WO2014179771A1 - Associations de dosages destinées à réduire les réponses immunitaires à médiation humorale non souhaitées - Google Patents

Associations de dosages destinées à réduire les réponses immunitaires à médiation humorale non souhaitées Download PDF

Info

Publication number
WO2014179771A1
WO2014179771A1 PCT/US2014/036698 US2014036698W WO2014179771A1 WO 2014179771 A1 WO2014179771 A1 WO 2014179771A1 US 2014036698 W US2014036698 W US 2014036698W WO 2014179771 A1 WO2014179771 A1 WO 2014179771A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
synthetic nanocarriers
therapeutic
factor
dosing
Prior art date
Application number
PCT/US2014/036698
Other languages
English (en)
Other versions
WO2014179771A8 (fr
Inventor
Roberto A. MALDONADO
Takashi Kei Kishimoto
Original Assignee
Selecta Biosciences, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to EA201592102A priority Critical patent/EA201592102A1/ru
Application filed by Selecta Biosciences, Inc. filed Critical Selecta Biosciences, Inc.
Priority to KR1020157034104A priority patent/KR20160003830A/ko
Priority to KR1020227004761A priority patent/KR20220025910A/ko
Priority to BR112015027281-9A priority patent/BR112015027281B1/pt
Priority to EP14791089.7A priority patent/EP2991628A4/fr
Priority to JP2016512092A priority patent/JP6760838B2/ja
Priority to CN201480031937.3A priority patent/CN105307641A/zh
Priority to AU2014262164A priority patent/AU2014262164B2/en
Priority to CA2910579A priority patent/CA2910579C/fr
Priority to MX2015015229A priority patent/MX2015015229A/es
Publication of WO2014179771A1 publication Critical patent/WO2014179771A1/fr
Priority to IL242271A priority patent/IL242271B/en
Publication of WO2014179771A8 publication Critical patent/WO2014179771A8/fr
Priority to AU2020203312A priority patent/AU2020203312B2/en
Priority to IL276205A priority patent/IL276205B/en
Priority to IL282045A priority patent/IL282045B/en
Priority to IL291394A priority patent/IL291394A/en
Priority to AU2022231724A priority patent/AU2022231724A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • 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
    • 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
    • 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

Definitions

  • This invention relates to doses of therapeutic macromolecules administered concomitantly with immunosuppressants, such as those attached to synthetic nanocarriers, in combination with doses of therapeutic macromolecules alone, and related methods.
  • the compositions and methods allow for efficient reduction of undesired humoral immune responses. Such undesired immune responses can neutralize the efficacy of the therapeutic treatment or cause hypersensitive reactions to the therapeutic.
  • the compositions and methods provided thus, can be used for subjects in which the administration of a therapeutic macromolecule thatresults in undesired humoral immune responses.
  • 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 may be reduced through the use of immunosuppressant drugs.
  • Conventional immunosuppressant drugs are broad-acting, and the use of broad- acting immunosuppressants are 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 (1) a first dosing that comprises concomitantly administering (a) therapeutic macromolecules that are not attached to any synthetic nanocarriers, and (b) immunosuppressants, such as those attached to synthetic nanocarriers, and that comprise no therapeutic macromolecule antigen-presenting cell (APC) presentable antigens of the therapeutic macromolecules; (2) a second dosing that comprises (c) administering the therapeutic macromolecules that are not attached to any synthetic nanocarriers, and not administering any synthetic nanocarriers; and (3) administering the first and second dosings to a subject according to an administration schedule that reduces an undesired humoral immune response to the therapeutic macromolecules is provided.
  • a first dosing that comprises concomitantly administering (a) therapeutic macromolecules that are not attached to any synthetic nanocarriers, and (b) immunosuppressants, such as those attached to synthetic nanocarriers, and that comprise no therapeutic macromolecule antigen-presenting cell (APC) presentable antigens of the therapeutic
  • the method further comprises (4) determining the administration schedule for the first and second dosings that reduces an undesired humoral immune response to the therapeutic macromolecules.
  • the undesired humoral immune response to the therapeutic macromolecules results from the second dosing without the first dosing.
  • the first dosing comprises administering (a) and (b) to the subject one or more times. In another embodiment of any one of the methods provided herein, (a) and (b) are administered at least 1, 2, 3, 4 or 5 times.
  • the second dosing is administered to the subject at least 1, 2, 3, 4, 5, 6, 7 or 8 weeks after the first dosing.
  • the method further comprises assessing the undesired humoral immune response in the subject prior to and/or after the administration of the first dosing and second dosing.
  • the administering of the first dosing and/or second dosing is by intravenous, intraperitoneal or subcutaneous administration.
  • the method further comprises identifying the subject as having or at risk of having an undesired humoral immune response to the therapeutic macromolecules.
  • the subject is expected or suspected to have an undesired humoral immune response to the therapeutic macromolecules.
  • compositions comprising (1) one or more first doses that each comprise (a) therapeutic macromolecules that are not attached to any synthetic nanocarriers, and (b) immunosuppressants, such as those attached to a population of synthetic nanocarriers and that comprise no therapeutic macromolecule APC presentable antigens of the therapeutic macromolecules; and (2) one or more second doses that comprises therapeutic macromolecules that are not attached to any synthetic nanocarriers is provided.
  • the composition is for use in a method of reducing an undesired humoral immune response to the therapeutic
  • the method comprises administering the first and second doses to a subject according to an administration schedule. In another embodiment, the method further comprises determining the administration schedule for the first and second doses that reduce an undesired humoral immune response to the therapeutic macromolecules. In another embodiment, the method is any method as provided herein.
  • the second dose does not comprise any synthetic nanocarriers.
  • the composition is a kit and one or more of the first doses and one or more of the second doses are each housed in a container in the kit.
  • the composition further comprises a pharmaceutically acceptable carrier.
  • the immunosuppressants comprise 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 phosphodiesterasse 4 inhibitor, an HDAC inhibitor or a proteasome inhibitor.
  • the mTOR inhibitor is rapamycin.
  • the therapeutic macromolecules are therapeutic proteins. In another embodiment of any one of the methods or compositions provided herein, the therapeutic macromolecules are therapeutic polynucleotides. In another embodiment of any one of the methods or compositions provided herein, the therapeutic proteins are for protein replacement or protein supplementation therapy. In another embodiment of any one of the methods or compositions provided herein, the therapeutic macromolecules comprise infusible or injectable therapeutic proteins, enzymes, enzyme cofactors, hormones, blood or blood coagulation factors, cytokines, interferons, growth factors, monoclonal antibodies, polyclonal antibodies or proteins associated with Pompe' s disease.
  • the infusible or injectable therapeutic proteins comprise Tocilizumab, alpha- 1 antitrypsin, Hematide, albinterferon alfa-2b, Rhucin, tesamorelin, ocrelizumab, belimumab, pegloticase, pegsiticase, taliglucerase alfa, agalsidase alfa or velaglucerase alfa.
  • the enzymes comprise an oxidoreductase, transferase, hydrolase, lyase, isomerase or ligase.
  • the enzymes comprise 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 a- glucosidase (GAA), alglucosidase alfa, LUMIZYME, MYOZYME, arylsulfatase B, laronidase, ALDURAZYME, idursulfase, ELAPRASE, arylsulfatase B or NAGLAZYME.
  • the enzymes comprise KRYSTEXXA (pegloticase) or pegsiticase.
  • the monoclonal antibodies comprises HUMIRA (adalimumab).
  • the cytokines comprise a lymphokine, interleukin, chemokine, type 1 cytokine or a type 2 cytokine.
  • the blood or blood coagulation factors comprise 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 factors comprise Factor VIII.
  • a load of the immunosuppressants on average across the population of synthetic nanocarriers is between 0.1% and 50%. In another embodiment of any one of the methods or compositions provided herein, the load of the immunosuppressants on average across the population of synthetic nanocarriers is between 0.1% and 20%.
  • the synthetic nanocarriers of the population comprise lipid nanoparticles, polymeric nanoparticles, metallic nanoparticles, surfactant-based emulsions, dendrimers, buckyballs, nanowires, virus-like particles or peptide or protein particles.
  • the synthetic nanocarriers of the population comprise lipid nanoparticles.
  • the synthetic nanocarriers of the population comprise liposomes.
  • the synthetic nanocarriers of the population comprise metallic nanoparticles.
  • the metallic nanoparticles comprise gold nanoparticles.
  • the synthetic nanocarriers of the population comprise polymeric nanoparticles.
  • the polymeric nanoparticles comprise polymer that is a non-methoxy- terminated, pluronic polymer.
  • the polymeric nanoparticles comprise a polyester, polyester coupled 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 coupled 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 of the population is a diameter greater than lOOnm. In another embodiment of any one of the methods or compositions provided herein, the diameter is greater than 150nm. In another embodiment of any one of the methods or compositions provided herein, the diameter is greater than 200nm. In another embodiment of any one of the methods or compositions provided herein, the diameter is greater than 250nm. In another embodiment of any one of the methods or compositions provided herein, the diameter is greater than 300nm. In another embodiment of any one of the methods or compositions provided herein, an aspect ratio of the synthetic nanocarriers of the population 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 one or more doses or dosage forms of a therapeutic macromolecule and producing one or more doses or dosage forms of an immunosuppressant.
  • the step of producing one or more doses or dosage forms of an immunosuppressant comprises attaching the immunosuppressant to synthetic nanocarriers.
  • the method further comprises combining the one or more doses or dosage forms of the immunosuppressant and one or more doses or dosage forms of the therapeutic
  • the composition or kit comprises one or more doses or dosage forms comprising an immunosuppressant and one or more doses or dosage forms comprising a therapeutic macromolecule, wherein the immunosuppressant and therapeutic macromolecule are administered according to any one of the method provided herein.
  • the immunosuppressant is attached to synthetic nanocarriers.
  • the immunosuppressant comprises no therapeutic macromolecule antigen-presenting cell (APC) presentable antigens of the therapeutic macromolecule.
  • the composition or kit further comprises one or more doses or dosage forms comprising the therapeutic macromolecule for use as one or more second dosings.
  • any one of the compositions or kits provided herein are provided for use in any one of the comprising an immunosuppressant and one or more doses or dosage forms comprising a therapeutic macromolecule.
  • the composition or kit further comprises one or more doses or dosage forms comprising the therapeutic macromolecule for use as one or more second dosings.
  • the immunosuppressant is attached to synthetic nanocarriers.
  • the immunosuppressant comprises no therapeutic macromolecule antigen-presenting cell (APC) presentable antigens of the therapeutic macromolecule.
  • APC therapeutic macromolecule antigen-presenting cell
  • Fig. 1A and B show the efficacy of nanocarrier and FVIII dosing in Hemophilia A mice.
  • Fig. 2 shows the antibody recall response to FVIII one month following the final nanocarrier and FVIII dosing.
  • Fig. 3A and B show immune responses to HUMIRA in mice that were treated with HUMIRA /adalimumab with or without nanocarriers attached to rapamycin.
  • Fig. 4 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. 5 shows anti-ovalbumin (OVA) antibody titers in mice that were treated with OVA with or without nanocarriers attached to rapamycin.
  • OVA anti-ovalbumin
  • Fig. 6 shows anti-KRYSTEXXA antibody titers in mice that were treated with
  • Fig. 7 shows antibody titers in mice that were treated with OVA and KLH either in the presence or absence of nanocarriers attached to rapamycin.
  • Fig. 8A and B show immune responses to KLH in mice that were treated with KLH with or without nanocarriers attached to rapamycin.
  • Fig. 9A and B show immune responses to HUMIRA /adalimumab in mice that were treated with HUMIRA /adalimumab with or without nanocarriers attached to rapamycin.
  • Fig. 10 shows immune responses to PEG-KLH in mice treated with or without nanocarriers attached to rapamycin.
  • Fig. 11 shows immune and therapeutic responses with HUMIRA with or without nanocarriers attached to rapamycin.
  • Fig. 12 shows an administration schedule
  • Fig. 13 demonstrates the effects of synthetic nanocarriers attached to rapamycin or nanocarriers attached to GSK1059615 administered concomitantly with encapsulated protein.
  • 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 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.
  • any recited integer e.g. a feature, element, characteristic, property, method/process step or limitation
  • group of integers e.g. features, element, characteristics, properties, method/process steps or limitations
  • the invention is useful for subjects in need of treatment with therapeutic macromolecules against which undesired humoral immune responses are generated or are expected to be generated.
  • the present invention prevents or suppresses undesired humoral immune responses that may neutralize the beneficial effect of certain therapeutic macromolecule treatments.
  • the inventors have unexpectedly and surprisingly discovered that the problems and limitations noted above can be overcome by practicing the invention disclosed herein.
  • the inventors have unexpectedly discovered that it is possible to provide immunosuppressant and therapeutic macromolecule dosings in certain combinations that reduce undesired humoral immune responses specific to the therapeutic macromolecule, particularly in embodiments wherein the immunosuppressant compositions and the therapeutic macromolecules are administered concomitantly followed by administration of the therapeutic macromolecules without synthetic nanocarriers.
  • 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.
  • administering schedule refers to administration of first dosings and second dosings according to a determined schedule.
  • the schedule can include the number of first and second doings as well as the frequency of such dosings or interval between dosings.
  • Such an administration schedule may include a number of parameters that are varied to achieve a particular objective, preferably reduction of an undesired humoral immune response to a therapeutic macromolecule.
  • the administration schedule is any of the administration schedules as provided below in the Examples.
  • administration schedules according to the invention may be used to administer first and second dosings to one or more test subjects. Immune responses in these test subjects can then be assessed to determine whether or not the schedule was effective in reducing an undesired humoral immune response.
  • Whether or not a schedule had a desired effect can be determined using any of the methods provided herein or otherwise known in the art.
  • a sample may be obtained from a subject to which dosings provided herein have been administered according to a specific administration schedule 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.
  • Useful methods for determining the level of antibody production are well known in the art and include the assays provided herein. Such assays include ELISA assays.
  • an amount effective in the context of a composition or dosage form for administration to a subject refers to an amount of the composition or dosage form that produces one or more desired immune responses in the subject, for example, a reduction in an undesired humoral immune response. Therefore, in some embodiments, an amount effective is any amount of a composition or dosage form provided herein that reduces an undesired humoral immune response. 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 as provided herein, such as one 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. Amounts effective, preferably, result in a reduction in an undesired humoral immune response in a subject specific to a therapeutic macromolecule. Amounts effective, can also result in a tolerogenic immune response in a subject to an antigen, such as a therapeutic macromolecule. In other embodiments, the amounts effective can involve enhancing the level of a desired response, such as a therapeutic endpoint or result. The achievement of any of the foregoing can be monitored by routine methods.
  • the amount effective is one in which the desired immune 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.
  • the amount effective is one which produces a measurable desired immune response, for example, a measurable decrease in a humoral immune response (e.g. , to a specific antigen), 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,
  • 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.
  • 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 an immune response” refers to any measurement or determination of the level, presence or absence, reduction, increase in, etc. of an immune response in vitro or in vivo. Such measurements or determinations may be performed on one or more samples obtained from a subject. Such assessing can be performed with any of the methods provided herein or otherwise known in the art.
  • attaching 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.
  • 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.
  • 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 humoral immune response as provided herein and would benefit from the compositions and methods provided.
  • the subjects are those that are expected to have an undesired humoral immune response to a therapeutic macromolecule.
  • 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
  • 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
  • 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
  • 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);
  • non-physically associated combined materials/agents examples include:
  • material e.g. , a non-unitary formulation
  • material 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
  • material 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;
  • 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.
  • each of the two or more materials/agents in a combination therapy may also differ with respect to the route of administration.
  • Conscomitantly means administering two or more materials/agents to a subject in a manner that is correlated in time, preferably sufficiently correlated in time so as to provide a modulation in an immune or physiologic response, and even more preferably the two or more materials/agents are administered in combination.
  • 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 an immune or physiologic response, and even more preferably the two or more materials/agents are administered in combination.
  • 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, 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 or administration schedule 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.
  • Dosing means the administration of a pharmacologically and/or immunologically active material or combination of pharmacologically and/or immunologically active materials to a subject.
  • 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
  • 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 an immune or physiologic response (e.g. , a tolerogenic immune response) to occur, either directly oneself or indirectly.
  • an action such as an immune or physiologic response (e.g. , a 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.
  • the identified subject is one who is in need of a therapeutic benefit from a therapeutic macromolecule and in which an undesired humoral immune response is expected to occur as provided herein.
  • 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
  • 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 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.
  • the immunosuppressant is one that causes an APC to promote a regulatory phenotype in one or more immune effector cells.
  • the regulatory phenotype may be characterized by the inhibition of the production, induction, stimulation or recruitment of antigen- specific CD4+ T cells or B cells, the inhibition of the production of antigen- specific antibodies, the production, induction, stimulation or recruitment of Treg cells (e.g., CD4+CD25highFoxP3+ Treg cells), etc. This may be the result of the conversion of CD4+ T cells or B cells to a regulatory phenotype. This may also be the result of induction of FoxP3 in other immune cells, such as CD8+ T cells, macrophages and iNKT cells.
  • the immunosuppressant is one that affects the response of the APC after it processes an antigen. 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- ⁇ signaling agents; TGF- ⁇ receptor agonists; histone deacetylase inhibitors, such as Trichostatin A; corticosteroids; inhibitors of mitochondrial function, such as rotenone; P38 inhibitors; NF- ⁇ inhibitors, such as 6Bio, Dexamethasone, TCPA-1, IKK VII; adenosine receptor agonists; prostaglandin E2 agonists (PGE2), such as Misoprostol; phosphodiesterase inhibitors, such as phosphodiesterase 4 inhibitor (PDE4), such as Rolipram; proteasome inhibitors; kinase inhibitors; G-protein coupled receptor agonists; G-protein coupled receptor antagonists; glucocorticoids; retinoids; cytokine inhibitors; cytokine receptor inhibitors; cytokine receptor activators
  • 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, nifhimic 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 indirectly i.e., after being processed in some way after
  • 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), biologies- 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;
  • a drug nanocrystal refers to a form of a drug (e.g., an immunosuppressant) that does not include a carrier or matrix material.
  • drug nanocrystals comprise 90%, 95%, 98%, or 99% or more drug.
  • Methods for producing drug nanocrystals include, without limitation, milling, high pressure homogenization, precipitation, spray drying, rapid expansion of supercritical solution
  • 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.
  • administration of a first dosing comprising therapeutic macromolecules and immunosuppressants in nanocrystalline form followed by a second dosing of the therapeutic macromolecule results in a reduction of the undesired therapeutic macromolecule- specific humoral immune response.
  • the load of the immunosuppressant on average across the synthetic nanocarriers is between 0.1% and 99%. In another embodiment, the load is between 0.1% and 50%. In yet another embodiment, the load of the immunosuppressant is between 0.1% and 20%. In a further embodiment, the load of the immunosuppressant is between 0.1% and 10%. In still a further embodiment, the load of the immunosuppressant is between 1% and 10%. In still a further embodiment, the load of the immunosuppressant is between 7% and 20%.
  • the load of the immunosuppressant 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% or at least 20%, at least 25%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% on average across the population of synthetic nanocarriers.
  • the load of the immunosuppressant 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 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 ⁇ .
  • 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 ⁇ , more preferably equal to or less than 2 ⁇ , more preferably equal to or less than 1 ⁇ , more preferably equal to or less than 800 nm, more preferably equal to or less than 600 nm, and more preferably still equal to or less than 500 nm.
  • a minimum dimension of at least 75%, preferably at least 80%, more preferably at least 90%, of the synthetic nanocarriers in a sample, based on the total number of synthetic nanocarriers in the sample is equal to or greater than 100 nm, more preferably equal to or greater than 120 nm, more preferably equal to or greater than 130 nm, more preferably equal to or greater than 140 nm, and more preferably still equal to or greater than 150 nm.
  • Measurement of synthetic nanocarrier dimensions e.g., effective diameter
  • a suspension of synthetic nanocarriers can be diluted from an aqueous buffer into purified water to achieve a final synthetic nanocarrier suspension concentration of approximately 0.01 to 0.1 mg/mL.
  • the diluted suspension may be prepared directly inside, or transferred to, a suitable cuvette for DLS analysis.
  • the cuvette may then be placed in the DLS, allowed to equilibrate to the controlled temperature, and then scanned for sufficient time to acquire a stable and reproducible distribution based on appropriate inputs for viscosity of the medium and refractive indicies of the sample. The effective diameter, or mean of the distribution, is then reported.
  • Determining the effective sizes of high aspect ratio, or non- spheroidal, synthetic nanocarriers may require augmentative techniques, such as electron microscopy, to obtain more accurate measurements.
  • "Dimension” or “size” or “diameter” of synthetic nanocarriers means the mean of a particle size distribution, for example, obtained using dynamic light scattering.
  • 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.
  • 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.
  • Subject means animals, including warm blooded mammals such as humans and primates; avians; domestic household or farm animals such as cats, dogs, sheep, goats, cattle, horses and pigs; laboratory animals such as mice, rats and guinea pigs; fish; reptiles; zoo and wild animals; and the like.
  • Synthetic nanocarrier(s) means a discrete object that is not found in nature, and that possesses at least one dimension that is less than or equal to 5 microns in size. Albumin nanoparticles are generally included as synthetic nanocarriers, however in certain
  • the synthetic nanocarriers do not comprise albumin nanoparticles. In embodiments, 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, viruslike 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 US Patent 5,543,158 to Gref et al., (2) the polymeric nanoparticles of Published US Patent Application 20060002852 to Saltzman et al., (3) the lithographically constructed nanoparticles of Published US Patent Application
  • 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 humoral immune responses, including production of anti-therapeutic
  • 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
  • therapeutic macromolecule antigen-presenting cell 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 a 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, 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.
  • one or more tolerogenic immune responses specific to the therapeutic macromolecule result with the methods, compositions or kits provided herein.
  • 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 humoral immune response refers to any undesired humoral 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 humoral 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 are specific to a therapeutic macromolecule and counteract the beneficial effects desired of administration with the therapeutic macromolecule.
  • first and second dosings that in combination can reduce undesired humoral immune responses.
  • first and second dosings can also result in improved efficacy of therapeutic macromolecules.
  • the methods and related compositions provided herein, therefore, can be used for subjects in need of treatment with a therapeutic macromolecule.
  • first dosings comprise immunosuppressants, in some embodiments attached to synthetic nanocarriers, in combination with therapeutic
  • macromolecules that are not attached to synthetic nanocarriers, and second dosings comprise therapeutic macromolecules that are also not attached to synthetic nanocarriers.
  • synthetic nanocarriers can be used to attach to immunosuppressants of the first dosings.
  • 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. 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.).
  • a non- polymeric core e.g., metal particle, quantum dot, ceramic particle, bone particle, viral particle, proteins, nucleic acids, carbohydrates, 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.).
  • lipid membrane e.g., lipid bilayer, lipid monolayer, etc.
  • 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 (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 phosphatidylinositol
  • sphingomyelin phosphatidylethanolamine (cephalin); cardiolipin
  • phosphatidic acid cerebrosides
  • dicetylphosphate dipalmitoylphosphatidylglycerol
  • amphiphilic entity component may be a mixture of different amphiphilic entities. Those skilled in the art will recognize that this is an exemplary, not comprehensive, list of substances with surfactant activity. Any amphiphilic entity may be used in the production of synthetic nanocarriers to be used in accordance with the present invention.
  • synthetic nanocarriers may optionally comprise one or more carbohydrates.
  • Carbohydrates may be natural or synthetic.
  • a carbohydrate may be a derivatized natural carbohydrate.
  • a carbohydrate comprises monosaccharide or disaccharide, including but not limited to glucose, fructose, galactose, ribose, lactose, sucrose, maltose, trehalose, cellbiose, mannose, xylose, arabinose, glucoronic acid, galactoronic acid, mannuronic acid, glucosamine, galatosamine, and neuramic acid.
  • a carbohydrate is a polysaccharide, including but not limited to pullulan, cellulose, microcrystalline cellulose, hydroxypropyl methylcellulose (HPMC), hydroxycellulose (HC), methylcellulose (MC), dextran, cyclodextran, glycogen,
  • 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.
  • 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 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 the synthetic nanocarriers. This bonding can result in the immunosuppressants being attached to the surface of the synthetic nanocarriers and/or contained (encapsulated) within the synthetic nanocarriers. In some embodiments, however, the immunosuppressants are encapsulated by the synthetic nanocarriers as a result of the structure of the synthetic nanocarriers rather than bonding to the synthetic nanocarriers.
  • the synthetic nanocarrier comprises a polymer as provided herein, and the immunosuppressants are attached to the polymer.
  • a coupling moiety can be any moiety through which an immunosuppressant is bonded to a synthetic nanocarrier.
  • moieties include covalent bonds, such as an amide bond or ester bond, as well as separate molecules that bond (covalently or non-covalently) the immunosuppressant to the synthetic nanocarrier.
  • molecules include linkers or polymers or a unit thereof.
  • the coupling moiety can comprise a charged polymer to which an
  • 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 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.
  • hydrophobic interactions e.g., hydrophobic interactions, charge interactions, van der Waals forces, etc.
  • 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(l,3-dioxan-2one)), polyanhydrides (e.g. poly(sebacic anhydride)), polypropylfumerates, polyamides (e.g. polycaprolactam), polyacetals, polyethers, polyesters (e.g., polylactide, polyglycolide, polylactide-co-glycolide, polycaprolactone, polyhydroxyacid (e.g.
  • polymers in accordance with the present invention include polymers which have been approved for use in humans by the U.S. Food and Drug
  • polyesters e.g., polylactic acid, poly(lactic-co-glycolic acid), polycaprolactone, polyvalerolactone, poly(l,3-dioxan-2one)
  • polyanhydrides e.g., poly(sebacic anhydride)
  • polyethers e.g., polyethylene glycol
  • polyurethanes polymethacrylates; polyacrylates; and
  • polymers can be hydrophilic.
  • polymers may comprise anionic groups (e.g., phosphate group, sulphate group, carboxylate group); cationic groups (e.g., quaternary amine group); or polar groups (e.g., hydroxyl group, thiol group, amine group).
  • a synthetic nanocarrier comprising a hydrophilic polymeric matrix generates a hydrophilic environment within the synthetic nanocarrier.
  • polymers can be hydrophobic.
  • a synthetic nanocarrier comprising a hydrophobic polymeric matrix generates a hydrophobic
  • hydrophobicity of the polymer may have an impact on the nature of materials that are incorporated (e.g. , 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
  • 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,
  • a polymer may be PLGA.
  • PLGA is a biocompatible and biodegradable co-polymer of lactic acid and glycolic acid, and various forms of PLGA are characterized by the ratio of lactic acid:glycolic acid.
  • Lactic acid can be L-lactic acid, D- lactic acid, or D,L-lactic acid.
  • the degradation rate of PLGA can be adjusted by altering the lactic acid:glycolic acid ratio.
  • PLGA to be used in accordance with the present invention is characterized by a lactic acid:glycolic acid ratio of approximately 85: 15, approximately 75:25, approximately 60:40, approximately 50:50, approximately 40:60, approximately 25:75, or approximately 15:85.
  • polymers may be one or more acrylic polymers.
  • acrylic polymers include, for example, acrylic acid and methacrylic acid copolymers, methyl methacrylate copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, aminoalkyl methacrylate copolymer, poly(acrylic acid), poly(methacrylic acid), methacrylic acid alkylamide copolymer, poly(methyl methacrylate), poly(methacrylic acid anhydride), methyl methacrylate, polymethacrylate, poly(methyl methacrylate) copolymer, polyacrylamide, aminoalkyl methacrylate copolymer, glycidyl methacrylate copolymers, polycyanoacrylates, and combinations comprising one or more of the foregoing polymers.
  • the acrylic polymer may comprise fully-polymerized copolymers of acrylic and methacrylic acid esters with a low content of quaternary ammonium groups.
  • 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;
  • 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
  • synthetic nanocarriers do not comprise a polymeric
  • 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).
  • First and/or second dosings according to the invention can comprise pharmaceutically acceptable excipients, such as preservatives, buffers, saline, or phosphate buffered saline.
  • the compositions may be made using conventional pharmaceutical manufacturing and compounding techniques to arrive at useful dosage forms.
  • the compositions of the dosings are suspended in sterile saline solution for injection together with a preservative.
  • synthetic nanocarriers are suspended in sterile saline solution for injection together with a preservative.
  • immunosuppressants 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 such as an immunosuppressant with the carboxylic acid group of a second component such as the nanocarrier.
  • the amide bond in the linker can be made using any of the conventional amide bond forming reactions with suitably protected amino acids and activated carboxylic acid such N-hydroxysuccinimide-activated ester.
  • a disulfide linker is made via the formation of a disulfide (S-S) bond between two sulfur atoms of the form, for instance, of R1-S-S-R2.
  • a disulfide bond can be formed by thiol exchange of a component containing thiol/mercaptan group(-SH) with another activated thiol group 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 , wherein Rl 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.
  • 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 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 preferably an immunosuppressant
  • a negative charged 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 antagonists; peroxisome prolifer
  • 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 ® , TOR VAST ® ), cerivastatin, fluvastatin (LESCOL ® , LESCOL ® XL), lovastatin (MEVACOR ® , ALTOCOR ® ,
  • ALTOPREV ® mevastatin (COMPACTIN ® ), pitavastatin (LIVALO ® , PIAVA ® ), rosuvastatin (PRAVACHOL ® , SELEKTINE ® , LIPOSTAT ® ), rosuvastatin (CRESTOR ® ), and simvastatin (ZOCOR ® , LIPEX ® ).
  • mTOR inhibitors examples include rapamycin and analogs thereof (e.g., CCL-779,
  • 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)
  • atractyloside dipotassium salt
  • bongkrekic acid triammonium salt
  • carbonyl cyanide m-chlorophenylhydrazone e.g., from Atractylis gummifera
  • CGP-37157 e.g., from CGP-37157
  • P38 inhibitors examples include SB-203580 (4-(4-Fluorophenyl)-2-(4- methylsulfinylphenyl)-5-(4-pyridyl)lH-imidazole), SB-239063 (trans- 1- (4hydroxycyclohexyl)-4-(fluorophenyl)-5-(2-methoxy-pyrimidin-4-yl) imidazole), SB- 220025 (5-(2amino-4-pyrimidinyl)-4-(4-fluorophenyl)-l-(4-piperidinyl)imidazole)), and ARPvY-797.
  • NF e.g., ⁇ - ⁇
  • NF e.g., ⁇ - ⁇
  • NF e.g., ⁇ - ⁇
  • examples of NF (e.g., ⁇ - ⁇ ) inhibitors include IFRD1, 2-(l,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]
  • NFKB Activation Inhibitor III NF- ⁇ Activation Inhibitor II
  • JSH-23 parthenolide
  • 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.
  • adenosine receptor agonists examples include CGS-21680 and ATL-146e.
  • prostaglandin E2 agonists examples include E-Prostanoid 2 and E-Prostanoid 4.
  • phosphodiesterase inhibitors include caffeine, aminophylline, IB MX (3-isobutyl-l-methylxanthine), paraxanthine, pentoxifylline, theobromine, theophylline, methylated xanthines, vinpocetine, EHNA
  • DALIRESPTM sildenafil
  • VIAGRA ® tadalafil
  • ADCIRCA ® CIALIS ®
  • vardenafil LEVITRA ®
  • STAXYN ® udenafil
  • icariin 4- methylpiperazine
  • pyrazolo pyrimidin-7-1 4- methylpiperazine
  • proteasome inhibitors examples include bortezomib, disulfiram, epigallocatechin- 3-gallate, and salinosporamide A.
  • kinase inhibitors examples include bevacizumab, BIBW 2992, cetuximab
  • ERP Error UX ®
  • imatinib GLEEVEC ®
  • trastuzumab HERCEPTIN ®
  • gefitinib IRESSA ®
  • ranibizumab LCENTIS ®
  • pegaptanib sorafenib
  • dasatinib sunitinib
  • erlotinib nilotinib
  • lapatinib panitumumab
  • panitumumab panitumumab
  • vandetanib E7080
  • pazopanib pazopanib
  • glucocorticoids examples include hydrocortisone (Cortisol), cortisone acetate, prednisone, prednisolone, methylprednisolone, dexamethasone, betamethasone,
  • retinoids include retinol, retinal, tretinoin (retinoic acid, RETIN-A ), isotretinoin (ACCUTANE ® , AMNESTEEM ® , CLARA VIS ® , SOTRET ® ), alitretinoin (PANRETIN ® ), etretinate (TEGISONTM) and its metabolite acitretin (S ORIAT ANE ® ) , tazarotene (TAZORAC ® , AVAGE ® , ZORAC ® ), bexarotene (TARGRETIN ® ), and adapalene (DIFFERIN ® ).
  • retinoids include retinol, retinal, tretinoin (retinoic acid, RETIN-A ), isotretinoin (ACCUTANE ® , AMNESTEEM ® , CLARA VIS ® , SOTRET ® ), alitret
  • cytokine inhibitors examples include ILlra, 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, PPARy antagonist III, G335, and T0070907 (EMD4Biosciences, USA).
  • peroxisome proliferator-activated receptor agonists examples include pioglitazone, ciglitazone, clofibrate, GW1929, GW7647, L-165,041, LY 171883, PPARy activator, Fmoc- Leu, troglitazone, and WY- 14643 (EMD4Biosciences, USA).
  • histone deacetylase inhibitors examples 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 (PXDIOI), LAQ824, and panobinostat (LBH589), benzamides such as entinostat (MS-275), CI994, and mocetinostat (MGCD0103), nicotinamide, derivatives of NAD, dihydrocoumarin, naphthopyranone, and 2-hydroxynaphaldehydes.
  • SAHA vorinostat
  • PXDIOI belinostat
  • LAQ824 panobinostat
  • benzamides such as entinostat (MS-275), CI994, and mocetinostat (MGCD0103), nicotin
  • calcineurin inhibitors examples include cyclosporine, pimecrolimus, voclosporin, and tacrolimus.
  • phosphatase inhibitors examples include BN82002 hydrochloride, CP-91149, calyculin A, cantharidic acid, cantharidin, cypermethrin, ethyl-3,4-dephostatin, fostriecin sodium salt, MAZ51, methyl-3,4-dephostatin, NSC 95397, norcantharidin, okadaic acid ammonium salt from prorocentrum concavum, okadaic acid, okadaic acid potassium salt, okadaic acid sodium salt, phenylarsine oxide, various phosphatase inhibitor cocktails, protein phosphatase 1C, protein phosphatase 2A inhibitor protein, protein phosphatase 2A1, protein phosphatase 2A2, and sodium ortho vanadate.
  • 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
  • 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 cc-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,
  • Gaucher' s disease e.g., CEREZYMETM
  • NAGLAZYMETM NAGLAZYMETM
  • pegloticase KRYSTEXXA
  • pegsiticase 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 examples include Melatonin (N-acetyl-5-methoxytryptamine),
  • Serotonin Thyroxine (or tetraiodo thyronine) (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, Gonadotrop
  • blood or blood coagulation factors examples include Factor I (fibrinogen), Factor II
  • 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-a), Transforming growth factor beta(TGF-P), Tumour_ne
  • monoclonal antibodies examples 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,
  • 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,
  • Nimotuzumab Nofetumomab merpentan, Ocrelizumab, Odulimomab, Ofatumumab,
  • Olaratumab Omalizumab
  • Oportuzumab monatox Oregovomab
  • Otelixizumab Otelixizumab
  • Ramucirumab Ranibizumab, Raxibacumab, Regavirumab Reslizumab, Rilotumumab,
  • Sonepcizumab Sontuzumab, Stamulumab, Sulesomab, Tacatuzumab tetraxetan, Tadocizumab, Talizumab, Tanezumab, Taplitumomab paptox, Tefibazumab, Telimomab aritox, Tenatumomab, Teneliximab, Teplizumab, Ticilimumab (tremelimumab),
  • Tigatuzumab 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-a antibodies.
  • infusion therapy or injectable therapeutic proteins examples include, for example, Tocilizumab (Roche/ Actemra®), alpha- 1 antitrypsin (Kamada/AAT), Hematide® (Affymax and Takeda, synthetic peptide), albinterferon alfa-2b (Novartis/ZalbinTM), Rhucin®
  • 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 FougeroUes et al. and in Published US Patent application 2012/0251618 to Schrum et al.
  • siRNAs small interfering RNAs
  • a component such as a therapeutic macromolecule or immunosuppressant
  • 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
  • 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.
  • An administration schedule can be determined by varying the number of first dosing(s) and second dosing(s) and/or the length of time between the first dosing(s) and second dosing(s) and assessing an undesired humoral immune response to a therapeutic macromolecule. For example, after administering first dosing(s) and second dosing(s) an undesired humoral immune response to a therapeutic macromolecule can be measured. This undesired humoral immune response can be compared to the same type of immune response that occurs without the first and second dosing(s), such as when only one or more dosings of therapeutic macromolecule has occurred without concomitant administration with
  • an administration schedule can be beneficial for subjects in need of treatment with a therapeutic macromolecule and can be used with the methods and compositions of the invention provided herein.
  • Administration schedules may be determined by starting with a test schedule of first dosing(s) and second dosing(s) and using known scaling techniques (such as allometric or isometric scaling) as appropriate.
  • the administration schedule may be determined by testing various schedules in a subject, e.g. , through direct experimentation based on experience and guiding data.
  • 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.
  • 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 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
  • synthetic nanocarriers can be attached to components directly or indirectly via non-covalent interactions.
  • the non- covalent coupling is mediated by non-covalent interactions including but not limited to charge interactions, affinity interactions, metal coordination, physical adsorption, host-guest interactions, hydrophobic interactions, TT stacking interactions, hydrogen bonding interactions, van der Waals interactions, magnetic interactions, electrostatic interactions, dipole-dipole interactions, and/or combinations thereof.
  • Such couplings may be arranged to be on an external surface or an internal surface of a synthetic nanocarrier.
  • encapsulation and/or absorption is a form of coupling.
  • 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
  • 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
  • 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 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 noninfectious, 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.
  • 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, according to the invention.
  • Doses of dosage forms may contain varying amounts of therapeutic macromolecules, according to the invention.
  • the amount of respective components present in the dosage forms can be varied according to the nature of the components, the therapeutic benefit to be accomplished, and other such parameters.
  • dose ranging studies can be conducted to establish optimal therapeutic amounts of the components to be present in the dosage forms.
  • the components are present in the dosage forms in an amount effective to reduce an undesired humoral immune response to the therapeutic macromolecule upon administration to a subject.
  • Dosage forms may be administered at a variety of frequencies (i.e. , according to an administration schedule). In a preferred embodiment, at least one administration of the dosage forms 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.
  • kits comprises one or more first doses and/or one or more second doses as provided herein.
  • Each of the doses of a kit can be contained within separate containers or within the same container in the kit.
  • the container is a vial or an ampoule.
  • each of the doses can be contained within a solution separate from the container, such that the dose may be added to a container at a subsequent time.
  • the doses 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.
  • kit further comprises one or more syringes.
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, MA 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, AL 35211 ; Product Code 7525 DLG 7A).
  • 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.
  • nanocarriers were washed by transferring the nanocarrier suspension to a centrifuge tube and centrifuging at 75,600xg 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.
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, MA 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, AL 35211; Product Code 5050 DLG 2.5A).
  • 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.
  • 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
  • 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, AL 35211; Product Code 100 DL mPEG 5000 5CE).
  • 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 IX 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.
  • the mice were challenged with second dosings of FVIII (i.v.
  • the inhibitor titer was determined by Bethesda assay using a chromogenic FVIII activity assay kit (Coatest SP4 FVIII), as presented in Fig. 2.
  • the data show that first dosings of immunosuppressant attached to synthetic nanocarriers with FVIII reduced the anti-Factor FVIII antibody response.
  • the data also show that such administration can result in improved activity of Factor VIII. This demonstrates the reduction of an undesired humoral immune response against a protein as well as improved efficacy with first and second dosings as provided herein.
  • the data also demonstrate an administration schedule that reduces an undesired humoral response to Factor VIII.
  • 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 dO, 7, 14, 22, 29, across all groups and conditions).
  • Another group received similar injections, but 0.9 mg of synthetic nanocarrier attached to rapamycin (Nanocarrier ID 1, 2 or 3) were admixed to the solution of HUMIRA on the priming day 0 (first dosings).
  • the results presented in Fig. 3A 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 (second dosings).
  • the animals received another challenge in one hind limb (a further second dosing) 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. 3B. Similar to the antibody results, the treatment with the synthetic nanocarriers abolished the inflammatory response induced by local administration of HUMIRA.
  • first and second dosings can reduce undesired anti-HUMIRA antibody response as well as eliminate undesired inflammatory reactions that can result from the administration of HUMIRA.
  • This demonstrates the reduction of undesired humoral immune responses against a protein with first and second dosings as provided herein.
  • the data also demonstrate an administration schedule that reduces an undesired humoral response to HumiraTM.
  • 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 dO, 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 synthetic nanocarriers attached to rapamycin
  • first and second dosings can reduce undesired anti-KLH antibody responses that can result from the administration of KLH.
  • This demonstrates the reduction of undesired humoral immune responses against a protein with first and second dosings as provided herein.
  • the data also demonstrate an administration schedule that reduces an undesired humoral response to KLH.
  • first and second dosings can reduce undesired 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 undesired humoral immune responses against a protein with first and second dosings as provided herein.
  • the data also demonstrate an administration schedule that reduces an undesired humoral response to OVA.
  • Example 6 Evaluating Undesired Humoral Immune 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 synthetic 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 with 20 ⁇ g of CpG s.c. in the hind limb (dO, 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. 6 show that treating animals with synthetic nanocarriers attached to rapamycin administered concomitantly with KRYSTEXXA in the same solution (first dosings) 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 -i-CpG without the synthetic nanocarriers (second dosings).
  • first and second dosings can reduce undesired anti-KRYSTEXXA antibody response, and such a reduction can remain even after being administered KRYSTEXXA in combination with a strong adjuvant.
  • This demonstrates the reduction of undesired humoral immune responses against a protein with first and second dosings as provided herein.
  • the data also demonstrate an administration schedule that reduces an undesired humoral response to KrystexxaTM.
  • 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 (dO, 7, 14, 20, 28, 35, 42, 49).
  • the other group of animals received pOVA and KLH in the same routes and amounts but admixed with 0.2mg of synthetic nanocarriers 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. 7 demonstrate that 3 doses of synthetic nanocarriers attached to rapamycin administered concomitantly with pOVA in the same solution (first dosings) 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 (second dosings).
  • 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 (dO, 7, 14, 21, 28, 35, 42, 49, 56, 63).
  • KLH Keyhole Limpet Hemocyanin
  • the other group received similar injections but 0.9 mg of synthetic 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. 8A 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. 8B.
  • first and second dosings can reduce undesired anti-KLH antibody responses as well as eliminate undesired anaphylactic reactions.
  • This demonstrates the reduction of undesired humoral immune responses against a protein with first and second dosings as provided herein.
  • the data also demonstrate an
  • 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 TNFa antibody HUMIRA
  • 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 synthetic 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) (first dosings) followed by 3 injections of HUMIRA alone (same amount) (day 49 to 63, weeks 13 to 15 of age) (second dosings).
  • the results in Fig. 9A show the antibody titers in the blood of all animals at day 21.
  • 9B 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.
  • Mesoporous Si02 nanoparticle cores are created through a sol-gel process.
  • CTAB Hexadecyltrimethyl-ammonium bromide
  • 2 M aqueous NaOH solution 3.5 mL
  • 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 Si02 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 ⁇ > 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 3-aminopropyl)- triethoxysilane (APTMS).
  • APTMS 50 ⁇ > 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
  • 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 Si02 nanoparticles formed above are first mixed with the gold seeds for 2 h at room temperature.
  • the gold- decorated Si02 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.
  • l,2-Dipalmitoyl-sn-glycero-3- phosphocholine (DPPC) (32 ⁇ ), cholesterol (32 ⁇ ), and cyclosporin A (6.4 ⁇ ) 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 30s 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 ⁇ of HS-PEG-rapamycin (10 ⁇ 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 12,000g for 30 minutes. The supernatant is decanted and the pellet containing AuNC-S- PEG-rapamycin is then pellet washed with lx PBS buffer.
  • the purified Gold-PEG-rapamycin nanocarriers are then resuspended in suitable buffer for further analysis and bioassays.
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, MA).
  • PLA-PEG-OMe block copolymer 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, AL 35211; Product Code 100 DL mPEG 5000 5CE).
  • 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.
  • nanocarriers were washed by transferring the nanocarrier suspension to a centrifuge tube and centrifuging at 75,600xg 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.
  • C57BL/6 age-matched (5-6 weeks) females were injected i.v. in the tail vein weekly (days 0, 7, 14, 21, 28, 35, 42, 49) with 250 ⁇ g of a conjugate of keyhole limpet hemocyanin and polyethylene glycol (KLH-PEG) with (first dosings) (days 0, 7, 14, 21, 28) or without 0.47mg of rapamycin-containing nanocarriers for the first 5 injections (NP[Rapa], 50 ⁇ g of rapamycin content).
  • the following 3 injections consisted of just KLH-PEG (second dosings) in the same amounts.
  • the IgM antibody response to PEG was monitored weekly (days 12, 20, 34, 40, 47, 54) in the blood of these animals.
  • Rapamycin-containing nanocarriers were generated using the materials and methods described above in Example 13.
  • 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 Rapamycin Content
  • Anti-HUMIRA immune and neutralizing responses in arthritic animals were evaluated.
  • Transgenic animals overexpressing human tumor necrosis factor alpha were evaluated.
  • HuTNFccTg develop progressive rheumatoid arthritis over the course of 20 weeks from birth. This process can be prevented by using the fully human anti-human TNFcc antibody Adalimumab or HUMIRA.
  • HUMIRA fully human anti-human TNFcc antibody
  • ADA anti-drug antibody formation
  • Humira (20C ⁇ g) can be injected to overcome this antagonizing immune response to allow for the therapeutic effect of Humira.
  • 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).
  • 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 developing titers (week 10).
  • Fig. 12 illustrates the dosing regimen.
  • Fig. 11 (right panel)
  • the scores 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.
  • Rapamycin was purchased from TSZ CHEM (185 Wilson Street, Framingham, MA 01702), product code RIO 17.
  • 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, AL 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, AL 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, NJ 08027), product code 1.41350.
  • Cellgro phosphate buffered saline IX (PBS IX) 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
  • 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).
  • 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, NJ 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, AL 35211), product code 5050 DLG 2.5A).
  • Phosphate-buffered saline IX 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 lOmM 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 lOOmM phosphate buffer, pH 8.
  • a primary (Wl/O) emulsion was first created by mixing Solutions 1 through 3.
  • Solution 1 (0.2 mL), Solution 2 (0.75 mL), and Solution 3 (0.25mL) 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 (W1/0/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 IX (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 IX. This washing procedure was repeated and then the pellet was re-suspended in PBS IX to achieve a nanocarrier suspension having a nominal concentration of 10 mg/mL on a polymer basis. The suspension was stored frozen at -20C until use. Effective Diameter Ovalbumin Content
  • GSK1059615 was purchased from MedChem Express (11 Deer Park Drive, Suite
  • 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, AL 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, AL 35211; Product Code 100 DL mPEG 5000 5K-E).
  • Cellgro phosphate buffered saline IX pH 7.4 (PBS IX) 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 IX.
  • the washing procedure was repeated, and the pellet was re-suspended in PBS IX 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 ⁇ 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 351nm.
  • 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 1.2mg of rapamycin-containing nanocarriers (NP[Rapa])(first dosings) or 8mg of GSK1059615-loaded nanocarriers (NP[GSK1059615]).
  • Ovalbumin-loaded nanocarriers NP[OVA]
  • NP[Rapa] rapamycin-containing nanocarriers
  • NP[GSK1059615] 8mg of GSK1059615-loaded nanocarriers
  • Example 16 Inventive Dosings Using Synthetic Nanocarriers and
  • the synthetic nanocarriers of Example 11 and HUMIRA are prepared for use in a first dosing in human subjects at risk of developing anti-drug antibody responses to HUMIRA.
  • the first dosing comprises 3 concomitant q2 week administrations of HUMIRA (40 mg) and synthetic nanocarriers of Example 11 (50 mg).
  • the second dosing comprises 3 q2week
  • HUMIRA HUMIRA alone (without the synthetic nanocarriers).
  • An administration schedule is generated in which the first dosing is followed by the second dosing.
  • the human subjects are evaluated for anti-HUMIRA antibodies (using standard ELISA techniques)
  • Example 17 Inventive Dosings Using Synthetic Nanocarriers
  • nanocarrier ID 1 The synthetic nanocarriers of Example 1, nanocarrier ID 1 are prepared. They are combined with HUMIRA and administered in first dosings and second dosings administered concomitantly according to the following schedules, as shown in Table 1:
  • Example 18 Inventive Dosings Using Synthetic Nanocarriers and mmRNA (Prophetic)
  • mmRNA de Fougerolles et al.
  • the first dosing comprises 3 concomitant q2week administrations of mmRNA (100 ng) and synthetic nanocarriers of
  • Example 11 (50 mg).
  • the second dosing comprises 3 q2week administrations of mmRNA alone (without the synthetic nanocarriers).
  • An administration schedule is generated in which the first dosing is followed by the second dosing.
  • the human subjects are evaluated for anti- mmRNA antibodies (using standard ELISA techniques) before the first dosing and following the second dosing. Any difference between the two anti-mmRNA antibody readouts is noted.
  • mice C57BL/6 age-matched (5-6 weeks) female mice are injected i.v. in the tail vein on days -21 and -14 with saline (No Treatment) or 1.1 mg of whole Ovalbumin and 1.2mg of nanocrystalline rapamycin (first dosings).
  • first dosings At day 0 all animals are injected s.c. in the hind limbs with a first dosing of 25 ⁇ g of particulate OVA (pOVA) admixed to 2 ⁇ g of CpG followed by injections of just of 25 ⁇ g pOVA on days 7 and 14 (second dosings).
  • pOVA particulate OVA
  • Antibody titers are measured on day 21. In the absence of any treatment, the animals develop a robust immune response against OVA that can be measured by the anti-OVA IgG antibody titers.
  • a reduction in OVA- specific IgG antibodies in the animals that received OVA in combination with nanocrystalline rapamycin indicates that the nanocrystal-form of the immunosuppressant when concomitantly delivered with a protein can prevent antibody formation to that protein for multiple challenges and periods of time.
  • Example 20 Inventive Dosings Using Nanocrystalline Immunosuppressants and HUMIRA/adalimumab (Prophetic)
  • Nanocrystalline rapamycin is purchased. The nanocrystalline rapamycin is combined with HUMIRA and administered in first dosings and second dosings administered concomitantly according to the following schedules, as shown in Table 1.
  • Example 21 Inventive Dosings Using Nanocrystalline Immunosuppressants and mmRNA (Prophetic)
  • Nanocrystalline rapamycin is purchased and mmRNA encoding asparaginase (made generally according to prepared according to US Patent application 2013/0115272 to de Fougerolles et al. ("mmRNA") ) are prepared for use in a first dosing in human subjects at risk of developing anti-drug antibody responses to mmRNA.
  • the first dosing comprises 3 concomitant q2week administrations of mmRNA (100 ng) and nanocrystalline rapamycin (50 mg).
  • the second dosing comprises 3 q2week administrations of mmRNA alone (without the nanocrystalline rapamycin).
  • An administration schedule is generated in which the first dosing is followed by the second dosing.
  • the human subjects are evaluated for anti-mmRNA antibodies (using standard ELISA techniques) before the first dosing and following the second dosing. Any difference between the two anti-mmRNA antibody readouts is noted.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Nanotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Biomedical Technology (AREA)
  • Optics & Photonics (AREA)
  • Inorganic Chemistry (AREA)
  • Ceramic Engineering (AREA)
  • Immunology (AREA)
  • Dispersion Chemistry (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

L'invention concerne des dosages de macromolécules thérapeutiques et d'immunosuppresseurs, qui, dans certains modes de réalisation, sont fixés à des nanovecteurs synthétiques, en association avec des dosages de macromolécules thérapeutiques sans nanovecteurs synthétiques, ainsi que des méthodes associées permettant de réduire les réponses immunitaires à médiation humorale.
PCT/US2014/036698 2013-05-03 2014-05-02 Associations de dosages destinées à réduire les réponses immunitaires à médiation humorale non souhaitées WO2014179771A1 (fr)

Priority Applications (16)

Application Number Priority Date Filing Date Title
AU2014262164A AU2014262164B2 (en) 2013-05-03 2014-05-02 Dosing combinations for reducing undesired humoral immune responses
CN201480031937.3A CN105307641A (zh) 2013-05-03 2014-05-02 用于降低不期望体液免疫应答的给药组合
KR1020157034104A KR20160003830A (ko) 2013-05-03 2014-05-02 바람직하지 않은 체액성 면역 반응을 감소시키기 위한 투약 조합물
KR1020227004761A KR20220025910A (ko) 2013-05-03 2014-05-02 바람직하지 않은 체액성 면역 반응을 감소시키기 위한 투약 조합물
BR112015027281-9A BR112015027281B1 (pt) 2013-05-03 2014-05-02 Composições compreendendo macromoléculas terapêuticas e uma população de nanocarreadores sintéticos que são fixados a imunossupressores
EP14791089.7A EP2991628A4 (fr) 2013-05-03 2014-05-02 Associations de dosages destinées à réduire les réponses immunitaires à médiation humorale non souhaitées
JP2016512092A JP6760838B2 (ja) 2013-05-03 2014-05-02 望ましくない液性免疫応答を低減するための投薬の組み合わせ
EA201592102A EA201592102A1 (ru) 2013-05-03 2014-05-02 Дозирование комбинаций для снижения нежелательных гуморальных иммунных ответов
MX2015015229A MX2015015229A (es) 2013-05-03 2014-05-02 Combinaciones de dosificaciones para reducir las respuestas inmunes humorales no deseadas.
CA2910579A CA2910579C (fr) 2013-05-03 2014-05-02 Associations de dosages destinees a reduire les reponses immunitaires a mediation humorale non souhaitees
IL242271A IL242271B (en) 2013-05-03 2015-10-26 Dose combinations to reduce unwanted humoral immune responses
AU2020203312A AU2020203312B2 (en) 2013-05-03 2020-05-21 Dosing combinations for reducing undesired humoral immune responses
IL276205A IL276205B (en) 2013-05-03 2020-07-21 Dose combinations to reduce unwanted humoral immune responses
IL282045A IL282045B (en) 2013-05-03 2021-04-04 Dose combinations to reduce unwanted humoral immune responses
IL291394A IL291394A (en) 2013-05-03 2022-03-15 Dose combinations to reduce unwanted humoral immune responses
AU2022231724A AU2022231724A1 (en) 2013-05-03 2022-09-15 Dosing combinations for reducing undesired humoral immune responses

Applications Claiming Priority (15)

Application Number Priority Date Filing Date Title
US201361819517P 2013-05-03 2013-05-03
US61/819,517 2013-05-03
US201361881851P 2013-09-24 2013-09-24
US201361881913P 2013-09-24 2013-09-24
US201361881921P 2013-09-24 2013-09-24
US61/881,913 2013-09-24
US61/881,851 2013-09-24
US61/881,921 2013-09-24
US201361907117P 2013-11-21 2013-11-21
US61/907,117 2013-11-21
US61/907,177 2013-11-21
US201461948313P 2014-03-05 2014-03-05
US201461948384P 2014-03-05 2014-03-05
US61/948,313 2014-03-05
US61/948,384 2014-03-05

Publications (2)

Publication Number Publication Date
WO2014179771A1 true WO2014179771A1 (fr) 2014-11-06
WO2014179771A8 WO2014179771A8 (fr) 2015-11-12

Family

ID=51844007

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/036698 WO2014179771A1 (fr) 2013-05-03 2014-05-02 Associations de dosages destinées à réduire les réponses immunitaires à médiation humorale non souhaitées

Country Status (3)

Country Link
AU (3) AU2014262164B2 (fr)
CA (1) CA2910579C (fr)
WO (1) WO2014179771A1 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017156513A1 (fr) * 2016-03-11 2017-09-14 Selecta Biosciences, Inc. Formulations et doses d'uricase pégylée
WO2018064215A1 (fr) * 2016-09-27 2018-04-05 Selecta Biosciences, Inc. Immunotoxines recombinantes destinées à être utilisées dans le traitement du cancer
US9987354B2 (en) 2011-04-29 2018-06-05 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for antigen-specific deletion of T effector cells
US10046064B2 (en) 2014-09-07 2018-08-14 Selecta Biosciences, Inc. Methods and compositions for attenuating exon skipping anti-viral transfer vector immune responses
WO2018169811A1 (fr) * 2017-03-11 2018-09-20 Selecta Biosciences, Inc. Procédés et compositions associés à un traitement combiné avec anti-inflammatoires et nanovecteurs synthétiques comprenant un immunosuppresseur
US10335395B2 (en) 2013-05-03 2019-07-02 Selecta Biosciences, Inc. Methods of administering immunosuppressants having a specified pharmacodynamic effective life and therapeutic macromolecules for the induction of immune tolerance

