US20200113874A1 - Methods related to administering immunosuppressants and therapeutic macromolecules at a reduced pharmacodynamically effective dose - Google Patents

Methods related to administering immunosuppressants and therapeutic macromolecules at a reduced pharmacodynamically effective dose Download PDF

Info

Publication number
US20200113874A1
US20200113874A1 US16/550,725 US201916550725A US2020113874A1 US 20200113874 A1 US20200113874 A1 US 20200113874A1 US 201916550725 A US201916550725 A US 201916550725A US 2020113874 A1 US2020113874 A1 US 2020113874A1
Authority
US
United States
Prior art keywords
therapeutic
dose
immunosuppressant
therapeutic macromolecule
synthetic nanocarriers
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
US16/550,725
Inventor
Roberto A. Maldonado
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 US16/550,725 priority Critical patent/US20200113874A1/en
Publication of US20200113874A1 publication Critical patent/US20200113874A1/en
Assigned to SELECTA BIOSCIENCES, INC. reassignment SELECTA BIOSCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KISHIMOTO, TAKASHI KEI, MALDONADO, Roberto A.
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • A61K38/13Cyclosporins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • A61K38/1761Apoptosis related proteins, e.g. Apoptotic protease-activating factor-1 (APAF-1), Bax, Bax-inhibitory protein(s)(BI; bax-I), Myeloid cell leukemia associated protein (MCL-1), Inhibitor of apoptosis [IAP] or Bcl-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1767Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/195Chemokines, e.g. RANTES
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/36Blood coagulation or fibrinolysis factors
    • A61K38/37Factors VIII
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/38Albumins
    • 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
    • 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)
    • A61K38/47Hydrolases (3) acting on glycosyl compounds (3.2), e.g. cellulases, lactases
    • 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)
    • 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)
    • A61K38/50Hydrolases (3) acting on carbon-nitrogen bonds, other than peptide bonds (3.5), e.g. asparaginase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0003Invertebrate antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/593Polyesters, e.g. PLGA or polylactide-co-glycolide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • 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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • 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
    • 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
    • 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
    • A61K9/5192Processes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/575Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
    • 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
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/62Medicinal preparations containing antigens or antibodies characterised by the link between antigen and carrier
    • A61K2039/622Medicinal preparations containing antigens or antibodies characterised by the link between antigen and carrier non-covalent binding
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y305/00Hydrolases acting on carbon-nitrogen bonds, other than peptide bonds (3.5)
    • C12Y305/01Hydrolases acting on carbon-nitrogen bonds, other than peptide bonds (3.5) in linear amides (3.5.1)
    • C12Y305/01001Asparaginase (3.5.1.1)

Definitions

  • This invention relates to immunosuppressant doses, in some embodiments attached to synthetic nanocarriers that are administered concomitantly with a therapeutic macromolecule, and related methods.
  • the compositions and methods allow for efficient pharmacodynamic effects specific to the therapeutic macromolecule.
  • the compositions and methods provided can, therefore, be used to generate pharmacodynamic responses in a subject even at reduced doses of the therapeutic macromolecule.
  • the compositions and methods provided herein can also be administered repeatedly concomitantly to generate desired pharmacodynamic and immunologic effects.
  • Therapeutic treatments such as protein or enzyme replacement therapies, often result in undesired immune responses to the particular therapeutic.
  • cells of the immune system recognize the therapeutic as foreign and attempt to neutralize or destroy it, just as they attempt to destroy infecting organisms such as bacteria and viruses.
  • Such undesired immune responses can neutralize the efficacy of the therapeutic treatment or cause hypersensitive reactions to the therapeutic.
  • immunosuppressant drugs Conventional immunosuppressant drugs, however, are broad-acting, and the use of broad-acting immunosuppressants is associated with a risk of severe side effects, such as tumors, infections, nephrotoxicity and metabolic disorders. Accordingly, new therapies would be beneficial.
  • a method comprising providing an immunosuppressant dose, wherein in some embodiments the immunosuppressant dose is attached to synthetic nanocarriers, and administering a reduced pharmacodynamically effective dose of a therapeutic macromolecule concomitantly with the immunosuppressant dose to a subject.
  • the concomitant administration is according to a protocol that has been demonstrated to result in a pharmacodynamic effect with the reduced pharmacodynamically effective dose of the therapeutic macromolecule upon concomitant administration with the immunosuppressant dose, as compared to administration of the therapeutic macromolecule when not administered concomitantly with the immunosuppressant dose, and each in the presence of an anti-therapeutic macromolecule antibody response.
  • the reduced pharmacodynamically effective dose of the therapeutic macromolecule is less than a pharmacodynamically effective dose of the therapeutic macromolecule that: (A) is administered in the presence of an anti-therapeutic macromolecule antibody response, and (B) is not administered concomitantly with the immunosuppressant dose.
  • a method comprising providing an immunosuppressant dose, wherein in some embodiments the immunosuppressant dose is attached to synthetic nanocarriers, and administering a pharmacodynamically effective dose of a therapeutic macromolecule concomitantly with the immunosuppressant dose.
  • the concomitant administration is according to a protocol that has been demonstrated to enhance a pharmacodynamic effect of the therapeutic macromolecule upon concomitant administration with the immunosuppressant dose, as compared to administration of the therapeutic macromolecule when not administered concomitantly with the immunosuppressant dose, and each in the presence of an anti-therapeutic macromolecule antibody response.
  • a method comprising providing an immunosuppressant dose, wherein in some embodiments the immunosuppressant dose is attached to synthetic nanocarriers, administering a pharmacodynamically effective dose of a therapeutic macromolecule concomitantly with the immunosuppressant dose, and recording an enhanced pharmacodynamic effect following the concomitant administration is provided.
  • a method comprising providing therapeutic macromolecules that cause or are expected to cause anti-therapeutic macromolecule antibodies upon repeated dosing in one or more subjects; and providing an immunosuppressant dose, wherein the immunosuppressant dose is attached to synthetic nanocarriers.
  • the method comprises repeatedly dosing at the same or a lower dose a subject with the therapeutic macromolecules concomitantly with the immunosuppressant dose.
  • the concomitant administration is according to a protocol that has been demonstrated to result in maintenance of a pharmacodynamic effect of the therapeutic macromolecule over two or more doses of the therapeutic macromolecule to a subject.
  • the method further comprises determining the protocol. In another embodiment of any one of the methods provided, the method further comprises determining the pharmacodynamically effective dose, such as the reduced or enhanced pharmacodynamically effective dose. In another embodiment of any one of the methods provided, the method further comprises assessing the pharmacodynamic effect in the subject prior to and/or after the administration. In another embodiment of any one of the methods provided, the concomitant administration is repeated one or more times. In another embodiment of any one of the methods provided, the administering is by intravenous, intraperitoneal or subcutaneous administration. In another embodiment of any one of the methods provided, the subject is at risk of an anti-therapeutic macromolecule antibody response. In another embodiment of any one of the methods provided, this subject is one in which the anti-therapeutic macromolecule response is expected to occur.
  • composition or kit comprising an immunosuppressant dose wherein, in some embodiments, the immunosuppressant is attached to synthetic nanocarriers, and a reduced pharmacodynamically effective dose of a therapeutic macromolecule is provided.
  • composition or kit comprising a reduced pharmacodynamically effective dose of a therapeutic macromolecule for use in any one of the methods provided herein is provided in combination with an immunosuppressant dose wherein in some embodiments the immunosuppressant is attached to synthetic nanocarriers.
  • composition or kit is for use in any one of the methods provided herein. In one embodiment of any one of the compositions or kits provided, the composition or kit further comprises a pharmaceutically acceptable carrier.
  • the therapeutic macromolecule is not attached to the synthetic nanocarriers. In another embodiment of any one of the methods or compositions or kits provided, the therapeutic macromolecule is attached to the synthetic nanocarriers. In another embodiment of any one of the methods or compositions or kits provided, the synthetic nanocarriers do not comprise therapeutic macromolecule APC presentable antigens.
  • the immunosuppressant dose and the therapeutic macromolecule are each contained in a container. In another embodiment of any one of the compositions or kits provided, the immunosuppressant dose and the therapeutic macromolecule are contained in separate containers. In another embodiment of any one of the compositions or kits provided, the immunosuppressant dose and the therapeutic macromolecule are contained in the same container.
  • the reduced pharmacodynamically effective dose of the therapeutic macromolecule is at least 30% less than a pharmacodynamically effective dose of the therapeutic macromolecule that (A) is administered in the presence of an anti-therapeutic macromolecule antibody response, and (B) is not administered concomitantly with the immunosuppressant dose.
  • the reduced pharmacodynamically effective dose is at least 40% less.
  • the reduced pharmacodynamically effective dose is at least 50% less.
  • the immunosuppressant dose comprises a statin, an mTOR inhibitor, a TGF- ⁇ signaling agent, a corticosteroid, an inhibitor of mitochondrial function, a P38 inhibitor, an NF- ⁇ inhibitor, an adenosine receptor agonist, a prostaglandin E2 agonist, a phosphodiesterase 4 inhibitor, an HDAC inhibitor or a proteasome inhibitor.
  • the mTOR inhibitor is rapamycin.
  • the therapeutic macromolecule comprises a therapeutic protein. In another embodiment of any one of the methods or compositions or kits provided, the therapeutic macromolecule comprises a therapeutic polynucleotide. In another embodiment of any one of the methods or compositions or kits provided, the therapeutic protein is for protein replacement or protein supplementation therapy. In another embodiment of any one of the methods or compositions or kits provided, the therapeutic macromolecule comprises a/an infusible or injectable therapeutic protein, enzyme, enzyme cofactor, hormone, blood or blood coagulation factor, cytokine, interferon, growth factor, monoclonal antibody, polyclonal antibody or protein associated with Pompe's disease.
  • the infusible or injectable therapeutic protein comprises Tocilizumab, alpha-1 antitrypsin, Hematide, albinterferon alfa-2b, Rhucin, tesamorelin, ocrelizumab, belimumab, pegloticase, pegsiticase, taliglucerase alfa, agalsidase alfa or velaglucerase alfa.
  • the enzyme comprises an oxidoreductase, transferase, hydrolase, lyase, isomerase or ligase.
  • the enzyme comprises an enzyme for enzyme replacement therapy for a lysosomal storage disorder.
  • the enzyme for enzyme replacement therapy for a lysosomal storage disorder comprises imiglucerase, a-galactosidase A (a-gal A), agalsidase beta, acid ⁇ -glucosidase (GAA), alglucosidase alfa, LUMIZYME, MYOZYME, arylsulfatase B, laronidase, ALDURAZYME, idursulfase, ELAPRASE, arylsulfatase B or NAGLAZYME.
  • the enzymes comprise KRYSTEXXA (pegloticase).
  • the monoclonal antibody comprises HUMIRA (adalimumab).
  • the cytokine comprises a lymphokine, interleukin, chemokine, type 1 cytokine or a type 2 cytokine.
  • the blood or blood coagulation factor comprises Factor I, Factor II, tissue factor, Factor V, Factor VII, Factor VIII, Factor IX, Factor X, Factor Xa, Factor XII, Factor XIII, von Willebrand factor, prekallikrein, high-molecular weight kininogen, fibronectin, antithrombin III, heparin cofactor II, protein C, protein S, protein Z, protein Z-related protease inhibitor (ZPI), plasminogen, alpha 2-antiplasmin, tissue plasminogen activator (tPA), urokinase, plasminogen activator inhibitor-1 (PAI1), plasminogen activator inhibitor-2 (PAI2), cancer procoagulant or epoetin alfa.
  • the blood or blood coagulation factor is Factor VIII.
  • a load of immunosuppressant attached to the synthetic nanocarriers, on average across the synthetic nanocarriers, is between 0.1% and 50%. In another embodiment of any one of the methods or compositions or kits provided, the load is between 0.1% and 20%.
  • 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 lipid nanoparticles.
  • the synthetic nanocarriers comprise liposomes.
  • the synthetic nanocarriers comprise metallic nanoparticles.
  • the metallic nanoparticles comprise gold nanoparticles.
  • the synthetic nanocarriers comprise polymeric nanoparticles.
  • the polymeric nanoparticles comprise 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 the synthetic nanocarriers is a diameter greater than 100 nm. In another embodiment of any one of the methods or compositions or kits provided, the diameter is greater than 150 nm. In another embodiment of any one of the methods or compositions or kits provided, the diameter is greater than 200 nm. In another embodiment of any one of the methods or compositions or kits provided, the diameter is greater than 250 nm. In another embodiment of any one of the methods or compositions or kits provided, the diameter is greater than 300 nm.
  • an aspect ratio 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.
  • a method of manufacturing any one of the compositions or kits provided herein comprises producing a dose or dosage form of a therapeutic macromolecule and producing a dose or dosage form of an immunosuppressant.
  • the dose or dosage form of a therapeutic macromolecule is a reduced pharamcodynamically effective dose of the therapeutic macromolecule.
  • the step of producing a dose or dosage form of an immunosuppressant comprises attaching the immunosuppressant to synthetic nanocarriers.
  • the method further comprises combining the dose or dosage form of the immunosuppressant and dose or dosage form of the therapeutic macromolecule in a kit.
  • compositions or kits provided herein for the manufacture of a medicament for reducing an anti-therapeutic macromolecule antibody response, in a subject.
  • the composition or kit comprises an immunosuppressant and a therapeutic macromolecule, wherein the therapeutic macromolecule may be provided in a reduced pharmacodynamically effective dose of the therapeutic macromolecule.
  • the immunosuppressant is attached to synthetic nanocarriers.
  • the use if for achieving any one of the methods provided herein.
  • any one of the composition or kits provided herein may be for use in any one of the methods provided herein.
  • the composition or kit comprises a one or more doses or dosage forms of a therapeutic macromolecule and/or one or more doses or dosage forms of an immunosuppressant.
  • the doses of a therapeutic macromolecule are reduced pharmacodynamically effective dose.
  • the immunosuppressant is attached to synthetic nanocarriers.
  • a method of manufacturing a medicament intended for reducing an anti-therapeutic macromolecule antibody response comprises an immunosuppressant and/or a therapeutic macromolecule, wherein the therapeutic macromolecule may be at a reduced pharmacodynamically effective dose.
  • the immunosuppressant is attached to synthetic nanocarriers.
  • FIG. 1 shows the level of circulating antigen-specific antibody production with a concomitant administration as provided herein.
  • FIG. 2 shows the level of circulating antigen-specific antibody production with a concomitant administration as provided herein.
  • FIG. 3 provides the anti-OVA antibody titers with a concomitant administration as provided herein.
  • FIG. 4 provides the anti-KLH antibody titers with a concomitant administration as provided herein.
  • FIG. 5 shows the antibody recall response to FVIII one month following a final nanocarrier and FVIII dosing.
  • FIGS. 6A and 6B show the efficacy of nanocarrier and FVIII dosing in Hemophilia A mice.
  • FIGS. 7A and 7B show immune responses to HUMIRA in mice that were treated with HUMIRA/adalimumab with or without nanocarriers attached to rapamycin.
  • FIG. 8 shows anti-Keyhole Limpet Hemocyanin (KLH) antibody titers in mice that were treated with KLH with or without nanocarriers attached to rapamycin.
  • KLH Keyhole Limpet Hemocyanin
  • FIG. 9 shows anti-ovalbumin (OVA) antibody titers in mice that were treated with OVA with or without nanocarriers attached to rapamycin.
  • OVA anti-ovalbumin
  • FIG. 10 shows anti-KRYSTEXXA antibody titers in mice that were treated with KRYSTEXXA with or with nanocarriers attached to rapamycin.
  • FIG. 11 shows antibody titers in mice that were treated with OVA and KLH either in the presence or absence of nanocarriers attached to rapamycin.
  • FIGS. 12A and 12B show immune responses to KLH in mice that were treated with KLH with or without nanocarriers attached to rapamycin.
  • FIGS. 13A and 13B show immune responses to HUMIRA/adalimumab in mice that were treated with HUMIRA/adalimumab with or without nanocarriers attached to rapamycin.
  • FIG. 14 provides an exemplary protocol for practicing the methods provided herein.
  • FIG. 15 shows the beneficial effects of practicing a method provided herein in regard to therapy with HUMIRA.
  • FIG. 16 provides an exemplary protocol for practicing the methods provided herein.
  • FIG. 17 shows the beneficial effects of practicing a method provided herein in regard to therapy with HUMIRA.
  • FIG. 18 demonstrates the reduction in anti-protein antibody responses as a result of two different immunosuppressants attached to synthetic nanocarriers.
  • FIG. 19 shows another exemplary protocol and the beneficial effects of practicing a method provided herein in regard to therapy with HUMIRA.
  • 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 RNA molecule” includes a mixture of two or more such RNA molecules or a plurality of such RNA molecules
  • reference to “an immunosuppressant” includes a mixture of two or more such materials 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, element, 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, element, characteristics, properties, method/process steps or limitations) alone.
  • the methods, composition or kits provided herein can be used to improve the pharmacodynamic effect(s) of a therapeutic macromolecule in a subject in which an antibody response against the therapeutic macromolecule has been mounted. Accordingly, the methods or compositions or kits provide herein can be used to improve the pharmacodynamics effect(s) of therapeutic macromolecules that are otherwise diminished because of an anti-therapeutic macromolecule antibody response. Without being bound by a particular theory, it is thought that undesirable humoral immune responses against therapeutic macromolecules can be reduced using the methods, compositions or kits provided.
  • the methods, compositions or kits can be used to tolerize a subject against a therapeutic macromolecule reducing undesirable immune responses that would otherwise result when the therapeutic macromolecule is administered without the concomitant administration of an immunosuppressant dose as provided, such dose may be repeatedly concomitantly administered.
  • undesirable immune responses can result in the enhanced clearance of the therapeutic macromolecule, or other interference with the therapeutic activity of the therapeutic macromolecule.
  • the pharmacodynamic effect(s) of a therapeutic macromolecule can be enhanced and/or reduced dosage amounts of the therapeutic macromolecule can be used to achieve the same level of effect with the methods, compositions or kits provided.
  • repeated dosings of a therapeutic macromolecule can be administered to a subject.
  • delivering immunosuppressants preferably in some embodiments when attached to synthetic nanocarriers, concomitantly with therapeutic macromolecules, and in the presence of an anti-therapeutic macromolecule antibody response, can result in enhanced pharmacodynamic effects.
  • the aforementioned combinations can help neutralize anti-therapeutic macromolecule-specific antibodies that interfere with the desired treatment effects of a therapeutic macromolecule.
  • the methods, compositions or kits provided herein in some embodiments, not only reduce the undesired immune responses against the therapeutic macromolecule but also result in the enhancement of the desired therapeutic effect of the therapeutic macromolecule that would otherwise be diminished when the therapeutic macromolecule is administered alone (as a result of an undesirable immune response to that therapeutic macromolecule).
  • compositions or kits provided herein can allow for a subject to obtain the treatment benefit of a therapeutic macromolecule without needing to increase the dose of the therapeutic macromolecule, which generally would be increased in order to compensate for undesired immune responses against the therapeutic macromolecule when administered without the benefit of the invention provided herein.
  • the methods, compositions or kits provided herein even allow for a subject to be administered reduced doses of the therapeutic macromolecule to achieve the same therapeutic benefit.
  • the invention is useful for achieving enhanced pharmacodynamic effects, or for using a reduced pharmacodynamically effective doses, in subjects in which undesired immune responses against the therapeutic macromolecule are generated or are expected to be generated.
  • the subject can be one at risk for such an undesired immune response.
  • administering means providing a material to a subject in a manner that is pharmacologically useful.
  • the term is intended to include causing to be administered in some embodiments.
  • “Causing to be administered” means causing, urging, encouraging, aiding, inducing or directing, directly or indirectly, another party to administer the material.
  • “Amount effective” in the context of a composition or dose for administration to a subject refers to an amount of the composition or dose that produces one or more desired responses in the subject, for example, the generation of a tolerogenic immune response (e.g., a reduction in the proliferation, activation, induction, survival, recruitment of therapeutic macromolecule-specific B cells or a reduction in the production of therapeutic macromolecule-specific antibodies).
  • the amount effective is a pharmacodynamically effective amount. Therefore, in some embodiments, an amount effective is any amount of a composition or dose provided herein (or multiple compositions or doses as provided herein) that produces one or more of the desired pharmacodynamic effects, therapeutic effects and/or immune responses as provided herein. This amount can be for in vitro or in vivo purposes. For in vivo purposes, the amount can be one that a clinician would believe may have a clinical benefit for a subject in need of therapeutic macromolecule administration and/or antigen-specific immune tolerance thereto.
  • 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 that is effective can also be an amount that produces a desired therapeutic endpoint or a desired therapeutic result. In other embodiments, the amounts effective can involve enhancing the level of a desired response, such as a therapeutic endpoint or result. Amounts effective, preferably, result in a tolerogenic immune response in a subject to an antigen, such as a therapeutic macromolecule. The achievement of any of the foregoing can be monitored by routine methods.
  • the amount effective is one in which the desired response persists in the subject for at least 1 week, at least 2 weeks, at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, or longer. In other embodiments of any of the compositions and methods provided, the amount effective is one which produces a measurable desired response for at least 1 week, at least 2 weeks, at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, or longer.
  • Amounts effective will depend, of course, on the particular subject being treated; the severity of a condition, disease or disorder; the individual patient parameters including age, physical condition, size and weight; the duration of the treatment; the nature of concurrent therapy (if any); the specific route of administration and like factors within the knowledge and expertise of the health practitioner. These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is generally preferred that a maximum dose be used, that is, the highest safe dose according to sound medical judgment. It will be understood by those of ordinary skill in the art, however, that a patient may insist upon a lower dose or tolerable dose for medical reasons, psychological reasons or for virtually any other reason.
  • doses of the immunosuppressants and/or therapeutic macromolecules in the compositions of the invention refer to the amount of the immunosuppressants and/or therapeutic macromolecules.
  • the dose can be administered based on the number of synthetic nanocarriers that provide the desired amount of immunosuppressants and/or therapeutic macromolecules.
  • Anti-therapeutic macromolecule antibody response or “anti-therapeutic macromolecule-specific antibody response” is the generation of anti-therapeutic macromolecule-specific antibodies, or induction of the process for producing such antibodies, as a result of administration of a therapeutic macromolecule. In embodiments, such a response counteracts the therapeutic effects of the therapeutic macromolecule.
  • 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 or are contained or expressed in cells.
  • the antigens may be contained within a cell or tissue preparation, cell debris, cell exosomes, conditioned media, etc.
  • Antigen-specific refers to any immune response that results from the presence of the antigen, or portion thereof, or that generates molecules that specifically recognize or bind the antigen.
  • antigen-specific may mean therapeutic macromolecule-specific.
  • the immune response is antigen-specific antibody production, such as therapeutic macromolecule-specific antibody production, antibodies are produced that specifically bind the antigen (e.g., therapeutic macromolecule).
  • the immune response is antigen-specific B cell or CD4+ T cell proliferation and/or activity, the proliferation and/or activity results from recognition of the antigen, or portion thereof, alone or in complex with MHC molecules, B cells, etc.
  • “Assessing a pharmacodynamic effect” refers to any measurement or determination of the level, presence or absence, reduction, increase in, etc. of a pharmacodynamic effect 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 in any one of the methods provided herein or otherwise known in the art.
  • an “at risk” subject is one in which a health practitioner believes has a chance of having a disease, disorder or condition or is one a health practitioner believes has a chance of experiencing an undesired anti-therapeutic macromolecule antibody response as provided herein and would benefit from the compositions and methods provided.
  • the subject is one that is at risk of having an anti-therapeutic macromolecule antibody response to a therapeutic macromolecule.
  • the subject is one that is expected to have an anti-therapeutic macromolecule antibody response to a therapeutic macromolecule.
  • 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.
  • therapeutic macromolecules and immunosuppressants are not attached to one another, meaning that the therapeutic macromolecules and immunosuppressants are not subjected to a process specifically intended to chemically associate one with another.
  • therapeutic macromolecules and/or immunosuppressants are not attached to synthetic nanocarriers, meaning that the therapeutic macromolecules (and/or immunosuppressants) and synthetic nanocarriers are not subjected to a process specifically intended to chemically associate one with another.
  • Average refers to the arithmetic mean unless otherwise noted.
  • Component as applied to two or more materials and/or agents (also referred to herein as the components), is intended to define material in which the two or more materials/agents are associated. Components may be separately identified, e.g., first component, second component, third component, etc. The terms “combined” and “combining” in this context are to be interpreted accordingly.
  • association of the two or more materials/agents in a combination may be physical or non-physical.
  • physically associated combined materials/agents include:
  • non-physically associated combined materials/agents examples include:
  • references to “combination therapy”, “combinations” and the use of materials/agents “in combination” in this application may refer to materials/agents that are administered as part of the same overall treatment regimen.
  • the posology of each of the two or more materials/agents may differ: each may be administered at the same time or at different times. It will, therefore, be appreciated that the materials/agents of the combination may be administered sequentially (e.g., before or after) or simultaneously, either in the same pharmaceutical formulation (i.e., together), or in different pharmaceutical formulations (i.e., separately).
  • 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 a physiologic or immunologic 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 may be provided in the Examples.
  • Determining or “determine” means to ascertain a factual relationship. Determining may be accomplished in a number of ways, including but not limited to performing experiments, or making projections. For instance, a dose of an immunosuppressant or therapeutic macromolecule 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 determine a protocol as provided herein. In another embodiment, the dose may be determined by testing various doses in a subject, i.e. through direct experimentation based on experience and guiding data.
  • determining comprises “causing to be determined.” “Causing to be determined” means causing, urging, encouraging, aiding, inducing or directing or acting in coordination with an entity for the entity to ascertain a factual relationship; including directly or indirectly, or expressly or impliedly.
  • 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.
  • 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.
  • Geneating means causing an action, such as a physiologic or immunologic response (e.g., tolerogenic immune response) to occur, either directly oneself or indirectly.
  • a physiologic or immunologic response e.g., tolerogenic immune response
  • Identifying a subject is any action or set of actions that allows a clinician to recognize a subject as one who may benefit from the methods, compositions or kits provided herein.
  • the identified subject is one who is in need of a therapeutic benefit from a therapeutic macromolecule as provided herein and in which an anti-therapeutic macromolecule-specific antibody response has occurred or is suspected to occur.
  • the action or set of actions may be either directly oneself or indirectly.
  • the method further comprises identifying a subject in need of a method, composition or kit as provided herein.
  • Immunosuppressant means a compound that causes an APC to have an immunosuppressive effect or a T cell or B cell to be suppressed (e.g., tolerogenic effect).
  • An immunosuppressive effect generally refers to the production or expression of cytokines or other factors by the APC that reduces, inhibits or prevents an undesired immune response or that promotes a desired immune response, such as a regulatory immune response.
  • the APC acquires an immunosuppressive function (under the immunosuppressive effect) on immune cells that recognize an antigen presented by this APC, the immunosuppressive effect is said to be specific to the presented antigen.
  • the immunosuppressive effect is a result of the immunosuppressant being delivered to the APC, preferably in the presence of an antigen.
  • 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.
  • 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.
  • the immunosuppressant is not an apoptotic-signaling molecule.
  • the immunosuppressant is not a phospholipid.
  • Immunosuppressants include, but are not limited to, statins; mTOR inhibitors, such as rapamycin or a rapamycin analog; TGF- ⁇ signaling agents; TGF- ⁇ receptor agonists; histone deacetylase inhibitors, such as Trichostatin A; corticosteroids; inhibitors of mitochondrial function, such as rotenone; P38 inhibitors; NF- ⁇ 3 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 inhibitors; cytokine receptor activ
  • Immunosuppressants also include IDO, vitamin D3, cyclosporins, such as cyclosporine A, aryl hydrocarbon receptor inhibitors, resveratrol, azathiopurine (Aza), 6-mercaptopurine (6-MP), 6-thioguanine (6-TG), FK506, sanglifehrin A, salmeterol, mycophenolate mofetil (MMF), aspirin and other COX inhibitors, niflumic acid, estriol, methotrexate and triptolide.
  • the immunosuppressant may comprise any of the agents provided herein.
  • the immunosuppressant can be a compound that directly provides the immunosuppressive effect on APCs or it can be a compound that provides the immunosuppressive effect indirectly (i.e., after being processed in some way after administration). Immunosuppressants, therefore, include prodrug forms of any of the compounds provided herein.
  • the immunosuppressants provided herein are attached to synthetic nanocarriers.
  • 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 attached to the one or more polymers.
  • the immunosuppressant is again in addition and attached to the one or more lipids.
  • the immunosuppressant is an element present in addition to the material of the synthetic nanocarrier that results in an immunosuppressive effect.
  • immunosuppressants include, but are not limited, small molecule drugs, natural products, antibodies (e.g., antibodies against CD20, CD3, CD4), biologics-based drugs, carbohydrate-based drugs, nanoparticles, liposomes, RNAi, antisense nucleic acids, aptamers, methotrexate, NSAIDs; fingolimod; natalizumab; alemtuzumab; anti-CD3; tacrolimus (FK506); cytokines and growth factors, such as TGF- ⁇ and IL-10; etc.
  • Further immunosuppressants are known to those of skill in the art, and the invention is not limited in this respect.
  • the immunosuppressant is in a form, such as a nanocrystalline form, whereby the form of the immunosuppressant itself is a particle or particle-like. In embodiments, such forms mimic a virus or other foreign pathogen.
  • a form such as a nanocrystalline form
  • such forms mimic a virus or other foreign pathogen.
  • 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.
  • 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 nanocrytalline 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.
  • concomitant administration of a reduced pharmacodynamically effective dose of a therapeutic macromolecule with an immunosuppressant in nanocrystalline form results in a reduced anti-therapeutic macromolecule antibody response.
  • “Load”, when attached to a synthetic nanocarrier, is the amount of the immunosuppressant and/or therapeutic macromolecule attached to a synthetic nanocarrier based on the total dry recipe weight of materials in an entire synthetic nanocarrier (weight/weight). Generally, the load is calculated as an average across a population of synthetic nanocarriers. In one embodiment, the load 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 of the immunosuppressant and/or therapeutic macromolecule is between 0.1% and 20%. In a further embodiment, the load of the immunosuppressant and/or therapeutic macromolecule is between 0.1% and 10%.
  • the load of the immunosuppressant and/or therapeutic macromolecule is between 1% and 10%. In still a further embodiment, the load of the immunosuppressant is between 7% and 20%. In yet another embodiment, the load of the immunosuppressant and/or the therapeutic macromolecule 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%,
  • the load of the immunosuppressant and/or the therapeutic macromolecule 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 of the immunosuppressant and/or the therapeutic macromolecule is no more than 25% on average across a population of synthetic nanocarriers. In embodiments, 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 may be obtained, in some embodiments, by suspending the synthetic nanocarriers in a liquid (usually aqueous) media and using dynamic light scattering (DLS) (e.g., using a Brookhaven ZetaPALS instrument).
  • 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.
  • 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.
  • “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.
  • “Pharmacodynamic effect” of “pharmacodynamic response” means any physiologic or immunologic response as a result of the administration of a therapeutic macromolecule. Such a response can be a desired response such as one that is associated with a therapeutic effect.
  • the methods, compositions or kits provided herein have been found to result in enhanced pharmacodynamic effects such as enhanced therapeutic effects, when administered in the presence of an anti-therapeutic macromolecule antibody response.
  • the enhanced pharmacodynamics effects can be obtained with a dose of the therapeutic macromolecule that is the same as or less than a dose of the therapeutic macromolecule when administered without the concomitant administration of an immunosuppressant dose as provided herein in the presence of an anti-therapeutic macromolecule antibody response.
  • a material/agent is pharmacodynamically effective can be evaluated by standard methods.
  • a pharmacodynamic effect using a method, composition or kit provided, in the presence of an anti-therapeutic macromolecule antibody response is compared to the pharmacodynamic effect when a therapeutic macromolecule is not so administered but also in the presence of an anti-therapeutic macromolecule antibody response.
  • the comparison is to the pharmacodynamic effect when the therapeutic macromolecule is administered alone in the presence of an anti-therapeutic macromolecule antibody response.
  • a pharmacodynamic effect is assessed with administration in the presence of an anti-therapeutic macromolecule antibody response, as it is desired that a method, composition or kit is effective to overcome such a response. Accordingly, pharmacodynamic effects are determined when such a response is occurring.
  • 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, dosing frequency, routes of administration, dosing duration, dosing rates, interval between dosing, combinations of any of the foregoing, and the like. In some embodiments, such 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 a pharmacodynamic effect.
  • any other therapeutic and/or immunologic effect may also be assessed instead of or in addition to the aforementioned immune responses.
  • 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.
  • 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 number of compositions as provided herein are administered to another subject using one or more or all or substantially all of the elements of which the protocol is comprised. In some embodiments, the protocol has been demonstrated to result in a reduction of an antibody response against a therapeutic macromolecule and/or improved pharmacodynamic effect.
  • Providing means an action or set of actions that an individual performs that supply a needed item or set of items or methods for practicing of the present invention.
  • the action or set of actions may be taken either directly oneself or indirectly.
  • Providing a subject is any action or set of actions that causes a clinician to come in contact with a subject and administer a composition provided herein thereto or to perform a method provided herein thereupon.
  • the subject is one who is in need of therapeutic macromolecule administration and antigen-specific immune tolerance thereto.
  • the action or set of actions may be taken either directly oneself or indirectly.
  • the method further comprises providing a subject.
  • “Recording an enhanced pharmacodynamic effect” means noting, or causing directly or indirectly activities in the expectation that such noting would take place, in any written or electronic form, that a therapeutic macromolecule dose achieved an enhanced pharmacodynamic effect in the presence of a real, expected or suspected anti-therapeutic macromolecule antibody response. Generally, in such circumstances the therapeutic macromolecule dose would not have been expected to achieve the enhanced pharmacodynamic effect in the presence of an anti-therapeutic macromolecule antibody response if administered without an immunosuppressant dose as provided herein (e.g., administered alone) based on information available at the time of concomitant administration as provided herein.
  • the recording occurs when an immunosuppressant dose in combination with a therapeutic macromolecule dose are administered to a subject or at some point thereafter.
  • the dose of the therapeutic macromolecule is reduced as compared to (or no greater than) a dose of therapeutic macromolecule that is administered without the immunosuppressant dose in the presence of an anti-therapeutic macromolecule antibody response.
  • “Written form”, as used herein, refers to any recordation on a medium such as paper.
  • Electrical form as used herein, refers to any recordation on electronic media. Any one of the methods provided herein can further comprise a step of recording a therapeutic and/or immune response in a subject receiving a treatment according to a method provided herein.
  • “Reduced pharmacodynamically effective dose” refers to a reduced amount of a therapeutic macromolecule that can achieve a similar pharmacodynamic effect when administered concomitantly with an immunosuppressant dose as provided herein as compared to the amount of the therapeutic macromolecule when not administered with the immunosuppressant dose (e.g., when the therapeutic macromolecule is administered alone).
  • a similar pharmacodynamic effect is a level of effect that is within one log of another level measured in the same way.
  • a similar pharmacodynamic effect is no more than 5-fold different. Still more preferably, a similar pharmacodynamic effect is no more than 2-fold different.
  • Subject means animals, including warm blooded mammals such as humans and primates; avians; domestic household or farm animals such as cats, dogs, sheep, goats, cattle, horses and pigs; laboratory animals such as mice, rats and guinea pigs; fish; reptiles; zoo and wild animals; and the like.
  • “Synthetic nanocarrier(s)” means a discrete object that is not found in nature, and that possesses at least one dimension that is less than or equal to 5 microns in size.
  • Albumin nanoparticles are generally included as synthetic nanocarriers, however in certain embodiments the synthetic nanocarriers do not comprise albumin nanoparticles.
  • synthetic nanocarriers do not comprise chitosan.
  • synthetic nanocarriers are not lipid-based nanoparticles.
  • synthetic nanocarriers do not comprise a phospholipid.
  • a synthetic nanocarrier can be, but is not limited to, one or a plurality of lipid-based nanoparticles (also referred to herein as lipid nanoparticles, i.e., nanoparticles where the majority of the material that makes up their structure are lipids), polymeric nanoparticles, metallic nanoparticles, surfactant-based emulsions, dendrimers, buckyballs, nanowires, virus-like particles (i.e., particles that are primarily made up of viral structural proteins but that are not infectious or have low infectivity), peptide or protein-based particles (also referred to herein as protein particles, i.e., particles where the majority of the material that makes up their structure are peptides or proteins) (such as albumin nanoparticles) and/or nanoparticles that are developed using a combination of nanomaterials such as lipid-polymer nanoparticles.
  • lipid-based nanoparticles also referred to herein as lipid nanoparticles, i
  • Synthetic nanocarriers may be a variety of different shapes, including but not limited to spheroidal, cuboidal, pyramidal, oblong, cylindrical, toroidal, and the like. Synthetic nanocarriers according to the invention comprise one or more surfaces. Exemplary synthetic nanocarriers that can be adapted for use in the practice of the present invention comprise: (1) the biodegradable nanoparticles disclosed in U.S. Pat. No.
  • synthetic nanocarriers may possess an aspect ratio greater than 1:1, 1:1.2, 1:1.5, 1:2, 1:3, 1:5, 1:7, or greater than 1:10.
  • Synthetic nanocarriers according to the invention that have a minimum dimension of equal to or less than about 100 nm, preferably equal to or less than 100 nm, do not comprise a surface with hydroxyl groups that activate complement or alternatively comprise a surface that consists essentially of moieties that are not hydroxyl groups that activate complement.
  • synthetic nanocarriers according to the invention that have a minimum dimension of equal to or less than about 100 nm, preferably equal to or less than 100 nm, do not comprise a surface that substantially activates complement or alternatively comprise a surface that consists essentially of moieties that do not substantially activate complement.
  • synthetic nanocarriers according to the invention that have a minimum dimension of equal to or less than about 100 nm, preferably equal to or less than 100 nm, do not comprise a surface that activates complement or alternatively comprise a surface that consists essentially of moieties that do not activate complement.
  • synthetic nanocarriers exclude virus-like particles.
  • 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.
  • a “therapeutic macromolecule” refers to any protein, carbohydrate, lipid or nucleic acid that may be administered to a subject and have a therapeutic effect.
  • administration of the therapeutic macromolecule to a subject may result in an undesired immune response, including production of anti-therapeutic macromolecule-specific antibodies.
  • administration of a therapeutic macromolecule concomitantly with an immunosuppressant dose can enhance the therapeutic effectiveness of the therapeutic macromolecule, such as by reducing undesired immune responses thereto.
  • the therapeutic macromolecule may be a therapeutic polynucleotide or therapeutic protein.
  • “Therapeutic polynucleotide” means any polynucleotide or polynucleotide-based therapy that may be administered to a subject and have a therapeutic effect. Such therapies include gene silencing. Examples of such therapy are known in the art, and include, but are not limited to, naked RNA (including messenger RNA, modified messenger RNA, and forms of RNAi). Examples of other therapeutic polynucleotides are provided elsewhere herein. Therapeutic polynucleotides may be produced in, on or by cells and also may be obtained using cell free or from fully synthetic in vitro methods. Subjects, therefore, include any subject that is in need of treatment with any of the foregoing. Such subject include those that will receive any of the foregoing.
  • “Therapeutic protein” means any protein or protein-based therapy that may be administered to a subject and have a therapeutic effect. Such therapies include protein replacement and protein supplementation therapies. Such therapies also include the administration of exogenous or foreign proteins, antibody therapies, and cell or cell-based therapies.
  • Therapeutic proteins comprise, but are not limited to, enzymes, enzyme cofactors, hormones, blood clotting factors, cytokines, growth factors, monoclonal antibodies, antibody-drug conjugates, and polyclonal antibodies. Examples of other therapeutic proteins are provided elsewhere herein. Therapeutic proteins may be produced in, on or by cells and may be obtained from such cells or administered in the form of such cells.
  • the therapeutic protein is produced in, on or by mammalian cells, insect cells, yeast cells, bacteria cells, plant cells, transgenic animal cells, transgenic plant cells, etc.
  • the therapeutic protein may be recombinantly produced in such cells.
  • the therapeutic protein may be produced in, on or by a virally transformed cell.
  • Subjects therefore, include any subject that is in need of treatment with any of the foregoing. Such subject include those that will receive any of the foregoing.
  • “Therapeutic macromolecule APC presentable antigen” means an antigen that is associated with a therapeutic macromolecule (i.e., the therapeutic macromolecule or a fragment thereof that can generate an immune response against the therapeutic macromolecule (e.g., the production of anti-therapeutic macromolecule-specific antibodies)).
  • therapeutic macromolecule antigen-presenting cell (APC) presentable antigens can be presented for recognition by the immune system (e.g., cells of the immune system, such as presented by antigen presenting cells, including but not limited to dendritic cells, B cells or macrophages).
  • the therapeutic macromolecule APC presentable antigen can be presented for recognition by, for example, T cells.
  • Such antigens may be recognized by and trigger an immune response in a T cell via presentation of an epitope of the antigen bound to Class I or Class II major histocompatability complex molecule (MHC).
  • MHC major histocompatability complex molecule
  • Therapeutic macromolecule APC presentable antigens generally include proteins, polypeptides, peptides, polynucleotides, lipoproteins, or are contained or expressed in, on or by cells.
  • the therapeutic macromolecule antigens in some embodiments, are attached to synthetic nanocarriers and comprise MHC Class I-restricted epitopes and/or MHC Class II-restricted epitopes and/or B cell epitopes.
  • MHC Class I-restricted epitopes and/or MHC Class II-restricted epitopes and/or B cell epitopes Preferably, one or more tolerogenic immune responses specific to the therapeutic macromolecule result with the methods, compositions or kits provided herein.
  • Undesired immune response refers to any undesired immune response that results from exposure to an antigen, promotes or exacerbates a disease, disorder or condition provided herein (or a symptom thereof), or is symptomatic of a disease, disorder or condition provided herein. Such immune responses generally have a negative impact on a subject's health or is symptomatic of a negative impact on a subject's health. Undesired immune responses include antigen-specific antibody production, antigen-specific B cell proliferation and/or activity or antigen-specific CD4+ T cell proliferation and/or activity. Generally, these undesired immune responses can be specific to a therapeutic macromolecule and counteract the beneficial effects desired of administration with the therapeutic macromolecule. Thus, in some embodiments, the undesired immune response is an anti-therapeutic macromolecule antibody response.
  • compositions comprising immunosuppressants and therapeutic macromolecules and related methods or kits.
  • Such methods, compositions or kits are useful to enhance the pharmacodynamics effect of a therapeutic macromolecule in the presence of an anti-therapeutic macromolecule antibody response, such as through the reduction or inhibition of undesired immune responses specific to the therapeutic macromolecule that diminish the therapeutic benefit of the therapeutic macromolecule.
  • Such methods, compositions or kits are also useful to allow for the repeated dosing of a therapeutic macromolecule.
  • the methods, compositions or kits provided herein are useful for achieving or enhancing desired therapeutic effects of the therapeutic macromolecules. In some embodiments, such therapeutic effects can be achieved or enhanced at reduced pharmacodynamically effective doses.
  • the methods, compositions or kits provided can be used for any subject in need of the therapeutic benefit of a therapeutic macromolecule.
  • the methods, compositions or kits can help neutralize anti-therapeutic macromolecule-specific antibodies that interfere with the desired treatment effects of a therapeutic macromolecule.
  • the methods, compositions or kits provided herein can, therefore, result in an enhancement of a desired therapeutic effect of a therapeutic macromolecule that would otherwise be diminished when the therapeutic macromolecule is administered without the immunosuppressant dose (e.g., when the therapeutic macromolecule is administered alone).
  • the methods, compositions or kit provided herein allow for a subject to obtain the treatment benefit of a therapeutic macromolecule without needing to increase the dose of the therapeutic macromolecule, which generally would be increased in order to compensate for undesired immune responses against the therapeutic macromolecule when administered without the benefit of the invention provided herein.
  • the methods, compositions or kits provided herein even allow for a subject to be administered reduced doses of the therapeutic macromolecule to achieve the same or better therapeutic benefit in the presence of an anti-therapeutic macromolecule antibody response.
  • a variety of immunosuppressants may be used in the practice of the present invention, which preferably, are attached to synthetic nanocarriers.
  • a wide variety of synthetic nanocarriers can be used according to the invention.
  • 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.
  • 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, various elements of the synthetic nanocarriers can be attached to the polymer.
  • a coating layer e.g., liposome
  • the immunosuppressants and/or therapeutic macromolecules can be attached to the synthetic nanocarriers by any of a number of methods.
  • the attaching can be a result of bonding between the immunosuppressants and/or therapeutic macromolecules and the synthetic nanocarriers. This bonding can result in the immunosuppressants and/or therapeutic macromolecules being attached to the surface of the synthetic nanocarriers and/or contained (encapsulated) within the synthetic nanocarriers.
  • the immunosuppressants and/or therapeutic macromolecules 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 and/or and/or therapeutic macromolecules are attached to the polymer.
  • a coupling moiety can be any moiety through which an immunosuppressant and/or therapeutic macromolecule 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 and/or therapeutic macromolecule 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 and/or therapeutic macromolecule 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 or therapeutic macromolecule, can be covalently associated with one or more polymers of the polymeric matrix.
  • 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 (e.g., attached) 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).
  • compositions according to the invention can comprise elements in combination with pharmaceutically acceptable excipients, such as preservatives, buffers, saline, or phosphate buffered saline.
  • pharmaceutically acceptable excipients such as preservatives, buffers, saline, or phosphate buffered saline.
  • the compositions may be made using conventional pharmaceutical manufacturing and compounding techniques to arrive at useful dosage forms.
  • compositions, such as those comprising the synthetic nanocarriers are suspended in sterile saline solution for injection together with a preservative.
  • the component when preparing synthetic nanocarriers as carriers, methods for attaching components to the synthetic nanocarriers may be useful. If the component is a small molecule it may be of advantage to attach the component to a polymer prior to the assembly of the synthetic nanocarriers. In embodiments, it may also be an advantage to prepare the synthetic nanocarriers with surface groups that are used to 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 attaching can be with a covalent linker.
  • components 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 on the surface of the nanocarrier with a component containing an alkyne group or by the 1,3-dipolar cycloaddition reaction of alkynes on the surface of the nanocarrier with a component containing an azido group.
  • Such cycloaddition reactions are preferably performed in the presence of a Cu(I) catalyst along with a suitable Cu(I)-ligand and a reducing agent to reduce Cu(II) compound to catalytic active Cu(I) compound.
  • This Cu(I)-catalyzed azide-alkyne cycloaddition (CuAAC) can also be referred as the click reaction.
  • the covalent attaching may comprise a covalent linker that comprises an amide linker, a disulfide linker, a thioether linker, a hydrazone linker, a hydrazide linker, an imine or oxime linker, an urea or thiourea linker, an amidine linker, an amine linker, and a sulfonamide linker.
  • a covalent linker that comprises an amide linker, a disulfide linker, a thioether linker, a hydrazone linker, a hydrazide linker, an imine or oxime linker, an urea or thiourea linker, an amidine linker, an amine linker, and a sulfonamide linker.
  • An amide linker is formed via an amide bond between an amine on one component with the carboxylic acid group of a second component, such as the nanocarrier.
  • the amide bond in the linker can be made using any of the conventional amide bond forming reactions with suitably protected amino acids and activated carboxylic acid such N-hydroxysuccinimide-activated ester.
  • a disulfide linker is made via the formation of a disulfide (S—S) bond between two sulfur atoms of the form, for instance, of R1-S—S—R2.
  • a disulfide bond can be formed by thiol exchange of a component containing thiol/mercaptan group (—SH) with another activated thiol group on a polymer or nanocarrier or a nanocarrier containing thiol/mercaptan groups with a component containing activated thiol group.
  • a triazole linker specifically a 1,2,3-triazole of the form
  • R1 and R2 may be any chemical entities, is made by the 1,3-dipolar cycloaddition reaction of an azide attached to a first component, such as the nanocarrier, with a terminal alkyne attached to a second component, such as the immunosuppressant or therapeutic macromolecule.
  • the 1,3-dipolar cycloaddition reaction is performed with or without a catalyst, preferably with Cu(I)-catalyst, which links the two components through a 1,2,3-triazole function.
  • This chemistry is described in detail by Sharpless et al., Angew. Chem. Int. Ed. 41(14), 2596, (2002) and Meldal, et al, Chem. Rev., 2008, 108(8), 2952-3015 and is often referred to as a “click” reaction or CuAAC.
  • a polymer containing an azide or alkyne group, terminal to the polymer chain is prepared.
  • This polymer is then used to prepare a synthetic nanocarrier in such a manner that a plurality of the alkyne or azide groups are positioned on the surface of that nanocarrier.
  • the synthetic nanocarrier can be prepared by another route, and subsequently functionalized with alkyne or azide groups.
  • the component is prepared with the presence of either an alkyne (if the polymer contains an azide) or an azide (if the polymer contains an alkyne) group.
  • the component is then allowed to react with the nanocarrier via the 1,3-dipolar cycloaddition reaction with or without a catalyst which covalently attaches the component to the particle through the 1,4-disubstituted 1,2,3-triazole linker.
  • a thioether linker is made by the formation of a sulfur-carbon (thioether) bond in the form, for instance, of R1-S—R2.
  • Thioether can be made by either alkylation of a thiol/mercaptan (—SH) group on one component with an alkylating group such as halide or epoxide on a second component.
  • 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 to the nanocarrier via non-covalent conjugation methods.
  • a negative charged therapeutic macromolecule or immunosuppressant can be conjugated to a positive charged nanocarrier through electrostatic adsorption.
  • a component containing a metal ligand can also be conjugated to a nanocarrier containing a metal complex via a metal-ligand complex.
  • the component can be attached to a polymer, for example polylactic acid-block-polyethylene glycol, prior to the assembly of the synthetic nanocarrier or the synthetic nanocarrier can be formed with reactive or activatible 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 is capable of attaching 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.
  • 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.
  • 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; cytokine receptor inhibitors; cytokine receptor activators; peroxisome proliferator-activated receptor
  • 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, AC
  • 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 IC, protein phosphatase 2A inhibitor protein, protein phosphatase 2A1, protein phosphatase 2A2, and sodium orthovanadate.
  • the therapeutic macromolecules as described herein are also attached to synthetic nanocarriers. In other embodiments, the therapeutic macromolecules are not attached to any synthetic nanocarriers. In some embodiments of either of these situations, the therapeutic macromolecules may be delivered in the form of the therapeutic macromolecule itself, or fragments or derivatives thereof.
  • Therapeutic macromolecules can include therapeutic proteins or therapeutic polynucleotides.
  • Therapeutic proteins include, but are not limited to, infusible therapeutic proteins, enzymes, enzyme cofactors, hormones, blood clotting factors, cytokines and interferons, growth factors, monoclonal antibodies, and polyclonal antibodies (e.g., that are administered to a subject as a replacement therapy), and proteins associated with Pompe's disease (e.g., acid glucosidase alfa, rhGAA (e.g., Myozyme and Lumizyme (Genzyme)).
  • Therapeutic proteins also include proteins involved in the blood coagulation cascade.
  • Therapeutic proteins include, but are not limited to, Factor VIII, Factor VII, Factor IX, Factor V, von Willebrand Factor, von Heldebrant Factor, tissue plasminogen activator, insulin, growth hormone, erythropoietin alfa, VEGF, thrombopoietin, lysozyme, antithrombin and the like.
  • Therapeutic proteins also include adipokines, such as leptin and adiponectin. Other examples of therapeutic proteins are as described below and elsewhere herein.
  • Examples of therapeutic proteins used in enzyme replacement therapy of subjects having a lysosomal storage disorder include, but are not limited to, imiglucerase for the treatment of Gaucher's disease (e.g., CEREZYMETM), a-galactosidase A (a-gal A) for the treatment of Fabry disease (e.g., agalsidase beta, FABRYZYMETM), acid ⁇ -glucosidase (GAA) for the treatment of Pompe disease (e.g., acid glucosidase alfa, LUMIZYMETM, MYOZYMETM), arylsulfatase B for the treatment of Mucopolysaccharidoses (e.g., laronidase, ALDURAZYMETM, idursulfase, ELAPRASETM, arylsulfatase B, NAGLAZYMETM), pegloticase (KRYSTEXXA) and pegsiticase.
  • enzymes include oxidoreductases, transferases, hydrolases, lyases, isomerases, asparaginases, uricases, glycosidases, asparaginases, uricases, proteases, nucleases, collagenases, hyaluronidases, heparinases, heparanases, lysins, and ligases.
  • Therapeutic proteins may also include any enzyme, toxin, or other protein or peptide isolated or derived from a bacterial, fungal, or viral source.
  • 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, prekallikrein (Fletcher factor), high-molecular weight kininogen (HMWK) (Fitzgerald factor), fibronectin, fibrin, thrombin, antithrombin III, heparin cofactor II, protein C, protein S, protein Z, protein Z-related protease inhibitot (ZPI), plasminogen, alpha 2-antiplasmin, tissue plasminogen activator (tPA),
  • 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- ⁇ ),
  • monoclonal antibodies include Abagovomab, Abciximab, Adalimumab, Adecatumumab, Afelimomab, Afutuzumab, Alacizumab pegol, ALD, Alemtuzumab, Altumomab pentetate, Anatumomab mafenatox, Anrukinzumab, Anti-thymocyte globin, Apolizumab, Arcitumomab, Aselizumab, Atlizumab (tocilizumab), Atorolimumab, Bapineuzumab, Basiliximab, Bavituximab, Bectumomab, Belimumab, Benralizumab, Bertilimumab, Besilesomab, Bevacizumab, Biciromab, Bivatuzumab mertansine, Blinatumomab, Brentuximab
  • infusion therapy 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), pegsiticase, taliglucerase alfa (Protalix/Uplyso), agalsidase alfa (Shire/Replagal®), velaglucerase alfa (Shire), and Keyhole Limpet Hemocyanin (KL)
  • Additional therapeutic proteins include, for example, engineered proteins, such as Fc fusion proteins, bispecific antibodies, multi-specific antibodies, nanobodies, antigen-binding proteins, antibody fragments, and protein conjugates, such as antibody drug conjugates.
  • engineered proteins such as Fc fusion proteins, bispecific antibodies, multi-specific antibodies, nanobodies, antigen-binding proteins, antibody fragments, and protein conjugates, such as antibody drug conjugates.
  • Therapeutic polynucleotides include, but are not limited to nucleic acid aptamers such as Pegaptanib (Macugen, a pegylated anti-VEGF aptamer), antisense therapeutics such as antisense poly- or oligonucleotides (e.g., antiviral drug Fomivirsen, or Mipomersen, an antisense therapeutic that targets the messenger RNA for apolipoprotein B for reduction of cholesterol level); small interfering RNAs (siRNAs) (e.g., dicer substrate siRNA molecules (DsiRNAs) which are 25-30 base pair asymmetric double-stranded RNAs that mediate RNAi with extremely high potency); or modified messenger RNAs (mmRNAs) such as those disclosed in US Patent application 2013/0115272 to de Fougerolles et al. and in Published US Patent application 2012/0251618 to Schrum et al.
  • nucleic acid aptamers such as Pegaptani
  • a component such as a therapeutic macromolecule or immunosuppressant may be isolated.
  • Isolated refers to the element being separated from its native environment and present in sufficient quantities to permit its identification or use. This means, for example, the element may be (i) selectively produced by expression cloning or (ii) purified as by chromatography or electrophoresis. Isolated elements may be, but need not be, substantially pure. Because an isolated element may be admixed with a pharmaceutically acceptable excipient in a pharmaceutical preparation, the element may comprise only a small percentage by weight of the preparation. The element is nonetheless isolated in that it has been separated from the substances with which it may be associated in living systems, i.e., isolated from other lipids or proteins. Any of the elements provided herein may be isolated and included in the compositions or used in the methods in isolated form.
  • aspects of the invention relate to determining a protocol for the methods of concomitant administration as provided herein.
  • a protocol can be determined by varying the frequency, dosage amount and other aspects of administration of the therapeutic macromolecule and the immunosuppressant and subsequently assessing the pharmacodynamic effect based on such variation.
  • the varied administration occurs in the presence of an anti-therapeutic macromolecule antibody response.
  • a preferred protocol for practice of the invention induces a desired pharmacodynamic effect but induces little to no anti-therapeutic macromolecule antibody response.
  • a desired pharmacodynamic effect of a therapeutic macromolecule includes stimulating or inhibiting a specific response.
  • the pharmacodynamic effect involves, without limitation, production or degradation of a cytokine, chemokine, signaling molecule or other molecule; inducing proliferation or death of a particular cell type; maturation or localization of a particular cell type; interaction with an enzyme, structural protein, carrier protein, or receptor protein; modulation of activity of an enzyme, structural protein or receptor protein, etc.
  • the pharmacodynamic effect is reducing production of a cytokine, for example an inflammation-associated cytokine such as TNF, IL-1.
  • the pharmacodynamic effect is reducing the activity of a cytokine. In some embodiments, the pharmacodynamic effect is reducing the production of an undesired molecule. In some embodiments, the pharmacodynamic effect is increasing degradation of an undesired molecule, for example uric acid crystals. In some embodiments, the pharmacodynamic effect is an activity of an enzyme.
  • a pharmacodynamic effect of a therapeutic macromolecule can be evaluated by standard methods.
  • the pharmacodynamic effect is a reduction of inflammation.
  • the level of inflammation can be assessed by any of the following exemplary methods without limitation, scoring of inflammatory symptoms such as redness or swelling; scoring of arthritic symptoms such as mobility, pain, or joint destruction; scoring of anaphylaxis symptoms such as swelling, blood pressure, shortness of breath; detecting and/or quantifying cell infiltration by histology, immunohistochemistry, flow cytometry; measuring the concentration of a protein or inflammation-associated cytokines such as TNF, IL-1 by ELISA, assessing the expression of gene or inflammation-associated genes by transcriptional analysis; measuring activity of an inflammation-associated cytokine, etc.
  • the pharmacodynamic effect is reduction or degradation of an undesired molecule.
  • the pharmacodynamic effect can be assessed, without limitation, by quantifying the undesired molecule in a tissue or blood sample by methods such as an ELISA.
  • the pharmacodynamic effect can be assessed by quantifying a molecule that is produced by the degradation of an undesired molecule such as by an ELISA.
  • the pharmacodynamic effect is the activity of an enzyme that was not previously present or not adequately present. In such embodiments, the activity of an enzyme can be assessed by detecting the presence or the concentration of a product of the enzyme activity.
  • a reduced dose of the therapeutic macromolecule is administered to produce a pharmacodynamic effect.
  • a reduced dose of a therapeutic macromolecule for such a purpose comprises any dose of the therapeutic macromolecule that achieves a pharmacodynamic effect in the presence of an anti-therapeutic macromolecule antibody response, with the concomitant administration of an immunosuppressant dose, that is less than the dose needed to achieve a similar pharmacodynamic effect with the therapeutic macromolecule when not administered concomitantly with the immunosuppressant dose in the presence of an anti-therapeutic macromolecule antibody response.
  • a reduced dose can be determined by administering a therapeutic macromolecule, with the concomitant administration of an immunosuppressant dose, at a certain dose in the presence of an anti-therapeutic macromolecule antibody response and assessing the pharmacodynamic effect.
  • the pharmacodynamic effect can then be compared with the pharmacodynamic effect that results through administration of the therapeutic macromolecule without the concomitant administration of an immunosuppressant dose in the presence of an anti-therapeutic macromolecule antibody response.
  • a lower dose that achieves a similar pharmacodynamic effect as determined by such a comparison is a reduced dose.
  • 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. Sci., 30:545; and Trindade et al., 2001, Chem. Mat., 13:3843). Additional methods have been described in the literature (see, e.g., Doubrow, Ed., “Microcapsules and Nanoparticles in Medicine and Pharmacy,” CRC Press, Boca Raton, 1992; Mathiowitz et al., 1987, J. Control.
  • Various materials may be encapsulated into synthetic nanocarriers as desirable using a variety of methods including but not limited to C. Astete et al., “Synthesis and characterization of PLGA nanoparticles” J. Biomater. Sci. Polymer Edn, Vol. 17, No. 3, pp. 247-289 (2006); K. Avgoustakis “Pegylated Poly(Lactide) and Poly(Lactide-Co-Glycolide) Nanoparticles: Preparation, Properties and Possible Applications in Drug Delivery” Current Drug Delivery 1:321-333 (2004); C. Reis et al., “Nanoencapsulation I. Methods for preparation of drug-loaded polymeric nanoparticles” Nanomedicine 2:8-21 (2006); P.
  • synthetic nanocarriers are prepared by a nanoprecipitation process or spray drying. Conditions used in preparing synthetic nanocarriers may be altered to yield particles of a desired size or property (e.g., hydrophobicity, hydrophilicity, external morphology, “stickiness,” shape, etc.). The method of preparing the synthetic nanocarriers and the conditions (e.g., solvent, temperature, concentration, air flow rate, etc.) used may depend on the materials to be 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, such synthetic nanocarriers can be sized, for example, using a sieve.
  • Elements (i.e., components) 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 couplings 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.
  • the synthetic nanocarriers can be combined with a therapeutic macromolecule or other composition by admixing in the same vehicle or delivery system.
  • 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 a sterile saline solution for injection together with a preservative.
  • compositions of the invention can be made in any suitable manner, and the invention is in no way limited to compositions that can be produced using the methods described herein. Selection of an appropriate method 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.
  • the compositions may be lyophilized and stored in suspension or as lyophilized powder depending on the formulation strategy for extended periods without losing activity.
  • Administration according to the present invention may be by a variety of routes, including but not limited to subcutaneous, intravenous, intraperitoneal, intramuscular, transmucosal, transdermal, transcutaneous or intradermal routes.
  • administration is via a subcutaneous route of administration.
  • the compositions referred to herein may be manufactured and prepared for administration, preferably concomitant administration, using conventional methods.
  • compositions of the invention can be administered in effective amounts, such as the effective amounts described elsewhere herein.
  • Doses of dosage forms may contain varying amounts of immunosuppressants and/or therapeutic macromolecules, according to the invention.
  • the amount of immunosuppressants and/or therapeutic macromolecules present in the inventive dosage forms can be varied according to the nature of the therapeutic macromolecules, and/or immunosuppressant, the therapeutic benefit to be accomplished, and other such parameters.
  • dose ranging studies can be conducted to establish optimal therapeutic amounts of immunosuppressant and/or therapeutic macromolecules to be present in the dosage form.
  • the immunosuppressants and/or therapeutic macromolecules are present in the dosage form in an amount effective to generate a desired pharmacodynamic effect and/or a reduced immune response to the therapeutic macromolecules upon administration to a subject. It may be possible to determine amounts of the immunosuppressants and/or therapeutic macromolecules effective to achieve a desired result using conventional dose ranging studies and techniques in subjects.
  • Inventive dosage forms may be administered at a variety of frequencies.
  • at least one administration of the compositions provided herein is sufficient to generate a pharmacologically relevant response.
  • at least two administrations or at least three administrations are utilized to ensure a pharmacologically relevant response.
  • repeated administrations are utilized to ensure a pharmacologically relevant response.
  • kits comprising a pharmacodynamically effective dose or more than one dose of a therapeutic macromolecule, such as a reduced pharmacodynamically effective dose.
  • the kit may also comprise an immunosuppressant dose or more than one dose of immunosuppressant.
  • the immunosuppressant dose and the pharmacodynamically effective dose can be contained within separate containers or within the same container in the kit.
  • the container is a vial or an ampoule.
  • the pharmacodynamically effective dose of a therapeutic macromolecule and/or immunosuppressant dose are contained within a solution separate from the container, such that the pharmacodynamically effective dose of a therapeutic macromolecule and/or immunosuppressant dose may be added to the container at a subsequent time.
  • the pharmacodynamically effective dose of a therapeutic macromolecule and/or immunosuppressant dose are in lyophilized form each in a separate container or in the same container, such that they may be reconstituted at a subsequent time.
  • the kit further comprises instructions for reconstitution, mixing, administration, etc.
  • the instructions include a description of the methods described herein. Instructions can be in any suitable form, e.g., as a printed insert or a label.
  • the kit further comprises one or more syringes.
  • Example 1 Evaluating Immune Responses with Synthetic Nanocarriers Comprising Immunosuppressant and APC Presentable Antigen In Vivo
  • Ovalbumin peptide 323-339 (OVA 323-339 ), a 17 amino acid peptide known to be a T and B cell epitope of Ovalbumin protein, was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance Calif. 90505; Part #4065609). Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Catalogue # R1017).
  • PLGA with a lactide:glycolide ratio of 3:1 and an inherent viscosity of 0.75 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 7525 DLG 7A).
  • Polyvinyl alcohol (85-89% hydrolyzed) was purchased from EMD Chemicals (Product Number 1.41350.1001).
  • Solution 1 OVA 323-339 at 20 mg/mL in dilute hydrochloric acid aqueous solution. The solution was prepared by dissolving ovalbumin peptide in 0.13 M hydrochloric acid solution at room temperature.
  • Solution 2 PLGA at 100 mg/mL in methylene chloride. The solution was prepared by dissolving PLGA in pure methylene chloride.
  • Solution 3 PLA-PEG at 100 mg/mL in methylene chloride. The solution was prepared by dissolving PLA-PEG in pure methylene chloride.
  • Solution 4 Rapamycin at 50 mg/mL in methylene chloride. The solution was prepared by dissolving rapamycin in pure methylene chloride.
  • Solution 5 Polyvinyl alcohol at 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • a primary water-in-oil emulsion was prepared first.
  • W1/O1 was prepared by combining solution 1 (0.2 mL), solution 2 (0.75 mL), solution 3 (0.25 mL), and solution 4 (0.2 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) was then prepared by combining solution 5 (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 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 synthetic nanocarriers to form.
  • a portion of the synthetic nanocarriers were washed by transferring the synthetic 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 was repeated, and the pellet was re-suspended in phosphate buffered saline for a final synthetic nanocarrier dispersion of about 10 mg/mL.
  • the amounts of peptide and rapamycin in the synthetic nanocarriers were determined by HPLC analysis.
  • the total dry-synthetic nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • a primary water-in-oil emulsion was prepared first.
  • W1/O1 was prepared by combining 0.13 M hydrochloric acid solution (0.2 mL), solution 2 (0.75 mL), solution 3 (0.25 mL), and solution 4 (0.2 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) was then prepared by combining solution 5 (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 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 synthetic nanocarriers to form.
  • a portion of the synthetic nanocarriers were washed by transferring the synthetic nanocarrier suspension to a centrifuge tube and centrifuging at 21,000 ⁇ g and 4° C. for one hour, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. The washing procedure was repeated, and the pellet was re-suspended in phosphate buffered saline for a final synthetic nanocarrier dispersion of about 10 mg/mL.
  • the amount of rapamycin in the synthetic nanocarrier was determined by HPLC analysis.
  • the total dry-synthetic nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • Tween-20 (Sigma, Catalog # P9416-100 mL) was used as wash buffer, prepared by adding 10 ml of Tween-20 ((Sigma, Catalog # P9416-100 mL) to 2 liters of a O1 ⁇ PBS stock (PBS: OmniPur® 10 ⁇ PBS Liquid Concentrate, 4 L, EMD Chemicals, Catalog #6505) and 18 Liters of deionized water.
  • PBS OmniPur® 10 ⁇ PBS Liquid Concentrate, 4 L, EMD Chemicals, Catalog #6505
  • OVA protein at a stock concentration of 5 mg/ml was used as a coating material. A 1:1000 dilution to 5 ⁇ g/ml was used as a working concentration.
  • Each well of the assay plates was coated with 100 ⁇ l diluted OVA per well, plates were sealed with sealing film (VWR catalog #60941-120), and incubated overnight at 4° C.
  • Costar 9017 96-well Flat bottom plates were used as assay plates (Costar 9017).
  • Low-binding polypropylene 96-well plate or tubes were used as set-up plates, in which samples were prepared before being transferred to the assay plate.
  • the setup plates did not contain any antigen and, therefore, serum antibodies did not bind to the plate during the setup of the samples.
  • Setup plates were used for sample preparation to minimize binding that might occur during preparation or pipetting of samples if an antigen-coated plate was used to prepare the samples.
  • wells were covered with diluent to block any non-specific binding and the plate was sealed and incubated at 4° C. overnight.
  • Assay plates were washed three times with wash buffer, and wash buffer was completely aspirated out of the wells after the last wash. After washing, 300 ⁇ l diluent were added to each well of assay plate(s) to block non-specific binding and plates were incubated at least 2 hours at room temperature. Serum samples were prepared in the setup plate at appropriate starting dilutions. Starting dilutions were sometimes also prepared in 1.5 ml tubes using diluent and then transferred to the set-up plate. Appropriate starting dilutions were determined based on previous data, where available. Where no previous data was available, the lowest starting dilution was 1:40. Once diluted, 200 ⁇ l of the starting dilution of the serum sample was transferred from the tube to the appropriate well of the setup plate.
  • An exemplary setup plate layout is described as follows: Columns 2 and 3 contained anti-Ovalbumin monoclonal IgG2b isotype (AbCam, ab17291) standard, diluted to 0.25 ⁇ g/mL (1:4000 dilution). Columns 3-11 contained serum samples (at appropriate dilutions). Columns 1 and 12 were not used for samples or standards to avoid any bias of measurements due to edge effect. Instead, columns 1 and 12 contained 200 ⁇ l diluent. Normal mouse serum diluted 1:40 was used as a negative control. Anti-mouse IgG2a diluted 1:500 from 0.5 mg/mL stock (BD Bioscience) was used as an isotype control.
  • Anti-mouse IgG2a diluted 1:500 from 0.5 mg/mL stock (BD Bioscience) was used as an isotype control.
  • serial dilutions were pipetted on the assay plate as follows: 50 ⁇ l of each serum sample was removed from row A using 12-channel pipet and mixed with the 100 ⁇ l of diluent previously added to each well of row B. This step was repeated down the entire plate. After pipetting the dilution of the final row, 50 ⁇ l of fluid was removed from the wells in the final row and discarded, resulting in a final volume of 100 ⁇ l in every well of the assay plate. Once sample dilutions were prepared in the assay plates, the plates were incubated at room temperature for at least 2 hours.
  • Detection antibody Goat anti-mouse anti-IgG, HRP conjugated, AbCam ab987157 was diluted 1:1500 (0.33 ⁇ g/mL) in diluent and 100 ⁇ l of the diluted antibody was added to each well. Plates were incubated for 1 hour at room temperature and then washed three times with wash buffer, with each washing step including a soak time of at least 30 seconds.
  • detection substrate was added to the wells. Equal parts of substrate A and substrate B (BD Biosciences TMB Substrate Reagent Set, catalog #555214) were combined immediately before addition to the assay plates, and 100 ⁇ l of the mixed substrate solution were added to each well and incubated for 10 minutes in the dark. The reaction was stopped by adding 50 ⁇ l of stop solution (2N H2SO4) to each well after the 10 minute period. The optical density (OD) of the wells was assessed immediately after adding the stop solution on a plate reader at 450 nm with subtraction at 570 nm. Data analysis was performed using Molecular Device's software SoftMax Pro v5.4.
  • a four-parameter logistic curve-fit graph was prepared with the dilution on the x-axis (log scale) and the OD value on the y-axis (linear scale), and the half maximum value (EC50) for each sample was determined.
  • the plate template at the top of the layout was adjusted to reflect the dilution of each sample (1 per column).
  • FIG. 1 shows a decrease in circulating antigen-specific antibody production with nanocarriers comprising peptide antigen and immunosuppressant when administered intravenously. Two independent experiments were performed using 5 animals in each experiment.
  • 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.
  • mice C57BL/6 age-matched (5-6 weeks) female mice (5 per group, 3 groups: na ⁇ ve control, untreated control, and treatment with synthetic nanocarriers with rapamycin were injected intravenously with 25 ⁇ g of chicken ovalbumin induced using high sheer to form aggregates in order to increase its immunogenicity (aggOVA).
  • aggOVA immunogenicity
  • subsequent boosts of 25 ⁇ g ovalbumin were performed intraperitoneally (i.p. on days 42 and 57), and subcutaneously a 12.5 ⁇ g pOVA challenge in the left hind limb (s.c. d62) followed by challenges using the same aggOVA combined with CpG (s.c.
  • FIG. 3 shows the results of such immunization protocol.
  • a prominent anti-OVA antibody (Ab) response can be detected in these animals using ELISA (technique generally as described in Example 1).
  • ELISA technique generally as described in Example 1.
  • mice were treated using the synthetic nanocarriers administered i.v. dose calculated to provide 100 ⁇ g of rapamycin) concomitantly with aggOVA during only the three first encounters with the antigen on d0, 14 and 28, no IgG response can be detected for the next 60 days even after 4 injections with the immunogenic mixture including aggOVA. See FIG. 3 .
  • mice as above were also injected with 0.05 ⁇ g of a second Ag, Keyhole Limpet Hemocyanin (KLH), with the KLH being mixed with the aggOVA, and administered according to the schedule noted above for the aggOVA.
  • KLH Keyhole Limpet Hemocyanin
  • Anti-KLH IgG antibody titer was determined using a method similar to the anti-OVA IgG titer method described in Example 1.
  • results demonstrate that an enhanced effect can be obtained with the compositions and methods provided herein when administered under conditions that would ordinarily produce a significant antibody response. Also, the results support that a protocol was established that was demonstrated to result in a pharmacodynamic effect.
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Code R1017).
  • PLGA with a lactide:glycolide ratio of 3:1 and an inherent viscosity of 0.75 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 7525 DLG 7A).
  • EMPROVE® Polyvinyl Alcohol 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).
  • Solution 1 PLGA at 75 mg/mL, PLA-PEG-OMe at 25 mg/mL, and rapamycin at 12.5 mg/mL in methylene chloride.
  • the solution was prepared by dissolving PLGA, PLA-PEG-OMe, and rapamycin in pure methylene chloride.
  • Solution 2 Polyvinyl alcohol @ 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • O/W emulsion An oil-in-water (O/W) emulsion was used to prepare the nanocarriers.
  • the O/W emulsion was prepared by combining Solution 1 (1.0 mL) and Solution 2 (3.0 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 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 75,600 ⁇ g and 4° C.
  • Nanocarrier size was determined by dynamic light scattering and presented in Table 1.
  • the amount of rapamycin in the nanocarrier was determined by HPLC analysis (Table 1).
  • the total dry-nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Code R1017).
  • PLGA with a lactide:glycolide ratio of 1:1 and an inherent viscosity of 0.24 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 5050 DLG 2.5A).
  • EMPROVE® Polyvinyl Alcohol 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).
  • Solution 1 PLGA at 75 mg/mL, PLA-PEG-OMe at 25 mg/mL, and rapamycin at 12.5 mg/mL in methylene chloride.
  • the solution was prepared by dissolving PLGA, PLA-PEG-OMe, and rapamycin in pure methylene chloride.
  • Solution 2 Polyvinyl alcohol at 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • Solution 3 70 mM phosphate buffer, pH 8.
  • a an oil-in-water emulsion was created by mixing Solutions 1 (1.0 mL) and Solution 2 (3.0 mL) in a small glass pressure tube and sonicating at 30% amplitude for 60 seconds using a Branson Digital Sonifier 250.
  • the emulsion was added to an open 50 mL beaker containing Solution 3 (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension.
  • a portion of the suspended nanocarriers was then washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 75,600 rcf for 40 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated and then the pellet was re-suspended in PBS 1 ⁇ to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis.
  • the suspension was stored frozen at ⁇ 20
  • 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.
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Code R1017).
  • PLGA with a lactide:glycolide ratio of 1:1 and an inherent viscosity of 0.24 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 5050 DLG 2.5A).
  • F8II.1723 peptide was purchased from AnaSpec (34801 Campus Drive, Fremont, Calif. 94555).
  • EMPROVE® Polyvinyl Alcohol 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).
  • Solution 1 PLGA at 75 mg/mL, PLA-PEG-OMe at 25 mg/mL, and rapamycin at 12.5 mg/mL in methylene chloride.
  • the solution was prepared by dissolving PLGA, PLA-PEG-OMe, and rapamycin in pure methylene chloride.
  • Solution 2 F8II.1723 (ERLWDYGMSSSPHVL) at 10 mg/mL and sucrose at 100 mg/mL in 50 mM pH 11.5 phosphate buffer.
  • the solution was prepared by dissolving the sucrose in 50 mM pH 11.5 phosphate buffer and then adding the F8II.1723 peptide as a dry powder.
  • Solution 3 Polyvinyl alcohol at 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • Solution 4 70 mM pH 8 phosphate buffer.
  • a primary (W1/O) emulsion was first created by mixing Solution 1 (1.0 mL) and Solution 2 (0.2 mL) in a small glass pressure tube and sonicating at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/O/W2) emulsion was then formed by adding Solution 3 (3.0 mL) to the primary emulsion, vortexing to create a crude dispersion, and then sonicating at 30% amplitude for 60 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to an open 50 mL beaker containing Solution 4 (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended nanocarriers was then washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 75,600 rcf for 35 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated and then the pellet was re-suspended in PBS 1 ⁇ to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at ⁇ 20° C. until use.
  • Nanocarrier size was determined by dynamic light scattering.
  • the amount rapamycin in the nanocarrier was determined by HPLC analysis.
  • the amount of F8II.1723 in the nanocarrier was determined using a fluorescamine-based assay.
  • the total dry-nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Code R1017).
  • PLGA with a lactide:glycolide ratio of 1:1 and an inherent viscosity of 0.24 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 5050 DLG 2.5A).
  • F8II.75 peptide was purchased from AnaSpec (34801 Campus Drive, Fremont, Calif. 94555).
  • EMPROVE® Polyvinyl Alcohol 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).
  • Solution 1 PLGA at 75 mg/mL, PLA-PEG-OMe at 25 mg/mL, and rapamycin at 12.5 mg/mL in methylene chloride.
  • the solution was prepared by dissolving PLGA, PLA-PEG-OMe, and rapamycin in pure methylene chloride.
  • Solution 2 F8II.75 (VHLFNIAKPRPPWMG) at 10 mg/mL and sucrose at 100 mg/mL in 50 mM pH 2 phosphate buffer.
  • the solution was prepared by dissolving the sucrose in 50 mM pH 2 phosphate buffer and then adding the F8II.75 peptide as a dry powder.
  • Solution 3 Polyvinyl alcohol at 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • Solution 4 70 mM pH 8 phosphate buffer.
  • a primary (W1/O) emulsion was first created by mixing Solution 1 (1.0 mL) and Solution 2 (0.2 mL) in a small glass pressure tube and sonicating at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/O/W2) emulsion was then formed by adding Solution 3 (3.0 mL) to the primary emulsion, vortexing to create a crude dispersion, and then sonicating at 30% amplitude for 60 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to an open 50 mL beaker containing Solution 4 (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended nanocarriers was then washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 75,600 rcf for 35 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated and then the pellet was re-suspended in PBS 1 ⁇ to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at ⁇ 20° C. until use.
  • Nanocarrier size was determined by dynamic light scattering.
  • the amount rapamycin in the nanocarrier was determined by HPLC analysis.
  • the amount of F8II.75 in the nanocarrier was determined using a fluorescamine-based assay.
  • the total dry-nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Code R1017).
  • PLGA with a lactide:glycolide ratio of 1:1 and an inherent viscosity of 0.24 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 5050 DLG 2.5A).
  • F8II.2210 peptide was purchased from AnaSpec (34801 Campus Drive, Fremont, Calif. 94555).
  • EMPROVE® Polyvinyl Alcohol 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).
  • Solution 1 PLGA at 75 mg/mL, PLA-PEG-OMe at 25 mg/mL, and rapamycin at 12.5 mg/mL in methylene chloride.
  • the solution was prepared by dissolving PLGA, PLA-PEG-OMe, and rapamycin in pure methylene chloride.
  • Solution 2 F8II.2210 (TASSYFTNMFATWSPSKAR) at 10 mg/mL and sucrose at 100 mg/mL in 50 mM pH 2 phosphate buffer.
  • the solution was prepared by dissolving the sucrose in 50 mM pH 2 phosphate buffer and then adding the F8II.2210 peptide as a dry powder.
  • Solution 3 Polyvinyl alcohol at 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • Solution 4 70 mM pH 8 phosphate buffer.
  • a primary (W1/O) emulsion was first created by mixing Solution 1 (1.0 mL) and Solution 2 (0.2 mL) in a small glass pressure tube and sonicating at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • a secondary (W1/O/W2) emulsion was then formed by adding Solution 3 (3.0 mL) to the primary emulsion, vortexing to create a crude dispersion, and then sonicating at 30% amplitude for 60 seconds using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to an open 50 mL beaker containing Solution 4 (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended nanocarriers was then washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 75,600 rcf for 35 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated and then the pellet was re-suspended in PBS 1 ⁇ to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at ⁇ 20° C. until use.
  • Nanocarrier size was determined by dynamic light scattering.
  • the amount rapamycin in the nanocarrier was determined by HPLC analysis.
  • the amount of F8II.2210 in the nanocarrier was determined using a fluorescamine-based assay.
  • the total dry-nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • PLA-PEG-OMe block co-polymer with a methyl ether terminated PEG block of approximately 5,000 Da and an overall inherent viscosity of 0.5 DL/g was purchased from Lakeshore Biochemicals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 100 DL mPEG 5000 5CE).
  • EMPROVE® Polyvinyl Alcohol 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).
  • Solution 1 PLGA at 75 mg/mL and PLA-PEG-OMe at 25 mg/mL in methylene chloride. The solution was prepared by dissolving PLGA and PLA-PEG-OMe in pure methylene chloride.
  • Solution 2 Polyvinyl alcohol at 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • Solution 3 70 mM phosphate buffer, pH 8.
  • a an oil-in-water emulsion was created by mixing Solutions 1 (1.0 mL) and Solution 2 (3.0 mL) in a small glass pressure tube and sonicating at 30% amplitude for 60 seconds using a Branson Digital Sonifier 250.
  • the emulsion was added to an open 50 mL beaker containing Solution 3 (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension.
  • a portion of the suspended nanocarriers was then washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 75,600 rcf for 40 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated and then the pellet was re-suspended in PBS 1 ⁇ to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis.
  • the suspension was stored frozen at ⁇ 20
  • Nanocarrier size was determined by dynamic light scattering. The total dry-nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • i.v. intravenous injections of the blood clotting protein, Factor VIII (FVIII), and the synthetic nanocarriers attached to rapamycin (Nanocarrier ID 4), or empty nanoparticles (NP) (Nanocarrier ID 8) and FVIII or IVIG (intravenous immunoglobulin) and FVIII for 5 consecutive weeks.
  • FVIII Factor VIII
  • IVIG intravenous immunoglobulin
  • the inhibitor titer was determined by Bethesda assay using a chromogenic FVIII activity assay kit (Coatest SP4 FVIII), as presented in FIG. 5 .
  • mice were challenged with FVIII (i.v. or i.p.) in the absence of further treatment. Anti-FVIII antibody levels were determined by ELISA. Results at selected time points are presented in FIG. 6B . Data at each time point are expressed as mean ⁇ SEM.
  • the nanocarrier groups (with protein or peptides) were compared with the empty nanoparticle control group using the student t-test with two tailed distribution. *p ⁇ 0.05 and **p ⁇ 0.01, between Nanocarrier ID 4 with protein and empty NP groups; # p ⁇ 0.05 and ## p ⁇ 0.01, between the nanocarrier and peptide (Nanocarrier IDs 5, 6 and 7) and empty NP groups. All injections of FVIII were 1 ⁇ g per injection. All injections of Nanocarrier ID 4 were 100 ⁇ g per injection.
  • Control C57BL/6 age-matched (5-6 weeks) females were injected subcutaneously (s.c.) in the front limbs with 60 ⁇ g of HUMIRA once a week until day 29 (on d0, 7, 14, 22, 29, across all groups and conditions).
  • Another group received similar injections, but 0.9 mg of nanocarrier attached to rapamycin (Nanocarrier ID 1, 2 or 3) were admixed to the solution of HUMIRA on the priming day 0.
  • the results presented in FIG. 7A show the antibody titers in the blood of all animals at day 21. The control animals develop a robust antibody response against HUMIRA while treated animals remain completely negative even after 20 days and two injections of HUMIRA without treatment.
  • the animals received another challenge in one hind limb while the other hind limb received saline in order to test the local antibody-mediated type I hypersensitivity response.
  • the thickness of the hind limbs were measured with the help of a caliper one hour after the injection. The difference in thickness between the two limbs is presented in FIG. 7B .
  • the treatment with the nanocarriers abolished the inflammatory response induced by local administration of HUMIRA.
  • Control C57BL/6 age-matched (5-6 weeks) females were injected intravenously (i.v.) in the tail vain with 200 ⁇ g of Keyhole Limpet Hemocyanin (KLH) starting at day 0 or day 21 once a week until 34 (on d0, 7, 14, 21, 28, 34).
  • KLH Keyhole Limpet Hemocyanin
  • Another group received similar injections from day 0 to day 34, but 0.47 mg of nanocarrier attached to rapamycin (Nanocarrier ID 1, 2 or 3) were admixed to the solution of KLH on days 0, 14 and 21 followed by KLH alone injections weekly between days 21 and 34 (on d21, 28, 34).
  • the results presented in FIG. 8 show the antibody titers in the blood of all animals at the indicated time points.
  • animals that received three injections of the nanocarriers remain completely negative after three injections of KLH alone.
  • a control group of C57BL/6 age-matched (5-6 weeks) female were injected i.v. in the tail vain with PBS while the treated group was injected with 0.9 mg of nanocarriers attached to rapamycin (Nanocarrier ID 1, 2 or 3) in combination with 40 ⁇ g of KRYSTEXXA on days ⁇ 21 and ⁇ 14. All animals received weekly injections from day 0 to day 28 of 100 ⁇ g of KRYSTEXXA combined to 20 ⁇ g of CpG s.c. in the hind limb (d0, 7, 12, 28). The control animals develop an immune response against KRYSTEXXA that can be measured by the anti-KRYSTEXXA IgM antibody titers. The results in FIG.
  • the control group of C57BL/6 age-matched (5-6 weeks) female were injected i.v. in the tail vein with 2.5 ⁇ g of an immunogenic form of particulate Ovalbumin (pOVA) and 2 ⁇ g of Keyhole Limpet Hemocyanin (KLH) s.c. in the hind limb once a week for 49 days (d0, 7, 14, 20, 28, 35, 42, 49).
  • the other group of animals received pOVA and KLH in the same routes and amounts but admixed to 0.2 mg of nanocarrier attached to rapamycin (Nanocarrier ID 1, 2 or 3) on days 0, 7 and 14 followed by pOVA injections between days 20 and 42 (same amounts as before).
  • the control animals develop a robust immune response against OVA and KLH that can be measured by the anti-OVA or anti-KLH IgG antibody titers.
  • the antibody titers at day 54 shown in FIG. 11 demonstrate that 3 doses of synthetic nanocarriers administered concomitantly with pOVA in the same solution were effective in reducing and preventing antibody formation to OVA for a prolonged period of time but not the KLH (that was injected in another location s.c.). The treated animals did not develop an anti-OVA response even after five injections of pOVA alone.
  • Control C57BL/6 age-matched (5-6 weeks) female were injected i.v. in the tail vain with 200 ⁇ g of Keyhole Limpet Hemocyanin (KLH) once a week for 63 days (d0, 7, 14, 21, 28, 35, 42, 49, 56, 63).
  • KLH Keyhole Limpet Hemocyanin
  • the other group received similar injections but 0.9 mg of nanocarriers attached to rapamycin (Nanocarrier ID 1, 2 or 3) were admixed to the solution of KLH at days 0, 7, 14 and 21 followed by KLH six injections (same amount) between days 28 and 63.
  • the control animals developed a robust response to KLH that can be measured by the anti-KLH IgG antibody titers as well as the anaphylactic reaction induced by the injections.
  • the results in FIG. 12A show the antibody titers in the blood of all animals at the indicated time points, and the anaphylaxis scores induced by the injection of the antigen are presented in FIG. 12B .
  • Four doses of synthetic nanocarriers administered concomitantly with KLH were effective in reducing and preventing antibody formation and anaphylaxis for a prolonged period of time. Indeed, treated animals did not develop an anti-KLH response even after four injections of KLH alone which in control animals was largely sufficient to create a response (day 26).
  • compositions provided herein when administered over a period of time concomitantly with a protein can reduce or prevent antibody formation and anaphylaxis for prolonged periods of time.
  • Transgenic animals overexpressing human tumor necrosis factor alpha develop progressive rheumatoid arthritis over the course of 20 weeks from birth. This process can be prevented by using the fully human anti-human TNF ⁇ antibody HUMIRA/adalimumab.
  • HUMIRA human anti-human TNF ⁇ antibody
  • ADA anti-drug antibody formation
  • Only very high doses can then maintain the therapeutic benefit and surmount the neutralizing immune response. For example, it is thought that about 200 ⁇ g is needed to effect maximal inhibition of inflammation in an example of such a mouse model (Binder et al., Arthritis & Rheumatism, Vol. 65 (No. 3), March 2013, pp. 608-617).
  • Age-matched huTNFaTg 5 week old female animals were either injected s.c. in the subscapular area with saline (PBS) or 60 ⁇ g of HUMIRA weekly or with a mixture of HUMIRA and 0.87 mg of nanocarriers attached to rapamycin (Nanocarrier ID 1, 2 or 3) for the first 7 injections (day 0, 7, 14, 21, 28, 35, 42, weeks 5 to 12 of age) followed by 3 injections of HUMIRA alone (same amount) (day 49 to 63, weeks 13 to 15 of age).
  • PBS subscapular area with saline
  • HUMIRA weekly a mixture of HUMIRA and 0.87 mg of nanocarriers attached to rapamycin
  • control animals that did not receive HUMIRA have no anti-HUMIRA titers, as expected; whereas a robust antibody response can be observed in the control animals that received only HUMIRA.
  • the animals treated with the nanocarriers remain completely negative even after three injections of HUMIRA without treatment.
  • the monitoring of the limbs of these animals revealed a progressive disease that was already evident at 10 weeks of age in the control animals.
  • the animals treated with HUMIRA alone had a significant blockade of the disease progression however adding nanocarriers to this regimen dramatically blocked the emergence of arthritic symptoms.
  • the scores here represent the total of 4 independent scorers as: 1) represents synovitis, joint effusions and soft tissue swelling 2) includes proliferating inflamed synovial tissue which grows into joint cavity and destroys cartilage 3) shows extensive loss of cartilage, erosion around the margins of joint, and deformities 4) is almost end stage of the disease with fibrous or bony stiffening of joint, which ends it's functional life.
  • HUMIRA based on the level of reduction in the arthritic score with the same dose of HUMIRA, reduced amounts of HUMIRA could also be used that would provide improved efficacy as compared to the amounts of HUMIRA that would be needed without the concomitant administration of immunosuppressant doses as provided herein. It is important to note that the amount of HUMIRA used in this example was 60 ⁇ g, much reduced as compared with amounts used in the art of about 200 ⁇ g in order to effect maximal inhibition of inflammation by HUMIRA (Binder et al., Arthritis & Rheumatism, Vol. 65 (No. 3), March 2013, pp. 608-617). Finally, the results support that a protocol was established as provided herein.
  • Example 12 Mesoporous Silica Nanoparticles with Attached Ibuprofen (Prophetic)
  • 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.
  • 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 resuspended in suitable buffer for further analysis and bioassays.
  • the liposomes are formed by thin film hydration. 1,2-Dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) (32 ⁇ mol), cholesterol (32 ⁇ mol), and rapamycin (6.4 ⁇ mol) are dissolved in pure chloroform (3 mL). This lipid solution is added to a 10 mL glass tube and the solvent is removed under nitrogen gas stream and desiccated for 6 hr. under vacuum. Multilamellar vesicles are obtained by hydration of the film with 2.0 ml of 25 mM MOPS buffer pH 8.5, containing excess amount of Ovalbumin. The tube is vortexed until the lipid film is peeled off from the tube surface.
  • DPPC 1,2-Dipalmitoyl-sn-glycero-3-phosphocholine
  • rapamycin 6.4 ⁇ mol
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Code R1017).
  • PLGA with a lactide:glycolide ratio of 3:1 and an inherent viscosity of 0.75 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 7525 DLG 7A).
  • EMPROVE® Polyvinyl Alcohol 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).
  • Solution 1 PLGA at 75 mg/mL, PLA-PEG-OMe at 25 mg/mL, and rapamycin at 12.5 mg/mL in methylene chloride. The solution was prepared by dissolving PLGA, PLA-PEG-OMe, and rapamycin in pure methylene chloride.
  • Solution 2 Polyvinyl alcohol @ 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • the O/W emulsion was prepared by combining Solution 1 (1.0 mL) and Solution 2 (3.0 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 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 75,600 ⁇ g and 4° 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.
  • Transgenic animals overexpressing human tumor necrosis factor alpha develop progressive rheumatoid arthritis over the course of 20 weeks from birth. This process can be prevented by using the fully human anti-human TNF ⁇ antibody Adalimumab or HUMIRA.
  • HUMIRA anti-drug antibody formation
  • Age-matched HuTNF ⁇ Tg 5 week old female animals were either injected s.c. in the subscapular area with saline (PBS) or 60 ⁇ g of HUMIRA weekly or with a mixture of HUMIRA and 0.87 mg of tolerogenic nanoparticles for the first 7 injections (day 0 to 42, weeks 5 to 11 of age) followed by 10 weekly injections of HUMIRA alone (same amounts) (day 49 to 107, weeks 12 to 20 of age).
  • PBS subsaline
  • HUMIRA subsaline
  • tolerogenic nanoparticles for the first 7 injections (day 0 to 42, weeks 5 to 11 of age) followed by 10 weekly injections of HUMIRA alone (same amounts) (day 49 to 107, weeks 12 to 20 of age).
  • the protocol is shown in FIG. 14 .
  • FIG. 15 shows the antibody titers in the blood of all animals at different time points.
  • the control mock-treated animals have no titers as expected whereas a robust anti-HUMIRA antibody response can be observed in the animals that received just HUMIRA.
  • Treatment with tolerogenic nanocarriers from week 5 to week 11 of age led to a complete resistance to develop anti-HUMIRA titers even after 10 injections of HUMIRA without the tolerogenic treatment (weeks 12 to 20).
  • the monitoring of the limbs of these animals revealed a progressive disease that was already evident at 10 weeks of age.
  • HUMIRA alone had a significant blockade of the disease progression however adding tolerogenic nanocarriers to this regimen dramatically blocked the emergence of arthritic symptoms.
  • the scores here represent the average total of 4 independent scorers as: 1) represents synovitis, joint effusions and soft tissue swelling 2) includes proliferating inflamed synovial tissue which grows into joint cavity and destroys cartilage 3) shows extensive loss of cartilage, erosion around the margins of joint, and deformities 4) is almost end stage of the disease with fibrous or bony stiffening of joint, which ends it's functional life.
  • compositions provided herein when administered over a period of time concomitantly with a protein can reduce or prevent antibody formation to a biological therapeutic and therefore improving its efficacy and therapeutic window for prolonged periods of time.
  • administration of immunosuppressant attached to synthetic nanocarriers with HUMIRA can reduce undesired anti-HUMIRA antibody response.
  • the benefit of the concomitant administration is also evidenced by the further reduced arthritic score which demonstrates improved efficacy of HUMIRA. This demonstrates not only the reduction of undesired immune responses against a therapeutic protein with concomitant administration of immunosuppressant attached to synthetic nanocarriers but also improved efficacy.
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Code R1017).
  • PLGA with a lactide:glycolide ratio of 3:1 and an inherent viscosity of 0.75 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 7525 DLG 7A).
  • EMPROVE® Polyvinyl Alcohol 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).
  • Solution 1 PLGA at 75 mg/mL, PLA-PEG-OMe at 25 mg/mL, and rapamycin at 12.5 mg/mL in methylene chloride. The solution was prepared by dissolving PLGA, PLA-PEG-OMe, and rapamycin in pure methylene chloride.
  • Solution 2 Polyvinyl alcohol @ 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • the O/W emulsion was prepared by combining Solution 1 (1.0 mL) and Solution 2 (3.0 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 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 75,600 ⁇ g and 4° 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.
  • Rapamycin-containing nanocarriers were generated using the materials and methods described above. 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.
  • Anti-HUMIRA immune and neutralizing responses in arthritic animals were evaluated.
  • Transgenic animals overexpressing human tumor necrosis factor alpha (huTNF ⁇ Tg) develop progressive rheumatoid arthritis over the course of 20 weeks from birth. This process can be prevented by using the fully human anti-human TNF ⁇ antibody Adalimumab or HUMIRA.
  • HUMIRA human tumor necrosis factor alpha
  • Adalimumab human anti-human TNF ⁇ antibody
  • Humira 200 ⁇ g
  • Humira 200 ⁇ g
  • Age-matched HuTNF ⁇ Tg 5 week old female animals were either injected s.c. in the subscapular area with saline (PBS) or 60 ⁇ g or 200 ⁇ g of HUMIRA weekly (weeks 5-10) or with a mixture of 60 ⁇ g of Humira and 0.87 mg of tolerogenic nanoparticles for the first 3 injections (week 5 to 7 of age) followed by 3 weekly injections of HUMIRA alone (same amounts) (weeks 8 to 10 of age).
  • the results in FIG. 17 show the antibody titers in the blood of all animals at different time points.
  • the control mock-treated animals have no titers as expected whereas a robust anti-HUMIRA antibody response can be observed in the animals that received just HUMIRA.
  • FIG. 16 illustrates the dosing regimen.
  • the monitoring of the limbs of these animals revealed a progressive disease that was already evident at 6 weeks of age. HUMIRA alone had a significant blockade of the disease progression however adding nanocarriers to this regimen dramatically blocked the emergence of arthritic symptoms.
  • the scores ( FIG. 17 (right panel)) represent the average total of 4 independent scorers as: 1) represents synovitis, joint effusions and soft tissue swelling 2) includes proliferating inflamed synovial tissue which grows into joint cavity and destroys cartilage 3) shows extensive loss of cartilage, erosion around the margins of joint, and deformities 4) is almost end stage of the disease with fibrous or bony stiffening of joint, which ends it's functional life.
  • HUMIRA based on the level of reduction in the arthritic score with the same dose of HUMIRA, reduced amounts of HUMIRA could also be used that would provide improved efficacy as compared to the amounts of HUMIRA that would be needed without the concomitant administration of immunosuppressant doses as provided herein. It is important to note that the amount of HUMIRA used in this example was 60 ⁇ g, much reduced as compared with amounts used in the art of about 200 ⁇ g in order to effect maximal inhibition of inflammation by HUMIRA (Binder et al., Arthritis & Rheumatism, Vol. 65 (No. 3), March 2013, pp. 608-617). Finally, the results support that a protocol was established as provided herein.
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702), product code R1017.
  • 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.
  • PLA-PEG-OMe block co-polymer with a methyl ether terminated PEG block of approximately 5,000 Da and an overall inherent viscosity of 0.50 DL/g was purchased from Lakeshore Biomaterials (756 Tom Martin Drive, Birmingham, Ala. 35211), product code 100 DL mPEG 5000 5CE.
  • EMPROVE® Polyvinyl Alcohol 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.
  • Cellgro phosphate buffered saline 1 ⁇ (PBS 1 ⁇ ) was purchased from Corning (9345 Discovery Blvd. Manassas, Va. 20109), product code 21-040-CV.
  • Solution 1 A polymer and rapamycin mixture was prepared by dissolving PLGA at 75 mg per 1 mL, PLA-PEG-Ome at 25 mg per 1 mL, and rapamycin as 12.5 mg per 1 mL in dichloromethane.
  • Solution 2 Polyvinyl alcohol was prepared at 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • O/W emulsions was prepared by combining Solution 1 (1.0 mL) and Solution 2 (3.0 mL) in a small glass pressure tube and sonicating at 30% amplitude for 60 seconds using a Branson Digital Sonifier 250.
  • the O/W emulsion was added to an open beaker containing 70 mM pH 8 phosphate buffer solution (60 mL).
  • Three additional, identical O/W emulsions were prepared and added to the same beaker as the first. These were 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 centrifuge tubes and centrifuging at 75,600 ⁇ g and 4° C. for 35 minutes, removing the supernatant, and re-suspending the pellet in PBS 1 ⁇ . The wash procedure was repeated and then 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.
  • An identical formulation was prepared as above in a separate beaker, and combined with the first after the wash step. The mixed nanocarrier solution was then filtered using 1.2 ⁇ m PES membrane syringe filters from Pall part number 4656, and 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.
  • Ovalbumin protein was purchased from Worthington Biochemical Corporation (730 Vassar Avenue, Lakewood, N.J. 08701), product code LS003054).
  • PLGA with 54% lactide and 46% glycolide content 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 ⁇ (PBS 1 ⁇ ) was purchased from Corning (9345 Discovery Blvd. Manassas, Va. 20109), product code 21-040-CV.
  • Solution 1 Ovalbumin protein @ 50 mg/mL was prepared in 10 mM phosphate buffer pH 8 with 10% by weight sucrose.
  • Solution 2 PLGA was prepared by dissolving PLGA at 100 mg per 1 mL of dichloromethane in the chemical fume hood.
  • Solution 3 PLA-PEG-OMe was prepared by dissolving PLA-PEG-OMe at 100 mg per 1 mL of dichloromethane in the chemical fume hood.
  • Solution 4 Polyvinyl alcohol @ 65 mg/mL in 100 mM phosphate buffer, pH 8.
  • a primary (W1/O) emulsion was first created by mixing Solutions 1 through 3.
  • Solution 1 (0.2 mL), Solution 2 (0.75 mL), and Solution 3 (0.25 mL) were combined in a small glass pressure tube which was pre-chilled >4 minutes in an ice water bath, and sonicated at 50% amplitude for 40 seconds over an ice bath using a Branson Digital Sonifier 250.
  • a secondary (W1/O/W2) emulsion was then formed by adding Solution 4 (3 mL) to the primary emulsion, vortex mixing to create a milky dispersion, and then sonicating at 30% amplitude for 60 seconds over an ice bath using the Branson Digital Sonifier 250.
  • the secondary emulsion was added to an open 50 mL beaker containing PBS 1 ⁇ (30 mL).
  • a second identical double emulsion formulation was prepared as described above, and added to the same 50 mL beaker as the first. The two preparations were stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 75,600 rcf for 50 minutes, removing the supernatant, and re-suspending the pellet in PBS 1 ⁇ . This washing procedure was repeated and then 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 suspension was stored frozen at ⁇ 20 C until use.
  • 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.
  • mice C57BL/6 age-matched (5-6 weeks) female mice were injected i.v. in the tail vein on days ⁇ 21 and ⁇ 14 with saline (No Treatment), 1.1 mg of whole Ovalbumin-loaded nanocarriers (NP[OVA]) combined to either 1.2 mg of rapamycin-containing nanocarriers (NP[Rapa]) or 8 mg of GSK1059615-loaded nanocarriers (NP[GSK1059615]).
  • Ovalbumin-loaded nanocarriers NP[OVA]
  • NP[Rapa] 1.2 mg
  • GSK1059615-loaded nanocarriers NP[GSK1059615]
  • Three thousand two hundred human subjects suffering from rheumatoid arthritis are recruited for a series of clinical trials.
  • 1,200 subjects are divided into four arms (placebo and 3 different dose levels of synthetic nanocarriers of Example 3).
  • Each subject in each of the four arms receives two rounds of HUMIRA 40 mg s.c. concomitantly with either placebo or synthetic nanocarrier.
  • the synthetic nanocarrier dose that most reduces the mean level of anti-HUMIRA antibodies in an arm is declared to be the “Immunosuppressant Dose” for the trial.
  • the recruited human subjects are divided into 4 Test Arms of 500 subjects each.
  • Placebo, HUMIRA, and synthetic nanocarriers of Example 3 are administered concomitantly (except for Test Arm 1) according to the following table, with the synthetic nanocarriers being administered at the Immunosuppressant Dose.
  • the target pharmacodynamic effects (“PD effect”) are evaluated, which in this case are the means of ACR 20, 50 and 70 responses for the subjects in each Test Arm.
  • the PD effect for subjects in Test Arm 1 is noted, and the HUMIRA dose in Test Arm 1 is arbitrarily defined as the pharmacodynamically effective dose (“PD Effective Dose”) of HUMIRA.
  • the PD effect for Test Arms 2-4 are then evaluated, and the lowest dose at which the PD effect is not significantly different from the Test Arm 1 PD effect is declared to be the reduced pharmacodynamically effective dose of HUMIRA.
  • a reduced pharmacodynamically effective dose of HUMIRA is administered concomitantly with the Immunosuppressant Dose (containing synthetic nanocarriers of Example 3) to subjects diagnosed with rheumatoid arthritis and at risk of developing antibodies to HUMIRA.
  • a protocol using the information established during the pilot trials is prepared to guide concomitant dosing of HumiraTM and synthetic nanocarriers of Example 3 to human subjects diagnosed with rheumatoid arthritis and have or are expected to have antibodies to HUMIRA. This protocol is then used to guide concomitant administration of a reduced pharmacodynamically effective dose of HUMIRA, and synthetic nanocarriers of Example 3, to human subjects.
  • Three thousand seven hundred human subjects suffering from rheumatoid arthritis are recruited for a series of clinical trials.
  • 1,200 subjects are divided into four arms (placebo and 3 different dose levels of synthetic nanocarriers of Example 3).
  • Each subject in each of the four arms receives two rounds of HUMIRA 40 mg s.c. concomitantly with either placebo or synthetic nanocarrier.
  • the synthetic nanocarrier dose that most reduces the mean level of anti-HUMIRA antibodies in an arm is declared to be the “Immunosuppressant Dose” for the trial.
  • the recruited human subjects are divided into 3 Test Arms, with two active Test Arms having 1000 subjects each, and one placebo arm of 500 subjects.
  • Placebo, HUMIRA, and synthetic nanocarriers of Example 3 are administered concomitantly (except for Test Arm 1) according to the following table, with the synthetic nanocarriers being administered at the Immunosuppressant Dose.
  • the target pharmacodynamic effects (“PD effect”) are evaluated, which in this case are the means of ACR 20, 50 and 70 responses for the subjects in each Test Arm.
  • the PD effect for subjects in Test Arm 1 is noted and compared to the PD effect in Test Arm 2.
  • Any enhancement of the pharmacodynamic effect of HUMIRA upon concomitant administration with the Immunosuppressant Dose in Test Arm 2 is noted, as compared with the PD effect observed in Test Arm 1.
  • the standard 40 mg dose of HUMIRA is administered concomitantly with the Immunosuppressant Dose (containing synthetic nanocarriers of Example 3) to subjects diagnosed with rheumatoid arthritis and at risk of developing antibodies to HUMIRA.
  • a protocol using the information established during the pilot trials is prepared to guide concomitant dosing of HUMIRA and synthetic nanocarriers of Example 3 to human subjects diagnosed with rheumatoid arthritis and known or believed to have antibodies to HUMIRA. This protocol is then used to guide concomitant administration of HUMIRA, and synthetic nanocarriers of Example 3, to human subjects. Any enhanced pharmacodynamic effect following the concomitant administration is recorded using the approaches disclosed herein.
  • modified mRNA encoding erythropoietin is prepared according to U.S. Patent application 2013/0115272 to de Fougerolles et al. (“mmRNA”). Twelve hundred subjects are divided into four arms (placebo and 3 different dose levels of synthetic nanocarriers of Example 3). Each subject in each of the four arms receives a therapeutic dose of mmRNA concomitantly with either placebo or synthetic nanocarrier.
  • the synthetic nanocarrier dose that most reduces the mean level of anti-mmRNA antibodies in an arm is declared to be the “Immunosuppressant Dose” for the trial.
  • the recruited human subjects are divided into 4 Test Arms of 500 subjects each.
  • Placebo, mmRNA, and synthetic nanocarriers of Example 3 are administered concomitantly (except for Test Arm 1) according to the following table, with the synthetic nanocarriers being administered at the Immunosuppressant Dose.
  • the target pharmacodynamic effects (“PD effect”) are evaluated, which in this case are the means of chemotherapy-induced anemia responses for the subjects in each Test Arm.
  • the PD effect for subjects in Test Arm 1 is noted, and the mmRNA dose in Test Arm 1 is arbitrarily defined as the pharmacodynamically effective dose (“PD Effective Dose”) of mmRNA.
  • PD Effective Dose the pharmacodynamically effective dose of mmRNA.
  • the PD effect for Test Arms 2-4 are then evaluated, and the lowest dose at which the PD effect is not significantly different from the Test Arm 1 PD effect is declared to be the reduced pharmacodynamically effective dose of mmRNA.
  • a reduced pharmacodynamically effective dose of mmRNA is administered concomitantly with the Immunosuppressant Dose (containing synthetic nanocarriers of Example 3) to subjects diagnosed with chemotherapy-related anemia and at risk of developing antibodies to mmRNA.
  • a protocol using the information established during the pilot trials is prepared to guide concomitant dosing of mmRNA and synthetic nanocarriers of Example 3 to human subjects diagnosed with chemotherapy-related anemia and known or believed to have antibodies to mmRNA. This protocol is then used to guide concomitant administration of a reduced pharmacodynamically effective dose of mmRNA, and synthetic nanocarriers of Example 3, to human subjects.
  • Example 22 Method to Maintain Efficacy of a Biologic Drug During a Period of Multidosing
  • Biologic drugs often lose activity over time after multiple doses.
  • a common cause of loss of efficacy is the formation of ADAs.
  • ADAs can cause loss of drug efficacy, for example by 1) enhancing clearance of the drug, such that the PK of the drug after multiple doses is substantially less than the PK of the drug after a single dose or by 2) neutralizing the activity of the drug, such that the biological activity of the drug after multiple doses is substantially less than the biological activity after a single dose. It would be desirable to maintain activity of the biologic drug over the course of multiple dosing.
  • mice expressing human TNF- ⁇ spontaneously develop arthritis over a period of 5-20 weeks of age were treated weekly with HUMIRA (60 ⁇ g/injection) with or without synthetic nanocarriers from Example 16 from weeks 5-11.
  • Panel B shows the serum levels of HUMIRA after the first dose (Day 1). After 6 doses of HUMIRA, the blood levels of HUMIRA are close to baseline in mice treated with HUMIRA alone and remain low through week 20 (Panel C, FIG. 19 , black squares). In contrast, mice treated with HUMIRA and the synthetic nanocarriers show serum levels of HUMIRA (panel C, FIG. 19 , blue triangles) that are similar to that after the first dose, indicating that synthetic nanocarrier treatment enabled effective blood levels of HUMIRA to be maintained after multiple dosing.
  • mice treated with HUMIRA alone developed high titer anti-drug antibodies (Panel A, FIG. 19 , black square symbols).
  • mice treated with the synthetic nanocarriers show little or no anti-HUMIRA antibody response (Panel A, FIG. 19 , blue triangles).
  • the impact of the reduction of HUMIRA serum levels with multiple dosing is apparent in the arthritis disease scores as above in Example 16 (Panel D, FIG. 19 ).
  • Mice treated with HUMIRA alone show suboptimal attenuation of arthritis scores from week 10-20 (Panel D, FIG. 19 , compare black squares to black circles).
  • mice treated with synthetic nanocarriers show strong inhibition of arthritis (Panel C, FIG. 19 , blue triangles).
  • Three thousand seven hundred human subjects suffering from rheumatoid arthritis are recruited for a series of clinical trials.
  • 1,200 subjects are divided into four arms (placebo and 3 different dose levels of nanocrystalline rapamycin).
  • Each subject in each of the four arms receives two rounds of HUMIRA 40 mg s.c. concomitantly with either placebo or nanocrystalline rapamycin.
  • the nanocrystalline rapamycin dose that most reduces the mean level of anti-HUMIRA antibodies in an arm is declared to be the “Immunosuppressant Dose” for the trial.
  • the recruited human subjects are divided into 3 Test Arms, with two active Test Arms having 1000 subjects each, and one placebo arm of 500 subjects.
  • Placebo, HUMIRA, and nanocrystalline rapamycin are administered concomitantly (except for Test Arm 1) according to the following table, with the synthetic nanocarriers being administered at the Immunosuppressant Dose.
  • the target pharmacodynamic effects (“PD effect”) are evaluated, which in this case are the means of ACR 20, 50 and 70 responses for the subjects in each Test Arm.
  • the PD effect for subjects in Test Arm 1 is noted and compared to the PD effect in Test Arm 2.
  • Any enhancement of the pharmacodynamic effect of HUMIRA upon concomitant administration with the Immunosuppressant Dose in Test Arm 2 is noted, as compared with the PD effect observed in Test Arm 1.
  • the standard 40 mg dose of HUMIRA is administered concomitantly with the Immunosuppressant Dose (containing nanocrystalline rapamycin) to subjects diagnosed with rheumatoid arthritis and at risk of developing antibodies to HUMIRA.
  • a protocol using the information established during the pilot trials is prepared to guide concomitant dosing of HUMIRA and nanocrystalline rapamycin to human subjects diagnosed with rheumatoid arthritis and known or believed to have antibodies to HUMIRA. This protocol is then used to guide concomitant administration of HUMIRA and nanocrystalline rapamycin, to human subjects. Any enhanced pharmacodynamic effect following the concomitant administration is recorded using the approaches disclosed herein.
  • modified mRNA encoding erythropoietin is prepared according to U.S. Patent application 2013/0115272 to de Fougerolles et al. (“mmRNA”). Twelve hundred subjects are divided into four arms (placebo and 3 different dose levels of nanocrystalline rapamycin). Each subject in each of the four arms receives a therapeutic dose of mmRNA concomitantly with either placebo or nanocrystalline rapamycin. The nanocrystalline rapamycin dose that most reduces the mean level of anti-mmRNA antibodies in an arm is declared to be the “Immunosuppressant Dose” for the trial.
  • the recruited human subjects are divided into 4 Test Arms of 500 subjects each.
  • Placebo, mmRNA, and synthetic nanocarriers of Example 3 are administered concomitantly (except for Test Arm 1) according to the following table, with the nanocrystalline rapamycin being administered at the Immunosuppressant Dose.
  • the target pharmacodynamic effects (“PD effect”) are evaluated, which in this case are the means of chemotherapy-induced anemia responses for the subjects in each Test Arm.
  • the PD effect for subjects in Test Arm 1 is noted, and the mmRNA dose in Test Arm 1 is arbitrarily defined as the pharmacodynamically effective dose (“PD Effective Dose”) of mmRNA.
  • the PD effect for Test Arms 2-4 are then evaluated, and the lowest dose at which the PD effect is not significantly different from the Test Arm 1 PD effect is declared to be the reduced pharmacodynamically effective dose of mmRNA.
  • a reduced pharmacodynamically effective dose of mmRNA is administered concomitantly with the Immunosuppressant Dose (containing nanocrystalline rapamycin) to subjects diagnosed with chemotherapy-related anemia and at risk of developing antibodies to mmRNA.
  • a protocol using the information established during the pilot trials is prepared to guide concomitant dosing of mmRNA and nanocrystalline rapamycin to human subjects diagnosed with chemotherapy-related anemia and known or believed to have antibodies to mmRNA. This protocol is then used to guide concomitant administration of a reduced pharmacodynamically effective dose of mmRNA and nanocrystalline rapamycin, to human subjects.

Abstract

Disclosed are compositions and methods that provide pharmacodynamic effects specific to therapeutic macromolecules. The effects may result from reduced doses of therapeutic macromolecules in combination with immunosuppressant doses. The effects may also be enhanced with such compositions.

Description

    RELATED APPLICATIONS
  • This application is a continuation of U.S. patent application Ser. No. 14/269,047, filed May 2, 2014, which claims the benefit under 35 U.S.C. § 119 of U.S. provisional applications 61/819,517, filed May 3, 2013; 61/881,851, filed Sep. 24, 2013; 61/881,913, filed Sep. 24, 2013; 61/881,921, filed Sep. 24, 2013; 61/907,177, filed Nov. 21, 2013; 61/948,313, filed Mar. 5, 2014; and 61/948,384, filed Mar. 5, 2014, the entire contents of each of which are incorporated herein by reference.
  • FIELD OF THE INVENTION
  • This invention relates to immunosuppressant doses, in some embodiments attached to synthetic nanocarriers that are administered concomitantly with a therapeutic macromolecule, and related methods. The compositions and methods allow for efficient pharmacodynamic effects specific to the therapeutic macromolecule. The compositions and methods provided can, therefore, be used to generate pharmacodynamic responses in a subject even at reduced doses of the therapeutic macromolecule. The compositions and methods provided herein can also be administered repeatedly concomitantly to generate desired pharmacodynamic and immunologic effects.
  • BACKGROUND OF THE INVENTION
  • Therapeutic treatments, such as protein or enzyme replacement therapies, often result in undesired immune responses to the particular therapeutic. In such cases, cells of the immune system recognize the therapeutic as foreign and attempt to neutralize or destroy it, just as they attempt to destroy infecting organisms such as bacteria and viruses. Such undesired immune responses can neutralize the efficacy of the therapeutic treatment or cause hypersensitive reactions to the therapeutic. These undesired responses may be reduced through the use of immunosuppressant drugs. Conventional immunosuppressant drugs, however, are broad-acting, and the use of broad-acting immunosuppressants is associated with a risk of severe side effects, such as tumors, infections, nephrotoxicity and metabolic disorders. Accordingly, new therapies would be beneficial.
  • SUMMARY OF THE INVENTION
  • In one aspect, a method comprising providing an immunosuppressant dose, wherein in some embodiments the immunosuppressant dose is attached to synthetic nanocarriers, and administering a reduced pharmacodynamically effective dose of a therapeutic macromolecule concomitantly with the immunosuppressant dose to a subject is provided. In one embodiment, the concomitant administration is according to a protocol that has been demonstrated to result in a pharmacodynamic effect with the reduced pharmacodynamically effective dose of the therapeutic macromolecule upon concomitant administration with the immunosuppressant dose, as compared to administration of the therapeutic macromolecule when not administered concomitantly with the immunosuppressant dose, and each in the presence of an anti-therapeutic macromolecule antibody response. In another embodiment of any one of the methods provided, the reduced pharmacodynamically effective dose of the therapeutic macromolecule is less than a pharmacodynamically effective dose of the therapeutic macromolecule that: (A) is administered in the presence of an anti-therapeutic macromolecule antibody response, and (B) is not administered concomitantly with the immunosuppressant dose.
  • In another aspect a method comprising providing an immunosuppressant dose, wherein in some embodiments the immunosuppressant dose is attached to synthetic nanocarriers, and administering a pharmacodynamically effective dose of a therapeutic macromolecule concomitantly with the immunosuppressant dose is provided. In one embodiment, the concomitant administration is according to a protocol that has been demonstrated to enhance a pharmacodynamic effect of the therapeutic macromolecule upon concomitant administration with the immunosuppressant dose, as compared to administration of the therapeutic macromolecule when not administered concomitantly with the immunosuppressant dose, and each in the presence of an anti-therapeutic macromolecule antibody response.
  • In another aspect, a method comprising providing an immunosuppressant dose, wherein in some embodiments the immunosuppressant dose is attached to synthetic nanocarriers, administering a pharmacodynamically effective dose of a therapeutic macromolecule concomitantly with the immunosuppressant dose, and recording an enhanced pharmacodynamic effect following the concomitant administration is provided.
  • In another aspect, a method comprising providing therapeutic macromolecules that cause or are expected to cause anti-therapeutic macromolecule antibodies upon repeated dosing in one or more subjects; and providing an immunosuppressant dose, wherein the immunosuppressant dose is attached to synthetic nanocarriers. In some embodiments, the method comprises repeatedly dosing at the same or a lower dose a subject with the therapeutic macromolecules concomitantly with the immunosuppressant dose. In some embodiments, the concomitant administration is according to a protocol that has been demonstrated to result in maintenance of a pharmacodynamic effect of the therapeutic macromolecule over two or more doses of the therapeutic macromolecule to a subject.
  • In one embodiment of any one of the methods provided, the method further comprises determining the protocol. In another embodiment of any one of the methods provided, the method further comprises determining the pharmacodynamically effective dose, such as the reduced or enhanced pharmacodynamically effective dose. In another embodiment of any one of the methods provided, the method further comprises assessing the pharmacodynamic effect in the subject prior to and/or after the administration. In another embodiment of any one of the methods provided, the concomitant administration is repeated one or more times. In another embodiment of any one of the methods provided, the administering is by intravenous, intraperitoneal or subcutaneous administration. In another embodiment of any one of the methods provided, the subject is at risk of an anti-therapeutic macromolecule antibody response. In another embodiment of any one of the methods provided, this subject is one in which the anti-therapeutic macromolecule response is expected to occur.
  • In another aspect, a composition or kit comprising an immunosuppressant dose wherein, in some embodiments, the immunosuppressant is attached to synthetic nanocarriers, and a reduced pharmacodynamically effective dose of a therapeutic macromolecule is provided.
  • In another aspect, a composition or kit comprising a reduced pharmacodynamically effective dose of a therapeutic macromolecule for use in any one of the methods provided herein is provided in combination with an immunosuppressant dose wherein in some embodiments the immunosuppressant is attached to synthetic nanocarriers.
  • In one embodiment of any one of the compositions or kits provided, the composition or kit is for use in any one of the methods provided herein. In one embodiment of any one of the compositions or kits provided, the composition or kit further comprises a pharmaceutically acceptable carrier.
  • In one embodiment of any one of the methods or compositions or kits provided, the therapeutic macromolecule is not attached to the synthetic nanocarriers. In another embodiment of any one of the methods or compositions or kits provided, the therapeutic macromolecule is attached to the synthetic nanocarriers. In another embodiment of any one of the methods or compositions or kits provided, the synthetic nanocarriers do not comprise therapeutic macromolecule APC presentable antigens.
  • In one embodiment of any one of the compositions or kits provided, the immunosuppressant dose and the therapeutic macromolecule are each contained in a container. In another embodiment of any one of the compositions or kits provided, the immunosuppressant dose and the therapeutic macromolecule are contained in separate containers. In another embodiment of any one of the compositions or kits provided, the immunosuppressant dose and the therapeutic macromolecule are contained in the same container.
  • In one embodiment of any one of the methods or compositions or kits provided, the reduced pharmacodynamically effective dose of the therapeutic macromolecule is at least 30% less than a pharmacodynamically effective dose of the therapeutic macromolecule that (A) is administered in the presence of an anti-therapeutic macromolecule antibody response, and (B) is not administered concomitantly with the immunosuppressant dose. In another embodiment of any one of the methods or compositions or kits provided, the reduced pharmacodynamically effective dose is at least 40% less. In another embodiment of any one of the methods or compositions or kits provided, the reduced pharmacodynamically effective dose is at least 50% less.
  • In one embodiment of any one of the methods or compositions or kits provided, the immunosuppressant dose comprises a statin, an mTOR inhibitor, a TGF-β signaling agent, a corticosteroid, an inhibitor of mitochondrial function, a P38 inhibitor, an NF-κβ inhibitor, an adenosine receptor agonist, a prostaglandin E2 agonist, a phosphodiesterase 4 inhibitor, an HDAC inhibitor or a proteasome inhibitor. In another embodiment of any one of the methods or compositions or kits provided, the mTOR inhibitor is rapamycin.
  • In one embodiment of any one of the methods or compositions or kits provided, the therapeutic macromolecule comprises a therapeutic protein. In another embodiment of any one of the methods or compositions or kits provided, the therapeutic macromolecule comprises a therapeutic polynucleotide. In another embodiment of any one of the methods or compositions or kits provided, the therapeutic protein is for protein replacement or protein supplementation therapy. In another embodiment of any one of the methods or compositions or kits provided, the therapeutic macromolecule comprises a/an infusible or injectable therapeutic protein, enzyme, enzyme cofactor, hormone, blood or blood coagulation factor, cytokine, interferon, growth factor, monoclonal antibody, polyclonal antibody or protein associated with Pompe's disease. In another embodiment of any one of the methods or compositions or kits provided, the infusible or injectable therapeutic protein comprises Tocilizumab, alpha-1 antitrypsin, Hematide, albinterferon alfa-2b, Rhucin, tesamorelin, ocrelizumab, belimumab, pegloticase, pegsiticase, taliglucerase alfa, agalsidase alfa or velaglucerase alfa. In another embodiment of any one of the methods or compositions or kits provided, the enzyme comprises an oxidoreductase, transferase, hydrolase, lyase, isomerase or ligase. In another embodiment of any one of the methods or compositions or kits provided, the enzyme comprises an enzyme for enzyme replacement therapy for a lysosomal storage disorder. In another embodiment of any one of the methods or compositions or kits provided, the enzyme for enzyme replacement therapy for a lysosomal storage disorder comprises imiglucerase, a-galactosidase A (a-gal A), agalsidase beta, acid α-glucosidase (GAA), alglucosidase alfa, LUMIZYME, MYOZYME, arylsulfatase B, laronidase, ALDURAZYME, idursulfase, ELAPRASE, arylsulfatase B or NAGLAZYME. In another embodiment of any one of the methods or compositions or kits provided, the enzymes comprise KRYSTEXXA (pegloticase). In another embodiment of any one of the methods or compositions or kits provided, the monoclonal antibody comprises HUMIRA (adalimumab). In another embodiment of any one of the methods or compositions or kits provided, the cytokine comprises a lymphokine, interleukin, chemokine, type 1 cytokine or a type 2 cytokine. In another embodiment of any one of the methods or compositions or kits provided, the blood or blood coagulation factor comprises Factor I, Factor II, tissue factor, Factor V, Factor VII, Factor VIII, Factor IX, Factor X, Factor Xa, Factor XII, Factor XIII, von Willebrand factor, prekallikrein, high-molecular weight kininogen, fibronectin, antithrombin III, heparin cofactor II, protein C, protein S, protein Z, protein Z-related protease inhibitor (ZPI), plasminogen, alpha 2-antiplasmin, tissue plasminogen activator (tPA), urokinase, plasminogen activator inhibitor-1 (PAI1), plasminogen activator inhibitor-2 (PAI2), cancer procoagulant or epoetin alfa. In another embodiment of any one of the methods or compositions or kits provided, the blood or blood coagulation factor is Factor VIII.
  • In one embodiment of any one of the methods or compositions or kits provided, a load of immunosuppressant attached to the synthetic nanocarriers, on average across the synthetic nanocarriers, is between 0.1% and 50%. In another embodiment of any one of the methods or compositions or kits provided, the load is between 0.1% and 20%.
  • In one embodiment of any one of the methods or compositions or kits provided, the synthetic nanocarriers comprise lipid nanoparticles, polymeric nanoparticles, metallic nanoparticles, surfactant-based emulsions, dendrimers, buckyballs, nanowires, virus-like particles or peptide or protein particles. In another embodiment of any one of the methods or compositions or kits provided, the synthetic nanocarriers comprise lipid nanoparticles. In another embodiment of any one of the methods or compositions or kits provided, the synthetic nanocarriers comprise liposomes. In another embodiment of any one of the methods or compositions or kits provided, the synthetic nanocarriers comprise metallic nanoparticles. In another embodiment of any one of the methods or compositions or kits provided, the metallic nanoparticles comprise gold nanoparticles. In another embodiment of any one of the methods or compositions or kits provided, the synthetic nanocarriers comprise polymeric nanoparticles. In another embodiment of any one of the methods or compositions or kits provided, the polymeric nanoparticles comprise polymer that is a non-methoxy-terminated, pluronic polymer. In another embodiment of any one of the methods or compositions or kits provided, the polymeric nanoparticles comprise a polyester, polyester attached to a polyether, polyamino acid, polycarbonate, polyacetal, polyketal, polysaccharide, polyethyloxazoline or polyethyleneimine. In another embodiment of any one of the methods or compositions or kits provided, the polyester comprises a poly(lactic acid), poly(glycolic acid), poly(lactic-co-glycolic acid) or polycaprolactone. In another embodiment of any one of the methods or compositions or kits provided, the polymeric nanoparticles comprise a polyester and a polyester attached to a polyether. In another embodiment of any one of the methods or compositions or kits provided, the polyether comprises polyethylene glycol or polypropylene glycol.
  • In one embodiment of any one of the methods or compositions or kits provided, the mean of a particle size distribution obtained using dynamic light scattering of the synthetic nanocarriers is a diameter greater than 100 nm. In another embodiment of any one of the methods or compositions or kits provided, the diameter is greater than 150 nm. In another embodiment of any one of the methods or compositions or kits provided, the diameter is greater than 200 nm. In another embodiment of any one of the methods or compositions or kits provided, the diameter is greater than 250 nm. In another embodiment of any one of the methods or compositions or kits provided, the diameter is greater than 300 nm.
  • In one embodiment of any one of the methods or compositions or kits provided, an aspect ratio 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.
  • In another aspect, a method of manufacturing any one of the compositions or kits provided herein is provided. In one embodiment, the method of manufacturing comprises producing a dose or dosage form of a therapeutic macromolecule and producing a dose or dosage form of an immunosuppressant. In some embodiments, the dose or dosage form of a therapeutic macromolecule is a reduced pharamcodynamically effective dose of the therapeutic macromolecule. In another embodiment of any one of the methods of manufacturing provided, the step of producing a dose or dosage form of an immunosuppressant comprises attaching the immunosuppressant to synthetic nanocarriers. In another embodiment of any one of the methods of manufacturing provided, the method further comprises combining the dose or dosage form of the immunosuppressant and dose or dosage form of the therapeutic macromolecule in a kit.
  • In another aspect, a use of any of the compositions or kits provided herein for the manufacture of a medicament for reducing an anti-therapeutic macromolecule antibody response, in a subject is provided. In one embodiment, the composition or kit comprises an immunosuppressant and a therapeutic macromolecule, wherein the therapeutic macromolecule may be provided in a reduced pharmacodynamically effective dose of the therapeutic macromolecule. In another embodiment of any one of the uses provided herein, the immunosuppressant is attached to synthetic nanocarriers. In another embodiment of any one of the uses provided herein, the use if for achieving any one of the methods provided herein.
  • In another aspect, any one of the composition or kits provided herein may be for use in any one of the methods provided herein. In one embodiment, the composition or kit comprises a one or more doses or dosage forms of a therapeutic macromolecule and/or one or more doses or dosage forms of an immunosuppressant. In one embodiment, the doses of a therapeutic macromolecule are reduced pharmacodynamically effective dose. In another embodiment, the immunosuppressant is attached to synthetic nanocarriers.
  • In another aspect, a method of manufacturing a medicament intended for reducing an anti-therapeutic macromolecule antibody response, is provided. In one embodiment, the medicament comprises an immunosuppressant and/or a therapeutic macromolecule, wherein the therapeutic macromolecule may be at a reduced pharmacodynamically effective dose. In another embodiment of any one of the methods of manufacturing provided herein, the immunosuppressant is attached to synthetic nanocarriers.
  • BRIEF DESCRIPTION OF FIGURES
  • FIG. 1 shows the level of circulating antigen-specific antibody production with a concomitant administration as provided herein.
  • FIG. 2 shows the level of circulating antigen-specific antibody production with a concomitant administration as provided herein.
  • FIG. 3 provides the anti-OVA antibody titers with a concomitant administration as provided herein.
  • FIG. 4 provides the anti-KLH antibody titers with a concomitant administration as provided herein.
  • FIG. 5 shows the antibody recall response to FVIII one month following a final nanocarrier and FVIII dosing.
  • FIGS. 6A and 6B show the efficacy of nanocarrier and FVIII dosing in Hemophilia A mice.
  • FIGS. 7A and 7B show immune responses to HUMIRA in mice that were treated with HUMIRA/adalimumab with or without nanocarriers attached to rapamycin.
  • FIG. 8 shows anti-Keyhole Limpet Hemocyanin (KLH) antibody titers in mice that were treated with KLH with or without nanocarriers attached to rapamycin.
  • FIG. 9 shows anti-ovalbumin (OVA) antibody titers in mice that were treated with OVA with or without nanocarriers attached to rapamycin.
  • FIG. 10 shows anti-KRYSTEXXA antibody titers in mice that were treated with KRYSTEXXA with or with nanocarriers attached to rapamycin.
  • FIG. 11 shows antibody titers in mice that were treated with OVA and KLH either in the presence or absence of nanocarriers attached to rapamycin.
  • FIGS. 12A and 12B show immune responses to KLH in mice that were treated with KLH with or without nanocarriers attached to rapamycin.
  • FIGS. 13A and 13B show immune responses to HUMIRA/adalimumab in mice that were treated with HUMIRA/adalimumab with or without nanocarriers attached to rapamycin.
  • FIG. 14 provides an exemplary protocol for practicing the methods provided herein.
  • FIG. 15 shows the beneficial effects of practicing a method provided herein in regard to therapy with HUMIRA.
  • FIG. 16 provides an exemplary protocol for practicing the methods provided herein.
  • FIG. 17 shows the beneficial effects of practicing a method provided herein in regard to therapy with HUMIRA.
  • FIG. 18 demonstrates the reduction in anti-protein antibody responses as a result of two different immunosuppressants attached to synthetic nanocarriers.
  • FIG. 19 shows another exemplary protocol and the beneficial effects of practicing a method provided herein in regard to therapy with HUMIRA.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Before describing the present invention in detail, it is to be understood that this invention is not limited to particularly exemplified materials or process parameters as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments of the invention only, and is not intended to be limiting of the use of alternative terminology to describe the present invention.
  • All publications, patents and patent applications cited herein, whether supra or infra, are hereby incorporated by reference in their entirety for all purposes.
  • As used in this specification and the appended claims, the singular forms “a,” “an” and “the” include plural referents unless the content clearly dictates otherwise. For example, reference to “a polymer” includes a mixture of two or more such molecules or a mixture of differing molecular weights of a single polymer species, reference to “a synthetic nanocarrier” includes a mixture of two or more such synthetic nanocarriers or a plurality of such synthetic nanocarriers, reference to “a RNA molecule” includes a mixture of two or more such RNA molecules or a plurality of such RNA molecules, reference to “an immunosuppressant” includes a mixture of two or more such materials or a plurality of such immunosuppressant molecules, and the like.
  • As used herein, 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, element, characteristics, properties, method/process steps or limitations) but not the exclusion of any other integer or group of integers. Thus, as used herein, the term “comprising” is inclusive and does not exclude additional, unrecited integers or method/process steps.
  • In embodiments of any one of the compositions and methods provided herein, “comprising” 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. As used herein, 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, element, characteristics, properties, method/process steps or limitations) alone.
  • A. Introduction
  • The methods, composition or kits provided herein can be used to improve the pharmacodynamic effect(s) of a therapeutic macromolecule in a subject in which an antibody response against the therapeutic macromolecule has been mounted. Accordingly, the methods or compositions or kits provide herein can be used to improve the pharmacodynamics effect(s) of therapeutic macromolecules that are otherwise diminished because of an anti-therapeutic macromolecule antibody response. Without being bound by a particular theory, it is thought that undesirable humoral immune responses against therapeutic macromolecules can be reduced using the methods, compositions or kits provided. In some embodiments, the methods, compositions or kits can be used to tolerize a subject against a therapeutic macromolecule reducing undesirable immune responses that would otherwise result when the therapeutic macromolecule is administered without the concomitant administration of an immunosuppressant dose as provided, such dose may be repeatedly concomitantly administered. Such undesirable immune responses can result in the enhanced clearance of the therapeutic macromolecule, or other interference with the therapeutic activity of the therapeutic macromolecule. As a result of the reduced undesirable immune responses, therefore, the pharmacodynamic effect(s) of a therapeutic macromolecule can be enhanced and/or reduced dosage amounts of the therapeutic macromolecule can be used to achieve the same level of effect with the methods, compositions or kits provided. As another result of the reduced undesirable immune responses, therefore, repeated dosings of a therapeutic macromolecule can be administered to a subject.
  • It has been unexpectedly and surprisingly found that delivering immunosuppressants, preferably in some embodiments when attached to synthetic nanocarriers, concomitantly with therapeutic macromolecules, and in the presence of an anti-therapeutic macromolecule antibody response, can result in enhanced pharmacodynamic effects. For example, the aforementioned combinations can help neutralize anti-therapeutic macromolecule-specific antibodies that interfere with the desired treatment effects of a therapeutic macromolecule. The methods, compositions or kits provided herein, in some embodiments, not only reduce the undesired immune responses against the therapeutic macromolecule but also result in the enhancement of the desired therapeutic effect of the therapeutic macromolecule that would otherwise be diminished when the therapeutic macromolecule is administered alone (as a result of an undesirable immune response to that therapeutic macromolecule). Thus, methods, compositions or kits provided herein can allow for a subject to obtain the treatment benefit of a therapeutic macromolecule without needing to increase the dose of the therapeutic macromolecule, which generally would be increased in order to compensate for undesired immune responses against the therapeutic macromolecule when administered without the benefit of the invention provided herein. Surprisingly, the methods, compositions or kits provided herein even allow for a subject to be administered reduced doses of the therapeutic macromolecule to achieve the same therapeutic benefit.
  • As undesired immune responses that are generated during therapeutic treatment with therapeutic macromolecules can be counteracted with the methods, compositions or kits provided, the invention is useful for achieving enhanced pharmacodynamic effects, or for using a reduced pharmacodynamically effective doses, in subjects in which undesired immune responses against the therapeutic macromolecule are generated or are expected to be generated. In one embodiment of any one of the methods provided herein, the subject can be one at risk for such an undesired immune response.
  • The invention will now be described in more detail below.
  • B. Definitions
  • “Administering” or “administration” or “administer” means providing a material to a subject in a manner that is pharmacologically useful. The term is intended to include causing to be administered in some embodiments. “Causing to be administered” means causing, urging, encouraging, aiding, inducing or directing, directly or indirectly, another party to administer the material.
  • “Amount effective” in the context of a composition or dose for administration to a subject refers to an amount of the composition or dose that produces one or more desired responses in the subject, for example, the generation of a tolerogenic immune response (e.g., a reduction in the proliferation, activation, induction, survival, recruitment of therapeutic macromolecule-specific B cells or a reduction in the production of therapeutic macromolecule-specific antibodies). In some embodiments, the amount effective is a pharmacodynamically effective amount. Therefore, in some embodiments, an amount effective is any amount of a composition or dose provided herein (or multiple compositions or doses as provided herein) that produces one or more of the desired pharmacodynamic effects, therapeutic effects and/or immune responses as provided herein. This amount can be for in vitro or in vivo purposes. For in vivo purposes, the amount can be one that a clinician would believe may have a clinical benefit for a subject in need of therapeutic macromolecule administration and/or antigen-specific immune tolerance thereto.
  • 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 that is effective can also be an amount that produces a desired therapeutic endpoint or a desired therapeutic result. In other embodiments, the amounts effective can involve enhancing the level of a desired response, such as a therapeutic endpoint or result. Amounts effective, preferably, result in a tolerogenic immune response in a subject to an antigen, such as a therapeutic macromolecule. The achievement of any of the foregoing can be monitored by routine methods.
  • In some embodiments of any one of the methods provided, the amount effective is one in which the desired response persists in the subject for at least 1 week, at least 2 weeks, at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, or longer. In other embodiments of any of the compositions and methods provided, the amount effective is one which produces a measurable desired response for at least 1 week, at least 2 weeks, at least 1 month, at least 2 months, at least 3 months, at least 4 months, at least 5 months, or longer.
  • Amounts effective will depend, of course, on the particular subject being treated; the severity of a condition, disease or disorder; the individual patient parameters including age, physical condition, size and weight; the duration of the treatment; the nature of concurrent therapy (if any); the specific route of administration and like factors within the knowledge and expertise of the health practitioner. These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is generally preferred that a maximum dose be used, that is, the highest safe dose according to sound medical judgment. It will be understood by those of ordinary skill in the art, however, that a patient may insist upon a lower dose or tolerable dose for medical reasons, psychological reasons or for virtually any other reason.
  • In general, doses of the immunosuppressants and/or therapeutic macromolecules in the compositions of the invention refer to the amount of the immunosuppressants and/or therapeutic macromolecules. Alternatively, the dose can be administered based on the number of synthetic nanocarriers that provide the desired amount of immunosuppressants and/or therapeutic macromolecules.
  • “Anti-therapeutic macromolecule antibody response” or “anti-therapeutic macromolecule-specific antibody response” is the generation of anti-therapeutic macromolecule-specific antibodies, or induction of the process for producing such antibodies, as a result of administration of a therapeutic macromolecule. In embodiments, such a response counteracts the therapeutic effects of the therapeutic macromolecule.
  • “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. In some embodiments, antigens may be proteins, polypeptides, peptides, lipoproteins, glycolipids, polynucleotides, polysaccharides or are contained or expressed in cells. In some embodiments, such as when the antigens are not well defined or characterized, the antigens may be contained within a cell or tissue preparation, cell debris, cell exosomes, conditioned media, etc.
  • “Antigen-specific” refers to any immune response that results from the presence of the antigen, or portion thereof, or that generates molecules that specifically recognize or bind the antigen. In some embodiments, when the antigen comprises the therapeutic macromolecule, antigen-specific may mean therapeutic macromolecule-specific. For example, where the immune response is antigen-specific antibody production, such as therapeutic macromolecule-specific antibody production, antibodies are produced that specifically bind the antigen (e.g., therapeutic macromolecule). As another example, where the immune response is antigen-specific B cell or CD4+ T cell proliferation and/or activity, the proliferation and/or activity results from recognition of the antigen, or portion thereof, alone or in complex with MHC molecules, B cells, etc.
  • “Assessing a pharmacodynamic effect” refers to any measurement or determination of the level, presence or absence, reduction, increase in, etc. of a pharmacodynamic effect 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 in any one of the methods provided herein or otherwise known in the art.
  • An “at risk” subject is one in which a health practitioner believes has a chance of having a disease, disorder or condition or is one a health practitioner believes has a chance of experiencing an undesired anti-therapeutic macromolecule antibody response as provided herein and would benefit from the compositions and methods provided. In an embodiment of any one of the methods, compositions or kits provided herein the subject is one that is at risk of having an anti-therapeutic macromolecule antibody response to a therapeutic macromolecule. In another embodiment of any one of the methods, compositions or kits provided herein, the subject is one that is expected to have an anti-therapeutic macromolecule antibody response to a therapeutic macromolecule.
  • “Attach” or “Attached” or “Couple” or “Coupled” (and the like) means to chemically associate one entity (for example a moiety) with another. In some embodiments, the attaching is covalent, meaning that the attachment occurs in the context of the presence of a covalent bond between the two entities. In non-covalent embodiments, 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. In embodiments, encapsulation is a form of attaching.
  • In embodiments, therapeutic macromolecules and immunosuppressants are not attached to one another, meaning that the therapeutic macromolecules and immunosuppressants are not subjected to a process specifically intended to chemically associate one with another. In embodiments, therapeutic macromolecules and/or immunosuppressants are not attached to synthetic nanocarriers, meaning that the therapeutic macromolecules (and/or immunosuppressants) and synthetic nanocarriers are not subjected to a process specifically intended to chemically associate one with another.
  • “Average”, as used herein, refers to the arithmetic mean unless otherwise noted.
  • “Combination”, as applied to two or more materials and/or agents (also referred to herein as the components), is intended to define material in which the two or more materials/agents are associated. Components may be separately identified, e.g., first component, second component, third component, etc. The terms “combined” and “combining” in this context are to be interpreted accordingly.
  • The association of the two or more materials/agents in a combination may be physical or non-physical. Examples of physically associated combined materials/agents include:
      • compositions (e.g., unitary formulations) comprising the two or more materials/agents in admixture (for example within the same unit dose);
      • compositions comprising material in which the two or more materials/agents are chemically/physicochemically linked (for example by crosslinking, molecular agglomeration or binding to a common vehicle moiety);
      • compositions comprising material in which the two or more materials/agents are chemically/physicochemically co-packaged (for example, disposed on or within lipid vesicles, particles (e.g., micro- or nanoparticles) or emulsion droplets);
      • pharmaceutical kits, pharmaceutical packs or patient packs in which the two or more materials/agents are co-packaged or co-presented (e.g., as part of an array of unit doses);
  • Examples of non-physically associated combined materials/agents include:
      • material (e.g., a non-unitary formulation) comprising at least one of the two or more materials/agents together with instructions for the extemporaneous association of the at least one compound/agent to form a physical association of the two or more materials/agents;
      • material (e.g., a non-unitary formulation) comprising at least one of the two or more materials/agents together with instructions for combination therapy with the two or more materials/agents;
      • material comprising at least one of the two or more materials/agents together with instructions for administration to a patient population in which the other(s) of the two or more materials/agents have been (or are being) administered;
      • material comprising at least one of the two or more materials/agents in an amount or in a form which is specifically adapted for use in combination with the other(s) of the two or more materials/agents.
  • As used herein, the term “combination therapy” is intended to define therapies which comprise the use of a combination of two or more materials/agents (as defined above). Thus, references to “combination therapy”, “combinations” and the use of materials/agents “in combination” in this application may refer to materials/agents that are administered as part of the same overall treatment regimen. As such, the posology of each of the two or more materials/agents may differ: each may be administered at the same time or at different times. It will, therefore, be appreciated that the materials/agents of the combination may be administered sequentially (e.g., before or after) or simultaneously, either in the same pharmaceutical formulation (i.e., together), or in different pharmaceutical formulations (i.e., separately). Simultaneously in the same formulation is as a unitary formulation whereas simultaneously in different pharmaceutical formulations is non-unitary. The posologies of each of the two or more materials/agents in a combination therapy may also differ with respect to the route of administration.
  • “Concomitantly” 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 a physiologic or immunologic response, and even more preferably the two or more materials/agents are administered in combination. In embodiments, 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. In embodiments, the materials/agents may be repeatedly administered concomitantly, that is concomitant administration on more than one occasion, such as may be provided in the Examples.
  • “Determining” or “determine” means to ascertain a factual relationship. Determining may be accomplished in a number of ways, including but not limited to performing experiments, or making projections. For instance, a dose of an immunosuppressant or therapeutic macromolecule 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 determine a protocol as provided herein. In another embodiment, the dose may be determined by testing various doses in a subject, i.e. through direct experimentation based on experience and guiding data. In embodiments, “determining” or “determine” comprises “causing to be determined.” “Causing to be determined” means causing, urging, encouraging, aiding, inducing or directing or acting in coordination with an entity for the entity to ascertain a factual relationship; including directly or indirectly, or expressly or impliedly.
  • “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.
  • “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.
  • “Generating” means causing an action, such as a physiologic or immunologic response (e.g., tolerogenic immune response) to occur, either directly oneself or indirectly.
  • “Identifying a subject” is any action or set of actions that allows a clinician to recognize a subject as one who may benefit from the methods, compositions or kits provided herein. Preferably, the identified subject is one who is in need of a therapeutic benefit from a therapeutic macromolecule as provided herein and in which an anti-therapeutic macromolecule-specific antibody response has occurred or is suspected to occur. The action or set of actions may be either directly oneself or indirectly. In one embodiment of any one of the methods provided herein, the method further comprises identifying a subject in need of a method, composition or kit as provided herein.
  • “Immunosuppressant” means a compound that causes an APC to have an immunosuppressive effect or a T cell or B cell to be suppressed (e.g., tolerogenic effect). An immunosuppressive effect generally refers to the production or expression of cytokines or other factors by the APC that reduces, inhibits or prevents an undesired immune response or that promotes a desired immune response, such as a regulatory immune response. When the APC acquires an immunosuppressive function (under the immunosuppressive effect) on immune cells that recognize an antigen presented by this APC, the immunosuppressive effect is said to be specific to the presented antigen. Without being bound by any particular theory, it is thought that the immunosuppressive effect is a result of the immunosuppressant being delivered to the APC, preferably in the presence of an antigen. In one embodiment, the immunosuppressant is one that causes an APC to promote a regulatory phenotype in one or more immune effector cells. For example, 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. In one embodiment, the immunosuppressant is one that affects the response of the APC after it processes an antigen. In another embodiment, 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; TGF-β signaling agents; TGF-β receptor agonists; histone deacetylase inhibitors, such as Trichostatin A; corticosteroids; inhibitors of mitochondrial function, such as rotenone; P38 inhibitors; NF-κ3 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 inhibitors; cytokine receptor activators; peroxisome proliferator-activated receptor antagonists; peroxisome proliferator-activated receptor agonists; histone deacetylase inhibitors; calcineurin inhibitors; phosphatase inhibitors; PI3KB inhibitors, such as TGX-221; autophagy inhibitors, such as 3-Methyladenine; aryl hydrocarbon receptor inhibitors; proteasome inhibitor I (PSI); and oxidized ATPs, such as P2X receptor blockers. Immunosuppressants also include IDO, vitamin D3, cyclosporins, such as cyclosporine A, aryl hydrocarbon receptor inhibitors, resveratrol, azathiopurine (Aza), 6-mercaptopurine (6-MP), 6-thioguanine (6-TG), FK506, sanglifehrin A, salmeterol, mycophenolate mofetil (MMF), aspirin and other COX inhibitors, niflumic acid, estriol, methotrexate and triptolide. In embodiments, the immunosuppressant may comprise any of the agents provided herein.
  • The immunosuppressant can be a compound that directly provides the immunosuppressive effect on APCs or it can be a compound that provides the immunosuppressive effect indirectly (i.e., after being processed in some way after administration). Immunosuppressants, therefore, include prodrug forms of any of the compounds provided herein.
  • In embodiments of any one of the methods, compositions or kits provided herein, the immunosuppressants provided herein are attached to synthetic nanocarriers. In preferable embodiments, the immunosuppressant is an element that is in addition to the material that makes up the structure of the synthetic nanocarrier. For example, in one embodiment, where the synthetic nanocarrier is made up of one or more polymers, the immunosuppressant is a compound that is in addition and attached to the one or more polymers. As another example, in one embodiment, where the synthetic nanocarrier is made up of one or more lipids, the immunosuppressant is again in addition and attached to the one or more lipids. In embodiments, such as where the material of the synthetic nanocarrier also results in an immunosuppressive effect, the immunosuppressant is an element present in addition to the material of the synthetic nanocarrier that results in an immunosuppressive effect.
  • Other exemplary immunosuppressants include, but are not limited, small molecule drugs, natural products, antibodies (e.g., antibodies against CD20, CD3, CD4), biologics-based drugs, carbohydrate-based drugs, nanoparticles, liposomes, RNAi, antisense nucleic acids, aptamers, methotrexate, NSAIDs; fingolimod; natalizumab; alemtuzumab; anti-CD3; tacrolimus (FK506); cytokines and growth factors, such as TGF-β and IL-10; etc. Further immunosuppressants, are known to those of skill in the art, and the invention is not limited in this respect.
  • In embodiments of any one of the methods, compositions or kits provided herein, the immunosuppressant is in a form, such as a nanocrystalline form, whereby the form of the immunosuppressant itself is a particle or particle-like. In embodiments, such forms mimic a virus or other foreign pathogen. Many drugs have been nanonized 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. As used herein a “drug nanocrystal” refers to a form of a drug (e.g., an immunosuppressant) that does not include a carrier or matrix material. In some embodiments, 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 Technology™ (Elan Corporation). In some embodiments, a surfactant or a stabilizer may be used for steric or electrostatic stability of the drug nanocrystal. In some embodiments the nanocrystal or nanocrytalline 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. In some embodiments, concomitant administration of a reduced pharmacodynamically effective dose of a therapeutic macromolecule with an immunosuppressant in nanocrystalline form results in a reduced anti-therapeutic macromolecule antibody response.
  • “Load”, when attached to a synthetic nanocarrier, is the amount of the immunosuppressant and/or therapeutic macromolecule attached to a synthetic nanocarrier based on the total dry recipe weight of materials in an entire synthetic nanocarrier (weight/weight). Generally, the load is calculated as an average across a population of synthetic nanocarriers. In one embodiment, the load 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 of the immunosuppressant and/or therapeutic macromolecule is between 0.1% and 20%. In a further embodiment, the load of the immunosuppressant and/or therapeutic macromolecule is between 0.1% and 10%. In still a further embodiment, the load of the immunosuppressant and/or therapeutic macromolecule is between 1% and 10%. In still a further embodiment, the load of the immunosuppressant is between 7% and 20%. In yet another embodiment, the load of the immunosuppressant and/or the therapeutic macromolecule 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. In yet a further embodiment, the load of the immunosuppressant and/or the therapeutic macromolecule 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. In some embodiments of the above embodiments, the load of the immunosuppressant and/or the therapeutic macromolecule is no more than 25% on average across a population of synthetic nanocarriers. In embodiments, the load is calculated as may be described in the Examples or as otherwise known in the art.
  • In some embodiments, when the form of the immunosuppressant is itself a particle or particle-like, such as a nanocrystalline immunosuppressant, 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. In an embodiment, a minimum dimension of at least 75%, preferably at least 80%, more preferably at least 90%, of the synthetic nanocarriers in a sample, based on the total number of synthetic nanocarriers in the sample, is equal to or greater than 100 nm. In an embodiment, a maximum dimension of at least 75%, preferably at least 80%, more preferably at least 90%, of the synthetic nanocarriers in a sample, based on the total number of synthetic nanocarriers in the sample, is equal to or less than 5 μm. Preferably, a minimum dimension of at least 75%, preferably at least 80%, more preferably at least 90%, of the synthetic nanocarriers in a sample, based on the total number of synthetic nanocarriers in the sample, is greater than 110 nm, more preferably greater than 120 nm, more preferably greater than 130 nm, and more preferably still greater than 150 nm. Aspects ratios of the maximum and minimum dimensions of synthetic nanocarriers may vary depending on the embodiment. For instance, aspect ratios of the maximum to minimum dimensions of the synthetic nanocarriers may vary from 1:1 to 1,000,000:1, preferably from 1:1 to 100,000:1, more preferably from 1:1 to 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. Preferably, a maximum dimension of at least 75%, preferably at least 80%, more preferably at least 90%, of the synthetic nanocarriers in a sample, based on the total number of synthetic nanocarriers in the sample is equal to or less than 3 μm, more preferably equal to or less than 2 μm, more preferably equal to or less than 1 μm, more preferably equal to or less than 800 nm, more preferably equal to or less than 600 nm, and more preferably still equal to or less than 500 nm. In preferred embodiments, a 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) may be obtained, in some embodiments, by suspending the synthetic nanocarriers in a liquid (usually aqueous) media and using dynamic light scattering (DLS) (e.g., using a Brookhaven ZetaPALS instrument). For example, 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.
  • “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.
  • “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.
  • “Pharmacodynamic effect” of “pharmacodynamic response” means any physiologic or immunologic response as a result of the administration of a therapeutic macromolecule. Such a response can be a desired response such as one that is associated with a therapeutic effect. The methods, compositions or kits provided herein, in some embodiments, have been found to result in enhanced pharmacodynamic effects such as enhanced therapeutic effects, when administered in the presence of an anti-therapeutic macromolecule antibody response. In some instances, the enhanced pharmacodynamics effects can be obtained with a dose of the therapeutic macromolecule that is the same as or less than a dose of the therapeutic macromolecule when administered without the concomitant administration of an immunosuppressant dose as provided herein in the presence of an anti-therapeutic macromolecule antibody response. Whether a material/agent is pharmacodynamically effective can be evaluated by standard methods. In some embodiments, a pharmacodynamic effect using a method, composition or kit provided, in the presence of an anti-therapeutic macromolecule antibody response, is compared to the pharmacodynamic effect when a therapeutic macromolecule is not so administered but also in the presence of an anti-therapeutic macromolecule antibody response. In embodiments, the comparison is to the pharmacodynamic effect when the therapeutic macromolecule is administered alone in the presence of an anti-therapeutic macromolecule antibody response. Generally, a pharmacodynamic effect is assessed with administration in the presence of an anti-therapeutic macromolecule antibody response, as it is desired that a method, composition or kit is effective to overcome such a response. Accordingly, pharmacodynamic effects are determined when such a response is occurring.
  • “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, dosing frequency, routes of administration, dosing duration, dosing rates, interval between dosing, combinations of any of the foregoing, and the like. In some embodiments, such 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 a pharmacodynamic effect. Any other therapeutic and/or immunologic effect may also be assessed instead of or in addition to the aforementioned immune responses. 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. 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. Such 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. In embodiments, a number of compositions as provided herein are administered to another subject using one or more or all or substantially all of the elements of which the protocol is comprised. In some embodiments, the protocol has been demonstrated to result in a reduction of an antibody response against a therapeutic macromolecule and/or improved pharmacodynamic effect.
  • “Providing” means an action or set of actions that an individual performs that supply a needed item or set of items or methods for practicing of the present invention. The action or set of actions may be taken either directly oneself or indirectly.
  • “Providing a subject” is any action or set of actions that causes a clinician to come in contact with a subject and administer a composition provided herein thereto or to perform a method provided herein thereupon. Preferably, the subject is one who is in need of therapeutic macromolecule administration and antigen-specific immune tolerance thereto. The action or set of actions may be taken either directly oneself or indirectly. In one embodiment of any one of the methods provided herein, the method further comprises providing a subject.
  • “Recording an enhanced pharmacodynamic effect” means noting, or causing directly or indirectly activities in the expectation that such noting would take place, in any written or electronic form, that a therapeutic macromolecule dose achieved an enhanced pharmacodynamic effect in the presence of a real, expected or suspected anti-therapeutic macromolecule antibody response. Generally, in such circumstances the therapeutic macromolecule dose would not have been expected to achieve the enhanced pharmacodynamic effect in the presence of an anti-therapeutic macromolecule antibody response if administered without an immunosuppressant dose as provided herein (e.g., administered alone) based on information available at the time of concomitant administration as provided herein. For example, in such circumstances, it would be expected that the effectiveness of the therapeutic macromolecule would be diminished if administered without an immunosuppressant dose, but an enhanced pharmacodynamic effect is instead observed with a concomitant administration as provided herein. In some embodiments, the recording occurs when an immunosuppressant dose in combination with a therapeutic macromolecule dose are administered to a subject or at some point thereafter. In some of these embodiments, the dose of the therapeutic macromolecule is reduced as compared to (or no greater than) a dose of therapeutic macromolecule that is administered without the immunosuppressant dose in the presence of an anti-therapeutic macromolecule antibody response. “Written form”, as used herein, refers to any recordation on a medium such as paper. “Electronic form”, as used herein, refers to any recordation on electronic media. Any one of the methods provided herein can further comprise a step of recording a therapeutic and/or immune response in a subject receiving a treatment according to a method provided herein.
  • “Reduced pharmacodynamically effective dose” refers to a reduced amount of a therapeutic macromolecule that can achieve a similar pharmacodynamic effect when administered concomitantly with an immunosuppressant dose as provided herein as compared to the amount of the therapeutic macromolecule when not administered with the immunosuppressant dose (e.g., when the therapeutic macromolecule is administered alone). As used herein, a similar pharmacodynamic effect is a level of effect that is within one log of another level measured in the same way. Preferably, a similar pharmacodynamic effect is no more than 5-fold different. Still more preferably, a similar pharmacodynamic effect is no more than 2-fold different.
  • “Subject” means animals, including warm blooded mammals such as humans and primates; avians; domestic household or farm animals such as cats, dogs, sheep, goats, cattle, horses and pigs; laboratory animals such as mice, rats and guinea pigs; fish; reptiles; zoo and wild animals; and the like.
  • “Synthetic nanocarrier(s)” means a discrete object that is not found in nature, and that possesses at least one dimension that is less than or equal to 5 microns in size. Albumin nanoparticles are generally included as synthetic nanocarriers, however in certain embodiments the synthetic nanocarriers do not comprise albumin nanoparticles. In embodiments, synthetic nanocarriers do not comprise chitosan. In other embodiments, synthetic nanocarriers are not lipid-based nanoparticles. In further embodiments, 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. 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. 5,543,158 to Gref et al., (2) the polymeric nanoparticles of Published US Patent Application 20060002852 to Saltzman et al., (3) the lithographically constructed nanoparticles of Published US Patent Application 20090028910 to DeSimone et al., (4) the disclosure of WO 2009/051837 to von Andrian et al., (5) the nanoparticles disclosed in Published US Patent Application 2008/0145441 to Penades et al., (6) the protein nanoparticles disclosed in Published US Patent Application 20090226525 to de los Rios et al., (7) the virus-like particles disclosed in published US Patent Application 20060222652 to Sebbel et al., (8) the nucleic acid attached virus-like particles disclosed in published US Patent Application 20060251677 to Bachmann et al., (9) the virus-like particles disclosed in WO2010047839A1 or WO2009106999A2, (10) the nanoprecipitated nanoparticles disclosed in P. Paolicelli et al., “Surface-modified PLGA-based Nanoparticles that can Efficiently Associate and Deliver Virus-like Particles” Nanomedicine. 5(6):843-853 (2010), (11) apoptotic cells, apoptotic bodies or the synthetic or semisynthetic mimics disclosed in U.S. Publication 2002/0086049, or (12) those of Look et al., Nanogel-based delivery of mycophenolic acid ameliorates systemic lupus erythematosus in mice” J. Clinical Investigation 123(4):1741-1749(2013). In embodiments, synthetic nanocarriers may possess an aspect ratio greater than 1:1, 1:1.2, 1:1.5, 1:2, 1:3, 1:5, 1:7, or greater than 1:10.
  • Synthetic nanocarriers according to the invention that have a minimum dimension of equal to or less than about 100 nm, preferably equal to or less than 100 nm, do not comprise a surface with hydroxyl groups that activate complement or alternatively comprise a surface that consists essentially of moieties that are not hydroxyl groups that activate complement. In a preferred embodiment, synthetic nanocarriers according to the invention that have a minimum dimension of equal to or less than about 100 nm, preferably equal to or less than 100 nm, do not comprise a surface that substantially activates complement or alternatively comprise a surface that consists essentially of moieties that do not substantially activate complement. In a more preferred embodiment, synthetic nanocarriers according to the invention that have a minimum dimension of equal to or less than about 100 nm, preferably equal to or less than 100 nm, do not comprise a surface that activates complement or alternatively comprise a surface that consists essentially of moieties that do not activate complement. In embodiments, synthetic nanocarriers exclude virus-like particles. In embodiments, synthetic nanocarriers may possess an aspect ratio greater than 1:1, 1:1.2, 1:1.5, 1:2, 1:3, 1:5, 1:7, or greater than 1:10.
  • A “therapeutic macromolecule” refers to any protein, carbohydrate, lipid or nucleic acid that may be administered to a subject and have a therapeutic effect. In some embodiments, administration of the therapeutic macromolecule to a subject may result in an undesired immune response, including production of anti-therapeutic macromolecule-specific antibodies. As described herein, administration of a therapeutic macromolecule concomitantly with an immunosuppressant dose can enhance the therapeutic effectiveness of the therapeutic macromolecule, such as by reducing undesired immune responses thereto. In some embodiments, the therapeutic macromolecule may be a therapeutic polynucleotide or therapeutic protein.
  • “Therapeutic polynucleotide” means any polynucleotide or polynucleotide-based therapy that may be administered to a subject and have a therapeutic effect. Such therapies include gene silencing. Examples of such therapy are known in the art, and include, but are not limited to, naked RNA (including messenger RNA, modified messenger RNA, and forms of RNAi). Examples of other therapeutic polynucleotides are provided elsewhere herein. Therapeutic polynucleotides may be produced in, on or by cells and also may be obtained using cell free or from fully synthetic in vitro methods. Subjects, therefore, include any subject that is in need of treatment with any of the foregoing. Such subject include those that will receive any of the foregoing.
  • “Therapeutic protein” means any protein or protein-based therapy that may be administered to a subject and have a therapeutic effect. Such therapies include protein replacement and protein supplementation therapies. Such therapies also include the administration of exogenous or foreign proteins, antibody therapies, and cell or cell-based therapies. Therapeutic proteins comprise, but are not limited to, enzymes, enzyme cofactors, hormones, blood clotting factors, cytokines, growth factors, monoclonal antibodies, antibody-drug conjugates, and polyclonal antibodies. Examples of other therapeutic proteins are provided elsewhere herein. Therapeutic proteins may be produced in, on or by cells and may be obtained from such cells or administered in the form of such cells. In embodiments, the therapeutic protein is produced in, on or by mammalian cells, insect cells, yeast cells, bacteria cells, plant cells, transgenic animal cells, transgenic plant cells, etc. The therapeutic protein may be recombinantly produced in such cells. The therapeutic protein may be produced in, on or by a virally transformed cell. Subjects, therefore, include any subject that is in need of treatment with any of the foregoing. Such subject include those that will receive any of the foregoing.
  • “Therapeutic macromolecule APC presentable antigen” means an antigen that is associated with a therapeutic macromolecule (i.e., the therapeutic macromolecule or a fragment thereof that can generate an immune response against the therapeutic macromolecule (e.g., the production of anti-therapeutic macromolecule-specific antibodies)). Generally, therapeutic macromolecule antigen-presenting cell (APC) presentable antigens can be presented for recognition by the immune system (e.g., cells of the immune system, such as presented by antigen presenting cells, including but not limited to dendritic cells, B cells or macrophages). The therapeutic macromolecule APC presentable antigen can be presented for recognition by, for example, T cells. Such antigens may be recognized by and trigger an immune response in a T cell via presentation of an epitope of the antigen bound to Class I or Class II major histocompatability complex molecule (MHC). Therapeutic macromolecule APC presentable antigens generally include proteins, polypeptides, peptides, polynucleotides, lipoproteins, or are contained or expressed in, on or by cells. The therapeutic macromolecule antigens, in some embodiments, are attached to synthetic nanocarriers and comprise MHC Class I-restricted epitopes and/or MHC Class II-restricted epitopes and/or B cell epitopes. Preferably, one or more tolerogenic immune responses specific to the therapeutic macromolecule result with the methods, compositions or kits provided herein. In embodiments, populations of the synthetic nanocarriers comprise no added therapeutic macromolecule APC presentable antigens, meaning that no substantial amounts of therapeutic macromolecule APC presentable antigens are intentionally added to the synthetic nanocarriers during the manufacturing thereof.
  • “Undesired immune response” refers to any undesired immune response that results from exposure to an antigen, promotes or exacerbates a disease, disorder or condition provided herein (or a symptom thereof), or is symptomatic of a disease, disorder or condition provided herein. Such immune responses generally have a negative impact on a subject's health or is symptomatic of a negative impact on a subject's health. Undesired immune responses include antigen-specific antibody production, antigen-specific B cell proliferation and/or activity or antigen-specific CD4+ T cell proliferation and/or activity. Generally, these undesired immune responses can be specific to a therapeutic macromolecule and counteract the beneficial effects desired of administration with the therapeutic macromolecule. Thus, in some embodiments, the undesired immune response is an anti-therapeutic macromolecule antibody response.
  • C. Compositions and Related Methods
  • Provided herein are compositions comprising immunosuppressants and therapeutic macromolecules and related methods or kits. Such methods, compositions or kits are useful to enhance the pharmacodynamics effect of a therapeutic macromolecule in the presence of an anti-therapeutic macromolecule antibody response, such as through the reduction or inhibition of undesired immune responses specific to the therapeutic macromolecule that diminish the therapeutic benefit of the therapeutic macromolecule. Such methods, compositions or kits are also useful to allow for the repeated dosing of a therapeutic macromolecule. Thus, the methods, compositions or kits provided herein are useful for achieving or enhancing desired therapeutic effects of the therapeutic macromolecules. In some embodiments, such therapeutic effects can be achieved or enhanced at reduced pharmacodynamically effective doses. The methods, compositions or kits provided can be used for any subject in need of the therapeutic benefit of a therapeutic macromolecule.
  • As mentioned above, it was found that delivering immunosuppressants, preferably in some embodiments when attached to synthetic nanocarriers, concomitantly with therapeutic macromolecules in the presence of an anti-therapeutic macromolecule antibody response can result in enhanced pharmacodynamic effects, including an enhancement of such effects even at reduced doses of the therapeutic macromolecules. For example, the methods, compositions or kits can help neutralize anti-therapeutic macromolecule-specific antibodies that interfere with the desired treatment effects of a therapeutic macromolecule. The methods, compositions or kits provided herein can, therefore, result in an enhancement of a desired therapeutic effect of a therapeutic macromolecule that would otherwise be diminished when the therapeutic macromolecule is administered without the immunosuppressant dose (e.g., when the therapeutic macromolecule is administered alone). Thus, in some embodiments, the methods, compositions or kit provided herein allow for a subject to obtain the treatment benefit of a therapeutic macromolecule without needing to increase the dose of the therapeutic macromolecule, which generally would be increased in order to compensate for undesired immune responses against the therapeutic macromolecule when administered without the benefit of the invention provided herein. Surprisingly, the methods, compositions or kits provided herein even allow for a subject to be administered reduced doses of the therapeutic macromolecule to achieve the same or better therapeutic benefit in the presence of an anti-therapeutic macromolecule antibody response.
  • A variety of immunosuppressants may be used in the practice of the present invention, which preferably, are attached to synthetic nanocarriers. A wide variety of synthetic nanocarriers can be used according to the invention. In some embodiments, synthetic nanocarriers are spheres or spheroids. In some embodiments, synthetic nanocarriers are flat or plate-shaped. In some embodiments, synthetic nanocarriers are cubes or cubic. In some embodiments, synthetic nanocarriers are ovals or ellipses. In some embodiments, synthetic nanocarriers are cylinders, cones, or pyramids.
  • In some embodiments, it is desirable to use a population of synthetic nanocarriers that is relatively uniform in terms of size or shape so that each synthetic nanocarrier has similar properties. For example, at least 80%, at least 90%, or at least 95% of the synthetic nanocarriers, based on the total number of synthetic nanocarriers, may have a minimum dimension or maximum dimension that falls within 5%, 10%, or 20% of the average diameter or average dimension of the synthetic nanocarriers.
  • Synthetic nanocarriers can be solid or hollow and can comprise one or more layers. In some embodiments, each layer has a unique composition and unique properties relative to the other layer(s). To give but one example, synthetic nanocarriers may have a core/shell structure, wherein the core is one layer (e.g., a polymeric core) and the shell is a second layer (e.g., a lipid bilayer or monolayer). Synthetic nanocarriers may comprise a plurality of different layers.
  • In some embodiments, synthetic nanocarriers may optionally comprise one or more lipids. In some embodiments, a synthetic nanocarrier may comprise a liposome. In some embodiments, a synthetic nanocarrier may comprise a lipid bilayer. In some embodiments, a synthetic nanocarrier may comprise a lipid monolayer. In some embodiments, a synthetic nanocarrier may comprise a micelle. In some embodiments, a synthetic nanocarrier may comprise a core comprising a polymeric matrix surrounded by a lipid layer (e.g., lipid bilayer, lipid monolayer, etc.). In some embodiments, a synthetic nanocarrier may comprise a non-polymeric core (e.g., metal particle, quantum dot, ceramic particle, bone particle, viral particle, proteins, nucleic acids, carbohydrates, etc.) surrounded by a lipid layer (e.g., lipid bilayer, lipid monolayer, etc.).
  • In other embodiments, synthetic nanocarriers may comprise metal particles, quantum dots, ceramic particles, etc. In some embodiments, a non-polymeric synthetic nanocarrier is an aggregate of non-polymeric components, such as an aggregate of metal atoms (e.g., gold atoms).
  • In some embodiments, synthetic nanocarriers may optionally comprise one or more amphiphilic entities. In some embodiments, an amphiphilic entity can promote the production of synthetic nanocarriers with increased stability, improved uniformity, or increased viscosity. In some embodiments, amphiphilic entities can be associated with the interior surface of a lipid membrane (e.g., lipid bilayer, lipid monolayer, etc.). Many amphiphilic entities known in the art are suitable for use in making synthetic nanocarriers in accordance with the present invention. Such amphiphilic entities include, but are not limited to, phosphoglycerides; phosphatidylcholines; dipalmitoyl phosphatidylcholine (DPPC); dioleylphosphatidyl ethanolamine (DOPE); dioleyloxypropyltriethylammonium (DOTMA); dioleoylphosphatidylcholine; cholesterol; cholesterol ester; diacylglycerol; diacylglycerolsuccinate; diphosphatidyl glycerol (DPPG); hexanedecanol; fatty alcohols such as polyethylene glycol (PEG); polyoxyethylene-9-lauryl ether; a surface active fatty acid, such as palmitic acid or oleic acid; fatty acids; fatty acid monoglycerides; fatty acid diglycerides; fatty acid amides; sorbitan trioleate (Span®85) glycocholate; sorbitan monolaurate (Span®20); polysorbate 20 (Tween®20); polysorbate 60 (Tween®60); polysorbate 65 (Tween®65); polysorbate 80 (Tween®80); polysorbate 85 (Tween®85); polyoxyethylene monostearate; surfactin; a poloxomer; a sorbitan fatty acid ester such as sorbitan trioleate; lecithin; lysolecithin; phosphatidylserine; phosphatidylinositol; sphingomyelin; phosphatidylethanolamine (cephalin); cardiolipin; phosphatidic acid; cerebrosides; dicetylphosphate; dipalmitoylphosphatidylglycerol; stearylamine; dodecylamine; hexadecyl-amine; acetyl palmitate; glycerol ricinoleate; hexadecyl sterate; isopropyl myristate; tyloxapol; poly(ethylene glycol)5000-phosphatidylethanolamine; poly(ethylene glycol)400-monostearate; phospholipids; synthetic and/or natural detergents having high surfactant properties; deoxycholates; cyclodextrins; chaotropic salts; ion pairing agents; and combinations thereof. An amphiphilic entity component may be a mixture of different amphiphilic entities. Those skilled in the art will recognize that this is an exemplary, not comprehensive, list of substances with surfactant activity. Any amphiphilic entity may be used in the production of synthetic nanocarriers to be used in accordance with the present invention.
  • In some embodiments, synthetic nanocarriers may optionally comprise one or more carbohydrates. Carbohydrates may be natural or synthetic. A carbohydrate may be a derivatized natural carbohydrate. In certain embodiments, a carbohydrate comprises monosaccharide or disaccharide, including but not limited to glucose, fructose, galactose, ribose, lactose, sucrose, maltose, trehalose, cellbiose, mannose, xylose, arabinose, glucoronic acid, galactoronic acid, mannuronic acid, glucosamine, galatosamine, and neuramic acid. In certain embodiments, a carbohydrate is a polysaccharide, including but not limited to pullulan, cellulose, microcrystalline cellulose, hydroxypropyl methylcellulose (HPMC), hydroxycellulose (HC), methylcellulose (MC), dextran, cyclodextran, glycogen, hydroxyethylstarch, carageenan, glycon, amylose, chitosan, N,O-carboxylmethylchitosan, algin and alginic acid, starch, chitin, inulin, konjac, glucommannan, pustulan, heparin, hyaluronic acid, curdlan, and xanthan. In embodiments, the synthetic nanocarriers do not comprise (or specifically exclude) carbohydrates, such as a polysaccharide. In certain embodiments, the carbohydrate may comprise a carbohydrate derivative such as a sugar alcohol, including but not limited to mannitol, sorbitol, xylitol, erythritol, maltitol, and lactitol.
  • In some embodiments, synthetic nanocarriers can comprise one or more polymers. In some embodiments, the synthetic nanocarriers comprise one or more polymers that is a non-methoxy-terminated, 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 are non-methoxy-terminated, pluronic polymers. In some embodiments, all of the polymers that make up the synthetic nanocarriers are non-methoxy-terminated, pluronic polymers. In some embodiments, 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. In some embodiments, all of the polymers that make up the synthetic nanocarriers are non-methoxy-terminated polymers. In some embodiments, 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, various elements of the synthetic nanocarriers can be attached to the polymer.
  • The immunosuppressants and/or therapeutic macromolecules can be attached to the synthetic nanocarriers by any of a number of methods. Generally, the attaching can be a result of bonding between the immunosuppressants and/or therapeutic macromolecules and the synthetic nanocarriers. This bonding can result in the immunosuppressants and/or therapeutic macromolecules being attached to the surface of the synthetic nanocarriers and/or contained (encapsulated) within the synthetic nanocarriers. In some embodiments, however, the immunosuppressants and/or therapeutic macromolecules are encapsulated by the synthetic nanocarriers as a result of the structure of the synthetic nanocarriers rather than bonding to the synthetic nanocarriers. In preferable embodiments, the synthetic nanocarrier comprises a polymer as provided herein, and the immunosuppressants and/or and/or therapeutic macromolecules are attached to the polymer.
  • When attaching occurs as a result of bonding between the immunosuppressants and/or therapeutic macromolecules and synthetic nanocarriers, the attaching may occur via a coupling moiety. A coupling moiety can be any moiety through which an immunosuppressant and/or therapeutic macromolecule is bonded to a synthetic nanocarrier. Such 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 and/or therapeutic macromolecule to the synthetic nanocarrier. Such molecules include linkers or polymers or a unit thereof. For example, the coupling moiety can comprise a charged polymer to which an immunosuppressant and/or therapeutic macromolecule electrostatically binds. As another example, the coupling moiety can comprise a polymer or unit thereof to which it is covalently bonded.
  • In preferred embodiments, 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.
  • In some embodiments, the polymers of a synthetic nanocarrier associate to form a polymeric matrix. In some of these embodiments, a component, such as an immunosuppressant or therapeutic macromolecule, can be covalently associated with one or more polymers of the polymeric matrix. In some embodiments, covalent association is mediated by a linker. In some embodiments, a component can be noncovalently associated with one or more polymers of the polymeric matrix. For example, in some embodiments, a component can be encapsulated within, surrounded by, and/or dispersed throughout a polymeric matrix. Alternatively or additionally, a component can be associated with 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.
  • In some embodiments, the polymer comprises a polyester, polycarbonate, polyamide, or polyether, or unit thereof. In other embodiments, 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. In some embodiments, it is preferred that 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. In other embodiments, the polymer does not solely comprise a polyether or unit thereof, such as poly(ethylene glycol) or polypropylene glycol or unit thereof.
  • Other examples of 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. poly(β-hydroxyalkanoate))), poly(orthoesters), polycyanoacrylates, polyvinyl alcohols, polyurethanes, polyphosphazenes, polyacrylates, polymethacrylates, polyureas, polystyrenes, and polyamines, polylysine, polylysine-PEG copolymers, and poly(ethyleneimine), poly(ethylene imine)-PEG copolymers.
  • In some embodiments, polymers in accordance with the present invention include polymers which have been approved for use in humans by the U.S. Food and Drug Administration (FDA) under 21 C.F.R. § 177.2600, including but not limited to polyesters (e.g., polylactic acid, poly(lactic-co-glycolic acid), polycaprolactone, polyvalerolactone, poly(1,3-dioxan-2one)); polyanhydrides (e.g., poly(sebacic anhydride)); polyethers (e.g., polyethylene glycol); polyurethanes; polymethacrylates; polyacrylates; and polycyanoacrylates.
  • In some embodiments, polymers can be hydrophilic. For example, polymers may comprise anionic groups (e.g., phosphate group, sulphate group, carboxylate group); cationic groups (e.g., quaternary amine group); or polar groups (e.g., hydroxyl group, thiol group, amine group). In some embodiments, a synthetic nanocarrier comprising a hydrophilic polymeric matrix generates a hydrophilic environment within the synthetic nanocarrier. In some embodiments, polymers can be hydrophobic. In some embodiments, a synthetic nanocarrier comprising a hydrophobic polymeric matrix generates a hydrophobic environment within the synthetic nanocarrier. Selection of the hydrophilicity or hydrophobicity of the polymer may have an impact on the nature of materials that are incorporated (e.g., attached) within the synthetic nanocarrier.
  • In some embodiments, polymers may be modified with one or more moieties and/or functional groups. A variety of moieties or functional groups can be used in accordance with the present invention. In some embodiments, polymers may be modified with polyethylene glycol (PEG), with a carbohydrate, and/or with acyclic polyacetals derived from polysaccharides (Papisov, 2001, ACS Symposium Series, 786:301). Certain embodiments may be made using the general teachings of U.S. Pat. No. 5,543,158 to Gref et al., or WO publication WO2009/051837 by Von Andrian et al.
  • In some embodiments, polymers may be modified with a lipid or fatty acid group. In some embodiments, a fatty acid group may be one or more of butyric, caproic, caprylic, capric, lauric, myristic, palmitic, stearic, arachidic, behenic, or lignoceric acid. In some embodiments, a fatty acid group may be one or more of palmitoleic, oleic, vaccenic, linoleic, alpha-linoleic, gamma-linoleic, arachidonic, gadoleic, arachidonic, eicosapentaenoic, docosahexaenoic, or erucic acid.
  • In some embodiments, polymers may be polyesters, including copolymers comprising lactic acid and glycolic acid units, such as poly(lactic acid-co-glycolic acid) and poly(lactide-co-glycolide), collectively referred to herein as “PLGA”; and homopolymers comprising glycolic acid units, referred to herein as “PGA,” and lactic acid units, such as poly-L-lactic acid, poly-D-lactic acid, poly-D,L-lactic acid, poly-L-lactide, poly-D-lactide, and poly-D,L-lactide, collectively referred to herein as “PLA.” In some embodiments, exemplary polyesters include, for example, polyhydroxyacids; PEG copolymers and copolymers of lactide and glycolide (e.g., PLA-PEG copolymers, PGA-PEG copolymers, PLGA-PEG copolymers, and derivatives thereof. In some embodiments, polyesters include, for example, poly(caprolactone), poly(caprolactone)-PEG copolymers, poly(L-lactide-co-L-lysine), poly(serine ester), poly(4-hydroxy-L-proline ester), polyα-(4-aminobutyl)-L-glycolic acid], and derivatives thereof.
  • In some embodiments, a polymer may be PLGA. PLGA is a biocompatible and biodegradable co-polymer of lactic acid and glycolic acid, and various forms of PLGA are characterized by the ratio of lactic acid:glycolic acid. Lactic acid can be L-lactic acid, D-lactic acid, or D,L-lactic acid. The degradation rate of PLGA can be adjusted by altering the lactic acid:glycolic acid ratio. In some embodiments, PLGA to be used in accordance with the present invention is characterized by a lactic acid:glycolic acid ratio of approximately 85:15, approximately 75:25, approximately 60:40, approximately 50:50, approximately 40:60, approximately 25:75, or approximately 15:85.
  • In some embodiments, polymers may be one or more acrylic polymers. In certain embodiments, acrylic polymers include, for example, acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, aminoalkyl methacrylate copolymer, poly(acrylic acid), poly(methacrylic acid), methacrylic acid alkylamide copolymer, poly(methyl methacrylate), poly(methacrylic acid anhydride), methyl methacrylate, polymethacrylate, poly(methyl methacrylate) copolymer, polyacrylamide, aminoalkyl methacrylate copolymer, glycidyl methacrylate copolymers, polycyanoacrylates, and combinations comprising one or more of the foregoing polymers. The acrylic polymer may comprise fully-polymerized copolymers of acrylic and methacrylic acid esters with a low content of quaternary ammonium groups.
  • In some embodiments, polymers can be cationic polymers. In general, cationic polymers are able to condense and/or protect negatively charged strands of nucleic acids. 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. Acad. Sci., USA, 93:4897; Tang et al., 1996, Bioconjugate Chem., 7:703; and Haensler et al., 1993, Bioconjugate Chem., 4:372) are positively-charged at physiological pH, form ion pairs with nucleic acids. In embodiments, the synthetic nanocarriers may not comprise (or may exclude) cationic polymers.
  • In some embodiments, polymers can be degradable polyesters bearing cationic side chains (Putnam et al., 1999, Macromolecules, 32:3658; Barrera et al., 1993, J. Am. Chem. Soc., 115:11010; Kwon et al., 1989, Macromolecules, 22:3250; Lim et al., 1999, J. Am. Chem. Soc., 121:5633; and Zhou et al., 1990, Macromolecules, 23:3399). Examples of these polyesters include poly(L-lactide-co-L-lysine) (Barrera et al., 1993, J. Am. Chem. Soc., 115:11010), poly(serine ester) (Zhou et al., 1990, Macromolecules, 23:3399), poly(4-hydroxy-L-proline ester) (Putnam et al., 1999, Macromolecules, 32:3658; and Lim et al., 1999, J. Am. Chem. Soc., 121:5633), and poly(4-hydroxy-L-proline ester) (Putnam et al., 1999, Macromolecules, 32:3658; and Lim et al., 1999, J. Am. Chem. Soc., 121:5633).
  • The properties of these and other polymers and methods for preparing them are well known in the art (see, for example, U.S. Pat. Nos. 6,123,727; 5,804,178; 5,770,417; 5,736,372; 5,716,404; 6,095,148; 5,837,752; 5,902,599; 5,696,175; 5,514,378; 5,512,600; 5,399,665; 5,019,379; 5,010,167; 4,806,621; 4,638,045; and U.S. Pat. No. 4,946,929; Wang et al., 2001, J. Am. Chem. Soc., 123:9480; Lim et al., 2001, J. Am. Chem. Soc., 123:2460; Langer, 2000, Acc. Chem. Res., 33:94; Langer, 1999, J. Control. Release, 62:7; and Uhrich et al., 1999, Chem. Rev., 99:3181). More generally, a variety of methods for synthesizing certain suitable polymers are described in Concise Encyclopedia of Polymer Science and Polymeric Amines and Ammonium Salts, Ed. by Goethals, Pergamon Press, 1980; Principles of Polymerization by Odian, John Wiley & Sons, Fourth Edition, 2004; Contemporary Polymer Chemistry by Allcock et al., Prentice-Hall, 1981; Deming et al., 1997, Nature, 390:386; and in U.S. Pat. Nos. 6,506,577, 6,632,922, 6,686,446, and 6,818,732.
  • In some embodiments, polymers can be linear or branched polymers. In some embodiments, polymers can be dendrimers. In some embodiments, polymers can be substantially cross-linked to one another. In some embodiments, polymers can be substantially free of cross-links. In some embodiments, polymers can be used in accordance with the present invention without undergoing a cross-linking step. It is further to be understood that 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.
  • In some embodiments, synthetic nanocarriers do not comprise a polymeric component. In some embodiments, synthetic nanocarriers may comprise metal particles, quantum dots, ceramic particles, etc. In some embodiments, a non-polymeric synthetic nanocarrier is an aggregate of non-polymeric components, such as an aggregate of metal atoms (e.g., gold atoms).
  • Compositions according to the invention can comprise elements in combination with pharmaceutically acceptable excipients, such as preservatives, buffers, saline, or phosphate buffered saline. The compositions may be made using conventional pharmaceutical manufacturing and compounding techniques to arrive at useful dosage forms. In an embodiment, compositions, such as those comprising the synthetic nanocarriers are suspended in sterile saline solution for injection together with a preservative.
  • In embodiments, when preparing synthetic nanocarriers as carriers, methods for attaching components to the synthetic nanocarriers may be useful. If the component is a small molecule it may be of advantage to attach the component to a polymer prior to the assembly of the synthetic nanocarriers. In embodiments, it may also be an advantage to prepare the synthetic nanocarriers with surface groups that are used to 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.
  • In certain embodiments, the attaching can be with a covalent linker. In embodiments, components 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 on the surface of the nanocarrier with a component containing an alkyne group or by the 1,3-dipolar cycloaddition reaction of alkynes on the surface of the nanocarrier with a component containing an azido group. Such cycloaddition reactions are preferably performed in the presence of a Cu(I) catalyst along with a suitable Cu(I)-ligand and a reducing agent to reduce Cu(II) compound to catalytic active Cu(I) compound. This Cu(I)-catalyzed azide-alkyne cycloaddition (CuAAC) can also be referred as the click reaction.
  • Additionally, the covalent attaching may comprise a covalent linker that comprises an amide linker, a disulfide linker, a thioether linker, a hydrazone linker, a hydrazide linker, an imine or oxime linker, an urea or thiourea linker, an amidine linker, an amine linker, and a sulfonamide linker.
  • An amide linker is formed via an amide bond between an amine on one component with the carboxylic acid group of a second component, such as the nanocarrier. The amide bond in the linker can be made using any of the conventional amide bond forming reactions with suitably protected amino acids and activated carboxylic acid such N-hydroxysuccinimide-activated ester.
  • A disulfide linker is made via the formation of a disulfide (S—S) bond between two sulfur atoms of the form, for instance, of R1-S—S—R2. A disulfide bond can be formed by thiol exchange of a component containing thiol/mercaptan group (—SH) with another activated thiol group on a polymer or nanocarrier or a nanocarrier containing thiol/mercaptan groups with a component containing activated thiol group.
  • A triazole linker, specifically a 1,2,3-triazole of the form
  • Figure US20200113874A1-20200416-C00001
  • wherein R1 and R2 may be any chemical entities, is made by the 1,3-dipolar cycloaddition reaction of an azide attached to a first component, such as the nanocarrier, with a terminal alkyne attached to a second component, such as the immunosuppressant or therapeutic macromolecule. The 1,3-dipolar cycloaddition reaction is performed with or without a catalyst, preferably with Cu(I)-catalyst, which links the two components through a 1,2,3-triazole function. This chemistry is described in detail by Sharpless et al., Angew. Chem. Int. Ed. 41(14), 2596, (2002) and Meldal, et al, Chem. Rev., 2008, 108(8), 2952-3015 and is often referred to as a “click” reaction or CuAAC.
  • In embodiments, a polymer containing an azide or alkyne group, terminal to the polymer chain is prepared. This polymer is then used to prepare a synthetic nanocarrier in such a manner that a plurality of the alkyne or azide groups are positioned on the surface of that nanocarrier. Alternatively, the synthetic nanocarrier can be prepared by another route, and subsequently functionalized with alkyne or azide groups. The 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.
  • 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. In another way, 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. Alternatively, 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 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 to the nanocarrier via non-covalent conjugation methods. For example, a negative charged therapeutic macromolecule or immunosuppressant can be conjugated to a positive charged nanocarrier through electrostatic adsorption. A component containing a metal ligand can also be conjugated to a nanocarrier containing a metal complex via a metal-ligand complex.
  • In embodiments, the component can be attached to a polymer, for example polylactic acid-block-polyethylene glycol, prior to the assembly of the synthetic nanocarrier or the synthetic nanocarrier can be formed with reactive or activatible groups on its surface. In the latter case, the component may be prepared with a group which is compatible with the attachment chemistry that is presented by the synthetic nanocarriers' surface. In other embodiments, a peptide component can be attached to VLPs or liposomes using a suitable linker. A linker is a compound or reagent that is capable of attaching two molecules together. In an embodiment, the linker can be a homobifuntional or heterobifunctional reagent as described in Hermanson 2008. For example, an VLP or liposome synthetic nanocarrier containing a carboxylic group on the surface can be treated with a homobifunctional linker, adipic dihydrazide (ADH), in the presence of EDC to form the corresponding synthetic nanocarrier with the ADH linker. The resulting ADH linked synthetic nanocarrier is then conjugated with 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.
  • For detailed descriptions of available conjugation methods, see Hermanson G T “Bioconjugate Techniques”, 2nd Edition Published by Academic Press, Inc., 2008. In addition to covalent attachment the 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.
  • Any immunosuppressant as provided herein can be used in the methods or compositions provided and can be, in some embodiments, attached to synthetic nanocarriers. Immunosuppressants include, but are not limited to, statins; mTOR inhibitors, such as rapamycin or a rapamycin analog; TGF-β signaling agents; TGF-β receptor agonists; histone deacetylase (HDAC) inhibitors; corticosteroids; inhibitors of mitochondrial function, such as rotenone; P38 inhibitors; NF-κβ 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; cytokine receptor inhibitors; cytokine receptor activators; peroxisome proliferator-activated receptor antagonists; peroxisome proliferator-activated receptor agonists; histone deacetylase inhibitors; calcineurin inhibitors; phosphatase inhibitors and oxidized ATPs. 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.
  • Examples of statins 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®).
  • Examples of 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. Chemistry & Biology 2006, 13:99-107)), AZD8055, BEZ235 (NVP-BEZ235), chrysophanic acid (chrysophanol), deforolimus (MK-8669), everolimus (RAD0001), KU-0063794, PI-103, PP242, temsirolimus, and WYE-354 (available from Selleck, Houston, Tex., USA).
  • Examples of TGF-β signaling agents include TGF-β ligands (e.g., activin A, GDF1, GDF11, bone morphogenic proteins, nodal, TGF-βs) and their receptors (e.g., ACVR1B, ACVR1C, ACVR2A, ACVR2B, BMPR2, BMPR1A, BMPR1B, TGFβRI, TGFβRII), 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).
  • Examples of 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).
  • Examples of P38 inhibitors 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.
  • Examples of NF (e.g., NK-κβ) 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], NFκB Activation Inhibitor III, NF-κB 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, rocaglaol, (R)-MG-132, sodium salicylate, triptolide (PG490), and wedelolactone.
  • Examples of adenosine receptor agonists include CGS-21680 and ATL-146e.
  • Examples of prostaglandin E2 agonists include E-Prostanoid 2 and E-Prostanoid 4.
  • Examples of phosphodiesterase inhibitors (non-selective and selective inhibitors) 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 (PERFAN™), milrinone, levosimendon, mesembrine, ibudilast, piclamilast, luteolin, drotaverine, roflumilast (DAXAS™, DALIRESP™), sildenafil (REVATION®, VIAGRA®), tadalafil (ADCIRCA®, CIALIS®), vardenafil (LEVITRA®, STAXYN®), udenafil, avanafil, icariin, 4-methylpiperazine, and pyrazolo pyrimidin-7-1.
  • Examples of proteasome inhibitors include bortezomib, disulfiram, epigallocatechin-3-gallate, and salinosporamide A.
  • Examples of kinase inhibitors 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.
  • Examples of glucocorticoids include hydrocortisone (cortisol), cortisone acetate, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone, triamcinolone, beclometasone, fludrocortisone acetate, deoxycorticosterone acetate (DOCA), and aldosterone.
  • Examples of retinoids include retinol, retinal, tretinoin (retinoic acid, RETIN-A®), isotretinoin (ACCUTANE®, AMNESTEEM®, CLARAVIS®, SOTRET®), alitretinoin (PANRETIN®), etretinate (TEGISON™) and its metabolite acitretin (SORIATANE®), tazarotene (TAZORAC®, AVAGE®, ZORAC®), bexarotene (TARGRETIN®), and adapalene (DIFFERIN®).
  • Examples of cytokine inhibitors include IL1ra, IL1 receptor antagonist, IGFBP, TNF-BF, uromodulin, Alpha-2-Macroglobulin, Cyclosporin A, Pentamidine, and Pentoxifylline (PENTOPAK®, PENTOXIL®, TRENTAL®).
  • Examples of peroxisome proliferator-activated receptor antagonists include GW9662, PPARγ antagonist III, G335, and T0070907 (EMD4Biosciences, USA).
  • Examples of peroxisome proliferator-activated receptor agonists include pioglitazone, ciglitazone, clofibrate, GW1929, GW7647, L-165,041, LY 171883, PPARγ activator, Fmoc-Leu, troglitazone, and WY-14643 (EMD4Biosciences, USA).
  • Examples of 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.
  • Examples of calcineurin inhibitors include cyclosporine, pimecrolimus, voclosporin, and tacrolimus.
  • Examples of phosphatase inhibitors 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 IC, protein phosphatase 2A inhibitor protein, protein phosphatase 2A1, protein phosphatase 2A2, and sodium orthovanadate.
  • In some embodiments of any one of the methods, compositions or kits provided, the therapeutic macromolecules as described herein are also attached to synthetic nanocarriers. In other embodiments, the therapeutic macromolecules are not attached to any synthetic nanocarriers. In some embodiments of either of these situations, the therapeutic macromolecules may be delivered in the form of the therapeutic macromolecule itself, or fragments or derivatives thereof.
  • Therapeutic macromolecules can include therapeutic proteins or therapeutic polynucleotides. Therapeutic proteins include, but are not limited to, infusible therapeutic proteins, enzymes, enzyme cofactors, hormones, blood clotting factors, cytokines and interferons, growth factors, monoclonal antibodies, and polyclonal antibodies (e.g., that are administered to a subject as a replacement therapy), and proteins associated with Pompe's disease (e.g., acid glucosidase alfa, rhGAA (e.g., Myozyme and Lumizyme (Genzyme)). Therapeutic proteins also include proteins involved in the blood coagulation cascade. Therapeutic proteins include, but are not limited to, Factor VIII, Factor VII, Factor IX, Factor V, von Willebrand Factor, von Heldebrant Factor, tissue plasminogen activator, insulin, growth hormone, erythropoietin alfa, VEGF, thrombopoietin, lysozyme, antithrombin and the like. Therapeutic proteins also include adipokines, such as leptin and adiponectin. Other examples of therapeutic proteins are as described below and elsewhere herein.
  • Examples of therapeutic proteins used in enzyme replacement therapy of subjects having a lysosomal storage disorder include, but are not limited to, imiglucerase for the treatment of Gaucher's disease (e.g., CEREZYME™), a-galactosidase A (a-gal A) for the treatment of Fabry disease (e.g., agalsidase beta, FABRYZYME™), acid α-glucosidase (GAA) for the treatment of Pompe disease (e.g., acid glucosidase alfa, LUMIZYME™, MYOZYME™), arylsulfatase B for the treatment of Mucopolysaccharidoses (e.g., laronidase, ALDURAZYME™, idursulfase, ELAPRASE™, arylsulfatase B, NAGLAZYME™), pegloticase (KRYSTEXXA) and pegsiticase.
  • Examples of enzymes include oxidoreductases, transferases, hydrolases, lyases, isomerases, asparaginases, uricases, glycosidases, asparaginases, uricases, proteases, nucleases, collagenases, hyaluronidases, heparinases, heparanases, lysins, and ligases.
  • Therapeutic proteins may also include any enzyme, toxin, or other protein or peptide isolated or derived from a bacterial, fungal, or viral source.
  • Examples of 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, Glucagon, Glucagon-like peptide (GLP-1), GIP, Gonadotropin-releasing hormone, Growth hormone-releasing hormone, Human chorionic gonadotropin, Human placental lactogen, Growth hormone, Inhibin, Insulin, Insulin-like growth factor (or somatomedin), Leptin, Luteinizing hormone, Melanocyte stimulating hormone, Orexin, Oxytocin, Parathyroid hormone, Prolactin, Relaxin, Secretin, Somatostatin, Thrombopoietin, Thyroid-stimulating hormone (or thyrotropin), Thyrotropin-releasing hormone, Cortisol, Aldosterone, Testosterone, Dehydroepiandrosterone, Androstenedione, Dihydrotestosterone, Estradiol, Estrone, Estriol, Progesterone, Calcitriol (1,25-dihydroxyvitamin D3), Calcidiol (25-hydroxyvitamin D3), Prostaglandins, Leukotrienes, Prostacyclin, Thromboxane, Prolactin releasing hormone, Lipotropin, Brain natriuretic peptide, Neuropeptide Y, Histamine, Endothelin, Pancreatic polypeptide, Renin, and Enkephalin.
  • Examples of 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, prekallikrein (Fletcher factor), high-molecular weight kininogen (HMWK) (Fitzgerald factor), fibronectin, fibrin, thrombin, antithrombin III, heparin cofactor II, protein C, protein S, protein Z, protein Z-related protease inhibitot (ZPI), plasminogen, alpha 2-antiplasmin, tissue plasminogen activator (tPA), urokinase, plasminogen activator inhibitor-1 (PAI1), plasminogen activator inhibitor-2 (PAI2), cancer procoagulant, and epoetin alfa (Epogen, Procrit).
  • Examples of cytokines include lymphokines, interleukins, and chemokines, type 1 cytokines, such as IFN-γ, TGF-β, and type 2 cytokines, such as IL-4, IL-10, and IL-13.
  • Examples of 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-α), Transforming growth factor beta(TGF-β), Tumour_necrosis_factor-alpha(TNF-α), Vascular endothelial growth factor (VEGF), Wnt Signaling Pathway, placental growth factor (P1GF), (Foetal Bovine Somatotrophin) (FBS), IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, and IL-7.
  • Examples of monoclonal antibodies include Abagovomab, Abciximab, Adalimumab, Adecatumumab, Afelimomab, Afutuzumab, Alacizumab pegol, ALD, Alemtuzumab, Altumomab pentetate, Anatumomab mafenatox, Anrukinzumab, Anti-thymocyte globin, Apolizumab, Arcitumomab, Aselizumab, Atlizumab (tocilizumab), Atorolimumab, Bapineuzumab, Basiliximab, Bavituximab, Bectumomab, Belimumab, Benralizumab, Bertilimumab, Besilesomab, Bevacizumab, Biciromab, Bivatuzumab mertansine, Blinatumomab, Brentuximab vedotin, Briakinumab, Canakinumab, Cantuzumab mertansine, Capromab pendetide, Catumaxomab, Cedelizumab, Certolizumab pegol, Cetuximab, Citatuzumab bogatox, Cixutumumab, Clenoliximab, Clivatuzumab tetraxetan, Conatumumab, Dacetuzumab, Daclizumab, Daratumumab, Denosumab, Detumomab, Dorlimomab aritox, Dorlixizumab, Ecromeximab, Eculizumab, Edobacomab, Edrecolomab, Efalizumab, Efungumab, Elotuzumab, Elsilimomab, Enlimomab pegol, Epitumomab cituxetan, Epratuzumab, Erlizumab, Ertumaxomab, Etaracizumab, Exbivirumab, Fanolesomab, Faralimomab, Farletuzumab, Felvizumab, Fezakinumab, Figitumumab, Fontolizumab, Foravirumab, Fresolimumab, Galiximab, Gantenerumab, Gavilimomab, Gemtuzumab ozogamicin, GC1008, Girentuximab, Glembatumumab vedotin, Golimumab, Gomiliximab, Ibalizumab, Ibritumomab tiuxetan, Igovomab, Imciromab, Infliximab, Intetumumab, Inolimomab, Inotuzumab ozogamicin, Ipilimumab, Iratumumab, Keliximab, Labetuzumab, Lebrikizumab, Lemalesomab, Lerdelimumab, Lexatumumab, Libivirumab, Lintuzumab, Lorvotuzumab mertansine, Lucatumumab, Lumiliximab, Mapatumumab, Maslimomab, Matuzumab, Mepolizumab, Metelimumab, Milatuzumab, Minretumomab, Mitumomab, Morolimumab, Motavizumab, Muromonab-CD3, Nacolomab tafenatox, Naptumomab estafenatox, Natalizumab, Nebacumab, Necitumumab, Nerelimomab, Nimotuzumab, Nofetumomab merpentan, Ocrelizumab, Odulimomab, Ofatumumab, Olaratumab, Omalizumab, Oportuzumab monatox, Oregovomab, Otelixizumab, Pagibaximab, Palivizumab, Panitumumab, Panobacumab, Pascolizumab, Pemtumomab, Pertuzumab, Pexelizumab, Pintumomab, Priliximab, Pritumumab, Rafivirumab, Ramucirumab, Ranibizumab, Raxibacumab, Regavirumab Reslizumab, Rilotumumab, Rituximab, Robatumumab, Rontalizumab, Rovelizumab, Ruplizumab, Satumomab pendetide, Sevirumab, Sibrotuzumab, Sifalimumab, Siltuximab, Siplizumab, Solanezumab, Sonepcizumab, Sontuzumab, Stamulumab, Sulesomab, Tacatuzumab tetraxetan, Tadocizumab, Talizumab, Tanezumab, Taplitumomab paptox, Tefibazumab, Telimomab aritox, Tenatumomab, Teneliximab, Teplizumab, Ticilimumab (tremelimumab), Tigatuzumab, Tocilizumab (atlizumab), Toralizumab, Tositumomab, Trastuzumab, Tremelimumab, Tucotuzumab celmoleukin, Tuvirumab, Urtoxazumab, Ustekinumab, Vapaliximab, Vedolizumab, Veltuzumab, Vepalimomab, Visilizumab, Volociximab, Votumumab, Zalutumumab, Zanolimumab, Ziralimumab, and Zolimomab aritox. Monoclonal antibodies further include anti-TNF-α antibodies.
  • Examples of infusion therapy 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/Zalbin™), Rhucin® (Pharming Group, C1 inhibitor replacement therapy), tesamorelin (Theratechnologies/Egrifta, synthetic growth hormone-releasing factor), ocrelizumab (Genentech, Roche and Biogen), belimumab (GlaxoSmithKline/Benlysta®), pegloticase (Savient Pharmaceuticals/Krystexxa™), pegsiticase, taliglucerase alfa (Protalix/Uplyso), agalsidase alfa (Shire/Replagal®), velaglucerase alfa (Shire), and Keyhole Limpet Hemocyanin (KLH).
  • Additional therapeutic proteins include, for example, engineered proteins, such as Fc fusion proteins, bispecific antibodies, multi-specific antibodies, nanobodies, antigen-binding proteins, antibody fragments, and protein conjugates, such as antibody drug conjugates.
  • Therapeutic polynucleotides include, but are not limited to nucleic acid aptamers such as Pegaptanib (Macugen, a pegylated anti-VEGF aptamer), antisense therapeutics such as antisense poly- or oligonucleotides (e.g., antiviral drug Fomivirsen, or Mipomersen, an antisense therapeutic that targets the messenger RNA for apolipoprotein B for reduction of cholesterol level); small interfering RNAs (siRNAs) (e.g., dicer substrate siRNA molecules (DsiRNAs) which are 25-30 base pair asymmetric double-stranded RNAs that mediate RNAi with extremely high potency); or modified messenger RNAs (mmRNAs) such as those disclosed in US Patent application 2013/0115272 to de Fougerolles et al. and in Published US Patent application 2012/0251618 to Schrum et al.
  • Additional therapeutic macromolecules useful in accordance with aspects of this invention will be apparent to those of skill in the art, and the invention is not limited in this respect.
  • In some embodiments, a component, such as a therapeutic macromolecule or immunosuppressant may be isolated. Isolated refers to the element being separated from its native environment and present in sufficient quantities to permit its identification or use. This means, for example, the element may be (i) selectively produced by expression cloning or (ii) purified as by chromatography or electrophoresis. Isolated elements may be, but need not be, substantially pure. Because an isolated element may be admixed with a pharmaceutically acceptable excipient in a pharmaceutical preparation, the element may comprise only a small percentage by weight of the preparation. The element is nonetheless isolated in that it has been separated from the substances with which it may be associated in living systems, i.e., isolated from other lipids or proteins. Any of the elements provided herein may be isolated and included in the compositions or used in the methods in isolated form.
  • D. Methods of Making and Using the Compositions and Related Methods
  • Aspects of the invention relate to determining a protocol for the methods of concomitant administration as provided herein. A protocol can be determined by varying the frequency, dosage amount and other aspects of administration of the therapeutic macromolecule and the immunosuppressant and subsequently assessing the pharmacodynamic effect based on such variation. The varied administration occurs in the presence of an anti-therapeutic macromolecule antibody response. A preferred protocol for practice of the invention induces a desired pharmacodynamic effect but induces little to no anti-therapeutic macromolecule antibody response.
  • In some aspects of the invention, a desired pharmacodynamic effect of a therapeutic macromolecule includes stimulating or inhibiting a specific response. In some embodiments, the pharmacodynamic effect involves, without limitation, production or degradation of a cytokine, chemokine, signaling molecule or other molecule; inducing proliferation or death of a particular cell type; maturation or localization of a particular cell type; interaction with an enzyme, structural protein, carrier protein, or receptor protein; modulation of activity of an enzyme, structural protein or receptor protein, etc. In some embodiments, the pharmacodynamic effect is reducing production of a cytokine, for example an inflammation-associated cytokine such as TNF, IL-1. In some embodiments, the pharmacodynamic effect is reducing the activity of a cytokine. In some embodiments, the pharmacodynamic effect is reducing the production of an undesired molecule. In some embodiments, the pharmacodynamic effect is increasing degradation of an undesired molecule, for example uric acid crystals. In some embodiments, the pharmacodynamic effect is an activity of an enzyme.
  • A pharmacodynamic effect of a therapeutic macromolecule can be evaluated by standard methods. In some aspects of the invention, the pharmacodynamic effect is a reduction of inflammation. The level of inflammation can be assessed by any of the following exemplary methods without limitation, scoring of inflammatory symptoms such as redness or swelling; scoring of arthritic symptoms such as mobility, pain, or joint destruction; scoring of anaphylaxis symptoms such as swelling, blood pressure, shortness of breath; detecting and/or quantifying cell infiltration by histology, immunohistochemistry, flow cytometry; measuring the concentration of a protein or inflammation-associated cytokines such as TNF, IL-1 by ELISA, assessing the expression of gene or inflammation-associated genes by transcriptional analysis; measuring activity of an inflammation-associated cytokine, etc.
  • In some aspects of the invention, the pharmacodynamic effect is reduction or degradation of an undesired molecule. In some embodiments, the pharmacodynamic effect can be assessed, without limitation, by quantifying the undesired molecule in a tissue or blood sample by methods such as an ELISA. In some embodiments, the pharmacodynamic effect can be assessed by quantifying a molecule that is produced by the degradation of an undesired molecule such as by an ELISA. In some aspects of the invention, the pharmacodynamic effect is the activity of an enzyme that was not previously present or not adequately present. In such embodiments, the activity of an enzyme can be assessed by detecting the presence or the concentration of a product of the enzyme activity.
  • In some aspects of the invention, a reduced dose of the therapeutic macromolecule is administered to produce a pharmacodynamic effect. A reduced dose of a therapeutic macromolecule for such a purpose comprises any dose of the therapeutic macromolecule that achieves a pharmacodynamic effect in the presence of an anti-therapeutic macromolecule antibody response, with the concomitant administration of an immunosuppressant dose, that is less than the dose needed to achieve a similar pharmacodynamic effect with the therapeutic macromolecule when not administered concomitantly with the immunosuppressant dose in the presence of an anti-therapeutic macromolecule antibody response. A reduced dose can be determined by administering a therapeutic macromolecule, with the concomitant administration of an immunosuppressant dose, at a certain dose in the presence of an anti-therapeutic macromolecule antibody response and assessing the pharmacodynamic effect. The pharmacodynamic effect can then be compared with the pharmacodynamic effect that results through administration of the therapeutic macromolecule without the concomitant administration of an immunosuppressant dose in the presence of an anti-therapeutic macromolecule antibody response. A lower dose that achieves a similar pharmacodynamic effect as determined by such a comparison is a reduced dose.
  • As mentioned previously, immunosuppressants may be attached to synthetic nanocarriers. Synthetic nanocarriers may be prepared using a wide variety of methods known in the art. For example, 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. Alternatively or additionally, aqueous and organic solvent syntheses for monodisperse semiconductor, conductive, magnetic, organic, and other nanomaterials have been described (Pellegrino et al., 2005, Small, 1:48; Murray et al., 2000, Ann. Rev. Mat. Sci., 30:545; and Trindade et al., 2001, Chem. Mat., 13:3843). Additional methods have been described in the literature (see, e.g., Doubrow, Ed., “Microcapsules and Nanoparticles in Medicine and Pharmacy,” CRC Press, Boca Raton, 1992; Mathiowitz et al., 1987, J. Control. Release, 5:13; Mathiowitz et al., 1987, Reactive Polymers, 6:275; and Mathiowitz et al., 1988, J. Appl. Polymer Sci., 35:755; U.S. Pat. Nos. 5,578,325 and 6,007,845; P. Paolicelli et al., “Surface-modified PLGA-based Nanoparticles that can Efficiently Associate and Deliver Virus-like Particles” Nanomedicine. 5(6):843-853 (2010)).
  • Various materials may be encapsulated into synthetic nanocarriers as desirable using a variety of methods including but not limited to C. Astete et al., “Synthesis and characterization of PLGA nanoparticles” J. Biomater. Sci. Polymer Edn, Vol. 17, No. 3, pp. 247-289 (2006); K. Avgoustakis “Pegylated Poly(Lactide) and Poly(Lactide-Co-Glycolide) Nanoparticles: Preparation, Properties and Possible Applications in Drug Delivery” Current Drug Delivery 1:321-333 (2004); C. Reis et al., “Nanoencapsulation I. Methods for preparation of drug-loaded polymeric nanoparticles” Nanomedicine 2:8-21 (2006); P. Paolicelli et al., “Surface-modified PLGA-based Nanoparticles that can Efficiently Associate and Deliver Virus-like Particles” Nanomedicine. 5(6):843-853 (2010). Other methods suitable for encapsulating materials into synthetic nanocarriers may be used, including without limitation methods disclosed in U.S. Pat. No. 6,632,671 to Unger issued Oct. 14, 2003.
  • In certain embodiments, synthetic nanocarriers are prepared by a nanoprecipitation process or spray drying. Conditions used in preparing synthetic nanocarriers may be altered to yield particles of a desired size or property (e.g., hydrophobicity, hydrophilicity, external morphology, “stickiness,” shape, etc.). The method of preparing the synthetic nanocarriers and the conditions (e.g., solvent, temperature, concentration, air flow rate, etc.) used may depend on the materials to be attached to the synthetic nanocarriers and/or the composition of the polymer matrix.
  • If synthetic nanocarriers prepared by any of the above methods have a size range outside of the desired range, such synthetic nanocarriers can be sized, for example, using a sieve.
  • Elements (i.e., components) of the synthetic nanocarriers (such as antigens, immunosuppressants, and the like) 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.
  • Alternatively or additionally, synthetic nanocarriers can be attached to components directly or indirectly via non-covalent interactions. In non-covalent embodiments, 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 couplings may be arranged to be on an external surface or an internal surface of a synthetic nanocarrier. In embodiments, encapsulation and/or absorption is a form of attaching. In embodiments, the synthetic nanocarriers can be combined with a therapeutic macromolecule or other composition by admixing in the same vehicle or delivery system.
  • 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-phenoxyethanol, EDTA), polymeric stabilizers and viscosity-adjustment agents (e.g., polyvinylpyrrolidone, poloxamer 488, carboxymethylcellulose) and co-solvents (e.g., glycerol, polyethylene glycol, ethanol).
  • Compositions according to the invention 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 a sterile saline solution for injection together with a preservative.
  • It is to be understood that the compositions of the invention can be made in any suitable manner, and the invention is in no way limited to compositions that can be produced using the methods described herein. Selection of an appropriate method of manufacture may require attention to the properties of the particular moieties being associated.
  • In some embodiments, 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. In some embodiments, the compositions may be lyophilized and stored in suspension or as lyophilized powder depending on the formulation strategy for extended periods without losing activity.
  • Administration according to the present invention may be by a variety of routes, including but not limited to subcutaneous, intravenous, intraperitoneal, intramuscular, transmucosal, transdermal, transcutaneous or intradermal routes. In a preferred embodiment, administration is via a subcutaneous route of administration. The compositions referred to herein may be manufactured and prepared for administration, preferably concomitant administration, using conventional methods.
  • The compositions of the invention can be administered in effective amounts, such as the effective amounts described elsewhere herein. Doses of dosage forms may contain varying amounts of immunosuppressants and/or therapeutic macromolecules, according to the invention. The amount of immunosuppressants and/or therapeutic macromolecules present in the inventive dosage forms can be varied according to the nature of the therapeutic macromolecules, and/or immunosuppressant, the therapeutic benefit to be accomplished, and other such parameters. In embodiments, dose ranging studies can be conducted to establish optimal therapeutic amounts of immunosuppressant and/or therapeutic macromolecules to be present in the dosage form. In embodiments, the immunosuppressants and/or therapeutic macromolecules are present in the dosage form in an amount effective to generate a desired pharmacodynamic effect and/or a reduced immune response to the therapeutic macromolecules upon administration to a subject. It may be possible to determine amounts of the immunosuppressants and/or therapeutic macromolecules effective to achieve a desired result using conventional dose ranging studies and techniques in subjects. Inventive dosage forms may be administered at a variety of frequencies. In a preferred embodiment, at least one administration of the compositions provided herein is sufficient to generate a pharmacologically relevant response. In more preferred embodiments, at least two administrations or at least three administrations are utilized to ensure a pharmacologically relevant response. In some embodiments, repeated administrations are utilized to ensure a pharmacologically relevant response.
  • Another aspect of the disclosure relates to kits. In some embodiments, the kit comprises a pharmacodynamically effective dose or more than one dose of a therapeutic macromolecule, such as a reduced pharmacodynamically effective dose. In such embodiments, the kit may also comprise an immunosuppressant dose or more than one dose of immunosuppressant. The immunosuppressant dose and the pharmacodynamically effective dose can be contained within separate containers or within the same container in the kit. In some embodiments, the container is a vial or an ampoule. In some embodiments, the pharmacodynamically effective dose of a therapeutic macromolecule and/or immunosuppressant dose are contained within a solution separate from the container, such that the pharmacodynamically effective dose of a therapeutic macromolecule and/or immunosuppressant dose may be added to the container at a subsequent time. In some embodiments, the pharmacodynamically effective dose of a therapeutic macromolecule and/or immunosuppressant dose are in lyophilized form each in a separate container or in the same container, such that they may be reconstituted at a subsequent time. In some embodiments, the kit further comprises instructions for reconstitution, mixing, administration, etc. In some embodiments, the instructions include a description of the methods described herein. Instructions can be in any suitable form, e.g., as a printed insert or a label. In some embodiments, the kit further comprises one or more syringes.
  • EXAMPLES Example 1: Evaluating Immune Responses with Synthetic Nanocarriers Comprising Immunosuppressant and APC Presentable Antigen In Vivo Materials
  • Ovalbumin peptide 323-339 (OVA323-339), a 17 amino acid peptide known to be a T and B cell epitope of Ovalbumin protein, was purchased from Bachem Americas Inc. (3132 Kashiwa Street, Torrance Calif. 90505; Part #4065609). Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Catalogue # R1017). PLGA with a lactide:glycolide ratio of 3:1 and an inherent viscosity of 0.75 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 7525 DLG 7A). Polyvinyl alcohol (85-89% hydrolyzed) was purchased from EMD Chemicals (Product Number 1.41350.1001).
  • Method for Preparing Synthetic Nanocarrier Containing Rapamycin and Ovalbumin (323-339)
  • Solutions were prepared as follows:
  • Solution 1: OVA323-339 at 20 mg/mL in dilute hydrochloric acid aqueous solution. The solution was prepared by dissolving ovalbumin peptide in 0.13 M hydrochloric acid solution at room temperature.
  • Solution 2: PLGA at 100 mg/mL in methylene chloride. The solution was prepared by dissolving PLGA in pure methylene chloride.
  • Solution 3: PLA-PEG at 100 mg/mL in methylene chloride. The solution was prepared by dissolving PLA-PEG in pure methylene chloride.
  • Solution 4: Rapamycin at 50 mg/mL in methylene chloride. The solution was prepared by dissolving rapamycin in pure methylene chloride.
  • Solution 5: Polyvinyl alcohol at 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • A primary water-in-oil emulsion was prepared first. W1/O1 was prepared by combining solution 1 (0.2 mL), solution 2 (0.75 mL), solution 3 (0.25 mL), and solution 4 (0.2 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) was then prepared by combining solution 5 (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 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 synthetic nanocarriers to form. A portion of the synthetic nanocarriers were washed by transferring the synthetic 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 was repeated, and the pellet was re-suspended in phosphate buffered saline for a final synthetic nanocarrier dispersion of about 10 mg/mL.
  • The amounts of peptide and rapamycin in the synthetic nanocarriers were determined by HPLC analysis. The total dry-synthetic nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • Method for Synthetic Nanocarrier Containing Rapamycin
  • A primary water-in-oil emulsion was prepared first. W1/O1 was prepared by combining 0.13 M hydrochloric acid solution (0.2 mL), solution 2 (0.75 mL), solution 3 (0.25 mL), and solution 4 (0.2 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) was then prepared by combining solution 5 (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 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 synthetic nanocarriers to form. A portion of the synthetic nanocarriers were washed by transferring the synthetic nanocarrier suspension to a centrifuge tube and centrifuging at 21,000×g and 4° C. for one hour, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. The washing procedure was repeated, and the pellet was re-suspended in phosphate buffered saline for a final synthetic nanocarrier dispersion of about 10 mg/mL.
  • The amount of rapamycin in the synthetic nanocarrier was determined by HPLC analysis. The total dry-synthetic nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • Method for Measuring Rapamycin Load
  • Approximately 3 mg of synthetic nanocarriers were collected and centrifuged to separate supernatant from synthetic nanocarrier pellet. Acetonitrile was added to the pellet, and the sample was sonicated and centrifuged to remove any insoluble material. The supernatant and pellet were injected on RP-HPLC and absorbance was read at 278 nm. The g found in the pellet were used to calculate % entrapped (load), μg in supernatant and pellet were used to calculate total g recovered.
  • Method for Measuring Ovalbumin (323-339) Load Approximately 3 mg of synthetic nanocarriers were collected and centrifuged to separate supernatant from synthetic nanocarrier pellet. Trifluoroethanol was added to the pellet and the sample was sonicated to dissolve the polymer, 0.2% trifluoroacetic acid was added and sample was sonicated and then centrifuged to remove any insoluble material. The supernatant and pellet were injected on RP-HPLC and absorbance was read at 215 nm. The g found in the pellet were used to calculate % entrapped (load), μg in supernatant and pellet were used to calculate total g recovered.
  • Immunization
  • The purpose of this experiment was to assess the effects of encapsulated immunosuppressant on ongoing antibody responses by measuring antigen-specific immunogloblulins. One group of animals remained unimmunized as a control. Two groups of animals were immunized using Chicken Ovalbumin (OVA) and CpG with 3 injections (d0, d14 and d28) in the footpad followed by an assessment of antibody titers. For immunization, animals received 20 μl/limb of OVA+CpG (12.5 μg OVA+10 μg CpG), both hind limbs s.c. The treatment, administered on the same days, included administration i.v. (200 μl) or s.c. (20 μl). Nanocarriers were diluted in such a manner that the same amounts of OVA323-339 were injected in the treated groups.
  • Measurement of IgG
  • The level of IgG antibodies were measured. Blocker Casein in PBS (Thermo Fisher, Catalog #37528) was used as diluent. 0.05% Tween-20 in PBS was used as wash buffer, prepared by adding 10 ml of Tween-20 ((Sigma, Catalog # P9416-100 mL) to 2 liters of a O1×PBS stock (PBS: OmniPur® 10×PBS Liquid Concentrate, 4 L, EMD Chemicals, Catalog #6505) and 18 Liters of deionized water.
  • OVA protein at a stock concentration of 5 mg/ml was used as a coating material. A 1:1000 dilution to 5 μg/ml was used as a working concentration. Each well of the assay plates was coated with 100 μl diluted OVA per well, plates were sealed with sealing film (VWR catalog #60941-120), and incubated overnight at 4° C. Costar 9017 96-well Flat bottom plates were used as assay plates (Costar 9017).
  • Low-binding polypropylene 96-well plate or tubes were used as set-up plates, in which samples were prepared before being transferred to the assay plate. The setup plates did not contain any antigen and, therefore, serum antibodies did not bind to the plate during the setup of the samples. Setup plates were used for sample preparation to minimize binding that might occur during preparation or pipetting of samples if an antigen-coated plate was used to prepare the samples. Before preparing samples in the setup plate, wells were covered with diluent to block any non-specific binding and the plate was sealed and incubated at 4° C. overnight.
  • Assay plates were washed three times with wash buffer, and wash buffer was completely aspirated out of the wells after the last wash. After washing, 300 μl diluent were added to each well of assay plate(s) to block non-specific binding and plates were incubated at least 2 hours at room temperature. Serum samples were prepared in the setup plate at appropriate starting dilutions. Starting dilutions were sometimes also prepared in 1.5 ml tubes using diluent and then transferred to the set-up plate. Appropriate starting dilutions were determined based on previous data, where available. Where no previous data was available, the lowest starting dilution was 1:40. Once diluted, 200 μl of the starting dilution of the serum sample was transferred from the tube to the appropriate well of the setup plate.
  • An exemplary setup plate layout is described as follows: Columns 2 and 3 contained anti-Ovalbumin monoclonal IgG2b isotype (AbCam, ab17291) standard, diluted to 0.25 μg/mL (1:4000 dilution). Columns 3-11 contained serum samples (at appropriate dilutions). Columns 1 and 12 were not used for samples or standards to avoid any bias of measurements due to edge effect. Instead, columns 1 and 12 contained 200 μl diluent. Normal mouse serum diluted 1:40 was used as a negative control. Anti-mouse IgG2a diluted 1:500 from 0.5 mg/mL stock (BD Bioscience) was used as an isotype control.
  • Once all samples were prepared in the setup plate, the plate was sealed and stored at 4° C. until blocking of the assay plates was complete. Assay plates were washed three times with wash buffer, and wash buffer was completely aspirated after the last wash. After washing, 100 μL of diluent was added to all wells in rows B-H of the assay plates. A 12-channel pipet was used to transfer samples from the setup plate to the assay plate. Samples were mixed prior to transfer by pipetting 150 μl of diluted serum up and down 3 times. After mixing, 150 μl of each sample was transferred from the setup plate and added to row A of the respective assay plate.
  • Once the starting dilutions of each sample were transferred from the setup plate to row A of the assay plate, serial dilutions were pipetted on the assay plate as follows: 50 μl of each serum sample was removed from row A using 12-channel pipet and mixed with the 100 μl of diluent previously added to each well of row B. This step was repeated down the entire plate. After pipetting the dilution of the final row, 50 μl of fluid was removed from the wells in the final row and discarded, resulting in a final volume of 100 μl in every well of the assay plate. Once sample dilutions were prepared in the assay plates, the plates were incubated at room temperature for at least 2 hours.
  • After the incubation, plates were washed three times with wash buffer. Detection antibody (Goat anti-mouse anti-IgG, HRP conjugated, AbCam ab98717) was diluted 1:1500 (0.33 μg/mL) in diluent and 100 μl of the diluted antibody was added to each well. Plates were incubated for 1 hour at room temperature and then washed three times with wash buffer, with each washing step including a soak time of at least 30 seconds.
  • After washing, detection substrate was added to the wells. Equal parts of substrate A and substrate B (BD Biosciences TMB Substrate Reagent Set, catalog #555214) were combined immediately before addition to the assay plates, and 100 μl of the mixed substrate solution were added to each well and incubated for 10 minutes in the dark. The reaction was stopped by adding 50 μl of stop solution (2N H2SO4) to each well after the 10 minute period. The optical density (OD) of the wells was assessed immediately after adding the stop solution on a plate reader at 450 nm with subtraction at 570 nm. Data analysis was performed using Molecular Device's software SoftMax Pro v5.4. A four-parameter logistic curve-fit graph was prepared with the dilution on the x-axis (log scale) and the OD value on the y-axis (linear scale), and the half maximum value (EC50) for each sample was determined. The plate template at the top of the layout was adjusted to reflect the dilution of each sample (1 per column).
  • Results
  • The results demonstrate the ability to reduce an antigen-specific antibody response using an immunosuppressant dose in combination with the antigen, both the immunosuppressant and antigen being attached to the nanocarriers. FIG. 1 shows a decrease in circulating antigen-specific antibody production with nanocarriers comprising peptide antigen and immunosuppressant when administered intravenously. Two independent experiments were performed using 5 animals in each experiment. FIG. 2 shows a decrease in circulating antigen-specific antibody production with nanocarriers comprising peptide antigen and immunosuppressant when administered subcutaneously. Three independent experiments were performed using 5 animals in each experiment. P values were calculated using a Bonferroni post-test of a regular one-way ANOVA test (*=p<0.05, **=p<0.01 and ***=p<0.001).
  • These results demonstrate that an enhanced effect can be obtained using the same amount of ovbalbumin protein, when administered under conditions that would ordinarily produce a significant anti-ovalbumin antibody response. Furthermore, this pharmacodynamic effect could have been obtained with a reduced pharmacodynamically effective dose, as the results suggest. Finally, the reproduction of the results supports that a protocol was established that was demonstrated to result in a pharmacodynamic effect with the reduced pharmacodynamically effective dose of the therapeutic macromolecule upon concomitant administration with the immunosuppressant dose.
  • Example 2: Evaluating Immune Responses Following Concomitant Administration of Synthetic Nanocarriers Comprising Immunosuppressant and Therapeutic Proteins Materials
  • 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. Product Code 7525 DLG 7A.) 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).
  • Method for Preparing Synthetic Nanocarriers
  • Solutions were prepared as follows:
  • 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.
  • An oil-in-water emulsion was used to prepare the nanocarriers. 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. for 35 min, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. The washing procedure was repeated, and the pellet was re-suspended in phosphate buffered saline for a final nanocarrier dispersion of about 10 mg/mL.
  • 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.
  • Effective Diameter Rapamycin Content
    (nm) (% w/w)
    227 6.4
  • Responses to Ovalbumin
  • C57BL/6 age-matched (5-6 weeks) female mice (5 per group, 3 groups: naïve control, untreated control, and treatment with synthetic nanocarriers with rapamycin were injected intravenously with 25 μg of chicken ovalbumin induced using high sheer to form aggregates in order to increase its immunogenicity (aggOVA). After these i.v. injections to prime and boost the immune response (administrations on day 0, 14 and 28), subsequent boosts of 25 μg ovalbumin were performed intraperitoneally (i.p. on days 42 and 57), and subcutaneously a 12.5 μg pOVA challenge in the left hind limb (s.c. d62) followed by challenges using the same aggOVA combined with CpG (s.c. day 90, 105 and 135). FIG. 3 shows the results of such immunization protocol. At days 25 and 40, a prominent anti-OVA antibody (Ab) response can be detected in these animals using ELISA (technique generally as described in Example 1). Subsequent injections of aggOVA were observed to maintain these titers or even boost the response. In contrast, when mice were treated using the synthetic nanocarriers administered i.v. (dose calculated to provide 100 μg of rapamycin) concomitantly with aggOVA during only the three first encounters with the antigen on d0, 14 and 28, no IgG response can be detected for the next 60 days even after 4 injections with the immunogenic mixture including aggOVA. See FIG. 3. It is only after the 3 s.c. injections (s.c. day 90, 105 and 135) with aggOVA/KLH and CpG, an extremely immunogenic combination, that a very modest anti-aggOVA IgG response can be detected in treated animals.
  • Responses to Keyhole Limpet Hemocyanin, a Second Antigen
  • The same mice as above were also injected with 0.05 μg of a second Ag, Keyhole Limpet Hemocyanin (KLH), with the KLH being mixed with the aggOVA, and administered according to the schedule noted above for the aggOVA. Anti-KLH IgG antibody titer was determined using a method similar to the anti-OVA IgG titer method described in Example 1.
  • The results in FIG. 4 show that, unlike aggOVA, KLH is not immunogenic in the first phase of encounters with the Ag (d0-40). However, KLH was immunogenic in animals that were not treated with the synthetic nanocarriers after 3 immunizations of KLH in presence of CpG s.c. (s.c. day 90, 105 and 135). A strong anti-KLH response can be observed in these control mice, whereas the mice that received the 3 treatments with synthetic nanocarriers with immunosuppressant in combination with the antigens at the beginning of this protocol have no detectable response even after the KLH/CpG challenges.
  • These results demonstrate that an enhanced effect can be obtained with the compositions and methods provided herein when administered under conditions that would ordinarily produce a significant antibody response. Also, the results support that a protocol was established that was demonstrated to result in a pharmacodynamic effect.
  • Example 3: Method for Preparing Nanocarriers Attached to Rapamycin Materials
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Code R1017). PLGA with a lactide:glycolide ratio of 3:1 and an inherent viscosity of 0.75 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 7525 DLG 7A). PLA-PEG-OMe block co-polymer with a methyl ether terminated PEG block of approximately 5,000 Da and an overall inherent viscosity of 0.5 DL/g was purchased from Lakeshore Biochemicals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 100 DL mPEG 5000 5CE). EMPROVE® Polyvinyl Alcohol 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).
  • Method
  • Solutions were prepared as follows:
  • Solution 1: PLGA at 75 mg/mL, PLA-PEG-OMe at 25 mg/mL, and rapamycin at 12.5 mg/mL in methylene chloride. The solution was prepared by dissolving PLGA, PLA-PEG-OMe, and rapamycin in pure methylene chloride.
  • Solution 2: Polyvinyl alcohol @ 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • An oil-in-water (O/W) emulsion was used to prepare the nanocarriers. The O/W emulsion was prepared by combining Solution 1 (1.0 mL) and Solution 2 (3.0 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 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 75,600×g and 4° C. for 50 min, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. The washing procedure was repeated, and the pellet was re-suspended in phosphate buffered saline for a final nanocarrier dispersion of about 10 mg/mL.
  • Nanocarrier size was determined by dynamic light scattering and presented in Table 1. The amount of rapamycin in the nanocarrier was determined by HPLC analysis (Table 1). The total dry-nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • Effective diameter and rapamycin content of nanocarriers attached to rapamycin
  • Effective Diameter Rapamycin Content
    Nanocarrier ID (nm) (% w/w)
    1 238 10.6
    2 241 11.1
    3 241 11.5
  • Materials
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Code R1017). PLGA with a lactide:glycolide ratio of 1:1 and an inherent viscosity of 0.24 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 5050 DLG 2.5A). PLA-PEG-OMe block co-polymer with a methyl ether terminated PEG block of approximately 5,000 Da and an overall inherent viscosity of 0.5 DL/g was purchased from Lakeshore Biochemicals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 100 DL mPEG 5000 5CE). EMPROVE® Polyvinyl Alcohol 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).
  • Method
  • Solutions were prepared as follows:
  • Solution 1: PLGA at 75 mg/mL, PLA-PEG-OMe at 25 mg/mL, and rapamycin at 12.5 mg/mL in methylene chloride. The solution was prepared by dissolving PLGA, PLA-PEG-OMe, and rapamycin in pure methylene chloride.
  • Solution 2: Polyvinyl alcohol at 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • Solution 3: 70 mM phosphate buffer, pH 8.
  • A an oil-in-water emulsion was created by mixing Solutions 1 (1.0 mL) and Solution 2 (3.0 mL) in a small glass pressure tube and sonicating at 30% amplitude for 60 seconds using a Branson Digital Sonifier 250. The emulsion was added to an open 50 mL beaker containing Solution 3 (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended nanocarriers was then washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 75,600 rcf for 40 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated and then the pellet was re-suspended in PBS 1× to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at −20° C. until use.
  • Nanocarrier 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.
  • Effective diameter and rapamycin content of nanocarriers attached to rapamycin
  • Nanocarrier Effective Rapamycin Load
    ID Diameter (nm) (% w/w)
    4 218 9.9
  • Materials
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Code R1017). PLGA with a lactide:glycolide ratio of 1:1 and an inherent viscosity of 0.24 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 5050 DLG 2.5A). PLA-PEG-OMe block co-polymer with a methyl ether terminated PEG block of approximately 5,000 Da and an overall inherent viscosity of 0.5 DL/g was purchased from Lakeshore Biochemicals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 100 DL mPEG 5000 5CE). F8II.1723 peptide was purchased from AnaSpec (34801 Campus Drive, Fremont, Calif. 94555). EMPROVE® Polyvinyl Alcohol 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).
  • Method
  • Solutions were prepared as follows:
  • Solution 1: PLGA at 75 mg/mL, PLA-PEG-OMe at 25 mg/mL, and rapamycin at 12.5 mg/mL in methylene chloride. The solution was prepared by dissolving PLGA, PLA-PEG-OMe, and rapamycin in pure methylene chloride.
  • Solution 2: F8II.1723 (ERLWDYGMSSSPHVL) at 10 mg/mL and sucrose at 100 mg/mL in 50 mM pH 11.5 phosphate buffer. The solution was prepared by dissolving the sucrose in 50 mM pH 11.5 phosphate buffer and then adding the F8II.1723 peptide as a dry powder.
  • Solution 3: Polyvinyl alcohol at 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • Solution 4: 70 mM pH 8 phosphate buffer.
  • A primary (W1/O) emulsion was first created by mixing Solution 1 (1.0 mL) and Solution 2 (0.2 mL) in a small glass pressure tube and sonicating at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • A secondary (W1/O/W2) emulsion was then formed by adding Solution 3 (3.0 mL) to the primary emulsion, vortexing to create a crude dispersion, and then sonicating at 30% amplitude for 60 seconds using the Branson Digital Sonifier 250.
  • The secondary emulsion was added to an open 50 mL beaker containing Solution 4 (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended nanocarriers was then washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 75,600 rcf for 35 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated and then the pellet was re-suspended in PBS 1× to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at −20° C. until use.
  • Nanocarrier size was determined by dynamic light scattering. The amount rapamycin in the nanocarrier was determined by HPLC analysis. The amount of F8II.1723 in the nanocarrier was determined using a fluorescamine-based assay. The total dry-nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • Effective diameter and rapamycin content of nanocarriers attached to rapamycin
  • Nanocarrier Effective Rapamycin Content Peptide content
    ID Diameter (nm) (% w/w) (% w/w)
    5 202 8.9 1.1
  • Materials
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Code R1017). PLGA with a lactide:glycolide ratio of 1:1 and an inherent viscosity of 0.24 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 5050 DLG 2.5A). PLA-PEG-OMe block co-polymer with a methyl ether terminated PEG block of approximately 5,000 Da and an overall inherent viscosity of 0.5 DL/g was purchased from Lakeshore Biochemicals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 100 DL mPEG 5000 5CE). F8II.75 peptide was purchased from AnaSpec (34801 Campus Drive, Fremont, Calif. 94555). EMPROVE® Polyvinyl Alcohol 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).
  • Method
  • Solutions were prepared as follows:
  • Solution 1: PLGA at 75 mg/mL, PLA-PEG-OMe at 25 mg/mL, and rapamycin at 12.5 mg/mL in methylene chloride. The solution was prepared by dissolving PLGA, PLA-PEG-OMe, and rapamycin in pure methylene chloride.
  • Solution 2: F8II.75 (VHLFNIAKPRPPWMG) at 10 mg/mL and sucrose at 100 mg/mL in 50 mM pH 2 phosphate buffer. The solution was prepared by dissolving the sucrose in 50 mM pH 2 phosphate buffer and then adding the F8II.75 peptide as a dry powder.
  • Solution 3: Polyvinyl alcohol at 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • Solution 4: 70 mM pH 8 phosphate buffer.
  • A primary (W1/O) emulsion was first created by mixing Solution 1 (1.0 mL) and Solution 2 (0.2 mL) in a small glass pressure tube and sonicating at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • A secondary (W1/O/W2) emulsion was then formed by adding Solution 3 (3.0 mL) to the primary emulsion, vortexing to create a crude dispersion, and then sonicating at 30% amplitude for 60 seconds using the Branson Digital Sonifier 250.
  • The secondary emulsion was added to an open 50 mL beaker containing Solution 4 (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended nanocarriers was then washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 75,600 rcf for 35 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated and then the pellet was re-suspended in PBS 1× to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at −20° C. until use.
  • Nanocarrier size was determined by dynamic light scattering. The amount rapamycin in the nanocarrier was determined by HPLC analysis. The amount of F8II.75 in the nanocarrier was determined using a fluorescamine-based assay. The total dry-nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • Effective diameter and rapamycin content of nanocarriers attached to rapamycin
  • Nanocarrier Effective Rapamycin Content Peptide content
    ID Diameter (nm) (% w/w) (% w/w)
    6 199 9.3 1.1
  • Materials
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Code R1017). PLGA with a lactide:glycolide ratio of 1:1 and an inherent viscosity of 0.24 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 5050 DLG 2.5A). PLA-PEG-OMe block co-polymer with a methyl ether terminated PEG block of approximately 5,000 Da and an overall inherent viscosity of 0.5 DL/g was purchased from Lakeshore Biochemicals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 100 DL mPEG 5000 5CE). F8II.2210 peptide was purchased from AnaSpec (34801 Campus Drive, Fremont, Calif. 94555). EMPROVE® Polyvinyl Alcohol 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).
  • Method
  • Solutions were prepared as follows:
  • Solution 1: PLGA at 75 mg/mL, PLA-PEG-OMe at 25 mg/mL, and rapamycin at 12.5 mg/mL in methylene chloride. The solution was prepared by dissolving PLGA, PLA-PEG-OMe, and rapamycin in pure methylene chloride.
  • Solution 2: F8II.2210 (TASSYFTNMFATWSPSKAR) at 10 mg/mL and sucrose at 100 mg/mL in 50 mM pH 2 phosphate buffer. The solution was prepared by dissolving the sucrose in 50 mM pH 2 phosphate buffer and then adding the F8II.2210 peptide as a dry powder.
  • Solution 3: Polyvinyl alcohol at 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • Solution 4: 70 mM pH 8 phosphate buffer.
  • A primary (W1/O) emulsion was first created by mixing Solution 1 (1.0 mL) and Solution 2 (0.2 mL) in a small glass pressure tube and sonicating at 50% amplitude for 40 seconds using a Branson Digital Sonifier 250.
  • A secondary (W1/O/W2) emulsion was then formed by adding Solution 3 (3.0 mL) to the primary emulsion, vortexing to create a crude dispersion, and then sonicating at 30% amplitude for 60 seconds using the Branson Digital Sonifier 250.
  • The secondary emulsion was added to an open 50 mL beaker containing Solution 4 (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended nanocarriers was then washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 75,600 rcf for 35 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated and then the pellet was re-suspended in PBS 1× to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at −20° C. until use.
  • Nanocarrier size was determined by dynamic light scattering. The amount rapamycin in the nanocarrier was determined by HPLC analysis. The amount of F8II.2210 in the nanocarrier was determined using a fluorescamine-based assay. The total dry-nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • Effective diameter and rapamycin content of nanocarriers attached to rapamycin
  • Nanocarrier Effective Rapamycin Content Peptide content
    ID Diameter (nm) (% w/w) (% w/w)
    7 215 9.3 1.6
  • Materials
  • PLGA with a lactide:glycolide ratio of 1:1 and an inherent viscosity of 0.24 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 5050 DLG 2.5A). PLA-PEG-OMe block co-polymer with a methyl ether terminated PEG block of approximately 5,000 Da and an overall inherent viscosity of 0.5 DL/g was purchased from Lakeshore Biochemicals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 100 DL mPEG 5000 5CE). EMPROVE® Polyvinyl Alcohol 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).
  • Method
  • Solutions were prepared as follows:
  • Solution 1: PLGA at 75 mg/mL and PLA-PEG-OMe at 25 mg/mL in methylene chloride. The solution was prepared by dissolving PLGA and PLA-PEG-OMe in pure methylene chloride.
  • Solution 2: Polyvinyl alcohol at 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • Solution 3: 70 mM phosphate buffer, pH 8.
  • A an oil-in-water emulsion was created by mixing Solutions 1 (1.0 mL) and Solution 2 (3.0 mL) in a small glass pressure tube and sonicating at 30% amplitude for 60 seconds using a Branson Digital Sonifier 250. The emulsion was added to an open 50 mL beaker containing Solution 3 (30 mL) and stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended nanocarriers was then washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 75,600 rcf for 40 minutes, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. This washing procedure was repeated and then the pellet was re-suspended in PBS 1× to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at −20° C. until use.
  • Nanocarrier size was determined by dynamic light scattering. The total dry-nanocarrier mass per mL of suspension was determined by a gravimetric method.
  • Nanocarrier ID Effective Diameter (nm)
    8 183
  • Example 4: Evaluating Immune Responses to FVIII
  • Hemophilia A mice (E16 mice) (n=8 at day 0) received weekly concomitant intravenous (i.v.) injections of the blood clotting protein, Factor VIII (FVIII), and the synthetic nanocarriers attached to rapamycin (Nanocarrier ID 4), or empty nanoparticles (NP) (Nanocarrier ID 8) and FVIII or IVIG (intravenous immunoglobulin) and FVIII for 5 consecutive weeks. To evaluate the antibody recall response to FVIII one month after the final dose of FVIII and nanocarrier attached to rapamycin, the inhibitor titer was determined by Bethesda assay using a chromogenic FVIII activity assay kit (Coatest SP4 FVIII), as presented in FIG. 5.
  • To further assess the lasting tolerance induced by treatment of FVIII and nanocarriers attached to rapamycin, Hemophilia A mice (E16 mice) (n=8 at day 0) received weekly concomitant i.v. injections of synthetic nanocarriers and FVIII peptides ( Nanocarrier IDs 5, 6 and 7 mixed to get a 1:1:1 peptide mass ratio and injected together), or nanocarriers attached to rapamycin (Nanocarrier ID 4) and FVIII protein, or empty NP (Nanocarrier ID 8) and FVIII, or IVIG (intravenous immunoglobulin) and FVIII for 5 weeks, as schematically presented in FIG. 6A. At days 57, 81, and 125, the mice were challenged with FVIII (i.v. or i.p.) in the absence of further treatment. Anti-FVIII antibody levels were determined by ELISA. Results at selected time points are presented in FIG. 6B. Data at each time point are expressed as mean±SEM. For statistical analysis, the nanocarrier groups (with protein or peptides) were compared with the empty nanoparticle control group using the student t-test with two tailed distribution. *p<0.05 and **p<0.01, between Nanocarrier ID 4 with protein and empty NP groups; # p<0.05 and ## p<0.01, between the nanocarrier and peptide ( Nanocarrier IDs 5, 6 and 7) and empty NP groups. All injections of FVIII were 1 μg per injection. All injections of Nanocarrier ID 4 were 100 μg per injection.
  • The data show that the administration of immunosuppressant attached to synthetic nanocarriers with FVIII or peptides thereof can reduce the anti-Factor FVIII antibody response. This was true using the immunosuppressant synthetic nanocarriers in combination with unattached Factor FVIII protein as well as Factor FVIII peptides also attached to the synthetic nanocarriers. The data also show that such administration can result in improved activity of Factor VIII. This demonstrates the reduction of an undesired immune response against a protein as well as improved efficacy with concomitant administration of immunosuppressant attached to synthetic nanocarriers. Finally, the results support that a protocol was established as provided herein.
  • Example 5: Evaluating Responses to HUMIRA/Adalimumab
  • Control C57BL/6 age-matched (5-6 weeks) females were injected subcutaneously (s.c.) in the front limbs with 60 μg of HUMIRA once a week until day 29 (on d0, 7, 14, 22, 29, across all groups and conditions). Another group received similar injections, but 0.9 mg of nanocarrier attached to rapamycin ( Nanocarrier ID 1, 2 or 3) were admixed to the solution of HUMIRA on the priming day 0. The results presented in FIG. 7A show the antibody titers in the blood of all animals at day 21. The control animals develop a robust antibody response against HUMIRA while treated animals remain completely negative even after 20 days and two injections of HUMIRA without treatment. At day 29, the animals received another challenge in one hind limb while the other hind limb received saline in order to test the local antibody-mediated type I hypersensitivity response. For this, the thickness of the hind limbs were measured with the help of a caliper one hour after the injection. The difference in thickness between the two limbs is presented in FIG. 7B. Similar to the antibody results, the treatment with the nanocarriers abolished the inflammatory response induced by local administration of HUMIRA.
  • These results show that administration of immunosuppressant attached to synthetic nanocarriers with HUMIRA can reduce undesired anti-HUMIRA antibody response as well as eliminate undesired inflammatory reactions that can result from the administration of HUMIRA without the immunosuppressant nanocarrier composition. This demonstrates the reduction of undesired immune responses against a protein with concomitant administration of immunosuppressant attached to synthetic nanocarriers. Finally, the results support that a protocol was established as provided herein.
  • Example 6: Evaluating Responses to Keyhole Limpet Hemocyanin (KLH)
  • Control C57BL/6 age-matched (5-6 weeks) females were injected intravenously (i.v.) in the tail vain with 200 μg of Keyhole Limpet Hemocyanin (KLH) starting at day 0 or day 21 once a week until 34 (on d0, 7, 14, 21, 28, 34). Another group received similar injections from day 0 to day 34, but 0.47 mg of nanocarrier attached to rapamycin ( Nanocarrier ID 1, 2 or 3) were admixed to the solution of KLH on days 0, 14 and 21 followed by KLH alone injections weekly between days 21 and 34 (on d21, 28, 34). The results presented in FIG. 8 show the antibody titers in the blood of all animals at the indicated time points. The control animals that started immunization at day 0 or at day 21 develop very similar responses (EC50=3-5×103) after three injections using the same amounts of KLH (day 26 and 40, respectively). In contrast, animals that received three injections of the nanocarriers remain completely negative after three injections of KLH alone.
  • The data show that the administration of immunosuppressant attached to synthetic nanocarriers with KLH can reduce the anti-KLH antibody response, and such a reduction can remain even after being administered KLH subsequent to the nanocarrier treatment. This demonstrates the reduction of an undesired immune response against a protein with concomitant administration of immunosuppressant attached to synthetic nanocarriers. Finally, the results support that a protocol was established as provided herein.
  • Example 7: Evaluating Responses to Ovalbumin
  • C57BL/6 age-matched (5-6 weeks) female were injected i.v. in the tail vein on days −21 and −13 with 1.2 mg of unattached nanoparticles (NP[Empty]) (Nanocarrier ID 8), free rapamycin (fRapa) (100 μg), 22 μg of free chicken Ovalbumin (fOVA), a combination of fRapa and fOVA, 1.2 mg of nanocarrier attached to rapamycin (NP[Rapa], 100 μg of rapamycin content) ( Nanocarrier ID 1, 2 or 3) alone or in combination with 22 μg of fOVA. At day 0 all animals were injected s.c. in the hind limbs with 2.5 μg of particulate OVA (pOVA) admixed to 2 μg of CpG followed by injections of 2.5 μg pOVA on days 7 and 14. In the absence of any treatment (NP[Empty]), the animals develop a robust immune response toward OVA that can be measured by the anti-OVA IgG antibody titers. The antibody titers at day 22 shown in FIG. 9 demonstrate that 2 doses of synthetic nanocarriers administered concomitantly with OVA in the same solution (fOVA+NP[Rapa]) were effective in preventing antibody formation to OVA even after 1 injection of OVA+CpG and 2 injections of OVA alone.
  • The data show that the administration of immunosuppressant attached to synthetic nanocarriers with OVA can reduce the anti-OVA antibody response, and such a reduction can remain even after being administered OVA in combination with a strong adjuvant. This demonstrates the reduction of an undesired immune response against a protein with concomitant administration of immunosuppressant attached to synthetic nanocarriers. Finally, the results support that a protocol was established as provided herein.
  • Example 8: Evaluating Responses to KRYSTEXXA
  • A control group of C57BL/6 age-matched (5-6 weeks) female were injected i.v. in the tail vain with PBS while the treated group was injected with 0.9 mg of nanocarriers attached to rapamycin ( Nanocarrier ID 1, 2 or 3) in combination with 40 μg of KRYSTEXXA on days −21 and −14. All animals received weekly injections from day 0 to day 28 of 100 μg of KRYSTEXXA combined to 20 μg of CpG s.c. in the hind limb (d0, 7, 12, 28). The control animals develop an immune response against KRYSTEXXA that can be measured by the anti-KRYSTEXXA IgM antibody titers. The results in FIG. 10 show that treating animals with synthetic nanocarriers administered concomitantly with KRYSTEXXA in the same solution were effective in preventing antibody formation to KRYSTEXXA for a prolonged period of time. The treated animals did not develop an anti-KRYSTEXXA response even after five injections of KRYSTEXXA+CpG without the nanocarriers.
  • The data show that the administration of immunosuppressant attached to synthetic nanocarriers with KRYSTEXXA can reduce the anti-KRYSTEXXA antibody response, and such a reduction can remain even after being administered KRYSTEXXA subsequent to the nanocarrier treatment with a strong adjuvant. This demonstrates the reduction of an undesired immune response against a protein with concomitant administration of immunosuppressant attached to synthetic nanocarriers. Finally, the results support that a protocol was established as provided herein.
  • Example 9: Evaluating Responses to Ovalbumin and KLH
  • The control group of C57BL/6 age-matched (5-6 weeks) female were injected i.v. in the tail vein with 2.5 μg of an immunogenic form of particulate Ovalbumin (pOVA) and 2 μg of Keyhole Limpet Hemocyanin (KLH) s.c. in the hind limb once a week for 49 days (d0, 7, 14, 20, 28, 35, 42, 49). The other group of animals received pOVA and KLH in the same routes and amounts but admixed to 0.2 mg of nanocarrier attached to rapamycin ( Nanocarrier ID 1, 2 or 3) on days 0, 7 and 14 followed by pOVA injections between days 20 and 42 (same amounts as before). The control animals develop a robust immune response against OVA and KLH that can be measured by the anti-OVA or anti-KLH IgG antibody titers. The antibody titers at day 54 shown in FIG. 11 demonstrate that 3 doses of synthetic nanocarriers administered concomitantly with pOVA in the same solution were effective in reducing and preventing antibody formation to OVA for a prolonged period of time but not the KLH (that was injected in another location s.c.). The treated animals did not develop an anti-OVA response even after five injections of pOVA alone.
  • The data show that the concomitant administration of immunosuppressant attached to synthetic nanocarriers with a protein can reduce the anti-protein antibody response but that such a response is specific to the protein administered by the same route. This demonstrates the reduction of an undesired immune response against a protein with concomitant administration of immunosuppressant attached to synthetic nanocarriers. Finally, the results support that a protocol was established as provided herein.
  • Example 10: Evaluating Responses to KLH
  • Control C57BL/6 age-matched (5-6 weeks) female were injected i.v. in the tail vain with 200 μg of Keyhole Limpet Hemocyanin (KLH) once a week for 63 days (d0, 7, 14, 21, 28, 35, 42, 49, 56, 63). The other group received similar injections but 0.9 mg of nanocarriers attached to rapamycin ( Nanocarrier ID 1, 2 or 3) were admixed to the solution of KLH at days 0, 7, 14 and 21 followed by KLH six injections (same amount) between days 28 and 63. The control animals developed a robust response to KLH that can be measured by the anti-KLH IgG antibody titers as well as the anaphylactic reaction induced by the injections. The results in FIG. 12A show the antibody titers in the blood of all animals at the indicated time points, and the anaphylaxis scores induced by the injection of the antigen are presented in FIG. 12B. Four doses of synthetic nanocarriers administered concomitantly with KLH were effective in reducing and preventing antibody formation and anaphylaxis for a prolonged period of time. Indeed, treated animals did not develop an anti-KLH response even after four injections of KLH alone which in control animals was largely sufficient to create a response (day 26).
  • These results show that compositions provided herein when administered over a period of time concomitantly with a protein can reduce or prevent antibody formation and anaphylaxis for prolonged periods of time. Anaphylaxis scores were determined by three independent observers as follows: 0=no symptom, 1=lethargy, 2=lethargy and inability to right, 3=moribund.
  • These results show that administration of immunosuppressant attached to synthetic nanocarriers with KLH can reduce undesired anti-KLH antibody response as well as eliminate undesired anaphylactic reactions that can result from the administration of KLH without the immunosuppressant nanocarrier composition. This demonstrates the reduction of undesired immune responses against a protein with concomitant administration of immunosuppressant attached to synthetic nanocarriers. Finally, the results support that a protocol was established as provided herein.
  • Example 11: Evaluating Anti-HUMIRA Immune Responses in Arthritic Animals
  • Transgenic animals overexpressing human tumor necrosis factor alpha (huTNFaTg) develop progressive rheumatoid arthritis over the course of 20 weeks from birth. This process can be prevented by using the fully human anti-human TNFα antibody HUMIRA/adalimumab. However, repetitive administration of the initial therapeutic dose of HUMIRA (60 μg) leads to anti-drug antibody formation (ADA) that neutralizes the therapeutic benefit. Only very high doses can then maintain the therapeutic benefit and surmount the neutralizing immune response. For example, it is thought that about 200 μg is needed to effect maximal inhibition of inflammation in an example of such a mouse model (Binder et al., Arthritis & Rheumatism, Vol. 65 (No. 3), March 2013, pp. 608-617).
  • Age-matched huTNFaTg 5 week old female animals were either injected s.c. in the subscapular area with saline (PBS) or 60 μg of HUMIRA weekly or with a mixture of HUMIRA and 0.87 mg of nanocarriers attached to rapamycin ( Nanocarrier ID 1, 2 or 3) for the first 7 injections ( day 0, 7, 14, 21, 28, 35, 42, weeks 5 to 12 of age) followed by 3 injections of HUMIRA alone (same amount) (day 49 to 63, weeks 13 to 15 of age). The results in FIG. 13A show the antibody titers in the blood of all animals at day 21. The control animals that did not receive HUMIRA have no anti-HUMIRA titers, as expected; whereas a robust antibody response can be observed in the control animals that received only HUMIRA. In contrast, the animals treated with the nanocarriers remain completely negative even after three injections of HUMIRA without treatment. The monitoring of the limbs of these animals revealed a progressive disease that was already evident at 10 weeks of age in the control animals. The animals treated with HUMIRA alone had a significant blockade of the disease progression however adding nanocarriers to this regimen dramatically blocked the emergence of arthritic symptoms. The scores here represent the total of 4 independent scorers as: 1) represents synovitis, joint effusions and soft tissue swelling 2) includes proliferating inflamed synovial tissue which grows into joint cavity and destroys cartilage 3) shows extensive loss of cartilage, erosion around the margins of joint, and deformities 4) is almost end stage of the disease with fibrous or bony stiffening of joint, which ends it's functional life.
  • These results show that administration of immunosuppressant attached to synthetic nanocarriers with HUMIRA can reduce undesired anti-HUMIRA antibody response. The benefit of the concomitant administration is also evidenced by the further reduced arthritic score which demonstrate improved efficacy of HUMIRA. This demonstrates not only the reduction of undesired immune responses against a therapeutic protein with concomitant administration of immunosuppressant attached to synthetic nanocarriers but also improved efficacy. This also demonstrates that a higher dose of the therapeutic is not needed with the inventive concomitant administration provided herein. Further, based on the level of reduction in the arthritic score with the same dose of HUMIRA, reduced amounts of HUMIRA could also be used that would provide improved efficacy as compared to the amounts of HUMIRA that would be needed without the concomitant administration of immunosuppressant doses as provided herein. It is important to note that the amount of HUMIRA used in this example was 60 μg, much reduced as compared with amounts used in the art of about 200 μg in order to effect maximal inhibition of inflammation by HUMIRA (Binder et al., Arthritis & Rheumatism, Vol. 65 (No. 3), March 2013, pp. 608-617). Finally, the results support that a protocol was established as provided herein.
  • Example 12: Mesoporous Silica Nanoparticles with Attached Ibuprofen (Prophetic)
  • 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. 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). To do this, the particles are suspended in ethanol (30 mL), and APTMS (50 μL) 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.
  • In 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.
  • To form the core-shell nanocarriers, 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.
  • Example 13: Liposomes Containing Cyclosporine A (Prophetic)
  • The liposomes are formed using thin film hydration. 1,2-Dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) (32 μmol), cholesterol (32 μmol), and cyclosporin A (6.4 μmol) are dissolved in pure chloroform (3 mL). 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. for 1 h to form a suspension. 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.
  • Example 14: Preparation of Gold Nanocarriers (AuNCs) Containing Rapamycin (Prophetic)
  • Preparation of HS-PEG-Rapamycin:
  • A solution of PEG acid disulfide (1.0 eq), rapamycin (2.0-2.5 eq), DCC (2.5 eq) and DMAP (3.0 eq) in dry DMF is stirred at rt overnight. The insoluble dicyclohexylurea is removed by filtration and the filtrate is added to isopropyl alcohol (IPA) to precipitate out the PEG-disulfide-di-rapamycin ester and washed with IPA and dried. The polymer is then treated with tris(2-carboxyethyl)phosphine hydrochloride in DMF to reduce the PEG disulfide to thiol PEG rapamycin ester (HS-PEG-rapamycin). The resulting polymer is recovered by precipitation from IPA and dried as previously described and analyzed by H NMR and GPC.
  • Formation of Gold NCs (AuNCs):
  • An aq. solution of 500 mL of 1 mM HAuCl4 is heated to reflux for 10 min with vigorous stirring in a 1 L round-bottom flask equipped with a condenser. A solution of 50 mL of 40 mM of trisodium citrate is then rapidly added to the stirring solution. The resulting deep wine red solution is kept at reflux for 25-30 min and the heat is withdrawn and the solution is cooled to room temperature. The solution is then filtered through a 0.8 μm membrane filter to give the AuNCs solution. The AuNCs are characterized using visible spectroscopy and transmission electron microscopy. The AuNCs are ca. 20 nm diameter capped by citrate with peak absorption at 520 nm.
  • AuNCs Conjugate with HS-PEG-Rapamycin:
  • A solution of 150 μl of HS-PEG-rapamycin (10 μM 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 resuspended in suitable buffer for further analysis and bioassays.
  • Example 15: Liposomes Containing Rapamycin and Ovalbumin (Prophetic)
  • The liposomes are formed by thin film hydration. 1,2-Dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) (32 μmol), cholesterol (32 μmol), and rapamycin (6.4 μmol) are dissolved in pure chloroform (3 mL). This lipid solution is added to a 10 mL glass tube and the solvent is removed under nitrogen gas stream and desiccated for 6 hr. under vacuum. Multilamellar vesicles are obtained by hydration of the film with 2.0 ml of 25 mM MOPS buffer pH 8.5, containing excess amount of Ovalbumin. The tube is vortexed until the lipid film is peeled off from the tube surface. To break the multilamellar vesicles into monolamellar, ten cycles of freezing (liquid nitrogen) and thawing (30° C. water bath) are applied. The sample is then diluted to 1 ml in 25 mM MOPS buffer pH 8.5. Size of the resulting liposome is homogenized by extrusion by passing the sample 10 fold through a 200 nm pore polycarbonate filters. The resulting liposomes are then used for further analysis and bioassays.
  • Example 16: Tolerogenic Responses to HUMIRA Prevent Formation of Neutralizing Anti-HUMIRA Responses in Arthritic Animals Materials
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Code R1017). PLGA with a lactide:glycolide ratio of 3:1 and an inherent viscosity of 0.75 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 7525 DLG 7A). PLA-PEG-OMe block co-polymer with a methyl ether terminated PEG block of approximately 5,000 Da and an overall inherent viscosity of 0.5 DL/g was purchased from Lakeshore Biochemicals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 100 DL mPEG 5000 5CE). EMPROVE® Polyvinyl Alcohol 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).
  • Method
  • Solutions were prepared as follows:
  • Solution 1: PLGA at 75 mg/mL, PLA-PEG-OMe at 25 mg/mL, and rapamycin at 12.5 mg/mL in methylene chloride. The solution was prepared by dissolving PLGA, PLA-PEG-OMe, and rapamycin in pure methylene chloride. Solution 2: Polyvinyl alcohol @ 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • An oil-in-water emulsion was used to prepare the nanocarriers. The O/W emulsion was prepared by combining Solution 1 (1.0 mL) and Solution 2 (3.0 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 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 75,600×g and 4° C. for 50 min, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. The washing procedure was repeated, and the pellet was re-suspended in phosphate buffered saline for a final nanocarrier dispersion of about 10 mg/mL.
  • 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.
  • Effective Diameter (nm) Rapamycin Content (% w/w)
    241 11.5
  • Transgenic animals overexpressing human tumor necrosis factor alpha (huTNFαTg) develop progressive rheumatoid arthritis over the course of 20 weeks from birth. This process can be prevented by using the fully human anti-human TNFα antibody Adalimumab or HUMIRA. However repetitive administration of the initial therapeutic dose of HUMIRA (60 μg) leads to anti-drug antibody formation (ADA) that neutralizes the therapeutic benefit.
  • Age-matched HuTNFαTg 5 week old female animals were either injected s.c. in the subscapular area with saline (PBS) or 60 μg of HUMIRA weekly or with a mixture of HUMIRA and 0.87 mg of tolerogenic nanoparticles for the first 7 injections (day 0 to 42, weeks 5 to 11 of age) followed by 10 weekly injections of HUMIRA alone (same amounts) (day 49 to 107, weeks 12 to 20 of age). The protocol is shown in FIG. 14.
  • The results in FIG. 15 (left panel) show the antibody titers in the blood of all animals at different time points. The control mock-treated animals have no titers as expected whereas a robust anti-HUMIRA antibody response can be observed in the animals that received just HUMIRA. Treatment with tolerogenic nanocarriers from week 5 to week 11 of age led to a complete resistance to develop anti-HUMIRA titers even after 10 injections of HUMIRA without the tolerogenic treatment (weeks 12 to 20). The monitoring of the limbs of these animals revealed a progressive disease that was already evident at 10 weeks of age. HUMIRA alone had a significant blockade of the disease progression however adding tolerogenic nanocarriers to this regimen dramatically blocked the emergence of arthritic symptoms. The scores here represent the average total of 4 independent scorers as: 1) represents synovitis, joint effusions and soft tissue swelling 2) includes proliferating inflamed synovial tissue which grows into joint cavity and destroys cartilage 3) shows extensive loss of cartilage, erosion around the margins of joint, and deformities 4) is almost end stage of the disease with fibrous or bony stiffening of joint, which ends it's functional life.
  • These results show that compositions provided herein when administered over a period of time concomitantly with a protein can reduce or prevent antibody formation to a biological therapeutic and therefore improving its efficacy and therapeutic window for prolonged periods of time. These results also show that administration of immunosuppressant attached to synthetic nanocarriers with HUMIRA can reduce undesired anti-HUMIRA antibody response. The benefit of the concomitant administration is also evidenced by the further reduced arthritic score which demonstrates improved efficacy of HUMIRA. This demonstrates not only the reduction of undesired immune responses against a therapeutic protein with concomitant administration of immunosuppressant attached to synthetic nanocarriers but also improved efficacy. This also demonstrates that a higher dose of the therapeutic is not needed with the inventive concomitant administration provided herein. Further, based on the level of reduction in the arthritic score with the same dose of HUMIRA, reduced amounts of HUMIRA could also be used that would provide improved efficacy as compared to the amounts of HUMIRA that would be needed without the concomitant administration of immunosuppressant doses as provided herein. It is important to note that the amount of HUMIRA used in this example was 60 μg, much reduced as compared with amounts used in the art of about 200 μg in order to effect maximal inhibition of inflammation by HUMIRA (Binder et al., Arthritis & Rheumatism, Vol. 65 (No. 3), March 2013, pp. 608-617). Finally, the results support that a protocol was established as provided herein.
  • Example 17: Evaluating Anti-HUMIRA Immune Responses Materials
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702; Product Code R1017). PLGA with a lactide:glycolide ratio of 3:1 and an inherent viscosity of 0.75 dL/g was purchased from SurModics Pharmaceuticals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 7525 DLG 7A). PLA-PEG-OMe block co-polymer with a methyl ether terminated PEG block of approximately 5,000 Da and an overall inherent viscosity of 0.5 DL/g was purchased from Lakeshore Biochemicals (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 100 DL mPEG 5000 5CE). EMPROVE® Polyvinyl Alcohol 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).
  • Method
  • Solutions were prepared as follows:
  • Solution 1: PLGA at 75 mg/mL, PLA-PEG-OMe at 25 mg/mL, and rapamycin at 12.5 mg/mL in methylene chloride. The solution was prepared by dissolving PLGA, PLA-PEG-OMe, and rapamycin in pure methylene chloride. Solution 2: Polyvinyl alcohol @ 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • An oil-in-water emulsion was used to prepare the nanocarriers. The O/W emulsion was prepared by combining Solution 1 (1.0 mL) and Solution 2 (3.0 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 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 75,600×g and 4° C. for 50 min, removing the supernatant, and re-suspending the pellet in phosphate buffered saline. The washing procedure was repeated, and the pellet was re-suspended in phosphate buffered saline for a final nanocarrier dispersion of about 10 mg/mL.
  • 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.
  • Effective Diameter (nm) Rapamycin Content (% w/w)
    238 10.6
  • Rapamycin-containing nanocarriers were generated using the materials and methods described above. 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.
  • Effective Diameter (nm) Rapamycin Content (% w/w)
    241 11.5
  • Anti-HUMIRA immune and neutralizing responses in arthritic animals were evaluated. Transgenic animals overexpressing human tumor necrosis factor alpha (huTNFαTg) develop progressive rheumatoid arthritis over the course of 20 weeks from birth. This process can be prevented by using the fully human anti-human TNFα antibody Adalimumab or HUMIRA. However repetitive administration of the initial therapeutic dose of HUMIRA (60 μg) leads to anti-drug antibody formation (ADA) that neutralizes the therapeutic benefit. Higher quantities of Humira (200 μg) can be injected to overcome this antagonizing immune response to allow for the therapeutic effect of Humira.
  • Age-matched HuTNFαTg 5 week old female animals were either injected s.c. in the subscapular area with saline (PBS) or 60 μg or 200 μg of HUMIRA weekly (weeks 5-10) or with a mixture of 60 μg of Humira and 0.87 mg of tolerogenic nanoparticles for the first 3 injections (week 5 to 7 of age) followed by 3 weekly injections of HUMIRA alone (same amounts) (weeks 8 to 10 of age). The results in FIG. 17 (left panel) show the antibody titers in the blood of all animals at different time points. The control mock-treated animals have no titers as expected whereas a robust anti-HUMIRA antibody response can be observed in the animals that received just HUMIRA. Treatment with the nanocarriers from week 5 to 7 of age led to a complete resistance to develop anti-HUMIRA titers even after 3 injections of HUMIRA without the treatment (weeks 8 to 10). Of note, three injections of HUMIRA led to very high titers on the control animals (week 7 titers) while three similar injections (for a total of six) in animals treated with the nanocarriers were totally resistant to develop titers (week 10). FIG. 16 illustrates the dosing regimen.
  • The monitoring of the limbs of these animals revealed a progressive disease that was already evident at 6 weeks of age. HUMIRA alone had a significant blockade of the disease progression however adding nanocarriers to this regimen dramatically blocked the emergence of arthritic symptoms. The scores (FIG. 17 (right panel)) represent the average total of 4 independent scorers as: 1) represents synovitis, joint effusions and soft tissue swelling 2) includes proliferating inflamed synovial tissue which grows into joint cavity and destroys cartilage 3) shows extensive loss of cartilage, erosion around the margins of joint, and deformities 4) is almost end stage of the disease with fibrous or bony stiffening of joint, which ends it's functional life.
  • These results show that methods and compositions provided herein can reduce or prevent antibody formation to a therapeutic protein and improve its efficacy. Specifically, as above, these results show that administration of immunosuppressant attached to synthetic nanocarriers with HUMIRA can reduce undesired anti-HUMIRA antibody response. The benefit of the concomitant administration is also evidenced by the further reduced arthritic score which demonstrate improved efficacy of HUMIRA. This demonstrates not only the reduction of undesired immune responses against a therapeutic protein with concomitant administration of immunosuppressant attached to synthetic nanocarriers but also improved efficacy. This also demonstrates that a higher dose of the therapeutic is not needed with the inventive concomitant administration provided herein. Further, based on the level of reduction in the arthritic score with the same dose of HUMIRA, reduced amounts of HUMIRA could also be used that would provide improved efficacy as compared to the amounts of HUMIRA that would be needed without the concomitant administration of immunosuppressant doses as provided herein. It is important to note that the amount of HUMIRA used in this example was 60 μg, much reduced as compared with amounts used in the art of about 200 μg in order to effect maximal inhibition of inflammation by HUMIRA (Binder et al., Arthritis & Rheumatism, Vol. 65 (No. 3), March 2013, pp. 608-617). Finally, the results support that a protocol was established as provided herein.
  • Example 18: Antigen-Specific Tolerogenic Responses to Chicken Ovalbumin with Encapsulated Rapamycin NP[Rapa] Materials and Methods Materials
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, Mass. 01702), product code R1017. 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. PLA-PEG-OMe block co-polymer with a methyl ether terminated PEG block of approximately 5,000 Da and an overall inherent viscosity of 0.50 DL/g was purchased from Lakeshore Biomaterials (756 Tom Martin Drive, Birmingham, Ala. 35211), product code 100 DL mPEG 5000 5CE. EMPROVE® Polyvinyl Alcohol 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. Cellgro phosphate buffered saline 1× (PBS 1×) was purchased from Corning (9345 Discovery Blvd. Manassas, Va. 20109), product code 21-040-CV.
  • Method
  • Solutions were prepared as follows:
  • Solution 1: A polymer and rapamycin mixture was prepared by dissolving PLGA at 75 mg per 1 mL, PLA-PEG-Ome at 25 mg per 1 mL, and rapamycin as 12.5 mg per 1 mL in dichloromethane. Solution 2: Polyvinyl alcohol was prepared at 50 mg/mL in 100 mM pH 8 phosphate buffer.
  • An O/W emulsions was prepared by combining Solution 1 (1.0 mL) and Solution 2 (3.0 mL) in a small glass pressure tube and sonicating at 30% amplitude for 60 seconds using a Branson Digital Sonifier 250. The O/W emulsion was added to an open beaker containing 70 mM pH 8 phosphate buffer solution (60 mL). Three additional, identical O/W emulsions were prepared and added to the same beaker as the first. These were then stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and for the nanocarriers to form. A portion of the nanocarriers was washed by transferring the nanocarrier suspension to centrifuge tubes and centrifuging at 75,600×g and 4° C. for 35 minutes, removing the supernatant, and re-suspending the pellet in PBS 1×. The wash procedure was repeated and then 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. An identical formulation was prepared as above in a separate beaker, and combined with the first after the wash step. The mixed nanocarrier solution was then filtered using 1.2 μm PES membrane syringe filters from Pall part number 4656, and 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.
  • Effective Diameter (nm) Rapamycin Content (% w/w)
    220 11.85
  • NP[OVA] Materials and Methods Materials
  • Ovalbumin protein, was purchased from Worthington Biochemical Corporation (730 Vassar Avenue, Lakewood, N.J. 08701), product code LS003054). PLGA with 54% lactide and 46% glycolide content 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). PLA-PEG block co-polymer with a methyl ether terminated PEG block of approximately 5,000 Da and Mw of 28,000 Da, inherent viscosity of 0.38 dL/g was purchased from Lakeshore Biomaterials (756 Tom Martin Drive, Birmingham, Ala. 35211), product code 100 DL mPEG 5000 4CE. EMPROVE® Polyvinyl Alcohol 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 1.41350.1001. Cellgro Phosphate-buffered saline 1× (PBS 1×) was purchased from Corning (9345 Discovery Blvd. Manassas, Va. 20109), product code 21-040-CV.
  • Method
  • Solutions were prepared as follows:
  • Solution 1: Ovalbumin protein @ 50 mg/mL was prepared in 10 mM phosphate buffer pH 8 with 10% by weight sucrose. Solution 2: PLGA was prepared by dissolving PLGA at 100 mg per 1 mL of dichloromethane in the chemical fume hood. Solution 3: PLA-PEG-OMe was prepared by dissolving PLA-PEG-OMe at 100 mg per 1 mL of dichloromethane in the chemical fume hood. Solution 4: Polyvinyl alcohol @ 65 mg/mL in 100 mM phosphate buffer, pH 8.
  • A primary (W1/O) emulsion was first created by mixing Solutions 1 through 3. Solution 1 (0.2 mL), Solution 2 (0.75 mL), and Solution 3 (0.25 mL) were combined in a small glass pressure tube which was pre-chilled >4 minutes in an ice water bath, and sonicated at 50% amplitude for 40 seconds over an ice bath using a Branson Digital Sonifier 250. A secondary (W1/O/W2) emulsion was then formed by adding Solution 4 (3 mL) to the primary emulsion, vortex mixing to create a milky dispersion, and then sonicating at 30% amplitude for 60 seconds over an ice bath using the Branson Digital Sonifier 250. The secondary emulsion was added to an open 50 mL beaker containing PBS 1× (30 mL). A second identical double emulsion formulation was prepared as described above, and added to the same 50 mL beaker as the first. The two preparations were stirred at room temperature for 2 hours to allow the dichloromethane to evaporate and the nanocarriers to form in suspension. A portion of the suspended nanocarriers was washed by transferring the nanocarrier suspension to a centrifuge tube, spinning at 75,600 rcf for 50 minutes, removing the supernatant, and re-suspending the pellet in PBS 1×. This washing procedure was repeated and then 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 suspension was stored frozen at −20 C until use.
  • Effective Diameter (nm) Ovalbumin Content (% w/w)
    164 5.81
  • NP[GSK1059615] Materials and Methods Materials
  • 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. PLA-PEG-OMe block co-polymer with a methyl ether terminated PEG block of approximately 5,000 Da and an overall inherent viscosity of 0.26 DL/g was purchased from Lakeshore Biomaterials (756 Tom Martin Drive, Birmingham, Ala. 35211; Product Code 100 DL mPEG 5000 5K-E). 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.
  • Method
  • Solutions were prepared as follows:
  • 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 μm 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.
  • Effective Diameter (nm) GSK1059615 Content (% w/w)
    143 1.02
  • C57BL/6 age-matched (5-6 weeks) female mice were injected i.v. in the tail vein on days −21 and −14 with saline (No Treatment), 1.1 mg of whole Ovalbumin-loaded nanocarriers (NP[OVA]) combined to either 1.2 mg of rapamycin-containing nanocarriers (NP[Rapa]) or 8 mg of GSK1059615-loaded nanocarriers (NP[GSK1059615]).
  • At day 0 all animals were injected s.c. in the hind limbs with 25 μg of particulate OVA (pOVA) admixed to 2 μg of CpG followed by injections of just 25 μg pOVA on days 7 and 14. Antibody titers were measured on day 21. In absence of any treatment, the animals developed a robust immune response against OVA that can be measured by the anti-OVA IgG antibody titers. The antibody titers at day 21 shown in FIG. 18 demonstrate that 2 doses of synthetic tolerogenic nanocarriers administered concomitantly with encapsulated OVA in the same solution (NP[OVA]+NP[Rapa] or NP[GSK1059615]) were effective in reducing antibody formation to OVA even after 1 injection of OVA+CpG and 2 injections of OVA alone.
  • These results show that encapsulated immunosuppressants (such as rapamycin and GSK1059615]) when concomitantly delivered with a protein can prevent antibody formation to that protein for multiple challenges and periods of time. This demonstrates not only the reduction of undesired immune responses against a protein with concomitant administration of two different kinds of immunosuppressant. Finally, the results support that a protocol was established as provided herein.
  • Example 19: Inventive Method Using Reduced Pharmacodynamically Effective Dose (Prophetic)
  • Three thousand two hundred human subjects suffering from rheumatoid arthritis are recruited for a series of clinical trials. In a pilot dose ranging trial, 1,200 subjects are divided into four arms (placebo and 3 different dose levels of synthetic nanocarriers of Example 3). Each subject in each of the four arms receives two rounds of HUMIRA 40 mg s.c. concomitantly with either placebo or synthetic nanocarrier. The synthetic nanocarrier dose that most reduces the mean level of anti-HUMIRA antibodies in an arm is declared to be the “Immunosuppressant Dose” for the trial.
  • In another pilot trial, the recruited human subjects are divided into 4 Test Arms of 500 subjects each. Placebo, HUMIRA, and synthetic nanocarriers of Example 3 are administered concomitantly (except for Test Arm 1) according to the following table, with the synthetic nanocarriers being administered at the Immunosuppressant Dose.
  • Test Arm Number HUMIRA dose NC Admin (sc)
    1 40 mg sc
    2 40 mg sc +
    3 20 mg sc +
    4 10 mg sc +
    5 Placebo Placebo
  • The target pharmacodynamic effects (“PD effect”) are evaluated, which in this case are the means of ACR 20, 50 and 70 responses for the subjects in each Test Arm. The PD effect for subjects in Test Arm 1 is noted, and the HUMIRA dose in Test Arm 1 is arbitrarily defined as the pharmacodynamically effective dose (“PD Effective Dose”) of HUMIRA.
  • The PD effect for Test Arms 2-4 are then evaluated, and the lowest dose at which the PD effect is not significantly different from the Test Arm 1 PD effect is declared to be the reduced pharmacodynamically effective dose of HUMIRA.
  • In an application of the information established during the pilot trials, a reduced pharmacodynamically effective dose of HUMIRA is administered concomitantly with the Immunosuppressant Dose (containing synthetic nanocarriers of Example 3) to subjects diagnosed with rheumatoid arthritis and at risk of developing antibodies to HUMIRA. In a further embodiment, a protocol using the information established during the pilot trials is prepared to guide concomitant dosing of Humira™ and synthetic nanocarriers of Example 3 to human subjects diagnosed with rheumatoid arthritis and have or are expected to have antibodies to HUMIRA. This protocol is then used to guide concomitant administration of a reduced pharmacodynamically effective dose of HUMIRA, and synthetic nanocarriers of Example 3, to human subjects.
  • Example 20: Inventive Method Demonstrating Enhanced Pharmacodynamic Effect (Prophetic)
  • Three thousand seven hundred human subjects suffering from rheumatoid arthritis are recruited for a series of clinical trials. In a pilot dose ranging trial, 1,200 subjects are divided into four arms (placebo and 3 different dose levels of synthetic nanocarriers of Example 3). Each subject in each of the four arms receives two rounds of HUMIRA 40 mg s.c. concomitantly with either placebo or synthetic nanocarrier. The synthetic nanocarrier dose that most reduces the mean level of anti-HUMIRA antibodies in an arm is declared to be the “Immunosuppressant Dose” for the trial.
  • In another pilot trial, the recruited human subjects are divided into 3 Test Arms, with two active Test Arms having 1000 subjects each, and one placebo arm of 500 subjects. Placebo, HUMIRA, and synthetic nanocarriers of Example 3 are administered concomitantly (except for Test Arm 1) according to the following table, with the synthetic nanocarriers being administered at the Immunosuppressant Dose.
  • Test Arm Number HUMIRA dose NC Admin (sc)
    1 40 mg sc
    2 40 mg sc +
    3 Placebo Placebo
  • The target pharmacodynamic effects (“PD effect”) are evaluated, which in this case are the means of ACR 20, 50 and 70 responses for the subjects in each Test Arm. The PD effect for subjects in Test Arm 1 is noted and compared to the PD effect in Test Arm 2. Any enhancement of the pharmacodynamic effect of HUMIRA upon concomitant administration with the Immunosuppressant Dose in Test Arm 2 is noted, as compared with the PD effect observed in Test Arm 1.
  • In an application of the information established during the pilot trials, the standard 40 mg dose of HUMIRA is administered concomitantly with the Immunosuppressant Dose (containing synthetic nanocarriers of Example 3) to subjects diagnosed with rheumatoid arthritis and at risk of developing antibodies to HUMIRA. In a further embodiment, a protocol using the information established during the pilot trials is prepared to guide concomitant dosing of HUMIRA and synthetic nanocarriers of Example 3 to human subjects diagnosed with rheumatoid arthritis and known or believed to have antibodies to HUMIRA. This protocol is then used to guide concomitant administration of HUMIRA, and synthetic nanocarriers of Example 3, to human subjects. Any enhanced pharmacodynamic effect following the concomitant administration is recorded using the approaches disclosed herein.
  • Example 21: Inventive Method Demonstrating Enhanced Pharmacodynamic Effect (Prophetic)
  • Three thousand two hundred human subjects suffering from chemotherapy-related anemia are recruited for a series of clinical trials. In a pilot dose ranging trial, modified mRNA encoding erythropoietin is prepared according to U.S. Patent application 2013/0115272 to de Fougerolles et al. (“mmRNA”). Twelve hundred subjects are divided into four arms (placebo and 3 different dose levels of synthetic nanocarriers of Example 3). Each subject in each of the four arms receives a therapeutic dose of mmRNA concomitantly with either placebo or synthetic nanocarrier. The synthetic nanocarrier dose that most reduces the mean level of anti-mmRNA antibodies in an arm is declared to be the “Immunosuppressant Dose” for the trial.
  • In another pilot trial, the recruited human subjects are divided into 4 Test Arms of 500 subjects each. Placebo, mmRNA, and synthetic nanocarriers of Example 3 are administered concomitantly (except for Test Arm 1) according to the following table, with the synthetic nanocarriers being administered at the Immunosuppressant Dose.
  • Test Arm Number mmRNA dose NC Admin (sc)
    1 Therapeutic dose sc
    2 Therapeutic dose sc +
    3 ½ Therapeutic dose sc +
    4 ¼ Therapeutic dose sc +
    5 Placebo Placebo
  • The target pharmacodynamic effects (“PD effect”) are evaluated, which in this case are the means of chemotherapy-induced anemia responses for the subjects in each Test Arm. The PD effect for subjects in Test Arm 1 is noted, and the mmRNA dose in Test Arm 1 is arbitrarily defined as the pharmacodynamically effective dose (“PD Effective Dose”) of mmRNA. The PD effect for Test Arms 2-4 are then evaluated, and the lowest dose at which the PD effect is not significantly different from the Test Arm 1 PD effect is declared to be the reduced pharmacodynamically effective dose of mmRNA. In an application of the information established during the pilot trials, a reduced pharmacodynamically effective dose of mmRNA is administered concomitantly with the Immunosuppressant Dose (containing synthetic nanocarriers of Example 3) to subjects diagnosed with chemotherapy-related anemia and at risk of developing antibodies to mmRNA.
  • In a further embodiment, a protocol using the information established during the pilot trials is prepared to guide concomitant dosing of mmRNA and synthetic nanocarriers of Example 3 to human subjects diagnosed with chemotherapy-related anemia and known or believed to have antibodies to mmRNA. This protocol is then used to guide concomitant administration of a reduced pharmacodynamically effective dose of mmRNA, and synthetic nanocarriers of Example 3, to human subjects.
  • Example 22: Method to Maintain Efficacy of a Biologic Drug During a Period of Multidosing
  • Biologic drugs often lose activity over time after multiple doses. A common cause of loss of efficacy is the formation of ADAs. ADAs can cause loss of drug efficacy, for example by 1) enhancing clearance of the drug, such that the PK of the drug after multiple doses is substantially less than the PK of the drug after a single dose or by 2) neutralizing the activity of the drug, such that the biological activity of the drug after multiple doses is substantially less than the biological activity after a single dose. It would be desirable to maintain activity of the biologic drug over the course of multiple dosing.
  • Transgenic mice expressing human TNF-α spontaneously develop arthritis over a period of 5-20 weeks of age. Mice were treated weekly with HUMIRA (60 μg/injection) with or without synthetic nanocarriers from Example 16 from weeks 5-11. Panel B (FIG. 19) shows the serum levels of HUMIRA after the first dose (Day 1). After 6 doses of HUMIRA, the blood levels of HUMIRA are close to baseline in mice treated with HUMIRA alone and remain low through week 20 (Panel C, FIG. 19, black squares). In contrast, mice treated with HUMIRA and the synthetic nanocarriers show serum levels of HUMIRA (panel C, FIG. 19, blue triangles) that are similar to that after the first dose, indicating that synthetic nanocarrier treatment enabled effective blood levels of HUMIRA to be maintained after multiple dosing.
  • The reduction in serum HUMIRA blood levels in mice treated with HUMIRA alone can be attributed to the development of ADAs. By week 11, mice treated with HUMIRA alone developed high titer anti-drug antibodies (Panel A, FIG. 19, black square symbols). In contrast mice treated with the synthetic nanocarriers show little or no anti-HUMIRA antibody response (Panel A, FIG. 19, blue triangles). The impact of the reduction of HUMIRA serum levels with multiple dosing is apparent in the arthritis disease scores as above in Example 16 (Panel D, FIG. 19). Mice treated with HUMIRA alone show suboptimal attenuation of arthritis scores from week 10-20 (Panel D, FIG. 19, compare black squares to black circles). In contrast, mice treated with synthetic nanocarriers show strong inhibition of arthritis (Panel C, FIG. 19, blue triangles).
  • These results indicate that maintenance of drug levels with multiple dosing is important for drug efficacy, and that the immunosuppressants attached to synthetic nanocarriers maintain drug levels with multiple dosing by inhibiting the ADA response. Accordingly, these results demonstrate the benefits of repeatedly dosing therapeutic macromolecules concomitantly with immunosuppressants as provided herein. The results further demonstrate the ability to determine protocols that can accomplish the desired therapeutic effects and/or reduced ADA responses in subjects.
  • Example 23: Inventive Method Demonstrating Enhanced Pharmacodynamic Effect (Prophetic)
  • Three thousand seven hundred human subjects suffering from rheumatoid arthritis are recruited for a series of clinical trials. In a pilot dose ranging trial, 1,200 subjects are divided into four arms (placebo and 3 different dose levels of nanocrystalline rapamycin). Each subject in each of the four arms receives two rounds of HUMIRA 40 mg s.c. concomitantly with either placebo or nanocrystalline rapamycin. The nanocrystalline rapamycin dose that most reduces the mean level of anti-HUMIRA antibodies in an arm is declared to be the “Immunosuppressant Dose” for the trial.
  • In another pilot trial, the recruited human subjects are divided into 3 Test Arms, with two active Test Arms having 1000 subjects each, and one placebo arm of 500 subjects. Placebo, HUMIRA, and nanocrystalline rapamycin are administered concomitantly (except for Test Arm 1) according to the following table, with the synthetic nanocarriers being administered at the Immunosuppressant Dose.
  • Test Arm Number HUMIRA dose Immunosuppressant dose (sc)
    1 40 mg sc
    2 40 mg sc +
    3 Placebo Placebo
  • The target pharmacodynamic effects (“PD effect”) are evaluated, which in this case are the means of ACR 20, 50 and 70 responses for the subjects in each Test Arm. The PD effect for subjects in Test Arm 1 is noted and compared to the PD effect in Test Arm 2. Any enhancement of the pharmacodynamic effect of HUMIRA upon concomitant administration with the Immunosuppressant Dose in Test Arm 2 is noted, as compared with the PD effect observed in Test Arm 1.
  • In an application of the information established during the pilot trials, the standard 40 mg dose of HUMIRA is administered concomitantly with the Immunosuppressant Dose (containing nanocrystalline rapamycin) to subjects diagnosed with rheumatoid arthritis and at risk of developing antibodies to HUMIRA. In a further embodiment, a protocol using the information established during the pilot trials is prepared to guide concomitant dosing of HUMIRA and nanocrystalline rapamycin to human subjects diagnosed with rheumatoid arthritis and known or believed to have antibodies to HUMIRA. This protocol is then used to guide concomitant administration of HUMIRA and nanocrystalline rapamycin, to human subjects. Any enhanced pharmacodynamic effect following the concomitant administration is recorded using the approaches disclosed herein.
  • Example 24: Inventive Method Demonstrating Enhanced Pharmacodynamic Effect (Prophetic)
  • Three thousand two hundred human subjects suffering from chemotherapy-related anemia are recruited for a series of clinical trials. In a pilot dose ranging trial, modified mRNA encoding erythropoietin is prepared according to U.S. Patent application 2013/0115272 to de Fougerolles et al. (“mmRNA”). Twelve hundred subjects are divided into four arms (placebo and 3 different dose levels of nanocrystalline rapamycin). Each subject in each of the four arms receives a therapeutic dose of mmRNA concomitantly with either placebo or nanocrystalline rapamycin. The nanocrystalline rapamycin dose that most reduces the mean level of anti-mmRNA antibodies in an arm is declared to be the “Immunosuppressant Dose” for the trial.
  • In another pilot trial, the recruited human subjects are divided into 4 Test Arms of 500 subjects each. Placebo, mmRNA, and synthetic nanocarriers of Example 3 are administered concomitantly (except for Test Arm 1) according to the following table, with the nanocrystalline rapamycin being administered at the Immunosuppressant Dose.
  • Nanocrystalline
    Test Arm Number mmRNA dose rapamycin (sc)
    1 Therapeutic dose sc
    2 Therapeutic dose sc +
    3 ½ Therapeutic dose sc +
    4 ¼ Therapeutic dose sc +
    5 Placebo Placebo
  • The target pharmacodynamic effects (“PD effect”) are evaluated, which in this case are the means of chemotherapy-induced anemia responses for the subjects in each Test Arm. The PD effect for subjects in Test Arm 1 is noted, and the mmRNA dose in Test Arm 1 is arbitrarily defined as the pharmacodynamically effective dose (“PD Effective Dose”) of mmRNA.
  • The PD effect for Test Arms 2-4 are then evaluated, and the lowest dose at which the PD effect is not significantly different from the Test Arm 1 PD effect is declared to be the reduced pharmacodynamically effective dose of mmRNA. In an application of the information established during the pilot trials, a reduced pharmacodynamically effective dose of mmRNA is administered concomitantly with the Immunosuppressant Dose (containing nanocrystalline rapamycin) to subjects diagnosed with chemotherapy-related anemia and at risk of developing antibodies to mmRNA.
  • In a further embodiment, a protocol using the information established during the pilot trials is prepared to guide concomitant dosing of mmRNA and nanocrystalline rapamycin to human subjects diagnosed with chemotherapy-related anemia and known or believed to have antibodies to mmRNA. This protocol is then used to guide concomitant administration of a reduced pharmacodynamically effective dose of mmRNA and nanocrystalline rapamycin, to human subjects.

Claims (31)

1. A method comprising:
providing an immunosuppressant dose, wherein the immunosuppressant dose is attached to synthetic nanocarriers; and
administering a reduced pharmacodynamically effective dose of a therapeutic macromolecule concomitantly with the immunosuppressant dose to a subject in which an anti-therapeutic macromolecule antibody response is expected to occur;
wherein the concomitant administration is according to a protocol that has been demonstrated to result in a pharmacodynamic effect with the reduced pharmacodynamically effective dose of the therapeutic macromolecule upon concomitant administration with the immunosuppressant dose, as compared to administration of the therapeutic macromolecule when not administered concomitantly with the immunosuppressant dose and in the presence of an anti-therapeutic macromolecule antibody response; or
wherein the reduced pharmacodynamically effective dose of the therapeutic macromolecule is less than a pharmacodynamically effective dose of the therapeutic macromolecule that: (A) is administered in the presence of an anti-therapeutic macromolecule antibody response, and (B) is not administered concomitantly with the immunosuppressant dose; or
wherein the concomitant administration is according to a protocol that has been demonstrated to enhance a pharmacodynamic effect of the therapeutic macromolecule upon concomitant administration with the immunosuppressant dose, as compared to administration of the therapeutic macromolecule when not administered concomitantly with the immunosuppressant dose, and each in the presence of an anti-therapeutic macromolecule antibody response.
2. The method of claim 1, wherein the method further comprises determining the protocol.
3. The method of claim 1, wherein the method further comprises determining the reduced pharmacodynamically effective dose.
4. The method of claim 1, wherein the method further comprises assessing the pharmacodynamic effect in the subject prior to and/or after the administration.
5-14. (canceled)
15. The method of claim 1, further comprising:
recording an enhanced pharmacodynamic effect following the concomitant administration.
16-18. (canceled)
19. A method comprising:
providing therapeutic macromolecules that cause or are expected to cause anti-therapeutic macromolecule antibodies upon repeated dosing in one or more subjects;
providing an immunosuppressant dose, wherein the immunosuppressant dose is attached to synthetic nanocarriers; and
repeatedly dosing at the same or a lower dose a subject with the therapeutic macromolecules concomitantly with the immunosuppressant dose.
20. The method of claim 19, wherein the concomitant administration is according to a protocol that has been demonstrated to result in maintenance of a pharmacodynamic effect of the therapeutic macromolecule over two or more doses of the therapeutic macromolecules to a subject.
21. The method of claim 20, wherein the method further comprises determining the protocol.
22. The method of claim 19, wherein the method further comprises assessing a pharmacodynamic effect in the subject prior to and/or after the administration of the two or more doses.
23. (canceled)
24. A composition or kit comprising:
an immunosuppressant dose wherein the immunosuppressant is attached to synthetic nanocarriers; and
a reduced pharmacodynamically effective dose of a therapeutic macromolecule.
25-28. (canceled)
29. The method of claim 1, wherein the therapeutic macromolecule is not attached to the synthetic nanocarriers.
30. The method of claim 1, wherein the therapeutic macromolecule is attached to the synthetic nanocarriers.
31-33. (canceled)
34. The method of claim 1, wherein the reduced pharmacodynamically effective dose of the therapeutic macromolecule is at least 30% less than a pharmacodynamically effective dose of the therapeutic macromolecule that: (A) is administered in the presence of an anti-therapeutic macromolecule antibody response, and (B) is not administered concomitantly with the immunosuppressant dose.
35. The method of claim 34, wherein the reduced pharmacodynamically effective dose is at least 40% less.
36. (canceled)
37. The method of claim 1, wherein the immunosuppressant dose comprises a statin, an mTOR inhibitor, a TGF-β signaling agent, a corticosteroid, an inhibitor of mitochondrial function, a P38 inhibitor, an NF-κβ inhibitor, an adenosine receptor agonist, a prostaglandin E2 agonist, a phosphodiesterase 4 inhibitor, an HDAC inhibitor or a proteasome inhibitor.
38-40. (canceled)
41. The method of claim 1, wherein the therapeutic macromolecule comprises a/an infusible or injectable therapeutic protein, enzyme, enzyme cofactor, hormone, blood or blood coagulation factor, cytokine, interferon, growth factor, monoclonal antibody, polyclonal antibody or protein associated with Pompe's disease.
42-50. (canceled)
51. The method of claim 1, wherein a load of immunosuppressant attached to the synthetic nanocarriers, on average across the synthetic nanocarriers, is between 0.1% and 50%.
52. (canceled)
53. The method of claim 1, wherein the synthetic nanocarriers comprise lipid nanoparticles, polymeric nanoparticles, metallic nanoparticles, surfactant-based emulsions, dendrimers, buckyballs, nanowires, virus-like particles or peptide or protein particles.
54-63. (canceled)
64. The method of claim 1, wherein the mean of a particle size distribution obtained using dynamic light scattering of the synthetic nanocarriers is a diameter greater than 100 nm.
65-68. (canceled)
69. The method of claim 1, wherein an aspect ratio 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.
US16/550,725 2013-05-03 2019-08-26 Methods related to administering immunosuppressants and therapeutic macromolecules at a reduced pharmacodynamically effective dose Pending US20200113874A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/550,725 US20200113874A1 (en) 2013-05-03 2019-08-26 Methods related to administering immunosuppressants and therapeutic macromolecules at a reduced pharmacodynamically effective dose

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US201361819517P 2013-05-03 2013-05-03
US201361881913P 2013-09-24 2013-09-24
US201361881921P 2013-09-24 2013-09-24
US201361881851P 2013-09-24 2013-09-24
US201361907177P 2013-11-21 2013-11-21
US201461948384P 2014-03-05 2014-03-05
US201461948313P 2014-03-05 2014-03-05
US14/269,047 US10434088B2 (en) 2013-05-03 2014-05-02 Methods related to administering immunosuppressants and therapeutic macromolecules at a reduced pharmacodynamically effective dose
US16/550,725 US20200113874A1 (en) 2013-05-03 2019-08-26 Methods related to administering immunosuppressants and therapeutic macromolecules at a reduced pharmacodynamically effective dose

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/269,047 Continuation US10434088B2 (en) 2013-05-03 2014-05-02 Methods related to administering immunosuppressants and therapeutic macromolecules at a reduced pharmacodynamically effective dose

Publications (1)

Publication Number Publication Date
US20200113874A1 true US20200113874A1 (en) 2020-04-16

Family

ID=51841530

Family Applications (13)

Application Number Title Priority Date Filing Date
US14/269,048 Active 2034-09-18 US10668053B2 (en) 2013-05-03 2014-05-02 Tolerogenic synthetic nanocarriers to reduce or prevent anaphylaxis in response to a non-allergenic antigen
US14/269,056 Active 2035-04-20 US10357483B2 (en) 2013-05-03 2014-05-02 Methods comprising dosing combinations for reducing undesired humoral immune responses
US14/269,047 Active 2035-03-28 US10434088B2 (en) 2013-05-03 2014-05-02 Methods related to administering immunosuppressants and therapeutic macromolecules at a reduced pharmacodynamically effective dose
US14/269,054 Active 2035-02-04 US10357482B2 (en) 2013-05-03 2014-05-02 Methods providing a therapeutic macromolecule and synthetic nanocarriers comprising immunosuppressant locally and concomitantly to reduce both type I and type IV hypersensitivity
US14/269,058 Active 2034-12-16 US10335395B2 (en) 2013-05-03 2014-05-02 Methods of administering immunosuppressants having a specified pharmacodynamic effective life and therapeutic macromolecules for the induction of immune tolerance
US14/269,042 Pending US20140335186A1 (en) 2013-05-03 2014-05-02 Methods and compositions for enhancing cd4+ regulatory t cells
US16/410,876 Abandoned US20200069659A1 (en) 2013-05-03 2019-05-13 Methods of administering immunosuppressants having a specified pharmacodynamic effective life and therapeutic macromolecules for the induction of immune tolerance
US16/433,622 Pending US20200069660A1 (en) 2013-05-03 2019-06-06 Methods comprising dosing combinations for reducing undesired humoral immune responses
US16/438,147 Active US11298342B2 (en) 2013-05-03 2019-06-11 Methods providing a therapeutic macromolecule and synthetic nanocarriers comprising immunosuppressant locally and concomitantly to reduce both type I and type IV hypersensitivity
US16/550,725 Pending US20200113874A1 (en) 2013-05-03 2019-08-26 Methods related to administering immunosuppressants and therapeutic macromolecules at a reduced pharmacodynamically effective dose
US16/858,349 Pending US20200360350A1 (en) 2013-05-03 2020-04-24 Tolerogenic synthetic nanocarriers to reduce or prevent anaphylaxis in response to a non-allergenic antigen
US17/211,459 Abandoned US20220071968A1 (en) 2013-05-03 2021-03-24 Methods of administering immunosuppressants having a specified pharmacodynamic effective life and therapeutic macromolecules for the induction of immune tolerance
US17/690,437 Pending US20230032226A1 (en) 2013-05-03 2022-03-09 Methods providing a therapeutic macromolecule and synthetic nanocarriers comprising immunosuppressant locally and concomitantly to reduce both type 1 and type iv hypersensitivity

Family Applications Before (9)

Application Number Title Priority Date Filing Date
US14/269,048 Active 2034-09-18 US10668053B2 (en) 2013-05-03 2014-05-02 Tolerogenic synthetic nanocarriers to reduce or prevent anaphylaxis in response to a non-allergenic antigen
US14/269,056 Active 2035-04-20 US10357483B2 (en) 2013-05-03 2014-05-02 Methods comprising dosing combinations for reducing undesired humoral immune responses
US14/269,047 Active 2035-03-28 US10434088B2 (en) 2013-05-03 2014-05-02 Methods related to administering immunosuppressants and therapeutic macromolecules at a reduced pharmacodynamically effective dose
US14/269,054 Active 2035-02-04 US10357482B2 (en) 2013-05-03 2014-05-02 Methods providing a therapeutic macromolecule and synthetic nanocarriers comprising immunosuppressant locally and concomitantly to reduce both type I and type IV hypersensitivity
US14/269,058 Active 2034-12-16 US10335395B2 (en) 2013-05-03 2014-05-02 Methods of administering immunosuppressants having a specified pharmacodynamic effective life and therapeutic macromolecules for the induction of immune tolerance
US14/269,042 Pending US20140335186A1 (en) 2013-05-03 2014-05-02 Methods and compositions for enhancing cd4+ regulatory t cells
US16/410,876 Abandoned US20200069659A1 (en) 2013-05-03 2019-05-13 Methods of administering immunosuppressants having a specified pharmacodynamic effective life and therapeutic macromolecules for the induction of immune tolerance
US16/433,622 Pending US20200069660A1 (en) 2013-05-03 2019-06-06 Methods comprising dosing combinations for reducing undesired humoral immune responses
US16/438,147 Active US11298342B2 (en) 2013-05-03 2019-06-11 Methods providing a therapeutic macromolecule and synthetic nanocarriers comprising immunosuppressant locally and concomitantly to reduce both type I and type IV hypersensitivity

Family Applications After (3)

Application Number Title Priority Date Filing Date
US16/858,349 Pending US20200360350A1 (en) 2013-05-03 2020-04-24 Tolerogenic synthetic nanocarriers to reduce or prevent anaphylaxis in response to a non-allergenic antigen
US17/211,459 Abandoned US20220071968A1 (en) 2013-05-03 2021-03-24 Methods of administering immunosuppressants having a specified pharmacodynamic effective life and therapeutic macromolecules for the induction of immune tolerance
US17/690,437 Pending US20230032226A1 (en) 2013-05-03 2022-03-09 Methods providing a therapeutic macromolecule and synthetic nanocarriers comprising immunosuppressant locally and concomitantly to reduce both type 1 and type iv hypersensitivity

Country Status (15)

Country Link
US (13) US10668053B2 (en)
EP (7) EP2991628A4 (en)
JP (17) JP6818549B2 (en)
KR (12) KR20160003830A (en)
CN (16) CN110841067A (en)
AU (15) AU2014262165B2 (en)
BR (7) BR122020023215B1 (en)
CA (5) CA2910575C (en)
DK (1) DK2991646T3 (en)
EA (6) EA201592107A1 (en)
ES (1) ES2842206T3 (en)
HU (1) HUE052599T2 (en)
IL (14) IL284924B2 (en)
MX (12) MX2015015229A (en)
WO (5) WO2014179773A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11235057B2 (en) 2011-04-29 2022-02-01 Selecta Biosciences, Inc. Methods for providing polymeric synthetic nanocarriers for generating antigen-specific tolerance 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
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
US11633422B2 (en) 2014-09-07 2023-04-25 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector immune responses

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EA022699B1 (en) 2009-05-27 2016-02-29 Селекта Байосайенсиз, Инк. Targeted synthetic nanocarriers with ph sensitive release of immunomodulatory agents
CN107029223A (en) 2010-05-26 2017-08-11 西莱克塔生物科技公司 Synthesize nano-carrier combined vaccine
US9994443B2 (en) 2010-11-05 2018-06-12 Selecta Biosciences, Inc. Modified nicotinic compounds and related methods
KR20140050698A (en) 2011-07-29 2014-04-29 셀렉타 바이오사이언시즈, 인크. Synthetic nanocarriers that generate humoral and cytotoxic t lymphocyte (ctl) immune responses
WO2017025889A1 (en) * 2015-08-13 2017-02-16 Pfizer Inc. Polymeric nanoparticles with dec-205 ligand and co-encapsulating an antigen subject to an autoimmune response and a glucocorticoid receptor agonist
US10744209B2 (en) 2015-11-12 2020-08-18 New York University Biodegradable polymeric nanoparticle conjugates and use thereof
CN109152819A (en) * 2016-03-11 2019-01-04 西莱克塔生物科技公司 The preparation and dosage of Pegylation uricase
JP7066632B2 (en) * 2016-05-09 2022-05-13 アストラゼネカ・アクチエボラーグ Lipid nanoparticles containing lipophilic anti-inflammatory agents and how to use them
US11365392B2 (en) * 2016-08-05 2022-06-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Ex vivo generation of MHCII restricted CD4+ FOXP3+ regulatory T cells and therapeutic uses thereof
JP2019530470A (en) * 2016-08-05 2019-10-24 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル Ex vivo generation of γδ Foxp3 + regulatory T cells and their therapeutic use
JP2020506890A (en) * 2017-01-07 2020-03-05 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. Patterned administration of immunosuppressants coupled to synthetic nanocarriers
EP3694543A1 (en) * 2017-10-13 2020-08-19 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector igm responses
CN108295303A (en) * 2018-02-08 2018-07-20 中山大学附属第三医院(中山大学肝脏病医院) A kind of titanium implantation material and its preparation method and application
CN110314236A (en) * 2018-03-29 2019-10-11 天津键凯科技有限公司 Polyethylene glycol and the conjugate of rapamycin and application thereof
CN109045304B (en) * 2018-04-13 2022-04-05 中山大学 Nucleolar targeting nano-carrier carrying Polymerase I inhibitor and preparation method and application thereof
US20220252574A1 (en) * 2019-07-22 2022-08-11 University Of Louisville Research Foundation, Inc. Immunomodulatory compositions and methods of using
US20220395563A1 (en) * 2019-11-11 2022-12-15 The Regents Of The University Of California Polymeric nanoparticles that target liver sinusoidal endothelial cells to induce antigen-specific immune tolerance
CN110747276B (en) * 2019-11-22 2020-06-30 山东大学齐鲁医院 Application of BACE2 as glioma prognosis/diagnosis/treatment marker
US20210290601A1 (en) * 2020-02-26 2021-09-23 Selecta Biosciences, Inc. Methods and compositions using synthetic nanocarriers comprising immunosuppressant
CA3180166A1 (en) * 2020-04-14 2021-10-21 Selecta Biosciences, Inc. Methods and compositions for inducing autophagy
WO2022022599A1 (en) * 2020-07-28 2022-02-03 浙江大学 Method for amplifying in-vitro induced cd4 +foxp3 +cd69 +treg and use thereof
CN113825832A (en) * 2020-07-28 2021-12-21 浙江大学 Amplification method for in vitro induction of CD4+ Foxp3+ CD69+ Treg and application thereof
CN112480244A (en) * 2020-11-24 2021-03-12 华科同济干细胞基因工程有限公司 Anti-allergic nano antibody composition, antibody determination method and spray
CN112961831B (en) * 2021-02-26 2022-10-11 仲恺农业工程学院 Preparation method of intestine-derived exosome
WO2023086615A1 (en) * 2021-11-14 2023-05-19 Selecta Biosciences, Inc. Multiple dosing with viral vectors

Family Cites Families (226)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1995A (en) 1841-03-03 Latch of door and other locks
US4946929A (en) 1983-03-22 1990-08-07 Massachusetts Institute Of Technology Bioerodible articles useful as implants and prostheses having predictable degradation rates
US4638045A (en) 1985-02-19 1987-01-20 Massachusetts Institute Of Technology Non-peptide polyamino acid bioerodible polymers
US4806621A (en) 1986-01-21 1989-02-21 Massachusetts Institute Of Technology Biocompatible, bioerodible, hydrophobic, implantable polyimino carbonate article
CA1340581C (en) 1986-11-20 1999-06-08 Joseph P. Vacanti Chimeric neomorphogenesis of organs by controlled cellular implantation using artificial matrices
US5736372A (en) 1986-11-20 1998-04-07 Massachusetts Institute Of Technology Biodegradable synthetic polymeric fibrous matrix containing chondrocyte for in vivo production of a cartilaginous structure
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
AU619740B2 (en) 1987-02-24 1992-02-06 Xoma Corporation Immunosuppression in immunotoxin based human therapy
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 (en) 1988-02-24 1997-10-29 株式会社ビーエムジー Polylactic acid microspheres containing physiologically active substance and method for producing the same
US5010167A (en) 1989-03-31 1991-04-23 Massachusetts Institute Of Technology Poly(amide-and imide-co-anhydride) for biological application
US5399665A (en) 1992-11-05 1995-03-21 Massachusetts Institute Of Technology Biodegradable polymers for cell transplantation
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
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
US5565215A (en) 1993-07-23 1996-10-15 Massachusettes Institute Of Technology Biodegradable injectable particles for imaging
TW249754B (en) 1993-10-26 1995-06-21 Alpha I Biomedicals Inc
WO1995031480A1 (en) 1994-05-18 1995-11-23 S.P.I. Synthetic Peptides Incorporated Heterodimer polypeptide immunogen carrier composition and method
US6007845A (en) 1994-07-22 1999-12-28 Massachusetts Institute Of Technology Nanoparticles and microparticles of non-linear hydrophilic-hydrophobic multiblock copolymers
WO1996012406A1 (en) 1994-10-19 1996-05-02 Genetic Therapy, Inc. Gene therapy involving concurrent and repeated administration of adenoviruses and immunosuppressive agents
US5716404A (en) 1994-12-16 1998-02-10 Massachusetts Institute Of Technology Breast tissue engineering
ATE252894T1 (en) 1995-01-05 2003-11-15 Univ Michigan SURFACE-MODIFIED NANOPARTICLES AND METHODS FOR THEIR PRODUCTION AND USE
US6251957B1 (en) 1995-02-24 2001-06-26 Trustees Of The University Of Pennsylvania Method of reducing an immune response to a recombinant virus
US6123727A (en) 1995-05-01 2000-09-26 Massachusetts Institute Of Technology Tissue engineered tendons and ligaments
JP3462313B2 (en) 1995-08-24 2003-11-05 キッコーマン株式会社 Mutant uricase, mutant uricase gene, novel recombinant DNA, and method for producing mutant uricase
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
EP0937082A2 (en) 1996-07-12 1999-08-25 Ariad Pharmaceuticals, Inc. Materials and method for treating or preventing pathogenic fungal infection
WO1998010056A1 (en) 1996-09-03 1998-03-12 Health Research Inc. Treatment of antigen presenting cells to modulate antigen presenting cell function
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
US5837752A (en) 1997-07-17 1998-11-17 Massachusetts Institute Of Technology Semi-interpenetrating polymer networks
US6350590B1 (en) 1997-10-30 2002-02-26 C.B.F. Leti, S.A. 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
JP2001526900A (en) 1997-12-23 2001-12-25 イントロヘーネ ベスローテン フェンノートシャップ Adeno-associated virus and chimeric adenovirus recombinant virus useful for integrating foreign gene information into chromosomal DNA of target cells
AU2064899A (en) 1998-01-09 1999-07-26 Circassia Limited Methods and compositions for desensitisation
US6632922B1 (en) 1998-03-19 2003-10-14 The Regents Of The University Of California Methods and compositions for controlled polypeptide synthesis
US6686446B2 (en) 1998-03-19 2004-02-03 The Regents Of The University Of California Methods and compositions for controlled polypeptide synthesis
US6506577B1 (en) 1998-03-19 2003-01-14 The Regents Of The University Of California Synthesis and crosslinking of catechol containing copolypeptides
SE9801288D0 (en) 1998-04-14 1998-04-14 Astra Ab Vaccine delivery system and method of production
US6306640B1 (en) 1998-10-05 2001-10-23 Genzyme Corporation Melanoma antigenic peptides
US6153217A (en) * 1999-01-22 2000-11-28 Biodelivery Sciences, Inc. Nanocochleate formulations, process of preparation and method of delivery of pharmaceutical agents
DE19951970A1 (en) 1999-10-28 2001-05-03 Bionetworks Gmbh Medicines for tolerance induction
MXPA02008361A (en) 2000-02-28 2004-05-17 Genesegues Inc Nanocapsule encapsulation system and method.
AU2001249182A1 (en) 2000-03-14 2001-09-24 Genetics Institute Inc. Use of rapamycin and agents that inhibit b7 activity in immunomodulation
WO2001085208A2 (en) 2000-05-05 2001-11-15 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
US20040204379A1 (en) 2000-06-19 2004-10-14 Cheng Seng H. Combination enzyme replacement, gene therapy and small molecule therapy for lysosomal storage diseases
US20020095135A1 (en) 2000-06-19 2002-07-18 David Meeker Combination enzyme replacement, gene therapy and small molecule therapy for lysosomal storage diseases
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
CA2319928A1 (en) 2000-09-18 2002-03-18 Vasogen Ireland Limited Apoptosis-mimicking synthetic entities and use thereof in medical treatments
GB0025414D0 (en) * 2000-10-16 2000-11-29 Consejo Superior Investigacion Nanoparticles
AU2002338571A1 (en) 2001-04-11 2002-11-11 Trustees Of The University Of Pennsylvania Compositions and methods for suppressing immune responses
US6913915B2 (en) 2001-08-02 2005-07-05 Phoenix Pharmacologics, Inc. PEG-modified uricase
US6818732B2 (en) 2001-08-30 2004-11-16 The Regents Of The University Of California Transition metal initiators for controlled poly (beta-peptide) synthesis from beta-lactam monomers
CN1294268C (en) 2001-09-03 2007-01-10 上海三维生物技术有限公司 Recombinant adenovirus vector capable of being duplicated and spread specifcally inside tumor cell
PL210795B1 (en) 2001-10-19 2012-03-30 Isotechnika Inc The method of producing the ISATX247 enriched isomer (E), the method of producing the ISATX247 enriched mixture of the (Z) isomer, the method of stereoselective synthesis of the (E) ISATX247 isomer, the method of stereoselective synthesis of the (Z) isomer ISATX247 and the method of producing the mixture of ISATX247 isomers
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
US20040043483A1 (en) 2002-06-04 2004-03-04 Shiguang Qian Novel tolerogenic dendritic cells and therapeutic uses therefor
EP1551221A4 (en) 2002-07-03 2007-08-01 Coley Pharm Group Inc Nucleic acid compositions for stimulating immune responses
US9809654B2 (en) 2002-09-27 2017-11-07 Vaccinex, Inc. Targeted CD1d molecules
US20060127357A1 (en) 2002-11-29 2006-06-15 Roncarolo Maria G Rapamycin and il-10 for the treatment of immune diseases
AU2004224762B2 (en) 2003-03-26 2009-12-24 Kuros Us Llc Packaging of immunostimulatory oligonucleotides into virus-like particles: method of preparation and use
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
AU2004311630A1 (en) 2003-12-02 2005-07-21 Cytimmune Sciences, Inc. Methods and compositions for the production of monoclonal antibodies
DK1704585T3 (en) 2003-12-19 2017-05-22 Univ North Carolina Chapel Hill Methods for preparing isolated micro- and nanostructures using soft lithography or printing lithography
WO2005097116A1 (en) 2004-04-08 2005-10-20 Applied Research Systems Ars Holding N.V. Composition comprising a jnk inhibitor and cyclosporin
JP2008512350A (en) * 2004-07-01 2008-04-24 イェール ユニバーシティ Polymeric substances that are targeted and loaded with drugs at high density
FR2874384B1 (en) 2004-08-17 2010-07-30 Genethon ADENO-ASSOCIATED VIRAL VECTOR FOR PRODUCING EXON JUMP IN A GENE ENCODING A PROTEIN WITH DISPENSABLE DOMAINS
US20090017016A1 (en) * 2004-10-05 2009-01-15 Tanox Inc. Treatment and prevention of hypersensitivity and/or anaphylaxis with anti-ige antibodies in patients receiving replacement therapy
BRPI0518101A (en) 2004-12-31 2008-10-28 Iceutica Pty Ltd methods of production and synthesis of nanoparticulate compositions, compositions obtained and their use
GB0504206D0 (en) * 2005-03-01 2005-04-06 Glaxo Group Ltd Combination therapy
CA2599758A1 (en) 2005-03-08 2006-09-14 Lifecycle Pharma A/S Pharmaceutical compositions comprising sirolimus and/or an analogue thereof
MX2007011494A (en) * 2005-03-17 2007-12-06 Elan Pharma Int Ltd Injectable compositions of nanoparticulate immunosuppressive compounds.
US7884109B2 (en) 2005-04-05 2011-02-08 Wyeth Llc Purine and imidazopyridine derivatives for immunosuppression
KR20080009196A (en) 2005-04-12 2008-01-25 위스콘신 얼럼나이 리서어치 화운데이션 Micelle composition of polymer and passenger drug
US20100062968A1 (en) 2005-05-10 2010-03-11 Bali Pulendran Novel strategies for delivery of active agents using micelles and particles
TW200711649A (en) * 2005-06-17 2007-04-01 Combinatorx Inc Combination therapy for the treatment of immunoinflammatory disorders
EP1932538A4 (en) 2005-08-25 2009-10-21 Taiho Pharmaceutical Co Ltd Biodegradable nanoparticle having t-cell recognizable epitope peptide immobilized thereon or encapsulated therein
CN1979166A (en) * 2005-11-30 2007-06-13 北京有色金属研究总院 Process for nano colloid gold for immune detection and reaction apparatus therefor
AU2006320162B2 (en) 2005-12-02 2013-07-25 The Johns Hopkins University Use of high-dose oxazaphosphorine drugs for treating immune disorders
US20070128289A1 (en) * 2005-12-07 2007-06-07 Zhao Jonathon Z Nano-and/or micro-particulate formulations for local injection-based treatment of vascular diseases
CA2633161A1 (en) 2005-12-08 2007-06-14 University Of Louisville Research Foundation, Inc. Methods and compositions for expanding t regulatory cells
US20100028450A1 (en) 2006-01-25 2010-02-04 The Board Of Trustees Of The University Of Illinoi S Tolerogenic biodegradable artificial antigen presenting system
DK1984007T3 (en) 2006-02-13 2015-12-07 Oncolytics Biotech Inc Application of Low-dose local immunosuppression for amplification of viral oncolytic therapy
US8021689B2 (en) 2006-02-21 2011-09-20 Ecole Polytechnique Federale de Lausanne (“EPFL”) Nanoparticles for immunotherapy
KR20090008290A (en) 2006-03-27 2009-01-21 글로브이뮨 Ras mutation and compositions and methods related thereto
US20070254897A1 (en) 2006-04-28 2007-11-01 Resolvyx Pharmaceuticals, Inc. Compositions and methods for the treatment of cardiovascular disease
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
JP5570808B2 (en) * 2006-08-18 2014-08-13 アルゴス・セラピューティクス・インコーポレーテッド Use of CD83 in combination therapy
US20120269774A1 (en) 2006-09-21 2012-10-25 Medistem Laboratories, Inc Allogeneic stem cell transplants in non-conditioned recipients
JP2010505877A (en) * 2006-10-05 2010-02-25 ザ・ジョンズ・ホプキンス・ユニバーシティー Water-dispersible oral, parenteral and topical formulations for low water-soluble drugs using smart polymer nanoparticles
CN101646418B (en) 2006-10-12 2013-07-17 昆士兰大学 Compositions and methods for modulating immune responses
WO2008043157A1 (en) 2006-10-12 2008-04-17 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
US20100172994A1 (en) 2006-11-22 2010-07-08 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
US9023984B2 (en) 2006-12-29 2015-05-05 The Regents Of The University Of Colorado, A Body Corporate Diagnostic and therapeutic target for autoimmune diseases and uses thereof
WO2008103392A2 (en) 2007-02-21 2008-08-28 Vaccinex, Inc. Modulation of nkt cell activity with antigen-loaded cdid molecules
KR20080078204A (en) 2007-02-22 2008-08-27 크레아젠 주식회사 Mesenchymal stem cell-mediated autologous dendritic cells with increased immunosuppression
KR102154459B1 (en) 2007-03-07 2020-09-09 아브락시스 바이오사이언스, 엘엘씨 Nanoparticle comprising rapamycin and albumin as anticancer agent
CN101730526A (en) * 2007-03-07 2010-06-09 阿布拉科斯生物科学有限公司 Nanoparticle comprising rapamycin and albumin as anticancer agent
AU2008222678B2 (en) 2007-03-07 2013-01-17 The General Hospital Corporation Compositions and methods for the prevention and treatment of autoimmune conditions
WO2008124639A2 (en) * 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Poly (amino acid) targeting moieties
EP2380564B1 (en) 2007-04-04 2014-10-22 Sigmoid Pharma Limited An oral pharmaceutical composition
AU2008236566A1 (en) 2007-04-09 2008-10-16 Chimeros, Inc. Self-assembling nanoparticle drug delivery system
JP2010523656A (en) 2007-04-12 2010-07-15 エモリー・ユニバーシティ A novel strategy for active substance delivery using micelles and particles
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
US20080311140A1 (en) 2007-05-29 2008-12-18 Baylor College Of Medicine Antigen specific immunosuppression by dendritic cell therapy
US20100183602A1 (en) 2007-06-05 2010-07-22 Novartis Ag Induction of Tolerogenic Phenotype in Mature Dendritic Cells
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
BRPI0814688A2 (en) 2007-07-09 2017-06-06 Astrazeneca Ab compound, use of a compound, methods for producing an antiproliferative effect on a warm-blooded animal, and for treating disease, and, pharmaceutical composition
GB0714963D0 (en) 2007-08-01 2007-09-12 Novartis Ag Compositions comprising antigens
BRPI0815395A2 (en) 2007-08-15 2015-02-10 Circassia Ltd COMPOSITION, VECTOR, PRODUCT, PHARMACEUTICAL FORMULATION, IN VITRO METHOD TO DETERMINE IF T-CELLS RECOGNIZE A POLYPEPTIDE, AND IN VITRO METHOD TO DETERMINE IF AN INDIVIDUAL IS OR IS AT RISK.
US20090104114A1 (en) 2007-09-21 2009-04-23 Cytimmune Sciences, Inc. Nanotherapeutic Colloidal Metal Compositions and Methods
CN105770878A (en) * 2007-10-12 2016-07-20 麻省理工学院 Vaccine Nanotechnology
WO2009062502A1 (en) 2007-11-13 2009-05-22 Dandrit Biotech A/S Method for generating tolerogenic dendritic cells employing decreased temperature
US20090142318A1 (en) * 2007-11-30 2009-06-04 Therakos, Inc. METHOD TO EXPAND nTREG CELLS USING p70 S6 KINASE ANTAGONIST
JP2011512326A (en) 2007-12-31 2011-04-21 ナノコア セラピューティクス,インコーポレイテッド RNA interference for the treatment of heart failure
JP5474831B2 (en) 2008-02-08 2014-04-16 テルモ株式会社 Bioactive substance intraluminal controlled drug delivery device and method for producing the same
EP2262489A2 (en) 2008-02-28 2010-12-22 Deutsches Krebsforschungszentrum, Stiftung des öffentlichen Rechts Hollow nanoparticles and uses thereof
CA2722184A1 (en) 2008-04-25 2009-10-29 Duke University Regulatory b cells and their uses
WO2009140626A2 (en) 2008-05-15 2009-11-19 Dynavax Technologies Corporation Long term disease modification using immunostimulatory oligonucleotides
WO2009145238A1 (en) 2008-05-27 2009-12-03 国立大学法人名古屋大学 Immunomodulatory agent and use thereof
MX2010014018A (en) 2008-06-16 2011-06-21 Bind Biosciences Inc Drug loaded polymeric nanoparticles and methods of making and using same.
WO2010005726A2 (en) 2008-06-16 2010-01-14 Bind Biosciences Inc. Therapeutic polymeric nanoparticles with mtor inhibitors and methods of making and using same
US20120034157A1 (en) 2010-08-03 2012-02-09 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Artificial cells
US8821887B2 (en) 2008-08-15 2014-09-02 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 (en) 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 Hydrogels for strengthening and repairing vocal cords and soft tissues
CN101676291B (en) 2008-09-18 2012-05-09 上海海和药物研究开发有限公司 Rapamycin carbonate analog, pharmaceutical composition thereof, and preparation method and uses thereof
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
US8343498B2 (en) 2008-10-12 2013-01-01 Massachusetts Institute Of Technology Adjuvant incorporation in 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 (en) 2008-10-24 2014-07-16 学校法人 聖マリアンナ医科大学 Medicament for treating or preventing HTLV-I related myelopathy and method for testing the effect of antibody therapy on patients with HTLV-I related myelopathy
US20120015899A1 (en) 2008-10-25 2012-01-19 Plant Bioscience, Limited Modified plant virus particles and uses therefor
WO2010075072A2 (en) 2008-12-15 2010-07-01 Bind Biosciences Long circulating nanoparticles for sustained release of therapeutic agents
KR20110117684A (en) 2009-01-20 2011-10-27 노쓰웨스턴 유니버시티 Compositions and methods for induction of antigen-specific tolerance
EP2620447B1 (en) 2009-02-04 2015-09-09 Universität Leipzig Vector(s) containing an inducible gene encoding a CDK4/CDK6 inhibitor useful for treating neurodegenerative disorders
EP2393830B8 (en) 2009-02-05 2015-03-18 Circassia Limited Grass peptides for vaccine
KR20100099849A (en) 2009-03-04 2010-09-15 동국대학교 산학협력단 Composition for treatment of atopic dermatitis comprising immunosuppressant and inhibitors of transglutaminase 2
GB0906159D0 (en) 2009-04-09 2009-05-20 Summit Corp Plc Drug combination for the treatment of proteostatic diseases
BRPI1011836A2 (en) 2009-04-21 2017-05-16 Selecta Biosciences Inc immunotherapeutic agents that provide a th1-induced response
WO2010123501A1 (en) 2009-04-22 2010-10-28 Massachusetts Institute Of Technology Innate immune suppression enables repeated delivery of long rna molecules
CA2760096C (en) 2009-04-27 2018-10-30 Immuron Limited Anti-lps enriched immunoglobulin preparation for use in treatment and/or prophylaxis of a pathologic disorder.
EA022699B1 (en) 2009-05-27 2016-02-29 Селекта Байосайенсиз, Инк. Targeted synthetic nanocarriers with ph sensitive release of immunomodulatory agents
WO2010151823A1 (en) 2009-06-25 2010-12-29 Savient Pharmaceuticals Inc. Methods and kits for predicting infusion reaction risk and antibody-mediated loss of response by monitoring serum uric acid during pegylated uricase therapy
EA201592264A1 (en) 2009-08-26 2016-08-31 Селекта Байосайенсиз, Инк. COMPOSITIONS WHICH INDUCE T-CELL HELPER EFFECT
AR078161A1 (en) 2009-09-11 2011-10-19 Hoffmann La Roche VERY CONCENTRATED PHARMACEUTICAL FORMULATIONS OF AN ANTIBODY ANTI CD20. USE OF THE FORMULATION. TREATMENT METHOD
EP2305277A1 (en) 2009-09-18 2011-04-06 Forskarpatent I Syd AB Use of tolerogenic dendritic cells in treatment and prevention of atherosclerosis
CN101703781A (en) * 2009-10-28 2010-05-12 陕西北美基因股份有限公司 Magnetic medicament loading method for immunosuppressant
KR101267813B1 (en) 2009-12-30 2013-06-04 주식회사 삼양바이오팜 An injectable composition comprising polymeric nanoparticles containing rapamycin with an improved water solubility and a method for preparing the same, and an anticancer composition comprising the same for a combination therapy with radiation
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
WO2011109833A2 (en) 2010-03-05 2011-09-09 President And Fellows Of Harvard College Induced dendritic cell compositions and uses thereof
US20110272836A1 (en) 2010-04-12 2011-11-10 Selecta Biosciences, Inc. Eccentric vessels
US20110262491A1 (en) 2010-04-12 2011-10-27 Selecta Biosciences, Inc. Emulsions and methods of making nanocarriers
JP5904645B2 (en) 2010-05-07 2016-04-13 ゾーマ (ユーエス) リミテッド ライアビリティ カンパニー Methods for the treatment of IL-1β related pathologies
CN107029223A (en) 2010-05-26 2017-08-11 西莱克塔生物科技公司 Synthesize nano-carrier combined vaccine
RU2577278C2 (en) 2010-06-07 2016-03-10 АБРАКСИС БАЙОСАЙЕНС, ЭлЭлСи Methods for combination therapy of proliferative diseases
EP2600878A4 (en) 2010-08-04 2014-06-11 Univ Duke Regulatory b cells and their uses
CA2807942C (en) 2010-08-10 2021-07-27 Ecole Polytechnique Federale De Lausanne Erythrocyte-binding therapeutics
WO2012024621A2 (en) 2010-08-20 2012-02-23 Selecta Biosciences, Inc. Synthetic nanocarrier vaccines comprising peptides obtained or derived from human influenza a virus hemagglutinin
JP2013538211A (en) 2010-08-23 2013-10-10 セレクタ バイオサイエンシーズ インコーポレーテッド Targeted multi-epitope dosage forms that elicit an immune response to an antigen
US9636309B2 (en) 2010-09-09 2017-05-02 Micell Technologies, Inc. Macrolide dosage forms
EP2630493A4 (en) 2010-10-22 2014-05-21 Univ Florida 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
US20120171229A1 (en) 2010-12-30 2012-07-05 Selecta Biosciences, Inc. Synthetic nanocarriers with reactive groups that release biologically active agents
CN103501793A (en) 2011-02-08 2014-01-08 夏洛特-梅克伦堡医院(商业用名:卡罗来纳保健系统) Antisense oligonucleotides
US8654487B2 (en) 2011-03-11 2014-02-18 Siemens Industry, Inc. Methods, systems, and apparatus and for detecting parallel electrical arc faults
BR112013024655A2 (en) 2011-03-25 2016-12-20 Selecta Biosciences Inc synthetic nanotransporters for osmotic mediated release
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US20120301498A1 (en) 2011-04-29 2012-11-29 Selecta Biosciences, Inc. Controlled release of immunosuppressants from synthetic nanocarriers
CN103648501A (en) 2011-05-16 2014-03-19 建新公司 Induction of immune tolerance by using methotrexate
KR20140050698A (en) * 2011-07-29 2014-04-29 셀렉타 바이오사이언시즈, 인크. Synthetic nanocarriers that generate humoral and cytotoxic t lymphocyte (ctl) immune responses
US20130058970A1 (en) 2011-09-06 2013-03-07 Selecta Biosciences, Inc. Induced tolerogenic dendritic cells to reduce systemic inflammatory cytokines
US8865487B2 (en) 2011-09-20 2014-10-21 General Electric Company Large area hermetic encapsulation of an optoelectronic device using vacuum lamination
WO2013052523A1 (en) 2011-10-03 2013-04-11 modeRNA Therapeutics Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
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
WO2013078400A1 (en) 2011-11-22 2013-05-30 The Children's Hospital Of Philadelphia Virus vectors for highly efficient transgene delivery
SG11201404956PA (en) 2012-02-17 2014-09-26 Philadelphia Children Hospital Aav vector compositions and methods for gene transfer to cells, organs and tissues
DK2841097T3 (en) 2012-04-24 2022-11-28 Ohio State Innovation Foundation COMPOSITIONS AND METHODS FOR THE TREATMENT AND PREVENTION OF PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME
CN102871966B (en) 2012-10-19 2013-11-20 东南大学 Nano drug carrier particles for improving bioavailability of rapamycin and preparation method thereof
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
AU2014251099B2 (en) 2013-04-08 2019-01-17 The University Of Kansas Chimeric adeno-associated virus/ bocavirus parvovirus vector
BR122020023215B1 (en) 2013-05-03 2022-11-22 Selecta Biosciences, Inc COMPOSITION AND KIT OF TOLEROGENIC SYNTHETIC NANOCARRIERS TO REDUCE OR PREVENT ANAPHYLAXIS IN RESPONSE TO A NON-ALLERGEN ANTIGEN
CA2910579C (en) 2013-05-03 2023-09-26 Selecta Biosciences, Inc. Dosing combinations for reducing undesired humoral immune responses
MX2015016691A (en) 2013-06-04 2016-04-04 Selecta Biosciences Inc Repeated administration of non-immunosupressive antigen specific immunotherapeutics.
CA2926215A1 (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
US9276815B2 (en) * 2013-12-27 2016-03-01 Dell Products L.P. N-node virtual link trunking (VLT) systems management plane
EP2916319A1 (en) 2014-03-07 2015-09-09 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Concept for encoding of information
US9890365B2 (en) 2014-03-09 2018-02-13 The Trustees Of The University Of Pennsylvania Compositions useful in treatment of ornithine transcarbamylase (OTC) deficiency
GB201407322D0 (en) 2014-04-25 2014-06-11 Ospedale San Raffaele Gene therapy
US20150358333A1 (en) * 2014-06-04 2015-12-10 Grandios Technologies, Llc Geo-location and biometric presence security
US20150359865A1 (en) 2014-06-17 2015-12-17 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for t-cell-mediated autoimmune disease
US20160220501A1 (en) 2015-02-03 2016-08-04 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers to reduce immune responses to therapeutic proteins
EP3160453A1 (en) 2014-06-25 2017-05-03 Selecta Biosciences, Inc. Methods and compositions for treatment with synthetic nanocarriers and immune checkpoint inhibitors
IL292574A (en) 2014-09-07 2022-06-01 Selecta Biosciences Inc Methods and compositions for attenuating anti-viral transfer vector immune responses
CN115212186A (en) 2014-11-05 2022-10-21 西莱克塔生物科技公司 Methods and compositions related to the use of low HLB surfactants in synthetic nanoparticles comprising RAPALOG
WO2017139212A1 (en) 2016-02-08 2017-08-17 Cyta Therapeutics, Inc. Particle delivery of rapamycin to the liver
CN108697815A (en) 2016-02-10 2018-10-23 辉瑞公司 Therapeutic nano particle and its preparation and application with EGFR ligands
CN109152819A (en) 2016-03-11 2019-01-04 西莱克塔生物科技公司 The preparation and dosage of Pegylation uricase
WO2018039465A1 (en) 2016-08-25 2018-03-01 Selecta Biosciences, Inc. Polyester polymer matrices for the delivery of allergens
CA3038089A1 (en) 2016-09-27 2018-04-05 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
JP2020506890A (en) 2017-01-07 2020-03-05 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. Patterned administration of immunosuppressants coupled to synthetic nanocarriers
JP2020510687A (en) 2017-03-11 2020-04-09 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. Methods and compositions relating to combination treatment with synthetic nanocarriers including anti-inflammatory and immunosuppressive agents
EP3694543A1 (en) 2017-10-13 2020-08-19 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector igm responses
US20210128534A1 (en) 2018-02-26 2021-05-06 Yissum Research Development Company Of The Hebrew University Of Jerusalem Ltd. Drug delivery systems
CN112771070A (en) 2018-07-16 2021-05-07 西莱克塔生物科技公司 Methods and compositions of OTC constructs and vectors
WO2020018587A1 (en) 2018-07-16 2020-01-23 Selecta Biosciences, Inc. Methods and compositions of mma constructs and vectors
KR20220004121A (en) 2019-04-28 2022-01-11 셀렉타 바이오사이언시즈, 인크. Methods of treating a subject with pre-existing immunity to a viral transfer vector
BR112021023594A2 (en) 2019-05-28 2022-02-08 Selecta Biosciences Inc Methods and compositions for attenuated antiviral transfer vector immune response
CA3138071A1 (en) 2019-06-04 2020-12-10 Selecta Biosciences, Inc. Formulations and doses of pegylated uricase
JP2022553345A (en) 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 Methods and compositions for treating liver disease and disorders
EP4054531A1 (en) 2019-11-08 2022-09-14 Selecta Biosciences, Inc. Formulations and doses of pegylated uricase

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11235057B2 (en) 2011-04-29 2022-02-01 Selecta Biosciences, Inc. Methods for providing polymeric synthetic nanocarriers for generating antigen-specific tolerance immune responses
US11717569B2 (en) 2011-04-29 2023-08-08 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers
US11779641B2 (en) 2011-04-29 2023-10-10 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for allergy therapy
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
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

Also Published As

Publication number Publication date
MX2021005697A (en) 2021-07-07
JP7440833B2 (en) 2024-02-29
BR112015027280A8 (en) 2018-01-30
MX2021001878A (en) 2021-04-19
JP6760838B2 (en) 2020-09-23
IL282045A (en) 2021-05-31
ES2842206T3 (en) 2021-07-13
WO2014179772A2 (en) 2014-11-06
AU2014262166B2 (en) 2020-02-27
AU2014262155A1 (en) 2015-11-19
AU2020200565A1 (en) 2020-02-13
CA2910578A1 (en) 2014-11-06
JP2019089784A (en) 2019-06-13
IL284732B (en) 2022-12-01
BR112015027291A2 (en) 2017-07-25
CN111068061A (en) 2020-04-28
BR112015027280A2 (en) 2017-07-25
IL276205B (en) 2021-04-29
AU2023202955A1 (en) 2023-06-15
AU2014262162B2 (en) 2019-11-07
US10335395B2 (en) 2019-07-02
AU2019250117A1 (en) 2019-11-07
MX2015015225A (en) 2016-07-06
JP7134885B2 (en) 2022-09-12
AU2020200065A1 (en) 2020-01-30
AU2014262165B2 (en) 2019-10-31
KR20160003214A (en) 2016-01-08
BR112015027281A2 (en) 2017-07-25
US20200360350A1 (en) 2020-11-19
BR122020023215B1 (en) 2022-11-22
BR112015027288A8 (en) 2019-12-24
US10434088B2 (en) 2019-10-08
WO2014179769A2 (en) 2014-11-06
IL292190A (en) 2022-06-01
MX2015015223A (en) 2016-07-05
IL274852B (en) 2021-07-29
IL284732B2 (en) 2023-04-01
AU2022201641A1 (en) 2022-03-31
JP2022122871A (en) 2022-08-23
KR20220025907A (en) 2022-03-03
US20140328924A1 (en) 2014-11-06
IL282045B (en) 2022-04-01
EP3763359A1 (en) 2021-01-13
CA2910584A1 (en) 2014-11-06
DK2991646T3 (en) 2021-01-11
AU2014262166A1 (en) 2015-11-19
EP2991630A4 (en) 2016-11-23
EP2991685A2 (en) 2016-03-09
CN105338968A (en) 2016-02-17
BR112015027288A2 (en) 2017-07-25
CN105307641A (en) 2016-02-03
CN105283175A (en) 2016-01-27
CN110639024A (en) 2020-01-03
JP2016518392A (en) 2016-06-23
MX2015015221A (en) 2016-07-06
JP2020015741A (en) 2020-01-30
CN105339012A (en) 2016-02-17
BR112015027297A8 (en) 2018-01-30
CA2910578C (en) 2024-02-27
MX2015015229A (en) 2016-07-06
KR20220025906A (en) 2022-03-03
EP2991628A4 (en) 2016-11-16
EA201592106A3 (en) 2016-08-31
US20230032226A1 (en) 2023-02-02
JP6580558B2 (en) 2019-09-25
AU2022200157A1 (en) 2022-02-10
US20140328854A1 (en) 2014-11-06
BR112015027297A2 (en) 2017-07-25
KR20220025909A (en) 2022-03-03
EA201592102A1 (en) 2016-10-31
IL242273B (en) 2018-10-31
BR112015027279A8 (en) 2018-01-30
BR112015027281B1 (en) 2022-03-15
US20200078341A1 (en) 2020-03-12
IL284924A (en) 2021-08-31
AU2022203142A1 (en) 2022-06-02
JP2023039981A (en) 2023-03-22
WO2014179773A2 (en) 2014-11-06
IL270133B (en) 2021-08-31
CN111686255A (en) 2020-09-22
AU2020203308A1 (en) 2020-07-09
BR112015027281A8 (en) 2019-12-24
US20200069660A1 (en) 2020-03-05
CN105263478A (en) 2016-01-20
JP2022025069A (en) 2022-02-09
JP2020045343A (en) 2020-03-26
EP2991630A2 (en) 2016-03-09
EA201592104A2 (en) 2016-05-31
JP2021073198A (en) 2021-05-13
BR112015027279A2 (en) 2017-09-26
AU2014262163A1 (en) 2015-11-19
WO2014179762A1 (en) 2014-11-06
KR20220025911A (en) 2022-03-03
AU2019250117B2 (en) 2021-10-14
AU2020200065B2 (en) 2022-02-17
EP2991646A4 (en) 2016-11-02
JP2016517890A (en) 2016-06-20
JP6983144B2 (en) 2021-12-17
MX2021012348A (en) 2021-11-04
IL242216B (en) 2019-11-28
JP7028556B2 (en) 2022-03-02
JP6818549B2 (en) 2021-01-20
CN110841066A (en) 2020-02-28
EA201592106A2 (en) 2016-05-31
CA2910575A1 (en) 2014-11-06
EP2991631A1 (en) 2016-03-09
AU2014262165A1 (en) 2015-11-19
EP2991646A2 (en) 2016-03-09
EA201592107A1 (en) 2016-10-31
BR112015027288B1 (en) 2022-03-15
EA201592105A1 (en) 2016-10-31
JP2016524599A (en) 2016-08-18
KR20160003215A (en) 2016-01-08
US20140335186A1 (en) 2014-11-13
JP2020073488A (en) 2020-05-14
JP7427419B2 (en) 2024-02-05
EP2991646B1 (en) 2020-10-07
KR20220025910A (en) 2022-03-03
IL274852A (en) 2020-07-30
US20140328923A1 (en) 2014-11-06
JP2022115998A (en) 2022-08-09
IL242272B (en) 2020-06-30
MX2022009538A (en) 2022-09-09
US10668053B2 (en) 2020-06-02
US20220071968A1 (en) 2022-03-10
KR20160003830A (en) 2016-01-11
AU2022203540A1 (en) 2022-06-16
CN110841067A (en) 2020-02-28
US10357483B2 (en) 2019-07-23
KR20160003217A (en) 2016-01-08
US20200069659A1 (en) 2020-03-05
IL276205A (en) 2020-09-30
IL242271B (en) 2020-11-30
CN110251676A (en) 2019-09-20
IL284924B (en) 2022-12-01
JP2022043071A (en) 2022-03-15
KR20220025908A (en) 2022-03-03
EA201592104A3 (en) 2016-09-30
JP2016520051A (en) 2016-07-11
CN111760025A (en) 2020-10-13
BR112015027291A8 (en) 2019-12-24
US20140328921A1 (en) 2014-11-06
IL242270B (en) 2019-11-28
CA2910583C (en) 2023-05-23
JP2016517891A (en) 2016-06-20
MX2015015230A (en) 2016-06-06
US20140328922A1 (en) 2014-11-06
CA2910583A1 (en) 2014-11-06
JP2016517889A (en) 2016-06-20
MX2021001982A (en) 2021-04-28
US11298342B2 (en) 2022-04-12
IL284732A (en) 2021-08-31
IL270135B (en) 2022-05-01
EP2991629A1 (en) 2016-03-09
EP2991685A4 (en) 2016-11-16
CN111053908A (en) 2020-04-24
EP2991629A4 (en) 2016-11-16
HUE052599T2 (en) 2021-05-28
CA2910575C (en) 2023-10-24
CN110251675A (en) 2019-09-20
JP2019069964A (en) 2019-05-09
CN110694077A (en) 2020-01-17
KR20160003212A (en) 2016-01-08
AU2014262162A1 (en) 2015-11-19
CN105338979A (en) 2016-02-17
EP2991631A4 (en) 2016-11-16
WO2014179770A1 (en) 2014-11-06
US10357482B2 (en) 2019-07-23
KR20160003219A (en) 2016-01-08
IL291394A (en) 2022-05-01
BR112015027297B1 (en) 2023-01-10
CA2910417C (en) 2023-10-03
AU2020200824A1 (en) 2020-02-20
EA201592103A3 (en) 2016-08-31
MX2015015231A (en) 2016-07-06
EP2991628A1 (en) 2016-03-09
CA2910417A1 (en) 2014-11-06
MX2021001879A (en) 2021-04-19
EA201592103A2 (en) 2016-04-29
IL284924B2 (en) 2023-04-01

Similar Documents

Publication Publication Date Title
AU2020200065B2 (en) Tolerogenic synthetic nanocarriers and therapeutic macromolecules for reduced or enhanced pharmacodynamic effects
AU2020203312B2 (en) Dosing combinations for reducing undesired humoral immune responses

Legal Events

Date Code Title Description
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: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

Free format text: FINAL REJECTION MAILED

AS Assignment

Owner name: SELECTA BIOSCIENCES, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MALDONADO, ROBERTO A.;KISHIMOTO, TAKASHI KEI;SIGNING DATES FROM 20140529 TO 20140604;REEL/FRAME:061239/0139

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: NON FINAL ACTION MAILED

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED