US20160074531A1 - Methods and compositions for attenuating gene therapy anti-viral transfer vector immune responses - Google Patents

Methods and compositions for attenuating gene therapy anti-viral transfer vector immune responses Download PDF

Info

Publication number
US20160074531A1
US20160074531A1 US14/846,952 US201514846952A US2016074531A1 US 20160074531 A1 US20160074531 A1 US 20160074531A1 US 201514846952 A US201514846952 A US 201514846952A US 2016074531 A1 US2016074531 A1 US 2016074531A1
Authority
US
United States
Prior art keywords
transfer vector
viral transfer
antigen
subject
immunosuppressant
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US14/846,952
Other languages
English (en)
Inventor
Takashi Kei Kishimoto
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cartesian Therapeutics Inc
Original Assignee
Selecta Biosciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Selecta Biosciences Inc filed Critical Selecta Biosciences Inc
Priority to US14/846,952 priority Critical patent/US20160074531A1/en
Assigned to SELECTA BIOSCIENCES, INC. reassignment SELECTA BIOSCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KISHIMOTO, TAKASHI KEI
Publication of US20160074531A1 publication Critical patent/US20160074531A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/439Heterocyclic 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 the ring forming part of a bridged ring system, e.g. quinuclidine
    • 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
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/482Serine endopeptidases (3.4.21)
    • A61K38/4846Factor VII (3.4.21.21); Factor IX (3.4.21.22); Factor Xa (3.4.21.6); Factor XI (3.4.21.27); Factor XII (3.4.21.38)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/001Preparations to induce tolerance to non-self, e.g. prior to transplantation
    • 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
    • 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
    • A61K47/69Medicinal 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
    • A61K47/6923Medicinal 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 an inorganic particle, e.g. ceramic particles, silica particles, ferrite or synsorb
    • 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
    • A61K47/6935Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer the polymer being obtained otherwise than by reactions involving carbon to carbon unsaturated bonds, e.g. polyesters, polyamides or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • A61K47/6931Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
    • A61K47/6935Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer the polymer being obtained otherwise than by reactions involving carbon to carbon unsaturated bonds, e.g. polyesters, polyamides or polyglycerol
    • A61K47/6937Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer the polymer being obtained otherwise than by reactions involving carbon to carbon unsaturated bonds, e.g. polyesters, polyamides or polyglycerol the polymer being PLGA, PLA or polyglycolic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • 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
    • A61K9/5115Inorganic compounds
    • 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
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • 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
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • GPHYSICS
    • G06COMPUTING; CALCULATING OR COUNTING
    • G06QINFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR ADMINISTRATIVE, COMMERCIAL, FINANCIAL, MANAGERIAL OR SUPERVISORY PURPOSES; SYSTEMS OR METHODS SPECIALLY ADAPTED FOR ADMINISTRATIVE, COMMERCIAL, FINANCIAL, MANAGERIAL OR SUPERVISORY PURPOSES, NOT OTHERWISE PROVIDED FOR
    • G06Q99/00Subject matter not provided for in other groups of this subclass
    • 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
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/577Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 tolerising response
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/00041Use of virus, viral particle or viral elements as a vector
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10032Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10041Use of virus, viral particle or viral elements as a vector
    • C12N2710/10043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/10041Use of virus, viral particle or viral elements as a vector
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15032Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16041Use of virus, viral particle or viral elements as a vector
    • C12N2740/16043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14132Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
    • 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • 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
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21022Coagulation factor IXa (3.4.21.22)

Definitions

  • the invention relates to methods and compositions for administering viral transfer vectors and antigen-presenting cell targeted immunosuppressants.
  • the viral transfer vector comprises a gene therapy transgene that may have a therapeutic benefit for any one of the purposes provided herein in any one of the methods or compositions provided herein.
  • a method comprising establishing an anti-viral transfer vector attenuated response in a subject by concomitant administration of an antigen-presenting cell targeted immunosuppressant and viral transfer vector to the subject.
  • the subject does not have pre-existing immunity against the viral transfer vector.
  • the anti-viral transfer vector attenuated response is a T cell response against the viral transfer vector
  • the method further comprises administering the viral transfer vector to the subject without an antigen-presenting cell targeted immunosuppressant prior to the concomitant administration of the antigen-presenting cell targeted immunosuppressant and viral transfer vector.
  • the concomitant administration of the antigen-presenting cell targeted immunosuppressant and viral transfer vector is repeated, concomitant administration of the antigen-presenting cell targeted immunosuppressant and viral transfer vector.
  • a method comprising establishing an anti-viral transfer vector attenuated response in a subject by concomitant administration of an antigen-presenting cell targeted immunosuppressant and viral transfer vector to the subject, and administering to the subject one or more repeat doses of the viral transfer vector.
  • the anti-viral transfer vector attenuated response is a T cell response against the viral transfer vector
  • the method further comprises administering the viral transfer vector to the subject without an antigen-presenting cell targeted immunosuppressant prior to both the concomitant administration of the antigen-presenting cell targeted immunosuppressant and viral transfer vector and the one or more repeat doses of the viral transfer vector.
  • the method further comprises providing or obtaining an antigen-presenting cell targeted immunosuppressant alone or in combination with a viral transfer vector.
  • a method comprising attenuating an anti-viral transfer vector response, wherein the anti-viral transfer vector response is a T cell response, by first administering to a subject a viral transfer vector without an antigen-presenting cell targeted immunosuppressant, and subsequently concomitantly administering the viral transfer vector and an antigen-presenting cell targeted immunosuppressant to the subject.
  • the method further comprises administering to the subject one or more repeat doses of the viral transfer vector subsequent to the concomitant administration of the viral transfer vector and the antigen-presenting cell targeted immunosuppressant to the subject.
  • a method comprising determining a level of pre-existing immunity to a viral transfer vector in a subject prior to administration of the viral transfer vector to the subject, concomitantly administering to the subject an antigen-presenting cell targeted immunosuppressant and viral transfer vector, and administering to the subject a dose of the viral transfer vector.
  • the determining comprises measuring a level of anti-viral transfer vector antibodies in the subject prior to administration of the viral transfer vector to the subject. In another embodiment of any one of the methods provided, the determining comprises measuring a level of a T cell response against the viral transfer vector in the subject prior to administration of the viral transfer vector to the subject.
  • the method further comprises one or more repeat doses of the viral transfer vector.
  • the level of pre-existing immunity is to a viral antigen of the viral transfer vector. In one embodiment of any one of the methods provided, the level of pre-existing immunity is to an antigen of a protein transgene expression product of the viral transfer vector.
  • a method comprising escalating transgene expression of a viral transfer vector in a subject by repeatedly, concomitantly administering to the subject an antigen-presenting cell targeted immunosuppressant and viral transfer vector.
  • the method further comprises determining the frequency and dosing of the repeated, concomitant administration of the antigen-presenting cell targeted immunosuppressant and viral transfer vector that increase the transgene expression in a subject.
  • a method comprising repeatedly, concomitantly administering to a subject an antigen-presenting cell targeted immunosuppressant and viral transfer vector, and selecting one or more doses of the viral transfer vector to be less than the dose of the viral transfer vector that would be selected for the subject if the subject were expected to develop anti-viral transfer vector immune responses due to the repeated administration of the viral transfer vector.
  • a method comprising inducing an entity to purchase or obtain an antigen-presenting cell targeted immunosuppressant alone or in combination with a viral transfer vector by communicating to the entity that concomitant administration of the antigen-presenting cell targeted immunosuppressant and viral transfer vector results in an anti-viral transfer vector attenuated response in a subject.
  • a method comprising inducing an entity to purchase or obtain an antigen-presenting cell targeted immunosuppressant alone or in combination with a viral transfer vector by communicating to the entity that efficacious repeated viral transfer vector dosing is possible by concomitant administration of the antigen-presenting cell targeted immunosuppressant and viral transfer vector to a subject.
  • the communicating further includes instructions for practicing any one of the methods described herein or information describing the benefits of concomitant administration of a viral transfer vector with an antigen-presenting cell targeted immunosuppressant.
  • the method further comprises distributing an antigen-presenting cell targeted immunosuppressant or a viral transfer vector or both to an entity.
  • a method comprising determining the frequency and dosing of concomitant administration of an antigen-presenting cell targeted immunosuppressant and viral transfer vector in order to generate an anti-viral transfer vector attenuated response in a subject.
  • the method further comprises directing the concomitant administration of the antigen-presenting cell targeted immunosuppressant and viral transfer vector to a subject according to the determined frequency and dosing.
  • a method comprising determining the frequency and dosing of concomitant administration of an antigen-presenting cell targeted immunosuppressant and viral transfer vector in combination with one or more repeat doses of the viral transfer vector in order to generate an anti-viral transfer vector attenuated response in a subject.
  • the method further comprises directing both the concomitant administration of the antigen-presenting cell targeted immunosuppressant and viral transfer vector and administration of the one or more repeat doses of the viral transfer vector to a subject according to the determined frequency and dosing.
  • the method further comprises directing the administration of a dose of the viral transfer vector to the subject prior to both the concomitant administration of the antigen-presenting cell targeted immunosuppressant and viral transfer vector and administration of the one or more repeat doses of the viral transfer vector to the subject.
  • the subject is one to which the viral transfer vector has not been previously administered.
  • the subject is one to which the viral transfer vector has been previously administered no more than once.
  • the amount of the viral transfer vector in the repeat dose(s) is at least equal to the amount of the viral transfer vector in a prior dose. In one embodiment of any one of the methods provided, the amount of the viral transfer vector in the repeat dose(s) is less than the amount of the viral transfer vector in a prior dose.
  • the antigen-presenting cell targeted immunosuppressant is also administered to the subject concomitantly with the one or more repeat doses of the viral transfer vector. In one embodiment of any one of the methods provided, the antigen-presenting cell targeted immunosuppressant is not also administered to the subject concomitantly with at least one of the one or more repeat doses of the viral transfer vector.
  • the subject does not have pre-existing immunity against the viral transfer vector.
  • the concomitant administration is simultaneous administration.
  • the method further comprises determining a level of pre-existing immunity to the viral transfer vector in the subject.
  • the viral transfer vector is a retroviral transfer vector, an adenoviral transfer vector, a lentiviral transfer vector or an adeno-associated viral transfer vector.
  • the viral transfer vector is an adenoviral transfer vector
  • the adenoviral transfer vector is a subgroup A, subgroup B, subgroup C, subgroup D, subgroup E, or subgroup F adenoviral transfer vector.
  • the viral transfer vector is a lentiviral transfer vector
  • the lentiviral transfer vector is an HIV, SIV, FIV, EIAV or ovine lentiviral vector.
  • the viral transfer vector is an adeno-associated viral transfer vector
  • the adeno-associated viral transfer vector is an AAV1, AAV2, AAV5, AAV6, AAV6.2, AAV7, AAV8, AAV9, AAV10 or AAV11 adeno-associated viral transfer vector.
  • the viral transfer vector is a chimeric viral transfer vector. In one embodiment of any one of the methods provided herein, the chimeric viral transfer vector is an AAV-adenoviral transfer vector.
  • the gene therapy transgene encodes a therapeutic protein.
  • the therapeutic protein is a/an infusible or injectable therapeutic protein, enzyme, enzyme cofactor, hormone, blood or blood coagulation factor, cytokine, interferon or growth factor.
  • the therapeutic protein is a protein associated with a disorder of lipid and sphingolipid degradation, disorder of mucopolysaccharide degradation, disorder of glycoprotein degradation, lysosomal storage disorder or a leukodystrophy.
  • the antigen-presenting cell targeted immunosuppressant comprises an erythrocyte-binding therapeutic.
  • the erythrocyte-binding therapeutic comprises ERY1, ERY19, ERY59, ERY64, ERY123, ERY141 and ERY162.
  • the erythrocyte-binding therapeutic further comprises a viral transfer vector antigen.
  • the viral transfer vector antigen is a viral antigen.
  • the antigen-presenting cell targeted immunosuppressant comprises a negatively-charged particle.
  • the negatively-charged particle is a polystyrene, PLGA, or diamond particle.
  • the zeta potential of the particle is negative.
  • the zeta potential of the particle is less than ⁇ 50 mV. In one embodiment of any one of the methods provided herein, the zeta potential of the particle is less than ⁇ 100 mV.
  • the antigen-presenting cell targeted immunosuppressant comprises an apoptotic-body mimic and one or more viral transfer vector antigens.
  • the apoptotic-body mimic is a particle that comprises the one or more viral transfer vector antigens.
  • the one or more viral transfer vector antigens comprise one or more viral antigens.
  • the particle may also comprise an apoptotic signaling molecule.
  • the particle comprises a polyglycolic acid polymer (PGA), polylactic acid polymer (PLA), polysebacic acid polymer (PSA), poly(lactic-co-glycolic) acid copolymer (PLGA), poly(lactic-co-sebacic) acid copolymer (PLSA), poly(glycolic-co-sebacic) acid copolymer (PGSA), polylactide co-glycolide (PLG), or polyethylene glycol (PEG).
  • PGA polyglycolic acid polymer
  • PLA polylactic acid polymer
  • PSA polysebacic acid polymer
  • PLGA poly(lactic-co-glycolic) acid copolymer
  • PLA poly(lactic-co-sebacic) acid copolymer
  • PGSA poly(glycolic-co-sebacic) acid copolymer
  • PEG polyethylene glycol
  • the average diameter of the particle is between 0.1 and 5 ⁇ m, between 0.1 and 4 ⁇ m, between 0.1 and 3 ⁇ m, between 0.1 and 2 ⁇ m, between 0.1 and 1 ⁇ m or between 0.1 and 500 nm.
  • the antigen-presenting cell targeted immunosuppressant comprises synthetic nanocarriers comprising an immunosuppressant.
  • the synthetic nanocarriers further comprise a viral transfer vector antigen.
  • the viral transfer vector antigen is a viral antigen.
  • the immunosuppressant and/or the antigen, if present, are/is encapsulated in the synthetic nanocarriers.
  • the synthetic nanocarriers comprise lipid nanoparticles, polymeric nanoparticles, metallic nanoparticles, surfactant-based emulsions, dendrimers, buckyballs, nanowires, virus-like particles or peptide or protein particles.
  • the synthetic nanocarriers comprise polymeric nanoparticles.
  • the polymeric nanoparticles comprise a polymer that is a non-methoxy-terminated, pluronic polymer.
  • the polymeric nanoparticles comprise a polyester, polyester attached to a polyether, polyamino acid, polycarbonate, polyacetal, polyketal, polysaccharide, polyethyloxazoline or polyethyleneimine.
  • the polyester comprises a poly(lactic acid), poly(glycolic acid), poly(lactic-co-glycolic acid) or polycaprolactone.
  • the polymeric nanoparticles comprise a polyester and a polyester attached to a polyether.
  • the polyether comprises polyethylene glycol or polypropylene glycol.
  • the mean of a particle size distribution obtained using dynamic light scattering of a population of the synthetic nanocarriers is a diameter greater than 110 nm. In one embodiment of any one of the methods provided herein, the diameter is greater than 150 nm. In one embodiment of any one of the methods provided herein, the diameter is greater than 200 nm. In one embodiment of any one of the methods provided herein, the diameter is greater than 250 nm. In one embodiment of any one of the methods provided herein, the diameter is less than 5 ⁇ m. In one embodiment of any one of the methods provided herein, the diameter is less than 4 ⁇ m. In one embodiment of any one of the methods provided herein, the diameter is less than 3 ⁇ m.
  • the diameter is less than 2 ⁇ m. In one embodiment of any one of the methods provided herein, the diameter is less than 1 ⁇ m. In one embodiment of any one of the methods provided herein, the diameter is less than 500 nm. In one embodiment of any one of the methods provided herein, the diameter is less than 450 nm. In one embodiment of any one of the methods provided herein, the diameter is less than 400 nm. In one embodiment of any one of the methods provided herein, the diameter is less than 350 nm. In one embodiment of any one of the methods provided herein, the diameter is less than 300 nm.
  • the load of immunosuppressant comprised in the synthetic nanocarriers, on average across the synthetic nanocarriers, is between 0.1% and 50% (weight/weight). In one embodiment of any one of the methods provided herein, the load is between 0.1% and 25%. In one embodiment of any one of the methods provided herein, the load is between 1% and 25%. In one embodiment of any one of the methods provided herein, the load is between 2% and 25%.
  • the immunosuppressant is an inhibitor of the NF-kB pathway. In one embodiment of any one of the methods provided herein, the immunosuppressant is rapamycin.
  • an aspect ratio of a population of the synthetic nanocarriers is greater than 1:1, 1:1.2, 1:1.5, 1:2, 1:3, 1:5, 1:7 or 1:10.
  • the method further comprises performing the method according to a protocol that attenuates an anti-viral transfer vector response, such as an antibody, T cell or B cell response, escalates transgene expression or that establishes an anti-viral transfer vector response.
  • a protocol that attenuates an anti-viral transfer vector response such as an antibody, T cell or B cell response, escalates transgene expression or that establishes an anti-viral transfer vector response.
  • the method further comprises assessing an antibody immune response against the viral transfer vector prior to, during or subsequent to the administering to the subject.
  • any one of the compositions is for use in any one of the methods provided.
  • any one of the methods is for use in treating any one of the disease or disorders described herein. In another aspect, any one of the methods is for use in attenuating an anti-viral transfer vector response, establishing an attenuated anti-viral transfer vector response, escalating transgene expression or for repeated administration of a viral transfer vector.
  • FIG. 1 shows GFP expression in livers of mice injected with AAV with or without synthetic nanocarriers comprising rapamycin at prime or boost. All cells in suspension have been analyzed for GFP expression with the exception of high side-scatter debris (2-3% of total, a by-product of collagenase treatment) excluded by the first ‘clean’ gate. All the remaining cells were gated for relative GFP strength (FL-1 channel). Numbers shown represent the percentage of GFP-positive cells of the total parent population.
  • FIG. 2 shows GFP expression in livers of AAV-injected mice as a function of boost with or without synthetic nanocarriers comprising rapamycin. Data presented are the same as in FIG. 1 , but are grouped according to whether AAV boost employed co-administration with the synthetic nanocarriers comprising rapamycin or not (unboosted samples from gr. 5 and 6 are also shown as a separate ‘supergroup’).
  • FIG. 3 demonstrates the GFP high cell share in livers of animals injected with AAV with or without synthetic nanocarriers comprising rapamycin.
  • GFP-positive cells (as presented in FIG. 1 ) were gated and then a population with an average GFP fluorescence intensity of 10 times higher than average in the parent population was gated again. Numbers presented are percentage from the parent GFP-positive population as seen in FIG. 1 .
  • FIG. 4 shows results from an experiment where mice were bled at d14 after receiving a single AAV-GFP inoculation with or without co-administration of synthetic nanocarriers comprising rapamycin and their sera assayed for antibodies against AAV. Top ODs for 1:40 serum dilutions are shown for all mice. Background normal mouse serum had an OD of 0.227.
  • FIG. 5 shows results from an experiment where mice were bled at days 14, 21 and 33 after receiving a single AAV-GFP inoculation with or without co-administration of synthetic nanocarriers comprising rapamycin (i.v.) and their sera assayed for antibodies against AAV. Top ODs for 1:40 serum dilutions are shown for all mice. Background normal mouse serum activity is shown. Statistical significance is calculated using two-way ANOVA.
  • FIG. 6 shows results from an experiment where mice were injected with AAV-GFP at days 0 and 21 with or without co-administration of synthetic nanocarriers comprising rapamycin (i.v.) at either or both injections, then bled at days 14 and 33 and their sera assayed for antibodies against AAV. Top ODs for 1:40 serum dilutions are shown for all mice. Background normal mouse serum activity is shown. Statistical significance is calculated using two-way ANOVA.
  • FIG. 7 provides data that are the same as in FIG. 6 with the readings for individual mice shown.
  • Two mice in the group treated with synthetic nanocarriers comprising rapamycin only at boost immunization (d21) did not show detectable antibodies at day 33 despite being positive at d14 (solid arrows).
  • One of five mice in both groups treated with synthetic nanocarriers comprising rapamycin at the prime had a detectable antibody level at d33 (dashed arrows) with the mouse from the group treated with synthetic nanocarriers comprising rapamycin at both prime and boost having a lower antibody level (open diamonds).
  • FIG. 10 shows results from an experiment where mice were injected with AAV8-GFP at days 0 and 21 with or without co-administration of synthetic nanocarriers comprising rapamycin (i.v.) at one or both injections, as indicated, and then bled at days 14 and 33.
  • Sera were assayed for antibodies against AAV8 by ELISA.
  • ODs for 1:40 serum dilutions are shown for all mice. Background level of normal mouse serum is indicated by the dotted line.
  • Statistical significance is calculated using two-way ANOVA.
  • FIG. 11 shows GFP expression in livers of mice injected with AAV with or without synthetic nanocarriers comprising rapamycin at prime or boost. All cells in suspension have been analyzed for GFP expression with the exception of high side-scatter debris (2-3% of total, a by-product of collagenase treatment) excluded by the first ‘clean’ gate. All the remaining cells were gated for relative GFP strength (FL-1 channel). Numbers shown represent the percentage of GFP-positive cells of the total parent population.
  • FIG. 12 shows RFP expression in livers of mice injected with AAV with or without synthetic nanocarriers comprising rapamycin at prime and/or boost. All cells in suspension have been analyzed for RFP expression with the exception of high side-scatter debris. Numbers shown represent the percentage of RFP-positive cells of the total parent population of liver cells.
  • FIG. 13 shows cytotoxic activity in mice immunized with AAV-GFP alone or in combination with synthetic nanocarriers comprising rapamycin.
  • Animals were injected with AAV8-GFP (i.v.) on days 0 and 21 with or without synthetic nanocarriers comprising rapamycin.
  • Target cells pulsed with a combination of dominant cytotoxic peptides from AAV capsid protein and the GFP transgene were administered at 7 days after the last injection (day 28) and their viability measured 18 hours later and compared to that of non-peptide pulsed control cells.
  • FIG. 14 shows AAV-specific IFN- ⁇ production in mice immunized with AAV-GFP alone or in combination with synthetic nanocarriers comprising rapamycin.
  • Animals were injected with AAV-GFP (i.v.) on days 0 and 17 with or without NCS.
  • Splenocytes were isolated on day 25 and incubated in vitro with dominant MHC class I-binding peptide from AAV capsid protein for 7 days and then assayed by ELISpot with the same peptide. Each sample was run in duplicate and presented with background subtracted.
  • FIG. 15 shows GFP-specific IFN- ⁇ production in mice immunized with AAV-GFP alone or in combination with synthetic nanocarriers comprising rapamycin.
  • Animals were injected (i.v.) with AAV8-GFP on days 0 and 17 with or without synthetic nanocarriers comprising rapamycin.
  • Splenocytes were isolated and incubated in vitro with MHC class I-binding peptide from GFP for 7 days and then assayed by ELISpot with the same peptide. Each sample was run in duplicate and presented with background subtracted.
  • FIG. 16 shows the design for an experiment.
  • FIG. 17 shows results from an experiment where mice were injected with rAAV2/8-luciferase on day 0 with or without co-administration of synthetic nanocarriers carrying 100 ⁇ g of rapamycin (i.v.) and then challenged with an i.v. injection of AAV-hFIX on day 14. Sera was collected at various time points, as indicated, and assayed for antibodies against AAV8 (left) and for the levels of human factor IX protein (right).
  • FIG. 18 shows the experimental design for an experiment.
  • FIG. 19 shows results from an experiment where male C57BL/6 mice were injected (i.v.) with rAAV2/8-luciferase concomitantly with synthetic nanocarriers carrying 100 ⁇ g of rapamycin on day 0 and then injected with rAAV2/8-hFIX concomitantly with synthetic nanocarriers carrying 100 ⁇ g of rapamycin on day 21. Control animals were treated similarly but with empty nanocarriers instead of synthetic nanocarriers comprising rapamycin. Sera were collected at various time points, as indicated, and assayed by ELISA for antibodies against AAV (left) and for levels of human FIX protein (right). AAV2/8-FIX vector copy number in the liver (middle) was determined by PCR.
  • FIG. 20 shows the experimental design for an experiment.
  • FIG. 21 provides results that showed that concomitant i.v. administration of synthetic nanocarriers carrying rapamycin with an rAAV2/8 vector (AAV2/8-Luc) on day 0 did not have a profound impact on the antibody response to an AAV5 vector (AAV5-hFIX) administered on day 21.
  • the results also showed that mice concomitantly treated with synthetic nanocarriers comprising rapamycin and rAAV2/8-Luc on day 0 showed a robust response to immunization with recombinant hFIX protein in complete Freund's adjuvant (CFA) on day 21.
  • CFA complete Freund's adjuvant
  • a polymer includes a mixture of two or more such molecules or a mixture of differing molecular weights of a single polymer species
  • a synthetic nanocarrier includes a mixture of two or more such synthetic nanocarriers or a plurality of such synthetic nanocarriers
  • reference to “a DNA molecule” includes a mixture of two or more such DNA molecules or a plurality of such DNA molecules
  • reference to “an immunosuppressant” includes a mixture of two or more such immunosuppressant molecules or a plurality of such immunosuppressant molecules, and the like.
  • the term “comprise” or variations thereof such as “comprises” or “comprising” are to be read to indicate the inclusion of any recited integer (e.g. a feature, element, characteristic, property, method/process step or limitation) or group of integers (e.g. features, elements, characteristics, properties, method/process steps or limitations) but not the exclusion of any other integer or group of integers.
  • the term “comprising” is inclusive and does not exclude additional, unrecited integers or method/process steps.
  • compositions and methods comprising or may be replaced with “consisting essentially of” or “consisting of”.
  • the phrase “consisting essentially of” is used herein to require the specified integer(s) or steps as well as those which do not materially affect the character or function of the claimed invention.
  • the term “consisting” is used to indicate the presence of the recited integer (e.g. a feature, element, characteristic, property, method/process step or limitation) or group of integers (e.g. features, elements, characteristics, properties, method/process steps or limitations) alone.
  • Anti-viral transfer vectors are promising therapeutics for a variety of applications. Viral transfer vectors, therefore, may comprise transgenes that encode any protein that may have a beneficial therapeutic purpose. Unfortunately, the promise of these therapeutics has not yet been realized in the art in a large part due to cellular and humoral immune responses against the viral transfer vector. These immune responses include antibody, B cell and T cell responses and can be specific to viral antigens of the viral transfer vector, such as viral capsid or coat proteins or peptides thereof.
  • viral vectors such as adeno-associated vectors
  • cellular and humoral immune responses against a viral transfer vector can develop after a single administration of the viral transfer vector.
  • neutralizing antibody titers can increase and remain high for several years and can reduce the effectiveness of readministration of the viral transfer vector, as repeated administration of a viral transfer vector generally results in enhanced undesired immune responses.
  • viral transfer vector-specific CD8+ T cells may arise that eliminate transduced cells expressing a desired transgene product, such as, for example, on reexposure to a viral antigen, such as a capsid protein.
  • AAV capsid antigen triggered immune-mediated destruction of hepatocytes transduced with an AAV viral transfer vector (e.g., Manno et al., Nature Medicine, Vol. 12, No. 3, 2006).
  • AAV viral transfer vector e.g., Manno et al., Nature Medicine, Vol. 12, No. 3, 2006.
  • multiple rounds of administration of viral transfer vectors will be needed for long-term benefits, and, without the methods and compositions provided herein, the ability to do so would be expected to be severely limited particularly if readministration is needed.
  • viral transfer vector antigens can persist for some time, such as for at least several weeks, after a single administration (e.g., Nathawani et al., N Engl J Med 365; 25, 2011; Nathwani, et al., N Engl J Med 371; 21, 2014).
  • AAV8 adeno-associated virus serotype 8
  • the inventors have surprisingly and unexpectedly discovered that the problems and limitations noted above can be overcome by practicing the invention disclosed herein.
  • Methods and compositions are provided that offer solutions to the aformentioned obstacles to effective use of viral transfer vectors for treatment.
  • anti-viral transfer vector immune responses can be attenuated with the methods and related compositions provided herein.
  • the methods and compositions can increase the efficacy of treatment with viral transfer vectors and provide for long-term immune attenuation even if the administration of the viral transfer vector need be repeated.
  • administering means giving or dispensing a material to a subject in a manner that is pharmacologically useful.
  • the term is intended to include “causing to be administered”.
  • “Causing to be administered” means causing, urging, encouraging, aiding, inducing or directing, directly or indirectly, another party to administer the material.
  • Any one of the methods provided herein may comprise or further comprise a step of administering concomitantly an antigen-presenting cell targeted immunosuppressant and a viral transfer vector.
  • the concomitant administration is performed repeatedly.
  • the concomitant administration is simultaneous administration.
  • “Amount effective” in the context of a composition or dosage form for administration to a subject as provided herein refers to an amount of the composition or dosage form that produces one or more desired results in the subject, for example, the reduction or elimination of an immune response against a viral transfer vector or the generation of an anti-viral transfer vector attenuated response.
  • the amount effective can be for in vitro or in vivo purposes.
  • the amount can be one that a clinician would believe may have a clinical benefit for a subject that may experience undesired immune responses as a result of administration of a viral transfer vector.
  • the composition(s) administered may be in any one of the amounts effective as provided herein.
  • Amounts effective can involve reducing the level of an undesired immune response, although in some embodiments, it involves preventing an undesired immune response altogether. Amounts effective can also involve delaying the occurrence of an undesired immune response. An amount effective can also be an amount that results in a desired therapeutic endpoint or a desired therapeutic result. Amounts effective, preferably, result in a tolerogenic immune response in a subject to an antigen, such as a viral transfer vector antigen. Amounts effective, can also preferably result in increased transgene expression (the transgene being delivered by the viral transfer vector). This can be determined by measuring transgene protein concentrations in various tissues or systems of interest in the subject. This increased expression may be measured locally or systemically. The achievement of any of the foregoing can be monitored by routine methods.
  • the amount effective is one in which the desired immune response, such as the reduction or elimination of an immune response against a viral transfer vector or the generation of an anti-viral transfer vector attenuated response, persists in the subject for at least 1 week, at least 2 weeks or at least 1 month.
  • the amount effective is one which produces a measurable desired immune response, such as the reduction or elimination of an immune response against a viral transfer vector or the generation of an anti-viral transfer vector attenuated response.
  • the amount effective is one that produces a measurable desired immune response (e.g., to a specific viral transfer vector antigen), for at least 1 week, at least 2 weeks or at least 1 month.
  • 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.
  • Anti-viral transfer vector immune response refers to any undesired immune response against a viral transfer vector.
  • the undesired immune response is an antigen-specific immune response against the viral transfer vector or an antigen thereof.
  • the immune response is specific to a viral antigen of the viral transfer vector.
  • the immune response is specific to a protein or peptide encoded by the transgene of the viral transfer vector.
  • the immune response is specific to a viral antigen of the viral transfer vector and not to a protein or peptide that is encoded by the transgene of the viral transfer vector.
  • the immune response may be an anti-viral transfer vector antibody response, an anti-viral transfer vector T cell immune response, such as a CD4+ T cell or CD8+ T cell immune response, or an anti-viral transfer vector B cell immune response.
  • an anti-viral transfer vector immune response is said to be an “anti-viral transfer vector attenuated response” when it is in some manner reduced or eliminated in the subject or as compared to an expected or measured response in the subject or another subject.
  • the anti-viral transfer vector attenuated response in a subject comprises a reduced anti-viral transfer vector immune response (such as a T cell, B cell or antibody response) measured using a biological sample obtained from the subject following a concomitant administration as provided herein as compared to an anti-viral transfer vector immune response measured using a biological sample obtained from another subject, such as a test subject, following administration to this other subject of the viral transfer vector without concomitant administration of the antigen-presenting cell targeted immunosuppressant.
  • the biological sample is obtained from the other subject following administration to this other subject of the viral transfer vector without any administration of the antigen-presenting cell targeted immunosuppressant.
  • the anti-viral transfer vector attenuated response is a reduced anti-viral transfer vector immune response (such as a T cell, B cell or antibody response) in a biological sample obtained from the subject following a concomitant administration as provided herein upon a subsequent viral transfer vector in vitro challenge performed on the subject's biological sample as compared to the anti-viral transfer vector immune response detected upon viral transfer vector in vitro challenge performed on a biological sample obtained from another subject, such as a test subject, following administration to this other subject of the viral transfer vector without concomitant administration of the antigen-presenting cell targeted immunosuppressant.
  • the anti-viral transfer vector attenuated response is a reduced anti-viral transfer vector immune response (such as a T cell, B cell or antibody response) in the subject following a concomitant administration as provided herein upon a subsequent viral transfer vector challenge administered to the subject as compared to the anti-viral transfer vector immune response in another subject, such as a test subject, upon a viral transfer vector challenge administered to this other subject following administration to this other subject of the viral transfer vector without concomitant administration of the antigen-presenting cell targeted immunosuppressant.
  • the viral transfer vector is administered without any administration of the antigen-presenting cell targeted immunosuppressant.
  • Antigen means a B cell antigen or T cell antigen.
  • Type(s) of antigens means molecules that share the same, or substantially the same, antigenic characteristics.
  • antigens may be proteins, polypeptides, peptides, lipoproteins, glycolipids, polynucleotides, polysaccharides, etc.
  • Antigen-presenting cell targeted immunosuppressant means an agent that results in antigen-presenting cells (APCs) having a tolerogenic effect.
  • APCs antigen-presenting cells
  • Such an immunosuppressant can include immunosuppressants coupled to a carrier that results in delivery to APCs and a tolerogenic effect as well as agents that by virtue of their form or characteristics can result in APC tolerogenic effects.
  • antigen-presenting cell targeted immunosuppressants include, but are not limited to synthetic nanocarriers that comprise an immunosuppressant as described herein; immunosuppressants, as described herein, coupled to antibodies or antigen-binding fragments thereof that target APCs (or other ligand that targets an APC), erythrocyte-binding therapeutics, as well as particles that by virtue of their characteristics lead to APC tolerogenic immune responses, etc.
  • the immunosuppressant is an element that is in addition to the material that makes up the structure of the synthetic nanocarrier.
  • the immunosuppressant is a compound that is in addition and, in some embodiments, attached to the one or more polymers.
  • the immunosuppressant is again in addition to and, in some embodiments, attached to the one or more lipids.
  • the antigen-presenting cell targeted immunosuppressant is a synthetic nanocarrier coupled to an immunosuppressant, and the material of the synthetic nanocarrier also results in a tolerogenic effect
  • the immunosuppressant is an element present in addition to the material of the synthetic nanocarrier that results in a tolerogenic effect.
  • Antigen-specific refers to an immune response that results from the presence of an antigen of interest or that generates molecules that specifically recognize or bind the antigen of interest. Generally, while such responses are measurable against the antigen of interest, the responses are reduced or negligible in regard to other antigens. For example, where the immune response is antigen-specific antibody production, antibodies are produced that selectively bind the antigen of interest but not to other antigens.
  • antigen-specific CD4+ or CD8+ T cells can bind to an antigen of interest or portion thereof when presented in the context of MHC class I or II antigens, respectively, by an antigen-presenting cell (APC) or, in case of CD8+ T cells, by any other cell in which the antigen is produced (e.g., a cell infected with a virus).
  • APC antigen-presenting cell
  • antigen specificity refers to the selective prevention or inhibition of a specific immune response to a target antigen versus other unrelated or unassociated antigens (e.g. antigens that are temporally or spatially dislocated from the target antigen).
  • “Assessing an immune response” refers to any measurement or determination of the level, presence or absence, reduction, increase in, etc. of an immune response in vitro or in vivo. Such measurements or determinations may be performed on one or more samples obtained from a subject. Such assessing can be performed with any one of the methods provided herein or otherwise known in the art. The assessing may be assessing the number or percentage of antibodies or T cells, such as those specific to a viral transfer vector, such as in a sample from a subject. The assessing also may be assessing any effect related to the immune response, such as measuring the presence or absence of a cytokine, cell phenotype, etc.
  • Any one of the methods provided herein may comprise or further comprise a step of assessing an immune response to a viral transfer vector or antigen thereof.
  • the assessing may be done directly or indirectly.
  • the term is intended to include actions that cause, urge, encourage, aid, induce or direct another party to assess an immune response.
  • Attaching or “Attached” or “Couple” or “Coupled” (and the like) means to chemically associate one entity (for example a moiety) with another.
  • the attaching is covalent, meaning that the attachment occurs in the context of the presence of a covalent bond between the two entities.
  • the non-covalent attaching 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 attaching.
  • Average refers to the arithmetic mean unless otherwise noted.
  • Conscomitantly means administering two or more materials/agents 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, and even more preferably the two or more materials/agents are administered in combination.
  • concomitant administration may encompass administration of two or more materials/agents 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.
  • the materials/agents may be repeatedly administered concomitantly; that is concomitant administration on more than one occasion, such as provided in the Examples.
  • Determining means objectively ascertaining something, such as a fact, relationship or quantity. In some embodiments, whether or not a subject has a pre-existing immunity to a viral transfer vector may be determined. The term is intended to include “causing to be determined”. “Causing to be determined” means causing, urging, encouraging, aiding, inducing or directing another party to perform a step of determining as provided herein. In some embodiments, the step of determining may be determining whether or not a subject has a pre-existing immunity to a viral transfer vector. Any one of the methods provided herein may comprise or further comprise a step of determining as described herein including a step of determining whether or not a subject has a pre-existing immunity to a viral transfer vector.
  • Directing means influencing, such as taking some action to influence, in some manner the actions of another party, such as causing or controlling the acts of the other party in such a manner that they perform one or more steps as provided herein.
  • the other party is an agent of the party that is doing the directing.
  • the other party is not an agent of the party that is doing the directing, but the step(s) performed by the other party is/are attributable to or the result of the directing.
  • directing includes instructing or providing instructions to perform one or more steps in order to receive a benefit conditioned on the performance of the one or more steps.
  • Dosage form means a pharmacologically and/or immunologically active material in a medium, carrier, vehicle, or device suitable for administration to a subject. Any one of the compositions or doses provided herein may be in a dosage form.
  • Dose refers to a specific quantity of a pharmacologically and/or immunologically active material for administration to a subject for a given time.
  • a “prior dose” refers to an earlier dose of a material.
  • doses of the antigen-presenting cell targeted immunosuppressants and/or viral transfer vectors in the methods and compositions of the invention refer to the amount of the antigen-presenting cell targeted immunosuppressants and/or viral transfer vectors.
  • the dose can be administered based on the number of synthetic nanocarriers that provide the desired amount of antigen-presenting cell targeted immunosuppressant, in instances where the antigen-presenting cell targeted immunosuppressant is a synthetic nanocarrier that comprises an immunosuppressant.
  • dose refers to the amount of each of the repeated doses, which may be the same or different.
  • Encapsulate means to enclose at least a portion of a substance within a synthetic nanocarrier. In some embodiments, a substance is enclosed completely within a synthetic nanocarrier. In other embodiments, most or all of a substance that is encapsulated is not exposed to the local environment external to the synthetic nanocarrier. In other embodiments, no more than 50%, 40%, 30%, 20%, 10% or 5% (weight/weight) is exposed to the local environment. Encapsulation is distinct from absorption, which places most or all of a substance on a surface of a synthetic nanocarrier, and leaves the substance exposed to the local environment external to the synthetic nanocarrier.
  • “Escalating transgene expression” refers to increasing the level of the transgene expression product of a viral transfer vector in a subject, the transgene being delivered by the viral transfer vector.
  • the level of the transgene expression product may be determined by measuring transgene protein concentrations in various tissues or systems of interest in the subject.
  • the transgene expression product is a nucleic acid
  • the level of transgene expression may be measured by transgene nucleic acid products.
  • Escalating transgene expression can be determined, for example, by measuring the amount of the transgene expression product in a sample obtained from a subject and comparing it to a prior sample. The sample may be a tissue sample.
  • the transgene expression product can be measured using flow cytometry.
  • “Establishing” or “establish” means to generate an outcome or result or to deduce something, such as a fact or relationship. Which use of this term will be apparent based on the context in which it is used.
  • the establishing may be accomplished in a number of ways, including but not limited to, taking steps to accomplish the outcome or result. For example, in some embodiments, administration of material(s) as provided herein can generate the outcome or result.
  • the establishing may be accomplished by performing experiments, making projections, etc.
  • establishing that administration of a viral transfer vector is likely to generate an anti-viral transfer vector immune response in a subject may be based on results of experiments on a subject, including on one or more samples obtained therefrom.
  • the likelihood of generating an anti-viral transfer vector immune response in a subject is the likelihood of generating such a response with the administration (or repeated administration, in some embodiments) of a viral transfer vector in the absence of administration of an antigen-presenting cell targeted immunosuppressant as provided herein.
  • establishing that a subject has a pre-existing immunity to a viral transfer vector may also be based on the result of experiments on a subject, including on one or more samples obtained therefrom.
  • such establishing may be determined by assessing an immune response in the subject.
  • a dose of an antigen-presenting cell targeted immunosuppressant or a viral transfer vector may be determined by starting with a test dose and using known scaling techniques (such as allometric or isometric scaling) to determine the dose for administration. Such may also be used to establish a protocol as provided herein. “Establishing” or “establish” comprises “causing to be established.” “Causing to be established” means causing, urging, encouraging, aiding, inducing or directing or acting in coordination with an entity for the entity to perform a step of establishing as provided herein. In some embodiments of any one of the methods provided herein, the method may comprise or further comprise any one of the steps of establishing as described herein.
  • “Frequency” refers to the interval of time at which the antigen-presenting cell targeted immunosuppressant, the viral transfer vector or both in combination (such as with concomitant administration) are administered to a subject.
  • Gene therapy transgene refers to a nucleic acid that encodes a protein and that when introduced into a cell directs the expression of the protein.
  • the protein is a therapeutic protein.
  • the subject to which the gene therapy transgene is administered by way of a viral transfer vector has a disease or disorder whereby the subject's endogenous version of the protein is defective or produced in limited amounts or not at all.
  • the encoded protein has no human counterpart but is predicted to provide therapeutically beneficial effects in the treatment of a disease or disorder.
  • Immunosuppressant means a compound that causes a tolerogenic effect, preferably through its effects on APCs.
  • a tolerogenic effect generally refers to the modulation by the APC or other immune cells systemically and/or locally, that reduces, inhibits or prevents an undesired immune response to an antigen in a durable fashion.
  • the immunosuppressant is one that causes an APC to promote a regulatory phenotype in one or more immune effector cells.
  • the regulatory phenotype may be characterized by the inhibition of the production, induction, stimulation or recruitment of antigen-specific CD4+ T cells or B cells, the inhibition of the production of antigen-specific antibodies, the production, induction, stimulation or recruitment of Treg cells (e.g., CD4+CD25highFoxP3+ Treg cells), etc.
  • This may be the result of the conversion of CD4+ T cells or B cells to a regulatory phenotype.
  • This may also be the result of induction of FoxP3 in other immune cells, such as CD8+ T cells, macrophages and iNKT cells.
  • the immunosuppressant is one that affects the response of the APC after it processes an antigen.
  • the immunosuppressant is not one that interferes with the processing of the antigen. In a further embodiment, the immunosuppressant is not an apoptotic-signaling molecule. In another embodiment, the immunosuppressant is not a phospholipid.
  • Immunosuppressants include, but are not limited to, statins; mTOR inhibitors, such as rapamycin or a rapamycin analog (i.e., rapalog); TGF- ⁇ signaling agents; TGF- ⁇ receptor agonists; histone deacetylase inhibitors, such as Trichostatin A; corticosteroids; inhibitors of mitochondrial function, such as rotenone; P38 inhibitors; NF- ⁇ inhibitors, such as 6Bio, Dexamethasone, TCPA-1, IKK VII; adenosine receptor agonists; prostaglandin E2 agonists (PGE2), such as Misoprostol; phosphodiesterase inhibitors, such as phosphodiesterase 4 inhibitor (PDE4), such as Rolipram; proteasome inhibitors; kinase inhibitors; G-protein coupled receptor agonists; G-protein coupled receptor antagonists; glucocorticoids; retinoids; cytokine inhibitors; cytokine receptor
  • Immunosuppressants also include IDO, vitamin D3, retinoic acid, cyclosporins, such as cyclosporine A, aryl hydrocarbon receptor inhibitors, resveratrol, azathiopurine (Aza), 6-mercaptopurine (6-MP), 6-thioguanine (6-TG), FK506, sanglifehrin A, salmeterol, mycophenolate mofetil (MMF), aspirin and other COX inhibitors, niflumic acid, estriol and triptolide.
  • cyclosporins such as cyclosporine A, aryl hydrocarbon receptor inhibitors, resveratrol, azathiopurine (Aza), 6-mercaptopurine (6-MP), 6-thioguanine (6-TG), FK506, sanglifehrin A, salmeterol, mycophenolate mofetil (MMF), aspirin and other COX inhibitors, niflumic acid, estriol and triptolide.
  • immunosuppressants include, but are not limited, small molecule drugs, natural products, antibodies (e.g., antibodies against CD20, CD3, CD4), biologics-based drugs, carbohydrate-based drugs, RNAi, antisense nucleic acids, aptamers, methotrexate, NSAIDs; fingolimod; natalizumab; alemtuzumab; anti-CD3; tacrolimus (FK506), abatacept, belatacept, etc.
  • “Rapalog” refers to a molecule that is structurally related to (an analog) of rapamycin (sirolimus).
  • rapalogs include, without limitation, temsirolimus (CCI-779), everolimus (RAD001), ridaforolimus (AP-23573), and zotarolimus (ABT-578). Additional examples of rapalogs may be found, for example, in WO Publication WO 1998/002441 and U.S. Pat. No. 8,455,510, the rapalogs of which are incorporated herein by reference in their entirety.
  • the immunosuppressant can be a compound that directly provides the tolerogenic effect on APCs or it can be a compound that provides the tolerogenic effect indirectly (i.e., after being processed in some way after administration). Immunosuppressants, therefore, include prodrug forms of any of the compounds provided herein. Further immunosuppressants, are known to those of skill in the art, and the invention is not limited in this respect. In embodiments, the immunosuppressant may comprise any one of the agents provided herein.
  • “Inducing to purchase” refers to any act that suggests to an entity to purchase an antigen-presenting cell targeted immunosuppressant, a viral transfer vector or both to achieve a beneficial effect as described herein or to perform any one of the methods provided herein.
  • Such acts includes packaging an antigen-presenting cell targeted immunosuppressant, a viral transfer vector or both that describes the benefits of concomitant administration of an antigen-presenting cell targeted immunosuppressant and a viral transfer vector in order to attenuate an anti-viral transfer vector response, escalate transgene expression or allow for repeated administration of a viral transfer vector.
  • the packaging may describe or suggest the performance of any one of the methods provided herein.
  • Acts that induce an entity to purchase also include marketing an antigen-presenting cell targeted immunosuppressant, a viral transfer vector or an antigen-presenting cell targeted immunosuppressant and a viral transfer vector product with information describing or suggesting the use of such product for carrying out any of the beneficial effects described herein or any one of the methods provided herein.
  • the marketing includes materials that describe or suggest the use of such product for attenuating an anti-viral transfer vector response, escalating transgene expression or for repeated administration of a viral transfer vector.
  • acts of inducing may also comprise acts of communicating information describing or suggesting any of the foregoing. The communicating is an action that can be performed in any form whether written, oral, etc.
  • acts of inducing also include acts of distributing an antigen-presenting cell targeted immunosuppressant, a viral transfer vector or both.
  • Acts of distributing include any action to make available the antigen-presenting cell targeted immunosuppressant, viral transfer vector or both to an entity with information, packaging, marketing materials, etc. that describes, instructs or communicates any of the benefits described herein or the steps of any one of the methods provided herein or the ability to attenuate an anti-viral transfer vector response, escalate transgene expression or allow for repeated administration of a viral transfer vector.
  • Acts of distributing include selling, offering for sale, and transporting for sale (e.g., transporting to pharmacies, hospitals, etc.)
  • Load when coupled to a synthetic nanocarrier, is the amount of the immunosuppressant coupled to the synthetic nanocarrier based on the total dry recipe weight of materials in an entire synthetic nanocarrier (weight/weight). Generally, such a load is calculated as an average across a population of synthetic nanocarriers. In one embodiment, the load on average across the synthetic nanocarriers is between 0.1% and 99%. In another embodiment, the load is between 0.1% and 50%. In another embodiment, the load is between 0.1% and 20%. In a further embodiment, the load is between 0.1% and 10%. In still a further embodiment, the load is between 1% and 10%. In still a further embodiment, the load is between 7% and 20%.
  • the load is at least 0.1%, at least 0.2%, at least 0.3%, at least 0.4%, at least 0.5%, at least 0.6%, at least 0.7%, at least 0.8%, at least 0.9%, at least 1%, at least 2%, at least 3%, at least 4%, at least 5%, at least 6%, at least at least 7%, at least 8%, at least 9%, at least 10%, at least 11%, at least 12%, at least 13%, at least 14%, at least 15%, at least 16%, at least 17%, at least 18%, at least 19%, at least 20%, at least 25%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% on average across the population of synthetic nanocarriers.
  • the load is 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19% or 20% on average across the population of synthetic nanocarriers.
  • the load is no more than 25% on average across a population of synthetic nanocarriers.
  • the load is calculated as may be described in the Examples or as otherwise known in the art.
  • the load of immunosuppressant is the amount of the immunosuppressant in the particles or the like (weight/weight). In such embodiments, the load can approach 97%, 98%, 99% or more.
  • “Maximum dimension of a synthetic nanocarrier” means the largest dimension of a nanocarrier measured along any axis of the synthetic nanocarrier. “Minimum dimension of a synthetic nanocarrier” means the smallest dimension of a synthetic nanocarrier measured along any axis of the synthetic nanocarrier. For example, for a spheroidal synthetic nanocarrier, the maximum and minimum dimension of a synthetic nanocarrier would be substantially identical, and would be the size of its diameter. Similarly, for a cuboidal synthetic nanocarrier, the minimum dimension of a synthetic nanocarrier would be the smallest of its height, width or length, while the maximum dimension of a synthetic nanocarrier would be the largest of its height, width or length.
  • a minimum dimension of at least 75%, preferably at least 80%, more preferably at least 90%, of the synthetic nanocarriers in a sample, based on the total number of synthetic nanocarriers in the sample is equal to or greater than 100 nm.
  • a maximum dimension of at least 75%, preferably at least 80%, more preferably at least 90%, of the synthetic nanocarriers in a sample, based on the total number of synthetic nanocarriers in the sample is equal to or less than 5 ⁇ m.
  • a minimum dimension of at least 75%, preferably at least 80%, more preferably at least 90%, of the synthetic nanocarriers in a sample, based on the total number of synthetic nanocarriers in the sample is greater than 110 nm, more preferably greater than 120 nm, more preferably greater than 130 nm, and more preferably still greater than 150 nm.
  • Aspects ratios of the maximum and minimum dimensions of synthetic nanocarriers may vary depending on the embodiment.
  • aspect ratios of the maximum to minimum dimensions of the synthetic nanocarriers may vary from 1:1 to 1,000,000:1, preferably from 1:1 to 100,000:1, more preferably from 1:1 to 10,000:1, more preferably from 1:1 to 1000:1, still more preferably from 1:1 to 100:1, and yet more preferably from 1:1 to 10:1.
  • a maximum dimension of at least 75%, preferably at least 80%, more preferably at least 90%, of the synthetic nanocarriers in a sample, based on the total number of synthetic nanocarriers in the sample is equal to or less than 3 ⁇ m, more preferably equal to or less than 2 ⁇ m, more preferably equal to or less than 1 ⁇ m, more preferably equal to or less than 800 nm, more preferably equal to or less than 600 nm, and more preferably still equal to or less than 500 nm.
  • a minimum dimension of at least 75%, preferably at least 80%, more preferably at least 90%, of the synthetic nanocarriers in a sample, based on the total number of synthetic nanocarriers in the sample is equal to or greater than 100 nm, more preferably equal to or greater than 120 nm, more preferably equal to or greater than 130 nm, more preferably equal to or greater than 140 nm, and more preferably still equal to or greater than 150 nm.
  • Measurement of synthetic nanocarrier dimensions e.g., effective diameter
  • a suspension of synthetic nanocarriers can be diluted from an aqueous buffer into purified water to achieve a final synthetic nanocarrier suspension concentration of approximately 0.01 to 0.1 mg/mL.
  • the diluted suspension may be prepared directly inside, or transferred to, a suitable cuvette for DLS analysis.
  • the cuvette may then be placed in the DLS, allowed to equilibrate to the controlled temperature, and then scanned for sufficient time to acquire a stable and reproducible distribution based on appropriate inputs for viscosity of the medium and refractive indicies of the sample. The effective diameter, or mean of the distribution, is then reported.
  • Determining the effective sizes of high aspect ratio, or non-spheroidal, synthetic nanocarriers may require augmentative techniques, such as electron microscopy, to obtain more accurate measurements.
  • “Dimension” or “size” or “diameter” of synthetic nanocarriers means the mean of a particle size distribution, for example, obtained using dynamic light scattering.
  • “Measurable level” refers to any level that is above a negative control level or would be considered to be above background or signal noise.
  • a measurable level is one that would be considered to be a level indicating the presence of the molecule being measured.
  • Non-methoxy-terminated polymer means a polymer that has at least one terminus that ends with a moiety other than methoxy. In some embodiments, the polymer has at least two termini that ends with a moiety other than methoxy. In other embodiments, the polymer has no termini that ends with methoxy.
  • Non-methoxy-terminated, pluronic polymer means a polymer other than a linear pluronic polymer with methoxy at both termini.
  • Polymeric nanoparticles as provided herein can comprise non-methoxy-terminated polymers or non-methoxy-terminated, pluronic polymers.
  • Obtaining means an act of acquiring a material(s) by any means.
  • the material may be acquired by producing it, purchasing it, receiving it, etc. This term is intended to include “causing to obtain”.
  • “Causing to obtain” means causing, urging, encouraging, aiding, inducing or directing or acting in coordination with an entity for the entity to obtain a material(s) as provided herein.
  • the method may comprise or further comprise any one of the steps of obtaining as described herein.
  • “Pharmaceutically acceptable excipient” or “pharmaceutically acceptable carrier” means a pharmacologically inactive material used together with a pharmacologically active material to formulate the compositions.
  • Pharmaceutically acceptable excipients comprise a variety of materials known in the art, including but not limited to saccharides (such as glucose, lactose, and the like), preservatives such as antimicrobial agents, reconstitution aids, colorants, saline (such as phosphate buffered saline), and buffers.
  • Pre-existing immunity against the viral transfer vector refers to the presence of antibodies, T cells and/or B cells in a subject, which cells have been previously primed by prior exposure to antigens of the viral transfer vector or to crossreactive antigens, including but not limited to other viruses.
  • this pre-existing immunity is at a level that is expected to result in anti-viral transfer vector immune response(s) that interferes with the efficacy of the viral transfer vector.
  • this pre-existing immunity is at a level that is expected to result in anti-viral transfer vector immune response(s) upon subsequent exposure to the viral transfer vector.
  • Pre-existing immunity can be assessed by determining the level of antibodies, such as neutralizing antibodies, against a viral transfer vector present in a sample, such as a blood sample, from the subject. Assays for assessing the level of antibodies, such as neutralizing antibodies, are described herein at least in the Examples and are also known to those of ordinary skill in the art. Such an assay is an ELISA assay. Pre-existing immunity can also be assessed by determining antigen recall responses of immune cells, such as B or T cells, stimulated in vivo or in vitro with viral transfer vector antigens presented by APCs or viral antigen epitopes presented on MHC class I or MHC class II molecules.
  • Assays for antigen-specific recall responses include, but are not limited to, ELISpot, intracellular cytokine staining, cell proliferation, and cytokine production assays. Generally, these and other assays are known to those of ordinary skill in the art.
  • a subject that does not exhibit pre-existing immunity against the viral transfer vector is one with a level of anti-viral transfer vector antibodies, such as neutralizing antibodies, or memory B or T cells that would be considered to be negative.
  • a subject that does not exhibit pre-existing immunity against the viral transfer vector is one with a level of an anti-viral transfer vector response that is no more than 3 standard deviations above a mean negative control.
  • “Producing” refers to any action that results in a material being made.
  • An act of producing includes preparing the material or processing it in some manner.
  • an act of producing includes any act that makes that material available for use by another. This term is intended to include “causing to produce”.
  • “Causing to produce” means causing, urging, encouraging, aiding, inducing or directing or acting in coordination with an entity for the entity to make a material(s) as provided herein.
  • the method may comprise or further comprise any one of the steps of producing as described herein.
  • Protocol means a pattern of administering to a subject and includes any dosing regimen of one or more substances to a subject. Protocols are made up of elements (or variables); thus a protocol comprises one or more elements. Such elements of the protocol can comprise dosing amounts (doses), dosing frequency, routes of administration, dosing duration, dosing rates, interval between dosing, combinations of any of the foregoing, and the like. In some embodiments, a protocol may be used to administer one or more compositions of the invention to one or more test subjects. Immune responses in these test subjects can then be assessed to determine whether or not the protocol was effective in generating a desired or desired level of an immune response or therapeutic effect. Any therapeutic and/or immunologic effect may be assessed.
  • One or more of the elements of a protocol may have been previously demonstrated in test subjects, such as non-human subjects, and then translated into human protocols. For example, dosing amounts demonstrated in non-human subjects can be scaled as an element of a human protocol using established techniques such as alimetric scaling or other scaling methods. Whether or not a protocol had a desired effect can be determined using any of the methods provided herein or otherwise known in the art. For example, a sample may be obtained from a subject to which a composition provided herein has been administered according to a specific protocol in order to determine whether or not specific immune cells, cytokines, antibodies, etc. were reduced, generated, activated, etc.
  • An exemplary protocol is one previously demonstrated to result in a tolerogenic immune response against a viral transfer vector antigen or to achieve any one of the beneficial results described herein.
  • Useful methods for detecting the presence and/or number of immune cells include, but are not limited to, flow cytometric methods (e.g., FACS), ELISpot, proliferation responses, cytokine production, and immunohistochemistry methods.
  • Antibodies and other binding agents for specific staining of immune cell markers are commercially available.
  • kits typically include staining reagents for antigens that allow for FACS-based detection, separation and/or quantitation of a desired cell population from a heterogeneous population of cells.
  • a composition as provided herein is administered to a subject using one or more or all or substantially all of the elements of which a protocol is comprised, provided the selected element(s) are expected to achieve the desired result in the subject. Such expectation may be based on protocols determined in test subjects and scaling if needed.
  • Any one of the methods provided herein may comprise or further comprise a step of administering a dose of the antigen-presenting cell targeted immunosuppressant alone or in combination as described herein with one or more doses of a viral transfer vector according to a protocol that has been shown to attenuate an anti-viral transfer vector immune response or allow for the repeated administration of a viral transfer vector.
  • Any one of the method provided herein may comprise or further comprise determining such a protocol that achieves any one of the beneficial results described herein.
  • Providing means an action or set of actions that an individual performs that supplies a material for practicing the invention. Providing may include acts of producing, distributing, selling, giving, making available, prescribing or administering the material. The action or set of actions may be taken either directly oneself or indirectly. Thus, this term is intended to include “causing to provide”. “Causing to provide” means causing, urging, encouraging, aiding, inducing or directing or acting in coordination with an entity for the entity to supply a material for practicing of the present invention. In some embodiments of any one of the methods provided herein, the method may comprise or further comprise any one of the steps of providing as described herein.
  • Repeat dose or “repeat dosing” or the like means at least one additional dose or dosing that is administered to a subject subsequent to an earlier dose or dosing of the same material.
  • a repeated dose of a viral transfer vector is at least one additional dose of the viral transfer vector after a prior dose of the same material. While the material may be the same, the amount of the material in the repeated dose may be different from the earlier dose. For example, in an embodiment of any one of the methods or compositions provided herein, the amount of the viral transfer vector in the repeated dose may be less than the amount of the viral transfer vector of the earlier dose.
  • the repeated dose may be in an amount that is at least equal to the amount of the viral transfer vector in the earlier dose.
  • a repeat dose may be administered weeks, months or years after the prior dose.
  • the repeat dose or dosing is administered at least 1 week after the dose or dosing that occurred just prior to the repeat dose or dosing. Repeat dosing is considered to be efficacious if it results in a beneficial effect for the subject. Preferably, efficacious repeat dosing results in a beneficial effect in conjunction with an attenuated anti-viral transfer vector response.
  • “Selecting the doses of the viral transfer vector to be less than” refers to the selection of the doses of the viral transfer vector that is less than the amount of the viral transfer vector that would be selected for administration to the subject if the subject were to develop an anti-viral transfer vector immune response to the viral transfer vector due to the repeated dosing of the viral transfer vector. This term is intended to include “causing to select”. “Causing to select” means causing, urging, encouraging, aiding, inducing or directing or acting in coordination with an entity for the entity to select the aforementioned lesser dosing. In some embodiments of any one of the methods provided herein, the method may comprise or further comprise any one of the steps of selecting as described herein.
  • “Simultaneous” means administration at the same time or substantially at the same time where a clinician would consider any time between administrations virtually nil or negligible as to the impact on the desired therapeutic outcome. In some embodiments, simultaneous means that the administrations occur with 5, 4, 3, 2, 1 or fewer minutes.
  • 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. As used herein, a subject may be in one need of any one of the methods or compositions provided herein.
  • “Synthetic nanocarrier(s)” means a discrete object that is not found in nature, and that possesses at least one dimension that is less than or equal to 5 microns in size.
  • Albumin nanoparticles are generally included as synthetic nanocarriers, however in certain embodiments the synthetic nanocarriers do not comprise albumin nanoparticles.
  • synthetic nanocarriers do not comprise chitosan.
  • synthetic nanocarriers are not lipid-based nanoparticles.
  • synthetic nanocarriers do not comprise a phospholipid.
  • a synthetic nanocarrier can be, but is not limited to, one or a plurality of lipid-based nanoparticles (also referred to herein as lipid nanoparticles, i.e., nanoparticles where the majority of the material that makes up their structure are lipids), polymeric nanoparticles, metallic nanoparticles, surfactant-based emulsions, dendrimers, buckyballs, nanowires, virus-like particles (i.e., particles that are primarily made up of viral structural proteins but that are not infectious or have low infectivity), peptide or protein-based particles (also referred to herein as protein particles, i.e., particles where the majority of the material that makes up their structure are peptides or proteins) (such as albumin nanoparticles) and/or nanoparticles that are developed using a combination of nanomaterials such as lipid-polymer nanoparticles.
  • lipid-based nanoparticles also referred to herein as lipid nanoparticles, i
  • Synthetic nanocarriers may be a variety of different shapes, including but not limited to spheroidal, cuboidal, pyramidal, oblong, cylindrical, toroidal, and the like. Synthetic nanocarriers according to the invention comprise one or more surfaces. Exemplary synthetic nanocarriers that can be adapted for use in the practice of the present invention comprise: (1) the biodegradable nanoparticles disclosed in U.S. Pat. No.
  • Synthetic nanocarriers according to the invention that have a minimum dimension of equal to or less than about 100 nm, preferably equal to or less than 100 nm, do not comprise a surface with hydroxyl groups that activate complement or alternatively comprise a surface that consists essentially of moieties that are not hydroxyl groups that activate complement.
  • synthetic nanocarriers according to the invention that have a minimum dimension of equal to or less than about 100 nm, preferably equal to or less than 100 nm, do not comprise a surface that substantially activates complement or alternatively comprise a surface that consists essentially of moieties that do not substantially activate complement.
  • synthetic nanocarriers according to the invention that have a minimum dimension of equal to or less than about 100 nm, preferably equal to or less than 100 nm, do not comprise a surface that activates complement or alternatively comprise a surface that consists essentially of moieties that do not activate complement.
  • synthetic nanocarriers exclude virus-like particles.
  • 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.
  • “Therapeutic protein” means any protein that may be expressed from a gene therapy transgene as provided herein.
  • the therapeutic protein may be one used for protein replacement or protein supplementation.
  • Therapeutic proteins include, but are not limited to, enzymes, enzyme cofactors, hormones, blood clotting factors, cytokines, growth factors, etc. Examples of other therapeutic proteins are provided elsewhere herein.
  • a subject may be one in need of treatment with any one of the therapeutic proteins provided herein.
  • Transgene of the viral transfer vector refers to the nucleic acid material the viral transfer vector is used to transport into a cell and, once in the cell, is expressed to produce a protein or nucleic acid molecule, respectively, such as for a therapeutic application as described herein.
  • the transgene may be a gene therapy transgene.
  • “Expressed” or “expression” or the like refers to the synthesis of a functional (i.e., physiologically active for the desired purpose) gene product after the transgene is transduced into a cell and processed by the transduced cell.
  • Such a gene product is also referred to herein as a “transgene expression product”.
  • the expressed products are, therefore, the resultant protein or nucleic acid, such as an antisense oligonucleotide or a therapeutic RNA, encoded by the transgene.
  • “Viral transfer vector” means a viral vector that has been adapted to deliver a transgene as provided herein.
  • “Viral vector” refers to all of the viral components of a viral transfer vector that delivers a transgene.
  • “viral antigen” refers to an antigen of the viral components of the viral transfer vector, such as a capsid or coat protein, but not to the transgene or to the product it encodes.
  • “Viral transfer vector antigen” refers to any antigen of the viral transfer vector including its viral components as well as a protein transgene expression product.
  • Viral vectors are engineered to transduce one or more desired nucleic acids into a cell.
  • the transgene may be a gene therapy transgene.
  • the transgene is one that encodes a protein provided herein, such as a therapeutic protein.
  • Viral vectors can be based on, without limitation, retroviruses (e.g., murine retrovirus, avian retrovirus, Moloney murine leukemia virus (MoMuLV), Harvey murine sarcoma virus (HaMuSV), murine mammary tumor virus (MuMTV), gibbon ape leukemia virus (GaLV) and Rous Sarcoma Virus (RSV)), lentiviruses, herpes viruses, adenoviruses, adeno-associated viruses, alphaviruses, etc. Other examples are provided elsewhere herein or are known in the art.
  • retroviruses e.g., murine retrovirus, avian retrovirus, Moloney murine leukemia virus (MoMuLV), Harvey murine sarcoma virus (HaMuSV), murine mammary tumor virus (MuMTV), gibbon ape
  • the viral vectors may be based on natural variants, strains, or serotypes of viruses, such as any one of those provided herein.
  • the viral vectors may also be based on viruses selected through molecular evolution.
  • the viral vectors may also be engineered vectors, recombinant vectors, mutant vectors, or hybrid vectors.
  • the viral vector is a “chimeric viral vector”. In such embodiments, this means that the viral vector is made up of viral components that are derived from more than one virus or viral vector.
  • cellular and humoral immune responses against the viral transfer vector can adversely effect the efficacy of viral transfer vector therapeutics and can also interfere with their readministration, making long-term treatment impossible for many patients.
  • treatment with viral transfer vectors would not be expected to be successful for some patients due to prior exposure to a virus on which the viral transfer is based.
  • a single administration of the viral transfer vector is likely to result in cellular and humoral immune responses, such as neutralizing antibody titers and/or the activation of memory T cells, that would not allow for successful readministration. Further compounding these issues is the long-term persistence of viral transfer vector antigens.
  • the methods and compositions provided herein have been found to overcome the aforementioned obstacles by attenuating immune responses against viral transfer vectors.
  • the methods and compositions provided herein have also been found to allow for the readministration of viral transfer vectors and provide for long-lasting tolerance against the viral transfer vector without the need for long-term immunosuppression. Accordingly, the methods and compositions provided herein are useful for the treatment of subjects with a viral transfer vector.
  • Viral transfer vectors can be used to deliver transgenes for a variety of purposes, including for gene therapy, the methods and compositions provided herein are also so applicable.
  • the transgene of the viral transfer vectors provided herein may be a gene therapy transgene and encode any protein or a portion thereof beneficial to a subject, such as one with a disease or disorder.
  • the protein may be an extracellular, intracellular or membrane-bound protein.
  • the protein is a therapeutic protein, and the subject to which the gene therapy transgene is administered by way of a viral transfer vector has a disease or disorder whereby the subject's endogenous version of the protein is defective or produced in limited amounts or not at all.
  • the subject may be one with any one of the diseases or disorders as provided herein, and the transgene is one that encodes any one of the therapeutic proteins or portion thereof as provided herein.
  • therapeutic proteins include, but are not limited to, infusible or injectable therapeutic proteins, enzymes, enzyme cofactors, hormones, blood or blood coagulation factors, cytokines and interferons, growth factors, adipokines, etc.
  • infusible or injectable therapeutic proteins include, for example, Tocilizumab (Roche/Actemra®), alpha-1 antitrypsin (Kamada/AAT), Hematide® (Affymax and Takeda, synthetic peptide), albinterferon alfa-2b (Novartis/ZalbinTM), Rhucin® (Pharming Group, C1 inhibitor replacement therapy), tesamorelin (Theratechnologies/Egrifta, synthetic growth hormone-releasing factor), ocrelizumab (Genentech, Roche and Biogen), belimumab (GlaxoSmithKline/Benlysta®), pegloticase (Sasilis/KrystexxaTM), taliglucerase alfa (Protalix/Uplyso), agalsidase alfa (Shire/Replagal®), and velaglucerase alfa (Shire).
  • Tocilizumab Roche/Actemra®
  • enzymes include lysozyme, oxidoreductases, transferases, hydrolases, lyases, isomerases, asparaginases, uricases, glycosidases, proteases, nucleases, collagenases, hyaluronidases, heparinases, heparanases, kinases, phosphatases, lysins and ligases.
  • enzymes include those that used for enzyme replacement therapy including, but not limited to, imiglucerase (e.g., CEREZYMETM), a-galactosidase A (a-gal A) (e.g., agalsidase beta, FABRYZYMETM), acid a-glucosidase (GAA) (e.g., alglucosidase alfa, LUMIZYMETM, MYOZYMETM), and arylsulfatase B (e.g., laronidase, ALDURAZYMETM, idursulfase, ELAPRASETM, arylsulfatase B, NAGLAZYMETM).
  • imiglucerase e.g., CEREZYMETM
  • a-gal A e.g., agalsidase beta, FABRYZYMETM
  • GAA acid a-glucosidase
  • hormones include Melatonin (N-acetyl-5-methoxytryptamine), Serotonin, Thyroxine (or tetraiodothyronine) (a thyroid hormone), Triiodothyronine (a thyroid hormone), Epinephrine (or adrenaline), Norepinephrine (or noradrenaline), Dopamine (or prolactin inhibiting hormone), Antimullerian hormone (or mullerian inhibiting factor or hormone), Adiponectin, Adrenocorticotropic hormone (or corticotropin), Angiotensinogen and angiotensin, Antidiuretic hormone (or vasopressin, arginine vasopressin), Atrial-natriuretic peptide (or atriopeptin), Calcitonin, Cholecystokinin, Corticotropin-releasing hormone, Erythropoietin, Follicle-stimulating hormone, Gastrin, Ghrelin
  • blood or blood coagulation factors include Factor I (fibrinogen), Factor II (prothrombin), tissue factor, Factor V (proaccelerin, labile factor), Factor VII (stable factor, proconvertin), Factor VIII (antihemophilic globulin), Factor IX (Christmas factor or plasma thromboplastin component), Factor X (Stuart-Prower factor), Factor Xa, Factor XI, Factor XII (Hageman factor), Factor XIII (fibrin-stabilizing factor), von Willebrand factor, von Heldebrant Factor, prekallikrein (Fletcher factor), high-molecular weight kininogen (HMWK) (Fitzgerald factor), fibronectin, fibrin, thrombin, antithrombin, such as antithrombin III, heparin cofactor II, protein C, protein S, protein Z, protein Z-related protease inhibitot (ZPI), plasminogen, alpha 2-
  • cytokines examples include lymphokines, interleukins, and chemokines, type 1 cytokines, such as IFN- ⁇ , TGF- ⁇ , and type 2 cytokines, such as IL-4, IL-10, and IL-13.
  • growth factors include Adrenomedullin (AM), Angiopoietin (Ang), Autocrine motility factor, Bone morphogenetic proteins (BMPs), Brain-derived neurotrophic factor (BDNF), Epidermal growth factor (EGF), Erythropoietin (EPO), Fibroblast growth factor (FGF), Glial cell line-derived neurotrophic factor (GDNF), Granulocyte colony-stimulating factor (G-CSF), Granulocyte macrophage colony-stimulating factor (GM-CSF), Growth differentiation factor-9 (GDF9), Hepatocyte growth factor (HGF), Hepatoma-derived growth factor (HDGF), Insulin-like growth factor (IGF), Migration-stimulating factor, Myostatin (GDF-8), Nerve growth factor (NGF) and other neurotrophins, Platelet-derived growth factor (PDGF), Thrombopoietin (TPO), Transforming growth factor alpha (TGF- ⁇ ), Transforming growth factor beta (TGF- ⁇ ), T
  • adipokines examples include leptin and adiponectin.
  • therapeutic proteins include, but are not limited to, receptors, signaling proteins, cytoskeletal proteins, scaffold proteins, transcription factors, structural proteins, membrane proteins, cytosolic proteins, binding proteins, nuclear proteins, secreted proteins, golgi proteins, endoplasmic reticulum proteins, mitochondrial proteins, and vesicular proteins, etc.
  • the transgene of the gene therapy viral transfer vectors provided herein may encode a functional version of any protein that through some defect in the endogenous version of which in a subject (including a defect in the expression of the endogenous version) results in a disease or disorder in the subject.
  • diseases or disorders include, but are not limited to, lysosomal storage diseases/disorders, such as Santavuori-Haltia disease (Infantile Neuronal Ceroid Lipofuscinosis Type 1), Jansky-Bielschowsky Disease (late infantile neuronal ceroid lipofuscinosis, Type 2), Batten disease (juvenile neuronal ceroid lipofuscinosis, Type 3), Kufs disease (neuronal ceroid lipofuscinosis, Type 4), Von Gierke disease (glycogen storage disease, Type Ia), glycogen storage disease, Type Ib, Pompe disease (glycogen storage disease, Type II), Forbes or Cori disease (glycogen storage disease, Type III), mucolipidosis II (I-Cell disease), mucolipidosis III (Pseudo-Hurler polydystrophy), mucolipdosis IV (sialolipidosis), cystinosis (adult nonnephropathic type),
  • Such diseases/disorders of a subject as provided herein include, but are not limited to, acid maltase deficiency (e.g., Pompe disease, glycogenosis type 2, lysosomal storage disease); carnitine deficiency; carnitine palmityl transferase deficiency; debrancher enzyme deficiency (e.g., Cori or Forbes disease, glycogenosis type 3); lactate dehydrogenase deficiency (e.g., glycogenosis type 11); myoadenylate deaminase deficiency; phosphofructokinase deficiency (e.g., Tarui disease, glycogenosis type 7); phosphogylcerate kinase deficiency (e.g., glycogenosis type 9); phosphogylcerate mutase deficiency (e.g., glycogenosis type 10); phosphorylase deficiency
  • diseases or disorders include, but are not limited to, Sickle Cell Anemia, Myotubular Myopathy, Hemophilia B, Lipoprotein lipase deficiency, Ornithine Transcarbamylase Deficiency, Crigler-Najjar Syndrome, Mucolipidosis IV, Niemann-Pick A, Sanfilippo A, Sanfilippo B, Sanfilippo C, Sanfilippo D, b-thalassaemia and Duchenne Muscular Dystrophy. Still futher examples of diseases or disorders include those that are the result of defects in lipid and sphingolipid degradation, mucopolysaccharide degradation, glycoprotein degradation, leukodystrophies, etc.
  • the functional versions of the defective proteins of any one of the disease or disorders provided herein may be encoded by the transgene of a gene therapy viral transfer vector and are also considered therapeutic proteins. It follows that therapeutic proteins also include Myophosphorylase, glucocerebrosidase, fibroblast growth factor receptor 3, huntingtin, HFE protein, CFTR, frataxin, VMD2, hemoglobin, phenylalanine hydroxylase, fibrillin, dystophia myotonica protein kinase, lignoceroyl-CoA ligase, dystrophin, methylCpG-binding protein 2, Beta hemoglobin, Myotubularin, Cathepsin A, Factor IX, Lipoprotein lipase, Beta galactosidase, Ornithine Transcarbamylase, Iduronate-2-Sulfatase, Acid-Alpha Glucosidase, UDP-glucuronosyltransferase 1-1, GlcNAc-1-phospho
  • therapeutic proteins also include functional versions of proteins associated with disorders of lipid and sphingolipid degradation (e.g., ⁇ -Galactosidase-1, ⁇ -Hexosaminidase A, ⁇ -Hexosaminidases A and B, GM2 Activator Protein, 8-Galactosidase A, Glucocerebrosidase, Glucocerebrosidase, Glucocerebrosidase, Arylsulfatase A, Galactosylceramidase, Sphingomyelinase, Sphingomyelinase, NPC1, HE1 protein (Cholesterol Trafficking Defect), Acid Ceramidase, Lysosomal Acid Lipase); disorders of mucopolysaccharide degradation (e.g., L-Iduronidase, L-Iduronidase, L-Iduronidase, Iduronate Sulfatase, Heparan N
  • the sequence of a transgene may also include an expression control sequence.
  • Expression control DNA sequences include promoters, enhancers, and operators, and are generally selected based on the expression systems in which the expression construct is to be utilized. In some embodiments, promoter and enhancer sequences are selected for the ability to increase gene expression, while operator sequences may be selected for the ability to regulate gene expression.
  • the transgene may also include sequences that facilitate, and preferably promote, homologous recombination in a host cell. The transgene may also include sequences that are necessary for replication in a host cell.
  • Exemplary expression control sequences include promoter sequences, e.g., cytomegalovirus promoter; Rous sarcoma virus promoter; and simian virus 40 promoter; as well as any other types of promoters that are disclosed elsewhere herein or are otherwise known in the art.
  • promoters are operatively linked upstream (i.e., 5′) of the sequence coding for a desired expression product.
  • the transgene also may include a suitable polyadenylation sequence (e.g., the SV40 or human growth hormone gene polyadenylation sequence) operably linked downstream (i.e., 3′) of the coding sequence.
  • Viruses have evolved specialized mechanisms to transport their genomes inside the cells that they infect; viral vectors based on such viruses can be tailored to transduce cells to specific applications.
  • viral vectors that may be used as provided herein are known in the art or described herein.
  • Suitable viral vectors include, for instance, retroviral vectors, lentiviral vectors, herpes simplex virus (HSV)-based vectors, adenovirus-based vectors, adeno-associated virus (AAV)-based vectors, and AAV-adenoviral chimeric vectors.
  • the viral transfer vectors provided herein may be based on a retrovirus.
  • Retrovirus is a single-stranded positive sense RNA virus capable of infecting a wide variety of host cells. Upon infection, the retroviral genome integrates into the genome of its host cell, using its own reverse transcriptase enzyme to produce DNA from its RNA genome. The viral DNA is then replicated along with host cell DNA, which translates and transcribes the viral and host genes.
  • a retroviral vector can be manipulated to render the virus replication-incompetent. As such, retroviral vectors are thought to be particularly useful for stable gene transfer in vivo. Examples of retroviral vectors can be found, for example, in U.S. Publication Nos. 20120009161, 20090118212, and 20090017543, the viral vectors and methods of their making being incorporated by reference herein in their entirety.
  • Lentiviral vectors are examples of retroviral vectors that can be used for the production of a viral transfer vector as provided herein.
  • Lentiviruses have the ability to infect non-dividing cells, a property that constitute a more efficient method of a gene delivery vector (see, e.g., Durand et al., Viruses. 2011 February; 3(2): 132-159).
  • lentiviruses include HIV (humans), simian immunodeficiency virus (SIV), feline immunodeficiency virus (FIV), equine infectious anemia virus (EIAV) and visna virus (ovine lentivirus).
  • HIV-based vectors are known to incorporate their passenger genes into non-dividing cells.
  • lentiviral vectors can be found, for example, in U.S. Publication Nos. 20150224209, 20150203870, 20140335607, 20140248306, 20090148936, and 20080254008, the viral vectors and methods of their making being incorporated by reference herein in their entirety.
  • Herpes simplex virus (HSV)-based viral vectors are also suitable for use as provided herein. Many replication-deficient HSV vectors contain a deletion to remove one or more intermediate-early genes to prevent replication. Advantages of the herpes vector are its ability to enter a latent stage that can result in long-term DNA expression, and its large viral DNA genome that can accommodate exogenous DNA up to 25 kb.
  • HSV-based viral vectors see, for example, U.S. Pat. Nos.
  • Adenoviruses are nonenveloped viruses that can transfer DNA in vivo to a variety of different target cell types.
  • the virus can be made replication-deficient by deleting select genes required for viral replication.
  • the expendable non-replication-essential E3 region is also frequently deleted to allow additional room for a larger DNA insert.
  • Viral transfer vectors can be based on adenoviruses.
  • Adenoviral transfer vectors can be produced in high titers and can efficiently transfer DNA to replicating and non-replicating cells. Unlike lentivirus, adenoviral DNA does not integrate into the genome and therefore is not replicated during cell division, instead they replicate in the nucleus of the host cell using the host's replication machinery.
  • the adenovirus on which a viral transfer vector may be based may be from any origin, any subgroup, any subtype, mixture of subtypes, or any serotype.
  • an adenovirus can be of subgroup A (e.g., serotypes 12, 18, and 31), subgroup B (e.g., serotypes 3, 7, 11, 14, 16, 21, 34, 35, and 50), subgroup C (e.g., serotypes 1, 2, 5, and 6), subgroup D (e.g., serotypes 8, 9, 10, 13, 15, 17, 19, 20, 22-30, 32, 33, 36-39, and 42-48), subgroup E (e.g., serotype 4), subgroup F (e.g., serotypes 40 and 41), an unclassified serogroup (e.g., serotypes 49 and 51), or any other adenoviral serotype.
  • subgroup A e.g., serotypes 12, 18, and 31
  • subgroup B e.g., serotypes 3, 7,
  • Adenoviral serotypes 1 through 51 are available from the American Type Culture Collection (ATCC, Manassas, Va.). Non-group C adenoviruses, and even non-human adenoviruses, can be used to prepare replication-deficient adenoviral vectors. Non-group C adenoviral vectors, methods of producing non-group C adenoviral vectors, and methods of using non-group C adenoviral vectors are disclosed in, for example, U.S. Pat. Nos. 5,801,030, 5,837,511, and 5,849,561, and International Patent Applications WO 97/12986 and WO 98/53087.
  • adenovirus even a chimeric adenovirus
  • a human adenovirus can be used as the source of the viral genome for a replication-deficient adenoviral vector.
  • adenoviral vectors can be found in U.S. Publication Nos. 20150093831, 20140248305, 20120283318, 20100008889, 20090175897 and 20090088398, the description of which viral vectors and methods of their making being incorporated by reference in its entirety.
  • the viral transfer vectors provided herein can also be based on adeno-associated viruses (AAVs).
  • AAV vectors have been of particular interest for use in therapeutic applications such as those described herein.
  • AAV is a DNA virus, which is not known to cause human disease.
  • AAV requires co-infection with a helper virus (e.g., an adenovirus or a herpes virus), or expression of helper genes, for efficient replication.
  • helper virus e.g., an adenovirus or a herpes virus
  • helper viruses e.g., an adenovirus or a herpes virus
  • AAV vectors that have been used in gene therapy applications generally have had approximately 96% of the parental genome deleted, such that only the terminal repeats (ITRs), which contain recognition signals for DNA replication and packaging, remain.
  • ITRs terminal repeats
  • AAV-based vectors see, for example, U.S. Pat. Nos. 8,679,837, 8,637,255, 8,409,842, 7,803,622, and 7,790,449, and U.S. Publication Nos. 20150065562, 20140155469, 20140037585, 20130096182, 20120100606, and 20070036757, the viral vectors of which and methods or their making being incorporated herein by reference in their entirety.
  • the AAV vectors may be recombinant AAV vectors.
  • the AAV vectors may also be self-complementary (sc) AAV vectors, which are described, for example, in U.S. Patent Publications 2007/01110724 and 2004/0029106, and U.S. Pat. Nos. 7,465,583 and 7,186,699, the vectors and methods of production of which are herein incorporated by reference.
  • sc self-complementary
  • the adeno-associated virus on which a viral transfer vector may be of any serotype or a mixture of serotypes.
  • AAV serotypes include AAV1, AAV 2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, and AAV11.
  • the viral transfer vector may contain the capsid signal sequences taken from one AAV serotype (for example selected from any one of AAV serotypes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, and 11) and packaging sequences from a different serotype (for example selected from any one of AAV serotypes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, and 11).
  • the AAV vector is an AAV 2/8 vector.
  • the AAV vector is an AAV 2/5 vector.
  • Alphaviruses include Sindbis (and VEEV) virus, Aura virus, Babanki virus, Barmah Forest virus, Bebaru virus, Cabassou virus, Chikungunya virus, Eastern equine encephalitis virus, Everglades virus, Fort Morgan virus, Getah virus, Highlands J virus, Kyzylagach virus, Mayaro virus, Me Tri virus, Middelburg virus, Mosso das Pedras virus, Mucambo virus, Ndumu virus, O′nyong-nyong virus, Pixuna virus, Rio Negro virus, Ross River virus, Salmon pancreas disease virus, Semliki Forest virus, Southern elephant seal virus, Tonate virus, Trocara virus, Una virus, Venezuelan equine encephalitis virus, Western equine encephalitis virus, and Whataroa virus.
  • viruses encode nonstructural (e.g., replicon) and structural proteins (e.g., capsid and envelope) that can be translated in the cytoplasm of the host cell.
  • Ross River virus, Sindbis virus, Semliki Forest virus (SFV), and Venezuelan equine encephalitis virus (VEEV) have all been used to develop viral transfer vectors for transgene delivery.
  • Pseudotyped viruses may be formed by combining alphaviral envelope glycoproteins and retroviral capsids. Examples of alphaviral vectors can be found in U.S. Publication Nos. 20150050243, 20090305344, and 20060177819; the vectors and methods of their making are incorporated herein by reference in their entirety.
  • Antigen-presenting cell targeted immunosuppressant can include agents that by virtue of their form or characteristics can result in APC tolerogenic effects. Antigen-presenting cell targeted immunosuppressant also include agents that comprise a carrier to which an immunosuppressant is conjugated.
  • Antigen-presenting cell targeted immunosuppressants include negatively-charged particles, such as polystyrene, PLGA, or diamond particles of a certain size and zeta potential, such as those described in U.S. Publication No. 20150010631, the description of such particles and methods of their production being incorporated herein by reference.
  • Such particles may have any particle shape or conformation. However, in some embodiments it is preferred to use particles that are less likely to clump in vivo. In one embodiment, these particles have a spherical shape. Generally, it is not necessary for such particles to be uniform in size, although such particles must generally be of a size sufficient to trigger phagocytosis in an antigen-presenting cell or other MPS cell. Preferably, these particles are microscopic or nanoscopic in size, in order to enhance solubility, avoid possible complications caused by aggregation in vivo and to facilitate pinocytosis.
  • These particles may an average diameter of from about 0.1 ⁇ m to about 10 ⁇ m, about 0.2 ⁇ m to about 2 ⁇ m, about 0.3 ⁇ m to about 5 ⁇ m, or about 0.5 ⁇ m to about 3 ⁇ m. In some embodiments, these particles may have an average diameter of about 0.1 ⁇ m, about 0.2 ⁇ m, about 0.3 ⁇ m, about 0.4 ⁇ m, about 0.5 ⁇ m, about 1.0 ⁇ m, about 1.5 ⁇ m, about 2.0 ⁇ m, about 2.5 ⁇ m, about 3.0 ⁇ m, about 3.5 ⁇ m, about 4.0 ⁇ m, about 4.5 ⁇ m, or about 5.0 ⁇ m. These particles need not be of uniform diameter, and a pharmaceutical formulation may contain a plurality of particles with a mixture of particle sizes.
  • these particles are non-metallic.
  • these particles may be formed from a polymer.
  • these particles are biodegradable.
  • suitable particles include polystyrene particles, PLGA particles, PLURONICS stabilized polypropylene sulfide particles, and diamond particles.
  • these particles can be formed from a wide range of other materials.
  • these particles may be composed of glass, silica, polyesters of hydroxy carboxylic acids, polyanhydrides of dicarboxylic acids, or copolymers of hydroxy carboxylic acids and dicarboxylic acids. More generally, these particles may be composed of other materials as described in U.S. Publication No. 20150010631.
  • the particles generally possess a particular zeta potential.
  • the zeta potential is negative.
  • the zeta potential may be less than about ⁇ 100 mV or less than about ⁇ 50 mV.
  • the particles possess a zeta potential between ⁇ 100 mV and 0 mV, between ⁇ 75 mV and 0 mV, between ⁇ 60 mV and 0 mV, between ⁇ 50 mV and 0 mV, between ⁇ 40 mV and 0 mV, between ⁇ 30 mV and 0 mV, between ⁇ 20 mV and +0 mV, between ⁇ 10 mV and ⁇ 0 mV, between ⁇ 100 mV and ⁇ 50 mV, between ⁇ 75 mV and ⁇ 50 mV, or between ⁇ 50 mV and ⁇ 40 mV.
  • these particles also comprise one or more antigens as provided herein.
  • the one or more antigens are encapsulated in the particles.
  • an antigen-presenting cell targeted immunosuppressant is an immunosuppressants in nanocrystalline form, whereby the form of the immunosuppressant itself is a particle or particle-like. In these embodiments, such forms mimic a virus or other foreign pathogen.
  • Many drugs have been nanosized and appropriate methods for producing such drug forms would be known to one of ordinary skill in the art.
  • Drug nanocrystals such as nanocrystalline rapamycin, are known to those of ordinary skill in the art (Katteboinaa, et al. 2009, International Journal of PharmTech Resesarch; Vol. 1, No. 3; pp 682-694.
  • a “drug nanocrystal” refers to a form of a drug (e.g., an immunosuppressant) that does not include a carrier or matrix material.
  • drug nanocrystals comprise 90%, 95%, 98%, or 99% or more drug.
  • Methods for producing drug nanocrystals include, without limitation, milling, high pressure homogenization, precipitation, spray drying, rapid expansion of supercritical solution (RESS), Nanoedge® technology (Baxter Healthcare), and Nanocrystal TechnologyTM (Elan Corporation).
  • a surfactant or a stabilizer may be used for steric or electrostatic stability of the drug nanocrystal.
  • the nanocrystal or nanocrystalline form of an immunosuppressant may be used to increase the solubility, stability, and/or bioavailability of the immunosuppressant, particularly immunosuppressants that are insoluble or labile.
  • Antigen-presenting call targeted immunosuppressants also may be an apoptotic-body mimic and cause an associated antigen(s) to be tolerized.
  • the apoptotic-body mimics may be particles, beads, branched polymers, dendrimers, or liposomes.
  • the mimic is particulate, and generally spherical, ellipsoidal, rod-shaped, globular, or polyhedral in shape. Alternatively, however, the mimic may be of an irregular or branched shape.
  • the mimic is composed of material which is biodegradable.
  • the mimic have a net neutral or negative charge, in order to reduce non-specific binding to cell surfaces which, in general, bear a net negative charge.
  • the mimic surface is composed of a material that minimizes non-specific or unwanted biological interactions.
  • the mimic surface may be coated with a material to prevent or decrease non-specific interactions.
  • Steric stabilization by coating particles with hydrophilic layers such as poly(ethylene glycol) (PEG) and its copolymers such as PLURONICS (including copolymers of poly(ethylene glycol)-bl-poly(propylene glycol)-bl-poly(ethylene glycol)) may reduce the non-specific interactions with proteins of the interstitium.
  • the mimics may be particles composed of glass, silica, polyesters of hydroxy carboxylic acids, polyanhydrides of dicarboxylic acids, or copolymers of hydroxy carboxylic acids and dicarboxylic acids.
  • These mimics may be quantum dots, or composed of quantum dots, such as quantum dot polystyrene particles.
  • These mimics may comprise materials including polyglycolic acid polymers (PGA), polylactic acid polymers (PLA), polysebacic acid polymers (PSA), poly(lactic-co-glycolic) acid copolymers (PLGA), poly(lactic-co-sebacic) acid copolymers (PLSA), poly(glycolic-co-sebacic) acid copolymers (PGSA), etc.
  • the mimics may also be polystyrene beads.
  • These mimics may comprise one or more antigens.
  • the mimics may be capable of being conjugated, either directly or indirectly, to one or more antigens to which tolerance is desired. In some instances, the mimic will have multiple binding sites in order to have multiple copies of the antigen exposed and increase the likelihood of a tolerogenic response.
  • the mimic may have one antigen on its surface or multiple different antigens on the surface. Alternatively, however, the mimic may have a surface to which conjugating moieties may be adsorbed without chemical bond formation.
  • the mimics may also comprise an apoptotic signaling molecule, although this is not necessarily required, such as with polystyrene beads.
  • Apoptotic signaling molecules include, but are not limited to, the apoptosis signaling molecules described in U.S. Publication No. 20050113297, which apoptosis signaling molecules are herein incorporated by reference.
  • Molecules suitable for use in these particles include molecules that target phagocytes, which include macrophages, dendritic cells, monocytes and neutrophils. Such molecules may be thrombospondins or Annexin I.
  • Antigen-presenting cell targeted immunosuppressants may also be erythrocyte-binding therapeutics, such as those described in U.S. Publication No. 20120039989, which therapeutics and methods of their making are incorporated herein by reference.
  • erythrocyte-binding therapeutics such as those described in U.S. Publication No. 20120039989, which therapeutics and methods of their making are incorporated herein by reference.
  • peptides that specifically bind to erythrocytes also known as red blood cells
  • an erythrocyte-binding therapeutic comprises one or more antigens to which tolerance is desired and an erythrocyte affinity ligand.
  • the one or more antigens may be viral transfer vector antigens as described herein, such as one or more viral antigens (e.g., of one or more viral capsid proteins). Also, the one or more antigens may also be or include one or more antigens of an expressed transgene as provided. The antigens may form a mixture to which tolerance is desired.
  • peptides that specifically bind erythrocytes include ERY1, ERY19, ERY59, ERY64, ERY123, ERY141 and ERY162.
  • proteins such as antibodies, for example single chain antibodies, and antigen binding fragments thereof may also be used as the affinity ligands.
  • the affinity ligands may also include nucleotide aptamer ligands for erythrocyte surface components. Accordingly, aptamers can be made and used in place of other erythrocyte affinity ligands. DNA and RNA aptamers may be used to provide non-covalent erythrocyte binding.
  • Aptamers can be classified as DNA aptamers, RNA aptamers, or peptide aptamers.
  • the affinity ligands may be a fusion of two or more affinity ligands, such as erythrocyte-binding peptides.
  • the components of the erythrocyte-binding therapeutics may be associated with a carrier such as a polymersome, a liposome or micelle or some types of nanoparticles. In some embodiments, the components are encapsulated in such a carrier.
  • the carrier comprises an affinity ligand as described herein and one or more antigens. In such an embodiment, the affinity ligand and one or more antigens do not necessarily need to be conjugated to each other.
  • Antigen-presenting cell targeted immunosuppressants also include any one of the immunosuppressants provided herein coupled to a carrier that targets APCs.
  • the carrier in some embodiments may be an antibody or antigen binding fragment thereof (or some other ligand) that is specific to an APC marker.
  • markers include, but are not limited to, CD1a (R4, T6, HTA-1); CD1b (R1); CD1c (M241, R7); CD1d (R3); CD1e (R2); CD11b ( ⁇ M Integrin chain, CR3, Mo1, C3niR, Mac-1); CD11c ( ⁇ X Integrin, p150, 95, AXb2); CDw117 (Lactosylceramide, LacCer); CD19 (B4); CD33 (gp67); CD 35 (CR1, C3b/C4b receptor); CD 36 (GpIIIb, GPIV, PASIV); CD39 (ATPdehydrogenase, NTPdehydrogenase-1); CD40 (Bp50); CD45 (LCA, T200, B220, Ly5); CD45RA; CD45RB; CD45RC; CD45RO (UCHL-1); CD49d (VLA-4 ⁇ , ⁇ 4 Integrin); CD49e (VLA-5 ⁇ , ⁇ 5 Integrin); CD58 (L
  • the antigen-presenting cell targeted immunosuppressant may also be synthetic nanocarriers that comprise any one of the immunosuppressants as described herein.
  • synthetic nanocarriers include those of U.S. Publication No. 20100151000, the synthetic nanocarriers of which, and methods of their making, are incorporated herein by reference.
  • particles e.g., liposomes or polymeric particles
  • particles that comprise an inhibitor of the NF- ⁇ B pathway and one or more viral transfer vector antigens can be used as antigen-presenting cell targeted immunosuppressants as provided herein.
  • the particle is liposomal.
  • the particle comprises a carrier particle, such as a metal particle (e.g., a tungsten, gold, platinum or iridium particle).
  • the particle comprises a polymeric matrix or carrier, illustrative examples of which include biocompatible polymeric particles (e.g., particles fabricated with poly(lactide-co-glycolide)).
  • the particle comprises a ceramic or inorganic matrix or carrier.
  • the inhibitor of the NF- ⁇ B pathway can decrease the level or functional activity of a member of the NF- ⁇ B pathway, and can be selected from BTK, LYN, BCR Ig.alpha., BCR Ig.beta., Syk, Blnk, PLC.gamma.2, PKC.beta., DAG, CARMA1, BCL10, MALT1, PI3K, PIPS, AKT, p38 MAPK, ERK, COT, IKK.alpha., IKK.beta., IKK.gamma., NIK, RelA/p65, P105/p50, c-Rel, RelB, p52, NIK, Leu13, CD81, CD19, CD21 and its ligands in the complement and coagulation cascade, TRAF6, ubiquitin ligase, Tab2, TAK1, NEMO, NOD2, RIP2, Lck, fyn, Zap70, LAT, GRB2,
  • synthetic nanocarriers can be used according to the invention, and in some embodiments, coupled to an immunosuppressant to provide still other antigen-presenting cell targeted immunosuppressants.
  • synthetic nanocarriers are spheres or spheroids.
  • synthetic nanocarriers are flat or plate-shaped.
  • synthetic nanocarriers are cubes or cubic.
  • synthetic nanocarriers are ovals or ellipses.
  • synthetic nanocarriers are cylinders, cones, or pyramids.
  • each synthetic nanocarrier has similar properties.
  • at least 80%, at least 90%, or at least 95% of the synthetic nanocarriers of any one of the compositions or methods provided, 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.
  • 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 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; diacylglycerolsuccinate; diphosphatidyl glycerol (DPPG); hexanedecanol; fatty alcohols such as polyethylene glycol (PEG); polyoxyethylene-9-lauryl ether; a surface active fatty acid, such as palmitic acid or oleic acid; fatty acids; fatty acid monoglycerides; fatty acid diglycerides; fatty acid amides; sorbitan trioleate (Span® 85) glycocholate; sorbitan monolaurate (Span® 20); polysorbate 20
  • amphiphilic entity component may be a mixture of different amphiphilic entities. Those skilled in the art will recognize that this is an exemplary, not comprehensive, list of substances with surfactant activity. Any amphiphilic entity may be used in the production of synthetic nanocarriers to be used in accordance with the present invention.
  • 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, hydroxyethylstarch, carageenan, glycon, amylose, chitosan, N,O-carboxylmethylchitosan, algin and alginic acid, starch, chitin, inulin, konjac, glucommannan, pustulan, heparin, hyaluronic acid, curdlan, and xanthan.
  • the synthetic nanocarriers do not comprise (or specifically exclude) carbohydrates, such as a polysaccharide.
  • the carbohydrate may comprise a carbohydrate derivative such as a sugar alcohol, including but not limited to mannitol, sorbitol, xylitol, erythritol, maltitol, and lactitol.
  • synthetic nanocarriers can comprise one or more polymers.
  • the synthetic nanocarriers comprise one or more polymers that is a non-methoxy-terminated, pluronic polymer.
  • at least 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% (weight/weight) of the polymers that make up the synthetic nanocarriers are non-methoxy-terminated, pluronic polymers.
  • all of the polymers that make up the synthetic nanocarriers are non-methoxy-terminated, pluronic polymers.
  • the synthetic nanocarriers comprise one or more polymers that is a non-methoxy-terminated polymer. In some embodiments, at least 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% (weight/weight) of the polymers that make up the synthetic nanocarriers are non-methoxy-terminated polymers.
  • all of the polymers that make up the synthetic nanocarriers are non-methoxy-terminated polymers.
  • the synthetic nanocarriers comprise one or more polymers that do not comprise pluronic polymer. In some embodiments, at least 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% (weight/weight) of the polymers that make up the synthetic nanocarriers do not comprise pluronic polymer. In some embodiments, all of the polymers that make up the synthetic nanocarriers do not comprise pluronic polymer. In some embodiments, such a polymer can be surrounded by a coating layer (e.g., liposome, lipid monolayer, micelle, etc.). In some embodiments, elements of the synthetic nanocarriers can be attached to the polymer.
  • a coating layer e.g., liposome,
  • Immunosuppressants can be coupled to the synthetic nanocarriers by any of a number of methods.
  • the attaching can be a result of bonding between the immunosuppressants and the synthetic nanocarriers. This bonding can result in the immunosuppressants being attached to the surface of the synthetic nanocarriers and/or contained (encapsulated) within the synthetic nanocarriers.
  • the immunosuppressants are encapsulated by the synthetic nanocarriers as a result of the structure of the synthetic nanocarriers rather than bonding to the synthetic nanocarriers.
  • the synthetic nanocarrier comprises a polymer as provided herein, and the immunosuppressants are attached to the polymer.
  • a coupling moiety can be any moiety through which an immunosuppressant is bonded to a synthetic nanocarrier.
  • moieties include covalent bonds, such as an amide bond or ester bond, as well as separate molecules that bond (covalently or non-covalently) the immunosuppressant to the synthetic nanocarrier.
  • molecules include linkers or polymers or a unit thereof.
  • the coupling moiety can comprise a charged polymer to which an immunosuppressant electrostatically binds.
  • the coupling moiety can comprise a polymer or unit thereof to which it is covalently bonded.
  • the synthetic nanocarriers comprise a polymer as provided herein. These synthetic nanocarriers can be completely polymeric or they can be a mix of polymers and other materials.
  • the polymers of a synthetic nanocarrier associate to form a polymeric matrix.
  • a component such as an immunosuppressant
  • covalent association is mediated by a linker.
  • a component can be noncovalently associated with one or more polymers of the polymeric matrix.
  • a component can be encapsulated within, surrounded by, and/or dispersed throughout a polymeric matrix.
  • a component can be associated with one or more polymers of 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.
  • 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.
  • the polymer comprises a polyester, polycarbonate, polyamide, or polyether, or unit thereof.
  • the polymer comprises poly(ethylene glycol) (PEG), polypropylene glycol, poly(lactic acid), poly(glycolic acid), poly(lactic-co-glycolic acid), or a polycaprolactone, or unit thereof.
  • the polymer is biodegradable. Therefore, in these embodiments, it is preferred that if the polymer comprises a polyether, such as poly(ethylene glycol) or polypropylene glycol or unit thereof, the polymer comprises a block-co-polymer of a polyether and a biodegradable polymer such that the polymer is biodegradable.
  • the polymer does not solely comprise a polyether or unit thereof, such as poly(ethylene glycol) or polypropylene glycol or unit thereof.
  • polymers suitable for use in the present invention include, but are not limited to polyethylenes, polycarbonates (e.g. poly(1,3-dioxan-2one)), polyanhydrides (e.g. poly(sebacic anhydride)), polypropylfumerates, polyamides (e.g. polycaprolactam), polyacetals, polyethers, polyesters (e.g., polylactide, polyglycolide, polylactide-co-glycolide, polycaprolactone, polyhydroxyacid (e.g.
  • polymers in accordance with the present invention include polymers which have been approved for use in humans by the U.S. Food and Drug Administration (FDA) under 21 C.F.R. ⁇ 177.2600, including but not limited to polyesters (e.g., polylactic acid, poly(lactic-co-glycolic acid), polycaprolactone, polyvalerolactone, poly(1,3-dioxan-2one)); polyanhydrides (e.g., poly(sebacic anhydride)); polyethers (e.g., polyethylene glycol); polyurethanes; polymethacrylates; polyacrylates; and polycyanoacrylates.
  • FDA U.S. Food and Drug Administration
  • polymers can be hydrophilic.
  • polymers may comprise anionic groups (e.g., phosphate group, sulphate group, carboxylate group); cationic groups (e.g., quaternary amine group); or polar groups (e.g., hydroxyl group, thiol group, amine group).
  • a synthetic nanocarrier comprising a hydrophilic polymeric matrix generates a hydrophilic environment within the synthetic nanocarrier.
  • polymers can be hydrophobic.
  • a synthetic nanocarrier comprising a hydrophobic polymeric matrix generates a hydrophobic environment within the synthetic nanocarrier. Selection of the hydrophilicity or hydrophobicity of the polymer may have an impact on the nature of materials that are incorporated within the synthetic nanocarrier.
  • polymers may be modified with one or more moieties and/or functional groups.
  • moieties or functional groups can be used in accordance with the present invention.
  • polymers may be modified with polyethylene glycol (PEG), with a carbohydrate, and/or with acyclic polyacetals derived from polysaccharides (Papisov, 2001, ACS Symposium Series, 786:301). Certain embodiments may be made using the general teachings of U.S. Pat. No. 5,543,158 to Gref et al., or WO publication WO2009/051837 by Von Andrian et al.
  • polymers may be modified with a lipid or fatty acid group.
  • a fatty acid group may be one or more of butyric, caproic, caprylic, capric, lauric, myristic, palmitic, stearic, arachidic, behenic, or lignoceric acid.
  • a fatty acid group may be one or more of palmitoleic, oleic, vaccenic, linoleic, alpha-linoleic, gamma-linoleic, arachidonic, gadoleic, arachidonic, eicosapentaenoic, docosahexaenoic, or erucic acid.
  • polymers may be polyesters, including copolymers comprising lactic acid and glycolic acid units, such as poly(lactic acid-co-glycolic acid) and poly(lactide-co-glycolide), collectively referred to herein as “PLGA”; and homopolymers comprising glycolic acid units, referred to herein as “PGA,” and lactic acid units, such as poly-L-lactic acid, poly-D-lactic acid, poly-D,L-lactic acid, poly-L-lactide, poly-D-lactide, and poly-D,L-lactide, collectively referred to herein as “PLA.”
  • exemplary polyesters include, for example, polyhydroxyacids; PEG copolymers and copolymers of lactide and glycolide (e.g., PLA-PEG copolymers, PGA-PEG copolymers, PLGA-PEG copolymers, and derivatives thereof.
  • polyesters include, for example, poly(caprolactone), poly(caprolactone)-PEG copolymers, poly(L-lactide-co-L-lysine), poly(serine ester), poly(4-hydroxy-L-proline ester), poly[ ⁇ -(4-aminobutyl)-L-glycolic acid], and derivatives thereof.
  • a polymer may be PLGA.
  • PLGA is a biocompatible and biodegradable co-polymer of lactic acid and glycolic acid, and various forms of PLGA are characterized by the ratio of lactic acid:glycolic acid.
  • Lactic acid can be L-lactic acid, D-lactic acid, or D,L-lactic acid.
  • the degradation rate of PLGA can be adjusted by altering the lactic acid:glycolic acid ratio.
  • PLGA to be used in accordance with the present invention is characterized by a lactic acid:glycolic acid ratio of approximately 85:15, approximately 75:25, approximately 60:40, approximately 50:50, approximately 40:60, approximately 25:75, or approximately 15:85.
  • polymers may be one or more acrylic polymers.
  • acrylic polymers include, for example, acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, aminoalkyl methacrylate copolymer, poly(acrylic acid), poly(methacrylic acid), methacrylic acid alkylamide copolymer, poly(methyl methacrylate), poly(methacrylic acid anhydride), methyl methacrylate, polymethacrylate, poly(methyl methacrylate) copolymer, polyacrylamide, aminoalkyl methacrylate copolymer, glycidyl methacrylate copolymers, polycyanoacrylates, and combinations comprising one or more of the foregoing polymers.
  • the acrylic polymer may comprise fully-polymerized copolymers of acrylic and methacrylic acid esters with a low content of quaternary ammoni
  • polymers can be cationic polymers.
  • cationic polymers are able to condense and/or protect negatively charged strands of nucleic acids.
  • Amine-containing polymers such as poly(lysine) (Zauner et al., 1998, Adv. Drug Del. Rev., 30:97; and Kabanov et al., 1995, Bioconjugate Chem., 6:7), poly(ethylene imine) (PEI; Boussif et al., 1995, Proc. Natl. Acad. Sci., USA, 1995, 92:7297), and poly(amidoamine) dendrimers (Kukowska-Latallo et al., 1996, Proc. Natl.
  • the synthetic nanocarriers may not comprise (or may exclude) cationic polymers.
  • polymers can be degradable polyesters bearing cationic side chains (Putnam et al., 1999, Macromolecules, 32:3658; Barrera et al., 1993, J. Am. Chem. Soc., 115:11010; Kwon et al., 1989, Macromolecules, 22:3250; Lim et al., 1999, J. Am. Chem. Soc., 121:5633; and Zhou et al., 1990, Macromolecules, 23:3399).
  • polyesters include poly(L-lactide-co-L-lysine) (Barrera et al., 1993, J. Am. Chem.
  • polymers can be linear or branched polymers. In some embodiments, polymers can be dendrimers. In some embodiments, polymers can be substantially cross-linked to one another. In some embodiments, polymers can be substantially free of cross-links. In some embodiments, polymers can be used in accordance with the present invention without undergoing a cross-linking step. It is further to be understood that the 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.
  • synthetic nanocarriers do 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).
  • Immunosuppressant can be, in some embodiments, coupled to synthetic nanocarriers, antibodies or antigen-binding fragments thereof (or other ligand that targets an APC), erythrocyte-binding peptides, etc.
  • Immunosuppressants include, but are not limited to, statins; mTOR inhibitors, such as rapamycin or a rapamycin analog; TGF- ⁇ signaling agents; TGF- ⁇ receptor agonists; histone deacetylase (HDAC) inhibitors; corticosteroids; inhibitors of mitochondrial function, such as rotenone; P38 inhibitors; NF- ⁇ inhibitors; adenosine receptor agonists; prostaglandin E2 agonists; phosphodiesterase inhibitors, such as phosphodiesterase 4 inhibitor; proteasome inhibitors; kinase inhibitors; G-protein coupled receptor agonists; G-protein coupled receptor antagonists; glucocorticoids; retinoids; cytokine inhibitors; cyto
  • Immunosuppressants also include IDO, vitamin D3, cyclosporine A, aryl hydrocarbon receptor inhibitors, resveratrol, azathiopurine, 6-mercaptopurine, aspirin, niflumic acid, estriol, tripolide, interleukins (e.g., IL-1, IL-10), cyclosporine A, siRNAs targeting cytokines or cytokine receptors and the like.
  • statins examples include atorvastatin (LIPITOR®, TORVAST®), cerivastatin, fluvastatin (LESCOL®, LESCOL® XL), lovastatin (MEVACOR®, ALTOCOR®, ALTOPREV®), mevastatin (COMPACTIN®), pitavastatin (LIVALO®, PIAVA®), rosuvastatin (PRAVACHOL®, SELEKTINE®, LIPOSTAT®), rosuvastatin (CRESTOR®), and simvastatin (ZOCOR®, LIPEX®).
  • atorvastatin LIPITOR®, TORVAST®
  • cerivastatin fluvastatin
  • fluvastatin LESCOL®, LESCOL® XL
  • lovastatin MEVACOR®, ALTOCOR®, ALTOPREV®
  • mevastatin COMPACTIN®
  • pitavastatin LIVALO®, PIAVA®
  • rosuvastatin
  • mTOR inhibitors include rapamycin and analogs thereof (e.g., CCL-779, RAD001, AP23573, C20-methallylrapamycin (C20-Marap), C16-(S)-butylsulfonamidorapamycin (C16-BSrap), C16-(S)-3-methylindolerapamycin (C16-iRap) (Bayle et al.
  • rapamycin and analogs thereof e.g., CCL-779, RAD001, AP23573, C20-methallylrapamycin (C20-Marap), C16-(S)-butylsulfonamidorapamycin (C16-BSrap), C16-(S)-3-methylindolerapamycin (C16-iRap) (Bayle et al.
  • TGF- ⁇ signaling agents include TGF- ⁇ ligands (e.g., activin A, GDF1, GDF11, bone morphogenic proteins, nodal, TGF- ⁇ s) and their receptors (e.g., ACVR1B, ACVR1C, ACVR2A, ACVR2B, BMPR2, BMPR1A, BMPR1B, TGF ⁇ RI, TGF ⁇ RII), R-SMADS/co-SMADS (e.g., SMAD1, SMAD2, SMAD3, SMAD4, SMAD5, SMAD8), and ligand inhibitors (e.g, follistatin, noggin, chordin, DAN, lefty, LTBP1, THBS1, Decorin).
  • TGF- ⁇ ligands e.g., activin A, GDF1, GDF11, bone morphogenic proteins, nodal, TGF- ⁇ s
  • their receptors e.g., ACVR1B, ACVR1C, ACVR
  • inhibitors of mitochondrial function include atractyloside (dipotassium salt), bongkrekic acid (triammonium salt), carbonyl cyanide m-chlorophenylhydrazone, carboxyatractyloside (e.g., from Atractylis gummifera ), CGP-37157, ( ⁇ )-Deguelin (e.g., from Mundulea sericea ), F16, hexokinase II VDAC binding domain peptide, oligomycin, rotenone, Ru360, SFK1, and valinomycin (e.g., from Streptomyces fulvissimus ) (EMD4Biosciences, USA).
  • atractyloside dipotassium salt
  • bongkrekic acid triammonium salt
  • carbonyl cyanide m-chlorophenylhydrazone e.g., from Atractylis gummifera
  • P38 inhibitors examples include SB-203580 (4-(4-Fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole), SB-239063 (trans-1-(4hydroxycyclohexyl)-4-(fluorophenyl)-5-(2-methoxy-pyrimidin-4-yl) imidazole), SB-220025 (5-(2amino-4-pyrimidinyl)-4-(4-fluorophenyl)-1-(4-piperidinyl)imidazole)), and ARRY-797.
  • NF (e.g., NK- ⁇ ) inhibitors include IFRD1, 2-(1,8-naphthyridin-2-yl)-Phenol, 5-aminosalicylic acid, BAY 11-7082, BAY 11-7085, CAPE (Caffeic Acid Phenethylester), diethylmaleate, IKK-2 Inhibitor IV, IMD 0354, lactacystin, MG-132 [Z-Leu-Leu-Leu-CHO], NF ⁇ B Activation Inhibitor III, NF- ⁇ B Activation Inhibitor II, JSH-23, parthenolide, Phenylarsine Oxide (PAO), PPM-18, pyrrolidinedithiocarbamic acid ammonium salt, QNZ, RO 106-9920, rocaglamide, rocaglamide AL, rocaglamide C, rocaglamide I, rocaglamide J, rocaglao
  • adenosine receptor agonists examples include CGS-21680 and ATL-146e.
  • prostaglandin E2 agonists examples include E-Prostanoid 2 and E-Prostanoid 4.
  • phosphodiesterase inhibitors examples include caffeine, aminophylline, IBMX (3-isobutyl-1-methylxanthine), paraxanthine, pentoxifylline, theobromine, theophylline, methylated xanthines, vinpocetine, EHNA (erythro-9-(2-hydroxy-3-nonyl)adenine), anagrelide, enoximone (PERFANTM), milrinone, levosimendon, mesembrine, ibudilast, piclamilast, luteolin, drotaverine, roflumilast (DAXASTM, DALIRESPTM), sildenafil (REVATION®, VIAGRA®), tadalafil (ADCIRCA®, CIALIS®), vardenafil (LEVITRA®, STAXYN®), udenafil, avanafil, icariin, 4-methylpiperaz
  • proteasome inhibitors examples include bortezomib, disulfiram, epigallocatechin-3-gallate, and salinosporamide A.
  • kinase inhibitors examples include bevacizumab, BIBW 2992, cetuximab (ERBITUX®), imatinib (GLEEVEC®), trastuzumab (HERCEPTIN®), gefitinib (IRESSA®), ranibizumab (LUCENTIS®), pegaptanib, sorafenib, dasatinib, sunitinib, erlotinib, nilotinib, lapatinib, panitumumab, vandetanib, E7080, pazopanib, and mubritinib.
  • glucocorticoids examples include hydrocortisone (cortisol), cortisone acetate, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, beclometasone, fludrocortisone acetate, deoxycorticosterone acetate (DOCA), and aldosterone.
  • retinoids examples include retinol, retinal, tretinoin (retinoic acid, RETIN-A®), isotretinoin (ACCUTANE®, AMNESTEEM®, CLARAVIS®, SOTRET®), alitretinoin (PANRETIN®), etretinate (TEGISONTM) and its metabolite acitretin (SORIATANE®), tazarotene (TAZORAC®, AVAGE®, ZORAC®), bexarotene (TARGRETIN®), and adapalene (DIFFERIN®).
  • retinoids include retinol, retinal, tretinoin (retinoic acid, RETIN-A®), isotretinoin (ACCUTANE®, AMNESTEEM®, CLARAVIS®, SOTRET®), alitretinoin (PANRETIN®), etretinate (TEGISONTM) and its metabolite acitretin (SORI
  • cytokine inhibitors include IL1ra, IL1 receptor antagonist, IGFBP, TNF-BF, uromodulin, Alpha-2-Macroglobulin, Cyclosporin A, Pentamidine, and Pentoxifylline (PENTOPAK®, PENTOXIL®, TRENTAL®).
  • peroxisome proliferator-activated receptor antagonists examples include GW9662, PPAR ⁇ antagonist III, G335, and T0070907 (EMD4Biosciences, USA).
  • peroxisome proliferator-activated receptor agonists examples include pioglitazone, ciglitazone, clofibrate, GW1929, GW7647, L-165,041, LY 171883, PPAR ⁇ activator, Fmoc-Leu, troglitazone, and WY-14643 (EMD4Biosciences, USA).
  • histone deacetylase inhibitors include hydroxamic acids (or hydroxamates) such as trichostatin A, cyclic tetrapeptides (such as trapoxin B) and depsipeptides, benzamides, electrophilic ketones, aliphatic acid compounds such as phenylbutyrate and valproic acid, hydroxamic acids such as vorinostat (SAHA), belinostat (PXD101), LAQ824, and panobinostat (LBH589), benzamides such as entinostat (MS-275), CI994, and mocetinostat (MGCD0103), nicotinamide, derivatives of NAD, dihydrocoumarin, naphthopyranone, and 2-hydroxynaphaldehydes.
  • hydroxamic acids such as trichostatin A, cyclic tetrapeptides (such as trapoxin B) and depsipeptides, benzamides, electrophilic keto
  • calcineurin inhibitors examples include cyclosporine, pimecrolimus, voclosporin, and tacrolimus.
  • phosphatase inhibitors examples include BN82002 hydrochloride, CP-91149, calyculin A, cantharidic acid, cantharidin, cypermethrin, ethyl-3,4-dephostatin, fostriecin sodium salt, MAZ51, methyl-3,4-dephostatin, NSC 95397, norcantharidin, okadaic acid ammonium salt from prorocentrum concavum, okadaic acid, okadaic acid potassium salt, okadaic acid sodium salt, phenylarsine oxide, various phosphatase inhibitor cocktails, protein phosphatase 1C, protein phosphatase 2A inhibitor protein, protein phosphatase 2A1, protein phosphatase 2A2, and sodium orthovanadate.
  • compositions according to the invention can comprise 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.
  • compositions are suspended in sterile saline solution for injection together with a preservative.
  • Viral transfer vectors can be made with methods known to those of ordinary skill in the art or as otherwise described herein.
  • viral transfer vectors can be constructed and/or purified using the methods set forth, for example, in U.S. Pat. No. 4,797,368 and Laughlin et al., Gene, 23, 65-73 (1983).
  • replication-deficient adenoviral vectors can be produced in complementing cell lines that provide gene functions not present in the replication-deficient adenoviral vectors, but required for viral propagation, at appropriate levels in order to generate high titers of viral transfer vector stock.
  • the complementing cell line can complement for a deficiency in at least one replication-essential gene function encoded by the early regions, late regions, viral packaging regions, virus-associated RNA regions, or combinations thereof, including all adenoviral functions (e.g., to enable propagation of adenoviral amplicons).
  • Complementing cell lines for producing adenoviral vectors include, but are not limited to, 293 cells (described in, e.g., Graham et al., J. Gen. Virol., 36, 59-72 (1977)), PER.C6 cells (described in, e.g., International Patent Application WO 97/00326, and U.S. Pat. Nos. 5,994,128 and 6,033,908), and 293-ORF6 cells (described in, e.g., International Patent Application WO 95/34671 and Brough et al., J. Virol., 71, 9206-9213 (1997)). In some instances, the complementing cell will not complement for all required adenoviral gene functions.
  • Helper viruses can be employed to provide the gene functions in trans that are not encoded by the cellular or adenoviral genomes to enable replication of the adenoviral vector.
  • Adenoviral vectors can be constructed, propagated, and/or purified using the materials and methods set forth, for example, in U.S. Pat. Nos. 5,965,358, 5,994,128, 6,033,908, 6,168,941, 6,329,200, 6,383,795, 6,440,728, 6,447,995, and 6,475,757, U.S. Patent Application Publication No.
  • Non-group C adenoviral vectors including adenoviral serotype 35 vectors, can be produced using the methods set forth in, for example, U.S. Pat. Nos. 5,837,511 and 5,849,561, and International Patent Applications WO 97/12986 and WO 98/53087.
  • AAV vectors may be produced using recombinant methods. Typically, the methods involve culturing a host cell which contains a nucleic acid sequence encoding an AAV capsid protein or fragment thereof; a functional rep gene; a recombinant AAV vector composed of AAV inverted terminal repeats (ITRs) and a transgene; and sufficient helper functions to permit packaging of the recombinant AAV vector into the AAV capsid proteins.
  • the viral transfer vector may comprise inverted terminal repeats (ITR) of AAV serotypes selected from the group consisting of: AAV1, AAV2, AAV5, AAV6, AAV6.2, AAV7, AAV8, AAV9, AAV10, AAV11 and variants thereof.
  • the components to be cultured in the host cell to package a rAAV vector in an AAV capsid may be provided to the host cell in trans.
  • any one or more of the required components e.g., recombinant AAV vector, rep sequences, cap sequences, and/or helper functions
  • a stable host cell which has been engineered to contain one or more of the required components using methods known to those of skill in the art.
  • such a stable host cell can contain the required component(s) under the control of an inducible promoter.
  • the required component(s) may be under the control of a constitutive promoter.
  • the recombinant AAV vector, rep sequences, cap sequences, and helper functions required for producing the rAAV of the invention may be delivered to the packaging host cell using any appropriate genetic element.
  • the selected genetic element may be delivered by any suitable method, including those described herein.
  • the methods used to construct any embodiment of this invention are known to those with skill in nucleic acid manipulation and include genetic engineering, recombinant engineering, and synthetic techniques. See, e.g., Sambrook et al, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, N.Y. Similarly, methods of generating rAAV virions are well known and the selection of a suitable method is not a limitation on the present invention. See, e.g., K. Fisher et al, J. Virol., 70:520-532 (1993) and U.S. Pat. No. 5,478,745.
  • recombinant AAV vectors may be produced using the triple transfection method (e.g., as described in detail in U.S. Pat. No. 6,001,650, the contents of which relating to the triple transfection method are incorporated herein by reference).
  • the recombinant AAVs are produced by transfecting a host cell with a recombinant AAV vector (comprising a transgene) to be packaged into AAV particles, an AAV helper function vector, and an accessory function vector.
  • an AAV helper function vector encodes AAV helper function sequences (rep and cap), which function in trans for productive AAV replication and encapsidation.
  • the AAV helper function vector supports efficient AAV vector production without generating any detectable wild-type AAV virions (i.e., AAV virions containing functional rep and cap genes).
  • the accessory function vector can encode nucleotide sequences for non-AAV derived viral and/or cellular functions upon which AAV is dependent for replication.
  • the accessory functions include those functions required for AAV replication, including, without limitation, those moieties involved in activation of AAV gene transcription, stage specific AAV mRNA splicing, AAV DNA replication, synthesis of cap expression products, and AAV capsid assembly.
  • Viral-based accessory functions can be derived from any of the known helper viruses such as adenovirus, herpesvirus (other than herpes simplex virus type-1), and vaccinia virus.
  • Lentiviral vectors may be produced using any of a number of methods known in the art. Examples of lentiviral vectors and/or methods of their production can be found, for example, in U.S. Publication Nos. 20150224209, 20150203870, 20140335607, 20140248306, 20090148936, and 20080254008, such lentiviral vectors and methods of production are incorporated herein by reference. As an example, when the lentiviral vector is integration-incompetent, the lentiviral genome further comprises an origin of replication (ori), whose sequence is dependent on the nature of cells where the lentiviral genome has to be expressed.
  • ori origin of replication
  • Said origin of replication may be from eukaryotic origin, preferably of mammalian origin, most preferably of human origin. Since the lentiviral genome does not integrate into the cell host genome (because of the defective integrase), the lentiviral genome can be lost in cells undergoing frequent cell divisions; this is particularly the case in immune cells, such as B or T cells. The presence of an origin of replication can be beneficial in some instances. Vector particles may be produced after transfection of appropriate cells, such as 293 T cells, by said plasmids, or by other processes.
  • all or some of the plasmids may be used to stably express their coding polynucleotides, or to transiently or semi-stably express their coding polynucleotides.
  • erythrocyte-binding peptides when preparing certain antigen-presenting cell targeted immunosuppressants, methods for attaching components to, for example, erythrocyte-binding peptides, antibodies or antigen-binding fragments thereof (or other ligand that targets an APC), or synthetic nanocarriers may be useful.
  • the attaching can be a covalent linker.
  • immunosuppressants according to the invention can be covalently attached to the external surface via a 1,2,3-triazole linker formed by the 1,3-dipolar cycloaddition reaction of azido groups with immunosuppressant containing an alkyne group or by the 1,3-dipolar cycloaddition reaction of alkynes with immunosuppressants 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.
  • 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 an immunosuppressant with the carboxylic acid group of a second component such as the nanocarrier.
  • the amide bond in the linker can be made using any of the conventional amide bond forming reactions with suitably protected amino acids and activated carboxylic acid such N-hydroxysuccinimide-activated ester.
  • a disulfide linker is made via the formation of a disulfide (S—S) bond between two sulfur atoms of the form, for instance, of R1-S—S—R2.
  • a disulfide bond can be formed by thiol exchange of a component containing thiol/mercaptan group (—SH) with another activated thiol group or a component containing thiol/mercaptan groups with a component containing activated thiol group.
  • a triazole linker specifically a 1,2,3-triazole of the form
  • R1 and R2 may be any chemical entities, is made by the 1,3-dipolar cycloaddition reaction of an azide attached to a first component with a terminal alkyne attached to a second component such as the immunosuppressant.
  • 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 thioether linker is made by the formation of a sulfur-carbon (thioether) bond in the form, for instance, of R1-S—R2.
  • Thioether can be made by either alkylation of a thiol/mercaptan (—SH) group on one component with an alkylating group such as halide or epoxide on a second component.
  • Thioether linkers can also be formed by Michael addition of a thiol/mercaptan group on one component to an electron-deficient alkene group on a second component containing a maleimide group or vinyl sulfone group as the Michael acceptor.
  • thioether linkers can be prepared by the radical thiol-ene reaction of a thiol/mercaptan group on one component with an alkene group on a second component.
  • a hydrazone linker is made by the reaction of a hydrazide group on one component with an aldehyde/ketone group on the second component.
  • a hydrazide linker is formed by the reaction of a hydrazine group on one component with a carboxylic acid group on the second component. Such 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 with an aldehyde or ketone group on the second component.
  • An urea or thiourea linker is prepared by the reaction of an amine group on one component with an isocyanate or thioisocyanate group on the second component.
  • An amidine linker is prepared by the reaction of an amine group on one component with an imidoester group on the second component.
  • An amine linker is made by the alkylation reaction of an amine group on one component with an alkylating group such as halide, epoxide, or sulfonate ester group on the second component.
  • an amine linker can also be made by reductive amination of an amine group on one component with an aldehyde or ketone group on the second component with a suitable reducing reagent such as sodium cyanoborohydride or sodium triacetoxyborohydride.
  • a sulfonamide linker is made by the reaction of an amine group on one component with a sulfonyl halide (such as sulfonyl chloride) group on the second component.
  • a sulfonyl halide such as sulfonyl chloride
  • a sulfone linker is made by Michael addition of a nucleophile to a vinyl sulfone.
  • Either the vinyl sulfone or the nucleophile may be on the surface of the nanocarrier or attached to a component.
  • the component can also be conjugated via non-covalent conjugation methods.
  • a negative charged immunosuppressant can be conjugated to a positive charged component through electrostatic adsorption.
  • a component containing a metal ligand can also be conjugated to a metal complex via a metal-ligand complex.
  • the component can be attached to a polymer, for example polylactic acid-block-polyethylene glycol, prior to the assembly of a synthetic nanocarrier or the synthetic nanocarrier can be formed with reactive or activatable groups on its surface.
  • the component may be prepared with a group which is compatible with the attachment chemistry that is presented by the synthetic nanocarriers' surface.
  • a peptide component can be attached to VLPs or liposomes using a suitable linker.
  • a linker is a compound or reagent that capable of coupling two molecules together.
  • the linker can be a homobifuntional or heterobifunctional reagent as described in Hermanson 2008.
  • an VLP or liposome synthetic nanocarrier containing a carboxylic group on the surface can be treated with a homobifunctional linker, adipic dihydrazide (ADH), in the presence of EDC to form the corresponding synthetic nanocarrier with the ADH linker.
  • ADH adipic dihydrazide
  • the resulting ADH linked synthetic nanocarrier is then conjugated with a peptide component containing an acid group via the other end of the ADH linker on nanocarrier to produce the corresponding VLP or liposome peptide conjugate.
  • a polymer containing an azide or alkyne group, terminal to the polymer chain is prepared.
  • This polymer is then used to prepare a synthetic nanocarrier in such a manner that a plurality of the alkyne or azide groups are positioned on the surface of that nanocarrier.
  • the synthetic nanocarrier can be prepared by another route, and subsequently functionalized with alkyne or azide groups.
  • the component is prepared with the presence of either an alkyne (if the polymer contains an azide) or an azide (if the polymer contains an alkyne) group.
  • the component is then allowed to react with the nanocarrier via the 1,3-dipolar cycloaddition reaction with or without a catalyst which covalently attaches the component to the particle through the 1,4-disubstituted 1,2,3-triazole linker.
  • the component is a small molecule it may be of advantage to attach the component to a polymer prior to the assembly of synthetic nanocarriers. In embodiments, it may also be an advantage to prepare the synthetic nanocarriers with surface groups that are used to attach the component to the synthetic nanocarrier through the use of these surface groups rather than attaching the component to a polymer and then using this polymer conjugate in the construction of synthetic nanocarriers.
  • the component can be attached by adsorption to a pre-formed synthetic nanocarrier or it can be attached 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 such 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.
  • Materials may be encapsulated into synthetic nanocarriers as desirable using a variety of methods including but not limited to C. Astete et al., “Synthesis and characterization of PLGA nanoparticles” J. Biomater. Sci. Polymer Edn, Vol. 17, No. 3, pp. 247-289 (2006); K. Avgoustakis “Pegylated Poly(Lactide) and Poly(Lactide-Co-Glycolide) Nanoparticles: Preparation, Properties and Possible Applications in Drug Delivery” Current Drug Delivery 1:321-333 (2004); C. Reis et al., “Nanoencapsulation I. Methods for preparation of drug-loaded polymeric nanoparticles” Nanomedicine 2:8-21 (2006); P.
  • synthetic nanocarriers are prepared by a nanoprecipitation process or spray drying. Conditions used in preparing synthetic nanocarriers may be altered to yield particles of a desired size or property (e.g., hydrophobicity, hydrophilicity, external morphology, “stickiness,” shape, etc.). The method of preparing the synthetic nanocarriers and the conditions (e.g., solvent, temperature, concentration, air flow rate, etc.) used may depend on the materials to be attached 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 attached to the synthetic nanocarriers and/or the composition of the polymer matrix.
  • synthetic nanocarriers prepared by any of the above methods have a size range outside of the desired range
  • synthetic nanocarriers can be sized, for example, using a sieve.
  • Elements of the synthetic nanocarriers may be attached to the overall synthetic nanocarrier, e.g., by one or more covalent bonds, or may be attached by means of one or more linkers. Additional methods of functionalizing synthetic nanocarriers may be adapted from Published US Patent Application 2006/0002852 to Saltzman et al., Published US Patent Application 2009/0028910 to DeSimone et al., or Published International Patent Application WO/2008/127532 A1 to Murthy et al.
  • synthetic nanocarriers can be attached to components directly or indirectly via non-covalent interactions.
  • the non-covalent attaching 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 attachments may be arranged to be on an external surface or an internal surface of a synthetic nanocarrier.
  • encapsulation and/or absorption is a form of attaching.
  • compositions provided herein 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-
  • compositions according to the invention may comprise 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, compositions are suspended in sterile saline solution for injection 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 of manufacture may require attention to the properties of the particular moieties being associated.
  • compositions are manufactured under sterile conditions or are terminally sterilized. This can ensure that resulting compositions are sterile and non-infectious, thus improving safety when compared to non-sterile compositions. This provides a valuable safety measure, especially when subjects receiving the compositions have immune defects, are suffering from infection, and/or are susceptible to infection.
  • Administration according to the present invention may be by a variety of routes, including but not limited to subcutaneous, intravenous, intramuscular and intraperitoneal routes.
  • the compositions referred to herein may be manufactured and prepared for administration, in some embodiments concomitant administration, using conventional methods.
  • compositions of the invention can be administered in effective amounts, such as the effective amounts described elsewhere herein.
  • the antigen-presenting cell targeted immunosuppressants and/or viral transfer vectors are present in dosage forms in an amount effective to attenuate an anti-viral transfer vector immune response or allow for readministration of a viral transfer vector to a subject.
  • the antigen-presenting cell targeted immunosuppressants and/or viral transfer vectors are present in dosage forms in an amount effective to escalate transgene expression in a subject.
  • the antigen-presenting cell targeted immunosuppressants and/or viral transfer vectors are present in dosage forms in an amount effective to reduce immune responses to the viral transfer vector, such as when concomitantly administered to a subject. Dosage forms may be administered at a variety of frequencies. In some embodiments, repeated administration of antigen-presenting cell targeted immunosuppressant with a viral transfer vector is undertaken.
  • a protocol can be determined by varying at least the frequency, dosage amount of the viral transfer vector and antigen-presenting cell targeted immunosuppressant and subsequently assessing a desired or undesired immune response.
  • a preferred protocol for practice of the invention reduces an immune response against the viral transfer vector, attenuates an anti-viral transfer vector response and/or escalates transgene expression.
  • the protocol comprises at least the frequency of the administration and doses of the viral transfer vector and antigen-presenting cell targeted immunosuppressant.
  • PLGA polymer with acid end group 7525 DLG1A, acid number 0.46 mmol/g, Lakeshore Biomaterials; 5 g, 2.3 mmol, 1.0 eq
  • DCM dichloromethane
  • N,N-Dicyclohexylcarbodimide 1.2 eq, 2.8 mmol, 0.57 g
  • rapamycin 1.0 eq, 2.3 mmol, 2.1 g
  • DMAP 4-dimethylaminopyridine
  • the filtrate is concentrated to ca. 10 mL in volume and added to 100 mL of isopropyl alcohol (IPA) to precipitate out the PLGA-rapamycin conjugate.
  • IPA isopropyl alcohol
  • the IPA layer is removed and the polymer is then washed with 50 mL of IPA and 50 mL of methyl t-butyl ether (MTBE).
  • MTBE methyl t-butyl ether
  • the polymer is then dried under vacuum at 35 C for 2 days to give PLGA-rapamycin as a white solid (ca. 6.5 g).
  • Nanocarrier containing PLGA-rapamycin is prepared as follows:
  • Solution 1 PLGA-rapamycin @ 100 mg/mL in methylene chloride. The solution is prepared by dissolving PLGA-rapamycin in pure methylene chloride.
  • Solution 2 PLA-PEG @ 100 mg/mL in methylene chloride. The solution is prepared by dissolving PLA-PEG in pure methylene chloride.
  • Solution 3 Polyvinyl alcohol @ 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • a primary water-in-oil emulsion is prepared first.
  • W1/O1 is prepared by combining solution 1 (0.75 mL), and solution 2 (0.25 mL) in a small pressure tube and sonicating at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary emulsion (W1/O1/W2) is then prepared by combining solution 3 (3.0 mL) with the primary W1/O1 emulsion, vortexing for 10 s, and sonicating at 30% amplitude for 60 seconds using the Branson Digital Sonifier 250.
  • the W1/O1/W2 emulsion is added to a beaker containing 70 mM pH 8 phosphate buffer solution (30 mL) and stirred at room temperature for 2 hours to allow the methylene chloride to evaporate and for the nanocarriers to form.
  • a portion of the nanocarriers is washed by transferring the nanocarrier suspension to a centrifuge tube and centrifuging at 75,600 ⁇ g and 4° C. for 35 min, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. The washing procedure is repeated, and the pellet is re-suspended in phosphate buffered saline for a final nanocarrier dispersion of about 10 mg/mL.
  • a solution of 150 ⁇ l of HS-PEG-rapamycin (10 ⁇ M in 10 mM pH 9.0 carbonate buffer) is added to 1 mL of 20 nm diameter citrate-capped gold nanocarriers (1.16 nM) to produce a molar ratio of thiol to gold of 2500:1.
  • the mixture is stirred at room temperature under argon for 1 hour to allow complete exchange of thiol with citrate on the gold nanocarriers.
  • the AuNCs with PEG-rapamycin on the surface is then purified by centrifuge at 12,000 g for 30 minutes. The supernatant is decanted and the pellet containing AuNC-S-PEG-rapamycin is then pellet washed with 1 ⁇ PBS buffer.
  • the purified Gold-PEG-rapamycin nanocarriers are then resuspend in suitable buffer for further analysis and bioassays.
  • Mesoporous SiO2 nanoparticle cores are created through a sol-gel process.
  • Hexadecyltrimethyl-ammonium bromide (CTAB) 0.5 g is dissolved in deionized water (500 mL), and then 2 M aqueous NaOH solution (3.5 mL) is added to the CTAB solution.
  • the solution is stirred for 30 min, and then Tetraethoxysilane (TEOS) (2.5 mL) is added to the solution.
  • TEOS Tetraethoxysilane
  • the resulting gel is stirred for 3 h at a temperature of 80° C.
  • the white precipitate which forms is captured by filtration, followed by washing with deionized water and drying at room temperature.
  • the remaining surfactant is then extracted from the particles by suspension in an ethanolic solution of HCl overnight.
  • the particles are washed with ethanol, centrifuged, and redispersed under ultrasonication. This wash procedure is repeated two additional times.
  • the SiO2 nanoparticles are then functionalized with amino groups using (3-aminopropyl)-triethoxysilane (APTMS).
  • APTMS (3-aminopropyl)-triethoxysilane
  • the particles are suspended in ethanol (30 mL), and APTMS (50 ⁇ L) is added to the suspension.
  • the suspension is allowed to stand at room temperature for 2 h and then is boiled for 4 h, keeping the volume constant by periodically adding ethanol. Remaining reactants are removed by five cycles of washing by centrifugation and redispersing in pure ethanol.
  • a separate reaction 1-4 nm diameter gold seeds are created. All water used in this reaction is first deionized and then distilled from glass. Water (45.5 mL) is added to a 100 mL round-bottom flask. While stirring, 0.2 M aqueous NaOH (1.5 mL) is added, followed by a 1% aqueous solution of tetrakis(hydroxymethyl)phosphonium chloride (THPC) (1.0 mL). Two minutes after the addition of THPC solution, a 10 mg/mL aqueous solution of chloroauric acid (2 mL), which has been aged at least 15 min, is added. The gold seeds are purified through dialysis against water.
  • THPC tetrakis(hydroxymethyl)phosphonium chloride
  • the amino-functionalized SiO2 nanoparticles formed above are first mixed with the gold seeds for 2 h at room temperature.
  • the gold-decorated SiO2 particles are collected through centrifugation and mixed with an aqueous solution of chloroauric acid and potassium bicarbonate to form the gold shell.
  • the particles are then washed by centrifugation and redispersed in water.
  • Ibuprofen is loaded by suspending the particles in a solution of sodium ibuprofen (1 mg/L) for 72 h. Free ibuprofen is then washed from the particles by centrifugation and redispersing in water.
  • the liposomes are formed using thin film hydration.
  • 1,2-Dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) 32 ⁇ mol
  • cholesterol 32 ⁇ mol
  • cyclosporin A 6.4 ⁇ mol
  • This lipid solution is added to a 50 mL round-bottom flask, and the solvent is evaporated on a rotary evaporator at a temperature of 60° C.
  • the flask is then flushed with nitrogen gas to remove remaining solvent.
  • Phosphate buffered saline (2 mL) and five glass beads are added to the flask, and the lipid film is hydrated by shaking at 60° C.
  • the suspension is transferred to a small pressure tube and sonicated at 60° C. for four cycles of 30 s pulses with a 30 s delay between each pulse.
  • the suspension is then left undisturbed at room temperature for 2 h to allow for complete hydration.
  • the liposomes are washed by centrifugation followed by resuspension in fresh phosphate buffered saline.
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Catalogue # R1017).
  • PLGA with 76% lactide and 24% glycolide content and an inherent viscosity of 0.69 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211.
  • PLA-PEG block co-polymer with a PEG block of approximately 5,000 Da and PLA block of approximately 40,000 Da was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 100 DL mPEG 5000 5CE).
  • Polyvinyl alcohol (85-89% hydrolyzed) was purchased from EMD Chemicals (Product Number 1.41350.1001).
  • Solution 1 PLGA at 75 mg/mL and PLA-PEG at 25 mg/mL in methylene chloride. The solution was prepared by dissolving PLGA and PLA-PEG in pure methylene chloride.
  • Solution 2 Rapamycin at 100 mg/mL in methylene chloride. The solution was prepared by dissolving rapamycin in pure methylene chloride.
  • Solution 3 Polyvinyl alcohol at 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • the O/W emulsion was prepared by combining solution 1 (1 mL), solution 2 (0.1 mL), and solution 3 (3 mL) in a small pressure tube and sonicating at 30% amplitude for 60 seconds using a Branson Digital Sonifier 250.
  • the O/W emulsion was added to a beaker containing 70 mM pH 8 phosphate buffer solution (30 mL) and stirred at room temperature for 2 hours to allow the methylene chloride to evaporate and for the nanocarriers to form.
  • a portion of the nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube and centrifuging at 75,000 ⁇ g and 4° C.
  • Nanocarrier size was determined by dynamic light scattering.
  • the amount rapamycin in the nanocarrier was determined by HPLC analysis.
  • the total dry-nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • GSK1059615 was purchased from MedChem Express (11 Deer Park Drive, Suite 102D Monmouth Junction, N.J. 08852), product code HY-12036.
  • PLGA with a lactide:glycolide ratio of 1:1 and an inherent viscosity of 0.24 dL/g was purchased from Lakeshore Biomaterials (756 Tom Martin Drive, Birmingham, Ala. 35211), product code 5050 DLG 2.5A.
  • Cellgro phosphate buffered saline 1 ⁇ pH 7.4 (PBS 1 ⁇ ) was purchased from Corning (9345 Discovery Blvd. Manassas, Va. 20109), product code 21-040-CV.
  • Solution 1 PLGA (125 mg), and PLA-PEG-OMe (125 mg), were dissolved in 10 mL of acetone.
  • Solution 2 GSK1059615 was prepared at 10 mg in 1 mL of N-methyl-2-pyrrolidinone (NMP).
  • Nanocarriers were prepared by combining Solution 1 (4 mL) and Solution 2 (0.25 mL) in a small glass pressure tube and adding the mixture drop wise to a 250 mL round bottom flask containing 20 mL of ultra-pure water under stirring. The flask was mounted onto a rotary evaporation device, and the acetone was removed under reduced pressure. A portion of the nanocarriers was washed by transferring the nanocarrier suspension to centrifuge tubes and centrifuging at 75,600 rcf and 4° C. for 50 minutes, removing the supernatant, and re-suspending the pellet in PBS 1 ⁇ .
  • the washing procedure was repeated, and the pellet was re-suspended in PBS 1 ⁇ to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis.
  • the washed nanocarrier solution was then filtered using 1.2 ⁇ m PES membrane syringe filters from Pall, part number 4656.
  • An identical nanocarrier solution was prepared as above, and pooled with the first after the filtration step. The homogenous suspension was stored frozen at ⁇ 20° C.
  • Nanocarrier size was determined by dynamic light scattering.
  • the amount of GSK1059615 in the nanocarrier was determined by UV absorption at 351 nm.
  • the total dry-nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • An erythrocyte-binding therapeutic is prepared based on the teachings of U.S. Publication No. 20120039989 and used as an antigen-presenting cell targeted immunosuppressant.
  • the erythrocyte-binding therapeutic may comprise any one of ERY1, ERY19, ERY59, ERY64, ERY123, ERY141 and ERY162 and any one of the viral transfer vector antigens described herein, such as a viral vector antigen, e.g., a capsid protein (or peptide antigen derived therefrom), or a protein (or peptide antigen derived therefrom), such as a therapeutic protein (or peptide antigen derived therefrom) that is encoded by a transgene as described herein.
  • a viral vector antigen e.g., a capsid protein (or peptide antigen derived therefrom), or a protein (or peptide antigen derived therefrom), such as a therapeutic protein (or peptide antigen derived therefrom
  • An antigen-presenting cell targeted immunosuppressant is prepared according to the teachings of U.S. Publication No. 20100151000.
  • the particle may be a liposome or polymeric particle and comprises any one of the immunosuppressants provided herein or any one of the inhibitors of the NF-kB pathway provided in U.S. Publication No. 20100151000, which inhibitors are incorporated herein by reference in their entirety.
  • the liposome or polymeric particle may further comprise any one of the viral transfer vector antigens described herein, such as a viral vector antigen, e.g., a capsid protein (or peptide antigen derived therefrom), or a protein (or peptide antigen derived therefrom), such as a therapeutic protein (or peptide antigen derived therefrom), that is encoded by a transgene as described herein.
  • a viral vector antigen e.g., a capsid protein (or peptide antigen derived therefrom), or a protein (or peptide antigen derived therefrom), such as a therapeutic protein (or peptide antigen derived therefrom), that is encoded by a transgene as described herein.
  • An adenoviral transfer vector is generated according to the methods provided in U.S. Patent Publication 2004/0005293.
  • Such a vector may comprise any one of the transgenes as provided herein.
  • an Ad-AAT-hFVIII vector that expresses the human B-domain deleted FVIII cDNA from the human alfa1-antitrypsin promoter (AAT) is prepared.
  • An HPRT stuffer fragment is employed to optimize vector size and to avoid vector rearrangements (Parks R J, Graham F L. A helper-dependent system for adenovirus vector production helps define a lower limit for efficient DNA packaging. J Virol. 1997, 71:3293-3298).
  • the Cre66 packaging cell line is used.
  • the viral transfer vector of any one of the Examples, such as Example 9, is administered concomitantly, such as on the same day, as any one of the antigen-presenting cell targeted immunosuppressants provided herein, such as in Examples 1-8 or 12, to subjects recruited for a clinical trial.
  • One or more immune responses against the viral transfer vector is evaluated.
  • the level(s) of the one or more immune responses against the viral transfer vector can be evaluated by comparison with the level(s) of the one or more immune responses in the subjects, or another group of subjects, administered the viral transfer vector in the absence of the antigen-presenting cell targeted immunosuppressant, such as when administered the viral transfer vector alone.
  • repeated concomitant administration is evaluated in a similar manner.
  • the viral transfer vector and antigen-presenting cell targeted immunosuppressant can be administered concomitantly to subjects in need of viral transfer vectors when such subjects are expected to have an undesired immune response against the viral transfer vector when not administered concomitantly with the antigen-presenting cell targeted immunosuppressant.
  • a protocol using the information established during the trials can be prepared to guide the concomitant dosing of the viral transfer vector and synthetic nanocarriers of subjects in need of treatment with a viral transfer vector and have or are expected to have an undesired immune response against the viral transfer vector without the benefit of the antigen-presenting cell targeted immunosuppressant. The protocol so prepared can then be used to treat subjects, particularly human subjects.
  • AAV-GFP adeno-associated virus expressing recombinant green fluorescent protein
  • NCs nanocarrier-encapsulated immunosuppressant
  • Tissue samples were then cut into ⁇ 2 mm squares, digested (collagenase, 400 U) with repeated agitation, filtered (nylon mesh), spun down (1,500 rpm), and pellets re-suspended in ice-cold 2% FBS.
  • VG viral genomes
  • mice were bled at d14 and their serum tested for the presence of antibodies to AAV.
  • all mice had been injected with AAV-GFP once with or without co-administration of NCs (resulting in two groups of 15 mice each).
  • mice were boosted with AAV-GFP at day 21, then antibody levels in untreated mice continued to grow significantly while being blunted in those mice that received NCs only at boost ( FIGS. 6 and 7 ).
  • 8/10 mice treated with NCs at prime had no detectable antibodies at d33 even after d21 boost ( FIGS. 6 and 7 ).
  • Application of NCs at AAV boost may have had a minor effect in blocking generation of antibodies to AAV although at this point it was not statistically significant from no NCs treatment at boost ( FIGS. 6 and 7 ).
  • NCs treatment at prime appears to be important for blocking the development of antibodies to AAV with its administration at later time-points also being beneficial.
  • PLA with an inherent viscosity of 0.41 dL/g was purchased from Lakeshore Biomaterials (756 Tom Martin Drive, Birmingham, Ala. 35211), product code 100 DL 4A.
  • Rapamycin was purchased from Concord Biotech Limited (1482-1486 Trasad Road, Dholka 382225, Ahmedabad India), product code SIROLIMUS.
  • Sorbitan monopalmitate was purchased from Sigma-Aldrich (3050 Spruce St., St. Louis, Mo. 63103), product code 388920.
  • EMPROVE® Polyvinyl Alcohol (PVA) 4-88, USP (85-89% hydrolyzed, viscosity of 3.4-4.6 mPa ⁇ S) was purchased from EMD Chemicals Inc. (480 South Democrat Road Gibbstown, N.J. 08027), product code 1.41350.
  • Dulbecco's phosphate buffered saline 1 ⁇ was purchased from Lonza (Muenchensteinerstrasse 38, CH-4002 Basel, Switzerland), product code 17-512Q.
  • a polymer, rapamycin, and sorbitan monopalmitate mixture was prepared by dissolving PLA at 37.5 mg/mL, PLA-PEG-Ome at 12.5 mg/mL, rapamycin at 8 mg/mL, and sorbitan monopalmitate at 2.5 in dichloromethane.
  • Polyvinyl alcohol was prepared at 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • An O/W emulsion was prepared by combining Solution 1 (1.0 mL) and Solution 2 (3 mL) in a small glass pressure tube, vortex mixed for 10 seconds. The formulation was then homogenized by sonication at 30% amplitude for 1 minute. The emulsion was then added to an open beaker containing DPBS (30 mL). A second O/W emulsion was prepared using the same materials and method as above and then added to the same beaker containing the first emulsion and DPBS. The combined emulsion was then stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and for the nanocarriers to form.
  • nanocarriers were washed by transferring the nanocarrier suspension to a centrifuge tube and centrifuging at 75,600 ⁇ g and 4° C. for 50 minutes, removing the supernatant, and re-suspending the pellet in DPBS containing 0.25% w/v PVA.
  • the wash procedure was repeated and then the pellet was re-suspended in DPBS containing 0.25% w/v PVA to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis.
  • the nanocarrier suspension was then filtered using a 0.22 ⁇ m PES membrane syringe filter (Millipore part number SLGP033RB). The filtered nanocarrier suspension was then stored at ⁇ 20° C.
  • Nanocarrier size was determined by dynamic light scattering.
  • the amount of rapamycin in the nanocarrier was determined by HPLC analysis.
  • the total dry-nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • AAV-GFP recombinant green fluorescent protein
  • mice Male C57BL/6 mice were used (5-15 mice/group). Animals were injected i.v. with 200 ⁇ L of AAV8-GFP or an admixture of AAV8-GFP+NCs, a PLGA nanocarrier containing rapamycin (see Table 2 below). On day 14 after treatment, all animals were bled and their sera analyzed for antibodies against AAV8 by ELISA. Briefly, 96-well plates were coated with 50 ⁇ L of AAV8 at 2 ⁇ 10 9 vector genomes (vg)/mL in carbonate buffer for 92 hours, and then blocked for 2 hours with 300 ⁇ L of casein.
  • mice Male C57BL/6 mice were used (5 mice/group). Animals were injected with 200 ⁇ L of AAV8-GFP (Virovek, Hayward, Calif.) or an admixture of AAV8-GFP+NCs (as produced in Example 12) on day 0 and/or day 21 as indicated in Table 3. Sera were collected on day 0 and 33 and analyzed for anti-AAV8 antibody levels by ELISA as described above.
  • mice injected with AAV8-GFP on day 0 in the absence of NCs showed a robust anti-AAV8 antibody response that increased significantly after the second injection of AAV8-GFP on day 21 ( FIG. 10 ).
  • NCs was concomitantly administered with the AAV8-GFP on day 21, the antibody response on average was significantly blunted.
  • two mice in this latter group which were antibody positive on d14 had no detectable levels of antibodies to AAV8 on d33 ( FIG. 10 ).
  • anti-AAV8 antibody titers increased in 2 other mice.
  • NCs concomitantly administered at the time of the first AAV8-GFP injection (day 0) completely inhibited the anti-AAV8 antibody response at day 14.
  • the anti-AAV8 antibody was also inhibited in 4 of 5 mice at day 33 after a second administration of AAV8-GFP alone on day 21. Concomitant administration of NCs at both day 0 and 21 showed a similar trend. Thus, NCs treatment at the time of the first administration of AAV is important for blocking the development of antibodies to AAV. Additional administration of NCs upon repeat dosing of AAV may be potentially beneficial.
  • NCs nanocarrier-encapsulated immunosuppressant
  • mice Male C57BL/6 mice were used (5 mice/group). Animals were injected with 200 ⁇ L of AAV8-GFP in the absence of NCs on day 0. One group of animals received no further treatment, while other groups received a second dose of AAV8-GFP on day 21 with or without concomitant administration of NCs carrying 50 ⁇ g rapamycin (see Table 4 below). At d33 after the first injection (12 days after the second injection for those groups that were injected twice) animals were sacrificed, their livers treated with collagenase 4 (Worthington, Lakewood, N.J.), meshed and total cell suspensions were analyzed by flow cytometry for GFP expression. Briefly, tissue was initially perfused with collagenase (100 U) and incubated at 37° C.
  • results demonstrate the benefit of administering synthetic nanocarriers coupled to an immunosuppressant in conjunction with a viral transfer vector for maintaining expression of the vector transgene.
  • Such benefits were seen with concomitant administration of synthetic nanocarriers coupled to an immunosuppressant in conjunction with a viral transfer vector comprising a transgene encoding a protein for expression.
  • mice Male C57BL/6 mice were used (5 mice/group). Animals were injected with 200 ⁇ L of AAV-red fluorescence protein (RFP) (Virovek, Hayward, Calif.) on day 0 (groups 1-5) and/or day 21 (groups 1-4, 6) (see Table 5 below). NCs carrying 50 ⁇ g rapamycin was concomitantly administered on day 0 (groups 2, 4) and/or day 21 (groups 3, 4). At d33 after the first injection (12 days after the second injection for those groups that were injected twice) animals were sacrificed, their livers treated with collagenase 4 (Worthington, Lakewood, N.J.), meshed and total cell suspensions were analyzed by flow cytometry for RFP expression.
  • RFP AAV-red fluorescence protein
  • tissue was initially perfused with collagenase (100 U) and incubated at 37° C. for 30 min. The collagenase supernatant was removed and quenched with 2% FBS. Tissue samples were then cut into ⁇ 2 mm squares, digested (collagenase, 400 U) with repeated agitation, filtered (nylon mesh), spun down (1,500 rpm), and pellets re-suspended in ice-cold 2% FBS.
  • mice administered one or two injections of AAV8-RFP in the absence of NCs showed similar low levels of RFP expression at day 33 ( FIG. 12 ).
  • Mice treated with NCs concomitantly at the time of the first injection of AAV8-RFP showed a trend towards increased expression of RFP that was not statistically significant.
  • mice that were treated concomitantly with NCs at the time of the second injection of AAV8-RFP showed a statistically significant increase in RFP expression.
  • Mice that were treated with NCs at both day 0 and day 21 also showed a significant increase in RFP expression compared to control animals that received AAV8-RFP on days 0 and 21 in the absence of NCs.
  • results demonstrate the benefit of administering synthetic nanocarriers coupled to an immunosuppressant in conjunction with a viral transfer vector for enhancing expression of the vector transgene.
  • Such benefits were seen with concomitant administration of synthetic nanocarriers coupled to an immunosuppressant in conjunction with a viral transfer vector comprising a transgene encoding a protein for expression.
  • AAV-GFP adeno-associated virus expressing recombinant green fluorescent protein
  • CTL cytolytic T cell
  • NCs nanocarrier-encapsulated immunosuppressant
  • CFSE high cells were incubated with 1 ⁇ g/mL dominant MHC class I-binding peptide from AAV capsid (sequence NSLANPGIA, amino acids 517-525) and dominant MHC class I peptide from GFP (HYLSTQSAL, aa 200-208) at 37° C. for 1 h, while CFSE low cells were incubated in medium alone.
  • the control CFSE low cells and peptide-pulsed CFSE high target cells were mixed in a 1:1 ratio (2.0 ⁇ 10 7 cells total) and injected i.v. Eighteen hours after the injection of labeled cells, spleens were harvested, processed and analyzed by flow cytometry.
  • mice injected at days 0 and 17 were assayed for antigen-specific IFN- ⁇ production on d25 after the first injection (7 days after the second injection). Briefly, splenocytes were isolated, plated in wells with pre-absorbed anti-IFN- ⁇ antibody and re-stimulated with AAV capsid or GFP peptides (1 ⁇ g/mL) for 7 days in vitro. ELISpots were developed by biotinylated anti-IFN- ⁇ antibody and streptavidin-HRP, and spots were counted. Nonspecific background was subtracted.
  • mice concomitantly treated with NCs showed a significant reduction in antigen-specific IFN- ⁇ -producing cells compared to the non-NCs-treated group ( FIGS. 14 and 15 ).
  • 4/6 mice demonstrated a recall response to the AAV capsid protein at 250,000 cells per well density, while no (0/6) mice responded to this peptide in the NCs-treated group ( FIG. 14 , p ⁇ 0.05).
  • mice Male C57BL/6 mice were used (5 mice/group). Animals were injected i.v. with 10 10 vg of rAAV2/8-luciferase (rAAV2/8-Luc)) (produced in a manner similar to the methods provided herein such as in Example 21 or 22) or rAAV2/8-Luc+synthetic nanocarriers containing 100 ⁇ g rapamycin (NCs) on Day 0 (see Table 7 below). On day 14, all animals received an i.v. injection of 10 10 vg of rAAV2/8 encoding human factor IX (hFIX) (rAAV2/8-hFIX)) (produced in a manner similar to the methods provided herein such as in Example 21 or 22). Sera were collected at various time points and assayed for anti-AAV antibody levels and hFIX protein levels by ELISA.
  • FIG. 16 illustrates the protocol and timing of administration of synthetic nanocarriers comprising an immunosuppressant.
  • the data show that a single administration of synthetic nanocarriers comprising immunosuppressant concomitantly administered with AAV8-Luc can prevent or delay the onset of anti-AAV8 antibodies ( FIG.
  • the experimental design is shown in FIG. 18 .
  • Male C57BL/6 mice were used (5 mice/group).
  • Mice were then challenged with rAAV2/8-hFIX) (produced in a manner similar to the methods provided herein such as in Example 21 or 22) concomitantly administered with synthetic nanocarriers containing rapamycin (100 ⁇ g rapamycin) on day 21.
  • the control group received empty NP instead of NCs on days 0 and 21.
  • FIG. 20 The experimental design is shown in FIG. 20 .
  • Synthetic nanocarriers comprising immunosuppressant (100 ⁇ g rapamycin) or control empty nanoparticles were administered i.v. concurrently with rAAV2/8-Luc vector) (produced in a manner similar to the methods provided herein such as in Example 21 or 22) (1 ⁇ 10 11 vg/mouse) on Day 0.
  • mice received either an i.v. injection of rAAV5-hFIX) (produced in a manner similar to the methods provided herein such as in Example 21 or 22) (1 ⁇ 10 11 vg/mouse) or an i.m. injection of human Factor IX (hFIX) protein emulsified in complete Freund's adjuvant (CFA) (Table 10).
  • hFIX human Factor IX
  • Nanoparticles (10 11 vg) (10 11 vg) CFA N 1 NCs, d 0 d 0 d 21 — 5 2 Empty NP, d 0 d 0 d 21 — 5 3 NCs, d 0 d 0 — d 21 5 4 Empty NP, d 0 d 0 — d 21 5
  • mice that received AAV2/8-FIX on day 21 showed little or no anti-AAV8 antibodies.
  • AAV8 the effect of the NCs treatment on inhibiting anti-AAV antibody responses were specific to AAV serotype with which it was co-administered (i.e., AAV8) and does not render the mice chronically immunosuppressed.
  • mice concomitantly treated with NCs and rAAV2/8-Luc on day 0 showed a robust response to immunization with recombinant hFIX protein in complete Freund's adjuvant (CFA) on day 21 ( FIG. 21 , right panel).
  • CFA complete Freund's adjuvant
  • ART-I02 is produced as described previously (Matsushita T, et al. Gene Ther. 1998; 5: 938-945).
  • the plasmid encodes any one of the transgenes provided herein under the control of the NF- ⁇ B promoter and a human growth hormone polyadenylation signal.
  • the gene of interest may also be under the control of the cytomegalovirus (CMV) promoter.
  • CMV cytomegalovirus
  • the transgene cassette is flanked by AAV-2 inverted terminal repeats and is packaged in capsid from AAV5 as described in Gao G P, et al. Proc Natl Acad Sci USA. 2002; 99: 11854-11859.
  • the vector is purified by combined chromatography and cesium chloride density gradient centrifugation, resulting in empty capsid-free fractions.
  • Vector titers can be determined by qPCR using specific primers and probe.
  • a rAAV5 vector can be produced coding for Firefly Luciferase.
  • An scAAV backbone plasmid is constructed by ligating MscIBsaI and BsaITsp45I fragments from AAV2-HCR-hAAT-FIX to the simian virus 40 late polyA (SV40 LpA).
  • the resulting plasmid contains the modified AAV2 backbone with an intact 5′ terminal resolution site (trs) and a deleted 3′ trs.
  • the LP1 enhancer/promoter can be constructed using standard polymerase chain reaction (PCR) methods with amplification of consecutive segments of the human apolipoprotein hepatic control region (HCR), the human alpha1antitrypsin (hAAT) gene promoter including the 5′ untranslated region and cloned upstream of a modified SV40 small t antigen intron (SV40 intron) modified at positions 4582 (g to c), 4580 (g to c), 4578 (a to c), and 4561 (a to t) into the modified AAV2 backbone.
  • PCR polymerase chain reaction
  • the wild-type hFIX cDNA, or other cDNA of interest, without the 3′ untranslated region (UTR) regions is PCR amplified from AAV-HCR-hAAT-hFIX and inserted downstream of the modified SV40 intron to make scAAV-LP1-hFIX.
  • a codon-optimized hFIX is generated using codons most frequently found in highly expressed eukaryotic genes, synthesized as oligonucleotides, and subsequently assembled by ligation, PCR amplified, and sequenced prior to cloning into the AAVLP1 backbone to create sc-AAV-LP1-hFIXco.
  • ss and scAAV vectors are made by the adenovirus-free transient transfection method.
  • AAV5-pseudotyped vector particles are generated using a chimeric AAV2 Rep-5Cap packaging plasmid called pLT-RCO3, which is based on XX2 and pAAV5-2. Additionally, AAV8-pseudotyped vectors are made using the packaging plasmid pAAV8-2. AAV2/5 and 2/8 vectors are purified by the ion exchange chromatography method. Vector genome (vg) titers can be determined by quantitative slotblot using supercoiled plasmid DNA as standards. Such a viral vector can comprise any one of the transgenes as provided herein.
  • a mouse genomic Alb segment (90474003-90476720 in NCBI reference sequence: NC — 000071.6) can be PCR-amplified and inserted between AAV2 inverted terminal repeats into BsrGI and SpeI restriction sites in a modified pTRUF backbone.
  • An optimized P2A coding sequence preceded by a linker coding sequence (glycine-serine-glycine) and followed by anNheI restriction site can be into the Bpu10I restriction site.
  • a codon-optimized F9 coding sequence can be inserted into the NheI site to get pAB269 that can serve in the construction of the rAAV8 vector.
  • an internal segment from the BsiWI restriction site to the NheI restriction site can be amplified using appropriate PCR primers.
  • Final rAAV production plasmids can be generated using an EndoFree Plasmid Megaprep Kit (Qiagen).
  • rAAV8 vectors can be produced as described in Grimm, et al., J. Virol. 80, 426-439 (2006) using a Ca 3 (PO 4 ) 2 transfection protocol followed by CsCl gradient purification. Vectors can be titred by quantitative dot blot.
  • Adenoviral constructs using a “first-generation” E1/E3-deleted replication-deficient adenovirus can be produced as described in Kypson, et al. J Thorac Cardiovasc Surg. 1998 and Akhter, et al. Proc Natl Acad Sci USA. 1997; 94:12100-12105.
  • the b 2 AR construct (Adeno-b 2 AR) and a transgene can be driven by an appropriate promoter.
  • Large-scale preparations of adenoviruses can be purified from infected Epstein-Barr nuclear antigen-transfected 293 cells.
  • a lentiviral expression plasmid containing a packaging sequence and a transgene inserted between the lentiviral LTRs to allow target cell integration; a packaging plasmid, encoding the pol, gag, rev and tat viral genes and containing the rev-response element; and a pseudotyping plasmid, encoding a protein, of a virus envelope gene.
  • HEK 293T cells can be transfected by the foregoing. After transfection of HEK 293T cells, the lentiviral vectors can be obtained from the cell supernatant which contains recombinant lentiviral vectors.
  • HIV Lentiviral Transfer Vector with a Gene Therapy Transgene (Prophetic) HIV Lentiviral Transfer Vector with a Gene Therapy Transgene (Prophetic)
  • An HIV lentiviral transfer vector is prepared according the methods of U.S. Publication No. 20150056696.
  • An hPEDF CDS fragment is PCR amplified from cDNA of the human Retinal pigment epithelium cell strain ARPE-19 (American Type Culture Collection, ATCC) as a template and using appropriate primers.
  • An alternative fragment can be similarly obtained for any one of the proteins described herein.
  • the hPEDF fragment is obtained by gel recovery and ligated into the pLenti6.3/V5-TOPO® vector (Invitrogen) by TA cloning procedure following the manufacturer's instruction. The sequence of the ligated hPEDF fragment can be verified by sequencing.
  • An SIV lentiviral transfer vector is prepared according the methods of U.S. Publication No. 20150056696.
  • a SIV gene transfer vector, a packaging vector, a rev expression vector, and a VSV-G expression vector are obtained, and an hPEDF fragment is introduced into the gene transfer vector.
  • An alternative fragment can be similarly obtained for any one of the proteins described herein for introduction into the gene transfer vector.
  • the cell line 293T cells derived from human fetal kidney cells are seeded at a cell density of approximate 1 ⁇ 10 7 cells per plastic Petri dish having the diameter of 15 cm (cell density of 70-80% next day) and cultured in 20 ml of D-MEM culture medium (Gibco BRL) supplemented with 10% fetal bovine serum for 24 hrs. After 24 h of cultivation, the culture medium is replaced with 10 ml of OPTI-MEM culture medium (Gibco BRL).
  • HSV transfer vector is prepared according the methods of U.S. Publication No. 20090186003.
  • HSV-1 (F) strain is a low passage clinical isolate used as the prototype HSV-1 strain.
  • M002 which expresses murine interleukin 12 (mIL-12) under the transcriptional control of the murine early-growth response-1 promoter (Egr-1), is constructed.
  • mIL-12 murine interleukin 12
  • Egr-1 murine early-growth response-1 promoter
  • similar constructs may be prepared encoding any one of the proteins described herein under the control of an appropriate promoter.
  • the plasmids containing the p40 and p35 subunits of mIL-12 in pBluescript-SK+ (Stratagene) are obtained.
  • the p40 subunit is removed by digestion with HindIII (5′ end) and BamHI (3′ end) and the p35 subunit is removed by digestion with NcoI (5′ end) and EcoRI (3′ end).
  • the internal ribosome entry site, or IRES, sequence is amplified from vector pCITE-4a+ (Novagen, Madison, Wis.) using polymerase chain reaction (PCR) and appropriate primers.
  • Plasmid pBS-IL12 is constructed by three-way ligation of the murine p40, murine p35 and IRES sequences into HindIII and EcoRI sites of pBS-SK+ such that the IRES sequence separates the p40 and p35 coding sequences.
  • Plasmid 4878-IL12 is constructed as follows: pBS-mIL-12 is digested with XhoI and SpeI to remove a 2.2 kb fragment containing the entire IL-12 subunit coding regions, including the IRES, ends filled in using the Klenow fragment, and ligated into a blunted KpnI site located between the Egr-1 promoter and hepatitis B virus polyA sequences within pRB4878.
  • M001 (tk ⁇ ) and M002 (tk repaired at native locus) are constructed via homologous recombination as described previously (Andreansky et al. (1998) Gene Ther. 5, 121-130).
  • Two tk-repaired viruses M002.29 and M002.211 are confirmed by Southern blot hybridization of restriction enzyme-digested viral DNAs which are electrophoretically separated on a 1% agarose, 1 ⁇ TPE gel and transferred to a Zeta-Probe membrane (Bio-Rad).
  • the blot is hybridized with the appropriate DNA probe labeled with alkaline phosphatase using the Gene Images AlkPhos Direct DNA labeling system (Amersham-Pharmacia Biotech, Piscataway, N.J.). IL-12 production is demonstrated by enzyme-linked immunosorbent assay (ELISA).
  • ELISA enzyme-linked immunosorbent assay
  • any one of the viral vectors described herein, such as in the above Examples, may be used to produce a viral transfer vector with a gene editing transgene.
  • the following provides a method for producing a viral transfer vector with a gene editing transgene that encodes Cas9, such as Cas9 wild-type (Type II).
  • HEK293T cells can be cultured in DMEM medium (Life Technologies, Darmstadt, Germany) containing 10% fetal bovine serum (Sigma, Steinheim, Germany), 100 U/mL penicillin and 100 ⁇ g/mL streptomycin (Life Technologies). Huh7 and Hep56D cell media can additionally contain 1% non-essential amino acids (Life Technologies).
  • Jurkat cells can be grown in RPMI 1640 medium (GE Healthcare, Pasching, Austria) containing 10% fetal bovine serum (Sigma), 100 U/mL penicillin, 100 ⁇ g/mL streptomycin, and 2 mM L-glutamine (all Life Technologies). All cell lines can be cultured at 37° C. and 5% CO2.
  • HEK293T cells can be seeded in ten 15 cm2 dishes (4 ⁇ 106 cells per dish). Two days later, they can be triple-transfected with (i) the AAV vector plasmid (encoding gRNA and/or Cas9), (ii) an AAV helper plasmid carrying AAV rep and cap genes, and (iii) an adenoviral plasmid providing helper functions for AAV production.
  • AAV vector plasmid encoding gRNA and/or Cas9
  • an AAV helper plasmid carrying AAV rep and cap genes
  • an adenoviral plasmid providing helper functions for AAV production.
  • the AAV cap gene can be either derived from the synthetic isolate AAV-DJ (Grimm, et al., J. Virol. 2008, 82, 5887-5911) or from a new variant AAVrh10A2.
  • AAVrh10A2 can be created through insertion of a seven amino acid long peptide into an exposed region of the capsid of AAV serotype rh10. Further details on AAV production plasmids and protocols can be performed as reported in Börner, et al., Nucleic Acids Res. 2013, 41, e199 and Grimm, Methods 2002, 28, 146-157.
  • HEK293T cells per well can be seeded in 6-well plates and the next day triple-transfected with the aforementioned plasmids. Three days later, the cells can be scraped off into the media, collected via 10 min centrifugation at 1500 rpm, resuspended in 300 ⁇ L 1 ⁇ PBS (Life Technologies) and subjected to three freeze-thaw cycles in liquid nitrogen and at 37° C. A 10 min centrifugation can be performed at 13,200 rpm to remove cell debris, and supernatants containing viral transfer vector particles can be used directly in transduction experiments or frozen at ⁇ 20° C.
  • 2.8 ⁇ 104 HEK293T or 1.2 ⁇ 104 Huh7 cells can be seeded per well in a 96-well plate and the next day transfected using lipofectamine 2000 (Life Technologies) following the manufacturer's recommendations for this format (200 ng DNA and 0.5 ⁇ L lipofectamine 2000, each in 25 ⁇ L serum-free medium).
  • the 200 ng DNA can consist of an all-in-one Cas9/gRNA vector, or, in the case of separate Cas9 and gRNA constructs, of 100 ng of each.
  • HEK293T cells can be transfected in 24-well plates (one well per lysate), using lipofectamine 2000 according to the manufacturer's recommendations for this format.
  • cells can be grown in 96-well plates and transduced with either 10 ⁇ L non-purified AAV or with purified vector 1 day after seeding. Following a three (transfections) to five (transductions) day incubation, the cells can be lysed with DirectPCR Lysis Reagent Cell (PeqLab, Er Weg, Germany) supplemented with 0.2 ⁇ g/mL proteinase K (Roche, Mannheim, Germany) following the manufacturer's protocol.
  • plasmids and vectors such as those above can achieve delivery of the CRISPR components—Cas9 and chimeric g(uide) RNA.
  • Cas9 expression could be directed to or away from hepatocytes, using a liver-specific promoter or a hepatic miRNA binding site, respectively. Further evidence was provided that such vectors can be used for gene engineering in vivo. This was accomplished in the exemplified liver of adult mice.
  • Example 30 can also be used to produce a viral transfer vector with a gene editing transgene, such as a transgene encoding a Cas9 variant, such as any one of the Cas9 variants described herein.
  • a gene editing transgene such as a transgene encoding a Cas9 variant, such as any one of the Cas9 variants described herein.
  • any one of the other viral vectors described herein may be used instead to produce such a viral transfer vector.
  • Any one of the Cas9 variants provided can be encoded by any one of the gene editing transgenes provided herein.
  • the human codon-optimized streptococcus pyogenes Cas9 nuclease with NLS and 3 ⁇ FLAG tag (Addgene plasmid 43861) can be used as the wild-type Cas9 expression plasmid.
  • PCR products of wild-type Cas9 expression plasmid as template with Cas9_Exp primers can be assembled with Gibson Assembly Cloning Kit (New England Biolabs) to construct Cas9 and FokI-dCas9 variants.
  • Expression plasmids encoding a single gRNA construct (gRNA G1 through G13) can also be cloned.
  • any one of the viral vectors described herein, such as in the above Examples, may be used to produce a viral transfer vector with a gene editing transgene that encodes a zinc finger nuclease.
  • the following provides a method for producing such a viral transfer vector with a gene editing transgene.
  • TRBC- and TRAC-ZFNs can be designed and assembled as described in Urnov, et al. Nature 435, 646-651 (2005).
  • the recognition helices used can be as provided Provasi, et al., Nature Medicine, Vol. 18, No. 5, May 2012.
  • Lentiviral vectors encoding TRBC- and TRAC-ZFNs can be generated from the HIV-derived self-inactivating transfer construct pCCLsin.cPPT.SFFV.eGFP.Wpre, which can be packaged by an integrase-defective third generation packaging construct carrying the D64V mutation in the HIV integrase and pseudotyped by the VSV envelope.
  • the Ad5/F35 adenoviral vectors can be generated on an E1-E3-deleted backbone.
  • the ZFNs targeting either the TRBC or TRAC gene can be linked using a 2A peptide sequence and cloned into the pAdEasy-1/F35 vector under the control of an appropriate promoter, and the Ad5/F35 virus for each construct can be generated using TREx 293T cells.
  • Lentiviral vectors encoding both WT1-specific TCR chains and single ⁇ 21 or ⁇ 21 WT1-specific TCR chains from the bidirectional self-inactivating transfer vector pCCLsin.cPPT. ⁇ LNGFR.minCMV.hPGK.eGFP.Wpre and from pCCLsin.cPPT.hPGK.eGFP.Wpre can be generated and packaged by an integrase-competent third-generation construct and pseudotyped by the VSV envelope.
  • Zinc finger nucleases targeting the hF9mut locus and F9-targeting vectors can be prepared as described in Li, et al. Nature. 2011; 475(7355):217-221. Such vectors have been shown to be successfully used for in vivo gene targeting in a neonatal mouse model of hemophilia B (HB).
  • Systemic codelivery of the AAV vectors, encoding the ZFN pair targeting the human F9 gene and a gene-targeting vector with arms of homology flanking a corrective cDNA cassette resulted in the correction of a defective hF9 gene engineered into the mouse genome in the livers of such mice. Further, stable levels of human factor IX expression sufficient to normalize clotting times was achieved.
  • any one of the viral vectors described herein, such as in the above Examples, may be used to produce a viral transfer vector with a gene editing transgene that encodes a meganuclease.
  • the following describes a general methodology for producing such a viral transfer vector with a gene editing transgene.
  • the meganuclease may be any one of the meganucleases provided in U.S. Publication Nos. 20110033935 and 20130224863.
  • particular viral genes are inactivated to prevent reproduction of the virus.
  • a virus is altered so that it is capable only of delivery and maintenance within a target cell, but does not retain the ability to replicate within the target cell or tissue.
  • One or more DNA sequences encoding a meganuclease can be introduced to the altered viral genome, so as to produce a viral genome that acts like a vector.
  • the viral vector is a retroviral vector such as, but not limited to, the MFG or pLJ vectors.
  • An MFG vector is a simplified Moloney murine leukemia virus vector (MoMLV) in which the DNA sequences encoding the pol and env proteins are deleted to render it replication defective.
  • MoMLV Moloney murine leukemia virus vector
  • a pll retroviral vector is also a form of the MoMLV (see, e.g., Korman et al. (1987), Proc. Nat'l Acad. Sci., 84:2150-2154).
  • a recombinant adenovirus or adeno-associated virus can be used to produce a viral vector.
  • any one of the viral vectors described herein, such as in the above Examples, may be used to produce a viral transfer vector with an exon skipping transgene.
  • the following provides a method for producing a viral transfer vector with a specific exon skipping transgene.
  • a three-plasmid transfection protocol can be used with pAAV(U7smOPT-SD23/BP22) and pAAV(U7smOPT-scr) plasmids for generation of single-strand AAV1-U7ex235 and AAV1-U7scr; and scAAV-U7ex23 plasmid for self-complementary scAAV9-U7ex239.
  • pAAV(U7smOPT-scr) plasmid can contain the non specific sequence GGTGTATTGCATGATATGT that does not match to any murine cDNAs.
  • compositions and methods provided herein can help maintain the effect of such a treatment.
  • Clone U1#23 can be obtained by inverse PCR on the human U1 snRNA gene, with oligos mU1anti5 (5′-CGAAATTTCAGGTAAGCCGAGGTTATGAGATCTTGGGCCTCTGC-3′ and mU1anti3 (5′-GAACTTTGCAGAGCCTCAAAATTAAATAGGGCAGGGGAGATACCATGATC-3′).
  • the antisense-containing insert can be amplified from corresponding plasmid with oligos U1cas-up-NheI (5′-CTAGCTAGCGGTAAGGACCAGCTTCTTTG-3′) and U1cas-down-NheI (5′-CTAGCTAGCGGTTAGCGTACAGTCTAC-3′).
  • the resulting fragment can be NheI-digested and cloned in the forward orientation of the pAAV2.1-CMV-EGFP plasmid.
  • AAV-U1#23 vector can be produced by triple transfection of 293 cells, purified by CsCl2 ultracentrifugation and titered by using both real-time PCR-based and dot-blot assays. The number of green-forming units can be assessed by serial dilution on 293 cells.
  • AAV vector can be produced by the AAV TIGEM Vector Core.
  • mice Six-week-old mdx mice can be administered with 3-4 ⁇ 10 12 genome copies of AAV vector via tail vein. Six and 12 weeks after virus administration, animals can be killed, and muscles from different districts can be harvested. EGFP analysis and dissections can be performed under a fluorescent stereomicroscope (Leica MZ16FA).
  • U7snRNA constructs specific to certain exons can be engineered, such as from U7smOPT-SD23/BP22 (modified murine U7snRNA gene).
  • Antisense sequences targeting certain exons can be replaced by antisense sequences targeting exons of dystrophin mRNA that induce exon skipping as antisense oligonucleotides. Sequences can be inserted into U7snRNA constructs. Resulting U7snRNA fragments can then be introduced either in a lentiviral vector construct for further lentiviral production or into an AAV vector construct for AAV production.
  • Lentiviral vectors can be based on pRRLcPPT-hPGK-eGFP-WPRE constructs where the hPGK-GFP cassette is removed and replaced with the U7snRNA construct.
  • Lentiviral vectors can be generated by transfection into 293T cells of a packaging construct, pCMV ⁇ R8.74, a plasmid producing the vesicular stomatosis virus-G envelope (pMD.G) and the vector itself as previously described.
  • Viral titers infectious particles
  • genomic DNA from transduced cells can be extracted using a genomic DNA purification kit (Qiagen, Crawley, UK).
  • the infectious particles titer infectious particle/ml
  • the infectious particles titer can be determined by quantitative real-time PCR as described elsewhere.
  • AAV2/1 pseudotyped vectors can be prepared by cotransfection in 293 cells of pAAV2-U7snRNA, pXX6 encoding adenovirus helper functions and pAAV1pITRCO2 that contains the AAV2 rep and AAV1 cap genes.
  • Vector particles can be purified on Iodixanol gradients from cell lysates obtained 48 hours after transfection and titers can be measured by quantitative real time PCR.
  • viral transfer vectors such as those produced and encoding U7 small-nuclear RNAs with the above methods can induce efficient exon skipping both in vitro and in vivo.
  • An HSV transfer vector can prepared according the methods of U.S. Publication No. 20090186003 and Example 28 above except that the methodology can be altered so that the transgene is instead an exon skipping transgene.
  • the exon skipping transgene may be any one of such transgenes as described herein or otherwise known in the art.
  • HIV lentiviral transfer vector is prepared according the methods of U.S. Publication No. 20150056696 and Example 26 above except that the methodology can be altered so that the transgene is instead an exon skipping transgene.
  • the exon skipping transgene may be any one of such transgenes as described herein or otherwise known in the art.
  • any one of the viral vectors described herein, such as in the above Examples, may be used to produce a viral transfer vector with a gene expression modulating transgene.
  • the following provides a method for producing a viral transfer vector with a specific gene expression modulating transgene.
  • a viral transfer vector is produced according to the methods described in Brown et al., Nat Med. 2006 May; 12(5):585-91. Briefly, a plasmid is constructed using reverse transcription of RNA, quantitative PCR analysis to quantify the concentration of mRNA, and GAPDH expression for normalization. VSV-pseudotyped third-generation lentiviral vectors (LVs) are produced by transient four-plasmid cotransfection into 293T cells and purified by ultracentrifugation. Vector particles can be measured by HIV-1 gag p24 antigen immunocapture.
  • an AAV2/1 serotype vector encoding an miRNA-based hairpin against a gene e.g., huntingtin gene; AAV2/1-miRNA-Htt
  • the cDNA for the specified gene e.g., human HTT
  • ITRs AAV2 inverted terminal repeats
  • CBA cytomegalovirus enhancer/chicken b-actin
  • Control vectors can also be developed and contain either an empty vector backbone (e.g., AAV2/1-Null) or express a reporter such as enhanced green fluorescent protein under the control of the same promoter (AAV2/1-eGFP).
  • Viral transfer vectors can be generated by triple-plasmid cotransfection of a cell line, such as human 293 cells, and the recombinant virions can then be column-purified as previously described in Stanek et al., Human Gene Therapy. 2014; 25:461-474. The resulting titer of AAV2/1-miRNA-Htt can then be determined using quantitative PCR.
  • AAV-mediated RNAi can be effective at transducing cells in the striatum and can partially reduce the levels of both wild-type and mutant Htt in this region.
  • the CD81 gene can be amplified by reverse transcription.
  • cDNA can be PCR amplified with appropriate primers.
  • the forward primer can contain a BamHI (Biolabs, Allschwill, Switzerland) restriction site followed by a 5′ CD81 cDNA-specific sequence;
  • the reverse primer can contain a 3′ CD81 cDNA-specific sequence, a 6 His-tag, a stop codon and an Xho I (Biolabs) restriction site.
  • the PCR product can be digested and cloned into similar sites in pTK431.
  • the pTK431 is a self-inactivating HIV-1 vector which contains the entire tet-off-inducible system, the central polypurine tract (cPPT) and the woodchuck hepatitis virus post-transcriptional regulatory element. Plasmids can be CsCl 2 purified.
  • Targets can be designed according to the CD81 mRNA sequence based on Hannon's design criterion (katandin.cshl.org:9331/RNAi/html/rnai.html).
  • Hannon's design criterion katandin.cshl.org:9331/RNAi/html/rnai.html.
  • each shRNA target can be added to the mouse U6 promoter by PCR.
  • the PCR product can be digested, cloned into similar sites in pTK431 and sequenced to verify the integrity of each construct.
  • the vector plasmids together with a packaging construct plasmid p ⁇ NRF and the envelope plasmid pMDG-VSVG, can be cotransfected into HEK293T cells to produce viral particles.
  • the viral titres can be determined by p24 antigen measurements (KPL, Lausanne, Switzerland).
  • lentiviruses expressing short hairpin RNA (shRNA) targeted against CD81 resulted in gene silencing after infection of HEK293T cells in vitro.
  • Lenti-CD81-shRNAs lentiviruses expressing short hairpin RNA targeted against CD81
  • in vivo delivery of Lenti-CD81-shRNA resulted in silencing of endogenous CD81.
  • any one of the viral vectors described herein, such as in the above Examples, may be used to produce a viral transfer vector with a gene expression modulating transgene that encodes a RNAi agent.
  • a gene expression modulating transgene that encodes a RNAi agent.
  • An example of a RNAi agent that can be encoded by a gene expression modulating transgene as provided herein is described below.
  • An expression construct can include a promoter driving the expression of three or more individual shRNA species.
  • the synthesis of small nuclear RNAs and transfer RNAs can be directed by RNA polymerase III (pol III) under the control of pol III-specific promoters. Because of the relatively high abundance of transcripts directed by these regulatory elements, pol III promoters, including those derived from the U6 and H1 genes, can be used to drive the expression of 1-x RNAi (see, e.g., Domitrovich and Kunkel. Nucl. Acids Res. 31(9): 2344-52 (2003); Boden, et al. Nucl. Acids Res. 31(17): 5033-38 (2003a); and Kawasaki, et al. Nucleic Acids Res.
  • RNAi expression constructs using the U6 promoter can comprise three RNAi agents targeting three different regions of the HCV genome.
  • Further examples of RNAi agents that may be encoded by a gene expression modulating transgene include any one of the RNAi agents described herein.
  • any one of the viral vectors described herein, such as in the above Examples, may be used to produce a viral transfer vector with a gene expression modulating transgene that encodes a Serpinal RNAi agent, such as one of such agents described in U.S. Patent Publication No. 20140350071.
  • a viral transfer vector with such a transgene can be produced following similar methodology as provided herein or otherwise known in the art.
  • adeno-associated virus (AAV) vectors may be used (Walsh et al., Proc. Soc. Exp. Biol. Med. 204:289-300 (1993); U.S. Pat. No. 5,436,146).
  • the iRNA can be expressed as two separate, complementary single-stranded RNA molecules from a recombinant AAV vector having, for example, either the U6 or H1 RNA promoters, or the cytomegalovirus (CMV) promoter.
  • Suitable AAV vectors for expressing the dsRNA featured in the invention, methods for constructing the recombinant AV vector, and methods for delivering the vectors into target cells are described in Samulski R et al. (1987), J.
  • any one of the viral transfer vectors provided herein, such as any one of the Examples, is administered concomitantly, such as simultaneously i.v., i.m., s.c. or i.p., with any one of the antigen-presenting cell targeted immunosuppressants as provided herein, such as any one of the Examples, that is also administered i.v., i.m., s.c. or i.p., respectively.
  • the administration occurs according to a protocol, including at least the frequency and dose of the viral transfer vector and antigen-presenting cell targeted immunosuppressant, that establishes an anti-viral transfer vector attenuated response in the subject.
  • the subject may be any one of the subjects described herein, such as one that does not have a pre-existing immunity to the viral transfer vector or one in which repeated administration of the viral transfer vector is desired.
  • the viral transfer vector when the anti-viral transfer vector attenuated response is a T cell response against the viral transfer vector, the viral transfer vector is administered to the subject without an antigen-presenting cell targeted immunosuppressant prior to the concomitant administration of the antigen-presenting cell targeted immunosuppressant and viral transfer vector.
  • one or more repeat doses of the viral transfer vector is administered to the subject subsequent to both the concomitant administration and the administration of the viral transfer vector prior thereto.
  • the subject when the anti-viral transfer vector attenuated response is a B cell response against the viral transfer vector, the subject is not administered the viral transfer vector prior to the concomitant administration of the viral transfer vector and antigen-presenting cell targeted immunosuppressant.
  • one or more repeat doses of the viral transfer vector is administered to the subject and each repeat dose is concomitantly administered with the antigen-presenting cell targeted immunosuppressant.
  • the subject when the anti-viral transfer vector attenuated response is an anti-viral transfer vector antibody response, the subject is not administered the viral transfer vector prior to the concomitant administration of the viral transfer vector and antigen-presenting cell targeted immunosuppressant.
  • one or more repeat doses of the viral transfer vector is administered to the subject and each repeat dose is concomitantly administered with the antigen-presenting cell targeted immunosuppressant.
  • the method for determining the level of antibodies may be with the use of an ELISA assay.
  • Assays for antigen-specific B cell or T cell recall responses include, but are not limited to, ELISpot, intracellular cytokine staining, cell proliferation, and cytokine production assays.
  • the anti-viral transfer vector attenuated response is evaluated after the concomitant administration of the viral transfer vector and the antigen-presenting cell targeted immunosuppressant.
  • a protocol for establishing the anti-viral transfer vector attenuated response may be determined.
  • the protocol is determined in another subject, such as a test subject. The protocol so determined can be used to treat other subjects subjects in need of treatment with the viral transfer vector.
  • a sample such as a blood sample, may be obtained from a subject that is in need of treatment with a viral transfer vector as provided herein, such as the viral transfer vector of any one of the viral transfer vectors provided herein, such as in any one of the Examples.
  • the level of antibodies such as neutralizing antibodies or antigen recall responses of immune cells, such as T cells or B cells
  • the method for determining the level of antibodies may be with the use of an ELISA assay.
  • Assays for antigen-specific B cell or T cell recall responses include, but are not limited to, ELISpot, intracellular cytokine staining, cell proliferation, and cytokine production assays.
  • the recall response can be assessed by contacting the sample with the viral transfer vector or an antigen thereof.
  • the recall response can also be assessed by taking the sample from the subject after administration of the viral transfer vector or an antigen thereof to the subject and then determining the level of antibodies or a B cell or T cell recall response that was generated.
  • the subject where the subject does not have a pre-existing immunity to the viral transfer vector, determined by the measurement of a level of anti-viral transfer vector antibodies in the subject (or a B cell response), the subject is administered, i.v., i.m., s.c. or i.p., any one of the viral transfer vectors provided herein, such as in any one of the Examples, concomitantly, such as simultaneously, with any one of the antigen-presenting cell targeted immunosuppressants provided herein, such as in any one of the Examples.
  • the antigen-presenting cell targeted immunosuppressant is administered by the same route.
  • the antigen-presenting cell targeted immunosuppressant and viral transfer vector are concomitantly, such as simultaneously, administered, i.v., i.m., s.c. or i.p., to the subject after the subject is administered a dose of the viral transfer vector without concomitant administration of the antigen-presenting cell targeted immunosuppressant.
  • one or more repeat doses of the viral transfer vector is/are administered to the subject. These repeat doses may be concomitantly administered with the antigen-presenting cell targeted immunosuppressant.
  • any one of the viral transfer vectors provided herein, such as in any one of the Examples, is administered concomitantly, such as simultaneously, i.v., i.m., s.c. or i.p., with any one of the antigen-presenting cell targeted immunosuppressants as provided herein, such as in any one of the Examples, according to a frequency and dosing that escalates transgene expression (the transgene being delivered by the viral transfer vector). This can be determined by measuring transgene protein concentrations in various tissues or systems of interest in the subject. Whether or not transgene expression is escalated can be determined according to a method, such as that described in the Examples above.
  • the administration occurs according to the frequency and dose of the viral transfer vector and antigen-presenting cell targeted immunosuppressant, that escalates transgene expression in the subject.
  • the subject may be any one of the subjects described herein, such as one that does not have a pre-existing immunity to the viral transfer vector or one in which repeated administration of the viral transfer vector is desired.
  • the frequency and dose that achieves escalating transgene expression is determined in another subject, such as a test subject. This can also be determined by measuring transgene protein concentrations in various tissues or systems of interest in the other subject, such as with a method as described above. If the frequency and dose achieves escalated transgene expression, as determined by the measured transgene protein concentrations, in the other subject, the concomitant, such as simultaneous, administration of the viral transfer vector and antigen-presenting cell targeted immunosuppressant according to the frequency and dose can be used to treat other subjects in need of treatment with the viral transfer vector.
  • the concomitant such as simultaneous, administration of the viral transfer vector and antigen-presenting cell targeted immunosuppressant according to the frequency and dose can be used to treat other subjects in need of treatment with the viral transfer vector.
  • a subject can be evaluated for the level of a pre-existing immunity to any one of the viral transfer vectors provided herein, such as any one of the viral transfer vectors any one of the Examples.
  • a clinician may evaluate a subject and determine whether or not, if administered the viral transfer vector, the subject is expected to develop an anti-viral transfer vector immune response if the viral transfer vector is repeatedly administered to the subject. This determination may be made based on the likelihood that the viral transfer vector will produce such a result and may be based on such a result in other subjects, such as test subjects, information about the virus that was used to generate the viral transfer vector, information about the subject, etc.
  • the clinician selects a certain dose of the viral transfer vector as a result of the expectation.
  • a clinician may now select and use lower doses of the viral transfer vector than would have been selected for the subject. Benefits of lower doses can include reduced toxicity associated with dosing of the viral transfer vector, and reduction of other off-target effects.
  • any one of the subjects provided herein can be treated with repeated, concomitant, such as simultaneous, administration of any one of the viral transfer vectors provided herein and any one of the antigen-presenting cell targeted immunosuppressants provided herein where the doses of the viral transfer vector are selected to be less than the dose of the viral transfer vector that would have been selected for the subject if the subject were expected to develop anti-viral transfer vector immune responses due to repeated dosing of the viral transfer vector.
  • Each dose of the viral transfer vector of the repeated, concomitant administration may be less than what would have otherwise been selected.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Nanotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Optics & Photonics (AREA)
  • Inorganic Chemistry (AREA)
  • Virology (AREA)
  • Biochemistry (AREA)
  • Transplantation (AREA)
  • Neurology (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Mycology (AREA)
  • General Physics & Mathematics (AREA)
  • Theoretical Computer Science (AREA)
  • General Business, Economics & Management (AREA)
  • Business, Economics & Management (AREA)
US14/846,952 2014-09-07 2015-09-07 Methods and compositions for attenuating gene therapy anti-viral transfer vector immune responses Pending US20160074531A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/846,952 US20160074531A1 (en) 2014-09-07 2015-09-07 Methods and compositions for attenuating gene therapy anti-viral transfer vector immune responses

Applications Claiming Priority (13)

Application Number Priority Date Filing Date Title
US201462047054P 2014-09-07 2014-09-07
US201462047044P 2014-09-07 2014-09-07
US201462047034P 2014-09-07 2014-09-07
US201462047051P 2014-09-07 2014-09-07
US201462051255P 2014-09-16 2014-09-16
US201462051267P 2014-09-16 2014-09-16
US201462051263P 2014-09-16 2014-09-16
US201462051258P 2014-09-16 2014-09-16
US201562101882P 2015-01-09 2015-01-09
US201562101872P 2015-01-09 2015-01-09
US201562101841P 2015-01-09 2015-01-09
US201562101861P 2015-01-09 2015-01-09
US14/846,952 US20160074531A1 (en) 2014-09-07 2015-09-07 Methods and compositions for attenuating gene therapy anti-viral transfer vector immune responses

Publications (1)

Publication Number Publication Date
US20160074531A1 true US20160074531A1 (en) 2016-03-17

Family

ID=54291579

Family Applications (7)

Application Number Title Priority Date Filing Date
US14/846,952 Pending US20160074531A1 (en) 2014-09-07 2015-09-07 Methods and compositions for attenuating gene therapy anti-viral transfer vector immune responses
US14/846,949 Active US11633422B2 (en) 2014-09-07 2015-09-07 Methods and compositions for attenuating anti-viral transfer vector immune responses
US14/846,958 Pending US20160074532A1 (en) 2014-09-07 2015-09-07 Methods and compositions for attenuating gene editing anti-viral transfer vector immune responses
US14/846,964 Active US10071114B2 (en) 2014-09-07 2015-09-07 Methods and compositions for attenuating gene expression modulating anti-viral transfer vector immune responses
US14/846,967 Active US10046064B2 (en) 2014-09-07 2015-09-07 Methods and compositions for attenuating exon skipping anti-viral transfer vector immune responses
US16/100,040 Pending US20190076458A1 (en) 2014-09-07 2018-08-09 Methods and compositions for attenuating gene expression modulating anti-viral transfer vector immune responses
US18/185,891 Pending US20230364124A1 (en) 2014-09-07 2023-03-17 Methods and compositions for attenuating anti-viral transfer vector immune responses

Family Applications After (6)

Application Number Title Priority Date Filing Date
US14/846,949 Active US11633422B2 (en) 2014-09-07 2015-09-07 Methods and compositions for attenuating anti-viral transfer vector immune responses
US14/846,958 Pending US20160074532A1 (en) 2014-09-07 2015-09-07 Methods and compositions for attenuating gene editing anti-viral transfer vector immune responses
US14/846,964 Active US10071114B2 (en) 2014-09-07 2015-09-07 Methods and compositions for attenuating gene expression modulating anti-viral transfer vector immune responses
US14/846,967 Active US10046064B2 (en) 2014-09-07 2015-09-07 Methods and compositions for attenuating exon skipping anti-viral transfer vector immune responses
US16/100,040 Pending US20190076458A1 (en) 2014-09-07 2018-08-09 Methods and compositions for attenuating gene expression modulating anti-viral transfer vector immune responses
US18/185,891 Pending US20230364124A1 (en) 2014-09-07 2023-03-17 Methods and compositions for attenuating anti-viral transfer vector immune responses

Country Status (12)

Country Link
US (7) US20160074531A1 (ja)
EP (5) EP3189147A1 (ja)
JP (11) JP7242177B2 (ja)
KR (7) KR20170045344A (ja)
CN (6) CN107073050A (ja)
AU (10) AU2015311704B2 (ja)
BR (5) BR112017001601A2 (ja)
CA (5) CA2957802A1 (ja)
EA (5) EA201790550A1 (ja)
IL (10) IL292580A (ja)
MX (10) MX2017002933A (ja)
WO (5) WO2016037162A1 (ja)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9764031B2 (en) 2010-05-26 2017-09-19 Selecta Biosciences, Inc. Dose selection of adjuvanted synthetic nanocarriers
US9884112B2 (en) 2009-05-27 2018-02-06 Selecta Biosciences, Inc. Immunomodulatory agent-polymeric compounds
US9987354B2 (en) 2011-04-29 2018-06-05 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for antigen-specific deletion of T effector cells
US9994443B2 (en) 2010-11-05 2018-06-12 Selecta Biosciences, Inc. Modified nicotinic compounds and related methods
US10046064B2 (en) 2014-09-07 2018-08-14 Selecta Biosciences, Inc. Methods and compositions for attenuating exon skipping anti-viral transfer vector immune responses
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
US10933129B2 (en) 2011-07-29 2021-03-02 Selecta Biosciences, Inc. Methods for administering synthetic nanocarriers that generate humoral and cytotoxic T lymphocyte responses
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
WO2022221529A1 (en) 2021-04-16 2022-10-20 Asklepios Biopharmaceutical, Inc. Rational polyploid aav virions that cross the blood brain barrier and elicit reduced humoral response

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2699578T3 (es) 2013-04-16 2019-02-11 Regeneron Pharma Modificación direccionada del genoma de rata
KR102170502B1 (ko) 2013-12-11 2020-10-28 리제너론 파마슈티칼스 인코포레이티드 게놈의 표적화된 변형을 위한 방법 및 조성물
GB201508025D0 (en) 2015-05-11 2015-06-24 Ucl Business Plc Fabry disease gene therapy
US10968253B2 (en) * 2015-10-20 2021-04-06 Institut National De La Sante Et De La Recherche Medicale (Inserm) Methods and products for genetic engineering
WO2017158422A1 (en) * 2016-03-16 2017-09-21 Crispr Therapeutics Ag Materials and methods for treatment of hereditary haemochromatosis
WO2017192512A2 (en) * 2016-05-02 2017-11-09 Massachusetts Institute Of Technology Amphiphilic nanoparticles for delivery of crispr based therapy
KR20190104194A (ko) * 2017-01-07 2019-09-06 셀렉타 바이오사이언시즈, 인크. 합성 나노담체에 커플링된 면역억제제의 패턴화된 투여
US20230190958A1 (en) * 2017-01-13 2023-06-22 Jichi Medical University AAV Vector for Disrupting Coagulation Factor-Related Gene on Liver Genome
KR20200086670A (ko) * 2017-10-13 2020-07-17 셀렉타 바이오사이언시즈, 인크. 항바이러스 전달 벡터 IgM 반응을 약화시키기 위한 방법 및 조성물
US20200338216A1 (en) * 2018-01-11 2020-10-29 Chameleon Biosciences, Inc. Immuno-evasive vectors and use for gene therapy
JP2021530571A (ja) * 2018-07-16 2021-11-11 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. Mmaコンストラクトおよびベクターの方法および組成物
EP3849564A4 (en) * 2018-09-10 2023-08-09 Ionis Pharmaceuticals, Inc. COMPOUNDS AND METHODS FOR MODULATION OF CLN3 EXPRESSION
EP3897691A4 (en) * 2018-12-21 2022-08-31 The Regents of the University of California IL-10 VACCINES AND THEIR USES
US20220142991A1 (en) * 2019-03-14 2022-05-12 Children's Hospital Medical Center Methods and compositions for treatment of l ysosomal storage disorder
CN110055278B (zh) * 2019-04-23 2021-10-15 深圳市弘际生物科技有限责任公司 一种新型腺病毒包装方法在CRISPR/Cas9基因编辑方法中的应用
MX2021013163A (es) * 2019-04-28 2022-02-21 Selecta Biosciences Inc Métodos para el tratamiento de sujetos con inmunidad preexistente a vectores de transferencia viral.
MX2021014566A (es) * 2019-05-28 2022-03-22 Selecta Biosciences Inc Métodos y composiciones para la respuesta inmune atenuada anti-vector de transferencia viral.
CN110396524B (zh) * 2019-05-31 2023-05-02 海南大学 一种用于蚊虫的RNAi纳米颗粒和制备方法及用途
WO2021211641A2 (en) * 2020-04-14 2021-10-21 University Of Florida Researchfoundation, Incorporated Enhancing aav-mediated delivery and transduction with polyvinyl alcohol
CN113769058A (zh) * 2020-06-10 2021-12-10 上海宝济药业有限公司 一种药物组合及其应用
CN112656763B (zh) * 2020-12-29 2022-07-15 吉林大学 一种基于剪切力响应的载药纳米胶束的制备方法
CN113876765A (zh) * 2021-10-28 2022-01-04 青岛大学附属医院 蟛蜞菊内酯在制备用于预防和/或治疗疱疹病毒感染的药物和保健品中的应用
CN116875621B (zh) * 2023-07-18 2024-03-29 浙江大学 一种提高转移载体中ires序列介导筛选基因表达效率的方法

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6251957B1 (en) * 1995-02-24 2001-06-26 Trustees Of The University Of Pennsylvania Method of reducing an immune response to a recombinant virus
US20020014242A1 (en) * 2000-07-31 2002-02-07 Abraham Scaria Use of rapamycin to inhibit immune response and induce tolerance to gene therapy vector and encoded transgene products
US20100129439A1 (en) * 2008-10-12 2010-05-27 Frank Alexis Adjuvant Incorporation in Immunonanotherapeutics
US20100273220A1 (en) * 2009-04-22 2010-10-28 Massachusetts Institute Of Technology Innate immune suppression enables repeated delivery of long rna molecules
US11633422B2 (en) * 2014-09-07 2023-04-25 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector immune responses

Family Cites Families (324)

* 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
US4797368A (en) 1985-03-15 1989-01-10 The United States Of America As Represented By The Department Of Health And Human Services Adeno-associated virus as eukaryotic expression vector
US5139941A (en) 1985-10-31 1992-08-18 University Of Florida Research Foundation, Inc. AAV transduction vectors
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
CA1340581C (en) 1986-11-20 1999-06-08 Joseph P. Vacanti Chimeric neomorphogenesis of organs by controlled cellular implantation using artificial matrices
US5759830A (en) 1986-11-20 1998-06-02 Massachusetts Institute Of Technology Three-dimensional fibrous scaffold containing attached cells for producing vascularized tissue in vivo
ATE81467T1 (de) 1987-02-24 1992-10-15 Xoma Corp Immunosuppression bei der auf immunotoxin basierten behandlung von menschen.
US5912017A (en) 1987-05-01 1999-06-15 Massachusetts Institute Of Technology Multiwall polymeric microspheres
US5019379A (en) 1987-07-31 1991-05-28 Massachusetts Institute Of Technology Unsaturated polyanhydrides
JP2670680B2 (ja) 1988-02-24 1997-10-29 株式会社ビーエムジー 生理活性物質含有ポリ乳酸系微小球およびその製造法
US5010167A (en) 1989-03-31 1991-04-23 Massachusetts Institute Of Technology Poly(amide-and imide-co-anhydride) for biological application
GB8918616D0 (en) 1989-08-15 1989-09-27 Univ Glasgow Herpes simplex virus type 1 mutant
US5436146A (en) 1989-09-07 1995-07-25 The Trustees Of Princeton University Helper-free stocks of recombinant adeno-associated virus vectors
US5849572A (en) 1990-10-10 1998-12-15 Regents Of The University Of Michigan HSV-1 vector containing a lat promoter
US5252479A (en) 1991-11-08 1993-10-12 Research Corporation Technologies, Inc. Safe vector for gene therapy
US5587308A (en) 1992-06-02 1996-12-24 The United States Of America As Represented By The Department Of Health & Human Services Modified adeno-associated virus vector capable of expression from a novel promoter
US5804413A (en) 1992-07-31 1998-09-08 University Of Pittsburgh Of The Commonwealth System Of Higher Education Herpes simplex virus strains for gene transfer
US5399665A (en) 1992-11-05 1995-03-21 Massachusetts Institute Of Technology Biodegradable polymers for cell transplantation
US5478745A (en) 1992-12-04 1995-12-26 University Of Pittsburgh Recombinant viral vector system
US5679347A (en) 1992-12-10 1997-10-21 Brigham And Women's Hospital Methods of isolating CD1-presented antigens, vaccines comprising CD1-presented antigens, and cell lines for use in said methods
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
US5565215A (en) 1993-07-23 1996-10-15 Massachusettes Institute Of Technology Biodegradable injectable particles for imaging
WO1995003035A1 (en) 1993-07-23 1995-02-02 Massachusetts Institute Of Technology Polymerized liposomes with enhanced stability for oral delivery
US5543158A (en) 1993-07-23 1996-08-06 Massachusetts Institute Of Technology Biodegradable injectable nanoparticles
BR9407956A (pt) 1993-10-25 1996-11-26 Canji Inc Composiçao farmacêutica vetor de express o de adenovirus recombinante e kit par reduzir a proliferaçao de células tumorais
TW249754B (ja) 1993-10-26 1995-06-21 Alpha I Biomedicals Inc
JP3872813B2 (ja) 1994-05-18 2007-01-24 エス.ピー.アイ. シンセティック ペプチデス インコーポレイテッド ヘテロダイマーポリペプチド免疫原キャリア組成物および方法
EP0784690B1 (en) 1994-06-10 2006-08-16 Genvec, Inc. Complementary adenoviral vector systems and cell lines
US6007845A (en) 1994-07-22 1999-12-28 Massachusetts Institute Of Technology Nanoparticles and microparticles of non-linear hydrophilic-hydrophobic multiblock copolymers
GB9415319D0 (en) 1994-07-29 1994-09-21 Medical Res Council HSV viral vector
US5846782A (en) 1995-11-28 1998-12-08 Genvec, Inc. Targeting adenovirus with use of constrained peptide motifs
EP0804076A4 (en) * 1994-10-19 1998-10-21 Genetic Therapy Inc GENTHERAPY THROUGH SIMULTANEOUS AND REPEATED ADMINISTRATION OF ADENOVIRUS AND IMMUNE SUPPRESSIVES
US5716404A (en) 1994-12-16 1998-02-10 Massachusetts Institute Of Technology Breast tissue engineering
DE69630514D1 (de) 1995-01-05 2003-12-04 Univ Michigan Oberflächen-modifizierte nanopartikel und verfahren für ihre herstellung und verwendung
US6123727A (en) 1995-05-01 2000-09-26 Massachusetts Institute Of Technology Tissue engineered tendons and ligaments
ES2333425T5 (es) 1995-06-15 2012-08-28 Crucell Holland B.V. Sistemas de empaquetado para adenovirus recombinante humano destinados a terapia génica
US6001650A (en) 1995-08-03 1999-12-14 Avigen, Inc. High-efficiency wild-type-free AAV helper functions
JP3462313B2 (ja) 1995-08-24 2003-11-05 キッコーマン株式会社 変異型ウリカーゼ、変異型ウリカーゼ遺伝子、新規な組み換え体dna及び変異型ウリカーゼの製造法
US5801030A (en) 1995-09-01 1998-09-01 Genvec, Inc. Methods and vectors for site-specific recombination
US5837511A (en) 1995-10-02 1998-11-17 Cornell Research Foundation, Inc. Non-group C adenoviral vectors
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
US6893664B1 (en) 1996-06-17 2005-05-17 Powderject Research Limited Particle delivery techniques
US6258823B1 (en) 1996-07-12 2001-07-10 Ariad Pharmaceuticals, Inc. Materials and method for treating or preventing pathogenic fungal infection
AU4176497A (en) * 1996-09-03 1998-03-26 Health Research Inc. Treatment of antigen presenting cells to modulate antigen presenting cell fun ction
US5814500A (en) 1996-10-31 1998-09-29 The Johns Hopkins University School Of Medicine Delivery construct for antisense nucleic acids and methods of use
CZ227199A3 (cs) 1996-12-27 1999-12-15 Icn, Pharmaceuticals, Inc. Aptamer
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
US5849561A (en) 1997-05-22 1998-12-15 Cornell Research Foundation, Inc. Method for the production of non-group C adenoviral vectors
EP0996735B1 (en) 1997-06-09 2003-12-10 Genvec, Inc. Chimeric vectors comprising a phage packaging site and a portion derived from the genome of a eukaryotic virus
US5837752A (en) 1997-07-17 1998-11-17 Massachusetts Institute Of Technology Semi-interpenetrating polymer networks
EP0894853A1 (en) 1997-07-24 1999-02-03 Gesellschaft für Biotechnologische Forschung mbH (GBF) Transcriptional silencer protein NRF, nucleic acid molecules encoding it and their use
EP1002119A1 (en) 1997-07-31 2000-05-24 University Of Pittsburgh Of The Commonwealth System Of Higher Education Targeted hsv vectors
DE69831417T2 (de) 1997-09-23 2006-06-22 Genvec, Inc. Duale selektionkassette und sie enthaltende plasmide
EP2042192A3 (en) 1997-10-30 2009-05-27 Laboratorios LETI, S.L. Tolerogenic fragments of natural allergens
US6018817A (en) 1997-12-03 2000-01-25 International Business Machines Corporation Error correcting code retrofit method and apparatus for multiple memory configurations
US6197229B1 (en) 1997-12-12 2001-03-06 Massachusetts Institute Of Technology Method for high supercoiled DNA content microspheres
CA2316414A1 (en) 1997-12-23 1999-07-01 Introgene B.V. Adeno-associated virus and adenovirus chimeric recombinant viruses useful for the integration of foreign genetic information into the chromosomal dna of target cells
GB2348808B (en) 1998-01-09 2003-03-19 Circassia Ltd Methods and compositions for desensitisation
US6506577B1 (en) 1998-03-19 2003-01-14 The Regents Of The University Of California Synthesis and crosslinking of catechol containing copolypeptides
US6686446B2 (en) 1998-03-19 2004-02-03 The Regents Of The University Of California Methods and compositions for controlled polypeptide synthesis
US6632922B1 (en) 1998-03-19 2003-10-14 The Regents Of The University Of California Methods and compositions for controlled polypeptide synthesis
SE9801288D0 (sv) 1998-04-14 1998-04-14 Astra Ab Vaccine delivery system and metod of production
WO1999054441A1 (en) 1998-04-22 1999-10-28 Genvec, Inc. Efficient purification of adenovirus
US6436392B1 (en) 1998-05-20 2002-08-20 University Of Iowa Research Foundation Adeno-associated virus vectors
US5965358A (en) 1998-08-26 1999-10-12 Genvec, Inc. Method for assessing the relative purity of viral gene transfer vector stocks
US6306640B1 (en) 1998-10-05 2001-10-23 Genzyme Corporation Melanoma antigenic peptides
WO2000020040A1 (en) 1998-10-08 2000-04-13 University Of Massachusetts Controlling gene expression in living cells
US6759237B1 (en) 1998-11-05 2004-07-06 The Trustees Of The University Of Pennsylvania Adeno-associated virus serotype 1 nucleic acid sequences, vectors and host cells containing same
US6764675B1 (en) 1999-06-08 2004-07-20 The Uab Research Foundation Herpes simplex virus expressing foreign genes and method for treating cancers therewith
DE19951970A1 (de) 1999-10-28 2001-05-03 Bionetworks Gmbh Arzneimittel für die Toleranzinduktion
JP4751556B2 (ja) 2000-02-28 2011-08-17 ジーンシーグス, インコーポレイテッド ナノカプセルカプセル化システムおよび方法
WO2001068133A1 (en) 2000-03-14 2001-09-20 Genetics Institute, Inc. Use of rapamycin and agents that inhibit b7 activity in immunomodulation
US6168941B1 (en) 2000-04-07 2001-01-02 Genvec, Inc. Method of producing adenoviral vector stocks
EP1278542A2 (en) 2000-05-05 2003-01-29 Cytos Biotechnology AG Molecular antigen arrays and vaccines
AU2001265187A1 (en) 2000-05-30 2001-12-11 Baylor College Of Medicine Chimeric viral vectors for gene therapy
CA2410828C (en) 2000-06-01 2012-01-24 University Of North Carolina At Chapel Hill Duplexed parvovirus vectors
US20040204379A1 (en) 2000-06-19 2004-10-14 Cheng Seng H. Combination enzyme replacement, gene therapy and small molecule therapy for lysosomal storage diseases
AU2001269923A1 (en) 2000-06-19 2002-01-02 Genzyme Corporation Combination enzyme replacement, gene therapy and small molecule therapy for lysosomal storage diseases
CA2319928A1 (en) 2000-09-18 2002-03-18 Vasogen Ireland Limited Apoptosis-mimicking synthetic entities and use thereof in medical treatments
EP1191097A1 (en) 2000-09-21 2002-03-27 Leids Universitair Medisch Centrum Induction of exon skipping in eukaryotic cells
US6447995B1 (en) 2000-10-04 2002-09-10 Genvec, Inc. Utilizing intrinsic fluorescence to detect adenovirus
GB0025414D0 (en) 2000-10-16 2000-11-29 Consejo Superior Investigacion Nanoparticles
CA2440367A1 (en) 2000-10-20 2002-08-15 Canji, Inc. Aptamer-mediated regulation of gene expression
US7122181B2 (en) 2000-12-19 2006-10-17 Research Development Foundation Lentiviral vector-mediated gene transfer and uses thereof
AU2002338571A1 (en) 2001-04-11 2002-11-11 Trustees Of The University Of Pennsylvania Compositions and methods for suppressing immune responses
AU2002303330B2 (en) 2001-05-31 2008-07-24 Novartis Vaccines And Diagnostics, Inc. Chimeric alphavirus replicon particles
US6913915B2 (en) 2001-08-02 2005-07-05 Phoenix Pharmacologics, Inc. PEG-modified uricase
AU2002327412A1 (en) 2001-08-02 2003-02-17 Institut Clayton De La Recherche Methods and compositions relating to improved lentiviral vector production systems
WO2003020797A1 (en) 2001-08-30 2003-03-13 The Regents Of The University Of California Transition metal initiators for controlled poly (beta-peptide) synthesis from beta-lactam monomers
CN1294268C (zh) * 2001-09-03 2007-01-10 上海三维生物技术有限公司 可在肿瘤细胞内特异性复制并扩散的重组腺病毒载体
JP4790984B2 (ja) 2001-09-06 2011-10-12 アルファヴァックス,インコーポレイテッド アルファウイルスレプリコンベクター系
EP1438075A4 (en) 2001-10-02 2006-04-19 Inst Clayton De La Rech METHOD AND COMPOSITIONS ASSOCIATED WITH LENTIVIRAL VECTORS WITH LIMITED EXPRESSION AND ITS APPLICATIONS
DK1436321T3 (da) 2001-10-19 2006-11-13 Isotechnika Inc Syntese af cyclosporinanaloger
AU2002360291A1 (en) 2001-12-17 2003-06-30 The Trustees Of The University Of Pennsylvania Adeno-associated virus (aav) serotype 8 sequences
GB0207440D0 (en) 2002-03-28 2002-05-08 Ppl Therapeutics Scotland Ltd Tolerogenic antigen-presenting cells
US20040038303A1 (en) 2002-04-08 2004-02-26 Unger Gretchen M. Biologic modulations with nanoparticles
US7485314B2 (en) 2002-05-06 2009-02-03 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Induction of antigen specific immunologic tolerance
WO2003102162A2 (en) 2002-06-04 2003-12-11 University Of Pittsburgh Of The Commonwealth System Of Higher Education Novel tolerogenic dendritic cells and therapeutic uses therefor
CA2494508A1 (en) 2002-07-03 2004-01-15 Coley Pharmaceutical Group, Inc. Nucleic acid compositions for stimulating immune responses
CA2392863A1 (en) 2002-07-08 2004-01-08 Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw A high capacity recombinant adenoviral vector for treatment of hemophilia a
US9809654B2 (en) 2002-09-27 2017-11-07 Vaccinex, Inc. Targeted CD1d molecules
ATE404196T1 (de) 2002-11-29 2008-08-15 Maria Grazia Roncarolo Rapamycin und il-10 zur behandlung von autoimmunerkrankungen
AU2003296958A1 (en) 2002-12-13 2004-07-09 Genetix Pharmaceuticals, Inc. Therapeutic retroviral vectors for gene therapy
US7510872B2 (en) 2003-02-26 2009-03-31 Nationwide Children's Hospital Recombinant adeno-associated virus production
NZ542323A (en) 2003-03-26 2008-07-31 Cytos Biotechnology Ag Melan-A peptide analogue-virus-like-particle conjugates
US7186699B2 (en) 2003-06-03 2007-03-06 Cell Genesys, Inc. Method for treating cancer by vector-mediated delivery of one or more anti-angiogenic or pro-apoptotic genes
EP1486567A1 (en) 2003-06-11 2004-12-15 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Improved adeno-associated virus (AAV) vector for gene therapy
WO2005019429A2 (en) 2003-08-22 2005-03-03 Potentia Pharmaceuticals, Inc. Compositions and methods for enhancing phagocytosis or phagocyte activity
US20060251711A1 (en) 2003-08-28 2006-11-09 Vgsk Technologies, Inc. Sterically stabilized carrier for aerosol therapeutics, compositions and methods for treating diseases of the respiratory tract of a mammal
US20080160089A1 (en) 2003-10-14 2008-07-03 Medivas, Llc Vaccine delivery compositions and methods of use
WO2005038035A2 (en) 2003-10-15 2005-04-28 University Of Iowa Research Foundation Methods for producing and using in vivo pseudotyped retroviruses
CA2548179A1 (en) 2003-12-02 2005-07-21 Cytimmune Sciences, Inc. Methods and compositions for the production of monoclonal antibodies
KR101376715B1 (ko) 2003-12-19 2014-03-27 더 유니버시티 오브 노쓰 캐롤라이나 엣 채플 힐 소프트 또는 임프린트 리소그래피를 이용하여 분리된 마이크로- 및 나노- 구조를 제작하는 방법
US7947659B2 (en) 2004-03-12 2011-05-24 Alnylam Pharmaceuticals, Inc. iRNA agents targeting VEGF
EA017893B1 (ru) 2004-04-08 2013-04-30 Лаборатуар Сероно Са Композиции, содержащие ингибитор jnk и циклоспорин
ES2361325T3 (es) 2004-06-28 2011-06-16 The University Of Western Australia Oligonucleótidos antisentido para inducir la omisión de exón y métodos de uso de los mismos.
CA2571899A1 (en) 2004-07-01 2006-08-03 Yale University Targeted and high density drug loaded polymeric materials
FR2874384B1 (fr) * 2004-08-17 2010-07-30 Genethon Vecteur viral adeno-associe pour realiser du saut d'exons dans un gene codant une proteine a domaines dispensables
JP2008514202A (ja) 2004-09-24 2008-05-08 ニュークレオニクス・インコーポレイテッド Rnaiによる一本鎖ウイルスの逆鎖複製中間体のターゲティング
EP1812061A2 (en) 2004-10-05 2007-08-01 Tanox Inc. Treatment and prevention of hypersensitivity and/or anaphylaxis with anti-ige antibodies in patients receiving replacement therapy
US20060089324A1 (en) 2004-10-22 2006-04-27 Sailen Barik RNAi modulation of RSV, PIV and other respiratory viruses and uses thereof
US7790878B2 (en) 2004-10-22 2010-09-07 Alnylam Pharmaceuticals, Inc. RNAi modulation of RSV, PIV and other respiratory viruses and uses thereof
US20090028948A1 (en) 2004-12-31 2009-01-29 Iceutica Pty Ltd Nanoparticle composition and methods of synthesis thereof
NZ560936A (en) 2005-02-03 2010-04-30 Benitec Inc RNAI expression constructs
MX2007010008A (es) 2005-02-16 2008-01-18 Lentigen Corp Vectores lentivirales y su uso.
GB0504206D0 (en) 2005-03-01 2005-04-06 Glaxo Group Ltd Combination therapy
CN101137365A (zh) 2005-03-08 2008-03-05 生命周期药物公司 包含西罗莫司和/或其类似物的药物组合物
CA2601312A1 (en) 2005-03-17 2006-09-28 Elan Pharma International Limited Injectable compositions of nanoparticulate immunosuppressive compounds
US7884109B2 (en) 2005-04-05 2011-02-08 Wyeth Llc Purine and imidazopyridine derivatives for immunosuppression
CA2603987A1 (en) 2005-04-12 2006-10-19 Wisconsin Alumni Research Foundation Micelle composition of polymer and passenger drug
AU2006243960A1 (en) 2005-05-10 2006-11-16 Emory University Strategies for delivery of active agents using micelles and particles
CA2609276A1 (en) 2005-05-23 2006-11-30 De-Chu C. Tang Rapid production of adenovirus-free recombinant adenovirus vectors
TW200711649A (en) 2005-06-17 2007-04-01 Combinatorx Inc Combination therapy for the treatment of immunoinflammatory disorders
WO2007002718A2 (en) 2005-06-27 2007-01-04 Alnylam Pharmaceuticals, Inc. Rnai modulation of hif-1 and theraputic uses thereof
WO2007024026A1 (ja) 2005-08-25 2007-03-01 Taiho Pharmaceutical Co., Ltd. T細胞認識エピトープペプチドを固定化又は内包化した生分解性ナノ粒子
CN1979166A (zh) 2005-11-30 2007-06-13 北京有色金属研究总院 一种制备免疫检测用纳米胶体金的工艺方法及其反应装置
EP2345412A1 (en) 2005-12-02 2011-07-20 The Johns Hopkins University Use of high-dose oxazaphosphorine drugs for treating immune disorders
AU2006321890A1 (en) 2005-12-08 2007-06-14 University Of Louisville Research Foundation, Inc. Methods and compositions for expanding T regulatory cells
US20070249043A1 (en) 2005-12-12 2007-10-25 Mayall Timothy P Adenoviral expression vectors
GB0526211D0 (en) 2005-12-22 2006-02-01 Oxford Biomedica Ltd Viral vectors
US20100028450A1 (en) 2006-01-25 2010-02-04 The Board Of Trustees Of The University Of Illinoi S Tolerogenic biodegradable artificial antigen presenting system
US20070190032A1 (en) 2006-02-13 2007-08-16 Oncolytics Biotech Inc. Use of Local Immune Suppression to Enhance Oncolytic Viral Therapy
US8021689B2 (en) 2006-02-21 2011-09-20 Ecole Polytechnique Federale de Lausanne (“EPFL”) Nanoparticles for immunotherapy
US8568487B2 (en) 2006-02-27 2013-10-29 Biomet Manufacturing, Llc Patient-specific hip joint devices
CN101448848B (zh) 2006-03-27 2013-12-04 全球免疫股份有限公司 Ras突变及其相关组合物和方法
WO2007127377A2 (en) 2006-04-28 2007-11-08 Resolvyx Pharmaceuticals, Inc. Combinations comprising omega-3 fatty acid compounds for the treatment of cardiovascular disease
JP5551432B2 (ja) 2006-05-25 2014-07-16 サンガモ バイオサイエンシーズ, インコーポレイテッド 遺伝子不活性化のための方法と組成物
DK3023500T3 (da) 2006-06-21 2020-04-20 Uniqure Ip Bv Insektceller til fremstilling af AAV-vektorer
EP1880729A1 (en) 2006-07-20 2008-01-23 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of soluble CD160 to suppress immunity
EP2051987B1 (en) 2006-08-18 2014-11-19 Argos Therapeutics, Inc. Use of cd83 in combination therapies
US20120269774A1 (en) 2006-09-21 2012-10-25 Medistem Laboratories, Inc Allogeneic stem cell transplants in non-conditioned recipients
WO2008073558A2 (en) 2006-10-05 2008-06-19 The Johns Hopkins University Water-dispersible oral, parenteral, and topical formulations for poorly water soluble drugs using smart polymeric nanoparticles
CN101646418B (zh) 2006-10-12 2013-07-17 昆士兰大学 调节免疫应答的组合物和方法
CA2702340C (en) 2006-10-12 2014-12-16 The University Of Queensland Compositions and methods for modulating immune responses
US20100112077A1 (en) 2006-11-06 2010-05-06 Abraxis Bioscience, Llc Nanoparticles of paclitaxel and albumin in combination with bevacizumab against cancer
WO2008064357A2 (en) 2006-11-22 2008-05-29 University Of Florida Research Foundation, Inc. Nanoparticles for protection of cells from oxidative stress
WO2008069942A2 (en) 2006-12-05 2008-06-12 Biogen Idec Ma Inc. Novel methods of enhancing delivery of a gene therapy vector using steroids
EP3070169B1 (en) 2006-12-14 2018-05-09 Dow AgroSciences LLC Optimized non-canonical zinc finger proteins
JP5694668B2 (ja) 2006-12-29 2015-04-01 ザ リージェンツ オブ ザ ユニバーシティ オブ コロラド,ア ボディー コーポレイトTHE REGENTS OF THE UNIVERSITY OF COLORADO,a body corporate 自己免疫疾患の診断上および治療上の標的ならびにその用途
JP5357782B2 (ja) 2007-02-21 2013-12-04 バクシネックス インコーポレーティッド 抗原負荷CD1d分子によるNKT細胞活性の調節
KR20080078204A (ko) 2007-02-22 2008-08-27 크레아젠 주식회사 면역억제능이 증가된 간엽줄기세포-매개 자가유래수지상세포
CA2680227C (en) 2007-03-07 2021-01-26 Uti Limited Partnership Compositions and methods for the prevention and treatment of autoimmune conditions
PT2131821T (pt) 2007-03-07 2018-10-10 Abraxis Bioscience Llc Nanopartícula que compreende rapamicina e albumina como agente anticancerígeno
US9114071B2 (en) 2007-04-04 2015-08-25 Sigmoid Pharma Limited Oral pharmaceutical composition
WO2008124639A2 (en) 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Poly (amino acid) targeting moieties
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
US8877917B2 (en) 2007-04-23 2014-11-04 Alnylam Pharmaceuticals, Inc. Glycoconjugates of RNA interference agents
US20080311140A1 (en) 2007-05-29 2008-12-18 Baylor College Of Medicine Antigen specific immunosuppression by dendritic cell therapy
WO2008150868A1 (en) 2007-05-29 2008-12-11 The Board Of Trustees Of The University Of Illinois Methods for inducing therapeutic t cells for immune diseases
EA200901621A1 (ru) 2007-06-05 2010-06-30 Новартис Аг Индукция толерогенного фенотипа у зрелых дендритных клеток
US20090004259A1 (en) 2007-06-14 2009-01-01 Consejo Nacional De Investigaciones Cientificas Y Tecnicas (Conicet) Methods of preparing a therapeutic formulation comprising galectin-induced tolerogenic dendritic cells
KR20100042280A (ko) 2007-07-09 2010-04-23 아스트라제네카 아베 Mtor 키나제 및/또는 pi3k와 연관된 질환에 사용되는 모르폴리노 피리미딘 유도체
GB0714963D0 (en) 2007-08-01 2007-09-12 Novartis Ag Compositions comprising antigens
ES2708856T3 (es) 2007-08-03 2019-04-11 Pasteur Institut Vectores de transferencia de gen lentivírico y sus aplicaciones medicinales
EP2083856B1 (en) 2007-08-15 2010-09-29 Circassia Limited Peptides for desensibilization against allergens
CA2700378A1 (en) 2007-09-21 2009-03-29 Cytimmune Sciences, Inc. Nanotherapeutic colloidal metal compositions and methods
US10736848B2 (en) 2007-10-12 2020-08-11 Massachusetts Institute Of Technology Vaccine nanotechnology
ES2422291T3 (es) 2007-10-31 2013-09-10 Prec Biosciences Inc Meganucleasas monocatenarias diseñadas racionalmente con secuencias de reconocimiento no palindrómicas
WO2009062502A1 (en) 2007-11-13 2009-05-22 Dandrit Biotech A/S Method for generating tolerogenic dendritic cells employing decreased temperature
JP2011505809A (ja) 2007-12-07 2011-03-03 プレシジョン バイオサイエンシズ,インク. ヒトゲノムのDNase高感受性領域に見出される認識配列を有する合理的に設計されたメガヌクレアーゼ
CN101970051A (zh) * 2007-12-31 2011-02-09 纳诺科尔治疗公司 用于治疗心力衰竭的rna干扰
WO2009100394A2 (en) 2008-02-08 2009-08-13 Terumo Kabushiki Kaisha Device for local intraluminal transport of a biologically and physiologically active agent
EP2262489A2 (en) 2008-02-28 2010-12-22 Deutsches Krebsforschungszentrum, Stiftung des öffentlichen Rechts Hollow nanoparticles and uses thereof
WO2009114321A2 (en) 2008-03-11 2009-09-17 Precision Biosciencs, Inc. Rationally-designed meganucleases for maize genome engineering
WO2009146179A1 (en) 2008-04-15 2009-12-03 University Of Iowa Research Foundation Zinc finger nuclease for the cftr gene and methods of use thereof
CA2722184A1 (en) 2008-04-25 2009-10-29 Duke University Regulatory b cells and their uses
JP5727367B2 (ja) 2008-05-15 2015-06-03 ダイナバックス テクノロジーズ コーポレイション 免疫賦活オリゴヌクレオチドを使用する長期疾患改良
JP5549014B2 (ja) 2008-05-27 2014-07-16 国立大学法人名古屋大学 免疫調節剤及びその利用
US8613951B2 (en) 2008-06-16 2013-12-24 Bind Therapeutics, Inc. Therapeutic polymeric nanoparticles with mTor inhibitors and methods of making and using same
ES2462090T5 (es) 2008-06-16 2017-07-12 Pfizer Inc. Nanopartículas poliméricas cargadas de fármaco y procedimientos de fabricación y uso de las mismas
US20110070154A1 (en) 2008-08-13 2011-03-24 Hyde Roderick A Artificial cells
EP2331124A1 (en) 2008-08-15 2011-06-15 Circassia Limited T-cell antigen peptide from allergen for stimulation of il-10 production
US8323696B2 (en) 2008-08-29 2012-12-04 Ecole Polytechnique Federale De Lausanne Nanoparticles for immunotherapy
JP5702723B2 (ja) 2008-09-04 2015-04-15 ザ ジェネラル ホスピタル コーポレイション 声帯および軟組織の増強および修復用ヒドロゲル
US8871515B2 (en) 2008-09-17 2014-10-28 Isogenis, Inc. Construction of fully-deleted adenovirus-based gene delivery vectors and uses thereof
CN101676291B (zh) 2008-09-18 2012-05-09 上海海和药物研究开发有限公司 一类雷帕霉素碳酸酯类似物、其药物组合物及其制备方法和用途
US8889124B2 (en) 2008-09-25 2014-11-18 The Board Of Trustees Of The Leland Stanford Junior University Tolerogenic populations of dendritic cells
US10369204B2 (en) 2008-10-02 2019-08-06 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
US8591905B2 (en) 2008-10-12 2013-11-26 The Brigham And Women's Hospital, Inc. Nicotine immunonanotherapeutics
US8277812B2 (en) 2008-10-12 2012-10-02 Massachusetts Institute Of Technology Immunonanotherapeutics that provide IgG humoral response without T-cell antigen
JP5552630B2 (ja) 2008-10-24 2014-07-16 学校法人 聖マリアンナ医科大学 Htlv−i関連脊髄症を治療または予防するための医薬、およびhtlv−i関連脊髄症の患者に対する抗体療法の効果を試験する方法
EP4174178A1 (en) 2008-10-24 2023-05-03 Sarepta Therapeutics, Inc. Multiple exon skipping compositions for dmd
US20120015899A1 (en) 2008-10-25 2012-01-19 Plant Bioscience, Limited Modified plant virus particles and uses therefor
ES2776126T3 (es) 2008-12-15 2020-07-29 Pfizer Nanopartículas de circulación prolongada para la liberación sostenida de agentes terapéuticos
TW201039843A (en) 2009-01-20 2010-11-16 Myelin Repair Foundation Inc Compositions and methods for induced tolerance
EP2218784A1 (en) * 2009-02-04 2010-08-18 Universität Leipzig Vector(s) containing an inducible gene encoding a CDK4/CD6 inhibitor useful for treating neurodegenerative disorders
CN102307893B (zh) 2009-02-05 2014-03-26 切尔卡西亚有限公司 用于疫苗的草肽
KR20100099849A (ko) 2009-03-04 2010-09-15 동국대학교 산학협력단 면역억제제와 트랜스글루타미나제 2의 억제제를 함유한 아토피 피부염 치료용 조성물
WO2010114948A2 (en) 2009-04-02 2010-10-07 University Of Florida Research Foundation, Inc. An inducible system for highly efficient production of recombinant adeno-associated virus (raav) vectors
GB0906159D0 (en) 2009-04-09 2009-05-20 Summit Corp Plc Drug combination for the treatment of proteostatic diseases
MX2011011134A (es) 2009-04-21 2012-01-31 Selecta Biosciences Inc Agentes inmunonanoterapeuticos que proporcionan una respuesta sesgada hacia th1.
JP5740390B2 (ja) 2009-04-27 2015-06-24 イミューロン リミテッドImmuron Limited 病的障害の治療および/または予防において使用するための抗lps強化免疫グロブリン製剤
KR20180026571A (ko) 2009-05-27 2018-03-12 셀렉타 바이오사이언시즈, 인크. 면역조절제를 pH 감응성으로 방출하는 표적화된 합성 나노운반체
US9377454B2 (en) 2009-06-25 2016-06-28 Crealta Pharmaceuticals Llc Methods and kits for predicting infusion reaction risk and antibody-mediated loss of response by monitoring serum uric acid during pegylated uricase therapy
AU2010293059B2 (en) 2009-08-26 2017-03-16 Selecta Biosciences, Inc. Compositions that induce T cell help
AR078161A1 (es) 2009-09-11 2011-10-19 Hoffmann La Roche Formulaciones farmaceuticas muy concentradas de un anticuerpo anti cd20. uso de la formulacion. metodo de tratamiento.
EP2305277A1 (en) 2009-09-18 2011-04-06 Forskarpatent I Syd AB Use of tolerogenic dendritic cells in treatment and prevention of atherosclerosis
EP2480659A2 (en) 2009-09-24 2012-08-01 Cellectis Meganuclease reagents of uses thereof for treating genetic diseases caused by frame shift/non sense mutations
US20120283318A1 (en) 2009-10-05 2012-11-08 Mei Ya-Fang Replicating viral vectors for gene therapy
CN101703781A (zh) 2009-10-28 2010-05-12 陕西北美基因股份有限公司 一种免疫抑制剂的磁性载药方法
US8956828B2 (en) 2009-11-10 2015-02-17 Sangamo Biosciences, Inc. Targeted disruption of T cell receptor genes using engineered zinc finger protein nucleases
EP3431603A1 (en) 2009-11-12 2019-01-23 The University Of Western Australia Antisense molecules and methods for treating pathologies
CN106834320B (zh) 2009-12-10 2021-05-25 明尼苏达大学董事会 Tal效应子介导的dna修饰
KR101267813B1 (ko) 2009-12-30 2013-06-04 주식회사 삼양바이오팜 향상된 수용해도를 갖는 라파마이신 함유 고분자나노입자 주사제형 조성물 및 그 제조방법, 및 방사선 요법과 병용하기 위한 항암 조성물
WO2011085231A2 (en) 2010-01-08 2011-07-14 Selecta Biosciences, Inc. Synthetic virus-like particles conjugated to human papillomavirus capsid peptides for use as vaccines
WO2011097511A1 (en) 2010-02-05 2011-08-11 The United States Of America, As Represented By The Secretary Department Of Health & Human Services REGULATORY B CELLS (tBREGS) AND THEIR USE
EP2660318A1 (en) 2010-02-09 2013-11-06 Sangamo BioSciences, Inc. Targeted genomic modification with partially single-stranded donor molecules
WO2011109833A2 (en) 2010-03-05 2011-09-09 President And Fellows Of Harvard College Induced dendritic cell compositions and uses thereof
US20110262491A1 (en) 2010-04-12 2011-10-27 Selecta Biosciences, Inc. Emulsions and methods of making nanocarriers
US20110272836A1 (en) 2010-04-12 2011-11-10 Selecta Biosciences, Inc. Eccentric vessels
WO2011139911A2 (en) 2010-04-29 2011-11-10 Isis Pharmaceuticals, Inc. Lipid formulated single stranded rna
US8927514B2 (en) 2010-04-30 2015-01-06 City Of Hope Recombinant adeno-associated vectors for targeted treatment
BR112012028557A2 (pt) 2010-05-07 2019-09-24 Xoma Technology Ltd. uso de um anticorpo anti-il-1b ou de fragmentos de ligação do mesmo.
MX2012013713A (es) 2010-05-26 2013-01-28 Selecta Biosciences Inc Composiciones de nanovehiculos con adyuvante no acoplado.
NZ707377A (en) 2010-06-07 2015-09-25 Abraxis Bioscience Llc Combination therapy methods for treating proliferative diseases
US10131875B2 (en) 2010-08-04 2018-11-20 Duke University Regulatory B cells and their uses
CA2807942C (en) 2010-08-10 2021-07-27 Ecole Polytechnique Federale De Lausanne Erythrocyte-binding therapeutics
US20120064110A1 (en) 2010-08-20 2012-03-15 Selecta Biosciences, Inc. Synthetic nanocarrier vaccines comprising peptides obtained or derived from human influenza a virus hemagglutinin
CN103079589A (zh) 2010-08-23 2013-05-01 西莱克塔生物科技公司 用于诱导对抗原的免疫应答的靶向多表位剂型
CA2810842C (en) 2010-09-09 2018-06-26 Micell Technologies, Inc. Macrolide dosage forms
CA2815422A1 (en) 2010-10-22 2012-04-26 University Of Florida Research Foundation, Inc. Antigen-specific, tolerance-inducing microparticles and uses thereof
US9994443B2 (en) 2010-11-05 2018-06-12 Selecta Biosciences, Inc. Modified nicotinic compounds and related methods
WO2012092552A1 (en) 2010-12-30 2012-07-05 Selecta Biosciences, Inc. Synthetic nanocarriers with reactive groups that release biologically active agents
CA2826836A1 (en) 2011-02-08 2012-08-16 The Charlotte-Mecklenburg Hospital Authority D/B/A Carolinas Medical Center Antisense oligonucleotides
WO2012112578A2 (en) 2011-02-14 2012-08-23 The Children's Hospital Of Philadelphia Improved aav8 vector with enhanced functional activity and methods of use thereof
US8654487B2 (en) 2011-03-11 2014-02-18 Siemens Industry, Inc. Methods, systems, and apparatus and for detecting parallel electrical arc faults
EA201391392A1 (ru) 2011-03-25 2014-04-30 Селекта Байосайенсиз, Инк. Синтетические наноносители с осмотически опосредованным высвобождением
WO2012145509A2 (en) 2011-04-19 2012-10-26 The Research Foundation Of State University Of New York Adeno-associated-virus rep sequences, vectors, and viruses
EP3699286A1 (en) 2011-04-20 2020-08-26 The Trustees of the University of Pennsylvania Regimens and compositions for aav-mediated passive immunization of airborne pathogens
EP3679933A1 (en) * 2011-04-29 2020-07-15 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers to reduce immune responses to therapeutic proteins
CN109620830A (zh) 2011-05-16 2019-04-16 建新公司 使用甲氨蝶呤诱导免疫耐受性
JP2014521687A (ja) 2011-07-29 2014-08-28 セレクタ バイオサイエンシーズ インコーポレーテッド 体液性および細胞傷害性tリンパ球(ctl)免疫応答を生成する合成ナノキャリア
US20130058894A1 (en) 2011-09-06 2013-03-07 Selecta Biosciences, Inc. Therapeutic protein-specific induced tolerogenic dendritic cells and methods of use
EP2753362A4 (en) 2011-09-08 2015-04-15 Univ Florida MATERIALS AND METHODS FOR MODULATING THE IMMUNE RESPONSE
US8865487B2 (en) 2011-09-20 2014-10-21 General Electric Company Large area hermetic encapsulation of an optoelectronic device using vacuum lamination
US20130085139A1 (en) 2011-10-04 2013-04-04 Royal Holloway And Bedford New College Oligomers
SG11201400701WA (en) 2011-10-11 2014-08-28 Aliophtha Ag Regulation of receptor expression through delivery of artificial transcription factors
WO2013058812A1 (en) 2011-10-19 2013-04-25 President And Fellows Of Harvard College Targeted delivery to pancreatic islet endothelial cells
EP2591801A1 (en) 2011-11-14 2013-05-15 Universitätsklinikum Hamburg-Eppendorf Nanoparticle compositions for generation of regulatory T cells and treatment of autoimmune diseases and other chronic inflammatory conditions
IL293434B2 (en) 2011-11-18 2024-05-01 Alnylam Pharmaceuticals Inc RNAI factors, preparations and methods of using them for the treatment of transthyretin-related diseases
JP6348064B2 (ja) 2011-11-22 2018-06-27 ザ チルドレンズ ホスピタル オブ フィラデルフィア 効率の高いトランスジーン送達のためのウイルスベクター
CN104520428B (zh) 2012-02-17 2018-09-21 费城儿童医院 将基因转移到细胞、器官和组织的aav载体组合物和方法
RU2650811C2 (ru) 2012-02-24 2018-04-17 Фред Хатчинсон Кэнсер Рисерч Сентер Композиции и способы лечения гемоглобинопатии
CN103316356B (zh) 2012-03-22 2016-08-17 北京三诺佳邑生物技术有限责任公司 一种重组慢病毒载体制剂
US9506041B2 (en) 2012-03-26 2016-11-29 The United States Of America, As Represented By The Secretary, Dept. Of Health And Human Services Delivery of packaged RNA to mammalian cells
RU2683497C2 (ru) 2012-04-18 2019-03-28 Дзе Чилдрен'З Хоспитал Оф Филадельфия Композиция и способы высокоэффективного переноса генов с помощью вариантов капсида aav
CN109876139A (zh) 2012-04-24 2019-06-14 俄亥俄州国家创新基金会 用于治疗和预防猪繁殖与呼吸综合征的组合物和方法
US9738879B2 (en) 2012-04-27 2017-08-22 Duke University Genetic correction of mutated genes
WO2014022739A2 (en) 2012-08-03 2014-02-06 Alnylam Pharmaceuticals, Inc. Modified rnai agents
US20150231241A1 (en) 2012-08-14 2015-08-20 Ibc Pharmaceuticals, Inc. Combination therapy for inducing immune response to disease
ES2878650T3 (es) 2012-09-06 2021-11-19 Univ Chicago Polinucleótidos antisentido para inducir la omisión de exón y procedimientos de tratamiento de distrofias
US20150224209A1 (en) 2012-09-14 2015-08-13 The Regents Of The University Of California Lentiviral vector for stem cell gene therapy of sickle cell disease
US9181535B2 (en) 2012-09-24 2015-11-10 The Chinese University Of Hong Kong Transcription activator-like effector nucleases (TALENs)
CN102871966B (zh) 2012-10-19 2013-11-20 东南大学 用于改善雷帕霉素生物利用度的纳米载药颗粒及其制备方法
US9624510B2 (en) 2013-03-01 2017-04-18 The Wistar Institute Adenoviral vectors comprising partial deletions of E3
RU2015142245A (ru) 2013-03-13 2017-04-18 Кур Фармасьютикалз Дивелопмент Компани Иммуномодифицирующие частицы для лечения воспаления
WO2014145524A2 (en) 2013-03-15 2014-09-18 Haplomics, Inc. Compositions and methods for immune tolerance induction to factor viii replacement therapies in subjects with hemophilia a
EA201591792A1 (ru) 2013-03-15 2016-02-29 Сарепта Терапьютикс, Инк. Улучшенные композиции для лечения мышечной дистрофии
US9828587B2 (en) 2013-04-08 2017-11-28 University Of Iowa Research Foundation Chimeric adeno-associated virus/ bocavirus parvovirus vector
KR20220025911A (ko) 2013-05-03 2022-03-03 셀렉타 바이오사이언시즈, 인크. Cd4+ 조절 t 세포를 증진시키기 위한 방법 및 조성물
CA2910579C (en) 2013-05-03 2023-09-26 Selecta Biosciences, Inc. Dosing combinations for reducing undesired humoral immune responses
RU2015154721A (ru) 2013-05-22 2017-06-27 Элнилэм Фармасьютикалз, Инк. КОМПОЗИЦИИ НА ОСНОВЕ RNAi SERPINA1 И СПОСОБЫ ИХ ПРИМЕНЕНИЯ
US11414695B2 (en) 2013-05-29 2022-08-16 Agilent Technologies, Inc. Nucleic acid enrichment using Cas9
HUE052233T2 (hu) 2013-06-04 2021-04-28 Selecta Biosciences Inc Nem immunszuppresszív, antigénre specifikus immunterápiás ágensek ismételt beadása
US9526784B2 (en) 2013-09-06 2016-12-27 President And Fellows Of Harvard College Delivery system for functional nucleases
US9388430B2 (en) 2013-09-06 2016-07-12 President And Fellows Of Harvard College Cas9-recombinase fusion proteins and uses thereof
WO2015051199A2 (en) 2013-10-06 2015-04-09 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Modified pseudomonas exotoxin a
US20150166985A1 (en) 2013-12-12 2015-06-18 President And Fellows Of Harvard College Methods for correcting von willebrand factor point mutations
US9300930B2 (en) * 2014-02-18 2016-03-29 Microvision, Inc. Multi-segment brightness compensation
CN105377481B (zh) * 2014-02-27 2018-02-06 宁夏东方钽业股份有限公司 一种高纯钽粉及其制备方法
US10339920B2 (en) * 2014-03-04 2019-07-02 Amazon Technologies, Inc. Predicting pronunciation in speech recognition
EP2916319A1 (en) * 2014-03-07 2015-09-09 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Concept for encoding of information
WO2015138357A2 (en) 2014-03-09 2015-09-17 The Trustees Of The University Of Pennsylvania Compositions useful in treatment of otc deficency
GB201407322D0 (en) 2014-04-25 2014-06-11 Ospedale San Raffaele Gene therapy
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
WO2015200728A1 (en) 2014-06-25 2015-12-30 Selecta Biosciences, Inc. Methods and compositions for treatment with synthetic nanocarriers and immune checkpoint inhibitors
EA201790977A1 (ru) 2014-11-05 2017-10-31 Селекта Байосайенсиз, Инк. Способы и композиции, относящиеся к применению поверхностно-активных веществ с низким показателем гидрофильно-липофильного баланса (hlb) при получении синтетических наноносителей, включающих рапалог
WO2017139212A1 (en) 2016-02-08 2017-08-17 Cyta Therapeutics, Inc. Particle delivery of rapamycin to the liver
CA3013896A1 (en) 2016-02-10 2017-08-17 Pfizer Inc. Therapeutic nanoparticles having egfr ligands and methods of making and using same
EP3426285B1 (en) 2016-03-11 2024-05-29 Cartesian Therapeutics, Inc. Formulations and doses of pegylated uricase
US10545815B2 (en) * 2016-08-03 2020-01-28 Futurewei Technologies, Inc. System and method for data redistribution in a database
WO2018039465A1 (en) 2016-08-25 2018-03-01 Selecta Biosciences, Inc. Polyester polymer matrices for the delivery of allergens
EP3518956A1 (en) 2016-09-27 2019-08-07 Selecta Biosciences, Inc. Recombinant immunotoxins for use in the treatment of cancer
US11104887B2 (en) 2017-01-03 2021-08-31 Ethris Gmbh Ornithine transcarbamylase coding polyribonucleotides and formulations thereof
KR20190104194A (ko) 2017-01-07 2019-09-06 셀렉타 바이오사이언시즈, 인크. 합성 나노담체에 커플링된 면역억제제의 패턴화된 투여
MX2019010757A (es) 2017-03-11 2020-01-20 Selecta Biosciences Inc Métodos y composiciones relacionados con el tratamiento combinado con antiinflamatorios y nanoportadores sintéticos que comprenden un inmunosupresor.
KR20200086670A (ko) 2017-10-13 2020-07-17 셀렉타 바이오사이언시즈, 인크. 항바이러스 전달 벡터 IgM 반응을 약화시키기 위한 방법 및 조성물
JP7416430B2 (ja) 2018-02-26 2024-01-17 イッサム リサーチ ディベロップメント カンパニー オブ ザ ヘブライ ユニバーシティー オブ エルサレム リミテッド 薬物送達システム
WO2019217552A1 (en) 2018-05-09 2019-11-14 Yale University Particles for spatiotemporal release of agents
JP2021530571A (ja) 2018-07-16 2021-11-11 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. Mmaコンストラクトおよびベクターの方法および組成物
CN112771070A (zh) 2018-07-16 2021-05-07 西莱克塔生物科技公司 Otc构建体和载体的方法和组合物
MX2021013163A (es) 2019-04-28 2022-02-21 Selecta Biosciences Inc Métodos para el tratamiento de sujetos con inmunidad preexistente a vectores de transferencia viral.
MX2021014566A (es) 2019-05-28 2022-03-22 Selecta Biosciences Inc Métodos y composiciones para la respuesta inmune atenuada anti-vector de transferencia viral.
CN114207440A (zh) 2019-06-04 2022-03-18 西莱克塔生物科技公司 聚乙二醇化尿酸酶的制剂和剂量
JP2022553345A (ja) 2019-10-21 2022-12-22 セレクタ バイオサイエンシーズ インコーポレーテッド 肝疾患および肝障害を処置するための方法および組成物
MX2022005506A (es) 2019-11-08 2022-08-10 Selecta Biosciences Inc Formulaciones y dosis de uricasa pegilada.
CA3174988A1 (en) 2020-03-11 2021-09-16 Selecta Biosciences, Inc. Methods and compositions related to synthetic nanocarriers

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6251957B1 (en) * 1995-02-24 2001-06-26 Trustees Of The University Of Pennsylvania Method of reducing an immune response to a recombinant virus
US20020014242A1 (en) * 2000-07-31 2002-02-07 Abraham Scaria Use of rapamycin to inhibit immune response and induce tolerance to gene therapy vector and encoded transgene products
US20100129439A1 (en) * 2008-10-12 2010-05-27 Frank Alexis Adjuvant Incorporation in Immunonanotherapeutics
US20100273220A1 (en) * 2009-04-22 2010-10-28 Massachusetts Institute Of Technology Innate immune suppression enables repeated delivery of long rna molecules
US11633422B2 (en) * 2014-09-07 2023-04-25 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector immune responses

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Aruda et al 2009, Molecular Therapy 17:1492-1503. *
Fang et al, Gene Therapy for Hemophilia B: Host Immunosuppression Prolongs the Therapeutic Effect of Adenovirus-Mediated Factor IX Expression, Human Gene Therapy 6: 1039-1044, 1995 *
Jhunjhunwala et al (2009, J. Controlled Release 133:191-197. *

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9884112B2 (en) 2009-05-27 2018-02-06 Selecta Biosciences, Inc. Immunomodulatory agent-polymeric compounds
US9764031B2 (en) 2010-05-26 2017-09-19 Selecta Biosciences, Inc. Dose selection of adjuvanted synthetic nanocarriers
US9994443B2 (en) 2010-11-05 2018-06-12 Selecta Biosciences, Inc. Modified nicotinic compounds and related methods
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
US10004802B2 (en) 2011-04-29 2018-06-26 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for generating CD8+ regulatory T cells
US10039822B2 (en) 2011-04-29 2018-08-07 Selecta Biosciences, Inc. Method for providing polymeric synthetic nanocarriers for generating antigen-specific tolerance immune responses
US11779641B2 (en) 2011-04-29 2023-10-10 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for allergy therapy
US11717569B2 (en) 2011-04-29 2023-08-08 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers
US9993548B2 (en) 2011-04-29 2018-06-12 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for inducing regulatory B cells
US11235057B2 (en) 2011-04-29 2022-02-01 Selecta Biosciences, Inc. Methods for providing polymeric synthetic nanocarriers for generating antigen-specific tolerance immune responses
US9987354B2 (en) 2011-04-29 2018-06-05 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for antigen-specific deletion of T effector cells
US10933129B2 (en) 2011-07-29 2021-03-02 Selecta Biosciences, Inc. Methods for administering synthetic nanocarriers that generate humoral and cytotoxic T lymphocyte responses
US10335395B2 (en) 2013-05-03 2019-07-02 Selecta Biosciences, Inc. Methods of administering immunosuppressants having a specified pharmacodynamic effective life and therapeutic macromolecules for the induction of immune tolerance
US10434088B2 (en) 2013-05-03 2019-10-08 Selecta Biosciences, Inc. Methods related to administering immunosuppressants and therapeutic macromolecules at a reduced pharmacodynamically effective dose
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
US10357482B2 (en) 2013-05-03 2019-07-23 Selecta Biosciences, Inc. Methods providing a therapeutic macromolecule and synthetic nanocarriers comprising immunosuppressant locally and concomitantly to reduce both type I and type IV hypersensitivity
US10357483B2 (en) 2013-05-03 2019-07-23 Selecta Biosciences, Inc. Methods comprising dosing combinations for reducing undesired humoral immune responses
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
US11633422B2 (en) 2014-09-07 2023-04-25 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector immune responses
US10071114B2 (en) 2014-09-07 2018-09-11 Selecta Biosciences, Inc. Methods and compositions for attenuating gene expression modulating anti-viral transfer vector immune responses
US10046064B2 (en) 2014-09-07 2018-08-14 Selecta Biosciences, Inc. Methods and compositions for attenuating exon skipping anti-viral transfer vector immune responses
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
WO2022221529A1 (en) 2021-04-16 2022-10-20 Asklepios Biopharmaceutical, Inc. Rational polyploid aav virions that cross the blood brain barrier and elicit reduced humoral response

Also Published As

Publication number Publication date
CN106714826A8 (zh) 2017-07-11
CN107073050A (zh) 2017-08-18
EP3193896A4 (en) 2018-04-18
MX2023003254A (es) 2023-04-11
JP2021152013A (ja) 2021-09-30
CN114146167A (zh) 2022-03-08
IL250873A0 (en) 2017-04-30
AU2015311705A1 (en) 2017-02-02
JP7395522B2 (ja) 2023-12-11
EP3189147A1 (en) 2017-07-12
CN107075499A (zh) 2017-08-18
IL250877B (en) 2022-06-01
EA201790551A1 (ru) 2017-07-31
JP2017529342A (ja) 2017-10-05
EP3189146A1 (en) 2017-07-12
JP2024004492A (ja) 2024-01-16
MX2023003252A (es) 2023-04-11
JP7242179B2 (ja) 2023-03-20
CN106715699A (zh) 2017-05-24
IL250872B (en) 2022-06-01
IL292574A (en) 2022-06-01
IL250865B2 (en) 2024-05-01
BR112017001359A2 (pt) 2017-11-21
WO2016037163A1 (en) 2016-03-10
US20190076458A1 (en) 2019-03-14
WO2016037165A1 (en) 2016-03-10
BR112017001245A2 (pt) 2017-12-05
IL250874A0 (en) 2017-04-30
AU2015311704B2 (en) 2021-12-09
JP2021100926A (ja) 2021-07-08
EP3188742A4 (en) 2018-04-18
KR20170045347A (ko) 2017-04-26
MX2023003246A (es) 2023-04-11
IL292577A (en) 2022-06-01
US20230364124A1 (en) 2023-11-16
JP2017532304A (ja) 2017-11-02
MX2017002935A (es) 2017-05-30
KR20230053710A (ko) 2023-04-21
IL250865B1 (en) 2024-01-01
JP2021152012A (ja) 2021-09-30
IL250877A0 (en) 2017-04-30
CA2957737A1 (en) 2016-03-10
JP7272746B2 (ja) 2023-05-12
US10046064B2 (en) 2018-08-14
EA201790550A1 (ru) 2017-08-31
WO2016037164A1 (en) 2016-03-10
JP2017527571A (ja) 2017-09-21
US20160074427A1 (en) 2016-03-17
AU2015311705B2 (en) 2021-12-09
JP2017532307A (ja) 2017-11-02
US20160067228A1 (en) 2016-03-10
WO2016037161A2 (en) 2016-03-10
KR20170045346A (ko) 2017-04-26
US11633422B2 (en) 2023-04-25
CN106714826A (zh) 2017-05-24
BR112017001311A2 (pt) 2017-11-14
AU2015311707A1 (en) 2017-02-02
JP2017527565A (ja) 2017-09-21
AU2021203379A1 (en) 2021-06-24
BR112017001601A2 (pt) 2017-11-21
KR20170045344A (ko) 2017-04-26
CA2957793A1 (en) 2016-03-10
MX2017002932A (es) 2017-05-30
CN106715699A8 (zh) 2017-07-11
MX2017002934A (es) 2017-05-30
CN107073091A (zh) 2017-08-18
BR112017001470A2 (pt) 2018-02-20
MX2017002933A (es) 2017-05-30
IL292575A (en) 2022-06-01
KR20170045345A (ko) 2017-04-26
IL250865A0 (en) 2017-04-30
WO2016037161A3 (en) 2016-05-06
US20160074372A1 (en) 2016-03-17
EA201790532A1 (ru) 2017-07-31
AU2015311707B2 (en) 2021-12-09
EP3188742A1 (en) 2017-07-12
JP2021176845A (ja) 2021-11-11
CA2957802A1 (en) 2016-03-10
IL250873B (en) 2022-06-01
JP7425767B2 (ja) 2024-01-31
JP2021152014A (ja) 2021-09-30
AU2015311708A1 (en) 2017-02-02
CA2957808A1 (en) 2016-03-10
WO2016037162A1 (en) 2016-03-10
JP7242177B2 (ja) 2023-03-20
EA201790534A1 (ru) 2017-07-31
KR20230053709A (ko) 2023-04-21
AU2022201350A1 (en) 2022-03-24
EP3189138A4 (en) 2018-04-18
AU2022201349A1 (en) 2022-03-24
IL250872A0 (en) 2017-04-30
IL309546A (en) 2024-02-01
US20160074532A1 (en) 2016-03-17
AU2021203376A1 (en) 2021-06-24
JP7242180B2 (ja) 2023-03-20
AU2015311704A1 (en) 2017-02-02
KR20170054434A (ko) 2017-05-17
EP3189138A2 (en) 2017-07-12
AU2015311706A1 (en) 2017-02-02
AU2022201323A1 (en) 2022-03-24
MX2017002931A (es) 2017-05-30
JP7242178B2 (ja) 2023-03-20
IL250874B (en) 2022-06-01
EP3193896A1 (en) 2017-07-26
CA2957800A1 (en) 2016-03-10
MX2023003249A (es) 2023-04-11
MX2023003253A (es) 2023-04-11
IL292580A (en) 2022-06-01
EA201790533A1 (ru) 2017-07-31
US10071114B2 (en) 2018-09-11

Similar Documents

Publication Publication Date Title
JP7395522B2 (ja) エクソンスキッピング抗ウイルス導入ベクター免疫応答を減弱化するための方法および組成物

Legal Events

Date Code Title Description
AS Assignment

Owner name: SELECTA BIOSCIENCES, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:KISHIMOTO, TAKASHI KEI;REEL/FRAME:037095/0097

Effective date: 20151009

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED