g., poly(sebacic anhydride)); polyethers (e.g., polyethylene glycol); polyurethanes; polymethacrylates; polyacrylates; and
polycyanoacrylates.
In some embodiments, polymers can be hydrophilic. For example, 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). In some embodiments, a synthetic nanocarrier comprising a hydrophilic polymeric matrix generates a hydrophilic environment within the synthetic nanocarrier. In some embodiments, polymers can be hydrophobic. In some embodiments, 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.
In some embodiments, polymers may be modified with one or more moieties and/or functional groups. A variety of moieties or functional groups can be used in accordance with the present invention. In some embodiments, 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. Patent No. 5543158 to Gref et al., or WO publication WO2009/051837 by Von Andrian et al.
In some embodiments, polymers may be modified with a lipid or fatty acid group. In some embodiments, a fatty acid group may be one or more of butyric, caproic, caprylic, capric, lauric, myristic, palmitic, stearic, arachidic, behenic, or lignoceric acid. In some embodiments, 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.
In some embodiments, 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." In some embodiments, 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. In some embodiments, 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[a-(4-aminobutyl)-L-glycolic acid], and derivatives thereof.
In some embodiments, 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. In some embodiments, 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.
In some embodiments, polymers may be one or more acrylic polymers. In certain embodiments, 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 ammonium groups.
In some embodiments, polymers can be cationic polymers. In general, 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), polyethylene imine) (PEI; Boussif et al., 1995, Proc. Natl. Acad. Sci., USA, 1995, 92:7297), and poly(amidoamine) dendrimers (Kukowska-Latallo et al., 1996, Proc. Natl. Acad. Sci., USA, 93:4897; Tang et al., 1996, Bioconjugate Chem., 7:703; and Haensler et al., 1993, Bioconjugate Chem., 4:372) are positively-charged at physiological pH, form ion pairs with nucleic acids, and mediate transfection in a variety of cell lines. In embodiments, the inventive synthetic nanocarriers may not comprise (or may exclude) cationic polymers.
In some embodiments, 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). Examples of these polyesters include poly(L-lactide-co-L-lysine) (Barrera et al., 1993, J. Am. Chem. Soc, 115:11010), poly(serine ester) (Zhou et al., 1990, Macromolecules, 23:3399), poly(4- hydroxy-L-proline ester) (Putnam et al., 1999, Macromolecules, 32:3658; and Lim et al., 1999, J. Am. Chem. Soc, 121:5633), and poly(4-hydroxy-L-proline ester) (Putnam et al., 1999, Macromolecules, 32:3658; and Lim et al., 1999, J. Am. Chem. Soc, 121:5633).
The properties of these and other polymers and methods for preparing them are well known in the art (see, for example, U.S. Patents 6,123,727; 5,804,178; 5,770,417; 5,736,372; 5,716,404; 6,095,148; 5,837,752; 5,902,599; 5,696,175; 5,514,378; 5,512,600; 5,399,665; 5,019,379; 5,010,167; 4,806,621; 4,638,045; and 4,946,929; Wang et al., 2001, J. Am. Chem. Soc, 123:9480; Lim et al., 2001, J. Am. Chem. Soc, 123:2460; Langer, 2000, Acc. Chem. Res., 33:94; Langer, 1999, J. Control. Release, 62:7; and Uhrich et al., 1999, Chem. Rev., 99:3181). More generally, a variety of methods for synthesizing certain suitable polymers are described in Concise Encyclopedia of Polymer Science and Polymeric Amines and
Ammonium Salts, Ed. by Goethals, Pergamon Press, 1980; Principles of Polymerization by Odian, John Wiley & Sons, Fourth Edition, 2004; Contemporary Polymer Chemistry by AUcock et al., Prentice-Hall, 1981; Deming et al., 1997, Nature, 390:386; and in U.S. Patents 6,506,577, 6,632,922, 6,686,446, and 6,818,732.
In some embodiments, 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 inventive 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.
In some embodiments, the synthetic nanocarriers comprise one or more polymers.
The polymeric synthetic nanocarriers, therefore, can also include those described in WO publication WO2009/051837 by Von Andrian et al., including, but not limited to those, with one or more hydrophilic components. Preferably, the one or more polymers comprise a polyester, such as a poly(lactic acid), poly(glycolic acid), poly(lactic-co-glycolic acid), or polycaprolactone. More preferably, the one or more polymers comprise or further comprise a polyester coupled to a hydrophilic polymer, such as a polyether. In embodiments, the polyether comprises polyethylene glycol. Still more preferably, the one or more polymers comprise a polyester and a polyester coupled to a hydrophilic polymer, such as a polyether. In other embodiments, the one or more polymers are coupled to one or more antigens and/or one or more adjuvants. In embodiments, at least some of the polymers are coupled to the antigen(s) and/or at least some of the polymers are coupled to the adjuvant(s). Preferably, when there are more than one type of polymer, one of the types of polymer is coupled to the antigen(s). In embodiments, one of the other types of polymer is coupled to the adjuvant(s). For example, in embodiments, when the nanocarriers comprise a polyester and a polyester coupled to a hydrophilic polymer, such as a polyether, the polyester is coupled to the adjuvant, while the polyester coupled to the hydrophilic polymer, such as a polyether, is coupled to the antigen(s). In embodiments, where the nanocarriers comprise a T helper cell antigen, the T helper cell antigen can be encapsulated in the nanocarrier.
In some embodiments, synthetic nanocarriers may not comprise a polymeric component. In some embodiments, synthetic nanocarriers may comprise metal particles, quantum dots, ceramic particles, etc. In some embodiments, a non-polymeric synthetic nanocarrier is an aggregate of non-polymeric components, such as an aggregate of metal atoms (e.g., gold atoms).
In some embodiments, synthetic nanocarriers may optionally comprise one or more amphiphilic entities. In some embodiments, an amphiphilic entity can promote the production of synthetic nanocarriers with increased stability, improved uniformity, or increased viscosity. In some embodiments, 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. Such 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 (Tween®20); polysorbate 60 (Tween®60);
polysorbate 65 (Tween®65); polysorbate 80 (Tween®80); polysorbate 85 (Tween®85); polyoxyethylene monostearate; surfactin; a poloxomer; a sorbitan fatty acid ester such as sorbitan trioleate; lecithin; lysolecithin; phosphatidylserine; phosphatidylinositol;sphingomyelin; phosphatidylethanolamine (cephalin); cardiolipin; phosphatidic acid; cerebrosides; dicetylphosphate; dipalmitoylphosphatidylglycerol;
stearylamine; dodecylamine; hexadecyl-amine; acetyl palmitate; glycerol ricinoleate;
hexadecyl sterate; isopropyl myristate; tyloxapol; poly(ethylene glycol)5000- phosphatidylethanolamine; poly(ethylene glycol)400-monostearate; phospholipids; synthetic and/or natural detergents having high surfactant properties; deoxycholates; cyclodextrins; chaotropic salts; ion pairing agents; and combinations thereof. An 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.
In some embodiments, synthetic nanocarriers may optionally comprise one or more carbohydrates. Carbohydrates may be natural or synthetic. A carbohydrate may be a derivatized natural carbohydrate. In certain embodiments, 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. In certain embodiments, a carbohydrate is a polysaccharide, including but not limited to pullulan, cellulose, microcrystalline cellulose, hydroxypropyl methylcellulose (HPMC), hydroxycellulose (HC), methylcellulose (MC), dextran, cyclodextran, glycogen, starch, hydroxyethylstarch, carageenan, glycon, amylose, chitosan, N,0-carboxylmethylchitosan, algin and alginic acid, starch, chitin, heparin, inulin, konjac, glucommannan, pustulan, heparin, hyaluronic acid, curdlan, and xanthan. In embodiments, the inventive synthetic nanocarriers do not comprise (or specifically exclude) carbohydrates, such as a
polysaccharide. In certain embodiments, the carbohydrate may comprise a carbohydrate derivative such as a sugar alcohol, including but not limited to mannitol, sorbitol, xylitol, erythritol, maltitol, and lactitol.
Compositions according to the invention comprise inventive synthetic nanocarriers in combination with 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. In an embodiment, inventive synthetic nanocarriers are suspended in sterile saline solution for injection together with a preservative. In embodiments, when preparing synthetic nanocarriers as carriers for agents (e.g., antigen or adjuvant) for use in vaccines methods for coupling the agents to the synthetic nanocarriers may be useful. If the agent is a small molecule it may be of advantage to attach the agent 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 agent to the synthetic nanocarrier through the use of these surface groups rather than attaching the agent to a polymer and then using this polymer conjugate in the construction of synthetic nanocarriers. A variety of reactions can be used for the purpose of attaching agents to synthetic nanocarriers.
In certain embodiments, the coupling can be a covalent linker. In embodiments, peptides 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 antigen or adjuvant containing an alkyne group or by the 1,3- dipolar cycloaddition reaction of alkynes on the surface of the nanocarrier with antigens or adjuvants 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.
Additionally, 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.
An amide linker is formed via an amide bond between an amine on one component such as the antigen or adjuvant 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 or antigens or adjuvants 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 Ri-S-S-R2. A disulfide bond can be formed by thiol exchange of an antigen or adjuvant containing thiol/mercaptan group(-SH) with another activated thiol group on a polymer or nanocarrier or a nanocarrier containing thiol/mercaptan groups with a antigen or adjuvant containing activated thiol group. A triazole linker, specifically a 1,2,3-triazole of the form
Figure imgf000036_0001
, wherein Ri 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 such as the peptide. 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.
In embodiments, 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. Alternatively, the synthetic nanocarrier can be prepared by another route, and subsequently functionalized with alkyne or azide groups. The antigen or adjuvant 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 antigen or adjuvant is then allowed to react with the nanocarrier via the 1,3-dipolar cycloaddition reaction with or without a catalyst which covalently couples the antigen or adjuvant 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 R S-R2. Thioether can be made by either alkylation of a
thiol/mercaptan (-SH) group on one component such as the antigen or adjuvant 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 such as a antigen or adjuvant 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. In another way, thioether linkers can be prepared by the radical thiol-ene reaction of a thiol/mercaptan group on one component such as a antigen or adjuvant 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 such as the antigen or adjuvant 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 such as the antigen or adjuvant 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 such as the antigen or adjuvant with 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 such as the antigen or adjuvant 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 such as the antigen or adjuvant 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 such as the antigen or adjuvant with an alkylating group such as halide, epoxide, or sulfonate ester group on the second component such as the nanocarrier. Alternatively, an amine linker can also be made by reductive amination of an amine group on one component such as the antigen or adjuvant 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 such as the antigen or adjuvant with a sulfonyl halide (such as sulfonyl chloride) group on the second component such as the nanocarrier.
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 nanoparticle or attached to the antigen or adjuvant.
The antigen or adjuvant can also be conjugated to the nanocarrier via non-covalent conjugation methods. For examples, a negative charged antigen or adjuvant can be conjugated to a positive charged nanocarrier through electrostatic adsorption. An antigen or adjuvant containing a metal ligand can also be conjugated to a nanocarner containing a metal complex via a metal-ligand complex.
In embodiments, the antigen or adjuvant 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 activatible groups on its surface. In the latter case, the antigen or adjuvant is prepared with a group which is compatible with the attachment chemistry that is presented by the synthetic nanocarriers' surface. In other embodiments, agents, such as a peptide antigen, 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. In an embodiment, the linker can be a homobifuntional or heterobifunctional reagent as described in Hermanson 2008. For example, 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. The resulting ADH linked synthetic nanocarrier is then conjugated with an agent containing an acid group via the other end of the ADH linker on the NC to produce the corresponding VLP or liposome peptide conjugate.
For detailed descriptions of available conjugation methods, see Hermanson G T "Bioconjugate Techniques", 2nd Edition Published by Academic Press, Inc., 2008. In addition to covalent attachment the antigen or adjuvant can be coupled by adsorbtion to a preformed synthetic nanocarrier or it can be coupled by encapsulation during the formation of the synthetic nanocarrier.
METHODS OF MAKING AND USING THE COMPOSITIONS AND RELATED METHODS
Synthetic nanocarriers may be prepared using a wide variety of methods known in the art. For example, synthetic nanocarriers can be formed by methods as nanoprecipitation, flow focusing using 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. Alternatively or additionally, 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. Sci., 30:545; and Trindade et al., 2001, Chem. Mat., 13:3843). Additional methods have been described in the literature (see, e.g., Doubrow, Ed., "Microcapsules and Nanoparticles in Medicine and Pharmacy," CRC Press, Boca Raton, 1992; Mathiowitz et al., 1987, J. Control. Release, 5:13; Mathiowitz et al., 1987, Reactive Polymers, 6:275; and Mathiowitz et al., 1988, J. Appl. Polymer Sci., 35:755, US Patents 5578325 and 6007845; P. Paolicelli et al., "Surface-modified PLGA-based
Nanoparticles that can Efficiently Associate and Deliver Virus-like Particles" Nanomedicine. 5(6):843-853 (2010)).
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.
Paolicelli et al., "Surface-modified PLGA-based Nanoparticles that can Efficiently Associate and Deliver Virus-like Particles" Nanomedicine. 5(6):843-853 (2010). Other methods suitable for encapsulating materials, such as oligonucleotides, into synthetic nanocarriers may be used, including without limitation methods disclosed in United States Patent 6,632,671 to Unger October 14, 2003.
In certain embodiments, 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.
If 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 of the inventive synthetic nanocarriers (such as targeting moieties, polymeric matrices, antigens, adjuvants, and the like), may be coupled to the 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 U.S. Patent Application 2006/0002852 to Saltzman et al., Published U.S. Patent Application 2009/0028910 to DeSimone et al., or Published International Patent Application WO/2008/127532 Al to Murthy et al.
Alternatively or additionally, synthetic nanocarriers can be coupled to an element, such as targeting moieties, adjuvants, various antigens, etc., directly or indirectly via non- covalent interactions. In non-covalent embodiments, 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 inventive synthetic nanocarrier. In embodiments, encapsulation and/or absorption is a form of coupling.
In embodiments, the inventive synthetic nanocarriers can be combined with other adjuvants by admixing in the same vehicle or delivery system. Such adjuvants may 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, ISCOMATRIX™, emulsions such as MF59™, Montanide® ISA 51 and ISA 720, AS02 (QS21+squalene+ MPL®), AS15, liposomes and liposomal formulations such as AS01, synthesized or specifically prepared microparticles and microcarriers such as bacteria-derived outer membrane vesicles (OMV) of N. 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. Additional useful adjuvants may be found in WO 2002/032450; US 7,357,936 "Adjuvant Systems and
Vaccines"; US 7,147,862 "Vaccine composition containing adjuvants"; US 6,544,518 "Vaccines"; US 5,750,110 "Vaccine composition containing adjuvants." The doses of such other adjuvants can be determined using conventional dose ranging studies. In embodiments, adjuvant that is not coupled to the recited synthetic nanocarriers may be the same or different from adjuvant that is coupled to the synthetic nanocarriers, if any. In other embodiments, the doses of such adjuvants may also be the same or different. In embodiments, any adjuvant coupled to the inventive synthetic nanocarriers can be different, similar or identical to those not coupled to any nanocarriers. The adjuvants (coupled and not coupled) can be administered separately at a different time-point and/or at a different body location and/or by a different immunization route. Additionally, the separate adjuvant and population of nanocarriers can be administered separately at a different time- point and/or at a different body location and/or by a different immunization route.
Populations of 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 subset 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.
Typical inventive 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., thimerosal, 2-phenoxyethanol, EDTA), polymeric stabilizers and viscosity-adjustment agents (e.g., polyvinylpyrrolidone, poloxamer 488,
carboxymethylcellulose) and co-solvents (e.g., glycerol, polyethylene glycol, ethanol).
Compositions according to the invention comprise inventive 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. In an embodiment, inventive synthetic nanocarriers are suspended in sterile saline solution for injection together with a preservative.
It is to be understood that the compositions of the invention can be made in any suitable manner, and the invention is in no way limited to 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.
In some embodiments, inventive synthetic nanocarriers are manufactured under sterile conditions or are terminally sterilized. This can ensure that resulting composition 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. In some embodiments, inventive synthetic nanocarriers may be lyophilized and stored in suspension or as lyophilized powder depending on the formulation strategy for extended periods without losing activity.
The inventive compositions may be administered by a variety of routes of
administration, including but not limited to subcutaneous, intramuscular, intradermal, oral, intranasal, transmucosal, sublingual, rectal, ophthalmic, transdermal, transcutaneous or by a combination of these routes.
Doses of dosage forms contain varying amounts of populations of synthetic nanocarriers and/or varying amounts of adjuvants and/or antigens, according to the invention. The amount of synthetic nanocarriers and/or adjuvants and/or antigens present in the inventive dosage forms can be varied according to the nature of the adjuvants and/or antigens, the therapeutic benefit to be accomplished, and other such parameters. In some embodiments, the doses of the dosage forms are systemic doses. In embodiments, dose ranging studies can be conducted to establish optimal therapeutic amount of the population of synthetic nanocarriers and/or the amount of adjuvants and/or antigens to be present in the dosage form. In embodiments, the synthetic nanocarriers and/or the adjuvants and/or antigens are present in the dosage form in an amount effective to generate an immune response upon administration to a subject. It may be possible to determine amounts of the adjuvants and/or antigens effective to generate an immune response using conventional dose ranging studies and techniques in subjects. Inventive 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.
The compositions and methods described herein can be used to induce, enhance, stimulate, 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, autoimmune diseases, inflammatory diseases, immunological 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 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.
In embodiments, the compositions provided can be used to induce a rapid and strong systemic induction of pro-inflammatory cytokines, such as TNF-oc, IL-6 and/or IL-12. The compositions provided, therefore, can be administered to subjects in need of an inflammatory response, preferably a systemic inflammatory response. In other embodiments, the compositions provided can be used for the rapid and strong systemic induction of cytokines that are important for a Thl immune response, such as IFN-γ, IL-12 and/or IL-18. The compositions provided, therefore, can be administered to subjects in need of a Thl response, preferably a systemic Thl response. In still other embodiments, the compositions provided can be used to induce a strong humoral response. The compositions provided, therefore, can be administered to subjects in need of a humoral response. In still further embodiments, the compositions provided can be used to induce a strong specific local CTL response. The compositions provided, therefore, can be administered to subjects in need of a specific local CTL response. Such a response can be specific to any of the antigens provided herein, preferably to one or more antigens in an inventive composition or that is administered according to an inventive method provided herein.
The subjects provided herein can have or be at risk of having cancer. 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 epithelial cells, stromal cells, germ cells and mesenchymal cells;
pancreatic cancer; prostate cancer; rectal cancer; sarcomas including leiomyosarcoma, rhabdomyosarcoma, liposarcoma, fibrosarcoma, and osteosarcoma; skin cancer including melanoma, Merkel cell carcinoma, Kaposi's sarcoma, basal cell carcinoma, and squamous cell cancer; testicular cancer including germinal tumors such as seminoma, non-seminoma (teratomas, choriocarcinomas), stromal tumors, and germ cell tumors; thyroid cancer including thyroid adenocarcinoma and medullar carcinoma; and renal cancer including adenocarcinoma and Wilms tumor.
The subjects provided herein can have or be at risk of having an infection or infectious disease. Infections or infectious diseases 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, Diphtheria, Epidemic Typhus, Gonorrhea, Impetigo, Legionellosis, Leprosy (Hansen's
Disease), Leptospirosis, Listeriosis, Lyme disease, Melioidosis, Rheumatic Fever, MRSA infection, Nocardiosis, Pertussis (Whooping Cough), Plague, Pneumococcal pneumonia, Psittacosis, Q fever, Rocky Mountain Spotted Fever (RMSF), Salmonellosis, Scarlet Fever, Shigellosis, Syphilis, Tetanus, Trachoma, Tuberculosis, Tularemia, Typhoid Fever, Typhus and Urinary Tract Infections; parasitic infectious diseases, such as African trypanosomiasis, Amebiasis, Ascariasis, Babesiosis, Chagas Disease, Clonorchiasis, Cryptosporidiosis, Cysticercosis, Diphyllobothriasis, Dracunculiasis, Echinococcosis, Enterobiasis, Fascioliasis, Fasciolopsiasis, Filariasis, Free-living amebic infection, Giardiasis, Gnathostomiasis, Hymenolepiasis, Isosporiasis, Kala-azar, Leishmaniasis, Malaria, Metagonimiasis, Myiasis, Onchocerciasis, Pediculosis, Pinworm Infection, Scabies, Schistosomiasis, Taeniasis, Toxocariasis, Toxoplasmosis, Trichinellosis, Trichinosis, Trichuriasis, Trichomoniasis and Trypanosomiasis; fungal infectious disease, such as Aspergillosis, Blastomycosis,
Candidiasis, Coccidioidomycosis, Cryptococcosis, Histoplasmosis, Tinea pedis (Athlete's Foot) and Tinea cruris; prion infectious diseases, such as Alpers' disease, Fatal Familial Insomnia, Gerstmann-Straussler-Scheinker syndrome, Kuru and Variant Creutzfeldt- Jakob disease.
Subject provided here also include those that have or are at risk of having an atopic condition, such as but not limited to allergy, allergic asthma, or atopic dermatitis; asthma; chronic obstructive pulmonary disease (COPD, e.g. emphysema or chronic bronchitis); and chronic infections due to chronic infectious agents such as chronic leishmaniasis, candidiasis or schistosomiasis and infections caused by plasmodia, Toxoplasma gondii, mycobacteria, HIV, HBV, HCV EBV or CMV, or any one of the above, or any subset of the above. Other indications treatable using the inventive compositions include but are not limited to indications in which a subject's Thl response is suboptimal and/or ineffective. Use of the present invention can enhance a subject's Thl immune response with an adjuvant that can stimulate a Thl immune response. The subjects, therefore, also include those that have or are at risk of having cancer, subjects with compromised or suboptimal immunity, such as infants, the elderly, cancer patients, individuals receiving immunosuppressive drugs or irradiation, hemodialysis patients and those with genetic or idiopathic immune dysfunction.
EXAMPLES Example I: Administration of Nanocarrier and Admixed R848 Adjuvant Results in Strong Systemic Production of Inflammatory Cytokines
Materials for NC-R848-1 Nanocarrier Formulations
Ovalbumin peptide 323-339 amide acetate salt, was purchased from Bachem
Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4065609.) PLGA-R848 conjugate of 75/25 lactide/glycolide monomer composition and of approximately 4100 Da molecular weight having 5.2% w/w R848 content was synthesized. PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized Polyvinyl alcohol (Mw = 11,000 - 31,000, 87- 89% hydrolyzed) was purchased from J.T. Baker (Part Number U232-08).
Methods for NC-R848-1 Nanocarrier Production
Solutions were prepared as follows:
Solution 1: Ovalbumin peptide 323 - 339 @ 70mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
Solution 2: PLGA-R848 @ 75 mg/mL and PLA-PEG-Nicotine @ 25 mg/mL in dichloromethane was prepared by dissolving PLGA-R848 at 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at 100 mg/mL in dichloromethane, then combining 3 parts of the PLGA-R848 solution to 1 part of the PLA-PEG-Nicotine solution.
Solution 3: Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
Solution 4: 70 mM phosphate buffer, pH 8.
A primary (Wl/O) emulsion was first created using Solution 1 & Solution 2. Solution
1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250. A secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250. The secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 13800 rcf for 60 minutes, removing the supernatant, and re- suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use. Table 1 : Characterization of the Nanocarriers Produced According to the Above
Nanocarrier ID Effective Diameter TLR Agonist, % w/w T-cell helper peptide, % w/w
(nm) NC-R848-1 220 R848, 1.3 Ova 323-339, 2.0
Materials for NC-R848-2 Formulations
Ovalbumin peptide 323-339 amide acetate salt was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4065609.) PLGA-R848 conjugate of 75/25 lactide/glycolide monomer composition and of approximately 4100 Da molecular weight having 5.2% w/w R848 content was synthesized. PLA-PEG-Nicotine with a nicotine- terminated PEG block of approximately 5,000 Da and DL-PLA block of approximately 17,000 Da was synthesized. Polyvinyl alcohol (Mw = 11,000 - 31,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232-08).
Methods for NC-R848-2 Nanocarrier Production
Solutions were prepared as follows:
Solution 1: Ovalbumin peptide 323 - 339 @ 70mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
Solution 2: PLGA-R848 @ 75 mg/mL and PLA-PEG-Nicotine @ 25 mg/mL in dichloromethane was prepared by dissolving PLGA-R848 at 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at 100 mg/mL in dichloromethane, then combining 3 parts of the PLGA-R848 solution to 1 part of the PLA-PEG-Nicotine solution.
Solution 3: Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
Solution 4: 70 mM phosphate buffer, pH 8.
A primary (Wl/O) emulsion was first created using Solution 1 & Solution 2. Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250. A secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250. The secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 13800 rcf for 60 minutes, removing the supernatant, and re- suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use.
Table 2: Characterization of the Nanocarriers Produced According to the Above
Figure imgf000048_0001
Materials for NC Only Formulations Ovalbumin peptide 323-339 amide acetate salt, was purchased from Bachem
Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4065609.) PLGA with 73% lactide and 27% glycolide content and an inherent viscosity of 0.12 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, AL 35211. Product Code 7525 DLG 1A.) PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized. Polyvinyl alcohol (Mw = 11,000 - 31,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232-08). Methods for NC Only Production
Solutions were prepared as follows:
Solution 1: Ovalbumin peptide 323 - 339 @ 70mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
Solution 2: PLGA @ 75 mg/mL and PLA-PEG-Nicotine @ 25 mg/mL in
dichloromethane was prepared by dissolving PLGA at 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at 100 mg/mL in dichloromethane, then combining 3 parts of the PLGA solution to 1 part of the PLA-PEG-Nicotine solution.
Solution 3: Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
Solution 4: 70 mM phosphate buffer, pH 8.
A primary (Wl/O) emulsion was first created using Solution 1 & Solution 2. Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250. A secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250. The secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution
(30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended
nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 13800 rcf for
60 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use. Table 3: Characterization of the Nanocarriers Produced According to the Above
Figure imgf000050_0001
Results
Groups of mice were injected subcutaneously into hind limbs with 100 μg of nanocarriers (NC) coupled, non-coupled or admixed with small molecule nucleoside analogue and known TLR7/8 agonist and adjuvant, R848. R848 amounts in nanocarrier were 2-3% resulting in
2-3 μg of coupled R848 per injection; amount of free R848 used was 20 μg per injection. Mouse serum was taken by terminal bleed and systemic cytokine production in serum was measured at different time-points by ELISA (BD Biosciences). As seen in Figs. 1A-1C, strong systemic production of major pro-inflammatory cytokines TNF-a, IL-6 and IL-12 was observed when admixed R848 (NC + R848) was used, while no expression of TNF-a, IL-6 and IL-12 was detected when two separate preparations of NC coupled with R848 (NC- R848-1 and NC-R848-2) were used. The difference in peak cytokine expression levels was > 100-fold for TNF-a and IL-6, and > 50-fold for IL-12. NC not coupled to R848 (labeled as NC only) did not induce any systemic cytokines when used without admixed R848.
Example 2: Coupling of Nanocarrier to R848 Adjuvant does not Inhibit Systemic Production of Immune Cytokine IFN-γ Materials for NC-R848 Nanocarrier Formulations
Ovalbumin peptide 323-339 amide acetate salt was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4065609.) PLGA-R848 conjugate of 75/25 lactide/glycolide monomer composition and of approximately 4100 Da molecular weight having 5.2% w/w R848 content was synthesized. PLA-PEG-Nicotine with a nicotine- terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized. Polyvinyl alcohol (Mw = 11,000 - 31,000, 87-89% hydrolyzed) purchased from J.T. Baker (Part Number U232-08).
Methods for NC-R848 Nanocarrier Production
Solutions were prepared as follows:
Solution 1: Ovalbumin peptide 323 - 339 @ 70mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
Solution 2: PLGA-R848 @ 75 mg/mL and PLA-PEG-Nicotine @ 25 mg/mL in dichloromethane was prepared by dissolving PLGA-R848 at 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at 100 mg/mL in dichloromethane, then combining 3 parts of the PLGA-R848 solution to 1 part of the PLA-PEG-Nicotine solution.
Solution 3: Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
Solution 4: 70 mM phosphate buffer, pH 8.
A primary (Wl/O) emulsion was first created using Solution 1 & Solution 2. Solution
1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250. A secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250. The secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution
(30 mL) and stirred at room temperature for 2 hours to allow for the dichloromethane to evaporate and for the nanocarriers to form in suspension. A portion of the suspended nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 13800 rcf for 60 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use.
Table 4: Characterization of the Nanocarriers Produced According to the Above
Nanocarrier ID Effective Diameter TLR Agonist, % w/w T-cell helper peptide, % w/w (nm)
NC-R848 213 R848, 2.6 Ova 323-339, 0.9
Materials for NC Only Nanocarrier Formulations
Ovalbumin peptide 323-339 amide acetate salt was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4065609.) PLGA with 73% lactide and 27% glycolide content and an inherent viscosity of 0.12 dL/g was purchased from
SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, AL 35211. Product Code 7525 DLG 1A.) PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized. Polyvinyl alcohol (Mw = 11,000 - 31,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232-08).
Methods for NC Only Nanocarrier Production
Solutions were prepared as follows:
Solution 1: Ovalbumin peptide 323 - 339 @ 70mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
Solution 2: PLGA @ 75 mg/mL and PLA-PEG-Nicotine @ 25 mg/mL in
dichloromethane was prepared by dissolving PLGA at 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at 100 mg/mL in dichloromethane, then combining 3 parts of the PLGA solution to 1 part of the PLA-PEG-Nicotine solution.
Solution 3: Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
Solution 4: 70 mM phosphate buffer, pH 8.
A primary (Wl/O) emulsion was first created using Solution 1 & Solution 2. Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250. A secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250. The secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution
(30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 13800 rcf for 60 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use.
Table 5: Characterization of the Nanocarriers Produced According to the Above
Figure imgf000053_0001
Results
While early proinflammatory cytokines are mostly associated with side effects during immunization, the production of other cytokines, such as immune IFN-γ is known to be involved in the induction of effective immune response. Therefore, systemic production of immune cytokine IFN-γ after injection with NC was measured 0-24 hours after inoculation. Briefly, groups of mice were injected subcutaneously into hind limbs with 100 μg of NCs coupled or admixed with small molecule nucleoside analogue and known TLR7/8 agonist and adjuvant, R848. R848 amounts in nanocarrier were 2% resulting in 2 μg of coupled R848 per injection; amount of free R848 used was 20 μg per injection. Mouse serum was taken by terminal bleed and systemic cytokine production in serum was measured at different time- points by ELISA (BD Biosciences). IFN-γ, which is important for Thl immune response, was seen with both NC-R848 (containing 2 μg of R848) and NC with admixed R848 (20 μg) (Fig. 2). Furthermore, higher levels of IFN- γ by NC with admixed R848 occurred earlier.
Example 3: Addition of Free Adjuvant Augments Immune Response
Materials for NC-Nic w/o R848 Nanocarrier Formulations
Ovalbumin peptide 323-339 amide TFA salt was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4064565.) PLA with an inherent viscosity of 0.19 dL/g was purchased from Boehringer Ingelheim (Ingelheim Germany.
Product Code R202H). PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized. Polyvinyl alcohol (Mw = 11,000 - 31,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232-08). Methods for NC-Nic w/o R848 Nanocarrier Production
Solutions were prepared as follows:
Solution 1: Ovalbumin peptide 323 - 339 @ 69mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
Solution 2: PLA @ 75 mg/mL and PLA-PEG-Nicotine @ 25mg/mL in
dichloromethane was prepared by dissolving PLA @ 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at lOOmg/mL in dichloromethane, then combining 3 parts of the PLA solution to 1 part of the PLA-PEG-Nicotine solution.
Solution 3: Polyvinyl alcohol @ 50 mg/mL in 100 mM in deionized water.
Solution 4: 70 mM phosphate buffer, pH 8.
A primary (Wl/O) emulsion was first created using Solution 1 & Solution 2. Solution
1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250. A secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 35% amplitude for 40 seconds using the Branson Digital Sonifier 250. The secondary emulsion was added to a beaker containing 70 mM phosphate buffer solution (30 mL) in an open 50ml beaker and stirred at room temperature for 2 hours to allow for the dichloromethane to evaporate and for the nanocarriers to form in suspension. A portion of the suspended nanocarriers were washed by transferring the nanocarrier suspension to centrifuge tubes, spinning at 5300 rcf for 60 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use. Table 6: Characterization of the Nanocarriers Produced According to the Above
Nanocarrier Effective Diameter TLR Agonist, % w/w T-cell helper peptide, % w/w
(nm) NC-Nic w/o 248 None Ova 323-339, 2.2
R848
Materials for NC-Nic w/ Entrapped R848 Nanocarrier Formulations
Ovalbumin peptide 323-339 amide TFA salt was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4064565.) R848 (Resiquimod) of approximately 98-99% purity was synthesized and purified. PLA with an inherent viscosity of 0.19 dL/g was purchased from Boehringer Ingelheim (Ingelheim Germany. Product Code R202H). PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized. Polyvinyl alcohol (Mw = 11,000 - 31,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232- 08).
Methods for NC-Nic w/ Entrapped R848 Nanocarrier Production
Solutions were prepared as follows:
Solution 1: Ovalbumin peptide 323 - 339 @ 69mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
Solution 2: PLA @ 75 mg/mL, R848 @ 7.5 mg/mL, and PLA-PEG-Nicotine @ 25mg/mL in dichloromethane was prepared by dissolving PLA @ 100 mg/mL in
dicholoromethane and adding R848 at lOmg/mL, also dissolving PLA-PEG-Nicotine at lOOmg/mL in dichloromethane, then combining 3 parts of the PLA/R848 solution to 1 part of the PLA-PEG-Nicotine.
Solution 3: Polyvinyl alcohol @ 50 mg/mL in 100 mM in deionized water.
Solution 4: 70 mM phosphate buffer, pH 8.
A primary (Wl/O) emulsion was first created using Solution 1 & Solution 2. Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250. A secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 35% amplitude for 40 seconds using the Branson Digital Sonifier 250. The secondary emulsion was added to a beaker containing 70 mM phosphate buffer solution (30 mL) in an open 50ml beaker and stirred at room temperature for 2 hours to allow for the dichloromethane to evaporate and for the nanocarriers to form in suspension. A portion of the suspended nanocarriers were washed by transferring the nanocarrier suspension to centrifuge tubes, spinning at 5300 rcf for 60 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use.
Table 7: Characterization of the Nanocarriers Produced According to the Above
Figure imgf000056_0001
Results
Mice were immunized with NC-Nic (nanocarrier exhibiting nicotine on the outer surface) carrying entrapped (non-conjugated) R848 with or without second adjuvant. Groups of five mice were immunized three times (subcutaneously, hind limbs) at 2-week intervals (days 0, 14 and 28) with 100 μg of NC-Nic. Serum anti-nicotine antibodies were then measured on days 26 and 40. EC50 for anti-nicotine antibodies were measured by standard ELISA against polylysine-nicotine (Fig. 3) (Group 1: NC-Nic w/o entrapped R848; group 2: NC-Nic w. 1.5% of entrapped R848; group 3: NC-Nic w. 1.5% of entrapped R848 + 80 μg of alum; group 4: NC-Nic w. 1.5% of entrapped R84 + 25 μg of CpG-1826). This demonstrates that utilization of entrapped R848 (Thl adjuvant, TLR7/8 agonist) within the nanocarriers (NC) generates an immune response, which is superior to one induced by NC without R848 (group 2 > group 1). Moreover, addition of free Th2 adjuvant (alum) to NC-Nic with R848 further augments humoral immune response (group 3 > group 2). At the same time, addition of another free Thl adjuvant (CpG, TLR9 agonist; group 4) also augments immune response, but is less potent in this combination than alum (group 4 > group 2 versus 4 < group 3). Example 4: Addition of Free Adjuvant Augments Immune Response to NC without Adjuvant
Materials for NC-Nic Nanocarrier Formulations Ovalbumin peptide 323-339 amide TFA salt, was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4064565.) PLA with an inherent viscosity of 0.19 dL/g was purchased from Boehringer Ingelheim (Ingelheim Germany.
Product Code R202H). PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized. Polyvinyl alcohol (Mw = 11,000 - 31,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232-08).
Methods for NC-Nic Nanocarrier Production
Solutions were prepared as follows:
Solution 1: Ovalbumin peptide 323 - 339 @ 69mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
Solution 2: PLA @ 75 mg/mL and PLA-PEG-Nicotine @ 25mg/mL in
dichloromethane was prepared by dissolving PLA @ 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at lOOmg/mL in dichloromethane, then combining 3 parts of the PLA solution to 1 part of the PLA-PEG-Nicotine solution.
Solution 3: Polyvinyl alcohol @ 50 mg/mL in 100 mM in deionized water.
Solution 4: 70 mM phosphate buffer, pH 8.
A primary (Wl/O) emulsion was first created using Solution 1 & Solution 2. Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250. A secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 35% amplitude for 40 seconds using the Branson Digital Sonifier 250. The secondary emulsion was added to a beaker containing 70 mM phosphate buffer solution (30 mL) in an open 50ml beaker and stirred at room temperature for 2 hours to allow for the dichloromethane to evaporate and for the nanocarriers to form in suspension. A portion of the suspended nanocarriers were washed by transferring the nanocarrier suspension to centrifuge tubes, spinning at 5300 rcf for 60 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use. Table 8: Characterization of the Nanocarriers Produced According to the Above
Figure imgf000058_0001
Materials for NC-Nic-R848 Nanocarrier Formulations
Ovalbumin peptide 323-339 amide TFA salt, was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4064565.) R848 (Resiquimod) of approximately 98-99% purity was synthesized and purified. PLA with an inherent viscosity of 0.19 dL/g was purchased from Boehringer Ingelheim (Ingelheim Germany. Product Code R202H). PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized. Polyvinyl alcohol (Mw = 11 ,000 - 31 ,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232- 08).
Methods for NC-Nic-R848 Nanocarrier Production
Solutions were prepared as follows:
Solution 1: Ovalbumin peptide 323 - 339 @ 69mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
Solution 2: PLA @ 75 mg/mL, R848 @ 7.5 mg/mL, and PLA-PEG-Nicotine @ 25mg/mL in dichloromethane was prepared by dissolving PLA @ 100 mg/mL in
dicholoromethane and adding R848 at lOmg/mL, also dissolving PLA-PEG-Nicotine at lOOmg/mL in dichloromethane, then combining 3 parts of the PLA/R848 solution to 1 part of the PLA-PEG-Nicotine.
Solution 3: Polyvinyl alcohol @ 50 mg/mL in 100 mM in deionized water.
Solution 4: 70 mM phosphate buffer, pH 8
A primary (Wl/O) emulsion was first created using Solution 1 & Solution 2. Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250. A secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 35% amplitude for 40 seconds using the Branson Digital Sonifier 250. The secondary emulsion was added to a beaker containing 70 mM phosphate buffer solution (30 mL) in an open 50ml beaker and stirred at room temperature for 2 hours to allow for the dichloromethane to evaporate and for the nanocarriers to form in suspension.
A portion of the suspended nanocarriers were washed by transferring the nanocarrier suspension to centrifuge tubes, spinning at 5300 rcf for 60 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use.
Table 9: Characterization of the Nanocarriers Produced According to the Above
Figure imgf000059_0001
Results
Mice were immunized with NC-Nic (nanocarrier exhibiting nicotine on the outer surface) that did not have adjuvant in the NC with or without admixed R848. Groups of five mice were immunized three times (subcutaneously, hind limbs) at 2-week intervals (days 0, 14 and 28) with 100 μg of NC-Nic. Serum anti-nicotine antibodies were then measured on days 26 and 40. EC50 for anti-nicotine antibodies were measured by standard ELISA against polylysine-nicotine (Fig. 4) (group 1: NC-Nic w/o entrapped R848; group 2: NC-Nic w/o entrapped R848 + 20 μg of free R848). This demonstrates that admixing of free R848 (Thl adjuvant, TLR7/8 agonist) to antigen-carrying NCs generates immune response, which is superior to one induced by NC without admixed R848 (group 2 > group 1).
Similarly, the immune response to NC-carried antigen in mice immunized with NC- Nic carrying encapsulated R848 adjuvant (NC-Nic-R848) admixed with free Thl adjuvant CpG-1826 (Enzo) or Th2 adjuvant alum (Pierce) was augmented. Groups of five mice were immunized three times (s.c, hind limbs) at 2-week intervals (days 0, 14 and 28) with 100 μg of NC-Nic-R848. Serum anti-nicotine antibodies were then measured on days 26 and 40.
EC50 for anti-nicotine antibodies as measured in standard ELISA against polylysine-nicotine (Fig. 5) (group 1: NC-Nic-R848; group 2: NC-NicR848 + 80 μg of free alum; group 3: NC- NicR848 + 25 μg of free CpG-1826). This demonstrates that admixing of a free adjuvant to antigen/adjuvant-carrying NCs generates immune response, which is superior to one induced by the same NC without admixed adjuvant (group 2 > group 1; group 3> group 1). Example 5: Addition of Free Adjuvant Augments Immune Response to NC Containing Encapsulated Adjuvant Materials for Nanocarrier Formulations
Ovalbumin peptide 323-339 amide acetate salt, was purchased from Bachem
Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4065609.) R848
(Resiquimod) of approximately 98-99% purity was synthesized and purified. PLA-R848 conjugate having molecular weight of approximately 1300 Da and R848 content of approximately 9% by weight was synthesized at by a ring-opening process. PLA-PEG- Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized. Polyvinyl alcohol (Mw = 11,000 - 31,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232-08). Methods for Nanocarrier Production
Solutions were prepared as follows:
Solution 1: Ovalbumin peptide 323 - 339 @ 70 mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
Solution 2: PLA-R848 @ 75 mg/mL, PLA-PEG-Nicotine @ 25 mg/mL, and R848 @ 1.9 mg/mL in dichloromethane was prepared by dissolving the polymers at 100 mg/mL, adding the R848 to the PLA-PEG-Nicotine solution, and then combining 3 parts of the PLA- R848 solution to 1 part of the PLA-PEG-Nicotine/R848 solution.
Solution 3: Polyvinyl alcohol @ 50 mg/mL in 100 mM in deionized water.
Solution 4: 70 mM phosphate buffer, pH 8.
A primary (Wl/O) emulsion was first created using Solution 1 & Solution 2. Solution
1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250. A secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 35% amplitude for 40 seconds using the Branson Digital Sonifier 250. The secondary emulsion was added to a beaker containing 70 mM phosphate buffer solution (30 mL) in an open 50ml beaker and stirred at room temperature for 2 hours to allow for the dichloromethane to evaporate and for the nanocarriers to form in suspension. A portion of the suspended nanocarriers were washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 13,800 rcf for 60 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use.
Table 10: Characterization of the Nanocarriers Produced According to the Above
Figure imgf000061_0001
Results
Mice were immunized with the nanocarriers, NC-Nic (nanocarrier exhibiting nicotine on the outer surface) which carried R848 and OP-II helper peptide, with or without admixed alum. Groups of five mice were immunized three times (subcutaneously, hind limbs) at 2- week intervals (days 0, 14 and 28) with 100 μg of NC[Nic,R848,OP-II] +/- 80 μg of admixed alum (Pierce). Serum anti-nicotine antibodies were then measured on days 40 and 70. EC50 for anti-nicotine antibodies were measured by standard ELISA against polylysine-nicotine (Fig. 6) (Group 1: NC[Nic,R848,OP-II]; group 2: NC[Nic,R848,OP-II] + 80 μg of admixed alum). This demonstrates that admixing of free alum (Th2 adjuvant) to antigen-carrying adjuvant-containing NCs generates immune response, which is superior to one induced by the same NC without admixed alum (group 2 > group 1).
Example 6: NC-encapsulated Antigen Generates a Stronger Cellular Immune Response than Free Antigen (Free Adjuvant Admixed)
Materials for Nanocarrier Formulations
Ovalbumin protein, was purchased from Worthington Biochemical Corporation (730
Vassar Avenue, Lakewood, NJ 08701. Product Code 3048.) PLA with an inherent viscosity of 0.21 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, AL 35211. Product Code 100 DL 2A.) PLA-PEG-OMe block co-polymer with a methyl ether terminated PEG block of approximately 2,000 Da and PLA block of approximately 19,000 Da was synthesized. Polyvinyl alcohol (Mw = 11,000 - 31,000, 87- 89% hydrolyzed) was purchased from J.T. Baker (Part Number U232-08).
Methods for Nanocarrier Production
Solutions were prepared as follows:
Solution 1: Ovalbumin protein @ 20mg/mL was prepared in phosphate buffered saline at room temperature.
Solution 2: PLA @ 75 mg/mL and PLA-PEG-OMe @ 25 mg/mL in dichloromethane was prepared by dissolving PLA at 100 mg/mL in dichloromethane and PLA-PEG-OMe at 100 mg/mL in dichloromethane, then combining 3 parts of the PLA solution to 1 part of the PLA-PEG-OMe solution.
Solution 3: Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
Solution 4: 70 mM phosphate buffer, pH 8.
A primary (Wl/O) emulsion was first created using Solution 1 & Solution 2. Solution 1 (0.2 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250. A secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (3.0 mL) to the primary emulsion, vortexing to create a course dispersion, and then sonicating at 30% amplitude for 60 seconds using the Branson Digital Sonifier 250. The secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 21,000 rcf for 45 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use.
Table 11 : Characterization of the Nanocarriers Produced According to the Above
Figure imgf000064_0001
Results
Mice were immunized either with the nanocarriers, NC-OVA (nanocarrier carrying encapsulated ovalbumin protein), or with free ovalbumin (OVA) with a free adjuvant admixed. Groups of 3 mice were immunized once (s.c, hind limbs) with 100 μg of NC-OVA (2.8% OVA) or with 2.5 μg of free OVA admixed with 10 μg of free 1826-CpG (TLR9 agonist). Draining popliteal lymph nodes were taken at day 4 after immunization, meshed, incubated in vitro for 4 days in complete RPMI medium supplemented with 10 units/ml of IL-2, and specific CTL (cytotoxic T cell) activity was determined (as % of lysis of ovalbumin-expressing cell line EG.7-OVA - % of lysis of its parental cell line EL-4) at different effector/target (E:T) ratios (Fig. 7). This demonstrates that utilization of NC- encapsulated antigen results in the generation of a stronger local cellular immune response than immunization with a free antigen.
Example 7: Addition of Free Adjuvant Augments Immune Response to NC without Adjuvant
Materials for Nanocarrier Formulations
Ovalbumin peptide 323-339 amide acetate salt was purchased from Bachem Americas
Inc. (3132 Kashiwa Street, Torrance CA 90505. Product code 4065609.) PLA with an inherent viscosity of 0.19 dL/g was purchased from Boehringer Ingelheim (Ingelheim
Germany. Product Code R202H). PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 5,000 Da and DL-PLA block of approximately 17,000 Da was synthesized. Polyvinyl alcohol (Mw = 11 ,000 - 31 ,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232-08). Methods for Nanocarrier Production
Solutions were prepared as follows:
Solution 1: Ovalbumin peptide 323 - 339 @ 17.5 mg/mL in dilute hydrochloric acid aqueous solution. The solution was prepared by dissolving ovalbumin peptide in 0.13N hydrochloric acid solution at room temperature.
Solution 2: 0.19-IV PLA @ 75 mg/mL and PLA-PEG-nicotine @ 25 mg/ml in dichloromethane. The solution was prepared by separately dissolving PLA @ 100 mg/mL in dichloromethane and PLA-PEG-nicotine @ 100 mg/mL in dichloromethane, then mixing the solutions by adding 3 parts PLA solution for each part of PLA-PEG-nicotine solution.
Solution 3: Polyvinyl alcohol @ 50 mg/mL in 100 mM pH 8 phosphate buffer.
A primary (Wl/O) emulsion was first created using Solution 1 & Solution 2. Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250. A secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and then sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250. The secondary emulsion was then added to an open 50 mL beaker containing 70mM pH 8 phosphate buffer solution (30 mL) and stirred at room temperature for 2 hours to evaporate dichloromethane and to form nanocarriers in aqueous suspension. A portion of the nanocarriers was washed by transferring the suspension to a centrifuge tube and spinning at 13,800g for one hour, removing the supernatant, and re- suspending the pellet in phosphate buffered saline. The washing procedure was repeated, and the pellet was re- suspended in phosphate buffered saline for a final nanocarrier dispersion of about 10 mg/mL. The amounts of oligonucleotide and peptide in the nanocarrier were determined by HPLC analysis. The total dry-nanocarrier mass per mL of suspension was determined by a gravimetric method and was adjusted to 5 mg/mL.
Table 12: Characterization of the Nanocarriers Produced According to the Above
Figure imgf000066_0001
Results
Mice were immunized with NC-Nic (nanocarrier exhibiting nicotine on the outer surface and containing OP-II helper peptide, no adjuvant in the NC) admixed with CpG in either the phosphodiester (PO) or phosphorothioate (PS) form. The PO form is degraded by nucleases and, therefore, is not stable once injected into mice. The PS form is nuclease- resistant and, therefore, stable once injected into mice. As a negative control, mice were immunized with PBS only. Groups of five mice were immunized three times
(subcutaneously, hind limbs) at 2-week intervals (days 0, 14 and 28) with 100 μg of NC-Nic + 20 μg of CpG (PS or PO) or PBS. Serum anti-nicotine antibody titers were measured on days 26 and 40. Anti-nicotine antibody titers (EC50) were measured by ELISA against polylysine-nicotine (Fig. 8) (group 1: NC-Nic (no adjuvant) + free CpG (PS); group 2: NC- Nic (no adjuvant) + free CpG (PO); group 3: PBS only). This demonstrates that admixing of free CpG (PS) (Thl adjuvant, TLR9 agonist) to antigen-carrying NCs generates an immune response which is superior to those induced by NC with admixed CpG (PO) or with PBS (group 1 > group 2 > group 3). Example 8: Addition of Free Adjuvant Augments Immune Response to NC without Adjuvant
Materials for Nanocarrier Formulations
Ovalbumin protein was purchased from Worthington Biochemical Corporation (730 Vassar Avenue, Lakewood, NJ 08701. Product Code 3048.) PLA with an inherent viscosity of 0.21 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, AL 35211. Product Code 100 DL 2A.) PLA-PEG-OMe block co-polymer with a methyl ether terminated PEG block of approximately 2,000 Da and PLA block of approximately 19,000 Da was synthesized. Polyvinyl alcohol (Mw = 11,000 - 31,000, 87- 89% hydrolyzed) was purchased from J.T. Baker (Part Number U232-08). Methods for Nanocarrier Production
Solutions were prepared as follows:
Solution 1: Ovalbumin protein @ 20mg/mL was prepared in phosphate buffered saline at room temperature.
Solution 2: PLA @ 75 mg/mL and PLA-PEG-OMe @ 25 mg/mL in dichloromethane was prepared by dissolving PLA at 100 mg/mL in dichloromethane and PLA-PEG-OMe at 100 mg/mL in dichloromethane, then combining 3 parts of the PLA solution to 1 part of the PLA-PEG-OMe solution.
Solution 3: Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
Solution 4: 70 mM phosphate buffer, pH 8.
A primary (Wl/O) emulsion was first created using Solution 1 & Solution 2. Solution 1 (0.2 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250. A secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (3.0 mL) to the primary emulsion, vortexing to create a course dispersion, and then sonicating at 30% amplitude for 60 seconds using the Branson Digital Sonifier 250. The secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 21,000 rcf for 45 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use.
Table 13: Characterization of the Nanocarriers Produced According to the Above
Nanocarrier Effective Diameter TLR Agonist, % w/w Antigen, % w/w
(nm)
NC-OVA 228 None OVA protein, 2.8 Results
Mice were immunized with NC-OVA (nanocarrier exhibiting ovalbumin (OVA) on the outer surface, no adjuvant in the NC) admixed with either 20 μg of R848 or CpG (PS; nuclease-resistant). Control mice received 2.5 μg of soluble antigen (OVA) admixed with 20 μg of CpG (PS). Groups of five mice were immunized three times (subcutaneously, hind limbs) at 2-week intervals (days 0, 14 and 28) with 100 μg of NC-OVA + 20 μg of R848 or CpG (PS) or 2.5 μg of soluble OVA + 20 μg of CpG (PS). Serum anti-OVA antibody titers were measured on days 26 and 44. Anti-OVA antibody titers (EC50) were measured by ELISA against OVA protein (Fig. 9) (group 1: NC-OVA (no adjuvant) + free R848; group 2: NC-OVA (no adjuvant) + free CpG (PS); group 3: soluble OVA + CpG (PS)). This demonstrates that admixing of free R848 (Thl adjuvant, TLR7/8 agonist) or CpG (PS) (Thl adjuvant, TLR9 agonist) to antigen-carrying NCs generates an immune response, which is superior to those induced by soluble antigen admixed with adjuvant (CpG (PS)) (groups 1 and 2 > group 3).
Example 9: Addition of Free Adjuvant Augments Immune Response to NC with
Adjuvant
Materials for Group 1 Nanocarrier Formulations
Ovalbumin peptide 323-339 amide acetate salt was purchased from Bachem Americas
Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4065609.) PLGA-R848 conjugate of 75/25 lactide/glycolide monomer composition and of approximately 4100 Da molecular weight having 5.2% w/w R848 content was synthesized. PLA-PEG-Nicotine with a nicotine- terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized. Polyvinyl alcohol (Mw = 11,000 - 31,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232-08).
Methods for Group 1 Nanocarrier Production
Solutions were prepared as follows:
Solution 1: Ovalbumin peptide 323 - 339 @ 70mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
Solution 2: PLGA-R848 @ 75 mg/mL and PLA-PEG-Nicotine @ 25 mg/mL in dichloromethane was prepared by dissolving PLGA-R848 at 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at 100 mg/mL in dichloromethane, then combining 3 parts of the PLGA-R848 solution to 1 part of the PLA-PEG-Nicotine solution.
Solution 3: Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
Solution 4: 70 mM phosphate buffer, pH 8.
A primary (Wl/O) emulsion was first created using Solution 1 & Solution 2. Solution 1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250. A secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250. The secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution (30 mL) and stirred at room temperature for 2 hours to allow for the dichloromethane to evaporate and for the nanocarriers to form in suspension. A portion of the suspended nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 13800 rcf for 60 minutes, removing the supernatant, and re- suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use.
Table 14: Characterization of the Nanocarriers Produced According to the Above
Figure imgf000069_0001
Materials for Group 2 Nanocarrier Formulations
Ovalbumin peptide 323-339 amide acetate salt was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4065609.) PLGA with 73% lactide and 27% glycolide content and an inherent viscosity of 0.12 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, AL 35211. Product Code 7525 DLG 1A.) PLA-PEG-Nicotine with a nicotine-terminated PEG block of approximately 3,500 Da and DL-PLA block of approximately 15,000 Da was synthesized. Polyvinyl alcohol (Mw = 11,000 - 31,000, 87-89% hydrolyzed) was purchased from J.T. Baker (Part Number U232-08).
Methods for Group 2 Nanocarrier Production
Solutions were prepared as follows:
Solution 1: Ovalbumin peptide 323 - 339 @ 70mg/mL was prepared in 0.13N hydrochloric acid at room temperature.
Solution 2: PLGA @ 75 mg/mL and PLA-PEG-Nicotine @ 25 mg/mL in
dichloromethane was prepared by dissolving PLGA at 100 mg/mL in dichloromethane and PLA-PEG-Nicotine at 100 mg/mL in dichloromethane, then combining 3 parts of the PLGA solution to 1 part of the PLA-PEG-Nicotine solution.
Solution 3: Polyvinyl alcohol @ 50 mg/mL in 100 mM in lOOmM phosphate buffer, pH 8.
Solution 4: 70 mM phosphate buffer, pH 8.
A primary (Wl/O) emulsion was first created using Solution 1 & Solution 2. Solution
1 (0.1 mL) and Solution 2 (1.0 mL) were combined in a small glass pressure tube and sonicated at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250. A secondary (W1/0/W2) emulsion was then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250. The secondary emulsion was added to an open 50 mL beaker containing 70 mM phosphate buffer solution (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 13800 rcf for 60 minutes, removing the supernatant, and re- suspending the pellet in phosphate buffered saline. This washing procedure was repeated, and then the pellet was re-suspended in phosphate buffered saline to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20°C until use. Table 15: Characterization of the Nanocarriers Produced According to the Above
Nanocarrier Effective Diameter TLR Agonist, % w/w T-cell helper peptide, % w/w
(nm) Group 2 NC 176 None Ova 323-339, 1.1
Mice were injected with 20 μg of CpG twice (subcutaneously, hind limbs) at 2-week intervals (days 0 and 14). At days 35 and 49, mice were immunized with 100 μg of NC-Nic (containing 2.6% R848 and 0.9% OP-II peptide) or 100 μg of NC-Nic (containing 1.1% OP- II peptide only). Serum anti-nicotine antibody titers were measured at days 12, 26, and 40 after immunization with NC. Anti-nicotine antibody titers (EC50) were measured by ELISA against polylysine-nicotine (Fig. 10 (group 1: NC-Nic (R848 + OP-II); group 2: NC-Nic (OP- II only)). This demonstrates that mice immunized with a combination of CpG followed at a later date by NC-Nic that contain R848 generate higher antibody titers to nicotine than mice immunized with CpG followed at a later date by NC-Nic that do not contain R848 (group 1 > group 2).
Example 10: Addition of Two Free Adjuvants Augments Immune Response to NC-Nic (Prophetic)
Materials for NC-Nic Nanocarrier Formulations
Ovalbumin peptide 323-339 amide acetate salt is purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance CA 90505. Part # 4064565.) PLA with an inherent viscosity of 0.19 dL/g is purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, AL 35211 (Product Code 100 DL 2A). PLA-PEG-Nicotine with a nicotine- terminated PEG block of approximately 5,000 Da and DL-PLA block of approximately 20,000 Da is synthesized. Polyvinyl alcohol (Mw = 11,000 - 31,000, 87-89% hydrolyzed) is purchased from J.T. Baker (Part Number U232-08). Methods for NC-Nic Nanocarrier Production
Solutions are prepared as follows:
Solution 1: Ovalbumin peptide 323 - 339 @ 20mg/mL is prepared in 0.13N hydrochloric acid at room temperature.
Solution 2: PLA @ 75 mg/mL and PLA-PEG-Nicotine @ 25mg/mL in
dichloromethane is prepared by dissolving PLA @ 100 mg/mL in dichloromethane and PLA- PEG-Nicotine at lOOmg/mL in dichloromethane, then combining 3 parts of the PLA solution to 1 part of the PLA-PEG-Nicotine solution. Solution 3: Polyvinyl alcohol @ 50 mg/mL in 100 mM in deionized water.
Solution 4: 70 mM phosphate buffer, pH 8.
A primary (Wl/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
(W1/0/W2) emulsion is then formed by adding Solution 3 (2.0 mL) to the primary emulsion and sonicating at 30% amplitude for 40 seconds using the Branson Digital Sonifier 250. The secondary emulsion is added to a beaker containing 70 mM phosphate buffer solution (30 mL) in an open 50ml beaker and stirred at room temperature for 2 hours to allow for the dichloromethane to evaporate and for the nanocarriers to form in suspension. A portion of the suspended nanocarriers are washed by transferring the nanocarrier suspension to centrifuge tubes, spinning at 21,000 rcf for 45 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 nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension is stored frozen at -20°C until use.
Table 16: Characterization of the Nanocarriers Produced According to the Above
Figure imgf000072_0001
Results
Mice are immunized with NC-Nic (nanocarrier exhibiting nicotine on the outer surface) admixed with a first (R848) and second adjuvant (alum). Groups of five mice are immunized three times (subcutaneously, hind limbs) at 2-week intervals (days 0, 14 and 28) with 100 μg of NC-Nic. Serum anti-nicotine antibodies are then measured on days 26 and 40. EC50 for anti-nicotine antibodies are measured by standard ELISA against polylysine- nicotine.

Claims

What is claimed is: 1. A composition comprising:
a dosage form that comprises
(1) a population of synthetic nanocarriers,
(2) a first adjuvant that is not coupled to any synthetic nanocarriers, and
(3) a pharmaceutically acceptable excipient.
2. The composition of claim 1, wherein the composition comprises a systemic dose of the first adjuvant.
3. The composition of claim 1, wherein the composition further comprises a second adjuvant.
4. The composition of claim 3, wherein the first adjuvant and second adjuvant are different.
5. The composition of claim 3 or 4, wherein the composition comprises a systemic dose of the first adjuvant and/or second adjuvant.
6. The composition of claim 2 or 5, wherein the systemic dose results in the systemic release of TNF-cc, IL-6 and/or IL-12.
7. The composition of claim 2 or 5, wherein the systemic dose results in the systemic release of IFN-γ, IL-12 and/or IL-18.
8. The composition of any of claims 3-7, wherein the second adjuvant is coupled to the synthetic nanocarriers.
9. The composition of any of claims 3-7, wherein the second adjuvant is not coupled to any synthetic nanocarriers.
10. The composition of any of claims 3-7, wherein the second adjuvant is coupled to another population of synthetic nanocarriers.
11. The composition of any of claims 1-10, further comprising one or more antigens.
12. The composition of claim 11, wherein the one or more antigens are coupled to the synthetic nanocarriers.
13. The composition of claim 11, wherein the one or more antigens are coupled to another population of synthetic nanocarriers.
14. The composition of claim 11, wherein the one or more antigens are not coupled to any synthetic nanocarriers.
15. The composition of any of claims 11-14, wherein the one or more antigens comprise a B cell antigen and/or a T cell antigen.
16. The composition of claim 15, wherein the T cell antigen is a T helper cell antigen.
17. The composition of any of claims 11-14, wherein the one or more antigens comprise a B cell antigen and/or a T cell antigen and a T helper cell antigen.
18. The composition of any of claims 1-10, wherein the composition does not comprise an antigen.
19. The composition of any of claims 1-18, wherein the first adjuvant and/or second adjuvant comprises a mineral salt, gel-type adjuvant, a microbial adjuvant, an oil-emulsion or emulsifier-based adjuvant, a particulate adjuvant, a synthetic adjuvant, a phosphate adjuvant, a polymer, a liposome, a microcarrier, an immunostimulatory nucleic acid, alum, a saponin, an interleukin, an interferon, a cytokine, a toll-like receptor (TLR) agonist, an imidazoquinoline, a cytokine receptor agonist, a CD40 agonist, an Fc receptor agonist, a complement receptor agonist, QS21, vitamin E, squalene, tocopherol, Quil A, ISCOMs, ISCOMATRIX, Ribi Detox, CRL-1005, L-121, tetrachlorodecaoxide, alum, MF59, AS02, AS15, cholera toxin, monophosphoryl lipid A, incomplete Freund's adjuvant, complete Freund's adjuvant, muramyl dipeptide or montanide.
20. The composition of claim 19, wherein the immunostimulatory nucleic acid comprises a CpG-containing nucleic acid.
21. The composition of claim 19, wherein the imidazoquinoline comprises resiquimod or imiquimod.
22. The composition of claim 19, wherein the first and/or second adjuvant comprises alum.
23. The composition of claim 20, wherein when the first adjuvant comprises a CpG- containing nucleic acid, the second adjuvant comprises an imidazoquinoline or alum.
24. The composition of claim 23, wherein the imidazoquinoline is resiquimod.
25. The composition of claim 19, wherein when the first adjuvant comprises an imidazoquinoline, the second adjuvant comprises a CpG-containing nucleic acid or alum.
26. The composition of claim 25, wherein the imidazoquinoline is resiquimod.
27. The composition of claim 22, wherein when the first adjuvant comprises alum, the second adjuvant comprises an imidazoquinoline or a CpG-containing nucleic acid.
28. The composition of claim 27, wherein the imidazoquinoline is resiquimod.
29. The composition of claim 19, wherein the TLR agonist comprises a TLR-1, TLR-2, TLR-3, TLR-4, TLR- 5, TLR- 6, TLR-7, TLR- 8, TLR-9, TLR-10, TLR-11 agonist or a combination thereof.
30. The composition of any of claims 1-18, wherein the first adjuvant and/or second adjuvant does not comprise a TLR agonist.
31. The composition of any of claims 1-18, wherein the first adjuvant and/or second adjuvant does not comprise a TLR-3, TLR-7, TLR-8 or TLR-9 agonist.
32. The composition of any of claims 1-31, wherein the synthetic nanocarriers comprise lipid nanoparticles, polymeric nanoparticles, metallic nanoparticles, surfactant-based emulsions, dendrimers, buckyballs, nanowires, virus-like particles, peptide or protein particles, nanoparticles that comprise a combination of nanomaterials, spheroidal
nanoparticles, cuboidal nanoparticles, pyramidal nanoparticles, oblong nanoparticles, cylindrical nanoparticles or toroidal nanoparticles.
33. The composition of claim 32, wherein the second adjuvant is coupled to the synthetic nanocarriers and comprises resiquimod.
34. The composition of claim 33, wherein the one or more antigens comprise nicotine and a T-helper cell antigen, each of which are coupled to the synthetic nanocarriers.
35. The composition of claim 34, wherein the T-helper cell antigen comprises a peptide obtained or derived from ovalbumin.
36. The composition of claim 35, wherein the peptide obtained or derived from
ovalbumin comprises the sequence as set forth in SEQ ID NO: 1.
37. The composition of any of claims 15-17 and 34-36, wherein the T helper cell antigen is coupled by encapsulation.
38. The composition of any of claims 32-37, wherein the synthetic nanocarriers comprise one or more polymers.
39. The composition of claim 38, wherein the one or more polymers comprise a polyester.
40. The composition of claim 38 or 39, wherein the one or more polymers comprise or further comprise a polyester coupled to a hydrophilic polymer.
41. The composition of claim 39 or 40, wherein the polyester comprises a poly(lactic acid), poly(glycolic acid), poly(lactic-co-glycolic acid), or polycaprolactone.
42. The composition of claim 40 or 41, wherein the hydrophilic polymer comprises a polyether.
43. The composition of claim 42, wherein the polyether comprises polyethylene glycol.
44. A method comprising:
administering the composition of any of claims 1-43 to a subject.
45. The method of claim 44, wherein the subject is a human.
46. A method comprising:
administering the composition of claim 1 and a second adjuvant to a subject, wherein the second adjuvant is administered at a time different from the administration of the composition.
47. The method of claim 46, wherein the subject is a human.
48. The method of claim 46 or 47, wherein the composition and second adjuvant are coadministered.
49. The method of claim 46 or 47, wherein the composition and second adjuvant are not coadministered.
50. The method of any of claims 46-49, wherein the second adjuvant is administered prior to the composition.
51. The method of any of claims 46-50, wherein the second adjuvant is not coupled to any synthetic nanocarriers.
52. The method of any of claims 46-50, wherein the second adjuvant is coupled to another population of synthetic nanocarriers.
53. The method of any of claims 44-52, wherein the method further comprises administering one or more antigens.
54. The method of claim 53, wherein the composition further comprises one or more antigens.
55. The method of claim 54, wherein the one or more antigens are coupled to the synthetic nanocarriers.
56. The method of claim 54, wherein the one or more antigens are coupled to another population of synthetic nanocarriers.
57. The method of claim 54, wherein the one or more antigens are not coupled to any synthetic nanocarriers.
58. The method of claim 53, 56 or 57, wherein the one or more antigens are
coadministered.
59. The method of any of claims 53-58, wherein the one or more antigens comprise a B cell antigen and/or a T cell antigen.
60. The method of claim 59, wherein the T cell antigen is a T helper cell antigen.
61. The method of any of claims 53-58, wherein the one or more antigens comprise a B cell antigen and/or a T cell antigen and a T helper cell antigen.
62. The method of any of claims 59-61, wherein the B cell antigen is nicotine.
63. The method of any of claims 60-62, wherein the T helper cell antigen comprises a peptide obtained or derived from ovalbumin.
64. The method of claim 63, wherein the peptide obtained or derived from ovalbumin comprises the sequence as set forth in SEQ ID NO: 1.
65. The method of any of claims 60-64, wherein the T helper cell antigen is coupled by encapsulation.
66. The method of any of claims 44-65, wherein the synthetic nanocarriers comprise one or more polymers.
67. The method of claim 66, wherein the one or more polymers comprise a polyester.
68. The method of claim 66 or 67, wherein the one or more polymers comprise or further comprise a polyester coupled to a hydrophilic polymer.
69. The method of claim 67 or 68, wherein the polyester comprises a poly(lactic acid), poly(glycolic acid), poly(lactic-co-glycolic acid), or polycaprolactone.
70. The method of claim 68 or 69, wherein the hydrophilic polymer comprises a polyether.
71. The method of claim 70, wherein the polyether comprises polyethylene glycol.
72. The method of any of claims 44-71, wherein the subject is in need of an inflammatory response.
73. The method of any of claims 44-71, wherein the subject is in need of a Thl immune response.
74. The method of any of claims 44-71, wherein the subject is in need of a humoral immune response.
75. The method of any of claims 44-71, wherein the subject is in need of a specific local cytotoxic T lymphocyte response.
76. The method of any of claims 44-75, wherein the subject has or is at risk of having cancer.
77. The method of any of claims 44-75, wherein the subject has or is at risk of having an infection or infectious disease.
78. The method of any of claims 44-71 and 73, the subject has or is at risk of having an atopic condition, asthma, COPD or a chronic infection.
79. The composition of any one of claims 1-43 for use in therapy or prophylaxis.
80. The composition of any one of claims 1-43 for use in a method as defined in any one of claims 44-78.
81. The composition of any one of claims 1-43 for use in a method of inducing an inflammatory response in a subject.
82. The composition of any one of claims 1-43 for use in a method of inducing a Thl immune response in a subject.
83. The composition of any one of claims 1-43 for use in a method of inducing a humoral immune response in a subject.
84. The composition of any one of claims 1-43 for use in a method of inducing a specific local cytotoxic T lymphocyte response in a subject.
85. The composition of any one of claims 1-43 for use in a method of treating or preventing cancer.
86. The composition of any one of claims 1-43 for use in a method of treating or preventing infection or infectious disease.
87. The composition of any one of claims 1-43 for use in a method of treating or preventing an atopic condition, asthma, COPD or a chronic infection.
88. Use of the composition of any one of claims 1-43 for the manufacture of a medicament for use in a method as defined in any one of claims 44-78 or 81-87.
PCT/US2011/038190 2010-05-26 2011-05-26 Nanocarrier compositions with uncoupled adjuvant WO2011150240A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
BR112012029917A BR112012029917A2 (en) 2010-05-26 2011-05-26 uncoupled adjuvant nanotransport compositions
EP11787434.7A EP2575886A4 (en) 2010-05-26 2011-05-26 Nanocarrier compositions with uncoupled adjuvant
KR1020127030841A KR20130108983A (en) 2010-05-26 2011-05-26 Nanocarrier compositions with uncoupled adjuvant
EA201291156A EA030863B1 (en) 2010-05-26 2011-05-26 Synthetic nanocarrier compositions for enhancing an immune response to an antigen and uses thereof
JP2013512250A JP6371058B2 (en) 2010-05-26 2011-05-26 Nanocarrier composition having unbound adjuvant
CN201180025504.3A CN102905728B (en) 2010-05-26 2011-05-26 There is the nanometer carrier combination of the adjuvant of not coupling
AU2011258147A AU2011258147B2 (en) 2010-05-26 2011-05-26 Nanocarrier compositions with uncoupled adjuvant
CA2798739A CA2798739A1 (en) 2010-05-26 2011-05-26 Nanocarrier compositions with uncoupled adjuvant
MX2012013713A MX2012013713A (en) 2010-05-26 2011-05-26 Nanocarrier compositions with uncoupled adjuvant.
IL222722A IL222722B (en) 2010-05-26 2012-10-28 Nanocarrier compositions with uncoupled adjuvant
AU2017201082A AU2017201082A1 (en) 2010-05-26 2017-02-17 Nanocarrier compositions with uncoupled adjuvant

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US34871310P 2010-05-26 2010-05-26
US34871710P 2010-05-26 2010-05-26
US34872810P 2010-05-26 2010-05-26
US61/348,728 2010-05-26
US61/348,713 2010-05-26
US61/348,717 2010-05-26
US35863510P 2010-06-25 2010-06-25
US61/358,635 2010-06-25

Publications (1)

Publication Number Publication Date
WO2011150240A1 true WO2011150240A1 (en) 2011-12-01

Family

ID=45004392

Family Applications (4)

Application Number Title Priority Date Filing Date
PCT/US2011/038200 WO2011150249A1 (en) 2010-05-26 2011-05-26 Multivalent synthetic nanocarrier vaccines
PCT/US2011/038218 WO2011150264A2 (en) 2010-05-26 2011-05-26 Synthetic nanocarrier combination vaccines
PCT/US2011/038210 WO2011150258A1 (en) 2010-05-26 2011-05-26 Dose selection of adjuvanted synthetic nanocarriers
PCT/US2011/038190 WO2011150240A1 (en) 2010-05-26 2011-05-26 Nanocarrier compositions with uncoupled adjuvant

Family Applications Before (3)

Application Number Title Priority Date Filing Date
PCT/US2011/038200 WO2011150249A1 (en) 2010-05-26 2011-05-26 Multivalent synthetic nanocarrier vaccines
PCT/US2011/038218 WO2011150264A2 (en) 2010-05-26 2011-05-26 Synthetic nanocarrier combination vaccines
PCT/US2011/038210 WO2011150258A1 (en) 2010-05-26 2011-05-26 Dose selection of adjuvanted synthetic nanocarriers

Country Status (17)

Country Link
US (6) US20110293723A1 (en)
EP (5) EP2575876B1 (en)
JP (12) JP6324067B2 (en)
KR (5) KR20130108983A (en)
CN (10) CN102917731A (en)
AU (8) AU2011258156B2 (en)
BR (3) BR112012029917A2 (en)
CA (4) CA2798739A1 (en)
DK (1) DK2575876T3 (en)
EA (6) EA201500857A1 (en)
ES (1) ES2661978T3 (en)
IL (6) IL222680B (en)
MX (4) MX352324B (en)
NO (1) NO2575876T3 (en)
PL (1) PL2575876T3 (en)
PT (1) PT2575876T (en)
WO (4) WO2011150249A1 (en)

Cited By (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103083663A (en) * 2013-02-04 2013-05-08 江苏省农业科学院 Immunity enhancing agent, inactivated vaccine, and preparation method thereof
WO2013106852A1 (en) * 2012-01-13 2013-07-18 President And Fellows Of Harvard College Controlled delivery of tlr agonists in structural polymeric devices
WO2013151736A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics In vivo production of proteins
WO2013151666A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of biologics and proteins associated with human disease
KR20140041134A (en) * 2012-09-27 2014-04-04 고려대학교 산학협력단 Protein nanoparticle based multivalent vaccines
US8728456B2 (en) 2009-07-31 2014-05-20 President And Fellows Of Harvard College Programming of cells for tolerogenic therapies
WO2014142653A1 (en) * 2013-03-11 2014-09-18 Cristal Delivery B.V. Vaccination composition
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
US8932583B2 (en) 2005-12-13 2015-01-13 President And Fellows Of Harvard College Scaffolds for cell transplantation
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
US9012399B2 (en) 2008-05-30 2015-04-21 President And Fellows Of Harvard College Controlled release of growth factors and signaling molecules for promoting angiogenesis
WO2015110957A2 (en) 2014-01-21 2015-07-30 De Beer Joel Hybridosomes, compositions comprising the same, processes for their production and uses thereof
US9297005B2 (en) 2009-04-13 2016-03-29 President And Fellows Of Harvard College Harnessing cell dynamics to engineer materials
WO2016055812A1 (en) * 2014-10-10 2016-04-14 Isis Innovation Limited Polymer adjuvant
EP2640190A4 (en) * 2010-11-05 2016-05-11 Selecta Biosciences Inc Modified nicotinic compounds and related methods
US9370558B2 (en) 2008-02-13 2016-06-21 President And Fellows Of Harvard College Controlled delivery of TLR agonists in structural polymeric devices
JP2016520051A (en) * 2013-05-03 2016-07-11 セレクタ バイオサイエンシーズ インコーポレーテッドSelecta Biosciences,Inc. Topical combination administration of tolerogenic synthetic nanocarriers to reduce type I and type IV hypersensitivity
US9486512B2 (en) 2011-06-03 2016-11-08 President And Fellows Of Harvard College In situ antigen-generating cancer vaccine
US9603894B2 (en) 2010-11-08 2017-03-28 President And Fellows Of Harvard College Materials presenting notch signaling molecules to control cell behavior
US9610328B2 (en) 2010-03-05 2017-04-04 President And Fellows Of Harvard College Enhancement of skeletal muscle stem cell engraftment by dual delivery of VEGF and IGF-1
US9675561B2 (en) 2011-04-28 2017-06-13 President And Fellows Of Harvard College Injectable cryogel vaccine devices and methods of use thereof
US9693954B2 (en) 2010-06-25 2017-07-04 President And Fellows Of Harvard College Co-delivery of stimulatory and inhibitory factors to create temporally stable and spatially restricted zones
US9770535B2 (en) 2007-06-21 2017-09-26 President And Fellows Of Harvard College Scaffolds for cell collection or elimination
US9821045B2 (en) 2008-02-13 2017-11-21 President And Fellows Of Harvard College Controlled delivery of TLR3 agonists in structural polymeric devices
US9937249B2 (en) 2012-04-16 2018-04-10 President And Fellows Of Harvard College Mesoporous silica compositions for modulating 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
US10047072B2 (en) 2013-09-16 2018-08-14 Astrazeneca Ab Therapeutic polymeric nanoparticles and methods of making and using same
US10045947B2 (en) 2011-04-28 2018-08-14 President And Fellows Of Harvard College Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration
US10420835B2 (en) 2011-04-29 2019-09-24 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for antigen-specific deletion of T effector cells
US10647959B2 (en) 2011-04-27 2020-05-12 President And Fellows Of Harvard College Cell-friendly inverse opal hydrogels for cell encapsulation, drug and protein delivery, and functional nanoparticle encapsulation
US10682400B2 (en) 2014-04-30 2020-06-16 President And Fellows Of Harvard College Combination vaccine devices and methods of killing cancer cells
US11150242B2 (en) 2015-04-10 2021-10-19 President And Fellows Of Harvard College Immune cell trapping devices and methods for making and using the same
US11202759B2 (en) 2010-10-06 2021-12-21 President And Fellows Of Harvard College Injectable, pore-forming hydrogels for materials-based cell therapies
WO2022081764A1 (en) 2020-10-14 2022-04-21 RNAimmune, Inc. PAN-RAS mRNA CANCER VACCINES
EP4011451A1 (en) 2015-10-22 2022-06-15 ModernaTX, Inc. Metapneumovirus mrna vaccines
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
EP4074834A1 (en) 2012-11-26 2022-10-19 ModernaTX, Inc. Terminally modified rna
US11555177B2 (en) 2016-07-13 2023-01-17 President And Fellows Of Harvard College Antigen-presenting cell-mimetic scaffolds and methods for making and using the same
EP4144378A1 (en) 2011-12-16 2023-03-08 ModernaTX, Inc. Modified nucleoside, nucleotide, and nucleic acid compositions
WO2023161350A1 (en) 2022-02-24 2023-08-31 Io Biotech Aps Nucleotide delivery of cancer therapy
US11752238B2 (en) 2016-02-06 2023-09-12 President And Fellows Of Harvard College Recapitulating the hematopoietic niche to reconstitute immunity
US11786457B2 (en) 2015-01-30 2023-10-17 President And Fellows Of Harvard College Peritumoral and intratumoral materials for cancer therapy

Families Citing this family (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2066354T3 (en) 2006-09-29 2013-05-27 Ligocyte Pharmaceuticals Inc Norovirus vaccine formulations
CA2698397C (en) 2007-09-18 2018-03-27 Ligocyte Pharmaceuticals, Inc. Method of conferring a protective immune response to norovirus
BRPI0916365A2 (en) 2008-07-21 2018-05-02 Brigham & Womens Hospital Inc methods and compositions related to synthetic beta-1,6 glycosine oligosaccharides
EP2376107B1 (en) * 2008-12-09 2014-05-21 Coley Pharmaceutical Group, Inc. Immunostimulatory oligonucleotides
EA201101530A1 (en) * 2009-04-21 2012-03-30 Селекта Байосайенсиз, Инк. IMMUNONANOTHERAPY, PROVIDING TH1-DISPERSED RESPONSE
AU2010254551B2 (en) * 2009-05-27 2016-10-20 Selecta Biosciences, Inc. Immunomodulatory agent-polymeric compounds
CN107715118A (en) * 2009-08-26 2018-02-23 西莱克塔生物科技公司 The composition of inducing T cell auxiliary
NO2575876T3 (en) * 2010-05-26 2018-05-05
DK2714017T3 (en) * 2011-06-02 2018-09-03 Univ California MEMBRANE-WRAPPED NANOPARTICLES AND METHOD OF USE
CN105031637A (en) 2011-07-11 2015-11-11 武田疫苗股份有限公司 Parenteral norovirus vaccine formulations
CA2843274A1 (en) 2011-07-29 2013-02-07 Selecta Biosciences, Inc. Synthetic nanocarriers that generate humoral and cytotoxic t lymphocyte (ctl) immune responses
US20130122106A1 (en) * 2011-10-19 2013-05-16 Aphios Corporation Dosage form, and methods of making and using the same, to produce immunization in animals and humans
KR101822941B1 (en) * 2012-02-06 2018-01-29 엘지전자 주식회사 Air cleaning filter and method for manufacturing the same
EP2634179A1 (en) * 2012-02-28 2013-09-04 Sanofi Functional PLA-PEG copolymers, the nanoparticles thereof, their preparation and use for targeted drug delivery and imaging
WO2014018931A1 (en) * 2012-07-26 2014-01-30 The General Hospital Corporation Methods and compositions for treating autoimmune disease
US10323279B2 (en) 2012-08-14 2019-06-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9701998B2 (en) 2012-12-14 2017-07-11 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11591637B2 (en) 2012-08-14 2023-02-28 10X Genomics, Inc. Compositions and methods for sample processing
US10752949B2 (en) 2012-08-14 2020-08-25 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10400280B2 (en) 2012-08-14 2019-09-03 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10533221B2 (en) 2012-12-14 2020-01-14 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10206985B2 (en) * 2013-02-05 2019-02-19 Nitto Denko Corporation WT1 peptide cancer vaccine composition for mucosal administration
KR102190198B1 (en) 2013-02-08 2020-12-14 10엑스 제노믹스, 인크. Polynucleotide barcode generation
JP6697384B2 (en) 2013-07-25 2020-05-20 イグジキュア, インコーポレーテッドExicure, Inc. Spherical nucleic acid-based constructs as immunostimulants for prophylactic and therapeutic use
CN105764524A (en) * 2013-08-06 2016-07-13 约翰斯·霍普金斯大学 Methods of treatment of HPV related diseases
CN104338126B (en) * 2013-08-08 2018-05-04 中国科学院过程工程研究所 It is a kind of that there is the vaccine combination for treating or preventing HPV viruse and its application
WO2015066715A1 (en) * 2013-11-04 2015-05-07 Viracell Advanced Products, Llc Virus-like particles and methods related thereto
JP2017503803A (en) 2014-01-10 2017-02-02 シャンハイ バーディー バイオテック インコーポレイテッド Compounds and compositions for treating EGFR expressing tumors
AU2015269412B2 (en) 2014-06-04 2020-03-12 Exicure Operating Company Multivalent delivery of immune modulators by liposomal spherical nucleic acids for prophylactic or therapeutic applications
WO2015200054A2 (en) 2014-06-24 2015-12-30 The Trustees Of Princeton University Process for encapsulating soluble biologics, therapeutics, and imaging agents
AU2015279738A1 (en) 2014-06-25 2016-12-22 Selecta Biosciences, Inc. Methods and compositions for treatment with synthetic nanocarriers and immune checkpoint inhibitors
MX2016016902A (en) 2014-06-26 2017-03-27 10X Genomics Inc Methods of analyzing nucleic acids from individual cells or cell populations.
WO2016004876A1 (en) 2014-07-09 2016-01-14 Shanghai Birdie Biotech, Inc. Anti-pd-l1 combinations for treating tumors
JP6836829B2 (en) * 2014-08-04 2021-03-03 日東電工株式会社 Immunity induction promoting composition and vaccine pharmaceutical composition containing nuclear receptor ligand
GB2549861A (en) * 2014-11-03 2017-11-01 Albert Einstein College Medicine Inc Modified paramagnetic nanoparticles for targeted delivery of therapeutics and methods thereof
RU2600031C2 (en) * 2014-11-11 2016-10-20 Публичное акционерное общество "Фармсинтез" Dosage form of specific immunobiological medicine for treating and preventing hiv infection and preparation method thereof
WO2016081911A2 (en) 2014-11-21 2016-05-26 Northwestern University The sequence-specific cellular uptake of spherical nucleic acid nanoparticle conjugates
US10339559B2 (en) * 2014-12-04 2019-07-02 Adobe Inc. Associating social comments with individual assets used in a campaign
KR101586466B1 (en) * 2014-12-31 2016-01-18 성균관대학교산학협력단 Immunoadjuvant and vaccine composition including the same
KR101595949B1 (en) * 2015-05-27 2016-02-19 성균관대학교산학협력단 Producing method of adjuvant composition and vaccine composition
EP3307313A1 (en) 2015-06-12 2018-04-18 GlaxoSmithKline Biologicals SA Adenovirus polynucleotides and polypeptides
CA2989332A1 (en) * 2015-06-15 2016-12-22 Emory University Multivalent enterovirus vaccine compositions and uses related thereto
JP6978079B2 (en) * 2015-07-02 2021-12-08 メディジェン, インコーポレイテッド Recombinant virus-like particles using bovine immunodeficiency virus GAG protein
JP2018522053A (en) 2015-08-06 2018-08-09 グラクソスミスクライン、インテレクチュアル、プロパテ TLR4 agonists and compositions thereof and their use in the treatment of cancer
WO2017112828A1 (en) 2015-12-22 2017-06-29 The Trustees Of Princeton University Process for encapsulating soluble biologics, therapeutics, and imaging agents
CN115554406A (en) 2016-01-07 2023-01-03 博笛生物科技有限公司 anti-CD 20 combinations for the treatment of tumors
CN115252792A (en) 2016-01-07 2022-11-01 博笛生物科技有限公司 anti-EGFR combinations for the treatment of tumors
WO2017120504A1 (en) 2016-01-08 2017-07-13 Durfee Paul N Osteotropic nanoparticles for prevention or treatment of bone metastases
CN105664152B (en) * 2016-01-27 2019-01-18 苏文全 A kind of double stranded polynucleotide with immunoregulation effect-epsilon-polylysine compound and its preparation application method
CN105535964B (en) * 2016-01-27 2019-01-18 苏文全 A kind of double stranded polynucleotide-epsilon-polylysine-sulfuric acid glycan compound and its preparation application method with immunoregulation effect
WO2018039629A2 (en) 2016-08-25 2018-03-01 Northwestern University Micellar spherical nucleic acids from thermoresponsive, traceless templates
US10369190B2 (en) 2016-09-13 2019-08-06 Allergan, Inc. Non-protein clostridial toxin compositions
CN106496309A (en) * 2016-11-24 2017-03-15 北京开景基因技术有限公司 Microballoon antigen and preparation method thereof and the preparation method of anti-cotinine antibody
KR101996538B1 (en) * 2017-02-13 2019-07-04 단디바이오사이언스 주식회사 Nanoemulsion containing imidazoquinoline-based material and uses thereof
WO2018160865A1 (en) 2017-03-01 2018-09-07 Charles Jeffrey Brinker Active targeting of cells by monosized protocells
CN106943592A (en) * 2017-03-02 2017-07-14 暨南大学 Application of the phosphonized chitosan as immunologic adjuvant in vaccine therapy
CN110582282A (en) * 2017-04-25 2019-12-17 佐剂技术公司 Triterpene saponin analogues
CN108794467A (en) 2017-04-27 2018-11-13 博笛生物科技有限公司 2- amino-quinoline derivatives
US11696954B2 (en) 2017-04-28 2023-07-11 Exicure Operating Company Synthesis of spherical nucleic acids using lipophilic moieties
JP7080501B2 (en) 2017-06-23 2022-06-06 バーディー バイオファーマシューティカルズ インコーポレイテッド Pharmaceutical composition
NL2019373B1 (en) * 2017-07-28 2019-02-19 Academisch Ziekenhuis Leiden Enhancement of pathogen immunogenicity
WO2019028387A1 (en) * 2017-08-03 2019-02-07 Rita Elena Serda Liposomal coated nanoparticles for immunotherapy applications
US11123415B2 (en) 2017-08-16 2021-09-21 Ohio State Innovation Foundation Nanoparticle compositions for Salmonella vaccines
WO2019055539A1 (en) 2017-09-12 2019-03-21 Prudhomme Robert K Cellulosic polymer nanoparticles and methods of forming them
CN107582564A (en) * 2017-09-14 2018-01-16 湖南晓林生物科技发展有限公司 A kind of medicine of targeted therapy thyroid cancer and preparation method thereof
US10590244B2 (en) * 2017-10-04 2020-03-17 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
US10837047B2 (en) * 2017-10-04 2020-11-17 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
EP3691703A1 (en) * 2017-10-04 2020-08-12 10X Genomics, Inc. Compositions, methods, and systems for bead formation using improved polymers
WO2019074842A1 (en) * 2017-10-09 2019-04-18 Keith Black Oncolytic cancer immunotherapies and methods of use
EP3625361A1 (en) 2017-11-15 2020-03-25 10X Genomics, Inc. Functionalized gel beads
US10829815B2 (en) 2017-11-17 2020-11-10 10X Genomics, Inc. Methods and systems for associating physical and genetic properties of biological particles
WO2019108928A1 (en) * 2017-11-30 2019-06-06 Ohio State Innovation Foundation Mucoadhesive nanoparticle entrapped influenza virus vaccine delivery system
CN108379562B (en) * 2018-03-20 2021-11-12 苏州杰纳生物科技有限公司 Polymer nano adjuvant and preparation method and application thereof
CN112020823A (en) 2018-05-10 2020-12-01 日产自动车株式会社 Motor system control method and motor system control device
EA038215B1 (en) * 2018-06-09 2021-07-26 Федеральное государственное бюджетное научное учреждение "Федеральный научный центр исследований и разработки иммунобиологических препаратов им. М.П. Чумакова РАН" Method for quantitative determination of yellow fever virus antigen by enzyme immunoassay using specific yolk antibodies and biotin-labelled detector antibodies
US11731099B2 (en) 2018-07-20 2023-08-22 The Trustees Of Princeton University Method for controlling encapsulation efficiency and burst release of water soluble molecules from nanoparticles and microparticles produced by inverse flash nanoprecipitation
CN109187982B (en) * 2018-08-02 2021-06-04 浙江康佰裕生物科技有限公司 Method for screening and identifying TLR vaccine adjuvant
WO2020083776A1 (en) 2018-10-24 2020-04-30 Alps Electric Europe Gmbh Asset tracking device, asset and a method of determining whether an asset tracking device is transported by a predetermined type of transportation means
US20220117997A1 (en) * 2019-02-05 2022-04-21 The Brigham And Women's Hospital Inc. Polysaccharide compositions for use in treating filariasis
CN110559432B (en) * 2019-10-11 2023-06-13 南京农业大学 Eimeria acervulina nano subunit vaccine and preparation method and application thereof
JP2021127868A (en) 2020-02-14 2021-09-02 株式会社デンソー Heat exchanger
US11559578B2 (en) 2020-06-30 2023-01-24 International Business Machines Corporation Biodegradable cationic polycarbonates as adjuvants for vaccines
WO2022092769A1 (en) * 2020-10-26 2022-05-05 한국과학기술원 Fusion protein comprising bp26 and antigenic polypeptide
WO2022109484A1 (en) * 2020-11-23 2022-05-27 Wisconsin Alumni Research Foundation Neutralizing vaccines against human coronavirus
CN112972673B (en) * 2021-02-02 2023-04-11 兰州大学 PLGA-PEG-Poly I: preparation of C nano-particles and application thereof in tuberculosis subunit vaccine
WO2022226035A1 (en) * 2021-04-21 2022-10-27 The Board Of Trustees Of The Leland Stanford Junior University Toll-like receptor agonist-nanoparticle vaccine adjuvant
WO2023032891A1 (en) * 2021-08-30 2023-03-09 東レ株式会社 Composition for enhancing immunogenicity
WO2023107371A1 (en) * 2021-12-06 2023-06-15 SURGE Therapeutics, Inc. Solid forms of resiquimod and formulations thereof
WO2023225503A2 (en) * 2022-05-16 2023-11-23 Dairy Management Inc. Protein particles including an active agent and methods of making and using the same
KR20230002334U (en) 2022-06-02 2023-12-11 성기봉 disposable ice pack
CN115645523B (en) * 2022-12-22 2023-03-21 深圳大学总医院 Application of polymer lipid hybrid nanoparticles as immunologic adjuvant and immunologic preparation
CN116478410B (en) * 2023-06-20 2023-09-12 觅投克(北京)生物医学技术有限公司 Inulin-modified polyethyleneimine derivative and preparation method and application thereof

Citations (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4638045A (en) 1985-02-19 1987-01-20 Massachusetts Institute Of Technology Non-peptide polyamino acid bioerodible polymers
US4806621A (en) 1986-01-21 1989-02-21 Massachusetts Institute Of Technology Biocompatible, bioerodible, hydrophobic, implantable polyimino carbonate article
US4946929A (en) 1983-03-22 1990-08-07 Massachusetts Institute Of Technology Bioerodible articles useful as implants and prostheses having predictable degradation rates
US5010167A (en) 1989-03-31 1991-04-23 Massachusetts Institute Of Technology Poly(amide-and imide-co-anhydride) for biological application
US5019379A (en) 1987-07-31 1991-05-28 Massachusetts Institute Of Technology Unsaturated polyanhydrides
US5399665A (en) 1992-11-05 1995-03-21 Massachusetts Institute Of Technology Biodegradable polymers for cell transplantation
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
US5543158A (en) 1993-07-23 1996-08-06 Massachusetts Institute Of Technology Biodegradable injectable nanoparticles
US5578325A (en) 1993-07-23 1996-11-26 Massachusetts Institute Of Technology Nanoparticles and microparticles of non-linear hydrophilic-hydrophobic multiblock copolymers
US5663153A (en) 1994-03-25 1997-09-02 Isis Pharmaceuticals, Inc. Immune stimulation by phosphorothioate oligonucleotide analogs
US5716404A (en) 1994-12-16 1998-02-10 Massachusetts Institute Of Technology Breast tissue engineering
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
US5750110A (en) 1992-06-25 1998-05-12 Smithkline Beecham Biologicals, S.A Vaccine composition containing adjuvants
US5770417A (en) 1986-11-20 1998-06-23 Massachusetts Institute Of Technology Children's Medical Center Corporation Three-dimensional fibrous scaffold containing attached cells for producing vascularized tissue in vivo
US5804178A (en) 1986-11-20 1998-09-08 Massachusetts Institute Of Technology Implantation of cell-matrix structure adjacent mesentery, omentum or peritoneum tissue
US5837752A (en) 1997-07-17 1998-11-17 Massachusetts Institute Of Technology Semi-interpenetrating polymer networks
US5902599A (en) 1996-02-20 1999-05-11 Massachusetts Institute Of Technology Biodegradable polymer networks for use in orthopedic and dental applications
US6007845A (en) 1994-07-22 1999-12-28 Massachusetts Institute Of Technology Nanoparticles and microparticles of non-linear hydrophilic-hydrophobic multiblock copolymers
US6095148A (en) 1995-11-03 2000-08-01 Children's Medical Center Corporation Neuronal stimulation using electrically conducting polymers
US6123727A (en) 1995-05-01 2000-09-26 Massachusetts Institute Of Technology Tissue engineered tendons and ligaments
US6130082A (en) 1988-05-05 2000-10-10 American Cyanamid Company Recombinant flagellin vaccines
US6194388B1 (en) 1994-07-15 2001-02-27 The University Of Iowa Research Foundation Immunomodulatory oligonucleotides
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6329381B1 (en) 1997-11-28 2001-12-11 Sumitomo Pharmaceuticals Company, Limited Heterocyclic compounds
WO2002032450A2 (en) 2000-10-18 2002-04-25 Glaxosmithkline Biologicals S.A. Vaccines
US6506577B1 (en) 1998-03-19 2003-01-14 The Regents Of The University Of California Synthesis and crosslinking of catechol containing copolypeptides
US6544518B1 (en) 1999-04-19 2003-04-08 Smithkline Beecham Biologicals S.A. Vaccines
US6585980B1 (en) 1997-04-11 2003-07-01 The University Of Toronto Flagellin gene, FlaC of Campylobacter
US6632922B1 (en) 1998-03-19 2003-10-14 The Regents Of The University Of California Methods and compositions for controlled polypeptide synthesis
US6632671B2 (en) 2000-02-28 2003-10-14 Genesegues, Inc. Nanoparticle encapsulation system and method
WO2003086280A2 (en) 2002-04-04 2003-10-23 Coley Pharmaceutical Gmbh Immunostimulatory g,u-containing oligoribonucleotides
US6686446B2 (en) 1998-03-19 2004-02-03 The Regents Of The University Of California Methods and compositions for controlled polypeptide synthesis
US6696076B2 (en) 1996-10-25 2004-02-24 3M Innovative Properties Company Immune response modifier compounds for treatment of TH2 mediated and related diseases
WO2004053056A2 (en) 2002-09-24 2004-06-24 University Of Kentucky Research Foundation Nanoparticle-based vaccine delivery system containing adjuvant
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
WO2004098509A2 (en) 2003-04-30 2004-11-18 Chiron Corporation Compositions for inducing immune responses
WO2005097993A2 (en) 2004-02-19 2005-10-20 Coley Pharmaceutical Group, Inc. Immunostimulatory viral rna oligonucleotides
US20060002852A1 (en) 2004-07-01 2006-01-05 Yale University Targeted and high density drug loaded polymeric materials
US20060222652A1 (en) 2000-05-05 2006-10-05 Cytos Biotechnology Ag Molecular antigen array
US20060241076A1 (en) 2005-04-26 2006-10-26 Coley Pharmaceutical Gmbh Modified oligoribonucleotide analogs with enhanced immunostimulatory activity
US20060251677A1 (en) 2003-03-26 2006-11-09 Cytos Biotechnology Ag Packaging of immunostimulatory oligonucleotides into virus-like particles: method of preparation and use
US7192725B2 (en) 2000-05-19 2007-03-20 University Of Toronto Flagellin gene, flaC of Campylobacter
US7202351B1 (en) 1993-09-14 2007-04-10 Pharmexa Inc. Alteration of immune response using pan DR-binding peptides
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
WO2007062107A2 (en) 2005-11-25 2007-05-31 Coley Pharmaceutical Gmbh Immunostimulatory oligoribonucleotides
US7250403B2 (en) 2000-03-10 2007-07-31 Dynavax Technologies Corporation Biodegradable immunomodulatory formulations and methods for use thereof
US7262286B2 (en) 2000-09-26 2007-08-28 Idera Pharmaceuticals, Inc. Modulation of immunostimulatory activity of immunostimulatory oligonucleotide analogs by positional chemical changes
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
WO2008033432A2 (en) 2006-09-12 2008-03-20 Coley Pharmaceutical Group, Inc. Immune modulation by chemically modified ribonucleosides and oligoribonucleotides
US7357936B1 (en) 1998-10-16 2008-04-15 Smithkline Beecham Biologicals, Sa Adjuvant systems and vaccines
US20080145441A1 (en) 2000-10-16 2008-06-19 Midatech Limited Nanoparticles
WO2008127532A1 (en) 2007-04-12 2008-10-23 Emory University Novel strategies for delivery of active agents using micelles and particles
US20090028910A1 (en) 2003-12-19 2009-01-29 University Of North Carolina At Chapel Hill Methods for Fabrication Isolated Micro-and Nano-Structures Using Soft or Imprint Lithography
WO2009051837A2 (en) 2007-10-12 2009-04-23 Massachusetts Institute Of Technology Vaccine nanotechnology
WO2009076158A1 (en) 2007-12-07 2009-06-18 Novartis Ag Compositions for inducing immune responses
US7566703B2 (en) 2004-10-20 2009-07-28 Coley Pharmaceutical Group, Inc. Semi-soft C-class immunostimulatory oligonucleotides
WO2009106999A2 (en) 2008-02-28 2009-09-03 Deutsches Krebsforschungszentrum, Stiftung Des Öffentlichen Rechts Hollow nanoparticles and uses thereof
US20090226525A1 (en) 2007-04-09 2009-09-10 Chimeros Inc. Self-assembling nanoparticle drug delivery system
WO2010018132A1 (en) 2008-08-11 2010-02-18 Smithkline Beecham Corporation Compounds
US20100075995A1 (en) 2008-08-11 2010-03-25 Smithkline Beecham Corporation Compounds
WO2010047839A1 (en) 2008-10-25 2010-04-29 Aura Biosciences Modified plant virus particles and uses therefor

Family Cites Families (306)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR7461M (en) 1968-06-19 1970-01-05
GB1355961A (en) 1970-02-27 1974-06-12 Wellcome Found Preparation of immunosuppressive antilymphocytic serum
CH594444A5 (en) 1972-12-04 1978-01-13 Gerd Birrenbach
US3996355A (en) 1975-01-02 1976-12-07 American Home Products Corporation Permanent suspension pharmaceutical dosage form
DK143689C (en) 1975-03-20 1982-03-15 J Kreuter PROCEDURE FOR THE PREPARATION OF AN ADVERTISED VACCINE
US4756907A (en) 1978-10-17 1988-07-12 Stolle Research & Development Corp. Active/passive immunization of the internal female reproductive organs
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
US4631211A (en) 1985-03-25 1986-12-23 Scripps Clinic & Research Foundation Means for sequential solid phase organic synthesis and methods using the same
JPS63122620A (en) 1986-11-12 1988-05-26 Sanraku Inc Polylactic acid microsphere and production thereof
FR2608988B1 (en) 1986-12-31 1991-01-11 Centre Nat Rech Scient PROCESS FOR THE PREPARATION OF COLLOIDAL DISPERSIBLE SYSTEMS OF A SUBSTANCE, IN THE FORM OF NANOPARTICLES
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
US4950432A (en) 1987-10-16 1990-08-21 Board Of Regents, The University Of Texas System Polyene microlide pre-liposomal powders
US4929624A (en) 1989-03-23 1990-05-29 Minnesota Mining And Manufacturing Company Olefinic 1H-imidazo(4,5-c)quinolin-4-amines
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
US6005087A (en) 1995-06-06 1999-12-21 Isis Pharmaceuticals, Inc. 2'-modified oligonucleotides
US6399754B1 (en) 1991-12-24 2002-06-04 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides
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
SG46492A1 (en) 1991-03-01 1998-02-20 Minnesota Mining & Mfg 1-Substituted 2-substituted 1H-imidazo [4,5-c] quinolin-4-amines
CA2084194C (en) 1991-04-02 2003-05-20 Gregory John Russell-Jones 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
EP0654973A4 (en) 1992-07-21 1995-08-09 Gen Hospital Corp 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 (en) 1992-09-11 1994-12-02 Pasteur Institut Microparticles carrying antigens and their use for the induction of humoral or cellular responses.
WO1994007469A1 (en) 1992-09-25 1994-04-14 Dynagen, Inc. An immunobooster for delayed release of immunogen
WO1994015635A1 (en) 1993-01-11 1994-07-21 Dana-Farber Cancer Institute Inducing cytotoxic t lymphocyte responses
US5395937A (en) 1993-01-29 1995-03-07 Minnesota Mining And Manufacturing Company Process for preparing quinoline amines
WO1994018955A1 (en) 1993-02-22 1994-09-01 Alza Corporation Compositions for oral delivery of active agents
JPH08508721A (en) 1993-03-17 1996-09-17 シリカゲル ゲス.エム.ビー.エイチ Superparamagnetic particles, their manufacturing method and their use
CH686761A5 (en) 1993-05-27 1996-06-28 Sandoz Ag Pharmaceutical formulations.
WO1995003035A1 (en) 1993-07-23 1995-02-02 Massachusetts Institute Of Technology Polymerized liposomes with enhanced stability for oral delivery
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
ATE228832T1 (en) 1994-02-28 2002-12-15 Nanopharm Ag SYSTEM FOR THE TARGETED DELIVERY OF ACTIVE INGREDIENTS, METHOD FOR THE PRODUCTION AND USE THEREOF
US5596091A (en) 1994-03-18 1997-01-21 The Regents Of The University Of California Antisense oligonucleotides comprising 5-aminoalkyl pyrimidine nucleotides
GB9412273D0 (en) 1994-06-18 1994-08-10 Univ Nottingham Administration means
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
EP0805678B1 (en) 1995-01-05 2003-10-29 THE BOARD OF REGENTS acting for and on behalf of THE UNIVERSITY OF MICHIGAN Surface-modified nanoparticles and method of making and using same
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
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
AU710347B2 (en) 1995-08-31 1999-09-16 Alkermes Controlled Therapeutics, Inc. Composition for sustained release of an agent
US5874064A (en) 1996-05-24 1999-02-23 Massachusetts Institute Of Technology Aerodynamically light particles for pulmonary drug delivery
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
AU6173196A (en) 1996-06-10 1998-01-07 Cytos Pharmaceuticals Llc Imidazole derivatives as protective agents in reperfusion injury and severe inflammatory responses
US5922695A (en) 1996-07-26 1999-07-13 Gilead Sciences, Inc. Antiviral phosphonomethyoxy nucleotide analogs having increased oral bioavarilability
JP2001504448A (en) 1996-08-30 2001-04-03 フュルステ,イェンス,ペーター Mirror selection and evolution of nucleic acids
US6043224A (en) 1996-09-05 2000-03-28 The Massachusetts Institute Of Technology Compositions and methods for treatment of neurological disorders and neurodegenerative diseases
US6042820A (en) 1996-12-20 2000-03-28 Connaught Laboratories Limited Biodegradable copolymer containing α-hydroxy acid and α-amino acid units
US6127533A (en) 1997-02-14 2000-10-03 Isis Pharmaceuticals, Inc. 2'-O-aminooxy-modified oligonucleotides
WO1998037919A1 (en) 1997-02-28 1998-09-03 University Of Iowa Research Foundation USE OF NUCLEIC ACIDS CONTAINING UNMETHYLATED CpG DINUCLEOTIDE IN THE TREATMENT OF LPS-ASSOCIATED DISORDERS
DE69841122D1 (en) 1997-03-10 2009-10-15 Coley Pharm Gmbh Use of non-methylated CpG dinucleotide in combination with aluminum as adjuvants
US5989591A (en) 1997-03-14 1999-11-23 American Home Products Corporation Rapamycin formulations for oral administration
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
CA2301575C (en) 1997-05-20 2003-12-23 Ottawa Civic Hospital Loeb Research Institute Vectors and methods for immunization or therapeutic protocols
US5985325A (en) 1997-06-13 1999-11-16 American Home Products Corporation Rapamycin formulations for oral administration
US6989435B2 (en) 1997-09-11 2006-01-24 Cambridge University Technical Services Ltd. Compounds and methods to inhibit or augment an inflammatory response
DE19745950A1 (en) 1997-10-17 1999-04-22 Dds Drug Delivery Service Ges Drug carrier particle for site specific drug delivery, especially to CNS
US6197229B1 (en) 1997-12-12 2001-03-06 Massachusetts Institute Of Technology Method for high supercoiled DNA content microspheres
US6254890B1 (en) 1997-12-12 2001-07-03 Massachusetts Institute Of Technology Sub-100nm biodegradable polymer spheres capable of transporting and releasing nucleic acids
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
FR2775435B1 (en) 1998-02-27 2000-05-26 Bioalliance Pharma NANOPARTICLES COMPRISING AT LEAST ONE POLYMER AND AT LEAST ONE COMPOUND CAPABLE OF COMPLEXING ONE OR MORE ACTIVE INGREDIENTS
ATE339223T1 (en) * 1998-03-09 2006-10-15 Glaxosmithkline Biolog Sa COMBINED VACCINE COMPOSITIONS
US6232287B1 (en) 1998-03-13 2001-05-15 The Burnham Institute Molecules that home to various selected organs or tissues
US6218371B1 (en) 1998-04-03 2001-04-17 University Of Iowa Research Foundation Methods and products for stimulating the immune system using immunotherapeutic oligonucleotides and cytokines
JP2002513763A (en) 1998-05-06 2002-05-14 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 Methods for preventing and treating parasitic infections and related diseases using CPG oligonucleotides
SE9801923D0 (en) 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 (en) 1998-07-29 2006-04-15 Chiron Corp MICROPARTICLES WITH ADSORBENT SURFACES, METHOD FOR THEIR PRODUCTION AND THEIR USE
DE19839214C1 (en) 1998-08-28 2000-05-25 Aventis Res & Tech Gmbh & Co Process for the production of spherical microparticles with a smooth surface which consist wholly or partly of at least one water-insoluble linear polysaccharide, and microparticles obtainable by this process and their use
CA2345817C (en) 1998-10-05 2013-02-12 M&E Biotech A/S Novel methods for therapeutic vaccination
US6306640B1 (en) 1998-10-05 2001-10-23 Genzyme Corporation Melanoma antigenic peptides
CN100444830C (en) 1998-11-02 2008-12-24 伊兰公司,Plc Multiparticulate modified release composition
US7521068B2 (en) 1998-11-12 2009-04-21 Elan Pharma International Ltd. Dry powder aerosols of nanoparticulate drugs
US6232082B1 (en) 1998-12-01 2001-05-15 Nabi Hapten-carrier conjugates for treating and preventing nicotine addiction
US6486168B1 (en) 1999-01-08 2002-11-26 3M Innovative Properties Company Formulations and methods for treatment of mucosal associated conditions with an immune response modifier
US6403779B1 (en) 1999-01-08 2002-06-11 Isis Pharmaceuticals, Inc. Regioselective synthesis of 2′-O-modified nucleosides
CA2361936C (en) 1999-01-08 2009-06-16 3M Innovative Properties Company Formulations comprising imiquimod or other immune response modifiers for treating mucosal conditions
US7238711B1 (en) 1999-03-17 2007-07-03 Cambridge University Technical Services Ltd. Compounds and methods to inhibit or augment an inflammatory response
DE19956568A1 (en) 1999-01-30 2000-08-17 Roland Kreutzer Method and medicament for inhibiting the expression of a given gene
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
EP1154790B1 (en) 1999-02-26 2004-10-20 Chiron S.r.l. Enhancement of bactericidal activity of neisseria antigens with oligonucleotides containing cg motifs
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US6110462A (en) 1999-03-03 2000-08-29 The Scripps Research Institute Enzymatic DNA molecules that contain modified nucleotides
US6800296B1 (en) 1999-05-19 2004-10-05 Massachusetts Institute Of Technology Modification of surfaces using biological recognition events
US6331539B1 (en) 1999-06-10 2001-12-18 3M Innovative Properties Company Sulfonamide and sulfamide substituted imidazoquinolines
US6815170B1 (en) 1999-06-30 2004-11-09 John Wayne Cancer Institute Methods for lymph node identification
AU6635900A (en) 1999-08-13 2001-03-13 Point Biomedical Corporation Microparticles useful as ultrasonic contrast agents and for lymphatic system
NZ517929A (en) 1999-09-25 2004-02-27 Univ Iowa Res Found Immunostimulatory nucleic acids
DE60041413D1 (en) 1999-10-12 2009-03-05 Ca Nat Research Council ARCHAEOSOME AS AN ADJUVANTIA AND SUPPORT FOR AZELLULAR IMPULSES FOR THE INDUCTION OF A CYTOTOXIC T-LYMPHOCYTE (CTL) IMMUNE RESPONSE
CA2391534A1 (en) 1999-11-15 2001-05-25 Drug Innovation & Design, Inc. Selective cellular targeting: multifunctional delivery vehicles
JP2004501340A (en) 2000-01-13 2004-01-15 ナノスフェアー インコーポレイテッド Oligonucleotide-attached nanoparticles and methods of use
AT409085B (en) 2000-01-28 2002-05-27 Cistem Biotechnologies Gmbh PHARMACEUTICAL COMPOSITION FOR IMMUNULATING AND PRODUCING VACCINES
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)
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
US7129222B2 (en) 2000-03-10 2006-10-31 Dynavax Technologies Corporation Immunomodulatory formulations and methods for use thereof
US7157437B2 (en) 2000-03-10 2007-01-02 Dynavax Technologies Corporation Methods of ameliorating symptoms of herpes infection using immunomodulatory polynucleotide sequences
SE0000933D0 (en) 2000-03-21 2000-03-21 Independent Pharmaceutica Ab Method of producing 6-substituted (S) -nicotine derivatives and intermediate compounds
EP2345742B1 (en) 2000-03-30 2014-06-11 The Whitehead Institute for Biomedical Research RNA sequence-specific mediators of RNA interference
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
GB0108364D0 (en) * 2001-04-03 2001-05-23 Glaxosmithkline Biolog Sa Vaccine composition
AU2001297913A1 (en) * 2000-10-13 2002-12-23 Ligocyte Pharmaceuticals, Inc. Polyvalent nanoparticles
WO2002055185A2 (en) 2000-10-19 2002-07-18 Eidgenoess Tech Hochschule 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
CZ308053B6 (en) 2000-12-01 2019-11-27 Max Planck Gesellschaft Isolated double-stranded RNA molecule, process for producing it and its use
US20060142202A1 (en) 2000-12-08 2006-06-29 3M Innovative Properties Company Compositions and methods for targeted delivery of immune response modifiers
CA2430691A1 (en) 2000-12-27 2002-07-04 Dynavax Technologies Corporation Immunomodulatory polynucleotides and methods of using the same
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
JP2006512401A (en) 2001-06-05 2006-04-13 ザ リージェンツ オブ ザ ユニバーシティ オブ ミシガン Nanoemulsion vaccine
EP1264603B8 (en) 2001-06-10 2010-03-03 Noxxon Pharma AG Use of L-polynucleotides for in vivo imaging
AU2002354644C1 (en) 2001-07-10 2009-04-30 Corixa Corporation Compositions and methods for delivery of proteins and adjuvants encapsulated in microspheres
JP4607452B2 (en) 2001-08-07 2011-01-05 ダイナバックス テクノロジーズ コーポレイション Immunomodulating composition, formulation and method of use thereof
US20030054042A1 (en) 2001-09-14 2003-03-20 Elaine Liversidge Stabilization of chemical compounds using nanoparticulate formulations
CA2465405A1 (en) 2001-11-02 2003-05-08 Wockhardt Limited Controlled release compositions for macrolide antimicrobial agents
US8088388B2 (en) 2002-02-14 2012-01-03 United Biomedical, Inc. Stabilized synthetic immunogen delivery system
CA2476626A1 (en) 2002-02-20 2003-08-28 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
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
AU2003234196A1 (en) 2002-04-22 2003-11-03 University Of Florida Functionalized nanoparticles and methods of use
US6824338B2 (en) * 2002-05-28 2004-11-30 Satco, Inc. Air transport modular container system
US20040092470A1 (en) 2002-06-18 2004-05-13 Leonard Sherry A. Dry powder oligonucleotide formualtion, preparation and its uses
WO2004005325A2 (en) 2002-07-10 2004-01-15 The Ohio State University Research Foundation Antigen-polymer compositions
CN101574518B (en) 2002-07-18 2012-08-22 赛托斯生物技术公司 Hapten-carrier conjugates and uses thereof
ATE488246T1 (en) 2002-08-15 2010-12-15 3M Innovative Properties Co IMMUNO-STIMULATORY COMPOSITIONS AND METHODS FOR STIMULATING AN IMMUNE RESPONSE
US20040091503A1 (en) * 2002-08-20 2004-05-13 Genitrix, Llc Lectin compositions and methods for modulating an immune response to an antigen
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
US7008411B1 (en) 2002-09-30 2006-03-07 Advanced Cardiovascular Systems, Inc. Method and apparatus for treating vulnerable plaque
NO20024755D0 (en) 2002-10-03 2002-10-03 Amersham Health As Method
US7670627B2 (en) 2002-12-09 2010-03-02 Salvona Ip Llc pH triggered targeted controlled release systems for the delivery of pharmaceutical active ingredients
WO2004053104A2 (en) 2002-12-11 2004-06-24 Coley Pharmaceutical Group, Inc. 5’ cpg nucleic acids and methods of use
SE0203687D0 (en) 2002-12-13 2002-12-13 Ian Harwigsson Med Adagit Fa Pharmaceutical Porous Particles
CN100546998C (en) 2002-12-23 2009-10-07 戴纳伐克斯技术股份有限公司 Immunostimulatory sequence oligonucleotides and using method
EP2572715A1 (en) 2002-12-30 2013-03-27 3M Innovative Properties Company Immunostimulatory Combinations
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
ATE371437T1 (en) 2003-02-17 2007-09-15 Peter Burkhard PEPTIDIC NANOPARTICLES AS DRUG DELIVERY AND ANTIGEN DISPLAY SYSTEMS
AU2004224277A1 (en) * 2003-03-21 2004-10-07 Wyeth Treating immunological disorder using agonists of interleukin-21/ interleukin-21 receptor
US20040191215A1 (en) 2003-03-25 2004-09-30 Michael Froix Compositions for induction of a therapeutic response
AU2004224530A1 (en) 2003-03-26 2004-10-07 Ltt Bio-Pharma Co., Ltd. Intravenous nanoparticles for targenting drug delivery and sustained drug release
CA2521682A1 (en) 2003-04-10 2004-12-16 3M Innovative Properties Company Delivery of immune response modifier compounds using metal-containing particulate support materials
CA2528007C (en) * 2003-06-02 2012-03-27 Chiron Corporation Immunogenic compositions based on microparticles comprising adsorbed toxoid and a polysaccharide-containing antigen
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
CA2544240A1 (en) 2003-07-22 2005-02-17 Cytos Biotechnology Ag Cpg-packaged liposomes
US20050042298A1 (en) 2003-08-20 2005-02-24 Pardridge William M. Immunonanoparticles
AU2004281634B2 (en) 2003-09-03 2011-01-27 Dendritherapeutics, Inc. Multiplex vaccines
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
JP2007514519A (en) 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 Method for producing microcapsules comprising polymer and charged nanoparticles
CN1608675A (en) * 2003-10-22 2005-04-27 四川大学 Medicine-carrying nanometer polymer particle and its prepn and use
SG122973A1 (en) 2003-10-30 2006-06-29 Coley Pharm Gmbh C-class oligonucleotide analogs with enhanced immunostimulatory potency
KR20060130057A (en) 2003-11-21 2006-12-18 알자 코포레이션 Gene delivery mediated by liposome-dna complex with cleavable peg surface modification
CN1544638A (en) * 2003-11-28 2004-11-10 中国药科大学 Virus-like particle capable of carrying and loading polypeptide
US20070116768A1 (en) 2003-12-09 2007-05-24 Michael Chorny Sustained release preparations composed of biocompatible complex microparticles
KR100581967B1 (en) 2003-12-18 2006-05-22 한국유나이티드제약 주식회사 Double pellet formulation of proton pump inhibitors and clarithromycin for the treatment of gastrointestinal ulcer, and method for producing the same
FR2863890B1 (en) * 2003-12-19 2006-03-24 Aventis Pasteur IMMUNOSTIMULATING COMPOSITION
US9040090B2 (en) 2003-12-19 2015-05-26 The University Of North Carolina At Chapel Hill Isolated and fixed micro and nano structures and methods thereof
EP1550458A1 (en) * 2003-12-23 2005-07-06 Vectron Therapeutics AG Synergistic liposomal adjuvants
WO2005065418A2 (en) 2003-12-31 2005-07-21 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
EP1735010A4 (en) 2004-04-09 2008-08-27 3M Innovative Properties Co Methods, compositions, and preparations for delivery of immune response modifiers
CA2562685C (en) 2004-04-27 2013-09-17 Alnylam Pharmaceuticals, Inc. Single-stranded and double-stranded oligonucleotides comprising a 2-arylpropyl moiety
ES2246695B1 (en) 2004-04-29 2007-05-01 Instituto Cientifico Y Tecnologico De Navarra, S.A. STIMULATING COMPOSITION OF THE IMMUNE RESPONSE THAT INCLUDES NANOPARTICLES BASED ON A COPYLIMER OF METHYL VINYL ETER AND MALEIC ANHYDRIDE.
ES2389080T3 (en) 2004-06-11 2012-10-23 Riken Drug presenting a regulatory cell ligand contained in the liposome
GB0413868D0 (en) * 2004-06-21 2004-07-21 Chiron Srl Dimensional anlaysis of saccharide conjugates
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 (en) 2004-09-17 2005-11-09 四川大学 Preparation and application of CpG DNA molecule anti-infection and immunity prepn
EP1793863B1 (en) 2004-10-01 2017-04-12 Midatech Ltd. Nanoparticles comprising antigens and adjuvants capable of stimulating t helper cells
CA2583389A1 (en) 2004-10-07 2006-04-20 Emory University Multifunctional nanoparticles conjugates and their use
WO2007001448A2 (en) 2004-11-04 2007-01-04 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
EP1812056B1 (en) 2004-11-15 2013-08-07 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
WO2006060710A2 (en) * 2004-12-02 2006-06-08 Becton, Dickinson And Company Vaccine formulations for intradermal delivery comprising adjuvants and antigenic agents
JP2008523157A (en) 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 RNAi regulation of MLL-AF4 and methods of use thereof
US20060257359A1 (en) 2005-02-28 2006-11-16 Cedric Francois Modifying macrophage phenotype for treatment of disease
EP1866416A2 (en) 2005-03-22 2007-12-19 Medstar Health Inc. Delivery systems and methods for diagnosing and treating cardiovascular diseases
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
KR101418369B1 (en) 2005-05-04 2014-07-24 녹손 파르마 아게 Novel use of spiegelmers
AU2006243960A1 (en) 2005-05-10 2006-11-16 Emory University Strategies for delivery of active agents using micelles and particles
WO2007003054A1 (en) 2005-07-06 2007-01-11 Shoichet Molly S Method of biomolecule immobilization on polymers using click-type chemistry
CA2618807C (en) 2005-08-12 2015-01-06 University Health Network Methods and devices for lymphatic targeting
TWI404537B (en) 2005-08-19 2013-08-11 Array Biopharma Inc 8-substituted benzoazepines as toll-like receptor modulators
TWI382019B (en) 2005-08-19 2013-01-11 Array Biopharma Inc Aminodiazepines as toll-like receptor modulators
US20090324551A1 (en) 2005-08-22 2009-12-31 The Regents Of The University Of California Office Of Technology Transfer Tlr agonists
WO2007028341A1 (en) 2005-09-09 2007-03-15 Beijing Diacrid Medical Technology Co., Ltd. Nano anticancer micelles of vinca alkaloids entrapped in polyethylene glycolylated phospholipids
EP1963308A4 (en) 2005-11-28 2010-12-15 Nabi Biopharmaceuticals Method for making nicotine hapten
WO2008051245A2 (en) 2005-12-02 2008-05-02 Novartis Ag Nanoparticles for use in immunogenic compositions
AU2006325225B2 (en) 2005-12-14 2013-07-04 Cytos Biotechnology Ag Immunostimulatory nucleic acid packaged particles for the treatment of hypersensitivity
WO2007070682A2 (en) 2005-12-15 2007-06-21 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
BRPI0706949A2 (en) 2006-01-23 2011-04-12 Yissum Res Dev Co microspheres, method of preparing microspheres comprising a plurality of nanocapsules accommodated in a gel-forming polymer, pharmaceutical composition, method for increasing the bioavailability of a lipophilic agent in the body of a human subject, and method of treating an individual for a pathological condition. requiring an effective blood level of an active agent
CA2676601A1 (en) 2006-01-31 2007-08-09 Medivas, Llc Vaccine delivery compositions and methods of use
EP2520287A1 (en) 2006-02-10 2012-11-07 Biocompatibles UK Limited Loading of hydrophobic drugs into hydrophilic polymer delivery systems
US8021689B2 (en) * 2006-02-21 2011-09-20 Ecole Polytechnique Federale de Lausanne (“EPFL”) Nanoparticles for immunotherapy
WO2007100699A2 (en) 2006-02-24 2007-09-07 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
WO2008105773A2 (en) 2006-03-31 2008-09-04 Massachusetts Institute Of Technology System for targeted delivery of therapeutic agents
CA2649149A1 (en) 2006-04-11 2007-10-25 Koko Kosmetikvertrieb Gmbh & Co. Kg Nanoparticle 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
US20110052697A1 (en) 2006-05-17 2011-03-03 Gwangju Institute Of Science & Technology Aptamer-Directed Drug Delivery
RU2476595C2 (en) 2006-06-12 2013-02-27 Цитос Биотехнологи Аг Methods of packaging of oligonucleotides into virus-like particles of rna-containing bacteriophages
US20080014281A1 (en) 2006-06-16 2008-01-17 Florida Atlantic University Chitin Micro-Particles As An Adjuvant
WO2007149802A2 (en) 2006-06-19 2007-12-27 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
EP2037888A2 (en) 2006-06-26 2009-03-25 Mutual Pharmaceutical Company, Inc. Active agent formulations, methods of making, and methods of use
WO2008019142A2 (en) 2006-08-04 2008-02-14 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
AU2007323007A1 (en) 2006-08-11 2008-05-29 Panacea Biotec Limited Particles for delivery of active ingredients, process of making and compositions thereof
EP2066344B2 (en) * 2006-09-07 2016-06-29 GlaxoSmithKline Biologicals S.A. Inactivated Poliovirus combination vaccine
CA2665090A1 (en) 2006-09-22 2008-03-27 Dana-Farber Cancer Institute, Inc. Methods for treating mica-related disorders
JP2010505883A (en) 2006-10-12 2010-02-25 ザ ユニバーシティー オブ クイーンズランド 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
EA200900784A1 (en) * 2006-12-06 2009-12-30 Новартис Аг VACCINES INCLUDING ANTIGENS FROM FOUR STRAINS OF THE INFLUENZA VIRUS
AU2007333225B2 (en) 2006-12-08 2014-06-12 Massachusetts Institute Of Technology Delivery of nanoparticles and/or agents to cells
EP1932516A1 (en) 2006-12-11 2008-06-18 Universiteit Utrecht Holding B.V. Anti-inflammatory compounds containing compositions for treatment of cancer
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
JP5389668B2 (en) 2007-01-31 2014-01-15 チョンシー ユー Positively charged water-soluble prodrugs of 1H-imidazo [4,5-c] quinolin-4-amine and related compounds with very high skin permeability
PL2125007T3 (en) 2007-02-07 2014-07-31 Univ California Conjugates of synthetic tlr agonists and uses therefor
WO2008098165A2 (en) 2007-02-09 2008-08-14 Massachusetts Institute Of Technology Oscillating cell culture bioreactor
US8889117B2 (en) 2007-02-15 2014-11-18 Yale University Modular nanoparticles for adaptable vaccines
AU2008222678B2 (en) 2007-03-07 2013-01-17 The General Hospital Corporation 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 (en) 2007-03-30 2015-07-30 Particle Sciences, Inc. Formulations in the form of particles and uses thereof
EP2144600A4 (en) 2007-04-04 2011-03-16 Massachusetts Inst Technology Poly (amino acid) targeting moieties
WO2008124634A1 (en) 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Polymer-encapsulated reverse micelles
EP1982729A1 (en) 2007-04-20 2008-10-22 Cytos Biotechnology AG Vaccination Regimen for B-Cell Vaccines
US20080294089A1 (en) * 2007-06-06 2008-11-27 Biovaluation & Analysis, Inc. Dendritic Polymers for Use in Acoustically Mediated Intracellular Drug Delivery in vivo
US20090047318A1 (en) 2007-08-16 2009-02-19 Abbott Cardiovascular Systems Inc. Nanoparticle-coated medical devices and formulations for treating vascular disease
US8394914B2 (en) 2007-08-24 2013-03-12 Board Of Trustees Of Michigan State University Functional polyglycolide nanoparticles derived from unimolecular micelles
WO2009027971A2 (en) 2007-08-27 2009-03-05 H2Q Water Industries Ltd. Antimicrobial polymers
US20090130210A1 (en) 2007-09-11 2009-05-21 Raheja Praveen Pharmaceutical compositions of sirolimus
EP2197457A4 (en) * 2007-09-26 2010-10-27 Aparna Biosciences Therapeutic and vaccine polyelectrolyte nanoparticle compositions
WO2009069448A1 (en) 2007-11-28 2009-06-04 Toray Industries, Inc. Adjuvant for japanese b encephalitis vaccine, and japanese b encephalitis vaccine
WO2009078754A1 (en) 2007-12-19 2009-06-25 Ardenia Investments, Ltd. Drug delivery system for administration of poorly water soluble pharmaceutically active substances
CN101932594A (en) * 2008-02-01 2010-12-29 阿尔法-O肽股份公司 Self-assembling peptide nanoparticles useful as vaccines
WO2009111588A1 (en) 2008-03-04 2009-09-11 Liquidia Technologies, Inc. Immunomodulator particles and methods of treating
EP2268289A4 (en) 2008-04-01 2014-07-02 Innate Therapeutics Ltd Anti-infective agents and uses thereof
US20090297621A1 (en) 2008-06-03 2009-12-03 Abbott Cardiovascular Systems Inc. Microparticles For The Treatment Of Disease
CA2728176C (en) 2008-06-16 2017-07-04 Bind Biosciences, Inc. Drug loaded polymeric nanoparticles and methods of making and using same
US8613951B2 (en) 2008-06-16 2013-12-24 Bind Therapeutics, Inc. Therapeutic polymeric nanoparticles with mTor inhibitors and methods of making and using same
WO2009158687A1 (en) 2008-06-26 2009-12-30 Anterios, Inc. Dermal delivery
US20110104293A1 (en) 2008-07-01 2011-05-05 Bali Pulendran Synergistic induction of humoral and cellular immunity by combinatorial activation of toll-like receptors
JP5667566B2 (en) 2008-08-06 2015-02-12 ノバルティス アーゲー Microparticles for use in immunogenic compositions
JP2011530562A (en) 2008-08-11 2011-12-22 グラクソスミスクライン エルエルシー Purine derivatives for the treatment of allergic, inflammatory and infectious diseases
EP2326646B1 (en) 2008-08-11 2013-07-31 GlaxoSmithKline LLC Purine derivatives for use in the treatment of allergic, inflammatory and infectious diseases
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
EP2172193A1 (en) 2008-10-02 2010-04-07 Capsulution Nanoscience AG Improved nanoparticulate compositions of poorly soluble compounds
WO2010037402A1 (en) 2008-10-02 2010-04-08 Dako Denmark A/S Molecular vaccines for infectious disease
US8343497B2 (en) 2008-10-12 2013-01-01 The Brigham And Women's Hospital, Inc. Targeting of antigen presenting cells with immunonanotherapeutics
US8277812B2 (en) 2008-10-12 2012-10-02 Massachusetts Institute Of Technology Immunonanotherapeutics that provide IgG humoral response without T-cell antigen
US8591905B2 (en) * 2008-10-12 2013-11-26 The Brigham And Women's Hospital, Inc. Nicotine immunonanotherapeutics
US8343498B2 (en) * 2008-10-12 2013-01-01 Massachusetts Institute Of Technology Adjuvant incorporation in immunonanotherapeutics
US20100098770A1 (en) 2008-10-16 2010-04-22 Manikandan Ramalingam Sirolimus pharmaceutical formulations
ES2705101T3 (en) 2008-11-06 2019-03-21 Ventirx Pharmaceuticals Inc Methods of synthesis of benzazepine derivatives
CN101822838B (en) * 2009-03-05 2012-06-27 无锡纳奥生物医药有限公司 Nano-medicament carrier material for target recognition of tumor cell as well as preparation and application thereof
US20100233231A1 (en) 2009-03-10 2010-09-16 Roger Labrecque Use of cryogenic processing to obtain a substantially-thickened formulation
RU2600798C2 (en) 2009-04-01 2016-10-27 Юниверсити Оф Майами Vaccine compositions and methods of use thereof
EA201101530A1 (en) 2009-04-21 2012-03-30 Селекта Байосайенсиз, Инк. IMMUNONANOTHERAPY, PROVIDING TH1-DISPERSED RESPONSE
GB0908129D0 (en) 2009-05-12 2009-06-24 Innovata Ltd Composition
AU2010254551B2 (en) 2009-05-27 2016-10-20 Selecta Biosciences, Inc. Immunomodulatory agent-polymeric compounds
CN102802624A (en) 2009-06-19 2012-11-28 太阳医药高级研发有限公司 Nanodispersion Of A Drug And Process For Its Preparation
EP3058953A1 (en) 2009-07-07 2016-08-24 The Research Foundation Of State University Of New York Lipidic compositions for induction of immune tolerance
CN107715118A (en) 2009-08-26 2018-02-23 西莱克塔生物科技公司 The composition of inducing T cell auxiliary
WO2011066414A1 (en) * 2009-11-25 2011-06-03 Cytometix, Inc. Arachidonic acid analogs and methods for analgesic treatment using same
JP5965844B2 (en) 2009-12-15 2016-08-10 バインド セラピューティックス インコーポレイテッド Therapeutic polymer nanoparticle compositions having high glass transition temperature or high molecular weight copolymers
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
NO2575876T3 (en) 2010-05-26 2018-05-05
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
MX2013002173A (en) 2010-08-23 2013-05-06 Selecta Biosciences Inc Targeted multi-epitope dosage forms for induction of an immune response to antigens.
EP2640190A4 (en) 2010-11-05 2016-05-11 Selecta Biosciences Inc Modified nicotinic compounds and related methods
US20120171229A1 (en) 2010-12-30 2012-07-05 Selecta Biosciences, Inc. Synthetic nanocarriers with reactive groups that release biologically active agents
BR112013024655A2 (en) 2011-03-25 2016-12-20 Selecta Biosciences Inc synthetic nanotransporters for osmotic mediated release
KR20230006042A (en) 2011-04-29 2023-01-10 셀렉타 바이오사이언시즈, 인크. Tolerogenic synthetic nanocarriers to reduce antibody responses
CA2843274A1 (en) 2011-07-29 2013-02-07 Selecta Biosciences, Inc. Synthetic nanocarriers that generate humoral and cytotoxic t lymphocyte (ctl) immune responses
US20130058902A1 (en) 2011-09-06 2013-03-07 Selecta Biosciences, Inc. Dendritic cell subsets for generating induced tolerogenic dendritic cells and related compositions and methods
CN110841067A (en) 2013-05-03 2020-02-28 西莱克塔生物科技公司 Tolerogenic synthetic nanocarriers and therapeutic macromolecules for reduced or enhanced pharmacodynamic effects
EP3003306B1 (en) 2013-06-04 2020-08-26 Selecta Biosciences, Inc. Repeated administration of non-immunosupressive antigen specific immunotherapeutics
US20160220501A1 (en) 2015-02-03 2016-08-04 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers to reduce immune responses to therapeutic proteins
US20150359865A1 (en) 2014-06-17 2015-12-17 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for t-cell-mediated autoimmune disease
AU2015279738A1 (en) 2014-06-25 2016-12-22 Selecta Biosciences, Inc. Methods and compositions for treatment with synthetic nanocarriers and immune checkpoint inhibitors
CN107073091A (en) 2014-09-07 2017-08-18 西莱克塔生物科技公司 Method and composition for weakening the antiviral transfer vector immune response of exon skipping
US20160128987A1 (en) 2014-11-05 2016-05-12 Selecta Biosciences, Inc. Methods and compositions related to synthetic nanocarriers with rapamycin in a stable, super-saturated state

Patent Citations (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4946929A (en) 1983-03-22 1990-08-07 Massachusetts Institute Of Technology Bioerodible articles useful as implants and prostheses having predictable degradation rates
US4638045A (en) 1985-02-19 1987-01-20 Massachusetts Institute Of Technology Non-peptide polyamino acid bioerodible polymers
US4806621A (en) 1986-01-21 1989-02-21 Massachusetts Institute Of Technology Biocompatible, bioerodible, hydrophobic, implantable polyimino carbonate article
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
US5770417A (en) 1986-11-20 1998-06-23 Massachusetts Institute Of Technology Children's Medical Center Corporation Three-dimensional fibrous scaffold containing attached cells for producing vascularized tissue in vivo
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
US5010167A (en) 1989-03-31 1991-04-23 Massachusetts Institute Of Technology Poly(amide-and imide-co-anhydride) for biological application
US7147862B1 (en) 1992-06-25 2006-12-12 Smithkline Beecham Biologicals (S.A.) Vaccine composition containing adjuvants
US5750110A (en) 1992-06-25 1998-05-12 Smithkline Beecham Biologicals, S.A Vaccine composition containing adjuvants
US5399665A (en) 1992-11-05 1995-03-21 Massachusetts Institute Of Technology Biodegradable polymers for cell transplantation
US5512600A (en) 1993-01-15 1996-04-30 Massachusetts Institute Of Technology Preparation of bonded fiber structures for cell implantation
US5696175A (en) 1993-01-15 1997-12-09 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
US5578325A (en) 1993-07-23 1996-11-26 Massachusetts Institute Of Technology Nanoparticles and microparticles of non-linear hydrophilic-hydrophobic multiblock copolymers
US5543158A (en) 1993-07-23 1996-08-06 Massachusetts Institute Of Technology Biodegradable injectable nanoparticles
US7202351B1 (en) 1993-09-14 2007-04-10 Pharmexa Inc. Alteration of immune response using pan DR-binding peptides
US5663153A (en) 1994-03-25 1997-09-02 Isis Pharmaceuticals, Inc. Immune stimulation by phosphorothioate oligonucleotide analogs
US6194388B1 (en) 1994-07-15 2001-02-27 The University Of Iowa Research Foundation Immunomodulatory oligonucleotides
US6207646B1 (en) 1994-07-15 2001-03-27 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
US6123727A (en) 1995-05-01 2000-09-26 Massachusetts Institute Of Technology Tissue engineered tendons and ligaments
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
US6696076B2 (en) 1996-10-25 2004-02-24 3M Innovative Properties Company Immune response modifier compounds for treatment of TH2 mediated and related diseases
US6585980B1 (en) 1997-04-11 2003-07-01 The University Of Toronto Flagellin gene, FlaC of Campylobacter
US5837752A (en) 1997-07-17 1998-11-17 Massachusetts Institute Of Technology Semi-interpenetrating polymer networks
US6329381B1 (en) 1997-11-28 2001-12-11 Sumitomo Pharmaceuticals Company, Limited Heterocyclic compounds
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
US7357936B1 (en) 1998-10-16 2008-04-15 Smithkline Beecham Biologicals, Sa Adjuvant systems and vaccines
US6544518B1 (en) 1999-04-19 2003-04-08 Smithkline Beecham Biologicals S.A. Vaccines
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
US6632671B2 (en) 2000-02-28 2003-10-14 Genesegues, Inc. Nanoparticle encapsulation system and method
US7250403B2 (en) 2000-03-10 2007-07-31 Dynavax Technologies Corporation Biodegradable immunomodulatory formulations and methods for use thereof
US20060222652A1 (en) 2000-05-05 2006-10-05 Cytos Biotechnology Ag Molecular antigen array
US7192725B2 (en) 2000-05-19 2007-03-20 University Of Toronto Flagellin gene, flaC of Campylobacter
US7262286B2 (en) 2000-09-26 2007-08-28 Idera Pharmaceuticals, Inc. Modulation of immunostimulatory activity of immunostimulatory oligonucleotide analogs by positional chemical changes
US20080145441A1 (en) 2000-10-16 2008-06-19 Midatech Limited Nanoparticles
WO2002032450A2 (en) 2000-10-18 2002-04-25 Glaxosmithkline Biologicals S.A. Vaccines
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
WO2003086280A2 (en) 2002-04-04 2003-10-23 Coley Pharmaceutical Gmbh Immunostimulatory g,u-containing oligoribonucleotides
WO2004053056A2 (en) 2002-09-24 2004-06-24 University Of Kentucky Research Foundation Nanoparticle-based vaccine delivery system containing adjuvant
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
US20060251677A1 (en) 2003-03-26 2006-11-09 Cytos Biotechnology Ag Packaging of immunostimulatory oligonucleotides into virus-like particles: method of preparation and use
WO2004098509A2 (en) 2003-04-30 2004-11-18 Chiron Corporation Compositions for inducing immune responses
US20090028910A1 (en) 2003-12-19 2009-01-29 University Of North Carolina At Chapel Hill Methods for Fabrication Isolated Micro-and Nano-Structures Using Soft or Imprint Lithography
WO2005097993A2 (en) 2004-02-19 2005-10-20 Coley Pharmaceutical Group, Inc. Immunostimulatory viral rna oligonucleotides
US20060002852A1 (en) 2004-07-01 2006-01-05 Yale University Targeted and high density drug loaded polymeric materials
US7566703B2 (en) 2004-10-20 2009-07-28 Coley Pharmaceutical Group, Inc. Semi-soft C-class immunostimulatory oligonucleotides
US20060241076A1 (en) 2005-04-26 2006-10-26 Coley Pharmaceutical Gmbh Modified oligoribonucleotide analogs with enhanced immunostimulatory activity
WO2007062107A2 (en) 2005-11-25 2007-05-31 Coley Pharmaceutical Gmbh Immunostimulatory oligoribonucleotides
WO2008033432A2 (en) 2006-09-12 2008-03-20 Coley Pharmaceutical Group, Inc. Immune modulation by chemically modified ribonucleosides and oligoribonucleotides
US20090226525A1 (en) 2007-04-09 2009-09-10 Chimeros Inc. Self-assembling nanoparticle drug delivery system
WO2008127532A1 (en) 2007-04-12 2008-10-23 Emory University Novel strategies for delivery of active agents using micelles and particles
WO2009051837A2 (en) 2007-10-12 2009-04-23 Massachusetts Institute Of Technology Vaccine nanotechnology
WO2009076158A1 (en) 2007-12-07 2009-06-18 Novartis Ag Compositions for inducing immune responses
WO2009106999A2 (en) 2008-02-28 2009-09-03 Deutsches Krebsforschungszentrum, Stiftung Des Öffentlichen Rechts Hollow nanoparticles and uses thereof
WO2010018132A1 (en) 2008-08-11 2010-02-18 Smithkline Beecham Corporation Compounds
US20100075995A1 (en) 2008-08-11 2010-03-25 Smithkline Beecham Corporation Compounds
WO2010047839A1 (en) 2008-10-25 2010-04-29 Aura Biosciences Modified plant virus particles and uses therefor

Non-Patent Citations (46)

* Cited by examiner, † Cited by third party
Title
"Concise Encyclopedia of Polymer Science and Polymeric Amines and Ammonium Salts", 1980, PERGAMON PRESS
"Handbook of Industrial Mixing: Science and Practice", 2004, JOHN WILEY & SONS, INC.
"Microcapsules and Nanoparticles in Medicine and Pharmacy", 1992, CRC PRESS
"Pharmaceutics: The Science of Dosage Form Design", 2001, CHURCHILL LIVINGSTONE
ALLCOCK ET AL.: "Contemporary Polymer Chemistry", 1981, PRENTICE-HALL
BARRERA ET AL., J. AM. CHEM. SOC., vol. 115, 1993, pages 11010
BOUSSIF ET AL., PROC. NATL. ACAD. SCI., USA, vol. 92, 1995, pages 7297
C. ASTETE ET AL.: "Synthesis and characterization of PLGA nanoparticles", J. BIOMATER. SCI. POLYMER EDN, vol. 17, no. 3, 2006, pages 247 - 289, XP009134610
C. REIS ET AL.: "Nanoencapsulation I. Methods for preparation of drug-loaded polymeric nanoparticles", NANOMEDICINE, vol. 2, 2006, pages 8 - 21
CHU ET AL.: "CpG oligodeoxynucleotides act as adjuvants that switch on T helper 1 (Thl) immunity", J. EXP. MED., vol. 186, 1997, pages 1623 - 1631, XP002910130, DOI: doi:10.1084/jem.186.10.1623
DAVIS ET AL.: "CpG DNA is a potent enhancer of specific immunity in mice immunized with recombinant hepatitis B surface antigen", J. IMMUNOL., vol. 160, 1998, pages 870 - 876
DEMING ET AL., NATURE, vol. 390, 1997, pages 386
F. HEIL ET AL.: "Species-Specific Recognition of Single-Stranded RNA via Toll-like Receptor 7 and 8", SCIENCE, vol. 303, no. 5663, 2004, pages 1526 - 1529, XP002371479
HAENSLER ET AL., BIOCONJUGATE CHEM., vol. 4, 1993, pages 372
HERMANSON G T: "Bioconjugate Techniques", 2008, ACADEMIC PRESS, INC.
K. AVGOUSTAKIS: "Pegylated Poly(Lactide) and Poly(Lactide-Co-Glycolide) Nanoparticles: Preparation, Properties and Possible Applications in Drug Delivery", CURRENT DRUG DELIVERY, vol. 1, 2004, pages 321 - 333, XP009134627
KABANOV ET AL., BIOCONJUGATE CHEM., vol. 6, 1995, pages 7
KRIEG ET AL.: "CpG motifs in bacterial DNA trigger direct B cell activation", NATURE, vol. 374, 1995, pages 546 - 549, XP002910391, DOI: doi:10.1038/374546a0
KUKOWSKA-LATALLO ET AL., PROC. NATL. ACAD. SCI., USA, vol. 93, 1996, pages 4897
KWON ET AL., MACROMOLECULES, vol. 22, 1989, pages 3250
LANGER, ACC. CHEM. RES., vol. 33, 2000, pages 94
LANGER, J. CONTROL. RELEASE, vol. 62, 1999, pages 7
LIM ET AL., J. AM. CHEM. SOC., vol. 121, 1999, pages 5633
LIM ET AL., J. AM. CHEM. SOC., vol. 123, 2001, pages 2460
LIPFORD ET AL.: "Bacterial DNA as immune cell activator", TRENDS MICROBIOL., vol. 6, 1998, pages 496 - 500, XP000952562, DOI: doi:10.1016/S0966-842X(98)01408-5
LIPFORD ET AL.: "CpG-containing synthetic oligonucleotides promote B and cytotoxic T cell responses to protein antigen: a new class of vaccine adjuvants", EUR. J. IMMUNOL., vol. 27, 1997, pages 2340 - 2344, XP002123073, DOI: doi:10.1002/eji.1830270931
MATHIOWITZ ET AL., J. APPL. POLYMER SCI., vol. 35, 1988, pages 755
MATHIOWITZ ET AL., J. CONTROL. RELEASE, vol. 5, 1987, pages 13
MATHIOWITZ ET AL., REACTIVE POLYMERS, vol. 6, 1987, pages 275
MELDAL ET AL., CHEM. REV., vol. 108, no. 8, 2008, pages 2952 - 3015
MURRAY ET AL., ANN. REV. MAT. SCI., vol. 30, 2000, pages 545
ODIAN: "Principles of Polymerization", 2004, JOHN WILEY & SONS
P. PAOLICELLI ET AL.: "Surface-modified PLGA-based Nanoparticles that can Efficiently Associate and Deliver Virus-like Particles", NANOMEDICINE, vol. 5, no. 6, 2010, pages 843 - 853
PAPISOV, ACS SYMPOSIUM SERIES, vol. 786, 2001, pages 301
PELLEGRINO ET AL., SMALL, vol. 1, 2005, pages 48
PUTNAM ET AL., MACROMOLECULES, vol. 32, 1999, pages 3658
ROMAN ET AL.: "Immunostimulatory DNA sequences function as T helper-1-promoting adjuvants", NAT. MED., vol. 3, 1997, pages 849 - 854, XP002188113, DOI: doi:10.1038/nm0897-849
See also references of EP2575886A4
SHARPLESS ET AL., ANGEW. CHEM. INT. ED., vol. 41, no. 14, 2002, pages 2596
TANG ET AL., BIOCONJUGATE CHEM., vol. 7, 1996, pages 703
TRINDADE ET AL., CHEM. MAT., vol. 13, 2001, pages 3843
UHRICH ET AL.: "99", CHEM. REV., 1999, pages 3181
V. CERUNDOLO ET AL.: "Harnessing invariant NKT cells in vaccination strategies", NATURE REV IMMUN, vol. 9, 2009, pages 28 - 38
WANG ET AL., J. AM. CHEM. SOC., vol. 123, 2001, pages 9480
ZAUNER ET AL., ADV. DRUG DEL. REV., vol. 30, 1998, pages 97
ZHOU ET AL., MACROMOLECULES, vol. 23, 1990, pages 3399

Cited By (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8932583B2 (en) 2005-12-13 2015-01-13 President And Fellows Of Harvard College Scaffolds for cell transplantation
US11096997B2 (en) 2005-12-13 2021-08-24 President And Fellows Of Harvard College Scaffolds for cell transplantation
US9132210B2 (en) 2005-12-13 2015-09-15 President And Fellows Of Harvard College Scaffolds for cell transplantation
US9446107B2 (en) 2005-12-13 2016-09-20 President And Fellows Of Harvard College Scaffolds for cell transplantation
US10149897B2 (en) 2005-12-13 2018-12-11 President And Fellows Of Harvard College Scaffolds for cell transplantation
US10137184B2 (en) 2005-12-13 2018-11-27 President And Fellows Of Harvard College Scaffolds for cell transplantation
US10695468B2 (en) 2007-06-21 2020-06-30 President And Fellows Of Harvard College Scaffolds for cell collection or elimination
US9770535B2 (en) 2007-06-21 2017-09-26 President And Fellows Of Harvard College Scaffolds for cell collection or elimination
US9821045B2 (en) 2008-02-13 2017-11-21 President And Fellows Of Harvard College Controlled delivery of TLR3 agonists in structural polymeric devices
US9370558B2 (en) 2008-02-13 2016-06-21 President And Fellows Of Harvard College Controlled delivery of TLR agonists in structural polymeric devices
US10258677B2 (en) 2008-02-13 2019-04-16 President And Fellows Of Harvard College Continuous cell programming devices
US10328133B2 (en) 2008-02-13 2019-06-25 President And Fellows Of Harvard College Continuous cell programming devices
US10568949B2 (en) 2008-02-13 2020-02-25 President And Fellows Of Harvard College Method of eliciting an anti-tumor immune response with controlled delivery of TLR agonists in porous polymerlc devices
US9539309B2 (en) 2008-05-30 2017-01-10 President And Fellows Of Harvard College Controlled release of growth factors and signaling molecules for promoting angiogenesis
US9012399B2 (en) 2008-05-30 2015-04-21 President And Fellows Of Harvard College Controlled release of growth factors and signaling molecules for promoting angiogenesis
US9297005B2 (en) 2009-04-13 2016-03-29 President And Fellows Of Harvard College Harnessing cell dynamics to engineer materials
US9381235B2 (en) 2009-07-31 2016-07-05 President And Fellows Of Harvard College Programming of cells for tolerogenic therapies
US8728456B2 (en) 2009-07-31 2014-05-20 President And Fellows Of Harvard College Programming of cells for tolerogenic therapies
US10080789B2 (en) 2009-07-31 2018-09-25 President And Fellows Of Harvard College Programming of cells for tolerogenic therapies
US9610328B2 (en) 2010-03-05 2017-04-04 President And Fellows Of Harvard College Enhancement of skeletal muscle stem cell engraftment by dual delivery of VEGF and IGF-1
US9693954B2 (en) 2010-06-25 2017-07-04 President And Fellows Of Harvard College Co-delivery of stimulatory and inhibitory factors to create temporally stable and spatially restricted zones
US11202759B2 (en) 2010-10-06 2021-12-21 President And Fellows Of Harvard College Injectable, pore-forming hydrogels for materials-based cell therapies
EP2640190A4 (en) * 2010-11-05 2016-05-11 Selecta Biosciences Inc Modified nicotinic compounds and related methods
US9994443B2 (en) 2010-11-05 2018-06-12 Selecta Biosciences, Inc. Modified nicotinic compounds and related methods
US9603894B2 (en) 2010-11-08 2017-03-28 President And Fellows Of Harvard College Materials presenting notch signaling molecules to control cell behavior
US10647959B2 (en) 2011-04-27 2020-05-12 President And Fellows Of Harvard College Cell-friendly inverse opal hydrogels for cell encapsulation, drug and protein delivery, and functional nanoparticle encapsulation
US9675561B2 (en) 2011-04-28 2017-06-13 President And Fellows Of Harvard College Injectable cryogel vaccine devices and methods of use thereof
US10045947B2 (en) 2011-04-28 2018-08-14 President And Fellows Of Harvard College Injectable preformed macroscopic 3-dimensional scaffolds for minimally invasive administration
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
US10420835B2 (en) 2011-04-29 2019-09-24 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for antigen-specific deletion of T effector cells
US10441651B2 (en) 2011-04-29 2019-10-15 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for generating CD8+ regulatory T cells
US11779641B2 (en) 2011-04-29 2023-10-10 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for allergy therapy
US9486512B2 (en) 2011-06-03 2016-11-08 President And Fellows Of Harvard College In situ antigen-generating cancer vaccine
US10406216B2 (en) 2011-06-03 2019-09-10 President And Fellows Of Harvard College In situ antigen-generating cancer vaccine
EP4144378A1 (en) 2011-12-16 2023-03-08 ModernaTX, Inc. Modified nucleoside, nucleotide, and nucleic acid compositions
WO2013106852A1 (en) * 2012-01-13 2013-07-18 President And Fellows Of Harvard College Controlled delivery of tlr agonists in structural polymeric devices
WO2013151666A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics Modified polynucleotides for the production of biologics and proteins associated with human disease
WO2013151736A2 (en) 2012-04-02 2013-10-10 modeRNA Therapeutics In vivo production of proteins
US9937249B2 (en) 2012-04-16 2018-04-10 President And Fellows Of Harvard College Mesoporous silica compositions for modulating immune responses
US11278604B2 (en) 2012-04-16 2022-03-22 President And Fellows Of Harvard College Mesoporous silica compositions comprising inflammatory cytokines comprising inflammatory cytokines for modulating immune responses
KR101953374B1 (en) 2012-09-27 2019-02-28 고려대학교 산학협력단 Protein nanoparticle based multivalent vaccines
KR20140041134A (en) * 2012-09-27 2014-04-04 고려대학교 산학협력단 Protein nanoparticle based multivalent vaccines
EP4074834A1 (en) 2012-11-26 2022-10-19 ModernaTX, Inc. Terminally modified rna
CN103083663A (en) * 2013-02-04 2013-05-08 江苏省农业科学院 Immunity enhancing agent, inactivated vaccine, and preparation method thereof
CN103083663B (en) * 2013-02-04 2014-12-10 江苏省农业科学院 Immunity enhancing agent, inactivated vaccine, and preparation method thereof
WO2014142653A1 (en) * 2013-03-11 2014-09-18 Cristal Delivery B.V. Vaccination composition
WO2014152211A1 (en) 2013-03-14 2014-09-25 Moderna Therapeutics, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
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
US10357483B2 (en) 2013-05-03 2019-07-23 Selecta Biosciences, Inc. Methods comprising 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
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
US10668053B2 (en) 2013-05-03 2020-06-02 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers to reduce or prevent anaphylaxis in response to a non-allergenic antigen
US10335395B2 (en) 2013-05-03 2019-07-02 Selecta Biosciences, Inc. Methods of administering immunosuppressants having a specified pharmacodynamic effective life and therapeutic macromolecules for the induction of immune tolerance
JP2016520051A (en) * 2013-05-03 2016-07-11 セレクタ バイオサイエンシーズ インコーポレーテッドSelecta Biosciences,Inc. Topical combination administration of tolerogenic synthetic nanocarriers to reduce type I and type IV hypersensitivity
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
US10047072B2 (en) 2013-09-16 2018-08-14 Astrazeneca Ab Therapeutic polymeric nanoparticles and methods of making and using same
US10577351B2 (en) 2013-09-16 2020-03-03 Astrazeneca Ab Therapeutic polymeric nanoparticles and methods of making and using same
WO2015110957A2 (en) 2014-01-21 2015-07-30 De Beer Joel Hybridosomes, compositions comprising the same, processes for their production and uses thereof
EP3791863A1 (en) 2014-01-21 2021-03-17 Anjarium Biosciences AG Process for the production of hybridosomes
US11944706B2 (en) 2014-01-21 2024-04-02 Anjarium Biosciences Ag Hybridosomes, compositions comprising the same, processes for their production and uses thereof
US10682400B2 (en) 2014-04-30 2020-06-16 President And Fellows Of Harvard College Combination vaccine devices and methods of killing cancer cells
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
WO2016055812A1 (en) * 2014-10-10 2016-04-14 Isis Innovation Limited Polymer adjuvant
US11786457B2 (en) 2015-01-30 2023-10-17 President And Fellows Of Harvard College Peritumoral and intratumoral materials for cancer therapy
US11150242B2 (en) 2015-04-10 2021-10-19 President And Fellows Of Harvard College Immune cell trapping devices and methods for making and using the same
EP4349404A2 (en) 2015-10-22 2024-04-10 ModernaTX, Inc. Respiratory virus vaccines
EP4349405A2 (en) 2015-10-22 2024-04-10 ModernaTX, Inc. Respiratory virus vaccines
EP4011451A1 (en) 2015-10-22 2022-06-15 ModernaTX, Inc. Metapneumovirus mrna vaccines
US11752238B2 (en) 2016-02-06 2023-09-12 President And Fellows Of Harvard College Recapitulating the hematopoietic niche to reconstitute immunity
US11555177B2 (en) 2016-07-13 2023-01-17 President And Fellows Of Harvard College Antigen-presenting cell-mimetic scaffolds and methods for making and using the same
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
WO2022081764A1 (en) 2020-10-14 2022-04-21 RNAimmune, Inc. PAN-RAS mRNA CANCER VACCINES
WO2023161350A1 (en) 2022-02-24 2023-08-31 Io Biotech Aps Nucleotide delivery of cancer therapy

Also Published As

Publication number Publication date
JP2018065813A (en) 2018-04-26
IL222680A0 (en) 2012-12-31
JP2017014217A (en) 2017-01-19
EP2582393A4 (en) 2014-04-02
AU2017201143A1 (en) 2017-03-09
AU2011258171B2 (en) 2016-11-24
CA2798493A1 (en) 2011-12-01
CA2798739A1 (en) 2011-12-01
WO2011150249A1 (en) 2011-12-01
DK2575876T3 (en) 2018-03-12
MX2012013716A (en) 2013-01-28
CN103118700A (en) 2013-05-22
IL222680B (en) 2020-03-31
EP2575876A4 (en) 2014-03-26
ES2661978T3 (en) 2018-04-04
PT2575876T (en) 2018-03-26
JP6371058B2 (en) 2018-08-15
JP2013526617A (en) 2013-06-24
MX352324B (en) 2017-11-17
EA201291156A1 (en) 2013-04-30
CA2798994A1 (en) 2011-12-01
KR20130108984A (en) 2013-10-07
EA201291154A1 (en) 2013-04-30
US20110293723A1 (en) 2011-12-01
EP2575876B1 (en) 2017-12-06
IL222725A0 (en) 2012-12-31
IL260015A (en) 2018-07-31
IL269615A (en) 2019-11-28
EA201291158A1 (en) 2013-05-30
WO2011150264A2 (en) 2011-12-01
KR20180099900A (en) 2018-09-05
CN107080839A (en) 2017-08-22
IL222722B (en) 2018-12-31
JP6407208B2 (en) 2018-10-17
US9764031B2 (en) 2017-09-19
JP2013528181A (en) 2013-07-08
JP2017014216A (en) 2017-01-19
US20150328309A1 (en) 2015-11-19
BR112012029912A2 (en) 2016-11-16
BR112012029823A2 (en) 2020-09-01
EP2575886A1 (en) 2013-04-10
CN107029223A (en) 2017-08-11
KR20130108987A (en) 2013-10-07
PL2575876T3 (en) 2018-07-31
CN105194665A (en) 2015-12-30
EA030863B1 (en) 2018-10-31
JP6367554B2 (en) 2018-08-01
JP2020023492A (en) 2020-02-13
EA201291157A1 (en) 2013-04-30
AU2011258147B2 (en) 2016-11-17
IL222722A0 (en) 2012-12-31
JP2018052940A (en) 2018-04-05
AU2011258147A1 (en) 2012-11-01
MX2012013715A (en) 2013-01-28
KR20130108983A (en) 2013-10-07
CA2798323A1 (en) 2011-12-01
MX2012013713A (en) 2013-01-28
MX355036B (en) 2018-04-02
EP2582393A1 (en) 2013-04-24
CN102905728B (en) 2015-11-25
IL222725B (en) 2018-06-28
CN107096021A (en) 2017-08-29
US20120027806A1 (en) 2012-02-02
AU2017201145A1 (en) 2017-03-09
EP2575773A2 (en) 2013-04-10
CN102905728A (en) 2013-01-30
WO2011150258A1 (en) 2011-12-01
AU2017201080A1 (en) 2017-03-09
CN106177940A (en) 2016-12-07
US20110293700A1 (en) 2011-12-01
CN107029222A (en) 2017-08-11
EA030620B1 (en) 2018-09-28
MX2012013714A (en) 2013-01-28
CN102905729A (en) 2013-01-30
AU2011258156A1 (en) 2012-11-08
EP2575886A4 (en) 2015-02-25
US20110293701A1 (en) 2011-12-01
WO2011150264A3 (en) 2013-04-04
EP3388081A1 (en) 2018-10-17
EP2575773A4 (en) 2014-06-25
AU2011258165A1 (en) 2012-11-08
US9066978B2 (en) 2015-06-30
AU2017201082A1 (en) 2017-03-09
JP2018052937A (en) 2018-04-05
BR112012029917A2 (en) 2017-02-21
NO2575876T3 (en) 2018-05-05
EA030813B1 (en) 2018-10-31
IL222724A0 (en) 2012-12-31
AU2011258165B2 (en) 2016-11-17
EP2575876A1 (en) 2013-04-10
JP2013530157A (en) 2013-07-25
JP2013530158A (en) 2013-07-25
CN102917731A (en) 2013-02-06
EA201890942A1 (en) 2018-09-28
US20180043023A1 (en) 2018-02-15
EA023397B1 (en) 2016-05-31
KR20130108988A (en) 2013-10-07
JP2017008054A (en) 2017-01-12
JP6324067B2 (en) 2018-05-16
EA201500857A1 (en) 2016-06-30
JP2017008055A (en) 2017-01-12
JP6324068B2 (en) 2018-05-23
AU2011258171A1 (en) 2012-11-08
AU2011258156B2 (en) 2016-11-24

Similar Documents

Publication Publication Date Title
AU2011258147B2 (en) Nanocarrier compositions with uncoupled adjuvant
US20230139671A1 (en) Osmotic mediated release synthetic nanocarriers
US20130039954A1 (en) Control of antibody responses to synthetic nanocarriers

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201180025504.3

Country of ref document: CN

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

Ref document number: 11787434

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2011258147

Country of ref document: AU

Date of ref document: 20110526

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2798739

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2013512250

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20127030841

Country of ref document: KR

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: MX/A/2012/013713

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 201291156

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 10971/DELNP/2012

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2011787434

Country of ref document: EP

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112012029917

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112012029917

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20121123