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002088304A2 (fr) * 2001-04-11 2002-11-07 Trustees Of The University Of Pennsylvania Compositions et procedes destines a supprimer des reponses immunitaires
US20090082260A1 (en) * 2005-03-01 2009-03-26 Jonathan Robert Lamb Combination of an immunosuppressant and a ppar gamma agonist for the treatment of an undesirable immune response
US20100055189A1 (en) * 2008-08-29 2010-03-04 Hubbell Jeffrey A Nanoparticles for immunotherapy
US20100151000A1 (en) * 2006-10-12 2010-06-17 The University Of Queensland Compositions and methods for modulating immune responses
WO2012149255A2 (fr) * 2011-04-29 2012-11-01 Selecta Biosciences, Inc. Nanovecteurs synthétiques tolérogènes destinés à réduire des réponses immunitaires à des protéines thérapeutiques

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2571899A1 (fr) * 2004-07-01 2006-08-03 Yale University Materiaux polymeres charges de medicaments cibles a forte densite
CN101583379B (zh) * 2006-10-05 2013-04-03 约翰斯霍普金斯大学 使用优良聚合物纳米粒子的水溶性差药物的水可分散性口服,肠胃外和局部制剂
US20100112077A1 (en) * 2006-11-06 2010-05-06 Abraxis Bioscience, Llc Nanoparticles of paclitaxel and albumin in combination with bevacizumab against cancer
DK2481402T3 (en) * 2007-03-07 2018-08-06 Abraxis Bioscience Llc Nanoparticle comprising rapamycin and albumin as anticancer agent
WO2008124632A1 (fr) * 2007-04-04 2008-10-16 Massachusetts Institute Of Technology Nanoparticules assistées par des composés amphiphiles pour délivrance ciblée
US20090104114A1 (en) * 2007-09-21 2009-04-23 Cytimmune Sciences, Inc. Nanotherapeutic Colloidal Metal Compositions and Methods
MX350501B (es) * 2007-10-12 2017-09-07 Massachusetts Inst Technology Nanotecnologia de vacuna.
US8343498B2 (en) * 2008-10-12 2013-01-01 Massachusetts Institute Of Technology Adjuvant incorporation in immunonanotherapeutics
EA028288B1 (ru) * 2009-05-27 2017-10-31 Селекта Байосайенсиз, Инк. Наноносители, имеющие компоненты с различными скоростями высвобождения
SG185798A1 (en) * 2010-06-07 2013-01-30 Abraxis Bioscience Llc Combination therapy methods for treating proliferative diseases
CN103702687A (zh) * 2011-07-29 2014-04-02 西莱克塔生物科技公司 产生体液和细胞毒性t淋巴细胞(ctl)免疫应答的合成纳米载体
WO2013058812A1 (fr) * 2011-10-19 2013-04-25 President And Fellows Of Harvard College Administration ciblée à des cellules endothéliales d'îlots pancréatiques

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002088304A2 (fr) * 2001-04-11 2002-11-07 Trustees Of The University Of Pennsylvania Compositions et procedes destines a supprimer des reponses immunitaires
US20090082260A1 (en) * 2005-03-01 2009-03-26 Jonathan Robert Lamb Combination of an immunosuppressant and a ppar gamma agonist for the treatment of an undesirable immune response
US20100151000A1 (en) * 2006-10-12 2010-06-17 The University Of Queensland Compositions and methods for modulating immune responses
US20100055189A1 (en) * 2008-08-29 2010-03-04 Hubbell Jeffrey A Nanoparticles for immunotherapy
WO2012149255A2 (fr) * 2011-04-29 2012-11-01 Selecta Biosciences, Inc. Nanovecteurs synthétiques tolérogènes destinés à réduire des réponses immunitaires à des protéines thérapeutiques

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10420835B2 (en) 2011-04-29 2019-09-24 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for antigen-specific deletion of T effector cells
US11779641B2 (en) 2011-04-29 2023-10-10 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for allergy therapy
US9987354B2 (en) 2011-04-29 2018-06-05 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for antigen-specific deletion of T effector cells
US9993548B2 (en) 2011-04-29 2018-06-12 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for inducing regulatory B cells
US10004802B2 (en) 2011-04-29 2018-06-26 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for generating CD8+ regulatory T cells
US10039822B2 (en) 2011-04-29 2018-08-07 Selecta Biosciences, Inc. Method 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
US11235057B2 (en) 2011-04-29 2022-02-01 Selecta Biosciences, Inc. Methods for providing polymeric synthetic nanocarriers for generating antigen-specific tolerance immune responses
US10441651B2 (en) 2011-04-29 2019-10-15 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers for generating CD8+ regulatory T cells
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
US10357483B2 (en) 2013-05-03 2019-07-23 Selecta Biosciences, Inc. Methods comprising dosing combinations for reducing undesired humoral immune responses
US10357482B2 (en) 2013-05-03 2019-07-23 Selecta Biosciences, Inc. Methods providing a therapeutic macromolecule and synthetic nanocarriers comprising immunosuppressant locally and concomitantly to reduce both type I and type IV hypersensitivity
US10335395B2 (en) 2013-05-03 2019-07-02 Selecta Biosciences, Inc. Methods of administering immunosuppressants having a specified pharmacodynamic effective life and therapeutic macromolecules for the induction of immune tolerance
US10434088B2 (en) 2013-05-03 2019-10-08 Selecta Biosciences, Inc. Methods related to administering immunosuppressants and therapeutic macromolecules at a reduced pharmacodynamically effective dose
US10668053B2 (en) 2013-05-03 2020-06-02 Selecta Biosciences, Inc. Tolerogenic synthetic nanocarriers to reduce or prevent anaphylaxis in response to a non-allergenic antigen
US11633422B2 (en) 2014-09-07 2023-04-25 Selecta Biosciences, Inc. Methods and compositions for attenuating anti-viral transfer vector immune responses
US10046064B2 (en) 2014-09-07 2018-08-14 Selecta Biosciences, Inc. Methods and compositions for attenuating exon skipping anti-viral transfer vector immune responses
US10071114B2 (en) 2014-09-07 2018-09-11 Selecta Biosciences, Inc. Methods and compositions for attenuating gene expression modulating anti-viral transfer vector immune responses
JP2019507797A (ja) * 2016-03-11 2019-03-22 セレクタ バイオサイエンシーズ インコーポレーテッドSelecta Biosciences,Inc. ペグ化ウリカーゼの処方物および用量
JP2022068180A (ja) * 2016-03-11 2022-05-09 セレクタ バイオサイエンシーズ インコーポレーテッド ペグ化ウリカーゼの処方物および用量
WO2017156513A1 (fr) * 2016-03-11 2017-09-14 Selecta Biosciences, Inc. Formulations et doses d'uricase pégylée
JP7417354B2 (ja) 2016-03-11 2024-01-18 セレクタ バイオサイエンシーズ インコーポレーテッド ペグ化ウリカーゼの処方物および用量
WO2018064215A1 (fr) * 2016-09-27 2018-04-05 Selecta Biosciences, Inc. Immunotoxines recombinantes destinées à être utilisées dans le traitement du cancer
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
WO2018169811A1 (fr) * 2017-03-11 2018-09-20 Selecta Biosciences, Inc. Procédés et compositions associés à un traitement combiné avec anti-inflammatoires et nanovecteurs synthétiques comprenant un immunosuppresseur

Also Published As

Publication number Publication date
AU2022231724A1 (en) 2022-10-06
AU2020203312A1 (en) 2020-06-11
AU2014262164A1 (en) 2015-11-19
AU2020203312B2 (en) 2022-06-16
CA2910579A1 (fr) 2014-11-06
CA2910579C (fr) 2023-09-26
WO2014179771A8 (fr) 2015-11-12
AU2014262164B2 (en) 2020-02-27

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
WWE Wipo information: entry into national phase

Ref document number: 201480031937.3

Country of ref document: CN

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

Ref document number: 14791089

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2910579

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 242271

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: MX/A/2015/015229

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2016512092

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2014262164

Country of ref document: AU

Date of ref document: 20140502

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2014791089

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20157034104

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 201592102

Country of ref document: EA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112015027281

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112015027281

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20151028