US20210346504A1 - Exosomes For Delivery Of Biotherapeutics - Google Patents

Exosomes For Delivery Of Biotherapeutics Download PDF

Info

Publication number
US20210346504A1
US20210346504A1 US17/380,388 US202117380388A US2021346504A1 US 20210346504 A1 US20210346504 A1 US 20210346504A1 US 202117380388 A US202117380388 A US 202117380388A US 2021346504 A1 US2021346504 A1 US 2021346504A1
Authority
US
United States
Prior art keywords
exosomes
exosome
protein
peptide
targeting moiety
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/380,388
Inventor
Matthew Wood
Samira Lakhal-Littleton
Samir El Andaloussi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Oxford University Innovation Ltd
Original Assignee
Oxford University Innovation Ltd
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=45541371&utm_source=***_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20210346504(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Oxford University Innovation Ltd filed Critical Oxford University Innovation Ltd
Priority to US17/380,388 priority Critical patent/US20210346504A1/en
Assigned to OXFORD UNIVERSITY INNOVATION LIMITED reassignment OXFORD UNIVERSITY INNOVATION LIMITED CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: ISIS INNOVATION LIMITED
Assigned to OXFORD UNIVERSITY INNOVATION LIMITED reassignment OXFORD UNIVERSITY INNOVATION LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: EL ANDALOUSSI, SAMIR
Assigned to ISIS INNOVATION LIMITED reassignment ISIS INNOVATION LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LAKHAL-LITTLETON, Samira, WOOD, MATTHEW
Publication of US20210346504A1 publication Critical patent/US20210346504A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/36Skin; Hair; Nails; Sebaceous glands; Cerumen; Epidermis; Epithelial cells; Keratinocytes; Langerhans cells; Ectodermal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/44Antibodies bound to carriers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/58Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. poly[meth]acrylate, polyacrylamide, polystyrene, polyvinylpyrrolidone, polyvinylalcohol or polystyrene sulfonic acid resin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • 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
    • A61K47/6901Conjugates being cells, cell fragments, viruses, ghosts, red blood cells or viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to compositions for the delivery of protein or peptide, including antibody and antibody fragments.
  • the present invention relates to exosomes, loaded with protein or peptide and methods of producing them.
  • Biotherapeutic agents are routinely used in the treatment and/or prevention of a wide range of diseases, including cancer, genetic disorders and autoimmune diseases.
  • Biotherapeutic agents are typically protein or peptides, for example antibodies or antibody fragments and tend to be administered in naked form, i.e. without any aid to delivery.
  • administering biotherapeutics in specialized delivery vehicles would be desirable.
  • Exosomes are small membrane-bound vesicles (30-100 nm) of endocytic origin that are released into the extracellular environment following fusion of multivesicular bodies with the plasma membrane. Exosome production has been described for many immune cells including B cells, T cells, and dendritic cells (DCs). Exosomes derived from B lymphocytes and mature DCs express MHC-II, MHC-I, CD86 and ICAM-1 [1, 2], and have been used to induce specific anti-tumor T cytotoxic responses and anti-tumor immunity in experimental models and clinical trials [2, 3].
  • exosome-mediated gene delivery has been shown with delivery of murine mRNAs and miRNAs to human mast cells [4] and glioma-derived exosomes [5] have been demonstrated to transfer mRNAs produced by exogenous DNA plasmids to heterologous cells. Exosomes with exogenous DNA, siRNAs and other modified oligonucleotides have also been disclosed [6].
  • the present inventors have successfully loaded exosomes with exogenous protein, and in particular exogenous antibodies.
  • the present invention relates to methods for loading exosomes with exogenous protein and/or peptide, including antibodies and fragments thereof, the exosomes loaded with such exogenous protein and/or peptides, and their use in delivery of the protein and/or peptides for therapy.
  • the exosomes of the present invention may have a targeting moiety expressed on their surface. Fusion proteins comprising exosomal transmembrane proteins and a targeting moiety allowing for the expression of the targeting moiety on the surface of the exosomes and nucleic acid constructs encoding such fusion proteins are also described.
  • the exosomes comprising targeting moieties can be loaded with exogenous protein and/or peptides, including antibodies or fragments thereof, according to the invention and used in delivery of the protein and/or peptides for therapy.
  • composition comprising an exosome, wherein the exosome is loaded with exogenous protein and/or peptide.
  • the invention provides a composition comprising an exosome containing protein and/or peptide, wherein the exosome is derived from an immature dendritic cell, for use in a method of delivering the protein and/or peptide in vivo.
  • the invention provides a method of loading exosomes with protein and/or peptide comprising providing a composition of exosomes, and loading the exosomes with protein and/or peptide by electroporation.
  • the invention provides a method of loading exosomes with protein and/or peptide comprising providing a composition of exosomes and loading the exosomes with protein and/or peptide by transfection, using a cationic liposome transfection agent.
  • FIGS. 1A and 1B Loading of HEK cells with HEK exosomes containing PE-conjugated IgG isotype antibody: FIG. 1A shows the localisation of the PE-conjugated antibody in HEK cells wherein the HEK exosomes were mixed, but not electroporated with, the antibody. FIG. 1B shows the localisation of the PE-conjugated antibody in HEK cells wherein the antibody was loaded into the HEK exosomes by electroporating at 200 mV.
  • FIG. 2 Loading of human fibroblasts with HEK exosomes containing FITC-avidin: Transportan-Lamp2 exosomes were loaded with FITC-avidin by electroporation at 0 mV, 100 mV, 200 mV and 400 mV. The exosomes were then incubated with human fibroblasts and the amount of fluorescence emitted by the fibroblasts measured.
  • the present invention is directed to exosomes, and their use as delivery vehicles for protein and/or peptide biotherapeutics, including antibodies and antibody fragments.
  • Exosomes are small membrane-bound vesicles of endocytic origin that are released into the extracellular environment following fusion of multivesicular bodies with the plasma membrane.
  • the present application is directed to a composition comprising such exosomes.
  • the exosomes are between 30 and 100 nm in diameter but can include membrane particles of similar origin up to 200 nm.
  • Exosomes as used herein refers to nanoparticles of endosomal origin that are secreted from multivesicular bodies.
  • Exosomes are produced by many different types of cells including immune cells such as B lymphocytes, T lymphocytes, dendritic cells (DCs) and mast cells. Exosomes are also produced, for example, by glioma cells, platelets, reticulocytes, neurons, intestinal epithelial cells, tumour cells, HELA cells, human embryonic kidney cells (HEK cells), B2M17 cells, Bend3 cells, primary bone marrow-derived dendritic cells, BV-2 microglia cells and NEURO2A cells. Exosomes for use in accordance with the present application can be derived from any suitable cell, including the cells identified above. Exosomes have also been isolated from physiological fluids, such as plasma, urine, amniotic fluid and malignant effusions.
  • physiological fluids such as plasma, urine, amniotic fluid and malignant effusions.
  • exosomes are derived from DCs, preferably immature DCs.
  • Exosomes produced from immature DCs do not express MHC-II, MHC-I or CD86. As such, such exosomes do not stimulate na ⁇ ve T cells to a significant extent and are unable to induce a response in a mixed lymphocyte reaction.
  • exosomes produced from immature dendritic cells are ideal candidates for use in delivery of protein and/or peptide biotherapeutics, particularly for in vivo use, for example, in the treatment of cancer and autoimmune conditions.
  • exosomes derived from dendritic cells are provided for use in in vivo delivery of protein and/or peptide.
  • exosomes are produced by many different types of cell and have also been isolated from physiological fluids.
  • exosomes can be obtained from any suitable cell type as discussed above, or by isolation from physiological fluids.
  • the methods of the present invention comprise isolation of the exosomes from cell culture medium or tissue supernatant.
  • Exosomes produced from cells can be collected from the culture medium by any suitable method.
  • a preparation of exosomes can be prepared from cell culture or tissue supernatant by centrifugation, filtration or combinations of these methods.
  • exosomes can be prepared by differential centrifugation, that is low speed ( ⁇ 20000 g) centrifugation to pellet larger particles followed by high speed (>100000 g) centrifugation to pellet exosomes, size filtration with appropriate filters (for example, 0.22 ⁇ m filter), gradient ultracentrifugation (for example, with sucrose gradient) or a combination of these methods.
  • the exosomes are loaded with exogenous protein and/or peptide.
  • exosomes are prepared and then loaded with the desired protein and/or peptide for delivery.
  • the protein or peptide can be loaded in the exosomes by overexpression of the protein or peptide in the cell which is used to produce the exosomes, so that the exosomes can be loaded with the peptide or protein.
  • Exogenous refers to a protein with which the exosome is not normally associated.
  • exosomes to deliver biotherapeutics offer a number of advantages over conventional means of delivering such compounds. For example, when biotherapeutics are delivered using exosomes they are protected from degradation and are more stable; biotherapeutics may be delivered to a target tissue, such as a specific type of cancer, more efficiently and/or more specifically than if not encapsulated inside exosomes; and/or the cargo may be less likely to elicit an immune response because it is contained within exosomes and so is not freely available for detection by immune cells and/or binding to antibodies.
  • Other potential advantages of the use of exosomes to deliver biotherapeutics include bypassing hepatic cells following systemic delivery of exosomes and/or avoiding drug resistance, such as the upregulation of drug transporters such as ABC-transporters.
  • Suitable protein and/or peptide for delivery include exogenous antibodies and antibody fragments.
  • the protein and/or peptide loaded into the exosome is not protein and/or peptide that is typically associated with the exosomes, for example, the protein and/or amino acid is preferably not an protein or a peptide which is incorporated into an exosome on its production from a cell.
  • the present invention is concerned with the ability to load protein and/or peptide into an exosome preparation that has already been isolated from cells.
  • exogenous protein and/or peptide refers to protein and/or peptide inclusive of proteins and peptides that are not normally associated with exosomes.
  • the protein and/or peptide is an antibody and/or antibody fragment which is not typically found in exosomes.
  • any biotherapeutic protein and/or peptide that has utility in the treatment and/or prevention of a condition, disease or disorder may be incorporated into exosomes according to the present invention.
  • the protein or peptide for incorporation into the exosomes is an antibody or an antibody fragment.
  • the protein and/or peptide to be loaded into the exosomes is chosen on the basis of the desired effect of that protein and/or peptide on the cell into which it is intended to be delivered and the mechanism by which that effect is to be carried out.
  • a single protein or peptide may be incorporated into the exosomes.
  • more than one protein and/or peptide may be incorporated into the exosomes.
  • the more than one proteins and/or peptides may act on the same or different targets to bring about their therapeutic and/or preventative effect.
  • the peptide or biotherapeutic protein for delivery is not one which is used to generate an immune response.
  • the protein is typically derived from a human protein, and is not a peptide entirely of non-human origin, when used to treat a human.
  • the peptide is not derived from a bacteria, virus or other infectious agent.
  • the peptide or protein is not selected from or derived from a tumour antigen.
  • the peptide or protein is selected to provide a therapeutic benefit itself, and is not intended to be used to generate an immune response against the peptide or protein.
  • the proteins and/or peptides to be incorporated into the exosomes may be useful, for example, in the treatment and/or prevention of cancer.
  • cancers that may be treated using protein or peptide biotherapeutics include leukaemia, colorectal cancer, head and neck cancer, non-Hodgkin lymphoma, breast cancer, ovarian cancer, prostate cancer, gastric cancer, pancreatic cancer, adenocarcinoma and solid tumours.
  • Such biotherapeutic proteins and/or peptides are typically antibodies or fragments thereof, particularly monoclonal antibodies or fragments thereof.
  • Protein and/or peptide biotherapeutics may also be used in the treatment and/or prevention of autoimmune conditions.
  • Autoimmune conditions arise from an overactive immune response of the body against substances, tissues and organs normally present in the body.
  • Biotherapeutic proteins and/or peptides loaded into exosomes according to the present invention may also be used to treat and/or prevent other conditions, including cardiovascular disease, haemophilia, sepsis, stroke, muscular dystrophy, including Duchenne muscular dystrophy (DMD), macular degeneration and Alzheimer's Disease.
  • cardiovascular disease haemophilia, sepsis, stroke, muscular dystrophy, including Duchenne muscular dystrophy (DMD), macular degeneration and Alzheimer's Disease.
  • DMD Duchenne muscular dystrophy
  • macular degeneration Alzheimer's Disease.
  • the biotherapeutic loaded into exosomes according to the present invention is a therapeutic antibody.
  • the therapeutic antibody is selected from a monoclonal antibody for blocking the active site of Bace-1 and b-amyloids; a monoclonal antibody against gliblastoma kinases and an antibody against a4-integrin for the treatment of MS, for example Natalizumab.
  • the biotherapeutic loaded into exosomes according to the present invention is a peptide.
  • the peptide is selected from an immune-dominant peptide for eliciting immune responses to viral and/or tumour antigens; a neuroprotective peptide such as a ⁇ -opioid receptor ligand, for example Biphalin; an immune-suppressive peptide for neuroinflammation, for example adrenomedullin and a NBD peptide that binds and inhibits NfKB signalling.
  • the exogenous protein and/or peptide can be introduced into the exosomes by a number of different techniques.
  • the exosomes are loaded by electroporation or the use of a transfection reagent.
  • the present inventors have identified that despite the small size of exosomes, it is still possible to use electroporation to load the exosomes with the exogenous protein and/or peptide. This is surprising in view of the small size of the exosomes compared to cells. Extrapolation of the voltages used for electroporation of cells to take into account the size of the exosomes would suggest that excessively high voltages would be required for electroporation of exosomes.
  • Electroporation conditions may vary depending on the charge and size of the biotherapeutic cargo. Typical voltages are in the range of 20V/cm to 1000V/cm, such as 20V/cm to 100V/cm with capacitance typically between 25 g and 250 g, such as between 25 g and 125 g. A voltage in the range of 150 mV to 250 mV, particularly a voltage of 200 mV is preferred for loading exosomes with an antibody according to the present invention.
  • the exosomes may be loaded with exogenous protein and/or peptide using a transfection reagent.
  • a transfection reagent may be used for transfection of exosomes with protein and/or peptide.
  • Preferred transfection reagents for use in accordance with the present invention include cationic liposomes.
  • exosomes may also be loaded by transforming or transfecting a host cell with a nucleic acid construct which expresses therapeutic protein or peptide of interest, such that the therapeutic protein or peptide is taken up into the exosomes as the exosomes are produced from the cell.
  • the protein and/or peptide to be incorporated into the exosomes is an antibody or antibody fragment.
  • antibody as referred to herein includes whole antibodies and any antigen binding fragment (i.e., “antigen-binding portion”) or single chains thereof.
  • An antibody refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as V H ) and a heavy chain constant region.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as V L ) and a light chain constant region.
  • variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the V H and V L regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (C1q) of the classical complement system.
  • An antibody of use in the invention may be a monoclonal antibody or a polyclonal antibody, and will preferably be a monoclonal antibody.
  • An antibody of use in the invention may be a chimeric antibody, a CDR-grafted antibody, a nanobody, a human or humanised antibody or an antigen binding portion of any thereof.
  • the experimental animal is typically a non-human mammal such as a goat, rabbit, rat or mouse but may also be raised in other species such as camelids.
  • Polyclonal antibodies may be produced by routine methods such as immunisation of a suitable animal, with the antigen of interest. Blood may be subsequently removed from the animal and the IgG fraction purified.
  • Monoclonal antibodies (mAbs) of use in the invention can be produced by a variety of techniques, including conventional monoclonal antibody methodology e.g., the standard somatic cell hybridization technique of Kohler and Milstein.
  • the preferred animal system for preparing hybridomas is the murine system.
  • Hybridoma production in the mouse is a very well-established procedure and can be achieved using techniques well known in the art.
  • antigen-binding portion of an antibody refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term “antigen-binding portion” of an antibody include a Fab fragment, a F(ab′) 2 fragment, a Fab′ fragment, a Fd fragment, a Fv fragment, a dAb fragment and an isolated complementarity determining region (CDR). Single chain antibodies such as scFv antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody. These antibody fragments may be obtained using conventional techniques known to those of skill in the art, and the fragments may be screened for utility in the same manner as intact antibodies.
  • An antibody of use in the invention may be prepared, expressed, created or isolated by recombinant means, such as (a) antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal for the immunoglobulin genes of interest or a hybridoma prepared therefrom, (b) antibodies isolated from a host cell transformed to express the antibody of interest, e.g., from a transfectoma, (c) antibodies isolated from a recombinant, combinatorial antibody library, and (d) antibodies prepared, expressed, created or isolated by any other means that involve splicing of immunoglobulin gene sequences to other DNA sequences.
  • recombinant means such as (a) antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal for the immunoglobulin genes of interest or a hybridoma prepared therefrom, (b) antibodies isolated from a host cell transformed to express the antibody of interest, e.g., from a transfectoma,
  • An antibody of use in the invention may be a human antibody or a humanised antibody.
  • the term “human antibody”, as used herein, is intended to include antibodies having variable regions in which both the framework and CDR regions are derived from human germline immunoglobulin sequences. Furthermore, if the antibody contains a constant region, the constant region also is derived from human germline immunoglobulin sequences.
  • the human antibodies of use in the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo). However, the term “human antibody”, as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • Such a human antibody may be a human monoclonal antibody.
  • Such a human monoclonal antibody may be produced by a hybridoma which includes a B cell obtained from a transgenic nonhuman animal, e.g., a transgenic mouse, having a genome comprising a human heavy chain transgene and a light chain transgene fused to an immortalized cell.
  • Human antibodies may be prepared by in vitro immunisation of human lymphocytes followed by transformation of the lymphocytes with Epstein-Barr virus.
  • human antibody derivatives refers to any modified form of the human antibody, e.g., a conjugate of the antibody and another agent or antibody.
  • humanized antibody is intended to refer to antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences. Additional framework region modifications may be made within the human framework sequences.
  • the exosomes of the present invention may be targeted to a desired cell type or tissue. This targeting is achieved by expressing on the surface of the exosome a targeting moiety which binds to a cell surface moiety expressed on the surface of the cell to be targeted.
  • the targeting moiety is a peptide which is expressed as a fusion protein with a transmembrane protein typically expressed on the surface of the exosome.
  • the exosomes of the invention can be targeted to particular cell types or tissues by expressing on their surface a targeting moiety such as a peptide.
  • Suitable peptides are those which bind to cell surface moieties such as receptors or their ligands found on the cell surface of the cell to be targeted.
  • suitable targeting moieties are short peptides, scFv and complete proteins, so long as the targeting moiety can be expressed on the surface of the exosome and does not interfere with insertion of the membrane protein into the exosome.
  • the targeting peptide is heterologous to the transmembrane exosomal protein.
  • Peptide targeting moieties may typically be less than 100 amino acids in length, for example less than 50 amino acids in length, less than 30 amino acids in length, to a minimum length of 10, 5 or 3 amino acids.
  • Targeting moieties can be selected to target particular tissue types such as muscle, brain, liver, pancreas and lung for example, or to target a diseased tissue such as a tumour.
  • the exosomes are targeted to brain tissue.
  • targeting moieties include muscle specific peptide, discovered by phage display, to target skeletal muscle, a 29 amino acid fragment of Rabies virus glycoprotein that binds to the acetylcholine receptor or a fragment of neural growth factor that targets its receptor to target neurons, the peptide transportan, and secretin peptide that binds to the secretin receptor can be used to target biliary and pancreatic epithelia.
  • immunoglobulins and their derivatives, including scFv antibody fragments can also be expressed as a fusion protein to target specific antigens, such as VEGFR for cancer gene therapy.
  • natural ligands for receptors can be expressed as fusion proteins to confer specificity, such as NGF which binds NGFR and confers neuron-specific targeting.
  • the peptide targeting moiety is expressed on the surface of the exosome by expressing it as a fusion protein with an exosomal transmembrane protein.
  • a number of proteins are known to be associated with exosomes; that is they are incorporated into the exosome as it is formed.
  • the preferred proteins for use in targeting the exosomes of the present invention are those which are transmembrane proteins.
  • Examples include but are not limited to Lamp-1, Lamp-2, CD13, CD86, Flotillin, Syntaxin-3, CD2, CD36, CD40, CD40L, CD41a, CD44, CD45, ICAM-1, Integrin alpha4, LiCAM, LFA-1, Mac-1 alpha and beta, Vti-1A and B, CD3 epsilon and zeta, CD9, CD18, CD37, CD53, CD63, CD81, CD82, CXCR4, FcR, GluR2/3, HLA-DM (MHC II), immunoglobulins, MHC-I or MHC-II components, TCR beta and tetraspanins.
  • MHC II HLA-DM
  • the exosomes loaded with biotherapeutic protein and/or peptide are targeted using a transmembrane protein selected from Lamp-1, Lamp-2, CD13, CD86, Flotillin, Syntaxin-3.
  • a transmembrane protein selected from Lamp-1, Lamp-2, CD13, CD86, Flotillin, Syntaxin-3.
  • the transmembrane protein is Lamp-2.
  • the sequence of Lamp-2 is set out in SEQ ID NO: 1.
  • Variants of polypeptides may be defined by particular levels of amino acid identity which are described in more detail in subsequent sections of this disclosure.
  • Amino acid identity may be calculated using any suitable algorithm.
  • PILEUP and BLAST algorithms can be used to calculate homology or line up sequences (such as identifying equivalent or corresponding sequences (typically on their default settings), for example as described in Altschul S. F. (1993) J Mol Evol 36:290-300; Altschul, S, F et al (1990) J Mol Biol 215:403-10.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (http://www.ncbi.nlm.nih.gov/).
  • This algorithm involves first identifying high scoring sequence pair (HSPs) by identifying short words of length W in the query sequence that either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence.
  • T is referred to as the neighbourhood word score threshold (Altschul et al, supra).
  • These initial neighbourhood word hits act as seeds for initiating searches to find HSPs containing them.
  • the word hits are extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Extensions for the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment.
  • the BLAST algorithm performs a statistical analysis of the similarity between two sequences; see e.g., Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90: 5873-5787.
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two polynucleotide or amino acid sequences would occur by chance.
  • P(N) the smallest sum probability
  • a sequence is considered similar to another sequence if the smallest sum probability in comparison of the first sequence to the second sequence is less than about 1, preferably less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001.
  • the UWGCG Package provides the BESTFIT program which can be used to calculate homology (for example used on its default settings) (Devereux et al (1984) Nucleic Acids Research 12, 387-395).
  • variants of polypeptides also includes substitution variants.
  • substitution variants preferably involve the replacement of one or more amino acids with the same number of amino acids and making conservative amino acid substitutions.
  • an amino acid may be substituted with an alternative amino acid having similar properties, for example, another basic amino acid, another acidic amino acid, another neutral amino acid, another charged amino acid, another hydrophilic amino acid, another hydrophobic amino acid, another polar amino acid, another aromatic amino acid or another aliphatic amino acid.
  • polypeptides for use in the invention may be modified to include non-naturally occurring chemistries or to increase the stability and targeting specificity of the compound.
  • polypeptides When the polypeptides are produced by synthetic means, such amino acids may be introduced during production.
  • the polypeptides may also be modified following either synthetic or recombinant production.
  • Variant polypeptides as described in this section are those for which the amino acid sequence varies from that in SEQ ID NO: 1, but which retain the ability to be inserted into the membrane of an exosome.
  • the variant sequences typically differ by at least 1, 2, 3, 5, 10, 20, 30, 50, 100 or more mutations (which may be substitutions, deletions or insertions of amino acids). For example, from 1 to 100, 2 to 50, 3 to 30 or 5 to 20 amino acid substitutions, deletions or insertions may be made, provided the modified polypeptide is inserted into the membrane of an exosome.
  • polypeptides which are variants of Lamp-2 have more than about 50%, 55% or 65% identity, preferably at least 70%, at least 80%, at least 90% and particularly preferably at least 95%, at least 97% or at least 99% identity, with the amino acid sequence of SEQ ID NO: 1.
  • the identity of variants of SEQ ID NO: 1 may be measured over a region of at least 30, 50, 100, 200, 250, 300, 350 or more contiguous amino acids of the sequence shown in SEQ ID NO: 1, or more preferably over the full length of SEQ ID NO: 1, excluding the signal sequence.
  • the fragment of the Lamp-2 polypeptide used in the targeting of exosomes of the invention is typically at least 55 amino acids, 100, 150, 200, or 250 amino acids in length.
  • the exosomal transmembrane protein is modified to incorporate a targeting moiety.
  • the exosomal transmembrane protein is expressed as a fusion protein comprising the targeting moiety.
  • the targeting moiety is incorporated into the transmembrane protein such that it is positioned in the portion of the transmembrane protein present on the surface of the exosomes.
  • the exosomal transmembrane protein is Lamp-2 and the targeting moiety is expressed as a fusion protein, wherein the targeting moiety is present near the N-terminus of Lamp-2 protein for example within 30, or within 20 amino acids of the Lamp-2 N terminal amino acid, not including the signal sequence.
  • Spacer or linker sequences may be provided between the targeting moiety and the remainder of the transmembrane protein for example to avoid interference from the targeting moiety in the folding of the transmembrane protein.
  • Linker or spacer sequences are typically 1 to 10 amino acids in length, typically 1 to 8 amino acids in length such as 2, 3 or 4 amino acids in length.
  • Suitable amino acids for incorporation in linkers are alanine, arginine, serine or glycine.
  • Suitable linkers include Ala-Arg and Ser-Gly-Gly.
  • the transmembrane protein is Lamp-2 and the targeting moiety is present at or near the N-terminus of the protein, separated from Lamp-2 with linker sequences.
  • the targeting moiety may be introduced into the exosome by expressing the fusion protein comprising the targeting moiety and exosomal transmembrane protein within a cell used to produce the exosomes. Expression of this fusion protein in the cell, allows for the fusion protein to be incorporated into the exosome as it is produced from the cell.
  • a polynucleotide construct such as a DNA plasmid, which expressed the fusion protein is transfected into the cell.
  • Any suitable method can be used for introduction of the polynucleotide construct into the cell.
  • the polynucleotide construct includes suitable promoter sequences so that the encoded fusion protein is expressed in the cell.
  • Signal peptide sequences are also included so that the protein is incorporated into the membrane of the endoplasmic reticulum as it is produced. The membrane protein is then subsequently exported to the exosomal/lysomal compartment before incorporation into the exosome.
  • the signal sequence is typically a signal peptide sequence for an exosomal transmembrane protein.
  • the signal peptide sequence is preferably derived from Lamp-2.
  • Preferred cells for production of exosomes are discussed in more detail above.
  • a preferred cell such as an immature dendritic cells is transfected with a polynucleotide construct as described above, such that the fusion protein of the invention is expressed in the cell.
  • Exosomes produced by the cell can then be collected.
  • Such exosomes have the fusion protein inserted into the membrane such that the exosomes are targeted to the desired tissue or cell type through the targeting moiety.
  • Exosomes produced from cells can be collected from the culture medium by any suitable method.
  • a preparation of exosomes can be prepared from cell culture or tissue supernatant by centrifugation, filtration or combinations of these methods.
  • exosomes can be prepared by differential centrifugation, that is low speed ( ⁇ 20000 g) centrifugation to pellet larger particles followed by high speed (>100000 g) centrifugation to pellet exosomes, size filtration with appropriate filters (for example, 0.22 ⁇ m filter), gradient ultracentrifugation (for example, with sucrose gradient) or a combination of these methods.
  • the targeted exosomes are loaded with exogenous protein and/or peptide.
  • exosomes are prepared with a targeting moiety as described herein and then loaded with the desired protein and/or for delivery or described above.
  • a specific targeting moiety does not need to be included in the exosome.
  • exosomes may be administered directly to the site where therapy is required.
  • direct targeting to a specific site may not be required and delivery, for example, intradermal or muscular delivery may be sufficient to generate the desired immune response without targeting exosomes to any specific cell type.
  • no targeting moiety is included on the surface of the exosomes.
  • the exosomes are selected such that they are more likely to target a specific tissue type.
  • exosomes derived from different cells may have natural affinities for specific cell subtypes as required by their physiological function such as the well-established affinity of mature dendritic cell-derived exosomes to T-cells. This affinity may be utilized to specifically deliver above-mentioned cargo to a tissue.
  • the constructs of the invention may be administered by any suitable means.
  • Administration to a human or animal subject may be selected from parenteral, intramuscular, intracerebral, intravascular (including intravenous), subcutaneous, intranasal, intracardiac, intracerebroventricular, intraperitoneal or transdermal administration.
  • the method of delivery is by injection.
  • the injection is intramuscular or intravascular (e.g. intravenous).
  • a physician will be able to determine the required route of administration for each particular patient.
  • the constructs are preferably delivered as a composition.
  • the composition may be formulated for any suitable means of administration, including parenteral, intramuscular, intracerebral, intravascular (including intravenous), intracardiac, intracerebroventricular, intraperitoneal, subcutaneous, intranasal or transdermal administration.
  • Compositions for parenteral administration may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives.
  • the constructs of the invention may be formulated in a pharmaceutical composition, which may include pharmaceutically acceptable carriers, thickeners, diluents, buffers, preservatives, and other pharmaceutically acceptable carriers or excipients and the like in addition to the exosomes.
  • a “pharmaceutically acceptable carrier” is a pharmaceutically acceptable solvent, suspending agent or any other pharmacologically inert vehicle for delivering one or more nucleic acids to a subject.
  • Typical pharmaceutically acceptable carriers include, but are not limited to, binding agents (e.g. pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose, etc); fillers (e.g. lactose and other sugars, microcrystalline cellulose, pectin, gelatin, calcium sulfate, ethyl cellulose, polyacrylates or calcium hydrogen phosphate, etc); lubricants (e.g.
  • compositions provided herein may additionally contain other adjunct components conventionally found in pharmaceutical compositions.
  • the compositions may contain additional compatible pharmaceutically-active materials or may contain additional materials useful in physically formulating various dosage forms of the composition of present invention, such as dyes, flavouring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers.
  • additional materials useful in physically formulating various dosage forms of the composition of present invention such as dyes, flavouring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers.
  • such materials when added, should not unduly interfere with the biological activities of the components of the compositions provided herein.
  • a therapeutically effective amount of composition is administered.
  • the dose may be determined according to various parameters, especially according to the severity of the condition, age, and weight of the patient to be treated; the route of administration; and the required regimen.
  • a physician will be able to determine the required route of administration and dosage for any particular patient.
  • Optimum dosages may vary depending on the relative potency of individual constructs, and can generally be estimated based on EC50s found to be effective in vitro and in in vivo animal models. In general, dosage is from 0.01 mg/kg to 100 mg per kg of body weight.
  • a typical daily dose is from about 0.1 to 50 mg per kg, preferably from about 0.1 mg/kg to 10 mg/kg of body weight, according to the potency of the specific construct, the age, weight and condition of the subject to be treated, the severity of the disease and the frequency and route of administration.
  • Different dosages of the construct may be administered depending on whether administration is by intramuscular injection or systemic (intravenous or subcutaneous) injection.
  • the dose of a single intramuscular injection is in the range of about 5 to 20 ⁇ g.
  • the dose of single or multiple systemic injections is in the range of 10 to 100 mg/kg of body weight.
  • the patient may have to be treated repeatedly, for example once or more daily, weekly, monthly or yearly. Persons of ordinary skill in the art can easily estimate repetition rates for dosing based on measured residence times and concentrations of the construct in bodily fluids or tissues. Following successful treatment, it may be desirable to have the patient undergo maintenance therapy, wherein the construct is administered in maintenance doses, ranging from 0.01 mg/kg to 100 mg per kg of body weight, once or more daily, to once every 20 years.
  • Example 1 Loading of HEK Cells with HEK Exosomes Loaded with IgG Antibody
  • HEK Human embryonic kidney cell exosomes were mixed with phycoerythrin (PE)-conjugated goat anti-rabbit IgG antibody in electroporation buffer (1.15 mM potassium phosphate pH 7.2, 25 mM KCl, 21% Optiprep) and either left unloaded (mixed with the antibody but not electroporated) or electroporated at 200 mV.
  • the antibody-loaded exosomes were then added to HEK cells and incubated for two hours, by fluorescence measuring at 575 nm
  • the HEK cells were then washed in phosphate-buffered saline (PBS) and imaged.
  • FIGS. 1A and 1B show the cytoplasmic and nuclear localisation of the PE-antibody signal when cells were treated with exosomes loaded at 200 mV (B) as opposed to control exosomes which were mixed with the antibody but not electroporated (A).
  • the transportan peptide GWTLNSAGYLLGKINLKALAALAKKIL (SEQ ID NO: 2) encoded by the polynucleotide sequence ggctggaccctgaacagcgcgggctatctgctgggcaaaattaacctgaaagcgctggcggcgctggcga aaaaattctg (SEQ ID NO: 3) was inserted into the Lamp2b-expression vector previously described in International Publication No. WO/2010/119256.
  • the expression vector is based on pEGFP-C1 vector (Clonetech) from which the eGFP gene has been removed.
  • Lamp2b was cloned with cDNA from C2C12 cells and Xhol and BspEI restriction sites were inserted after the signal peptide sequence together with glycine linkers (Ala-Arg- ⁇ Targeting Peptide ⁇ -Ser-Gly-Gly).
  • the signal peptide of Lamp2b is required for membrane insertion but is cleaved off in the mature protein.
  • the full construct was then cloned downstream of the CMV promoter with NheI and BamHI restriction sites into a pEGFP-C1 vector, removing the eGFP in the process.
  • the glycine linkers flanking the transportan targeting peptide prevent the transportan targeting peptide from influencing the folding of the Lamp2b protein.
  • the transportan targeting peptide should be located on the external surface of the exosomes, hence conferring targeting capabilities to the exosomes.
  • the transportan targeting peptide of SEQ ID NO: 2 encoded by the polynucleotide sequence of SEQ ID NO: 3 was cloned into Lamp2b and transfected into dendritic cells 4 days before exosome purification.
  • a major hindrance to the ability to express targeting ligands on the surface of exosomes is that primary dendritic cells are difficult to transfect and can potentially differentiate after transfection. Infection with viral vectors is not ideal either as dendritic cells are likely to be activated by the virus [30], hence producing immunostimulatory molecules that will be incorporated into the resultant exosomes.
  • Minis Bio's TransIT-LT1 reagent was selected as it appeared to efficiently transduce dendritic cells without significantly activating dendritic cells.
  • Transfection of HEK cells was performed with 5 ⁇ g of pLamp2b-transportan peptide expression vector and 5 ⁇ l of TransIT LT1 transfection reagent (Minis Bio) in a 6-well plate with 106 cells on day 4 after harvesting and isolation of exosomes is done on Day 8.
  • Transportan-Lamp2 exosomes (5 ⁇ g) derived from HEK cells were mixed with 5 FITC-avidin and electroporated at 0 mV, 100 mV, 200 mV and 400 mV. A control population of the exosomes was not treated with the FITC-avidin.
  • Patient fibroblasts were treated for 3 h with the different exosome populations, after which the cells were washed three times with PBS and trypsinised to remove membrane-bound material. The fibroblast cells were then lysed and fluorescence measured at 490/520 nm.
  • FIG. 2 shows that fibroblasts incubated with the untreated exosomes had the lowest level of fluorescence.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • Virology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Dispersion Chemistry (AREA)
  • Hematology (AREA)
  • Zoology (AREA)
  • Dermatology (AREA)
  • Biomedical Technology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Inorganic Chemistry (AREA)
  • Transplantation (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicinal Preparation (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

The present invention relates to exosomes, loaded with biotherapeutic protein and/or peptide and methods of producing them and to the use of such exosomes for delivering protein and/or peptide in vivo, in particular the use of such exosomes in methods of therapy.

Description

    RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 14/363,685, filed Dec. 7, 2012 which is the U.S. National Stage of International Application No. PCT/GB2012/053052, filed Dec. 7, 2012, which designates the U.S., published in English, and claims priority under 35 U.S.C. §§ 119 or 365(c) to Great Britain Application No. 1121070.5, filed Dec. 7, 2011. The entire teachings of the above application are incorporated herein by reference.
  • INCORPORATION BY REFERENCE OF MATERIAL IN ASCII TEXT FILE
  • This application incorporates by reference the Sequence Listing contained in the following ASCII text file being submitted concurrently herewith:
  • File name: 47761005001_SEQUENCE_LISTING.txt; created Jun. 17, 2021, 5 KB in size.
  • FIELD OF THE INVENTION
  • The present invention relates to compositions for the delivery of protein or peptide, including antibody and antibody fragments. In particular the present invention relates to exosomes, loaded with protein or peptide and methods of producing them.
  • BACKGROUND TO THE INVENTION
  • Biotherapeutic agents are routinely used in the treatment and/or prevention of a wide range of diseases, including cancer, genetic disorders and autoimmune diseases. Biotherapeutic agents are typically protein or peptides, for example antibodies or antibody fragments and tend to be administered in naked form, i.e. without any aid to delivery. However, in some instances, for example due to rapid clearance, lack of organ-specific distribution and low efficacy of cellular uptake, administering biotherapeutics in specialized delivery vehicles would be desirable.
  • Exosomes are small membrane-bound vesicles (30-100 nm) of endocytic origin that are released into the extracellular environment following fusion of multivesicular bodies with the plasma membrane. Exosome production has been described for many immune cells including B cells, T cells, and dendritic cells (DCs). Exosomes derived from B lymphocytes and mature DCs express MHC-II, MHC-I, CD86 and ICAM-1 [1, 2], and have been used to induce specific anti-tumor T cytotoxic responses and anti-tumor immunity in experimental models and clinical trials [2, 3]. The potential of exosome-mediated gene delivery has been shown with delivery of murine mRNAs and miRNAs to human mast cells [4] and glioma-derived exosomes [5] have been demonstrated to transfer mRNAs produced by exogenous DNA plasmids to heterologous cells. Exosomes with exogenous DNA, siRNAs and other modified oligonucleotides have also been disclosed [6].
  • SUMMARY OF THE INVENTION
  • The present inventors have successfully loaded exosomes with exogenous protein, and in particular exogenous antibodies. Thus the present invention relates to methods for loading exosomes with exogenous protein and/or peptide, including antibodies and fragments thereof, the exosomes loaded with such exogenous protein and/or peptides, and their use in delivery of the protein and/or peptides for therapy.
  • The exosomes of the present invention may have a targeting moiety expressed on their surface. Fusion proteins comprising exosomal transmembrane proteins and a targeting moiety allowing for the expression of the targeting moiety on the surface of the exosomes and nucleic acid constructs encoding such fusion proteins are also described. The exosomes comprising targeting moieties can be loaded with exogenous protein and/or peptides, including antibodies or fragments thereof, according to the invention and used in delivery of the protein and/or peptides for therapy.
  • In accordance with one aspect of the present invention, there is provided a composition comprising an exosome, wherein the exosome is loaded with exogenous protein and/or peptide.
  • In another aspect, the invention provides a composition comprising an exosome containing protein and/or peptide, wherein the exosome is derived from an immature dendritic cell, for use in a method of delivering the protein and/or peptide in vivo.
  • In another embodiment, the invention provides a method of loading exosomes with protein and/or peptide comprising providing a composition of exosomes, and loading the exosomes with protein and/or peptide by electroporation.
  • In another embodiment, the invention provides a method of loading exosomes with protein and/or peptide comprising providing a composition of exosomes and loading the exosomes with protein and/or peptide by transfection, using a cationic liposome transfection agent.
  • DESCRIPTION OF THE DRAWINGS
  • FIGS. 1A and 1B. Loading of HEK cells with HEK exosomes containing PE-conjugated IgG isotype antibody: FIG. 1A shows the localisation of the PE-conjugated antibody in HEK cells wherein the HEK exosomes were mixed, but not electroporated with, the antibody. FIG. 1B shows the localisation of the PE-conjugated antibody in HEK cells wherein the antibody was loaded into the HEK exosomes by electroporating at 200 mV.
  • FIG. 2. Loading of human fibroblasts with HEK exosomes containing FITC-avidin: Transportan-Lamp2 exosomes were loaded with FITC-avidin by electroporation at 0 mV, 100 mV, 200 mV and 400 mV. The exosomes were then incubated with human fibroblasts and the amount of fluorescence emitted by the fibroblasts measured.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is directed to exosomes, and their use as delivery vehicles for protein and/or peptide biotherapeutics, including antibodies and antibody fragments. Exosomes are small membrane-bound vesicles of endocytic origin that are released into the extracellular environment following fusion of multivesicular bodies with the plasma membrane. Thus, the present application is directed to a composition comprising such exosomes. Typically, the exosomes are between 30 and 100 nm in diameter but can include membrane particles of similar origin up to 200 nm. Exosomes as used herein refers to nanoparticles of endosomal origin that are secreted from multivesicular bodies.
  • Exosomes are produced by many different types of cells including immune cells such as B lymphocytes, T lymphocytes, dendritic cells (DCs) and mast cells. Exosomes are also produced, for example, by glioma cells, platelets, reticulocytes, neurons, intestinal epithelial cells, tumour cells, HELA cells, human embryonic kidney cells (HEK cells), B2M17 cells, Bend3 cells, primary bone marrow-derived dendritic cells, BV-2 microglia cells and NEURO2A cells. Exosomes for use in accordance with the present application can be derived from any suitable cell, including the cells identified above. Exosomes have also been isolated from physiological fluids, such as plasma, urine, amniotic fluid and malignant effusions.
  • In a preferred aspect of the present invention, exosomes are derived from DCs, preferably immature DCs. Exosomes produced from immature DCs do not express MHC-II, MHC-I or CD86. As such, such exosomes do not stimulate naïve T cells to a significant extent and are unable to induce a response in a mixed lymphocyte reaction. Thus exosomes produced from immature dendritic cells are ideal candidates for use in delivery of protein and/or peptide biotherapeutics, particularly for in vivo use, for example, in the treatment of cancer and autoimmune conditions.
  • Thus, in accordance with the first aspect of the present invention, exosomes derived from dendritic cells are provided for use in in vivo delivery of protein and/or peptide.
  • As outlined above, exosomes are produced by many different types of cell and have also been isolated from physiological fluids. Thus, in accordance with the present invention, exosomes can be obtained from any suitable cell type as discussed above, or by isolation from physiological fluids. Typically, the methods of the present invention comprise isolation of the exosomes from cell culture medium or tissue supernatant.
  • Exosomes produced from cells can be collected from the culture medium by any suitable method. Typically a preparation of exosomes can be prepared from cell culture or tissue supernatant by centrifugation, filtration or combinations of these methods. For example, exosomes can be prepared by differential centrifugation, that is low speed (<20000 g) centrifugation to pellet larger particles followed by high speed (>100000 g) centrifugation to pellet exosomes, size filtration with appropriate filters (for example, 0.22 μm filter), gradient ultracentrifugation (for example, with sucrose gradient) or a combination of these methods.
  • In accordance with the present invention, the exosomes are loaded with exogenous protein and/or peptide. In particular, in accordance with the present invention, exosomes are prepared and then loaded with the desired protein and/or peptide for delivery. In another aspect, the protein or peptide can be loaded in the exosomes by overexpression of the protein or peptide in the cell which is used to produce the exosomes, so that the exosomes can be loaded with the peptide or protein. Exogenous refers to a protein with which the exosome is not normally associated.
  • The use of exosomes to deliver biotherapeutics offers a number of advantages over conventional means of delivering such compounds. For example, when biotherapeutics are delivered using exosomes they are protected from degradation and are more stable; biotherapeutics may be delivered to a target tissue, such as a specific type of cancer, more efficiently and/or more specifically than if not encapsulated inside exosomes; and/or the cargo may be less likely to elicit an immune response because it is contained within exosomes and so is not freely available for detection by immune cells and/or binding to antibodies. Other potential advantages of the use of exosomes to deliver biotherapeutics include bypassing hepatic cells following systemic delivery of exosomes and/or avoiding drug resistance, such as the upregulation of drug transporters such as ABC-transporters.
  • Suitable protein and/or peptide for delivery include exogenous antibodies and antibody fragments. In accordance with one aspect of the present invention, the protein and/or peptide loaded into the exosome is not protein and/or peptide that is typically associated with the exosomes, for example, the protein and/or amino acid is preferably not an protein or a peptide which is incorporated into an exosome on its production from a cell. In particular, the present invention is concerned with the ability to load protein and/or peptide into an exosome preparation that has already been isolated from cells. Thus exogenous protein and/or peptide refers to protein and/or peptide inclusive of proteins and peptides that are not normally associated with exosomes. In particularly preferred embodiments, the protein and/or peptide is an antibody and/or antibody fragment which is not typically found in exosomes.
  • Any biotherapeutic protein and/or peptide that has utility in the treatment and/or prevention of a condition, disease or disorder may be incorporated into exosomes according to the present invention. In a preferred embodiment the protein or peptide for incorporation into the exosomes is an antibody or an antibody fragment.
  • The protein and/or peptide to be loaded into the exosomes is chosen on the basis of the desired effect of that protein and/or peptide on the cell into which it is intended to be delivered and the mechanism by which that effect is to be carried out. A single protein or peptide may be incorporated into the exosomes. Alternatively, more than one protein and/or peptide may be incorporated into the exosomes. The more than one proteins and/or peptides may act on the same or different targets to bring about their therapeutic and/or preventative effect.
  • In a preferred aspect of the present invention, the peptide or biotherapeutic protein for delivery is not one which is used to generate an immune response. In particular the protein is typically derived from a human protein, and is not a peptide entirely of non-human origin, when used to treat a human. Typically the peptide is not derived from a bacteria, virus or other infectious agent. Also the peptide or protein is not selected from or derived from a tumour antigen. Thus the peptide or protein is selected to provide a therapeutic benefit itself, and is not intended to be used to generate an immune response against the peptide or protein.
  • The proteins and/or peptides to be incorporated into the exosomes may be useful, for example, in the treatment and/or prevention of cancer. Examples of cancers that may be treated using protein or peptide biotherapeutics include leukaemia, colorectal cancer, head and neck cancer, non-Hodgkin lymphoma, breast cancer, ovarian cancer, prostate cancer, gastric cancer, pancreatic cancer, adenocarcinoma and solid tumours. Such biotherapeutic proteins and/or peptides are typically antibodies or fragments thereof, particularly monoclonal antibodies or fragments thereof.
  • Protein and/or peptide biotherapeutics may also be used in the treatment and/or prevention of autoimmune conditions. Autoimmune conditions arise from an overactive immune response of the body against substances, tissues and organs normally present in the body.
  • Biotherapeutic proteins and/or peptides loaded into exosomes according to the present invention may also be used to treat and/or prevent other conditions, including cardiovascular disease, haemophilia, sepsis, stroke, muscular dystrophy, including Duchenne muscular dystrophy (DMD), macular degeneration and Alzheimer's Disease.
  • In a preferred embodiment, the biotherapeutic loaded into exosomes according to the present invention is a therapeutic antibody. In a more preferred embodiment, the therapeutic antibody is selected from a monoclonal antibody for blocking the active site of Bace-1 and b-amyloids; a monoclonal antibody against gliblastoma kinases and an antibody against a4-integrin for the treatment of MS, for example Natalizumab.
  • In a preferred embodiment the biotherapeutic loaded into exosomes according to the present invention is a peptide. In a more preferred embodiment the peptide is selected from an immune-dominant peptide for eliciting immune responses to viral and/or tumour antigens; a neuroprotective peptide such as a δ-opioid receptor ligand, for example Biphalin; an immune-suppressive peptide for neuroinflammation, for example adrenomedullin and a NBD peptide that binds and inhibits NfKB signalling.
  • The exogenous protein and/or peptide can be introduced into the exosomes by a number of different techniques. In particularly preferred embodiments of the invention, the exosomes are loaded by electroporation or the use of a transfection reagent. The present inventors have identified that despite the small size of exosomes, it is still possible to use electroporation to load the exosomes with the exogenous protein and/or peptide. This is surprising in view of the small size of the exosomes compared to cells. Extrapolation of the voltages used for electroporation of cells to take into account the size of the exosomes would suggest that excessively high voltages would be required for electroporation of exosomes. Surprisingly however, the present inventors have identified that it is possible to use electroporation to load exosomes with antibodies. Electroporation conditions may vary depending on the charge and size of the biotherapeutic cargo. Typical voltages are in the range of 20V/cm to 1000V/cm, such as 20V/cm to 100V/cm with capacitance typically between 25 g and 250 g, such as between 25 g and 125 g. A voltage in the range of 150 mV to 250 mV, particularly a voltage of 200 mV is preferred for loading exosomes with an antibody according to the present invention.
  • Alternatively, the exosomes may be loaded with exogenous protein and/or peptide using a transfection reagent. Despite the small size of the exosomes, conventional transfection agents may be used for transfection of exosomes with protein and/or peptide. Preferred transfection reagents for use in accordance with the present invention include cationic liposomes.
  • In another embodiment, exosomes may also be loaded by transforming or transfecting a host cell with a nucleic acid construct which expresses therapeutic protein or peptide of interest, such that the therapeutic protein or peptide is taken up into the exosomes as the exosomes are produced from the cell.
  • Antibodies
  • As described herein, in a preferred embodiment, the protein and/or peptide to be incorporated into the exosomes is an antibody or antibody fragment. The term “antibody” as referred to herein includes whole antibodies and any antigen binding fragment (i.e., “antigen-binding portion”) or single chains thereof. An antibody refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (C1q) of the classical complement system.
  • An antibody of use in the invention may be a monoclonal antibody or a polyclonal antibody, and will preferably be a monoclonal antibody. An antibody of use in the invention may be a chimeric antibody, a CDR-grafted antibody, a nanobody, a human or humanised antibody or an antigen binding portion of any thereof. For the production of both monoclonal and polyclonal antibodies, the experimental animal is typically a non-human mammal such as a goat, rabbit, rat or mouse but may also be raised in other species such as camelids.
  • Polyclonal antibodies may be produced by routine methods such as immunisation of a suitable animal, with the antigen of interest. Blood may be subsequently removed from the animal and the IgG fraction purified.
  • Monoclonal antibodies (mAbs) of use in the invention can be produced by a variety of techniques, including conventional monoclonal antibody methodology e.g., the standard somatic cell hybridization technique of Kohler and Milstein. The preferred animal system for preparing hybridomas is the murine system. Hybridoma production in the mouse is a very well-established procedure and can be achieved using techniques well known in the art.
  • The term “antigen-binding portion” of an antibody refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen. It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term “antigen-binding portion” of an antibody include a Fab fragment, a F(ab′)2 fragment, a Fab′ fragment, a Fd fragment, a Fv fragment, a dAb fragment and an isolated complementarity determining region (CDR). Single chain antibodies such as scFv antibodies are also intended to be encompassed within the term “antigen-binding portion” of an antibody. These antibody fragments may be obtained using conventional techniques known to those of skill in the art, and the fragments may be screened for utility in the same manner as intact antibodies.
  • An antibody of use in the invention may be prepared, expressed, created or isolated by recombinant means, such as (a) antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal for the immunoglobulin genes of interest or a hybridoma prepared therefrom, (b) antibodies isolated from a host cell transformed to express the antibody of interest, e.g., from a transfectoma, (c) antibodies isolated from a recombinant, combinatorial antibody library, and (d) antibodies prepared, expressed, created or isolated by any other means that involve splicing of immunoglobulin gene sequences to other DNA sequences.
  • An antibody of use in the invention may be a human antibody or a humanised antibody. The term “human antibody”, as used herein, is intended to include antibodies having variable regions in which both the framework and CDR regions are derived from human germline immunoglobulin sequences. Furthermore, if the antibody contains a constant region, the constant region also is derived from human germline immunoglobulin sequences. The human antibodies of use in the invention may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo). However, the term “human antibody”, as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • Such a human antibody may be a human monoclonal antibody. Such a human monoclonal antibody may be produced by a hybridoma which includes a B cell obtained from a transgenic nonhuman animal, e.g., a transgenic mouse, having a genome comprising a human heavy chain transgene and a light chain transgene fused to an immortalized cell.
  • Human antibodies may be prepared by in vitro immunisation of human lymphocytes followed by transformation of the lymphocytes with Epstein-Barr virus.
  • The term “human antibody derivatives” refers to any modified form of the human antibody, e.g., a conjugate of the antibody and another agent or antibody.
  • The term “humanized antibody” is intended to refer to antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences. Additional framework region modifications may be made within the human framework sequences.
  • Targeting
  • The exosomes of the present invention may be targeted to a desired cell type or tissue. This targeting is achieved by expressing on the surface of the exosome a targeting moiety which binds to a cell surface moiety expressed on the surface of the cell to be targeted. Typically the targeting moiety is a peptide which is expressed as a fusion protein with a transmembrane protein typically expressed on the surface of the exosome.
  • In more detail, the exosomes of the invention can be targeted to particular cell types or tissues by expressing on their surface a targeting moiety such as a peptide. Suitable peptides are those which bind to cell surface moieties such as receptors or their ligands found on the cell surface of the cell to be targeted. Examples of suitable targeting moieties are short peptides, scFv and complete proteins, so long as the targeting moiety can be expressed on the surface of the exosome and does not interfere with insertion of the membrane protein into the exosome. Typically the targeting peptide is heterologous to the transmembrane exosomal protein. Peptide targeting moieties may typically be less than 100 amino acids in length, for example less than 50 amino acids in length, less than 30 amino acids in length, to a minimum length of 10, 5 or 3 amino acids.
  • Targeting moieties can be selected to target particular tissue types such as muscle, brain, liver, pancreas and lung for example, or to target a diseased tissue such as a tumour. In a particularly preferred embodiment of the present invention, the exosomes are targeted to brain tissue.
  • Specific examples of targeting moieties include muscle specific peptide, discovered by phage display, to target skeletal muscle, a 29 amino acid fragment of Rabies virus glycoprotein that binds to the acetylcholine receptor or a fragment of neural growth factor that targets its receptor to target neurons, the peptide transportan, and secretin peptide that binds to the secretin receptor can be used to target biliary and pancreatic epithelia. As an alternative, immunoglobulins and their derivatives, including scFv antibody fragments can also be expressed as a fusion protein to target specific antigens, such as VEGFR for cancer gene therapy. As an alternative, natural ligands for receptors can be expressed as fusion proteins to confer specificity, such as NGF which binds NGFR and confers neuron-specific targeting.
  • The peptide targeting moiety is expressed on the surface of the exosome by expressing it as a fusion protein with an exosomal transmembrane protein. A number of proteins are known to be associated with exosomes; that is they are incorporated into the exosome as it is formed. The preferred proteins for use in targeting the exosomes of the present invention are those which are transmembrane proteins. Examples include but are not limited to Lamp-1, Lamp-2, CD13, CD86, Flotillin, Syntaxin-3, CD2, CD36, CD40, CD40L, CD41a, CD44, CD45, ICAM-1, Integrin alpha4, LiCAM, LFA-1, Mac-1 alpha and beta, Vti-1A and B, CD3 epsilon and zeta, CD9, CD18, CD37, CD53, CD63, CD81, CD82, CXCR4, FcR, GluR2/3, HLA-DM (MHC II), immunoglobulins, MHC-I or MHC-II components, TCR beta and tetraspanins. In particularly preferred embodiments of the present invention, the exosomes loaded with biotherapeutic protein and/or peptide are targeted using a transmembrane protein selected from Lamp-1, Lamp-2, CD13, CD86, Flotillin, Syntaxin-3. In a particularly preferred embodiment the transmembrane protein is Lamp-2. The sequence of Lamp-2 is set out in SEQ ID NO: 1.
  • The following section relates to general features of all polypeptides, and in particular to variations, alterations, modifications or derivatisations of amino acid sequence. It will be understood that such variations, alterations, modifications or derivatisations of polypeptides as are described herein are subject to the requirement that the polypeptides retain any further required activity or characteristic as may be specified subsequent sections of this disclosure.
  • Variants of polypeptides may be defined by particular levels of amino acid identity which are described in more detail in subsequent sections of this disclosure. Amino acid identity may be calculated using any suitable algorithm. For example the PILEUP and BLAST algorithms can be used to calculate homology or line up sequences (such as identifying equivalent or corresponding sequences (typically on their default settings), for example as described in Altschul S. F. (1993) J Mol Evol 36:290-300; Altschul, S, F et al (1990) J Mol Biol 215:403-10. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (http://www.ncbi.nlm.nih.gov/). This algorithm involves first identifying high scoring sequence pair (HSPs) by identifying short words of length W in the query sequence that either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighbourhood word score threshold (Altschul et al, supra). These initial neighbourhood word hits act as seeds for initiating searches to find HSPs containing them. The word hits are extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Extensions for the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment. The BLAST program uses as defaults a word length (W) of 11, the BLOSUM62 scoring matrix (see Henikoff and Henikoff (1992) Proc. Natl. Acad. Sci. USA 89: 10915-10919) alignments (B) of 50, expectation (E) of 10, M=5, N=4, and a comparison of both strands.
  • The BLAST algorithm performs a statistical analysis of the similarity between two sequences; see e.g., Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90: 5873-5787. One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two polynucleotide or amino acid sequences would occur by chance. For example, a sequence is considered similar to another sequence if the smallest sum probability in comparison of the first sequence to the second sequence is less than about 1, preferably less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001. Alternatively, the UWGCG Package provides the BESTFIT program which can be used to calculate homology (for example used on its default settings) (Devereux et al (1984) Nucleic Acids Research 12, 387-395).
  • It will be understood that variants of polypeptides also includes substitution variants. Substitution variants preferably involve the replacement of one or more amino acids with the same number of amino acids and making conservative amino acid substitutions. For example, an amino acid may be substituted with an alternative amino acid having similar properties, for example, another basic amino acid, another acidic amino acid, another neutral amino acid, another charged amino acid, another hydrophilic amino acid, another hydrophobic amino acid, another polar amino acid, another aromatic amino acid or another aliphatic amino acid. Some properties of the 20 main amino acids which can be used to select suitable substituents are as follows:
  • Ala aliphatic, hydrophobic, neutral
    Cys polar, hydrophobic, neutral
    Asp polar, hydrophilic, charged (−)
    Glu polar, hydrophilic, charged (−)
    Phe aromatic, hydrophobic, neutral
    Gly aliphatic, neutral
    His aromatic, polar, hydrophilic, charged (+)
    Ile aliphatic, hydrophobic, neutral
    Lys polar, hydrophilic, charged (+)
    Leu aliphatic, hydrophobic, neutral
    Met hydrophobic, neutral
    Asn polar, hydrophilic, neutral
    Pro hydrophobic, neutral
    Gln polar, hydrophilic, neutral
    Arg polar, hydrophilic, charged (+)
    Ser polar, hydrophilic, neutral
    Thr polar, hydrophilic, neutral
    Val aliphatic, hydrophobic, neutral
    Trp aromatic, hydrophobic, neutral
    Tyr aromatic, polar, hydrophobic
  • The amino acid sequence of polypeptides for use in the invention may be modified to include non-naturally occurring chemistries or to increase the stability and targeting specificity of the compound. When the polypeptides are produced by synthetic means, such amino acids may be introduced during production. The polypeptides may also be modified following either synthetic or recombinant production.
  • A number of side chain modifications are known in the art and may be made to the side chains of the polypeptides, subject to the polypeptides retaining any further required activity or characteristic as may be specified herein.
  • Variant polypeptides as described in this section are those for which the amino acid sequence varies from that in SEQ ID NO: 1, but which retain the ability to be inserted into the membrane of an exosome.
  • The variant sequences typically differ by at least 1, 2, 3, 5, 10, 20, 30, 50, 100 or more mutations (which may be substitutions, deletions or insertions of amino acids). For example, from 1 to 100, 2 to 50, 3 to 30 or 5 to 20 amino acid substitutions, deletions or insertions may be made, provided the modified polypeptide is inserted into the membrane of an exosome.
  • Typically, polypeptides which are variants of Lamp-2 have more than about 50%, 55% or 65% identity, preferably at least 70%, at least 80%, at least 90% and particularly preferably at least 95%, at least 97% or at least 99% identity, with the amino acid sequence of SEQ ID NO: 1. The identity of variants of SEQ ID NO: 1 may be measured over a region of at least 30, 50, 100, 200, 250, 300, 350 or more contiguous amino acids of the sequence shown in SEQ ID NO: 1, or more preferably over the full length of SEQ ID NO: 1, excluding the signal sequence.
  • The fragment of the Lamp-2 polypeptide used in the targeting of exosomes of the invention is typically at least 55 amino acids, 100, 150, 200, or 250 amino acids in length.
  • The exosomal transmembrane protein is modified to incorporate a targeting moiety. Thus the exosomal transmembrane protein is expressed as a fusion protein comprising the targeting moiety. The targeting moiety is incorporated into the transmembrane protein such that it is positioned in the portion of the transmembrane protein present on the surface of the exosomes. In a preferred aspect of the present invention, the exosomal transmembrane protein is Lamp-2 and the targeting moiety is expressed as a fusion protein, wherein the targeting moiety is present near the N-terminus of Lamp-2 protein for example within 30, or within 20 amino acids of the Lamp-2 N terminal amino acid, not including the signal sequence.
  • Spacer or linker sequences may be provided between the targeting moiety and the remainder of the transmembrane protein for example to avoid interference from the targeting moiety in the folding of the transmembrane protein.
  • Linker or spacer sequences are typically 1 to 10 amino acids in length, typically 1 to 8 amino acids in length such as 2, 3 or 4 amino acids in length. Suitable amino acids for incorporation in linkers are alanine, arginine, serine or glycine. Suitable linkers include Ala-Arg and Ser-Gly-Gly.
  • In a particularly preferred aspect of the present invention, the transmembrane protein is Lamp-2 and the targeting moiety is present at or near the N-terminus of the protein, separated from Lamp-2 with linker sequences.
  • The targeting moiety may be introduced into the exosome by expressing the fusion protein comprising the targeting moiety and exosomal transmembrane protein within a cell used to produce the exosomes. Expression of this fusion protein in the cell, allows for the fusion protein to be incorporated into the exosome as it is produced from the cell.
  • For example, a polynucleotide construct such as a DNA plasmid, which expressed the fusion protein is transfected into the cell. Any suitable method can be used for introduction of the polynucleotide construct into the cell. The polynucleotide construct includes suitable promoter sequences so that the encoded fusion protein is expressed in the cell. Signal peptide sequences are also included so that the protein is incorporated into the membrane of the endoplasmic reticulum as it is produced. The membrane protein is then subsequently exported to the exosomal/lysomal compartment before incorporation into the exosome. The signal sequence is typically a signal peptide sequence for an exosomal transmembrane protein. For example the signal peptide sequence is preferably derived from Lamp-2.
  • Preferred cells for production of exosomes are discussed in more detail above. Typically a preferred cell, such as an immature dendritic cells is transfected with a polynucleotide construct as described above, such that the fusion protein of the invention is expressed in the cell. Exosomes produced by the cell can then be collected. Such exosomes have the fusion protein inserted into the membrane such that the exosomes are targeted to the desired tissue or cell type through the targeting moiety.
  • Exosomes produced from cells can be collected from the culture medium by any suitable method. Typically a preparation of exosomes can be prepared from cell culture or tissue supernatant by centrifugation, filtration or combinations of these methods. For example, exosomes can be prepared by differential centrifugation, that is low speed (<20000 g) centrifugation to pellet larger particles followed by high speed (>100000 g) centrifugation to pellet exosomes, size filtration with appropriate filters (for example, 0.22 μm filter), gradient ultracentrifugation (for example, with sucrose gradient) or a combination of these methods.
  • In accordance with a preferred aspect of the present invention, the targeted exosomes are loaded with exogenous protein and/or peptide. In particular, in accordance with the present invention, exosomes are prepared with a targeting moiety as described herein and then loaded with the desired protein and/or for delivery or described above.
  • In other embodiments of the invention, a specific targeting moiety does not need to be included in the exosome. For example, exosomes may be administered directly to the site where therapy is required. Alternatively, direct targeting to a specific site may not be required and delivery, for example, intradermal or muscular delivery may be sufficient to generate the desired immune response without targeting exosomes to any specific cell type.
  • In some embodiments of the invention, no targeting moiety is included on the surface of the exosomes. However, the exosomes are selected such that they are more likely to target a specific tissue type. For example, exosomes derived from different cells may have natural affinities for specific cell subtypes as required by their physiological function such as the well-established affinity of mature dendritic cell-derived exosomes to T-cells. This affinity may be utilized to specifically deliver above-mentioned cargo to a tissue.
  • Delivery/Administration
  • The constructs of the invention may be administered by any suitable means. Administration to a human or animal subject may be selected from parenteral, intramuscular, intracerebral, intravascular (including intravenous), subcutaneous, intranasal, intracardiac, intracerebroventricular, intraperitoneal or transdermal administration. Typically the method of delivery is by injection. Preferably the injection is intramuscular or intravascular (e.g. intravenous). A physician will be able to determine the required route of administration for each particular patient.
  • The constructs are preferably delivered as a composition. The composition may be formulated for any suitable means of administration, including parenteral, intramuscular, intracerebral, intravascular (including intravenous), intracardiac, intracerebroventricular, intraperitoneal, subcutaneous, intranasal or transdermal administration. Compositions for parenteral administration may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives. The constructs of the invention may be formulated in a pharmaceutical composition, which may include pharmaceutically acceptable carriers, thickeners, diluents, buffers, preservatives, and other pharmaceutically acceptable carriers or excipients and the like in addition to the exosomes.
  • A “pharmaceutically acceptable carrier” (excipient) is a pharmaceutically acceptable solvent, suspending agent or any other pharmacologically inert vehicle for delivering one or more nucleic acids to a subject. Typical pharmaceutically acceptable carriers include, but are not limited to, binding agents (e.g. pregelatinised maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose, etc); fillers (e.g. lactose and other sugars, microcrystalline cellulose, pectin, gelatin, calcium sulfate, ethyl cellulose, polyacrylates or calcium hydrogen phosphate, etc); lubricants (e.g. magnesium stearate, talc, silica, colloidal silicon dioxide, stearic acid, metallic stearates, hydrogenated vegetable oils, corn starch, polyethylene glycols, sodium benzoate, sodium acetate, etc); disintegrates (e.g. starch, sodium starch glycolate, etc); or wetting agents (e.g. sodium lauryl sulphate, etc).
  • The compositions provided herein may additionally contain other adjunct components conventionally found in pharmaceutical compositions. Thus, for example, the compositions may contain additional compatible pharmaceutically-active materials or may contain additional materials useful in physically formulating various dosage forms of the composition of present invention, such as dyes, flavouring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers. However, such materials, when added, should not unduly interfere with the biological activities of the components of the compositions provided herein.
  • A therapeutically effective amount of composition is administered. The dose may be determined according to various parameters, especially according to the severity of the condition, age, and weight of the patient to be treated; the route of administration; and the required regimen. A physician will be able to determine the required route of administration and dosage for any particular patient. Optimum dosages may vary depending on the relative potency of individual constructs, and can generally be estimated based on EC50s found to be effective in vitro and in in vivo animal models. In general, dosage is from 0.01 mg/kg to 100 mg per kg of body weight. A typical daily dose is from about 0.1 to 50 mg per kg, preferably from about 0.1 mg/kg to 10 mg/kg of body weight, according to the potency of the specific construct, the age, weight and condition of the subject to be treated, the severity of the disease and the frequency and route of administration. Different dosages of the construct may be administered depending on whether administration is by intramuscular injection or systemic (intravenous or subcutaneous) injection. Preferably, the dose of a single intramuscular injection is in the range of about 5 to 20 μg. Preferably, the dose of single or multiple systemic injections is in the range of 10 to 100 mg/kg of body weight.
  • Due to construct clearance (and breakdown of any targeted molecule), the patient may have to be treated repeatedly, for example once or more daily, weekly, monthly or yearly. Persons of ordinary skill in the art can easily estimate repetition rates for dosing based on measured residence times and concentrations of the construct in bodily fluids or tissues. Following successful treatment, it may be desirable to have the patient undergo maintenance therapy, wherein the construct is administered in maintenance doses, ranging from 0.01 mg/kg to 100 mg per kg of body weight, once or more daily, to once every 20 years.
  • The invention is hereinafter described in more detail with reference to the following Examples.
  • EXAMPLES Example 1—Loading of HEK Cells with HEK Exosomes Loaded with IgG Antibody
  • Human embryonic kidney cell (HEK) exosomes were mixed with phycoerythrin (PE)-conjugated goat anti-rabbit IgG antibody in electroporation buffer (1.15 mM potassium phosphate pH 7.2, 25 mM KCl, 21% Optiprep) and either left unloaded (mixed with the antibody but not electroporated) or electroporated at 200 mV. The antibody-loaded exosomes were then added to HEK cells and incubated for two hours, by fluorescence measuring at 575 nm The HEK cells were then washed in phosphate-buffered saline (PBS) and imaged. FIGS. 1A and 1B show the cytoplasmic and nuclear localisation of the PE-antibody signal when cells were treated with exosomes loaded at 200 mV (B) as opposed to control exosomes which were mixed with the antibody but not electroporated (A).
  • Example 2—Loading of Human Fibroblasts with HEK Exosomes Loaded with FITC-Avidin
  • The transportan peptide GWTLNSAGYLLGKINLKALAALAKKIL (SEQ ID NO: 2) encoded by the polynucleotide sequence ggctggaccctgaacagcgcgggctatctgctgggcaaaattaacctgaaagcgctggcggcgctggcga aaaaaattctg (SEQ ID NO: 3) was inserted into the Lamp2b-expression vector previously described in International Publication No. WO/2010/119256.
  • The expression vector is based on pEGFP-C1 vector (Clonetech) from which the eGFP gene has been removed. Lamp2b was cloned with cDNA from C2C12 cells and Xhol and BspEI restriction sites were inserted after the signal peptide sequence together with glycine linkers (Ala-Arg-{Targeting Peptide}-Ser-Gly-Gly). The signal peptide of Lamp2b is required for membrane insertion but is cleaved off in the mature protein. The full construct was then cloned downstream of the CMV promoter with NheI and BamHI restriction sites into a pEGFP-C1 vector, removing the eGFP in the process.
  • The additional sequence added after the signal peptide containing the Xhol and BspEI sites enabled insertion of the transportan-coding sequence at the N-terminal part of Lamp2b.
  • The glycine linkers flanking the transportan targeting peptide prevent the transportan targeting peptide from influencing the folding of the Lamp2b protein. Ultimately, the transportan targeting peptide should be located on the external surface of the exosomes, hence conferring targeting capabilities to the exosomes.
  • The transportan targeting peptide of SEQ ID NO: 2 encoded by the polynucleotide sequence of SEQ ID NO: 3 was cloned into Lamp2b and transfected into dendritic cells 4 days before exosome purification. A major hindrance to the ability to express targeting ligands on the surface of exosomes is that primary dendritic cells are difficult to transfect and can potentially differentiate after transfection. Infection with viral vectors is not ideal either as dendritic cells are likely to be activated by the virus [30], hence producing immunostimulatory molecules that will be incorporated into the resultant exosomes. Minis Bio's TransIT-LT1 reagent was selected as it appeared to efficiently transduce dendritic cells without significantly activating dendritic cells. Transfection of HEK cells was performed with 5 μg of pLamp2b-transportan peptide expression vector and 5 μl of TransIT LT1 transfection reagent (Minis Bio) in a 6-well plate with 106 cells on day 4 after harvesting and isolation of exosomes is done on Day 8.
  • Transportan-Lamp2 exosomes (5 μg) derived from HEK cells were mixed with 5 FITC-avidin and electroporated at 0 mV, 100 mV, 200 mV and 400 mV. A control population of the exosomes was not treated with the FITC-avidin. Patient fibroblasts were treated for 3 h with the different exosome populations, after which the cells were washed three times with PBS and trypsinised to remove membrane-bound material. The fibroblast cells were then lysed and fluorescence measured at 490/520 nm. FIG. 2 shows that fibroblasts incubated with the untreated exosomes had the lowest level of fluorescence. Fibroblasts incubated with exosomes treated with FICT-avidin but and electroporated at 0 mV, 200 mV and 400 mV emitted more fluorescence than the cells treated with the control exosomes. However, cells incubated with exosomes treated with FITC-avidin and electroporated at 100 mV emitted the highest level of fluorescence, with an RFU of over 35,000.
    • 1 Raposo G, Nijman H W, Stoorvogel W, Liejendekker R, Harding C V, Melief D J Geuze H J (1996), B lymphocytes secrete antigen-presenting vesicles, J Exp Med 183:1161-1172.
    • 2 Zitvogel L, Regnault A, Lozier A, Wolfers J, Flament C, Tenza D, Ricciardi-Castagnoli P, Raposo G, Amigorena S (1998), Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes, Nat Med 4:594-600.
    • 3 Hao S, Moyana T, Xiang J (2007), Review: cancer immunotherapy by exosome-based vaccines, Cancer Biother Radiopharm 22:692-703.
    • 4 Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee J J, Lövall J O (2007), Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells, Nat Cell Biol 9(6):654-9.
    • 5 Skog J, Würdinger T, van Rijn S, Meijer D H, Gainche L, Sena-Esteves M, Curry W T Jr, Carter B S, Krichevsky A M, Breakefield X O (2008), Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers, Nat Cell Biol 10(12):1470-6.
    • 6 WO 2010/119256.

Claims (8)

1. (canceled)
2. A composition comprising a therapeutic delivery exosome and a pharmaceutically acceptable carrier, wherein the exosome is loaded with a n exogenous recombinant therapeutic protein, and wherein the exogenous recombinant therapeutic protein is encapsulated inside the exosome and does not elicit an immune response when inside the exosome, and wherein the exosome further comprises a targeting moiety expressed on the surface of the exosome.
3. The composition of claim 2, wherein the targeting moiety is 5-100 amino acids in length.
4. The composition of claim 2, wherein the targeting moiety comprises an antibody, scFv, nanobody, muscle specific protein, rabies glycoprotein or NGF.
5. The composition of claim 2, wherein the targeting moiety comprises a peptide which binds to a moiety present on a cell type to be targeted.
6. The composition of claim 5, wherein the exosome comprises an exosomal transmembrane protein which has been modified to incorporate the targeting moiety.
7. The composition of claim 6, wherein the exosomal transmembrane protein is Lamp-1, Lamp-2, CD 13, CD86, Flotillin, Syntaxin-3, CD2, CD36, CD40, CD40L, CD41a, CD44, CD45, ICAM-1, Integrin alpha4, LiCAM, LFA-1, Mac-1 alpha and beta, Vti-1A and B, CD3 epsilon and zeta, CD9, CD18, CD37, CD53, CD63, CD81, CD82, CXCR4 or tetraspanins.
8. A method of producing a therapeutic delivery exosome with an exogenous therapeutic protein comprising (a) transforming or transfecting a host cell with a nucleic acid which expresses the therapeutic protein and a nucleic acid that expresses a targeting moiety and (b) producing an exosome from the host cell, wherein the exosome comprises the therapeutic protein and the targeting moiety, and wherein the therapeutic protein is encapsulated inside the exosome and does not elicit an immune response when inside the exosome, and wherein the targeting moiety is expressed on the surface of the exosome.
US17/380,388 2011-12-07 2021-07-20 Exosomes For Delivery Of Biotherapeutics Pending US20210346504A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/380,388 US20210346504A1 (en) 2011-12-07 2021-07-20 Exosomes For Delivery Of Biotherapeutics

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
GBGB1121070.5A GB201121070D0 (en) 2011-12-07 2011-12-07 composition for delivery of biotherapeutics
GB1121070.5 2011-12-07
PCT/GB2012/053052 WO2013084000A2 (en) 2011-12-07 2012-12-07 Exosomes for delivery of biotherapeutics
US201414363685A 2014-06-06 2014-06-06
US17/380,388 US20210346504A1 (en) 2011-12-07 2021-07-20 Exosomes For Delivery Of Biotherapeutics

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/GB2012/053052 Continuation WO2013084000A2 (en) 2011-12-07 2012-12-07 Exosomes for delivery of biotherapeutics
US14/363,685 Continuation US11103586B2 (en) 2011-12-07 2012-12-07 Exosomes for delivery of biotherapeutics

Publications (1)

Publication Number Publication Date
US20210346504A1 true US20210346504A1 (en) 2021-11-11

Family

ID=45541371

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/363,685 Active 2035-02-07 US11103586B2 (en) 2011-12-07 2012-12-07 Exosomes for delivery of biotherapeutics
US17/380,388 Pending US20210346504A1 (en) 2011-12-07 2021-07-20 Exosomes For Delivery Of Biotherapeutics

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US14/363,685 Active 2035-02-07 US11103586B2 (en) 2011-12-07 2012-12-07 Exosomes for delivery of biotherapeutics

Country Status (8)

Country Link
US (2) US11103586B2 (en)
EP (3) EP2788019B1 (en)
JP (3) JP6145595B2 (en)
CN (1) CN104053451A (en)
DK (1) DK2788019T3 (en)
ES (1) ES2625863T3 (en)
GB (1) GB201121070D0 (en)
WO (1) WO2013084000A2 (en)

Families Citing this family (73)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010119256A1 (en) 2009-04-17 2010-10-21 Isis Innovation Limited Composition for delivery of genetic material
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
EP2625189B1 (en) 2010-10-01 2018-06-27 ModernaTX, Inc. Engineered nucleic acids and methods of use thereof
JP2014511687A (en) 2011-03-31 2014-05-19 モデルナ セラピューティクス インコーポレイテッド Engineered nucleic acid delivery and formulation
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
HUE057725T2 (en) 2011-10-03 2022-06-28 Modernatx Inc Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
HRP20220717T1 (en) 2011-12-16 2022-07-22 Modernatx, Inc. Modified mrna compositions
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
AU2013243947A1 (en) 2012-04-02 2014-10-30 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
EP2912177A1 (en) 2012-10-23 2015-09-02 Cornell University Treatment of metastatic breast cancer
ES2921623T3 (en) 2012-11-26 2022-08-30 Modernatx Inc terminally modified RNA
EP2971010B1 (en) 2013-03-14 2020-06-10 ModernaTX, Inc. Formulation and delivery of modified nucleoside, nucleotide, and nucleic acid compositions
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
ES2662326T5 (en) * 2013-04-12 2021-08-04 Evox Therapeutics Ltd Therapeutic delivery vesicles
EP3039126B1 (en) 2013-08-26 2019-10-09 The J. David Gladstone Institutes, A Testamentary Trust Established under The Will of J. David Gladstone Small molecule cellular reprogramming to generate neuronal cells
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
WO2015034928A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Chimeric polynucleotides
WO2015038585A1 (en) * 2013-09-11 2015-03-19 Trustees Of Dartmouth College Method for selectively inhibiting acat1 in the treatment of alzheimer's disease
EP3052106A4 (en) 2013-09-30 2017-07-19 ModernaTX, Inc. Polynucleotides encoding immune modulating polypeptides
EP3052521A1 (en) 2013-10-03 2016-08-10 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
US10513710B2 (en) 2014-04-18 2019-12-24 University Of Massachusetts Exosomal loading using hydrophobically modified oligonucleotides
US20170204152A1 (en) 2014-07-16 2017-07-20 Moderna Therapeutics, Inc. Chimeric polynucleotides
US20170210788A1 (en) 2014-07-23 2017-07-27 Modernatx, Inc. Modified polynucleotides for the production of intrabodies
NZ738149A (en) * 2015-06-10 2024-02-23 Univ Texas Use of exosomes for the treatment of disease
AU2016357303B2 (en) * 2015-11-18 2023-11-30 Aruna Bio, Inc. Neural cell extracellular vesicles
SI3394093T1 (en) 2015-12-23 2022-05-31 Modernatx, Inc. Methods of using ox40 ligand encoding polynucleotides
US20190241658A1 (en) 2016-01-10 2019-08-08 Modernatx, Inc. Therapeutic mRNAs encoding anti CTLA-4 antibodies
WO2017175253A1 (en) * 2016-04-04 2017-10-12 国立研究開発法人医薬基盤・健康・栄養研究所 Exosome-targeted dna vaccine
EP3235908A1 (en) 2016-04-21 2017-10-25 Ecole Normale Superieure De Lyon Methods for selectively modulating the activity of distinct subtypes of cells
GB201609216D0 (en) 2016-05-25 2016-07-06 Evox Therapeutics And Isis Innovation Ltd Exosomes comprising therapeutic polypeptides
GB2552301A (en) * 2016-07-11 2018-01-24 Evox Therapeutics Ltd Metabolic drug loading of EVs
GB2552460A (en) * 2016-07-11 2018-01-31 Evox Therapeutics Ltd CPP-Mediated EV Loading
GB2552774A (en) * 2016-07-12 2018-02-14 Evox Therapeutics Ltd EV-Mediated delivery of binding protein-small molecule conjugates
GB2552473A (en) 2016-07-21 2018-01-31 Evox Therapeutics Ltd Surface decoration of extracellular vesicles
WO2018049284A1 (en) * 2016-09-09 2018-03-15 Cornell University Delivery of nucleic acids, proteins and small molecules in vitreous vesicular bodies
GB201702863D0 (en) 2017-02-22 2017-04-05 Evox Therapeutics Ltd Improved loading of EVs with therapeutic proteins
WO2018208728A1 (en) 2017-05-08 2018-11-15 Flagship Pioneering, Inc. Compositions for facilitating membrane fusion and uses thereof
AU2018270111B2 (en) 2017-05-18 2022-07-14 Modernatx, Inc. Polynucleotides encoding tethered interleukin-12 (IL12) polypeptides and uses thereof
ES2962985T3 (en) 2017-06-15 2024-03-22 Univ Chicago Compositions for the treatment of cancer
MX2020000406A (en) 2017-07-10 2020-10-01 Univ Cornell Targeting chromosomal instability and downstream cytosolic dna signaling for cancer treatment.
WO2019027847A1 (en) * 2017-07-29 2019-02-07 University Of Southern California Synthetic extracellular vesicles for novel therapies
EP3710014A1 (en) 2017-11-14 2020-09-23 Henry Ford Health System Compositions for use in the treatment and prevention of cardiovascular disorders resulting from cerebrovascular injury
JP7253762B2 (en) * 2017-12-27 2023-04-07 慈濟大學 Drug delivery to autophagic and apoptotic cells by vesicles with surface-expressed proteins
KR102143160B1 (en) * 2018-01-15 2020-08-10 연세대학교 산학협력단 Pharmaceutical composition for delivering microvesicle including cardiac targeting peptide to heart and method using the same
WO2019199941A1 (en) * 2018-04-10 2019-10-17 Northwestern University Extracellular vesicles comprising targeting affinity domain-based membrane proteins
EP3598978B1 (en) * 2018-07-26 2024-05-29 EXOFIX S.r.l. Fibroadipogenic progenitor-derived exosomes for regeneration of dystrophic muscles
EP3884958A4 (en) * 2018-07-31 2023-01-25 Pineda Olvera, Benjamín Vaccination with microvesicles derived from tumour cells for cancer treatment
WO2020030561A1 (en) 2018-08-10 2020-02-13 Unicyte Ev Ag Extracellular vesicles loaded with an exogenous molecule
US20220000932A1 (en) 2018-09-28 2022-01-06 Henry Ford Health System Use of extracellular vesicles in combination with tissue plasminogen activator and/or thrombectomy to treat stroke
CA3125511A1 (en) 2019-02-08 2020-08-13 Curevac Ag Coding rna administered into the suprachoroidal space in the treatment of ophthalmic diseases
EP3946269A4 (en) * 2019-03-27 2023-05-03 University of Georgia Research Foundation, Inc. Methods of making targeted vesicles, and compositions made thereby
WO2020198696A1 (en) * 2019-03-28 2020-10-01 Agex Therapeutics, Inc. Induced tissue regeneration using extracellular vesicles
WO2020231700A1 (en) * 2019-05-11 2020-11-19 Youngsuk Yi Neurotoxin compositions and methods
US20230147602A1 (en) * 2020-03-20 2023-05-11 Orgenesis Inc. Ribonucleases for treating viral infections
WO2021205459A1 (en) 2020-04-07 2021-10-14 Ramot At Tel-Aviv University Ltd. Cannabidiol-containing compositions and uses thereof
US20210330760A1 (en) * 2020-04-22 2021-10-28 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Methods and materials for treating osteoarthritis
KR102341138B1 (en) 2020-05-31 2021-12-21 주식회사 엑소코바이오 Exosomes comprising exosomal membrane protein variant and manufacturing method thereof
WO2022023559A1 (en) 2020-07-31 2022-02-03 Curevac Ag Nucleic acid encoded antibody mixtures
KR20220101559A (en) 2021-01-11 2022-07-19 주식회사 엑소코바이오 Exosome comprising overexpressed Fc receptor or its part and manufacturing method thereof
WO2022149779A1 (en) * 2021-01-11 2022-07-14 주식회사 엑소코바이오 Exosome comprising overexpressed fc receptor or portion thereof, and method for preparing same
CN116997651A (en) 2021-01-19 2023-11-03 J·大卫格莱斯顿研究所-根据J·大卫格莱斯顿遗嘱的遗嘱信托 Gene activation targets for enhancing human T cell function
KR20220106696A (en) 2021-01-21 2022-07-29 주식회사 엑소코바이오 Recombinant exosome comprising Fc receptor or its part which can be fused to target protein or peptide and its use
CN114874990A (en) * 2021-02-05 2022-08-09 中国科学院苏州纳米技术与纳米仿生研究所 Functional exosome and preparation method and application thereof
WO2022215078A1 (en) * 2021-04-07 2022-10-13 Ramot At Tel-Aviv University Ltd. Modified cannabinoids and uses thereof
IL307544A (en) 2021-04-08 2023-12-01 Sana Biotechnology Inc Cd8-specific antibody constructs and compositions thereof
US11180534B1 (en) * 2021-06-04 2021-11-23 Morehouse School Of Medicine Compositions and methods for treating SARS-CoV-2 infections
WO2023133422A1 (en) * 2022-01-04 2023-07-13 The Broad Institute, Inc. Compositions and methods for delivering cargo to a target cell
WO2023176750A1 (en) * 2022-03-15 2023-09-21 株式会社プロジェニサイトジャパン Novel method for producing exosome containing target nucleic acid and serving as nucleic acid medicine
CN117802045A (en) * 2022-09-30 2024-04-02 北京恩康医药有限公司 Construction and application of engineering extracellular vesicles
WO2024097418A2 (en) 2022-11-03 2024-05-10 The J. David Gladstone Institutes, A Testamentary Trust Established Under The Will Of J. David Gladstone Gene activation and interference targets for exhaustion/dysfunction-resistant t cell products and uses thereof
NL2034796B1 (en) * 2023-05-10 2024-01-09 Univ Henan Science & Tech Preparation method and application of fluorescent-labeled exosomes

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE4203413C2 (en) 1992-02-06 1993-11-25 Fraunhofer Ges Forschung Multiple scanning method
DE69635895D1 (en) 1995-08-03 2006-05-04 Rijksuniversiteit Te Leiden Le ANTIGEN PRESENTING BUBBLES LEAVING FROM CELLS
US6669835B1 (en) 1997-02-18 2003-12-30 Atofina Chemicals, Inc. Aqueous dispersions of polymerizable reactants and a water incompatible catalyst sorbed on an inorganic particulate carrier
FR2766205B1 (en) 1997-07-16 2002-08-30 Inst Nat Sante Rech Med NOVEL METHOD FOR SENSITIZING ANTIGEN PRESENTING CELLS AND NOVEL MEANS FOR IMPLEMENTING THE METHOD
US6645490B2 (en) * 1998-03-02 2003-11-11 Yissum Research Development Company Of The Hebrew University Of Jerusalem Chimeric proteins with cell-targeting specificity and apoptosis-inducing activities
FR2785543B1 (en) * 1998-11-05 2003-02-28 Inst Nat Sante Rech Med MODIFIED EXOSOMES AND USES
US6627421B1 (en) * 1999-04-13 2003-09-30 Imarx Therapeutics, Inc. Methods and systems for applying multi-mode energy to biological samples
GB9927320D0 (en) 1999-11-18 2000-01-12 Chiron Spa Exosome separation
US20040241176A1 (en) * 2000-04-27 2004-12-02 Ap Cells. Inc. Method of producing membrane vesicles
FR2812813B1 (en) 2000-08-09 2004-11-26 Neovacs USE OF IMMUNOGENS TO TREAT OR PREVENT WITHIN MALIGNANT TUMORS IMMUNE DISORDERS INDUCED BY EXTRACELLULAR FACTORS
AU2002331244B2 (en) 2001-08-17 2007-02-15 Exothera L.L.C. Methods and compounds for the targeting of protein to exosomes
KR100519384B1 (en) 2002-08-13 2005-10-06 (주)누백스 Manufacturing method of exosomes using gene transfection and use of the same
US7914792B2 (en) * 2003-02-14 2011-03-29 Exothera L.L.C. Methods and compounds for raising antibodies and for screening antibody repertoires
WO2004074451A2 (en) 2003-02-18 2004-09-02 Maxcyte, Inc. Loading of cells with antigens by electroporation
US20050153304A1 (en) * 2003-04-10 2005-07-14 Government Of The Usa, As Represented By The Secretary, Department Of Health And Human Services Multivariate profiling of complex biological regulatory pathways
EP1537858A1 (en) 2003-12-04 2005-06-08 Vectron Therapeutics AG Drug delivery vehicles and uses thereof
US9085778B2 (en) * 2006-05-03 2015-07-21 VL27, Inc. Exosome transfer of nucleic acids to cells
CA2676143A1 (en) * 2007-01-26 2008-07-31 University Of Louisville Research Foundation, Inc. Modification of exosomal components for use as a vaccine
JP2011524164A (en) 2008-06-06 2011-09-01 サントル・ナシオナル・ドウ・ラ・ルシエルシユ・シアンテイフイク(セー・エヌ・エール・エス) Use of endolysosomal systems and secretory vesicles (exosome-like) in small RNA based therapeutics and diagnostics and experimental studies of small RNAs
WO2010119256A1 (en) * 2009-04-17 2010-10-21 Isis Innovation Limited Composition for delivery of genetic material
CN102459308B (en) * 2009-05-01 2016-05-04 迈世耐特股份公司 For the method for dissolving insoluble protein and/or peptide
US20120135542A1 (en) * 2009-06-05 2012-05-31 Mayo Foundation For Medical Education And Research Methods and materials for diagnosing light chain amyloidosis
CN102596177B (en) * 2009-07-01 2014-05-28 阿昂梅迪克斯公司 Microvesicles derived from nucleated, mammalian cells and use thereof
CN102470167A (en) * 2009-07-02 2012-05-23 Ith免疫治疗控股公司 Exosome based treatment of cancer
ATE545193T1 (en) 2009-07-02 2012-02-15 Converteam Technology Ltd CONTROL METHOD FOR PARALLEL-CONNECTED CURRENT TRANSFORMERS
US9023798B2 (en) * 2009-07-24 2015-05-05 The Regents Of The University Of Michigan Cystinosin replacement factor
GB201121069D0 (en) * 2011-12-07 2012-01-18 Isis Innovation Delivery system

Also Published As

Publication number Publication date
JP2015500825A (en) 2015-01-08
EP2788019B1 (en) 2017-04-05
CN104053451A (en) 2014-09-17
EP3192526A2 (en) 2017-07-19
JP2020023503A (en) 2020-02-13
DK2788019T3 (en) 2017-06-19
US11103586B2 (en) 2021-08-31
EP3192526A3 (en) 2017-08-30
WO2013084000A3 (en) 2013-10-10
JP6145595B2 (en) 2017-06-14
EP2788019A2 (en) 2014-10-15
US20140356382A1 (en) 2014-12-04
JP2017119701A (en) 2017-07-06
WO2013084000A2 (en) 2013-06-13
ES2625863T3 (en) 2017-07-20
EP3563866A1 (en) 2019-11-06
GB201121070D0 (en) 2012-01-18

Similar Documents

Publication Publication Date Title
US20210346504A1 (en) Exosomes For Delivery Of Biotherapeutics
JP7420866B2 (en) Nucleic acid polypeptide compositions and uses thereof
US11364302B1 (en) Nucleic acid-polypeptide compositions and uses thereof
US20140348904A1 (en) Exosomes With Transferrin Peptides
CN103930442A (en) Anti-CD40 antibodies, uses and methods
CN114340681A (en) Macrophage specific adapter compositions and methods of use thereof
EP4309677A1 (en) Pd-l1 and tlr7 double-targeting nanobody coupling drug and use thereof in anti-tumor
WO2023215560A1 (en) Tumor cell/immune cell multivalent receptor engager – bio-nanoparticle (timre-bnp)
CA3194929A1 (en) Modified cxcl10 for immunotherapy of cancer diseases
WO2013117735A1 (en) Conjugate comprising an agent and an antiviral ligand and its use in a method for delivering the agent intracellularly

Legal Events

Date Code Title Description
AS Assignment

Owner name: OXFORD UNIVERSITY INNOVATION LIMITED, GREAT BRITAIN

Free format text: CHANGE OF NAME;ASSIGNOR:ISIS INNOVATION LIMITED;REEL/FRAME:056944/0392

Effective date: 20160616

Owner name: OXFORD UNIVERSITY INNOVATION LIMITED, GREAT BRITAIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:EL ANDALOUSSI, SAMIR;REEL/FRAME:056935/0657

Effective date: 20210531

Owner name: ISIS INNOVATION LIMITED, GREAT BRITAIN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WOOD, MATTHEW;LAKHAL-LITTLETON, SAMIRA;REEL/FRAME:056935/0523

Effective date: 20140701

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED