US20150182588A1 - Synthetic membrane-receiver complexes - Google Patents

Synthetic membrane-receiver complexes Download PDF

Info

Publication number
US20150182588A1
US20150182588A1 US14/581,486 US201414581486A US2015182588A1 US 20150182588 A1 US20150182588 A1 US 20150182588A1 US 201414581486 A US201414581486 A US 201414581486A US 2015182588 A1 US2015182588 A1 US 2015182588A1
Authority
US
United States
Prior art keywords
receiver
polypeptide
synthetic membrane
complex
approximately
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.)
Abandoned
Application number
US14/581,486
Other languages
English (en)
Inventor
Avak Kahvejian
Jordi Mata-Fink
John Round
David Arthur Berry
Noubar B. Afeyan
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.)
Flagship Ventures Management Inc
Rubius Therapeutics Inc
Original Assignee
Flagship Ventures Management Inc
Rubius Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=52293163&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20150182588(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Flagship Ventures Management Inc, Rubius Therapeutics Inc filed Critical Flagship Ventures Management Inc
Priority to US14/581,486 priority Critical patent/US20150182588A1/en
Priority to US14/738,414 priority patent/US9644180B2/en
Publication of US20150182588A1 publication Critical patent/US20150182588A1/en
Assigned to FLAGSHIP VENTURES MANAGEMENT, INC. reassignment FLAGSHIP VENTURES MANAGEMENT, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: AFEYAN, NOUBAR B., MATA-FINK, Jordi, KAHVEJIAN, AVAK, ROUND, JOHN
Assigned to FLAGSHIP VENTURES MANAGEMENT, INC. reassignment FLAGSHIP VENTURES MANAGEMENT, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BERRY, DAVID ARTHUR
Assigned to VL26, Inc. reassignment VL26, Inc. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FLAGSHIP VENTURES MANAGEMENT, INC.
Assigned to RUBIUS THERAPEUTICS, INC. reassignment RUBIUS THERAPEUTICS, INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: VL26, Inc.
Priority to US15/473,421 priority patent/US10344263B2/en
Priority to US15/783,921 priority patent/US10329531B2/en
Priority to US15/824,877 priority patent/US10253296B2/en
Priority to US15/900,029 priority patent/US20180187154A1/en
Priority to US15/899,895 priority patent/US20180187153A1/en
Priority to US15/925,212 priority patent/US10301593B2/en
Priority to US15/940,215 priority patent/US20180216067A1/en
Priority to US15/940,069 priority patent/US20180265847A1/en
Priority to US16/195,811 priority patent/US10301594B1/en
Priority to US16/409,573 priority patent/US20190264177A1/en
Priority to US16/409,576 priority patent/US10557119B2/en
Priority to US16/426,905 priority patent/US20190316091A1/en
Priority to US16/426,880 priority patent/US20190316090A1/en
Priority to US16/431,270 priority patent/US20190309262A1/en
Priority to US16/431,236 priority patent/US20190309261A1/en
Priority to US16/549,840 priority patent/US20190376034A1/en
Priority to US16/572,129 priority patent/US20200002674A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0641Erythrocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K35/14Blood; Artificial blood
    • A61K35/18Erythrocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1774Immunoglobulin superfamily (e.g. CD2, CD4, CD8, ICAM molecules, B7 molecules, Fc-receptors, MHC-molecules)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/001Preparations to induce tolerance to non-self, e.g. prior to transplantation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/385Haptens or antigens, bound to carriers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K47/69Medicinal 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
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5063Compounds of unknown constitution, e.g. material from plants or animals
    • A61K9/5068Cell membranes or bacterial membranes enclosing drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/081Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from DNA viruses
    • C07K16/082Hepadnaviridae, e.g. hepatitis B virus
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/88Lyases (4.)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/13Coculture with; Conditioned medium produced by connective tissue cells; generic mesenchyme cells, e.g. so-called "embryonic fibroblasts"
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y204/00Glycosyltransferases (2.4)
    • C12Y204/02Pentosyltransferases (2.4.2)
    • C12Y204/02004Thymidine phosphorylase (2.4.2.4)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/22Cysteine endopeptidases (3.4.22)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y403/00Carbon-nitrogen lyases (4.3)
    • C12Y403/01Ammonia-lyases (4.3.1)
    • C12Y403/01024Phenylalanine ammonia-lyase (4.3.1.24)
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the field of the invention is pharmaceutical compositions for the treatment of diseases and disorders.
  • the circulatory system permits blood and lymph circulation to transport, e.g., nutrients, oxygen, carbon dioxide, cellular waste products, hormones, cytokines, blood cells, and pathogens to and from cells in the body.
  • Blood is a fluid comprising, e.g., plasma, red blood cells, white blood cells, and platelets that is circulated by the heart through the vertebrate vascular system.
  • the circulatory system becomes a reservoir for many toxins and pathogenic molecules upon their introduction to or production by the body.
  • the circulatory system also serves as a reservoir for cellular secretions or detritus from within the body.
  • the perpetual or aberrant circulation and proliferation of such molecules and entities can drive disease and/or exacerbate existing conditions.
  • the efficacy of therapeutic compositions that alleviate or prevent diseases and conditions associated with the circulatory system is often limited by their half-life, which is typically up to a few days.
  • the short half-life often necessitates repeated injections and hospitalizations. It is thought that the short half-life may be due to both renal clearance, e.g., of proteins smaller than 60 kDa, and non-renal clearance, e.g., via liver excretion or immune-mediated removal.
  • the activity of therapies is also often limited by an immune reaction elicited against them (see, e.g., Wang et al., Leukemia 2003, 17:1583).
  • erythrocyte ghosts that are derived from a hemolyzed red blood cell.
  • red blood cells undergo hypotonic lysis.
  • the red blood cells are exposed to low ionic strength buffer causing them to burst.
  • the resulting lysed cell membranes are isolated by centrifugation.
  • the pellet of lysed red blood cell membranes is resuspended and incubated in the presence of the therapeutic agent, for example, such as an antibiotic or chemotherapeutic agent in a low ionic strength buffer.
  • the therapeutic agent distributes within the cells.
  • Erythrocyte ghosts also elicit an immune response in mammalian subjects. These vesicles are typically constituted of both lipids and proteins, including potentially high amounts of phosphatidylserine, which is normally found on the inner leaflet of the plasma membrane. This leads to potential immunological reactions in the recipient mammalian subjects. The undesirable effects seriously limit the potential for therapeutic applications of technologies based on erythrocyte ghosts.
  • exosomes include cell-derived vesicles that are present in many and perhaps all biological fluids, including blood, urine, and cultured medium of cell cultures. The reported diameter of exosomes is between 30 and 100 nm, which is larger than low-density lipoprotein (LDL), but smaller than, for example, red blood cells. Exosomes are either released from the cell when multivesicular bodies fuse with the plasma membrane or they are released directly from the plasma membrane. Exosome delivery methods require a better understanding of their biology, as well as the development of production, characterization, targeting and cargo-loading nanotechnologies.
  • LDL low-density lipoprotein
  • hESC-MSCs human embryonic stem cell derived mesenchymal stem cells
  • a “liposome” includes an artificially-prepared spherical vesicle composed of a lamellar phase lipid bilayer. The liposome can be used as a vehicle for administration of nutrients and pharmaceutical agents. Liposomes can be prepared by disrupting biological membranes, e.g., by sonication. Liposomes are often composed of phosphatidylcholine-enriched phospholipids and may also contain mixed lipid chains with surfactant properties such as egg phosphatidylethanolamine A liposome design may employ surface ligands for attaching to a target, e.g., unhealthy tissue.
  • liposomes include the multilamellar vesicle (MLV), the small unilamellar liposome vesicle (SUV), the large unilamellar vesicle (LUV), and the cochleate vesicle.
  • MLV multilamellar vesicle
  • SUV small unilamellar liposome vesicle
  • LUV large unilamellar vesicle
  • cochleate vesicle Liposomes as cariers of anthracycline antibiotics have been a subject of a great number of studies. As a result, liposome formulations of daunorubicin (DaunoXomeTM) and doxorubicin (DoxilTM) are now commercially available. The pharmacokinetics of the liposomal forms of anthracycline antibiotics differ from that of their free forms in higher peak concentrations and longer circulations times of the drugs.
  • DaunoXomeTM daunorubi
  • the kinetics of DaunoXome and Doxil clearance from plasma is close to mono-exponential.
  • the half-life of DaumoXome in patient plasma is on the order of a few hours.
  • Doxil polyethylene glycol-coated liposomes are used.
  • the immune system poorly recognizes such liposomes; therefore the plasma half-life of Doxil is in the order of tens of hours.
  • Red blood cells have been considered for use, e.g., to degrade toxic metabolites or inactivate xenobiotics, as drug delivery systems, as carriers of antigens for vaccination, and in other biomedical applications (Magnani Ed. 2003, Erythrocyte Engineering for Drug Delivery and Targeting). Many of these applications require procedures for the transient opening of pores across the red cell membrane. Drugs have commonly been loaded into freshly isolated red blood cells, without culturing, using disruptive methods based on hypotonic shock.
  • hypotonic dialysis can induce a high degree of hemolysis, irreversible modifications in the morphology of the cells and phosphotidyl serine exposure, which has been recognized as an important parameter associated with premature red blood cells removal and induction of transfusion-related pathologies (Favretto 2013 J Contr Rel).
  • Many drugs stimulate immunogenic responses that include B cell antibody production, T cell activation, and macrophage phagocytosis.
  • the causes of immunogenicity can be extrinsic or intrinsic to the protein. Extrinsic factors are drug formulation, aggregate formation, degradation products, contaminants and dosing.
  • the administration mode, as well as the drug regimen, also strongly influences how immunogenicity is assessed. That is, immunogenicity will have different effects for drugs that are given in acute indications compared to drugs to treat chronic diseases. In the latter case, patients are exposed to the drug over a longer period of time and as such can mount a complete response. Pegylation is a technology designed to prolong the half-life, as well as minimize immunogenic responses.
  • anti-PEG polyethylene glycol
  • PEG-ASNase PEG-asparaginase
  • kidney's glomerular filtration apparatus is the primary site in the body where blood components are filtered, see for reference e.g., Osicka et al. Clin Sci 1997 93:65 and Myers et al. Kidney Int 1982 21:633.
  • the main determinant of filtration is the hydrodynamic radius of the molecule in the blood; smaller molecules ( ⁇ 80 kDa) are filtered out of the blood to a higher extent than larger molecules.
  • PEG polyethylene glycol
  • Numerous PEGylated protein and small molecule therapeutics are currently offered in the clinic (Pasut and Veronese, 2009 Adv Drug Deliv Rev 61(13):1177; Fishburn, 2008 J Pharm Sci 97(10):4167). Though effective in many cases in increasing circulation half-life, especially as the hydrodynamic radius of the graft or fusion increases (Gao, Liu, et al., 2009 PNAS 106(36):15231), these methods offer challenges in manufacturing and maintenance of biological effector function.
  • Albumin may be used to bind a therapeutic protein for increased circulation of the drug (Dennis et al, 2002 J Bil Chem 277(38):35035; Walker, Dunlevy, et al., 2010 Prot Engr Des Sel 23(4):271) to increase the apparent size of the therapeutic by engineering it to bind another protein in the blood. In this manner, the drug attains its large molecular size only after administration into the blood stream.
  • affinity-matured serum albumin-binding peptides to antibody fragments increased their circulation time 24 fold in mice (Dennis et al, 2002 J Bil Chem 277(38):35035).
  • T-cell therapies chimeric antigen receptor T-cell therapies, antibody-coupled T-cell receptor (ACTR) therapies and other adoptive T-cell therapies in effecting complete and durable responses has been demonstrated in a number of malignant and infectious diseases.
  • ACTR antibody-coupled T-cell receptor
  • T-cell therapies in effecting complete and durable responses has been demonstrated in a number of malignant and infectious diseases.
  • the development of more potent T cells is limited, however, by safety concerns, highlighted by the occurrence of on-target and off-target toxicities that, although uncommon, have been fatal on occasions.
  • Timely pharmacological intervention can be effective in the management of adverse events but adoptively transferred T cells can persist long term, along with any unwanted effects.
  • T cells targeting differentiation antigens can be expected to also recognize nonmalignant cells that express the same antigens, resulting in adverse events.
  • melanoma patients treated with T cells targeting melanocyte differentiation antigens often develop vitiligo and uveitis.
  • melanocyte differentiation antigens such as MART-1 and gp100
  • on-target toxicities have been observed across all forms of therapeutic approaches, including tumor-infiltrating cells, in vitro-expanded T-cell clones and TCR-transgenic cells.
  • on-target autoimmunity is associated with tumor regression and is more prominent in treatment approaches that are more efficacious.
  • On-target but off-tumour toxicities can be immediately life-threatening.
  • patients with colorectal cancer with lung and liver metastases may develop respiratory distress within 15 min of HER2-specific CAR T-cell infusion and may subsequently die from multiorgan failure 5 days later.
  • Cytokine release syndrome which is characterized by fevers, rigors, hypotension and hypoxia, has been observed in a number of CD19 CAR T-cell studies as a result of large-scale T-cell activation upon the recognition of CD19+ malignant cells.
  • a method of reducing the circulatory concentration of a target self-antibody comprises the steps of administering to a human subject suffering from or at risk of developing a self-antibody mediated disease, disorder or condition, a pharmaceutical composition comprising a synthetic membrane-receiver polypeptide complex, wherein the pharmaceutical composition is administered in an amount effective to substantially reduce the circulatory concentration of the target self-antibody.
  • the synthetic membrane-receiver polypeptide complex has a volume of distribution equal to the plasma volume of the subject.
  • the synthetic membrane-receiver polypeptide complex has a volume of distribution of less than 0.09 l/kg.
  • the method comprises administering the pharmaceutical composition at least twice over a treatment period such that the self-antibody mediated disease, disorder or condition is treated, or a symptom thereof is decreased.
  • the method comprises administering the pharmaceutical composition at least twice over a treatment period such that the self-antibody mediated disease, disorder or condition is prevented.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of the target self-antibody is substantially decreased during the treatment period.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of the target self-antibody is substantially decreased during the treatment period such that one or more symptoms of the self-antibody mediated disease, disorder or condition is prevented, decreased or delayed.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of the target self-antibody is decreased at a rate greater than i) the endogenous clearance rate of the target self-antibody by the human subject, or ii) the endogenous production rate of the target self-antibody by the human subject, or iii) both i) and ii).
  • the circulatory concentration of the target self-antibody is decreased by at least about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, 99.99%, or greater than 99.99% during part or the entirety of the treatment period.
  • the circulatory concentration of the target self-antibody is decreased by at least about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, 99.99%, or greater than 99.99% within about 1, 5, 10, 15, 20, 30, 40, or 50 minutes, or about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, or 23 hours, or 1, 2, 3, 4, 5, or 6 days or about 1, 2, 3, 4, 5, or 6 weeks of the administration.
  • the method comprises administering the pharmaceutical composition a sufficient number of times a treatment period such that the circulatory concentration of the target self-antibody is substantially decreased for at least about one week, two weeks, three weeks, four weeks, one month, two months, three months, four months, five months, six months, or greater than six months.
  • the method comprises administering the pharmaceutical composition a sufficient number of times a treatment period such that the circulatory concentration of the target self-antibody is substantially decreased for a period of time at least as long as the treatment period.
  • the treatment period is not longer than a year, six months, three months, two months, one month, two weeks, one week, three days, two days, one day.
  • the time interval between administrations within a treatment period is no longer than the period in which the number of synthetic membrane-receiver polypeptide complexes in circulation is reduced to less than about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% of the number of synthetic membrane-receiver polypeptide complexes present in the administered pharmaceutical composition.
  • the frequency of administration is sufficient to effectively reduce the circulatory concentration of the target self-antibody below a level that is associated with a symptom of the self-antibody mediated disease, disorder or condition.
  • the administering of the pharmaceutical composition reduces the concentration of unbound target self-antibody or the concentration of total target self-antibody in the circulatory system of the subject.
  • the concentration of total target self-antibody is approximately equal to the concentration of unbound and bound target self-antibody in the circulatory system of the subject.
  • the pharmaceutical composition further comprises a pharmaceutically active agent.
  • the method further comprises the step of administering a pharmaceutically active agent, wherein the pharmaceutically active agent is administered prior to, after, or concurrent with the pharmaceutical composition.
  • the pharmaceutical composition is administered topically or parenterally.
  • the pharmaceutically active agent is selected from a biological agent, a small molecule agent, or a nucleic acid agent.
  • the pharmaceutical composition further comprises a pharmaceutically acceptable carrier.
  • the method further comprises the step of selecting for treatment a subject suffering from or at risk of a self-antibody mediated disease, disorder or condition selected from the group consisting of: type I diabetes, multiple sclerosis, ulcerative colitis, lupus, immune thrombocytopenia purpura, warm antibody hemolytic anemia, cold agglutinin disease, Goodpasture syndrome, antiphospholipid antibody syndrome, and membranous glomerulonephritis.
  • a self-antibody mediated disease disorder or condition selected from the group consisting of: type I diabetes, multiple sclerosis, ulcerative colitis, lupus, immune thrombocytopenia purpura, warm antibody hemolytic anemia, cold agglutinin disease, Goodpasture syndrome, antiphospholipid antibody syndrome, and membranous glomerulonephritis.
  • the synthetic membrane-receiver polypeptide complex is formulated for short-term duration in the circulatory system of the subject.
  • the synthetic membrane-receiver polypeptide complex is formulated for long-term duration in the circulatory system of the subject.
  • the receiver polypeptide is not substantially disassociated from the membrane in the circulatory system of the subject.
  • the receiver polypeptide is present in the circulatory system for at least 21 days.
  • the synthetic membrane-receiver polypeptide complex comprises phosphatidylcholine, sphingomyelin, lysophosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, or phosphatidic acid.
  • the synthetic membrane-receiver polypeptide complex further comprises i) a CD47, CD55, or CD59 polypeptide or a functional fragment thereof, or ii) a cell membrane polypeptide, or iii) both i) and ii).
  • the synthetic membrane-receiver polypeptide complex comprises a CD47, CD55, or CD59 polypeptide or a functional fragment thereof in an amount effective for the polypeptide complex to reside in the circulatory system for long-term duration.
  • the synthetic membrane-receiver polypeptide complex does not contain a substantial amount of a replicating nucleic acid.
  • the synthetic membrane-receiver polypeptide complex comprises at least 10 copies, 100 copies, 1,000 copies, 10,000 copies, 25,000 copies, 50,000 copies, or 100,000 copies of the receiver polypeptide, and/or wherein the synthetic membrane-receiver polypeptide complex comprises a ratio of the receiver polypeptide relative to a membrane lipid selected from the group consisting of phosphatidylcholine, sphingomyelin, lysophosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, or phosphatidic acid.
  • a membrane lipid selected from the group consisting of phosphatidylcholine, sphingomyelin, lysophosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, or phosphatidic acid.
  • the synthetic membrane-receiver polypeptide complex comprises at least a second polypeptide in addition to the receiver polypeptide.
  • the synthetic membrane-receiver polypeptide complex has catalytic activity for more than one substrate independent of the receiver polypeptide.
  • the second polypeptide is associated with the membrane.
  • the receiver polypeptide is encoded by an exogenous nucleic acid.
  • the exogenous nucleic acid is not substantially retained by the synthetic membrane-receiver polypeptide complex.
  • the expression of the receiver polypeptide is effectively terminated.
  • the receiver polypeptide is associated with the membrane.
  • the receiver polypeptide is a fusion or a chimera.
  • the fusion or chimera comprises at least one of an S domain, an A domain or a U domain, wherein the S domain is a surface domain exposed to the environment around the synthetic membrane-receiver polypeptide complex, wherein the A domain is an anchor, wherein the U domain faces the unexposed side of the synthetic membrane-receiver polypeptide complex, and wherein the S domain, the A domain, and/or the U domain are of different polypeptide origin.
  • the S domain and/or the A domain comprises at least 6 or at least 30 amino acids.
  • the target self-antibody specifically recognizes glycoprotein (GP Ib-IX, IIb-IIIa, IV, or Ia-IIa), the NC1 domain of collagen ⁇ 3 (IV), B2 glycoprotein-1, or phospholipase A2 receptor.
  • glycoprotein GP Ib-IX, IIb-IIIa, IV, or Ia-IIa
  • IV the NC1 domain of collagen ⁇ 3
  • B2 glycoprotein-1 the NC1 domain of collagen ⁇ 3
  • phospholipase A2 receptor phospholipase A2 receptor
  • the receiver polypeptide comprises an antigenic polypeptide selected from the group consisting of glycoprotein (GP Ib-IX, IIb-IIIa, IV, or Ia-IIa), the NC1 domain of collagen ⁇ 3 (IV), B2 glycoprotein-1, or phospholipase A2 receptor, or an antigenic fragment thereof.
  • an antigenic polypeptide selected from the group consisting of glycoprotein (GP Ib-IX, IIb-IIIa, IV, or Ia-IIa), the NC1 domain of collagen ⁇ 3 (IV), B2 glycoprotein-1, or phospholipase A2 receptor, or an antigenic fragment thereof.
  • the S domain comprises the antigenic polypeptide or antigenic fragment thereof.
  • provided herein is a pharmaceutical composition administered by the methods disclosed herein.
  • the pharmaceutical composition further comprises a pharmaceutically acceptable carrier.
  • the pharmaceutical composition comprises a population of synthetic membrane-receiver polypeptide complexes.
  • the pharmaceutical composition comprises at least 1 ⁇ 10 5 synthetic membrane-receiver polypeptide complexes.
  • the synthetic membrane-receiver polypeptide complexes are provided in a volume of about 10 nl, 100 nl, 1 ⁇ l, 10 ⁇ l, 100 ⁇ l, 1 ml, 10 ml, 20 ml, or 50 ml.
  • the pharmaceutical composition comprises at least 1 ⁇ 10 11 synthetic membrane-receiver polypeptide complexes.
  • the synthetic membrane-receiver polypeptide complexes are provided in a volume of about 1 ml, 10 ml, 20 ml, 50 ml, 100 ml, 250 ml, or 500 ml.
  • the pharmaceutical composition is a composition formulated for long-term storage.
  • the pharmaceutical composition is a composition which is frozen.
  • the pharmaceutical composition comprises a pharmaceutically active agent.
  • the pharmaceutically active agent is selected from a biological agent, a small molecule agent, or a nucleic acid agent.
  • a dosage form comprising the compositions disclosed herein formulated as a liquid suspension for intravenous injection.
  • a medical device comprising a container holding the pharmaceutical compositions disclosed herein and an applicator for intravenous injection of the pharmaceutical composition to the subject.
  • kits comprising the pharmaceutical compositions disclosed herein and a medical device for intravenous injection of the pharmaceutical composition to the subject.
  • provided herein is the synthetic membrane-receiver polypeptide complex of the pharmaceutical composition administered by the methods disclosed herein.
  • provided herein is a population of synthetic membrane-receiver polypeptide complexes as disclosed herein.
  • the population of synthetic membrane-receiver polypeptide complexes are formulated as a liquid.
  • the population of synthetic membrane-receiver polypeptide complexes are frozen.
  • an isolated receiver polypeptide of the synthetic membrane-receiver polypeptide complex as disclosed herein is provided herein.
  • provided herein is an exogenous nucleic acid encoding the receiver polypeptide disclosed herein.
  • a synthetic membrane-receiver polypeptide complex comprising: a receiver polypeptide capable of interacting with a target, and a membrane comprising a second polypeptide, wherein the synthetic membrane-receiver polypeptide complex has catalytic activity independent of the receiver.
  • the synthetic membrane-receiver polypeptide complex is formulated for intravenous administration to the circulatory system of a mammalian subject, which for example can be a human.
  • the receiver polypeptide is capable of reducing the concentration of unbound target or total target in the circulatory system of the subject.
  • the synthetic membrane-receiver polypeptide complex has a volume of distribution approximately equal or equivalent to the plasma volume of the subject.
  • the synthetic membrane-receiver polypeptide complex has a volume of distribution of less than 0.09 l/kg.
  • the receiver polypeptide is present in the circulatory system for substantially the duration of the synthetic membrane-receiver polypeptide complex in the circulatory system of the subject.
  • the synthetic membrane-receiver polypeptide complex is formulated for short-term duration in the circulatory system of the subject.
  • the synthetic membrane-receiver polypeptide complex is formulated for long-term duration in the circulatory system of the subject.
  • the receiver polypeptide is present in the circulatory system for at least about 21 days.
  • the synthetic membrane-receiver polypeptide complex comprises phosphatidylcholine, sphingomyelin, lysophosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, or phosphatidic acid.
  • the synthetic membrane-receiver polypeptide complex further comprises a CD47, CD55, or CD59 polypeptide or a functional fragment thereof.
  • the synthetic membrane-receiver polypeptide complex comprises a CD47, CD55, or CD59 polypeptide or a functional fragment thereof in an amount effective for the complex to reside in the circulatory system for long-term duration.
  • the interaction of the complex with a target comprises binding, degrading, cleaving and/or sequestering the target.
  • the interaction of the complex with a target comprises altering an activity of the target.
  • the interaction of the complex with a target comprises reducing an activity of the target.
  • the interaction of the complex with a target comprises inactivating the target.
  • the target is a self-antibody, a complement protein, an immune complex, a serum amyloid protein, a metabolite or a toxin.
  • the target is an inflammatory molecule, a cytokine or a chemokine.
  • the target is a lipid or a carbohydrate, an amino acid.
  • the target is a virus, a viral antigen, an envelope antigen or a capsid antigen.
  • the target is a bacterium, a bacterial antigen, a bacterial surface antigen, a secreted bacterial toxin, or a secreted bacterial antigen.
  • the target is a fungus, a fungal antigen, a fungal cell surface antigen, a secreted fungal toxin, or a secreted fungal antigen.
  • the target is DNA or RNA.
  • the target is a circulating cell, an inflammatory cell, a tumor cell, or a metastatic cancer cell.
  • the receiver polypeptide is a complement receptor 1 (CR1) polypeptide, a variant or functional fragment thereof.
  • CR1 complement receptor 1
  • the CR1 polypeptide comprises one or more Short Consensus Repeats (SCRs), Complement Control Proteins (CCPs) and/or Long Homologous Repeats (LHRs).
  • SCRs Short Consensus Repeats
  • CCPs Complement Control Proteins
  • LHRs Long Homologous Repeats
  • the receiver polypeptide is a duffy antigen receptor complex (DARC), a variant or functional fragment thereof.
  • DARC duffy antigen receptor complex
  • the receiver polypeptide is an antibody, a single-chain variable fragment, a nanobody, a diabody, or a DARPin.
  • the receiver polypeptide is a lyase, a hydrolase, a protease, or a nuclease.
  • the receiver polypeptide is exposed to the environment around the synthetic receiver polypeptide complex.
  • the receiver polypeptide is located at the unexposed side of the synthetic receiver polypeptide complex.
  • the receiver polypeptide is associated with the membrane.
  • the receiver polypeptide is a fusion or a chimera.
  • the fusion or chimera comprises at least one of an S domain, an A domain or a U domain, wherein the S domain is a surface domain exposed to the environment around the synthetic membrane-receiver polypeptide complex, wherein the A domain is an anchor, wherein the U domain faces the unexposed side of the synthetic membrane-receiver polypeptide complex, and wherein the S domain, the A domain, and/or the U domain are of different polypeptide origin.
  • the S domain and/or the A domain comprises at least 6 or at least 30 amino acids.
  • the synthetic membrane-receiver polypeptide complex comprises at least 10 copies, 100 copies, 1,000 copies, 10,000 copies, 25,000 copies, 50,000 copies, or 100,000 copies of the receiver polypeptide, and/or wherein the synthetic membrane-receiver polypeptide complex comprises a ratio of the receiver polypeptide relative to a membrane lipid selected from the group consisting of phosphatidylcholine, sphingomyelin, lysophosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, or phosphatidic acid.
  • a membrane lipid selected from the group consisting of phosphatidylcholine, sphingomyelin, lysophosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, or phosphatidic acid.
  • the receiver polypeptide is encoded by a recombinant nucleic acid.
  • the recombinant nucleic acid is not retained by the synthetic membrane-receiver polypeptide complex.
  • the expression of the receiver polypeptide is effectively terminated.
  • the synthetic membrane-receiver polypeptide complex does not contain a substantial amount of a replicating nucleic acid.
  • a pharmaceutical composition comprising a population of synthetic membrane-receiver polypeptide complexes as disclosed herein and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition comprises at least 1 ⁇ 10 5 synthetic membrane-receiver complexes.
  • the synthetic membrane-receiver complexes are provided in a volume of about 10 nl, 100 nl, 1 ⁇ l, 10 ⁇ l, 100 ⁇ l, 1 ml, 10 ml, 20 ml, or 50 ml.
  • the pharmaceutical composition comprises at least 1 ⁇ 10 11 synthetic membrane-receiver complexes.
  • the synthetic membrane-receiver complexes are provided in a volume of about 1 ml, 10 ml, 20 ml, 50 ml, 100 ml, 250 ml, or 500 ml.
  • the pharmaceutical composition is a composition formulated for long-term storage.
  • the pharmaceutical composition is a composition which is frozen.
  • the pharmaceutical composition comprises a pharmaceutically active agent.
  • the pharmaceutically active agent is selected from a biological agent, a small molecule agent, or a nucleic acid agent.
  • a dosage form comprising the pharmaceutical compositions disclosed herein formulated as a liquid suspension for intravenous injection.
  • a medical device comprising a container holding the pharmaceutical composition disclosed herein and an applicator for intravenous injection of the pharmaceutical composition to a subject.
  • kits comprising the pharmaceutical composition disclosed herein and a medical device for intravenous injection of the pharmaceutical composition to a subject.
  • provided herein is a method of treating or preventing a disease, disorder or condition associated with the presence of or the concentration of a target in the circulatory system of a mammalian subject.
  • the method comprises administering intravenously to the subject the pharmaceutical compositions disclosed herein in an amount effective to treat or prevent disease, disorder or condition.
  • the target is associated with the disease, disorder or condition.
  • a method of modulating the circulatory concentration of a target comprises administering to a mammalian subject suffering from or at risk of developing a disease, disorder or condition associated with the presence, absence, elevated or depressed concentration of the target in the circulatory system of the subject, a pharmaceutical composition comprising a synthetic membrane-receiver polypeptide complex in an amount effective to substantially modulate the circulatory concentration of the target.
  • the synthetic membrane-receiver polypeptide complex has a volume of distribution equal to the plasma volume of the subject.
  • the administration is repeated when the amount of synthetic membrane-receiver polypeptide complexes in circulation is reduced to 50% of i) the concentration of the complexes that were first administered or ii) Cmax of the synthetic membrane-receiver polypeptide complexes in circulation.
  • the synthetic membrane-receiver polypeptide complex interacts with the target in circulation.
  • the interaction with the target comprises binding, degrading, cleaving and/or sequestering the target.
  • the interaction with a target comprises altering an activity of the target.
  • the interaction with the target comprises reducing an activity of the target.
  • the interaction with the target comprises inactivating the target.
  • the interaction with the target comprises catalytically converting the target.
  • modulating consists of reducing the circulatory concentration of the target.
  • the presence or elevated level of the target in the circulatory system of the subject is associated with the disease, disorder or condition.
  • the method further comprises increasing the circulatory concentration of a non-target compound.
  • the absence or depressed level of the non-target compound in the circulatory system of the subject is associated with the disease, disorder or condition.
  • the target is a biological compound, an inorganic compound, an organic compound, a gaseous compound or an element.
  • the target is less than 1000 Da, less than 500 Da, less than 250 Da, or less than 100 Da.
  • the target is more than 1 kDa.
  • the target is a polypeptide, a lipid, a carbohydrate, a nucleic acid, an amino acid, metabolite, or a small molecule.
  • the target is an antibody, a complement factor, an immune complex, a serum amyloid protein, a bacterial pathogen, a fungal pathogen, a viral pathogen, or an infected, pathogenic, apoptotic, necrotic, aberrant or oncogenic mammalian cell.
  • the target is an amyloid polypeptide.
  • the target is a complement polypeptide.
  • the substantial modulation of the circulatory concentration of the target is reversible.
  • the substantial modulation of the circulatory concentration of the target is temporally restricted.
  • the substantial modulation of the circulatory concentration of the target is spatially restricted.
  • a method of reducing the circulatory concentration of a target serum amyloid protein comprises the steps of administering to a mammalian subject suffering from or at risk of developing an amyloidosis, a pharmaceutical composition comprising a synthetic membrane-receiver complex, wherein the pharmaceutical composition is administered in an amount effective to substantially reduce the circulatory concentration of the target serum amyloid protein.
  • the synthetic membrane-receiver complex has a volume of distribution equal to the plasma volume of the subject. In some embodiments, the synthetic membrane-receiver complex has a volume of distribution of less than 0.09 l/kg.
  • the method comprises administering the pharmaceutical composition at least twice over a treatment period such that the amyloidosis is treated, or a symptom thereof is decreased.
  • the method comprises administering the pharmaceutical composition at least twice over a treatment period such that the amyloidosis is prevented.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of the target serum amyloid protein is substantially decreased during the treatment period.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of the target serum amyloid protein is substantially decreased during the treatment period such that one or more symptom of the amyloidosis is prevented, decreased or delayed.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of the target serum amyloid protein is decreased at a rate greater than i) the endogenous clearance rate of the target serum amyloid protein by the mammalian subject, or ii) the endogenous production rate of the target serum amyloid protein by the mammalian subject, or iii) both i) and ii).
  • the circulatory concentration of the target serum amyloid protein is decreased by at least about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, 99.99%, or greater than 99.99% during part or the entirety of the treatment period.
  • the circulatory concentration of the target serum amyloid protein is decreased by at least about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, 99.99%, or greater than 99.99% within about 1, 5, 10, 15, 20, 30, 40, or 50 minutes, or about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, or 23 hours, or 1, 2, 3, 4, 5, or 6 days or about 1, 2, 3, 4, 5, or 6 weeks of the administration.
  • the method comprises administering the pharmaceutical composition a sufficient number of times a treatment period such that the circulatory concentration of the target serum amyloid protein is substantially decreased for at least about one week, two weeks, three weeks, four weeks, one month, two months, three months, four months, five months, six months, or greater than six months.
  • the method comprises administering the pharmaceutical composition a sufficient number of times a treatment period such that the circulatory concentration of the target serum amyloid protein is substantially decreased for a period of time at least as long as the treatment period.
  • the treatment period is not longer than a year, six months, three months, two months, one month, two weeks, one week, three days, two days, one day.
  • the time interval between administrations within a treatment period is no longer than the period in which the number of synthetic membrane-receiver complexes in circulation is reduced to less than about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% of the number of synthetic membrane-receiver complexes present in the administered pharmaceutical composition.
  • the frequency of administration is sufficient to effectively reduce the circulatory concentration of the target serum amyloid protein below a level that is associated with a symptom of the amyloidosis.
  • the administering of the pharmaceutical composition reduces the concentration of unbound target serum amyloid protein or the concentration of total target serum amyloid protein in the circulatory system of the subject.
  • the concentration of total target serum amyloid protein is approximately equal to the concentration of unbound and bound target serum amyloid protein in the circulatory system of the subject.
  • the method further comprises the step of selecting for treatment a subject suffering from or at risk of an amyloidosis selected from the group consisting of: A amyloidosis (AA), Ig light chain amyloidosis (AL), transthyretin (TTR) amyloidosis, and fibrinogen amyloidosis.
  • AA A amyloidosis
  • AL Ig light chain amyloidosis
  • TTR transthyretin amyloidosis
  • fibrinogen amyloidosis fibrinogen amyloidosis
  • the target serum amyloid protein is selected from the group consisting of: amyloid P protein, amyloid A protein, light chain, misfolded transthyretin, and fibrinogen alpha chain.
  • the receiver is associated with the membrane.
  • the receiver is a fusion or a chimera.
  • the fusion or chimera may comprise at least one of an S domain, an A domain or a U domain, wherein the S domain is a surface domain exposed to the environment around the synthetic membrane-receiver complex, wherein the A domain is an anchor, wherein the U domain faces the unexposed side of the synthetic membrane-receiver complex, and wherein the S domain, the A domain, and/or the U domain are of different polypeptide origin.
  • the S domain and/or the A domain comprise a polypeptide comprising at least 6 or at least 30 amino acids.
  • the S domain comprises the antigenic polypeptide or antigenic fragment thereof.
  • a method of reducing the circulatory concentration of a target immune complex comprises the steps of administering to a mammalian subject suffering from or at risk of developing an immune complex-associated disease, disorder or condition, a pharmaceutical composition comprising a synthetic membrane-complement receptor 1 (CR1) receiver complex, wherein the pharmaceutical composition is administered in an amount effective to substantially reduce the circulatory concentration of the target immune complex.
  • a pharmaceutical composition comprising a synthetic membrane-complement receptor 1 (CR1) receiver complex, wherein the pharmaceutical composition is administered in an amount effective to substantially reduce the circulatory concentration of the target immune complex.
  • CR1 synthetic membrane-complement receptor 1
  • the synthetic membrane-CR1 receiver complex has a volume of distribution equal to the plasma volume of the subject. In some embodiments, the synthetic membrane-CR1 receiver complex has a volume of distribution of less than 0.09 l/kg.
  • the method comprises administering the pharmaceutical composition at least twice over a treatment period such that the immune complex-associated disease, disorder or condition is treated, or a symptom thereof is decreased.
  • the method comprises administering the pharmaceutical composition at least twice over a treatment period such that the immune complex-associated disease, disorder or condition is prevented.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of the target immune complex is substantially decreased during the treatment period.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of the target immune complex is substantially decreased during the treatment period such that one or more symptom of the a immune complex-associated disease, disorder or condition is prevented, decreased or delayed.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of the target immune complex is decreased at a rate greater than i) the endogenous clearance rate of the target immune complex by the mammalian subject, or ii) the endogenous production rate of the target immune complex by the mammalian subject, or iii) both i) and ii).
  • the circulatory concentration of the target immune complex is decreased by at least about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, 99.99%, or greater than 99.99% during part or the entirety of the treatment period.
  • the circulatory concentration of the target immune complex is decreased by at least about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, 99.99%, or greater than 99.99% within about 1, 5, 10, 15, 20, 30, 40, or 50 minutes, or about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, or 23 hours, or 1, 2, 3, 4, 5, or 6 days or about 1, 2, 3, 4, 5, or 6 weeks of the administration.
  • the method comprises administering the pharmaceutical composition a sufficient number of times a treatment period such that the circulatory concentration of the target immune complex is substantially decreased for at least about one week, two weeks, three weeks, four weeks, one month, two months, three months, four months, five months, six months, or greater than six months.
  • the method comprises administering the pharmaceutical composition a sufficient number of times a treatment period such that the circulatory concentration of the target immune complex is substantially decreased for a period of time at least as long as the treatment period.
  • the treatment period is not longer than a year, six months, three months, two months, one month, two weeks, one week, three days, two days, one day.
  • the time interval between administrations within a treatment period is no longer than the period in which the number of synthetic membrane-CR1 receiver complexes in circulation is reduced to less than about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% of the number of synthetic membrane-CR1 receiver complexes present in the administered pharmaceutical composition.
  • the frequency of administration is sufficient to effectively reduce the circulatory concentration of the target immune complex below a level that is associated with a symptom of the immune complex-associated disease, disorder or condition.
  • the administering of the pharmaceutical composition reduces the concentration of unbound target immune complex or the concentration of total target immune complex in the circulatory system of the subject.
  • the concentration of total target immune complex is approximately equal to the concentration of unbound and bound target immune complex in the circulatory system of the subject.
  • the method further comprises the step of selecting for treatment a subject suffering from or at risk of a immune complex-associated disease, disorder or condition selected from the group consisting of: IgA nephropathy and lupus nephritis.
  • the target immune complex comprises i) IgM or IgG, and ii) C3b and/or C4b.
  • the receiver is associated with the membrane.
  • the receiver is a fusion or a chimera.
  • the fusion or chimera may comprise at least one of an S domain, an A domain or a U domain, wherein the S domain is a surface domain exposed to the environment around the synthetic membrane-CR1 receiver complex, wherein the A domain is an anchor, wherein the U domain faces the unexposed side of the synthetic membrane-CR1 receiver complex, and wherein the S domain, the A domain, and/or the U domain are of different polypeptide origin.
  • the S domain and/or the A domain comprise a polypeptide comprising at least 6 or at least 30 amino acids.
  • the S domain comprises the antigenic polypeptide or antigenic fragment thereof.
  • the CR1 receiver polypeptide comprises one or more of any one of a complement control protein (CCP) module, a short consensus repeat (SCR), and/or a long homologous repeat (LHRs)
  • CCP complement control protein
  • SCR short consensus repeat
  • LHRs long homologous repeat
  • a method of reducing the circulatory concentration of a target complement protein comprises the steps of administering to a mammalian subject suffering from or at risk of developing a disease, disorder or condition associated with the dysregulation of a complement protein, a pharmaceutical composition comprising a synthetic membrane-receiver complex, wherein the pharmaceutical composition is administered in an amount effective to substantially reduce the circulatory concentration of the target complement protein.
  • the synthetic membrane-receiver complex has a volume of distribution equal to the plasma volume of the subject. In some embodiments, the synthetic membrane-receiver complex has a volume of distribution of less than 0.09 l/kg.
  • the method comprises administering the pharmaceutical composition at least twice over a treatment period such that the disease, disorder or condition associated with the dysregulation of a complement protein is treated, or a symptom thereof is decreased.
  • the method comprises administering the pharmaceutical composition at least twice over a treatment period such that the disease, disorder or condition associated with the dysregulation of a complement protein is prevented.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of the target complement protein is substantially decreased during the treatment period.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of the target complement protein is substantially decreased during the treatment period such that one or more symptom of the disease, disorder or condition associated with the dysregulation of a complement protein is prevented, decreased or delayed.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of the target complement protein is decreased at a rate greater than i) the endogenous clearance rate of the target complement protein by the mammalian subject, or ii) the endogenous production rate of the target complement protein by the mammalian subject, or iii) both i) and ii).
  • the circulatory concentration of the target complement protein is decreased by at least about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, 99.99%, or greater than 99.99% during part or the entirety of the treatment period.
  • the circulatory concentration of the target complement protein is decreased by at least about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, 99.99%, or greater than 99.99% within about 1, 5, 10, 15, 20, 30, 40, or 50 minutes, or about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, or 23 hours, or 1, 2, 3, 4, 5, or 6 days or about 1, 2, 3, 4, 5, or 6 weeks of the administration.
  • the method comprises administering the pharmaceutical composition a sufficient number of times a treatment period such that the circulatory concentration of the target complement protein is substantially decreased for at least about one week, two weeks, three weeks, four weeks, one month, two months, three months, four months, five months, six months, or greater than six months.
  • the method comprises administering the pharmaceutical composition a sufficient number of times a treatment period such that the circulatory concentration of the target complement protein is substantially decreased for a period of time at least as long as the treatment period.
  • the treatment period is not longer than a year, six months, three months, two months, one month, two weeks, one week, three days, two days, one day.
  • the time interval between administrations within a treatment period is no longer than the period in which the number of synthetic membrane-receiver complexes in circulation is reduced to less than about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% of the number of synthetic membrane-receiver complexes present in the administered pharmaceutical composition.
  • the frequency of administration is sufficient to effectively reduce the circulatory concentration of the target complement protein below a level that is associated with a symptom of the disease, disorder or condition associated with the dysregulation of a complement protein.
  • the administering of the pharmaceutical composition reduces the concentration of unbound target complement protein or the concentration of total target complement protein in the circulatory system of the subject.
  • the concentration of total target complement protein is approximately equal to the concentration of unbound and bound target complement protein in the circulatory system of the subject.
  • the method further comprises the step of selecting for treatment a subject suffering from or at risk of a disease, disorder or condition associated with the dysregulation of a complement protein selected from the group consisting of: atypical hemolytic-uremic syndrome (aHUS), paroxysmal nocturnal hemoglobinuria (PNH), age-related macular degeneration, autoimmune hemolytic anemia, complement factor I deficiency, and non-alcoholic steatohepatitis.
  • aHUS atypical hemolytic-uremic syndrome
  • PNH paroxysmal nocturnal hemoglobinuria
  • age-related macular degeneration age-related macular degeneration
  • autoimmune hemolytic anemia complement factor I deficiency
  • non-alcoholic steatohepatitis non-alcoholic steatohepatitis
  • the target complement protein is selected from the group consisting of: C1q, C1r, C1s, C2, C3, C4, C5, C6, C7, C8, and C9.
  • a method of modulating the circulatory concentration of a target metabolite comprises the steps of administering to a mammalian subject suffering from or at risk of developing a metabolic disease, disorder or condition, a pharmaceutical composition comprising a synthetic membrane-receiver complex, wherein the pharmaceutical composition is administered in an amount effective to substantially modulate the circulatory concentration of the target metabolite.
  • the synthetic membrane-receiver complex has a volume of distribution equal to the plasma volume of the subject. In some embodiments, the synthetic membrane-receiver complex has a volume of distribution of less than 0.09 l/kg.
  • the method comprises administering the pharmaceutical composition at least twice over a treatment period such that the metabolic disease, disorder or condition is treated, or a symptom thereof is decreased.
  • the method comprises administering the pharmaceutical composition at least twice over a treatment period such that the metabolic disease, disorder or condition is prevented.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of the target metabolite is substantially decreased during the treatment period.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of a metabolite is substantially increased during the treatment period.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of the target metabolite is substantially decreased during the treatment period such that one or more symptom of the a metabolic disease, disorder or condition is prevented, decreased or delayed.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of a metabolite is substantially increased during the treatment period such that one or more symptom of the a metabolic disease, disorder or condition is prevented, decreased or delayed.
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of the target metabolite is decreased at a rate greater than i) the endogenous clearance rate of the target metabolite by the mammalian subject, or ii) the endogenous production rate of the target metabolite by the mammalian subject, or iii) both i) and ii).
  • the method comprises administering the pharmaceutical composition a sufficient number of times over a treatment period such that the circulatory concentration of a metabolite is increased at a rate greater than i) the endogenous clearance rate of a metabolite by the mammalian subject, or ii) the endogenous production rate of a metabolite by the mammalian subject, or iii) both i) and ii).
  • the circulatory concentration of the target metabolite is decreased by at least about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, 99.99%, or greater than 99.99% during part or the entirety of the treatment period.
  • the circulatory concentration of a metabolite is increased by at least about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, 99.99%, or greater than 99.99% during part or the entirety of the treatment period.
  • the circulatory concentration of the target metabolite is decreased by at least about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, 99.99%, or greater than 99.99% within about 1, 5, 10, 15, 20, 30, 40, or 50 minutes, or about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, or 23 hours, or 1, 2, 3, 4, 5, or 6 days or about 1, 2, 3, 4, 5, or 6 weeks of the administration.
  • the circulatory concentration of a metabolite is increased by at least about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, 99.99%, or greater than 99.99% within about 1, 5, 10, 15, 20, 30, 40, or 50 minutes, or about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, or 23 hours, or 1, 2, 3, 4, 5, or 6 days or about 1, 2, 3, 4, 5, or 6 weeks of the administration.
  • the method comprises administering the pharmaceutical composition a sufficient number of times a treatment period such that the circulatory concentration of the target metabolite is substantially decreased for at least about one week, two weeks, three weeks, four weeks, one month, two months, three months, four months, five months, six months, or greater than six months.
  • the method comprises administering the pharmaceutical composition a sufficient number of times a treatment period such that the circulatory concentration of a metabolite is substantially increased for at least about one week, two weeks, three weeks, four weeks, one month, two months, three months, four months, five months, six months, or greater than six months.
  • the method comprises administering the pharmaceutical composition a sufficient number of times a treatment period such that the circulatory concentration of the target metabolite is substantially decreased for a period of time at least as long as the treatment period.
  • the method comprises administering the pharmaceutical composition a sufficient number of times a treatment period such that the circulatory concentration of a metabolite is substantially increased for a period of time at least as long as the treatment period.
  • the treatment period is not longer than a year, six months, three months, two months, one month, two weeks, one week, three days, two days, one day.
  • the time interval between administrations within a treatment period is no longer than the period in which the number of synthetic membrane-receiver complexes in circulation is reduced to less than about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or 95% of the number of synthetic membrane-receiver complexes present in the administered pharmaceutical composition.
  • the frequency of administration is sufficient to effectively reduce the circulatory concentration of the target metabolite below a level that is associated with a symptom of the metabolic disease, disorder or condition.
  • the frequency of administration is sufficient to effectively increase the circulatory concentration of a metabolite above a level that is associated with a symptom of the metabolic disease, disorder or condition.
  • the administering of the pharmaceutical composition reduces the concentration of unbound target metabolite or the concentration of total target metabolite in the circulatory system of the subject.
  • the concentration of total target metabolite is approximately equal to the concentration of unbound and bound target metabolite in the circulatory system of the subject.
  • the administering of the pharmaceutical composition increases the concentration of an unbound metabolite or the concentration of total metabolite in the circulatory system of the subject.
  • the method further comprises the step of selecting for treatment a subject suffering from or at risk of a metabolic disease, disorder or condition selected from the group consisting of: Phenylketonuria (PKU), Adenosine Deaminase Deficiency-Severe Combined Immunodeficiency (ADA-SCID), Mitochondrial Neurogastrointestinal Encephalopathy (MNGIE), Primary Hyperoxaluria, Alkaptonuria, and Thrombotic Thrombocytopenic Purpura (TTP).
  • PKU Phenylketonuria
  • ADA-SCID Adenosine Deaminase Deficiency-Severe Combined Immunodeficiency
  • MNGIE Mitochondrial Neurogastrointestinal Encephalopathy
  • TTP Thrombotic Thrombocytopenic Purpura
  • the target metabolite is selected from the group consisting of: Phenylalanine, Adenosine, Thymidine, Deoxyuridine, Oxalate, Homogentisate, von Willenbrand Factor.
  • the receiver is associated with the membrane.
  • the receiver is a fusion or a chimera.
  • the fusion or chimera may comprise at least one of an S domain, an A domain or a U domain, wherein the S domain is a surface domain exposed to the environment around the synthetic membrane-receiver complex, wherein the A domain is an anchor, wherein the U domain faces the unexposed side of the synthetic membrane-receiver complex, and wherein the S domain, the A domain, and/or the U domain are of different polypeptide origin.
  • the S domain and/or the A domain comprise a polypeptide comprising at least 6 or at least 30 amino acids.
  • the S domain comprises the antigenic polypeptide or antigenic fragment thereof.
  • the receiver polypeptide is selected from the group consisting of: Phenylalanine Hydroxylase, Adenosine Deaminase, Thymidine Phosphorylase, Glyoxalate Reductase, Homogentisate Reductase, ADAMTS13.
  • the receiver is associated with the membrane.
  • the receiver is a fusion or a chimera with a polypeptide.
  • the fusion or chimera may comprise at least one of an S domain, an A domain or a U domain, wherein the S domain is a surface domain exposed to the environment around the synthetic membrane-receiver complex, wherein the A domain is an anchor, wherein the U domain faces the unexposed side of the synthetic membrane-receiver complex, and wherein the S domain, the A domain, and/or the U domain are of different origin.
  • the S domain and/or the A domain comprise a polypeptide comprising at least 6 or at least 30 amino acids.
  • the S domain comprises the antigenic polypeptide or antigenic fragment thereof.
  • the pharmaceutical compositions described herein comprise a population of synthetic membrane-receiver complexes such as at least 1 ⁇ 10 5 synthetic membrane-receiver complexes, optionally in a volume of about 10 nl, 100 nl, 1 ⁇ l, 10 ⁇ l, 100 ⁇ l, 1 ml, 10 ml, 20 ml, or 50 ml.
  • the pharmaceutical compositions described herein comprise a population of synthetic membrane-receiver complexes such as at least 1 ⁇ 10 11 synthetic membrane-receiver complexes, optionally in a volume of about 1 ml, 10 ml, 20 ml, 50 ml, 100 ml, 250 ml, or 500 ml.
  • provided herein is the synthetic membrane-receiver complex of the pharmaceutical composition administered by the methods disclosed herein.
  • a population of synthetic membrane-receiver complexes as disclosed herein.
  • the population of synthetic membrane-receiver complexes is formulated as a liquid.
  • the population of synthetic membrane-receiver complexes is frozen.
  • an isolated receiver of the synthetic membrane-receiver complex as disclosed herein is an isolated receiver of the synthetic membrane-receiver complex as disclosed herein.
  • provided herein is an exogenous nucleic acid encoding the receiver disclosed herein.
  • a synthetic membrane-receiver complex comprising: a receiver capable of interacting with a target, and a membrane comprising a polypeptide that is not the receiver, wherein the synthetic membrane-receiver complex has catalytic activity independent of the receiver.
  • the synthetic membrane-receiver complex comprises a receiver that is not a polypeptide.
  • any synthetic membrane-receiver complex described herein including those comprising a polypeptide receiver, optionally comprise a payload, such as a therapeutic agent.
  • aspects of the invention relate to isolated, enucleated erythroid cell comprising a receiver polypeptide that is functionally active when the enucleated erythroid cell is administered to the circulatory system of a subject.
  • the erythroid cell is a human cell.
  • aspects of the invention relate to isolated, functional erythroid precursor cell comprising a receiver polypeptide that is encoded by an exogenous nucleic acid, wherein the expression of the receiver polypeptide does not substantially alter: the expression of a surface marker, selected from the group consisting of GPA, cKit, and TR when the functional erythroid precursor cell differentiates; the rate of enucleation when the functional erythroid precursor cell terminally matures; and/or the rate of expansion when the functional erythroid precursor cell expands in culture, wherein the alteration is compared to an isolated, uncultured erythroid precursor cell of the same stage and lineage not comprising the polypeptide receiver.
  • a surface marker selected from the group consisting of GPA, cKit, and TR when the functional erythroid precursor cell differentiates
  • the rate of enucleation when the functional erythroid precursor cell terminally matures
  • the rate of expansion when the functional erythroid precursor cell expands in culture
  • aspects of the invention relate to isolated erythroid cell populations comprising a plurality of functional erythroid cells comprising a receiver polypeptide localized to an exterior surface of the erythroid cells, wherein the population is substantially free of non-erythroid cells.
  • the population comprises greater than 5-95% of enucleated erythroid cells.
  • aspects of the invention relate to isolated erythroid cell populations comprising a plurality of functional erythroid cells comprising a receiver polypeptide encoded by an exogenous nucleic acid, wherein during enucleation the receiver polypeptide is retained by the erythroid cell whereas the exogenous nucleic acid is not retained.
  • the population comprises greater than 5-95% of enucleated erythroid cells, optionally in the absence of: i) an enrichment step and/or ii) co-culturing with non-erythroid cells.
  • aspects of the invention relate to isolated erythroid cell populations comprising a plurality of functional erythroid cells comprising a receiver polypeptide encoded by an exogenous nucleic acid, wherein during enucleation the receiver polypeptide is retained by the erythroid cell whereas the exogenous nucleic acid is not retained, and wherein the resulting functional enucleated erythroid cell exhibits substantially the same osmotic membrane fragility as an isolated, uncultured erythroid cell not comprising the polypeptide receiver.
  • aspects of the invention relate to isolated erythroid cell populations comprising a plurality of functional erythroid precursor cells in substantially the same stage of differentiation and/or cell cycle stage, wherein the precursor cells comprise an exogenous nucleic acid encoding a receiver polypeptide, and wherein a majority of erythrocyte precursor cells is capable of differentiating into mature erythrocytes that retain the receiver polypeptide without retaining the exogenous nucleic acid.
  • aspects of the invention relate to isolated erythroid cell populations comprising a plurality of functional erythroid cells comprising a receiver polypeptide, wherein an exogenous nucleic acid encoding the receiver polypeptide is introduced into a cultured or freshly isolated erythroid cell precursor and wherein after introduction of the exogenous nucleic acid the functional erythroid cells expand from the precursor cells by more than 20,000-fold in culture.
  • the population comprises greater than 5-95% of enucleated erythroid cells, optionally in the absence of: i) an enrichment step and/or ii) co-culturing with non-erythroid cells.
  • aspects of the invention relate to an isolated erythroid cell population that is cultured from a functional erythrocyte precursor cell comprising an exogenous nucleic acid, the population comprising: a pyrenocyte, a functional nucleated erythroid cell and a functional enucleated erythroid cell, wherein the functional nucleated erythroid cell and the functional enucleated erythroid cell comprise an receiver polypeptide encoded by the exogenous nucleic acid, and wherein the receiver polypeptide is retained by the functional enucleated erythroid cell, whereas the exogenous nucleic acid is not retained by the enucleated erythroid cell.
  • the enucleated, functional erythroid cell exhibits substantially the same osmotic membrane fragility as an isolated, uncultured erythroid cell not comprising the polypeptide receiver.
  • the erythroid cell populations described herein comprise greater than 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9% or greater than 10% fetal hemoglobin.
  • the functional erythroid cell exhibits at least 10 copies, 100 copies, 1,000 copies, 10,000 copies, 25,000 copies, 50,000 copies, or 100,000 copies of the receiver polypeptide per cell.
  • a plurality of functional erythroid cells loses a substantial portion of its cell membrane after being administered to the circulatory system of a subject.
  • the functional erythroid cells comprise a receiver polypeptide that interacts with a target.
  • interacting with a target comprises binding to the target, degrading the target, cleaving the target, and/or sequestering the target.
  • the receiver polypeptide is displayed on the cell surface. In other embodiments, the receiver polypeptide is localized in the interior of the functional erythroid cell.
  • the functional erythroid cells comprise a receiver polypeptide that is selected from the group consisting of: an antibody, a single-chain variable fragment, a nanobody, a diabody, a darbin, a lyase, a hydrolase, a protease, a nuclease, and a DNase.
  • the functional erythroid cells comprise a receiver polypeptide that interacts with a target that is selected from the group consisting of: an immune complex, an inflammatory molecule, an inflammatory cell, a lipid, a carbohydrate, an amino acid, a virus, a bacterium, a bacterial toxin, a fungus, a fungal toxin, a DNA, an RNA, a cell, a circulating cell, a tumor cell, a metastatic cancer cell, a metabolite, a plant toxin, a cytokine, a chemokine, a complement cascade factor, and a clotting cascade factor.
  • a target that is selected from the group consisting of: an immune complex, an inflammatory molecule, an inflammatory cell, a lipid, a carbohydrate, an amino acid, a virus, a bacterium, a bacterial toxin, a fungus, a fungal toxin, a DNA, an RNA,
  • the functional erythroid cells comprise a receiver polypeptide that is fused to an endogenous polypeptide.
  • the endogenous polypeptide is an intracellular polypeptide.
  • the endogenous polypeptide is an extracellular polypeptide.
  • the endogenous polypeptide is membrane-bound.
  • the functional erythroid cells comprise a receiver polypeptide that is fused to an endogenous extracellular polypeptide.
  • the functional erythroid cells comprise a receiver polypeptide that conjugated to the erythroid cell.
  • the functional erythroid cells comprise a receiver polypeptide that interacts with the target intercellularly.
  • the functional erythroid cells comprise a receiver polypeptide that is localized in the cytosol of the erythroid cell.
  • the functional erythroid cells comprise a receiver polypeptide that is located in the cell membrane of the erythroid cell.
  • the functional erythroid cells comprise a plurality of receiver polypeptides.
  • a first receiver polypeptide is located in the cytosol of the functional erythroid cell and a second receiver polypeptide is located on the cell surface of the functional erythroid cell.
  • the functional erythroid cells comprise a receiver polypeptide that is an Fv portion of an antibody that binds a botulinum toxin and the target is a botulinum toxin.
  • the functional erythroid cells comprise a receiver polypeptide that is a complement receptor 1 and the target is a circulating immune complex.
  • the functional erythroid cells comprise a receiver polypeptide that is a duffy antigen receptor complex (DARC) and the target is a circulating chemokine.
  • DARC duffy antigen receptor complex
  • the functional erythroid cells comprise a receiver polypeptide that is phenylalanine hydroxylase (PAH) and the target is phenylalanine.
  • PAH phenylalanine hydroxylase
  • the functional erythroid cells comprise a receiver polypeptide that is expressed as a fusion of the C-terminus of a cytoplasmic beta globin protein.
  • the functional erythroid cells comprise a receiver polypeptide that is an exonuclease and wherein the target is a circulating cell-free DNA molecule.
  • the functional erythroid cells comprise a receiver polypeptide that is expressed as a fusion of the N-terminus of endogenous glycophorin A.
  • the functional erythroid cells comprise a receiver polypeptide that is attached extracellularly on the erythroid cell by covalent bond formation.
  • the covalent bond is formed by an isopeptidase.
  • the isopeptidase is SpyTag/SpyCatcher.
  • the SpyTag is expressed on the surface of the cell.
  • the SpyTag is fused to an extracellular terminus of a transmembrane protein.
  • the SpyTag is an in-frame fusion in an extracellular region of a multi-pass membrane protein.
  • the SpyTag is fused to a GPI-linked protein.
  • the SpyCatcher is fused to the receiver polypeptide.
  • the receiver polypeptide fused to SpyCatcher is expressed and/or secreted in the same functional erythroid cell that expresses the SpyTag fusion.
  • the receiver polypeptide fused to SpyCatcher is expressed by an exogenous protein production system and then contacted with the functional erythroid cell that expresses the SpyTag fusion.
  • the SpyTag is replaced with SpyCatcher and the SpyCatcher is replaced with SpyTag.
  • the receiver polypeptide is anchored intracellularly in the functional erythroid cell by covalent bond formation.
  • the covalent bond is formed by an isopeptidase.
  • the isopeptidase is SpyTag/SpyCatcher.
  • the SpyTag is expressed in the intracellular space of the cell.
  • the SpyTag is fused to an intracellular terminus of a membrane protein.
  • the SpyTag is an in-frame fusion in an intracellular region of a multi-pass membrane protein.
  • the SpyTag is fused to an endogenous intracellular protein.
  • the SpyTag is fused to a cytoskeletal protein.
  • the SpyCatcher is fused to the receiver polypeptide.
  • the receiver polypeptide fused to SpyCatcher is expressed in the intracellular space of the same functional erythroid cell that expresses the SpyTag fusion.
  • the SpyTag is replaced with SpyCatcher and the SpyCatcher is replaced with SpyTag.
  • aspects of the invention relate to methods of generating functional erythroid cells comprising a receiver polypeptide, the methods comprising contacting an erythroid cell with a receiver and exposing the erythroid cell to a controlled cell injury.
  • the controlled cell injury is cell deformation, electroporation, sonoporation, liposomal transfection, or salt-based transfection.
  • the cell is contacted with an mRNA that encodes the receiver polypeptide.
  • the contacting results in an uptake and translation of the mRNA encoding the receiver polypeptide by the erythroid cell or erythriod cell precursor.
  • the populations of erythroid cells described herein are maintained and/or propagated in vitro. In other embodiments, the populations of erythroid cells described herein are lyophilized. In yet other embodiments, the populations of erythroid cells described herein are frozen.
  • aspects of the invention relate to methods of contacting a target comprising: introducing into a biological sample or a subject the erythroid cell populations described herein, and maintaining the contact of the erythroid cell population with the sample or subject for a time sufficient for a functional erythroid cell from the population to interact with a target in the sample or subject.
  • interacting with a target comprises binding to the target, degrading the target, cleaving the target, and/or sequestering the target.
  • the methods of contacting a target are carried out in vitro.
  • the methods of contacting a target are carried out in vivo, e.g. in an animal.
  • the methods of contacting a target further comprise contacting the target with an assayable moiety.
  • the assayable moiety is used to determine the rate and/or degree of interaction between the functional erythroid cell and the target.
  • compositions comprising the erythroid cell populations comprising the functional erythroid cells comprising a receiver described herein.
  • the pharmaceutical compositions comprising the erythroid cell populations further comprise a pharmaceutically acceptable carrier.
  • the pharmaceutical compositions comprising the erythroid cell populations further comprise a therapeutic agent.
  • aspects of the invention relate to methods of treating, preventing, or managing a disease or condition, comprising administering to a subject in need of such treatment, prevention or management, a therapeutically or prophylactically effective amount of the pharmaceutical composition comprising a population of functional erythroid cells comprising a receiver, thereby treating, preventing, or managing the disease or condition.
  • compositions comprising a population of functional erythroid cells comprising a receiver for use in any of the methods of treatment or prevention described herein.
  • the receiver polypeptide interacts with a target residing in the circulatory system of the subject.
  • the presence, absence, elevated or depressed level of the target is associated with a disease, disorder or condition.
  • interacting with a target comprises binding to the target, degrading the target, cleaving the target, and/or sequestering the target.
  • the administration of the pharmaceutical compositions comprising a population of functional erythroid cells comprising a receiver results in a substantial reduction of the concentration or number of the target in the circulatory system of the subject.
  • compositions comprising a plurality of functional erythroid cells comprising a receiver polypeptide, wherein the erythroid cells exhibit the receiver polypeptide in or on the cell, and wherein the receiver polypeptide when the functional erythroid cell is administered to the circulatory system of a subject: does not substantially affect the circulation clearance time of the functional erythroid cell when compared to a unmodified erythroid cell in a control animal, and/or does not activate fibrinogen breakdown, measured by circulating levels of fibrinopeptide A and/or fibrinopeptide B, compared to an unmodifed erythroid cell.
  • aspects of the invention relate to methods for culturing the functional erythroid cell population of described herein, comprising using one or more culturing factors selected from the group consisting of stem cell factor, IL-3, IL-6, insulin, transferrin, erythropoietin, hydrocortisone, and estrogens to culture the functional erythroid cells.
  • aspects of the invention relate to populations of at least 10 10 cells comprising at least 10% reticulocytes of the same blood group, wherein a plurality of the reticulocytes comprises a receiver polypeptide.
  • aspects of the invention relate to a pharmaceutical composition comprising a synthetic membrane-receiver polypeptide complex for use in the treatment of any of the diseases, disorders, or conditions disclosed herein.
  • aspects of the invention relate to a pharmaceutical composition comprising a synthetic membrane-receiver polypeptide complex for use in the treatment of a disease, disorder, or condition associated with the presence of or the concentration of a target in the circulatory system of a mammalian subject.
  • aspects of the invention relate to a pharmaceutical composition
  • a pharmaceutical composition comprising a synthetic membrane-receiver polypeptide complex for use in the modulation of the circulatory concentration of a target.
  • FIG. 1 is a collection of flow cytometry plots of red blood cells contacted with fluorescently labeled IgG encapsulated within liposomes.
  • Cells are shown that are incubated with no liposomes (A, D), a low dose of liposomes (B, E), and a high dose of liposomes (C, F).
  • A, D no liposomes
  • B, E low dose of liposomes
  • C, F high dose of liposomes
  • FIG. 2 is a plot of cell surface expression levels assessed by quantitative flow cytometry. The plot shows of various cell surface receptors—glycophorin A (solid triangles), cKIT (dashed squares), transferrin receptor (dotted diamonds)—and an exogenous surface transgene (open circles) during the course of erythroid cell differentiation.
  • various cell surface receptors glycophorin A (solid triangles), cKIT (dashed squares), transferrin receptor (dotted diamonds)—and an exogenous surface transgene (open circles) during the course of erythroid cell differentiation.
  • FIG. 3A-FIG . 3 AP is a collection of flow cytometry plots and Western blots that demonstrate the expression of a vast array of exemplary receivers on the surface, in the cytoplasm, as fusions, and as intact proteins, in three cell types, enucleated erythroid cells, nucleated erythroid precursor cells, and erythroleukemic cells.
  • FIG. 3A-FIG . 3 N shows the exogenous expression of surface and cytoplasmic proteins on enucleated cultured erythroid cells.
  • FIG. 3 Example of glycophorin A with an HA epitope tag at the cytoplasmic C terminus assessed by expression of co-translated GFP.
  • FIG. 3 B Expression of glycophorin A with an HA epitope tag at the N terminus between the leader sequence and the body of the gene assessed by anti-HA staining.
  • FIG. 3 C Example of complement receptor 1-derived fragment of ⁇ 70 kDa with an HA epitope tag at the N terminus assessed by anti-HA staining.
  • FIG. 3 D Example of antibody scFv as N terminal fusion to glycophorin A assessed by anti-HA staining.
  • FIG. 3 E Example of antibody scFv fused to C terminus of Kell-derived fragment of 71 amino acids assessed by anti-HA staining.
  • FIG. 3 F—Expression of antibody scFv fused to C terminus of Kell-derived fragment of 79 amino acids assessed by anti-HA staining.
  • FIG. 3 G Expression of CD55 with HA epitope tag at the extracellular N terminus after the leader sequence assessed by anti-HA staining.
  • FIG. 3 H Expression of CD59 with HA epitope tag at the extracellular N terminus after the leader sequences assessed by anti-HA staining.
  • FIG. 3 I Example of antibody scFv fused to N-terminus of CD55-derived fragment of 37 amino acids, assessed by anti-HA Western blot.
  • FIG. 3 J—Cytoplasmic expression of adenosine deaminase fused to HA tag assessed by anti-HA Western blot. Expected size approximately 40 kDa.
  • FIG. 3 K—Cytoplasmic expression of phenylalanine hydroxylase fused to HA tag assessed by anti-HA Western blot. Expected size approximately 33 kDa.
  • FIG. 3 L—Cytoplasmic expression of phenylalanine hydroxylase fused to adenosine deaminase and an HA tag assessed by anti-HA Western blot.
  • FIG. 3 M—Cytoplasmic expression of adenosine deaminase fused to the intracellular C terminus of glycophorin A assessed by anti-HA Western blot. Expected size approximately 55 kDa.
  • FIG. 3 N—Cytoplasmic expression of phenylalanine hydroxylase fused to the intracellular C terminus of glycophorin A assessed by anti-HA Western blot. Expected size approximately 50 kDa.
  • FIG. 3O-Fig . AJ shows the exogenous expression of surface and cytoplasmic proteins on nucleated cultured erythroid precursor cells.
  • FIG. 3 O—Expression of glycophorin A with an HA epitope tag at the cytoplasmic C terminus assessed by expression of co-translated GFP.
  • FIG. 3 P—Expression of glycophorin A with an HA epitope tag at the N terminus between the leader sequence and the body of the gene assessed by anti-HA staining.
  • FIG. 3 Q Overexpression of complement receptor 1 assessed by anti-CR1 staining.
  • FIG. 3 Expression of complement receptor 1-derived fragment of ⁇ 70 kDa with an HA epitope tag at the N terminus assessed by anti-HA staining.
  • FIG. 3 S—Expression of complement receptor 1-derived fragment of ⁇ 210 kDa with an HA epitope tag at the N terminus assessed by anti-HA staining.
  • FIG. 3 T—Expression of complement receptor 1-derived fragment of ⁇ 230 kDa fused to the N terminus of glycophorin A with an HA epitope tag at the N terminus assessed by anti-HA staining.
  • FIG. 3 U—Expression of antibody scFv as N terminal fusion to glycophorin A assessed by anti-HA staining.
  • FIG. 3 Example of antibody scFv fused to the extracellular C terminus of Kell, assessed by anti-HA staining. Expected size approximately 108 kDa.
  • FIG. 3 W Example of HA tag fused to the extracellular C terminus of Kell, assessed by anti-HA staining.
  • FIG. 3 X Example of Kell-derived fragment of 71 amino acids with HA tag at the C (extracellular) terminus assessed by anti-HA staining.
  • FIG. 3 Y Expression of Kell-derived fragment of 79 amino acids with HA tag at the C terminus assessed by anti-HA staining.
  • FIG. 3 Z Expression of antibody scFv fused to C terminus of Kell-derived fragment of 71 amino acids assessed by anti-HA staining.
  • FIG. 3 AA Example of antibody scFv fused to C terminus of Kell-derived fragment of 79 amino acids assessed by anti-HA staining.
  • FIG. 3 AB—Expression of CD55 with HA epitope tag at the extracellular N terminus after the leader sequence assessed by anti-HA staining.
  • FIG. 3 AC—Expression of CD59 with HA epitope tag at the extracellular N terminus after the leader sequences assessed by anti-HA staining.
  • FIG. 3 AD—Expression of antibody scFv fused to N-terminus of CD55-derived fragment of 37 amino acids, assessed by anti-HA staining.
  • FIG. 3 AE Example of antibody scFv fused to N-terminus of CD59 assessed by anti-HA staining.
  • FIG. 3 AF—Cytoplasmic expression of adenosine deaminase fused to HA tag assessed by anti-HA Western blot. Expected size approximately 40 kDa.
  • FIG. 3 Cytoplasmic expression of phenylalanine hydroxylase fused to HA tag assessed by anti-HA Western blot. Expected size approximately 33 kDa.
  • FIG. 3 AH—Cytoplasmic expression of phenylalanine hydroxylase fused to adenosine deaminase and an HA tag assessed by flow cytometry for fluorescence from co-translated GFP.
  • FIG. 3 Cytoplasmic expression of adenosine deaminase fused to the intracellular C terminus of glycophorin A assessed by anti-HA Western blot. Expected size approximately 55 kDa.
  • FIG. 3 AJ—Cytoplasmic expression of phenylalanine hydroxylase fused to the intracellular C terminus of glycophorin A assessed by anti-HA Western blot. Expected size approximately 50 kDa.
  • FIG. 3 AK-FIG. 3 AP shows the exogenous expression of surface and cytoplasmic proteins on K562 erythroleukemia cells.
  • FIG. 3 AK Overexpression of complement receptor 1 assessed by anti-CR1 staining.
  • FIG. 3 AL—Expression of antibody scFv as N terminal fusion to glycophorin A assessed by anti-HA staining.
  • FIG. 3 AM—Expression of antibody scFv fused to N-terminus of CD55-derived fragment of 37 amino acids, assessed by anti-HA staining.
  • FIG. 3 AN—Expression of antibody scFv fused to N-terminus of CD59 assessed by anti-HA staining.
  • FIG. 3 AO—Cytoplasmic expression of adenosine deaminase fused to HA tag assessed by anti-HA Western blot. Expected size approximately 40 kDa.
  • FIG. 3 AP—Cytoplasmic expression of phenylalanine hydroxylase fused to HA tag assessed by anti-HA Western blot. Expected size approximately 33 kDa.
  • FIG. 4 is a collection of flow cytometry histograms that measure fluorescence in primary platelets that have been transfected with mRNA encoding a fluorescent protein (GFP).
  • A Untransfected platelets.
  • B Platelets transfected with 3 ug GFP mRNA.
  • C Platelets transfected with 6.8 ug GFP mRNA.
  • FIG. 5 shows protein expression and enzymatic activity of transgenic erythroid cells in culture.
  • A is a Western blot of exogenously expressed adenosine deaminase detected with an anti-HA antibody over the course of differentiation, from nucleated precursor cells (“Diff I D5”) through to enucleated erythroid cells (“Diff III D8”).
  • B is a bar chart of inosine produced from adenosine by intact adenosine deaminase-expressing 293T cells.
  • (C) is a Western blot of the exogenously expressed phenylalanine hydroxylase detected with an anti-HA antibody at various time points over the course of differentiation, from nucleated precursor cells (“Diff I D5”) through to enucleated erythroid cells (“Diff III D8”).
  • D is a bar chart of tyrosine produced from phenylalanine by lysates of cultured phenylalanine hydroxylase-expressing enucleated erythroid cells.
  • FIG. 6 shows immune complex capture and transfer to macrophages by cultured erythroid cells that overexpress complement receptor 1 (CR1).
  • A is a flow cytometry plot that shows the capture of fluorescent immune complexes (white histogram) and complement-deficient immune complexes (shaded histogram) by cultured erythroid cells that overexpress CR1.
  • B is a bar chart of flow cytometry data assessing the uptake of fluorescent immune complexes (hashed bars), complement deficient immune complexes (gray bars), or no immune complexes (black bars) by macrophages (left set) or macrophages incubated with cultured erythroid cells that overexpress CR1 (right set).
  • FIG. 7 shows the activity of an antibody scFv that binds hepatitis B surface antigen (scFv) on the surface of a cultured erythroid cell.
  • A is a flow cytometry histogram showing binding of 450 nM antigen (white histogram) or no antigen (gray histogram).
  • B is a titration of binding signal assessed by flow cytometry for a range of antigen concentrations.
  • C-D are flow cytometry plots of blood cells from mice that had been injected with fluorescent antigen and cultured erythroid cells that (C) do not or (D) do express scFv.
  • the y-axis measures antigen fluorescence.
  • the x-axis measures fluorescence of the cultured cells.
  • FIG. 8 shows the specific clearance of circulating antibodies mediated by membrane-receiver complexes in vivo.
  • A is a set of flow cytometry plots that show no binding (right) and binding (left) of circulating Dylight650-labeled mouse anti-HA antibody to CFSE-labeled cultured human erythroid cells isolated from a recipient mouse that either do not (right) or do (left) express HA epitope tag on their surface.
  • the x-axis measures CFSE fluorescence.
  • the y-axis measures anti-HA antibody Dylight650 fluorescence.
  • (B) is data from an HA epitope tag substrate ELISA comparing anti-HA antibody levels over time in plasma collected from mice injected with anti-HA antibody (open circles, solid line), anti-HA antibody followed by cultured human erythroid cells that do not express HA epitope tag (dashed line), or anti-HA antibody followed by cultured human erythroid cells that do express HA epitope tag (dotted line).
  • (C) is a set of flow cytometry plots that show no binding (right) and binding (left) of Dylight650-labeled mouse anti-biotin antibody to CFSE-labeled primary human erythrocytes that either are not (right) or are (left) conjugated with biotin on their surface.
  • the x-axis measures CFSE fluorescence.
  • the y-axis measures anti-biotin antibody Dylight650 fluorescence.
  • D is data from a biotin substrate ELISA comparing anti-biotin antibody levels over time in plasma collected from mice injected with anti-biotin antibody (open circles, solid line), anti-biotin antibody followed by cultured human erythroid cells that are not conjugated to biotin (dashed line), or anti-biotin antibody followed by cultured human erythroid cells that are conjugated to biotin (dotted line).
  • FIG. 9 shows the clearance rate of cultured human eyrthroid cells in a mouse.
  • A is a representative flow cytometry dot-plot of drawn blood, stained for human glycophorin A (y-axis) and CFSE (x-axis), in which human cultured cells are double-positive.
  • B is a plot of the clearance rate over time as a percentage of double-positive cells remaining after NSG mice were injected with human red blood cells (solid circles), cultured enucleated erythroid cells (dashed diamonds), cultured enucleated erythroid cells that express an intracellular exogenous protein (dotted squares) and cultured enucleated erythroid cells that express a surface exogenous protein (open triangles).
  • FIG. 10 is an assessment of adverse events following injection of cultured human erythroid cells into a mouse.
  • A-B show levels of (A) fibrinopeptide A and (B) fibrinopeptide B assessed by ELISA in plasma collected from mice 20 minutes (black), 6 hours (gray), and 48 hours (white) after injection with (1) human red blood cells, (2) cultured human erythroid cells, (3) cultured human erythroid cells expressing an exogenous cytoplasmic protein, (4) cultured human erythroid cells expressing an exogenous surface transgene, or (5) recombinant protein.
  • C-D show microscope images of histologically stained sections of spleen for mice injected with (C) cultured human erythroid cells and (D) recombinant protein.
  • FIG. 11 tracks the expression of exogenous protein on cultured erythroid cells in circulation.
  • A is flow cytometry data of blood drawn from a mouse that was injected with cultured human erythroid cells expressing an exogenous surface protein, showing the percent of cultured human erythroid cells that are HA-positive over time.
  • B is a Western blot of blood drawn from two mice, wherein one mouse was injected with cultured human erythroid cells expressing an exogenous cytoplasmic protein, and wherein the other mouse was injected with the purified recombinantly-produced exogenous protein in the absence of any cells, showing the level of HA-containing protein in the blood over time.
  • FIG. 12 is an assessment of expansion and differentiation of cultured human erythroid cells.
  • A is a plot of expansion rates for distinct cultures of in vitro differentiated erythroid cells that contain transgenes (dashed line and dotted line) and cells that do not contain a transgene (solid line).
  • B is a flow cytometry plot of cell surface markers GPA and CKIT for distinct cultures of cultured human erythroid cells that do not (left) or do (right) contain a transgene.
  • (C) is a flow cytometry plot of cultured human erythroid cells that do not (left) or do (right) contain a transgene, wherein the cells are stained with DNA stain DRAQ5 (y-axis) and anti-glycophorin A (x-axis), which identifies distinct populations of (1) enucleated cells, (2) nucleated cells, and (3) nuclei.
  • FIG. 13A is a schematic of a synthetic membrane-receiver complex comprising a receiver polypeptide.
  • the left panel depicts the flux of a target substrate across the membrane of the synthetic membrane-receiver complex.
  • the target substrate is altered by an internally localized enzymatic receiver polypeptide and the resulting product of the enzymatic reaction either remains in the synthetic membrane-receiver complex or exits through the membrane.
  • the right panel depicts a synthetic membrane-receiver complex that contains at least two receivers (e.g., receiver polypeptides), one being localized on the surface and one being internally localized.
  • the surface-localized receiver aids a substrate to enter the synthetic membrane-receiver complex, e.g., by carrying out a transporter function.
  • the second receiver localized internally, alters the substrate enzymatically.
  • the resulting product of the enzymatic reaction either remains in the synthetic membrane-receiver complex or exits through the membrane, optionally aided by the first surface-localized receiver.
  • FIG. 13B is a schematic of another synthetic membrane-receiver complex comprising a receiver polypeptide.
  • FIG. 13B depicts a receiver polypeptide localized on the surface of the synthetic membrane-receiver complex.
  • a target substrate can be acted upon directly by the receiver.
  • the target substrate does not need to cross the membrane to be enzymatically converted to a product.
  • the surface-localized enzymatic receiver polypeptide can be made cleavable, e.g., if the complex enters a specific microenvironment. In that instance, the receiver polypeptide will be cleaved and become active in the extracellular space.
  • FIG. 13C is a schematic of yet another synthetic membrane-receiver complex comprising a receiver.
  • FIG. 13C depicts the lysis of a synthetic membrane-receiver complex containing internally-localized receiver (e.g., a polypeptide) and optional payload (e.g., a therapeutic agent) which may result from a variety of stimuli.
  • internally-localized receiver e.g., a polypeptide
  • optional payload e.g., a therapeutic agent
  • FIG. 14A is a schematic of three ways in which a receiver may be localized in a synthetic membrane-receiver complex.
  • FIG. 14B is a schematic of three ways in which a receiver localized in or on a synthetic membrane-receiver complex may act on a target in circulation.
  • FIG. 14C is a schematic of an auto-catalytic fusion of an endogenous polypeptide anchor to a receiver utilizing a SpyTag-SpyCatcher mechanism.
  • aspects of the invention relate to compositions and methods for performing, e.g., functions related to circulating clearance and functions related to metabolic enzyme delivery, and methods for treating or preventing a variety of diseases, disorders and conditions.
  • compositions and methods disclosed herein address the long sought after need for therapeutic compositions that are distributed through the circulatory system that have increased half-life, safety profile, and/or efficacy that avoid shortcomings associated with previous approaches such as undesirable immunological reactions, short half-life due to rapid clearance from the circulation, and off-target effects, among others.
  • Functions related to circulating clearance include activities characterized by, e.g., the specific binding, degradation, and/or sequestration of a target (e.g., a pathogenic substance or toxic molecule) in the circulatory system of a subject by a synthetic membrane-receiver complex comprising a receiver capable of interacting with a target as described herein.
  • a target e.g., a pathogenic substance or toxic molecule
  • Synthetic membrane-receiver complexes are introduced or capable of being introduced into the circulation of a subject.
  • the bound or sequestered targets are guided to the liver, spleen, or any other site in which they may be removed from the circulatory system.
  • Functions related to metabolic enzyme delivery include activities characterized by, e.g., removal of a target (e.g., a pathogenic substance or toxic molecule), in circulation of a subject by a synthetic membrane-receiver complex as described herein that comprises, e.g., one or more metabolic enzyme receiver polypeptides within the complex or on the surface of the complex, such that the receiver polypeptide interacts with and modifies the target.
  • Modification of the target includes, e.g., alteration of the bioavailability of the target, cleaving, degrading, and/or otherwise inactivating the target by the receiver.
  • the enzymatic polypeptide is protected from the immune system.
  • the half-life of the enzyme is extended and/or an immunogenic reaction is reduced when administered in the subject.
  • any concentration range, percentage range, ratio range, or integer range is to be understood to include the value of any integer within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an integer), unless otherwise indicated.
  • administering means introducing a composition, such as a synthetic membrane-receiver complex, or agent into a subject and includes concurrent and sequential introduction of a composition or agent.
  • the introduction of a composition or agent into a subject is by any suitable route, including orally, pulmonarily, intranasally, parenterally (intravenously, intramuscularly, intraperitoneally, or subcutaneously), rectally, intralymphatically, or topically.
  • Administration includes self-administration and the administration by another.
  • a suitable route of administration allows the composition or the agent to perform its intended function. For example, if a suitable route is intravenous, the composition is administered by introducing the composition or agent into a vein of the subject. Administration can be carried out by any suitable route,
  • “Anchor” or “anchor domain” or “A domain” is used to refer to the portion of a receiver polypeptide, including a fusion or chimeric receiver polypeptide that is in contact with the lipid layer of a synthetic membrane-receiver polypeptide complex.
  • the receiver polypeptide may interact with the lipid layer via a phospholipid tail insertion, covalent binding to a lipid layer constituent, an ionic bond, hydrogen bond, or via a single or multi-pass transmembrane polypeptide domain that cross one or more of the lipid layers.
  • antibody encompasses an immunoglobulin whether natural or partly or wholly synthetically produced, and fragments thereof.
  • the term also covers any protein having a binding domain which is homologous to an immunoglobulin binding domain. These proteins can be derived from natural sources, or partly or wholly synthetically produced.
  • Antibody further includes a polypeptide comprising a framework region from an immunoglobulin gene or fragments thereof that specifically binds and recognizes an antigen.
  • antibody is meant to include whole antibodies, polyclonal, monoclonal and recombinant antibodies, fragments thereof, and further includes single-chain antibodies, humanized antibodies; murine antibodies; chimeric, mouse-human, mouse-primate, primate-human monoclonal antibodies, anti-idiotype antibodies, antibody fragments, such as, e.g., scFv, (scFv)2, Fab, Fab′, and F(ab′)2, F(ab1)2, Fv, dAb, and Fd fragments, diabodies, and antibody-related polypeptides.
  • Antibody includes bispecific antibodies and multispecific antibodies so long as they exhibit the desired biological activity or function.
  • antigen binding fragment refers to fragments of an intact immunoglobulin, and any part of a polypeptide including antigen binding regions having the ability to specifically bind to the antigen.
  • the antigen binding fragment may be a F(ab′)2 fragment, a Fab′ fragment, a Fab fragment, a Fv fragment, or a scFv fragment, but is not limited thereto.
  • a Fab fragment has one antigen binding site and contains the variable regions of a light chain and a heavy chain, the constant region of the light chain, and the first constant region CH1 of the heavy chain.
  • a Fab′ fragment differs from a Fab fragment in that the Fab′ fragment additionally includes the hinge region of the heavy chain, including at least one cysteine residue at the C-terminal of the heavy chain CH1 region.
  • the F(ab′)2 fragment is produced whereby cysteine residues of the Fab′ fragment are joined by a disulfide bond at the hinge region.
  • a Fv fragment is the minimal antibody fragment having only heavy chain variable regions and light chain variable regions, and a recombinant technique for producing the Fv fragment is well known in the art.
  • Two-chain Fv fragments may have a structure in which heavy chain variable regions are linked to light chain variable regions by a non-covalent bond.
  • Single-chain Fv (scFv) fragments generally may have a dimer structure as in the two-chain Fv fragments in which heavy chain variable regions are covalently bound to light chain variable regions via a peptide linker or heavy and light chain variable regions are directly linked to each other at the C-terminal thereof.
  • the antigen binding fragment may be obtained using a protease (for example, a whole antibody is digested with papain to obtain Fab fragments, and is digested with pepsin to obtain F(ab′)2 fragments), and may be prepared by a genetic recombinant technique.
  • a dAb fragment consists of a VH domain.
  • Single-chain antibody molecules may comprise a polymer with a number of individual molecules, for example, dimmer, trimer or other polymers.
  • Applicator refers to any device used to connect to a subject. This includes, e.g., needles, cannulae, catheters, and tubing.
  • Associated with when used to describe the relationships among multiple compounds or molecules encompasses such as, e.g., any interaction between a receiver and a target or between a synthetic membrane-receiver complex and a target. This includes enzymatic interaction, ionic binding, covalent binding, non-covalent binding, hydrogen bonding, London forces, van der Waals forces, hydrophobic interaction, lipophilic interactions, magnetic interactions, electrostatic interactions, and the like.
  • Associated with when used to describe the relationships among a target, entity, compound, agent, or molecule and a disease, disorder, condition, symptom or phenotype is any link that may reasonably be made between them, including a causal link, or a statistical significant link, an empirically established link, a suggested link, whether or not causative of the disease, disorder, condition, symptom or phenotype.
  • Autoimmune disorders generally are conditions in which a subject's immune system attacks the body's own cells, causing tissue destruction. Autoimmune disorders may be diagnosed using blood tests, cerebrospinal fluid analysis, electromyogram (measures muscle function), and magnetic resonance imaging of the brain, but antibody testing in the blood, for self-antibodies (or auto-antibodies) is particularly useful. Usually, IgG class antibodies are associated with autoimmune diseases.
  • Binding describes an interaction among compounds or molecules, e.g., between a receiver and a target or between a synthetic membrane-receiver complex and a target, that comes about by covalent binding or non-covalent binding, including ionic binding, electrostatic interactions, hydrogen bonding, London forces, van der Waals forces, hydrophobic interaction, lipophilic interactions, and similar.
  • biological activity of a polypeptide refers to any molecular activity or phenotype (such as, e.g., binding, signal transduction, catalytic, etc.) that is caused by the polypeptide, such as a receiver polypeptide.
  • biological sample refers to any type of material of biological origin isolated from a subject, including, for example, DNA, RNA, lipids, carbohydrates, and protein.
  • biological sample includes tissues, cells and biological fluids isolated from a subject.
  • Biological samples include, e.g., but are not limited to, whole blood, plasma, serum, semen, saliva, tears, urine, fecal material, sweat, buccal, skin, cerebrospinal fluid, bone marrow, bile, hair, muscle biopsy, organ tissue or other material of biological origin known by those of ordinary skill in the art.
  • Biological samples can be obtained from, e.g., biopsies of internal organs or from cancers.
  • Biological samples can be obtained from subjects for diagnosis or research or can be obtained from healthy subjects, as controls or for basic research.
  • the “clearance rate” as used herein is calculated by measuring the amount or concentration of, e.g., target, receiver, target-receiver, or synthetic membrane-receiver complexes remaining in the circulatory system of a subject over time. For example, 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99% of target detected in a first sample may still be detected in a second sample that is taken 1 hour, 5 hours, 10 hours, 24 hours, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 2 weeks, 3 weeks, 4 weeks, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 12 months, 2 years, 3 years, 4 years, or 5 years later.
  • the clearance rate may alternatively be expressed as: number of entities (e.g., target/receiver) per unit of time (e.g., per day).
  • An increase in clearance rate is a rate greater than that exhibited in an untreated or healthy suitable control subject.
  • a decrease in clearance rate is a rate less than that exhibited in an untreated or healthy suitable control subject.
  • the increase or decrease may be 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 500%, 1000% or may be 1.1-fold, 1.2-fold, 1.3 fold, 1.4-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 10-fold, 20-fold, 50-fold, 100-fold, 500-fold, or 1000-fold.
  • An increase in clearance rate of a target includes, e.g., a slow down in the accumulation of a target, a reaching of a new equilibrium of generation and degradation, and a reversal of an accumulation, e.g., a decrease in the number or concentration of the target in circulation.
  • “Cleaving” as used herein is a process that disrupts a bonding interaction present in a target, such as a polypeptide or nucleic e.g., to produce two or more entities that after cleaving can be separated from one another.
  • the separation can involve, e.g., disrupt an ionic bond, a covalent bond, a polar covalent bond, a non-polar covalent bond, or a metallic bond.
  • cleaving applies to polypeptide targets, cleavage can involve breaking one or more peptide bonds.
  • cleaving applies to small molecule targets, cleavage can involve breaking one or more carbon or sulfide bonds.
  • cleavage can involve breaking one or more phosphodiester bonds.
  • cleaving applies to microbes such as bacteria, fungi, or viruses
  • cleavage can involve lysis of a membrane or capsid structure.
  • Cleaving can be carried out by an enzyme, e.g., a catalytically active receiver polypeptide.
  • Receivers can comprise, e.g., exonuclease, endonuclease, or protease activity.
  • the “circulatory concentration” is the concentration of a target, e.g., a cell, polypeptide (such as an antibody, pathogenic antigen, etc.), therapeutic agent, small molecule, metabolite or other entity, a receiver or a synthetic membrane-receiver complex in the space defined as the circulatory system.
  • the concentration may be defined as the number of free (unbound) entities in a given volume. In other embodiments, the concentration may be defined as the total number of entities in a given volume.
  • CDR complementarity determining region
  • a “complex” as used herein comprises an association of two or more entities.
  • a complex may comprise one or more polypeptides, nucleic acid, lipids, carbohydrates, inorganic compounds, organic compounds, and the like.
  • a complex can be functional (multiunit polypeptides) or non-functional (e.g., aggregates or precipitates) and may have beneficial or detrimental properties (e.g., immune complexes).
  • Complexes may be naturally occurring or may be man-made or synthetic. Synthetic complexes include higher order entities, e.g., subcellular structures and cells if they comprise a synthetic compound or molecule.
  • a synthetic membrane-receiver complex includes a cell comprising a receiver.
  • amino acid sequences one of skill will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a “conservatively modified variant” where the alteration results in the substitution of an amino acid with a chemically similar amino acid.
  • conservative amino acid substitution is illustrated by a substitution among amino acids within each of the following groups: (1) glycine, alanine, valine, leucine, and isoleucine, (2) phenylalanine, tyrosine, and tryptophan, (3) serine and threonine, (4) aspartate and glutamate, (5) glutamine and asparagine, and (6) lysine, arginine and histidine.
  • “Decrease,” in the context of a symptom of a treated disease, disorder or condition, refers to a reduction in measurable or conveyable parameters associated with the disease or condition that manifest as symptoms.
  • measurable parameters are a reduction in the subject's body temperature, a reduction in the concentration of targets in a sample taken from the subject, reduction in the intensity of inflammation or size of an inflamed area, reduction in the number of infiltrating cells, reduction in the number of episodes associated with the disease, disorder or condition, increase/decrease in organ size, weight gain/loss, etc.
  • conveyable parameters are, e.g., the subject's own assessment of well being and quality of life.
  • the decrease may be quantified as one, or a combination of, the following parameters: reduced inflammation, reduced flare-ups, reduced fatigue, reduced blood clotting, reduced swelling, increased energy, or increased hair growth, etc.
  • the parameters that may be quantified are those appropriate for assessing the specific disease, disorder or condition that is being treated. Delay, in the context of symptoms of a treated disease, disorder or condition, refers to the significant extension of a manageable health condition that would otherwise become exacerbated, using a treatment.
  • Degrading is defined as the process in which a target is either directly, or indirectly, reduced, inactivated, decomposed, deconstructed, lysed, dissolved, broken, lessened, impaired, weakened, deteriorated, diminished, or partitioned.
  • “Different polypeptide origin” refers to the organism or species from which a genetic sequence encoding the polypeptide, the polypeptide, or portion thereof, is sourced.
  • a fusion comprising polypeptides of different polypeptide origin may include a receiver polypeptide that is encoded by the genetic sequence for human adenosine deaminase and the genetic sequence for phenylalanine hydroxylase from chromobacterium violaceum.
  • a “domain” is a part of a polypeptide, such as a receiver polypeptide that is generally having a 3-dimensional structure and may exhibit a distinct activity, function, such as, e.g., a catalytic, an enzymatic, a structural role, or a binding function.
  • Duration refers to the period of time that a portion of the synthetic membrane-receiver polypeptide complex exists in a specific tissue or an organism as a whole. This applies to 0.1% 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 100% of the initial dose or concentration of the synthetic membrane-receiver polypeptide complex.
  • the synthetic membrane-receiver complex is formulated for long-term duration. In some embodiments, the synthetic membrane-receiver complex is formulated for short-term duration.
  • an “enriched population of cells” it is meant a population of cells that is substantially comprised of a particular cell of interest.
  • 50% or more of the cells in the population are the cells of interest, e.g., 50%, 60%, 70%, usually 80%, 85%, 90%, more usually 92%, 95%, 96%, 97%, 98%, or 99%, sometimes as much as 100% of the cells in the population.
  • the separation of cells of interest from a complex mixture or heterogeneous culture of cells may be performed by any convenient means known in the art, for example, by affinity separation techniques such as magnetic separation using magnetic beads coated with an affinity reagent, affinity chromatography, or “panning” with an affinity reagent attached to a solid matrix, e.g., plate, or other convenient technique.
  • affinity separation techniques such as magnetic separation using magnetic beads coated with an affinity reagent, affinity chromatography, or “panning” with an affinity reagent attached to a solid matrix, e.g., plate, or other convenient technique.
  • Other techniques providing accurate separation include fluorescence activated cell sorters, which can have varying degrees of sophistication, such as multiple color channels, low angle and obtuse light scattering detecting channels, impedance channels, etc.
  • the cells may be selected against dead cells by employing dyes associated with dead cells. Any technique may be employed which is not unduly detrimental to the viability of the desired cells.
  • Enucleation is the rendering of a cell to a non-replicative state, either through inactivation or removal of the nucleus.
  • An “epitope” includes any segment on an antigen to which an antibody or other ligand or binding molecule binds.
  • An epitope may consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics.
  • receivers comprise specific epitopes.
  • targets comprise specific epitopes.
  • Erythroid cells include nucleated red blood cells, red blood cell precursors, and enucleated red blood cells and those listed in Table 2.
  • the erythroid cells are a cord blood stem cell, a CD34+ cell, a hematopoietic stem cell (HSC), a spleen colony forming (CFU-S) cell, a common myeloid progenitor (CMP) cell, a blastocyte colony-forming cell, a burst forming unit-erythroid (BFU-E), a megakaryocyte-erythroid progenitor (MEP) cell, an erythroid colony-forming unit (CFU-E), a reticulocyte, an erythrocyte, an induced pluripotent stem cell (iPSC), a mesenchymal stem cell (MSC), a polychromatic normoblast, an orthochromatic normoblast, or a combination thereof.
  • HSC hematopoietic stem cell
  • CFU-S
  • the erythroid cells are immortal or immortalized cells.
  • immortalized erythroblast cells can be generated by retroviral transduction of CD34+ hematopoietic progenitor cells to express Oct4, Sox2, Klf4, cMyc, and suppress TP53 (e.g., as described in Huang et al., Mol Ther 2013, epub ahead of print September 3).
  • the cells may be intended for autologous use or provide a source for allogeneic transfusion.
  • Erythroid cells can be contacted with a receiver to generate a synthetic membrane-receiver complex. Erythroid cells comprising a receiver are one example of a synthetic membrane-receiver complex.
  • erythroid cells a recultured.
  • erythroid progenitor cells are contacted with a receiver to generate a synthetic membrane-receiver complex.
  • excipient refers to an inert substance added to a pharmaceutical composition to further facilitate administration of a compound.
  • excipients include, but are not limited to, calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, anti-coagulants, and polyethylene glycols.
  • the receiver including a receiver polypeptide is “exogenous” or “heterologous”, thus it may either not naturally exist, such as a fusion or chimera comprising domains of different polypeptide or species origin, it may not naturally occur in a naturally occurring cell, such as an unmodified erythrocyte or platelet, it may not function in the same way as a naturally occurring polypeptide would, or it may not naturally occur in the quantity that the receiver polypeptide occurs, e.g., in embodiments in which the synthetic membrane-receiver polypeptide complex is a cell-derived polypeptide receiver that is overexpressed as compared to the expression of a naturally occurring polypeptide in an unmodified cell.
  • the polypeptide receiver is expressed from an exogenous nucleic acid.
  • the receiver is isolated from a source and loaded into or conjugated to a synthetic membrane-receiver complex.
  • exogenous when used in the context of nucleic acid includes a transgene and recombinant nucleic acids.
  • expression refers to the process to produce a polypeptide, such as a receiver polypeptide including transcription and translation.
  • Expression may be, e.g., increased by a number of approaches, including: increasing the number of genes encoding the polypeptide, increasing the transcription of the gene (such as by placing the gene under the control of a constitutive promoter), increasing the translation of the gene, knocking out of a competitive gene, or a combination of these and/or other approaches.
  • a synthetic membrane-receiver complex that is “formulated for long-term duration” is, in some embodiments, one that is part of a population of synthetic membrane-receiver complexes wherein a substantial fraction of the population resides in the circulatory system for more than 10 days, e.g., 15, 21, 25, 35, 45, 50, 60, 90, 100, 110, or 120 days.
  • the population may have an increased half-life, e.g., 1.5 ⁇ , 2 ⁇ , 5 ⁇ , 10 ⁇ , 20 ⁇ , 50 ⁇ , 100 ⁇ more time in circulation, when formulated for long-term duration compared to the duration exhibited by a population of unformulated complexes.
  • an entity such as a receiver may have an increased half-life, e.g., 1.5 ⁇ , 2 ⁇ , 5 ⁇ , 10 ⁇ , 20 ⁇ , 50 ⁇ , 100 ⁇ more time in circulation, when formulated for long-term duration compared to the duration that entity would exhibit in an unmodified state.
  • a synthetic membrane-receiver complex that is “formulated for short-term duration” is, in some embodiments, one that is part of a population of synthetic membrane-receiver complexes wherein a substantial fraction of the population resides in the circulatory system for less than 10 days, e.g., 9, 8, 7, 6, 5, 4, 3, 2 days, 1 day, 12 hours, or 6 hours.
  • the population may have a decreased half-life, e.g., 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% less time in circulation, when formulated for short-term duration compared to the duration exhibited by a population of unformulated complexes.
  • an entity such as a receiver may have a reduced half-life, e.g., 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% less time in circulation, when formulated for short-term duration compared to the duration that entity would exhibit in an unmodified state.
  • a reduced half-life e.g., 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99% less time in circulation, when formulated for short-term duration compared to the duration that entity would exhibit in an unmodified state.
  • Formated for residency in the circulatory system describes one or more modifications to an entity, such as a synthetic membrane-receiver complex formulated for administration to the circulatory system of a subject that substantially decrease recognition, modification, degradation, and/or destruction of the entity by components of the circulatory system (e.g., circulating immune cells, antibodies, enzymatic activities) thereby increasing the half-life of the entity when compared to an unmodified entity.
  • an entity such as a synthetic membrane-receiver complex formulated for administration to the circulatory system of a subject that substantially decrease recognition, modification, degradation, and/or destruction of the entity by components of the circulatory system (e.g., circulating immune cells, antibodies, enzymatic activities) thereby increasing the half-life of the entity when compared to an unmodified entity.
  • a “functional” receiver or synthetic membrane-receiver complex refers to a synthetic membrane-receiver complex or a receiver that exhibits a desired or specified activity or characteristic, including enzymatic, catalytic or metabolic activity, structural integrity, immunogenic complementarity, target binding, and correct localization or is capable of promoting a desired or specified effect or phenotype.
  • Fusion or chimera is defined as a polypeptide sequence, or corresponding encoding nucleotide sequence, that is derived from the combination of two or more sequences that are not found together in nature. This may be a combination of separate sequences derived from separate genes within the same genome, or from heterologous genes derived from distinctly different species' genomes.
  • Genetic material refers to nucleic acid molecules having nucleotide sequences of adenosine, thymine, uracil, cytosine, and guanine capable of encoding a gene.
  • heavy chain used herein is understood to include a full-length heavy chain including a variable region (VH) having amino acid sequences that determine specificity for antigens and a constant region having three constant domains (CHL CH2, and CH3), and fragments thereof.
  • light chain used herein is understood to include a full-length light chain including a variable region (VL) having amino acid sequences that determine specificity for antigens and a constant region (CL), and fragments thereof.
  • homolog indicates polypeptides, including receiver polypeptide that have the same or conserved residues at a corresponding position in their primary, secondary or tertiary structure. Functional homologs include receivers and other polypeptides that exhibit similar function and/or specificity (e.g., for a particular target).
  • a naturally occurring intact antibody, or immunoglobulin includes four polypeptides: two full-length light chains and two full-length heavy chains, in which each light chain is linked to a heavy chain by disulfide bonds.
  • Each heavy chain has a constant region and a variable region.
  • each light chain has a constant region and a variable region.
  • the light chain constant region can be either kappa ( ⁇ ) or lambda ( ⁇ ) type.
  • the variable regions differ in sequence among antibodies and are used in the binding and specificity of a given antibody to its particular antigen.
  • the term “increase,” “enhance,” “stimulate,” and/or “induce” generally refers to the act of improving or increasing, either directly or indirectly, a concentration, level, function, activity, or behavior relative to the natural, expected, or average, or relative to a control condition.
  • the term “inhibit,” “suppress,” “decrease,” “interfere,” and/or “reduce” generally refers to the act of reducing, either directly or indirectly, a concentration, level, function, activity, or behavior relative to the natural, expected, or average, or relative to a control condition.
  • a “library” as used herein includes a collection of nucleic acid molecules (e.g., DNA, RNA) having diverse nucleic acid sequences, a genetically diverse collection of clones, a collection of diverse polypeptides, a diverse collection of cells, etc.
  • nucleic acid molecules e.g., DNA, RNA
  • a mammalian subject includes all mammals, including without limitation, humans, domestic animals (e.g., dogs, cats and the like), farm animals (e.g., cows, sheep, pigs, horses and the like) and laboratory animals (e.g., monkey, rats, mice, rabbits, guinea pigs and the like).
  • the terms “individual,” “subject,” “host,” and “patient,” are used interchangeably herein and refer to any mammalian subject for whom diagnosis, treatment, or therapy is desired, particularly humans. The methods described herein are applicable to both human therapy and veterinary applications.
  • the subject is a mammal, and in other embodiments the subject is a human.
  • Medical device refers to any device, apparatus or machine used to deliver a dose of a synthetic membrane-receiver complex and/or a therapeutic agent. This includes containers, bottles, vials, syringes, bags, cartridges, cassettes, magazines, cylinders, or canisters.
  • Medical kit refers to a packaged unit that includes a medical device, applicator, appropriate dosage of synthetic membrane-receiver complex optionally including a therapeutic agent, and relevant labeling and instructions.
  • the term “modulate,” “modulating”, “modify,” and/or “modulator” generally refers to the ability to alter, by increase or decrease, e.g., directly or indirectly promoting/stimulating/upregulating or interfering with/inhibiting/downregulating a specific concentration, level, expression, function or behavior, such as, e.g., to act as an antagonist or agonist.
  • a modulator may increase and/or decrease a certain concentration, level, activity or function relative to a control, or relative to the average level of activity that would generally be expected or relative to a control level of activity.
  • Membrane as used herein is a boundary layer that separates an interior space from an exterior space comprising one or more biological compounds, typically lipids, and optionally polypeptides.
  • Membranes can be lipid bilayers.
  • membranes comprise one or more of phosphatidylcholine, sphingomyelin, lysophosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, or phosphatidic acid.
  • membranes comprise one or more polypeptides such as ankyrin and coenzyme Q10. Included in the definition of membrane are cell membranes comprising, e.g., a phospholipid bilayer and cell membrane associated polypeptides.
  • the synthetic membrane-receiver complex comprises a membrane as defined herein.
  • nucleic acid molecule refers to a single or double-stranded polymer of deoxyribonucleotide or ribonucleotide bases. It includes chromosomal DNA and self-replicating plasmids, vectors, mRNA, tRNA, siRNA, etc. which may be recombinant and from which exogenous polypeptides may be expressed when the nucleic acid is introduced into a cell.
  • Orthologs are defined as genes in different species that evolved from a common ancestral gene by speciation.
  • pharmaceutically-acceptable refers to compositions, carriers, diluents and reagents capable of administration to or upon a subject without the production of undesirable physiological effects to a degree that would prohibit administration of the composition.
  • pharmaceutically-acceptable excipient includes an excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic, and desirable, and includes excipients that are acceptable for veterinary use as well as for human pharmaceutical use. Such excipients can be solid, liquid, semisolid, or, in the case of an aerosol composition, gaseous.
  • the term “pharmaceutically acceptable carrier” includes any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, emulsions such as an oil/water or water/oil, and various types of wetting agents.
  • the term also encompasses any of the agents approved by a regulatory agency of the US Federal government or listed in the US Pharmacopeia for use in animals, including humans, as well as any carrier or diluent that does not cause significant irritation to a subject and does not abrogate the biological activity and properties of the administered compound.
  • Some agents may be administered as “pharmaceutically acceptable salt”, e.g., prepared from inorganic and organic acids.
  • Salts derived from inorganic acids include hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Salts derived from organic acids include acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluene-sulfonic acid, salicylic acid, and the like.
  • Salts can also be prepared from inorganic and organic bases. Salts derived from inorganic bases, include by way of example only, sodium, potassium, lithium, ammonium, calcium and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary and tertiary amines Any ordinary skilled person in the art will know how to select a proper pharmaceutically acceptable carrier, a pharmaceutically acceptable salt thereof for implementing this invention without undue experimentation.
  • the term “pharmaceutical composition” refers to one or more of the compounds described herein, such as, e.g., a synthetic membrane-receiver polypeptide complex mixed or intermingled with, or suspended in one or more other chemical components, such as physiologically acceptable carriers and excipients.
  • a pharmaceutical composition is to facilitate administration of a compound to a subject.
  • polypeptide molecules having sequences associated with a desired function or activity, such as receiver polypeptides.
  • a polypeptide is a term that refers to a chain of amino acid residues, regardless of post-translational modification (e.g., phosphorylation or glycosylation) and/or complexation with additional polypeptides, synthesis into multisubunit complexes, with nucleic acids and/or carbohydrates, or other molecules.
  • Proteoglycans therefore also are referred to herein as polypeptides.
  • the synthetic membrane-receiver complex comprises a polypeptide receiver and is referred to a “synthetic membrane-receiver polypeptide complex.”
  • the synthetic membrane-receiver complex comprises one or more non-receiver polypeptides that are optionally membrane-associated and that exhibit catalytic and/or metabolic activity independent of the receiver.
  • the non-receiver polypeptides may have catalytic activity for an organic compound including a metabolite.
  • the synthetic membrane-receiver complex comprises a sufficient number of non-receiver polypeptides (and optionally non-protein co-factors) to support a metabolic pathway.
  • pharmaceutically active agent or “pharmaceutical agent” is defined as any compound, e.g., a small molecule drug, or a biologic (e.g., a polypeptide drug or a nucleic acid drug) that when administered to a subject has a measurable or conveyable effect on the subject, e.g., it alleviates or decreases a symptom of a disease, disorder or condition.
  • the pharmaceutical agent may be administered prior to, in combination with, or following the delivery of a synthetic membrane-receiver polypeptide complex.
  • the pharmaceutically active agent exerts a synergistic treatment effect with the synthetic membrane-receiver polypeptide complex.
  • the pharmaceutically active agents exerts an additive treatment effect with the synthetic membrane-receiver polypeptide complex.
  • a “promoter” is defined as an array of nucleic acid control sequences that direct transcription of an operably linked nucleic acid. Promoters include necessary nucleic acid sequences near the start site of transcription. A promoter also optionally includes distal enhancer or repressor elements. A “constitutive” promoter is a promoter that is active under most environmental and developmental conditions. An “inducible” promoter is a promoter that is active under environmental or developmental regulation.
  • the term “operably linked” refers to a functional linkage between a nucleic acid expression control sequence (such as a promoter, or array of transcription factor binding sites) and a second nucleic acid sequence, wherein the expression control sequence directs transcription of the nucleic acid corresponding to the second sequence.
  • a “receiver,” as used herein, is an entity capable of interacting with a target, e.g., to associate with or bind to a target.
  • a receiver can comprise or can consist essentially of a polypeptide.
  • the receiver comprises a polypeptide, a carbohydrate, a nucleic acid, a lipid, a small molecule, or a combination thereof.
  • the receiver is “synthetic” in the sense that it is an exogenous or heterologous compound or molecule with regard to its presence in the synthetic membrane-receiver complex.
  • the receiver is “synthetic” in the sense that it is a man-made compound or molecule, such as a fusion or chimera, a non-naturally occurring polypeptide, carbohydrate, nucleic acid, lipid, or combination thereof, or a man-made small molecule or other therapeutic agent.
  • the receiver may comprise a fusion or chimera comprising one or more of an S domain, an A domain and a U domain.
  • the S domain is a surface domain exposed to the environment around the synthetic membrane-receiver complex, such as the circulatory system of a subject.
  • the A domain is an anchor domain that attaches the S domain to the synthetic membrane of the synthetic membrane-receiver polypeptide complex.
  • the U domain faces the unexposed side of or is located within the synthetic membrane-receiver complex, i.e. the side that is not exposed to the external environment of the circulatory system of a subject.
  • a receiver may be located on the surface of the synthetic membrane-receiver polypeptide complex or may be located within the complex.
  • the receiver may be associated with the membrane of the synthetic membrane-receiver complex, e.g., the receiver is anchored in, conjugated to or otherwise bound to the membrane.
  • the receiver may be conjugated to the membrane of the synthetic membrane-receiver complex by chemical or enzymatic conjugation. In other embodiments, the receiver is not conjugated to the membrane.
  • the receiver is not associated with the membrane of the synthetic membrane-receiver complex and is located within the membrane-encapsulated volume of the complex. In some embodiments, a receiver located within the synthetic membrane-receiver complex does not substantially diffuse out of the complex and/or may not permeate the membrane. In other embodiments, the receiver may substantially diffuse out of the complex and/or may permeate the membrane. In some embodiments, the receiver is loaded, e.g., introduced into or put onto the synthetic membrane-receiver complex. A receiver that is loaded is not biologically synthesized by the synthetic membrane-receiver complex.
  • a receiver suitable for loading may be e.g., produced in a cell-based expression system, isolated from a biological sample, or chemically or enzymatically synthesized, and then loaded into or onto the synthetic membrane-receiver complex.
  • the receiver may be further modified by the synthetic membrane-receiver complex after loading.
  • the receiver is not modified after loading.
  • the receiver polypeptide is not loaded onto or into the complex.
  • the receiver is made, e.g., biologically synthesized by the synthetic membrane-receiver complex.
  • a receiver polypeptide is expressed by the synthetic membrane-receiver complex from an exogenous nucleic acid molecule (e.g., a DNA or mRNA) that was introduced into the complex.
  • the receiver may bind to and/or sequester a target.
  • the receiver may exhibit a catalytic activity toward the target, e.g., the receiver may convert or modify the target, or may degrade the target.
  • a product may then optionally be released from the receiver.
  • “Residency” of a synthetic membrane-receiver complex refers to the period of time it spends in a physiological location.
  • the specific location of the synthetic membrane-receiver complex may change during its lifetime and “residency” applies to the period of time spent in various environments, including vascular circulation, peripheral tissues, capillaries, digestive system, pulmonary system, nasal tissues, epidermal surface, and interstitial tissue.
  • the synthetic membrane-receiver complex resides in the circulatory system of a subject.
  • Replicating nucleic acid refers to deoxyribonucleic acid (DNA) that is capable of being copied by enzymes dedicated to the increasing the number of copies of the DNA.
  • DNA replication leads to the production of two identical replicas from one original DNA molecule.
  • DNA replication comprises the incorporation of nucleotides into a growing DNA strand by DNA polymerase matched to the template strand one at a time via the creation of phosphodiester bonds.
  • “Sequestering” is defined as cloistering, occluding, separating, segregating, hiding, insulating, or isolating of a target and preventing it from freely interacting with its environment.
  • Specifically binding or “specifically interacting”, as used herein, describes any interaction between two entities (e.g., a target with a receiver, such as an antibody with an antigen, a receptor with a ligand, an enzyme with a substrate, biotin with avidin, etc.) that is saturable, often reversible and so competitive, as these terms are understood by those of ordinary skill in the chemical and biochemical arts.
  • a target with a receiver such as an antibody with an antigen, a receptor with a ligand, an enzyme with a substrate, biotin with avidin, etc.
  • Kd binding constant
  • the Kd may range from a mM range to a pM range, including ⁇ M ranges and nM ranges. Typical Kd values are below about 10 ⁇ 6 M, below about 10 ⁇ 7 M, below about 10 ⁇ 8 M, and in some embodiments below about 10 ⁇ 9 M.
  • the term “substantially” or “substantial” refers, e.g., to the presence, level, or concentration of an entity in a particular space, the effect of one entity on another entity, or the effect of a treatment.
  • an activity, level or concentration of an entity is substantially increased if the increase is 2-fold, 3-fold, 4-fold, 5-fold, 10-fold, 50-fold, 100-fold, or 1000-fold relative to a baseline.
  • An activity, level or concentration of an entity is also substantially increased if the increase is 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, or 500% relative to a baseline.
  • An entity may be substantially present in a particular space if it can be detected by methods known in the art.
  • An entity may not be substantially present in a particular space if it is present at levels below the limit of detection for assays and methods known in the art. In some embodiments, an entity may not be substantially present in a particular space if it is barely detectable but only in non-functional quantities or minute quantities that do not cause or change a phenotype. In other embodiments, an entity may not be substantially present in a particular population if it is present and can be detected only in a small number of constituents making up the population, e.g., less than 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3% 2% or less than 1%, 0.5%, 0.1% of constituents of the population.
  • an exogenous nucleic acid may not be retained upon enucleation, the cell is rendered non-replicative, and the enucleated cell is incapable of continued expression of the receiver polypeptide encoded by the exogenous nucleic acid.
  • the loss of the ability of the cell to continue to significantly translate the exogenous polypeptide “effectively terminates” protein expression.
  • the synthetic membrane-receiver complex is substantially incapable of self-replication, e.g., the replication of nucleic acids.
  • the synthetic membrane-receiver polypeptide complex does not substantially incorporate a nucleoside if contacted with labeled nucleoside, such as thymidine, in an incorporation assay.
  • the synthetic membrane-receiver polypeptide complex does not contain a substantial amount of self-replicating nucleic acids.
  • substantially identity of polynucleotide or nucleic acid sequences means that a polynucleotide comprises a sequence that has at least 25% sequence identity. Alternatively, percent identity can be any integer from 25% to 100%. More preferred embodiments include at least: 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% compared to a reference sequence using the programs described herein; preferably BLAST using standard parameters.
  • Synthetic refers to a compound or molecule that is either man-made and non-naturally occurring, or if it is naturally occurring is placed in a context or location that it would not naturally exist, or if it naturally exists in the context or location is in a state of purity, or is present in an amount, concentration or number that it would not naturally be present in the context or location.
  • Synthetic entities can be isolated or purified compounds that are optionally chemically or enzymatically modified from their natural state, exogenous nucleic acids, exogenous (heterologous) receivers, and the like.
  • the presence of a synthetic compound or molecule, as defined herein, in any entity renders the entire entity “synthetic”.
  • a cell comprising a receiver is a synthetic cell.
  • a “target,” as used herein, is an entity capable of interacting with a receiver, e.g., to associate with or bind to a receiver.
  • a “target” includes, but is not limited to a polypeptide (e.g., an antibody or antibody-related polypeptide, a complement constituent, an amyloid protein, a pathogen, a toxin, a prion), a molecule (e.g., a metabolite, a steroid, a hormone, a carbohydrate; an oligosaccharide; a chemical; a polysaccharide, a DNA; an RNA; a lipid, an amino acid, an element, a toxin or pathogen), a complex (e.g., an immune complex), or a cell (e.g., a cancer cell, a macrophage, a bacterium, a fungus, a virus, or a parasite).
  • a target is intended to be detected, diagnosed, impaired, destroyed
  • a “target self-antibody,” as used herein, is a self-antibody associated with an autoimmune disease. Such self-antibodies may be detected and analyzed using antibody binding tests involving contacting the subject's antibodies to samples of the subject's own tissue, usually thyroid, stomach, liver, and kidney tissue. Antibodies binding to the “self” tissue (comprising self-antigens) indicate an autoimmune disorder.
  • Transgene or “exogenous nucleic acid” refers to a foreign or native nucleotide sequence that is introduced into a synthetic membrane-receiver complex. Transgene and exogenous nucleic acid are used interchangeably herein and encompass recombinant nucleic acids.
  • treat are an approach for obtaining beneficial or desired clinical results, pharmacologic and/or physiologic effect, e.g., alleviation of the symptoms, preventing or eliminating said symptoms, and refer to both therapeutic treatment and prophylactic or preventative treatment of the specific disease, disorder or condition.
  • pharmacologic and/or physiologic effect include, but are not limited to, preventing the disease, disorder or condition from occurring in a subject that may be predisposed to the disease, disorder or condition but does not yet experience or exhibit symptoms of the disease (prophylactic treatment), alleviation of symptoms of the disease, disorder or condition, diminishment of extent of the disease, disorder or condition, stabilization (i.e., not worsening) of the disease, disorder or condition, preventing spread of the disease, disorder or condition, delaying or slowing of the disease, disorder or condition progression, amelioration or palliation of the disease, disorder or condition, and combinations thereof, as well as prolonging survival as compared to expected survival if not receiving treatment.
  • a “therapeutic agent” or “therapeutic molecule” includes a compound or molecule that, when present in an effective amount, produces a desired therapeutic effect, pharmacologic and/or physiologic effect on a subject in need thereof.
  • terapéuticaally effective amount is an amount of an agent being administered to a subject sufficient to effect beneficial or desired clinical results, pharmacologic and/or physiologic effects.
  • An effective amount can be administered in one or more administrations.
  • An effective amount is typically sufficient to palliate, ameliorate, stabilize, reverse, slow or delay the progression of the disease state.
  • the effective amount thus refers to a quantity of an agent or frequency of administration of a specific quantity of an agent sufficient to reasonably achieve a desired therapeutic and/or prophylactic effect.
  • it may include an amount that results in the prevention of, treatment of, or a decrease in, the symptoms associated with a disease or condition that is being treated, e.g., the diseases or medical conditions associated with a target polypeptide.
  • the amount of a therapeutic composition administered to the subject will depend on the type and severity of the disease and on the characteristics of the individual, such as general health, pathologic conditions, diets, age, sex, body weight and tolerance to drugs. It will also depend on the degree, severity and type of disease. Further, the effective amount will depend on the methods of formulation and administration used, e.g., administration time, administration route, excretion speed, and reaction sensitivity. The skilled artisan will be able to determine appropriate dosages depending on these and other factors.
  • the compositions can also be administered in combination with one or more additional therapeutic compounds.
  • a desirable dosage of the pharmaceutical composition may be in the range of about 0.001 to 100 mg/kg for an adult.
  • an intravenous administration is initiated at a dose which is minimally effective, and the dose is increased over a pre-selected time course until a positive effect is observed. Subsequently, incremental increases in dosage are made limiting to levels that produce a corresponding increase in effect while taking into account any adverse affects that may appear.
  • suitable dosages can range, for example, from 1 ⁇ 10 10 to 1 ⁇ 10 14 , from 1 ⁇ 10 11 to 1 ⁇ 10 13 , or from 5 ⁇ 10 11 to 5 ⁇ 10 12 synthetic membrane-receiver polypeptide complexes of the present invention.
  • each dose of synthetic membrane-receiver polypeptide complexes can be administered at intervals such as once daily, once weekly, twice weekly, once monthly, or twice monthly.
  • Unbound refers to the state of a target with which the receiver is capable of interacting. An unbound target is not associated with another entity or a receiver. An unbound receiver is not associated with another entity or a target. A target is considered “bound” once it is associated with the receiver or another entity. Unbound targets include soluble forms of the target in circulation. Bound targets include targets that are embedded, associated with, linked to, or otherwise interacting with entities in circulation or peripheral tissue. Entities with which a target may interact include circulating cells, peripheral endothelial tissue, immune complexes, glycolipids, microbes, immunoglobulins, serum albumin, clotting factors, lipoproteins, and electrolytes.
  • a “variant” is a polypeptide which differs from the original protein by one or more amino acid substitutions, deletions, insertions, or other modifications. These modifications do not significantly change the biological activity of the original protein. In many cases, a variant retains at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100% of the biological activity of original protein. The biological activity of a variant can also be higher than that of the original protein.
  • a variant can be naturally-occurring, such as by allelic variation or polymorphism, or be deliberately engineered.
  • the amino acid sequence of a variant is substantially identical to that of the original protein.
  • a variant shares at least 50%, 60%, 70%, 80%, 85%, 90%, 95%, 99%, or more global sequence identity or similarity with the original protein.
  • Sequence identity or similarity can be determined using various methods known in the art, such as Basic Local Alignment Tool (BLAST), dot matrix analysis, or the dynamic programming method.
  • BLAST Basic Local Alignment Tool
  • GCG Genetics Computer Group
  • GAP Needleman-Wunsch algorithm
  • the term “vector” is a nucleic acid molecule, preferably self-replicating, which transfers and/or replicates an inserted nucleic acid molecule, such as a transgene or exogenous nucleic acid into and/or between host cells. It includes a plasmid or viral chromosome into whose genome a fragment of recombinant DNA is inserted and used to introduce recombinant DNA, or a transgene, into a synthetic membrane-receiver polypeptide complex.
  • the “volume of distribution” is a pharmacological, theoretical volume that the total amount of administered drug would have to occupy (if it were uniformly distributed), to provide the same concentration as it is in blood plasma.
  • a VD greater than the blood plasma indicates that an agent is distributed in tissue in the rest of the body.
  • the VD is influenced by solubility, charge, size, etc. Generally, non-polar agents with high lipid solubility, agents with low rates of ionization or low plasma binding capabilities have higher volumes of distribution than agents that are more polar, more highly ionized or exhibit high plasma binding.
  • the units for Volume of Distribution are typically reported in (ml or liter)/kg body weight.
  • a volume of distribution “equal to plasma volume” is relative to the volume of the circulatory system exclusive of circulating cells.
  • synthetic membrane-receiver complexes populations, pharmaceutical compositions, and dosage forms thereof, as well as medical devices and kits comprising a formulation of the synthetic membrane-receiver complexes.
  • the synthetic membrane-receiver complexes described herein comprise a receiver (e.g., a polypeptide) that is capable of interacting with a target and further comprise a membrane comprising a polypeptide that is not the receiver.
  • the synthetic membrane-receiver complex has catalytic activity independent of the receiver.
  • the synthetic membrane-receiver complexes comprise a payload, for example a therapeutic agent.
  • synthetic membrane-receiver complex are generated using cells as a source material.
  • generating a synthetic membrane-receiver complex comprises the step of contacting an erythroid cell and platelets with a receiver.
  • generating a synthetic membrane-receiver complex comprises the step of contacting a cell derived from a hematopoietic stell cell with a receiver.
  • synthetic membrane-receiver complexes are administered, e.g., intravenously to the circulatory system of a mammalian subject, such as a human.
  • the membrane-receiver complexes provide a natural barrier between a receiver and optionally a payload (e.g., therapeutic agent) and the immune system.
  • the synthetic membrane-receiver complexes are capable of residing in the circulatory system of a subject for an extended period of time allowing delivery of a therapeutic effect for a longer period of time than what can be achieved by delivery through other methods currently used.
  • Synthetic membrane-receiver complexes may interact with a target in the circulatory system of the subject.
  • concentration of an unbound target or total target in the circulatory system of the subject is reduced subsequent to its interaction with the receiver exhibited in or on the synthetic membrane-receiver complex.
  • the presence or elevated concentration of a target in circulation is associated with a disease, disorder or condition and reducing the concentration of the target leads to a reduction in disease burden, may alleviate a symptom of the disease or has some other treatment effect.
  • a reduction in the concentration of the target prevents the onset of a disease, disorder or condition.
  • Biodistribution is a substantial hurdle in drug delivery and efficacy. After a drug enters the systemic circulation, it is distributed to the body's tissues. Distribution is generally uneven because of differences in blood perfusion, tissue binding (e.g., because of lipid content), regional pH, and permeability of cell membranes.
  • the entry rate of a drug into a tissue depends on the rate of blood flow to the tissue, tissue mass, and partition characteristics between blood and tissue. Distribution equilibrium (when entry and exit rates are the same) between blood and tissue is reached more rapidly in richly vascularized areas, unless diffusion across cell membranes is the rate-limiting step. After equilibrium, drug concentrations in tissues and in extracellular fluids are reflected by the plasma concentration. Metabolism and excretion occur simultaneously with distribution, making the process dynamic and complex.
  • the synthetic membrane-receiver complexes when formulated in a pharmaceutical compositions suitable for administration into the circulatory system of a subject can have a volume of distribution equal to the plasma volume of the subject.
  • Advantages of the volume of distribution characteristic of the synthetic membrane-receiver complexes include that the biodistribution of the receiver when administered as a synthetic membrane-receiver complex into the circulatory system of a subject may be accurately predicted and/or that potential adverse extravascular effects of the receiver (e.g., an inflammatory response, an immune response, toxicity, etc.) are substantially reduced.
  • Distribution of a therapeutic composition out of the bloodstream and into surrounding tissue increases the apparent volume of distribution to be greater than the plasma volume of the subject.
  • Therapeutic compositions that exit the bloodstream and interact with surrounding tissue e.g., adipose tissue or muscle, may interact with those tissues in unpredictable ways and trigger adverse events.
  • a therapeutic composition such as a composition comprising a synthetic membrane-receiver complex described herein, whose volume of distribution does not substantially exceed the plasma volume of the subject typically has a safety profile that is superior to a therapeutic composition with a large volume of distribution.
  • the amount of a therapeutic composition that must be loaded to be effective is in part dependent on the bioavailability of the therapeutic composition.
  • Bioavailability is related to the composition's profile and rate of distribution into extra-vascular tissues, and thus its volume of distribution.
  • a therapeutic composition such as a composition comprising a synthetic membrane-receiver complex described herein, will have a more precise and predictable dose-effect relationship than a therapeutic composition with a less precise and predictable volume of distribution.
  • the drug distribution rate for interstitial fluids of most tissues is determined primarily by perfusion.
  • perfusion For poorly perfused tissues (e.g., muscle, fat), distribution is very slow, especially if the tissue has a high affinity for the drug.
  • Endothelial cells lining the vessel wall are connected by adherens, tight and gap junctions. These junctional complexes are related to those found at epithelial junctions but with notable changes in terms of specific molecules and organization. Endothelial junctional proteins play important roles in tissue integrity but also in vascular permeability, leukocyte extravasation and angiogenesis.
  • Small molecules, protein therapeutics, and viruses measure 1-30 nm and are capable of diffusing far beyond the vasculature based on lipophilicity, ability to bind plasma proteins, and charge.
  • a drug that is confined to the vasculature has a lesser volume of tissue to occupy and thus may remain at an effective, therapeutic concentration.
  • the drug is unable to interact with peripheral tissues and potential off-target toxicity effects are limited.
  • Larger circulatory agents e.g., between 1 micron and 20 microns
  • do not pass through endothelial tight junctions which are less than 100 nm in width and endothelial cells are incapable of facilitating the transcytosis of agents of that size.
  • the synthetic membrane-receiver complexes described herein measure between 1 micron and 20 microns. The vascular properties of these agents limit their diffusive capabilities to the bloodstream and concentrate the therapeutic effect of any receiver or payload.
  • synthetic membrane-receiver complexes described herein exhibit advantageous clearance properties.
  • synthetic membrane-receiver complexes may be degraded using a natural degradation process, through the reticulo-endothelial system. Such degradation typically does not cause any or little side effects.
  • receivers displayed on the synthetic membrane-receiver complexes can be selectively trapped by organs of the reticulo-endothelial system.
  • the synthetic membrane-receiver complexes described herein are, in some embodiments, incapable of self-replication. In some embodiments, the synthetic membrane-receiver complexes do not contain self-replicating nucleic acids. Thus, such complexes do not carry a risk of uncontrolled cellular division, undesired protein expression and/or the potential of triggering cytokine release syndrome.
  • the synthetic membrane-receiver polypeptide complex comprises a membrane that has a mass of approximately 1 ⁇ 10 ⁇ -12 g and a density of approximately 1.15 g/cm ⁇ 3.
  • the mass of the membrane component can be assessed by separating it from the remainder of the complex using hypotonic solutions of mildly alkaline buffer, see e.g., protocols in Dodge et al 1963, Arch Biochem Biophys 100:119.
  • the synthetic membrane-receiver complex comprises a membrane.
  • the membrane comprises phosphatidylcholine, sphingomyelin, lysophosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, or phosphatidic acid.
  • the membrane is a cell membrane.
  • the synthetic membrane-receiver polypeptide complex comprises lipid molecules of the class of choline phospholipids, acidic phospholipids, and phosphatidylethanolamine.
  • the synthetic membrane-receiver polypeptide complex comprises phosphatidylcholine, sphingomyelin, lysophosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, or phosphatidic acid.
  • the synthetic membrane-receiver polypeptide complex comprises choline phospholipids in an approximate amount of 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, or 65% relative to the total lipid content of the complex.
  • the synthetic membrane-receiver polypeptide complex comprises acidic phospholipids in an approximate amount of 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, or 20% relative to the total lipid content of the complex.
  • the synthetic membrane-receiver polypeptide complex comprises phosphatidylcholine in an amount greater than 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%. 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, or greater than 50% relative to the total lipid content of the complex.
  • the synthetic membrane-receiver polypeptide complex comprises sphingomyelin in an amount greater than 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%. 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, or greater than 50% relative to the total lipid content of the complex.
  • the synthetic membrane-receiver polypeptide complex comprises lysophosphatidylcholine in an amount greater than 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.5%, 2%, 2.5%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, or greater than 10% relative to the total lipid content of the complex.
  • the synthetic membrane-receiver polypeptide complex comprises phosphatidylethanolamine in an amount greater than 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%. 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, or greater than 50% relative to the total lipid content of the complex.
  • the synthetic membrane-receiver polypeptide complex comprises phosphatidylserine in an amount greater than 1%, 1.5%, 2%, 2.5%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%.
  • the synthetic membrane-receiver polypeptide complex comprises phosphatidylinositol in an amount greater than 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.5%, 2%, 2.5%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, or greater than 10% relative to the total lipid content of the complex.
  • the synthetic membrane-receiver polypeptide complex comprises phosphatidic acid in an amount greater than 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1%, 1.5%, 2%, 2.5%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, or greater than 10% relative to the total lipid content of the complex.
  • the synthetic membrane-receiver polypeptide complex comprises molecules from at least one, two, or three, of the following classes of molecules, including, but not limited to, choline phospholipids, acidic phospholipids, and phosphatidylethanolamine.
  • the molar ratio of choline phospholipids to acidic phospholipids in the synthetic membrane-receiver polypeptide complex is less than 1:1000, approximately 1:1000, approximately 1:500, approximately 1:250, approximately 1:100, approximately 1:50, approximately 1:25, approximately 1:10, approximately 1:9, approximately 1:8, approximately 1:7, approximately 1:6, approximately 1:5, approximately 1:4, approximately 1:3, approximately 1:2, approximately 1:1, approximately 2:1, approximately 3:1, approximately 4:1, approximately 5:1, approximately 6:1, approximately 7:1, approximately 8:1, approximately 9:1, approximately 10:1, approximately 25:1, approximately 50:1, approximately 100:1, approximately 250:1, approximately 500:1, approximately 1000:1, or greater than approximately 1000:1.
  • the molar ratio of choline phospholipids to phosphatidyl ethanolamine in the synthetic membrane-receiver polypeptide complex is less than 1:1000, approximately 1:1000, approximately 1:500, approximately 1:250, approximately 1:100, approximately 1:50, approximately 1:25, approximately 1:10, approximately 1:9, approximately 1:8, approximately 1:7, approximately 1:6, approximately 1:5, approximately 1:4, approximately 1:3, approximately 1:2, approximately 1:1, approximately 2:1, approximately 3:1, approximately 4:1, approximately 5:1, approximately 6:1, approximately 7:1, approximately 8:1, approximately 9:1, approximately 10:1, approximately 25:1, approximately 50:1, approximately 100:1, approximately 250:1, approximately 500:1, approximately 1000:1, or greater than approximately 1000:1.
  • the molar ratio of phosphatidylethanolamine to acidic phospholipids in the synthetic membrane-receiver polypeptide complex is less than 1:1000, approximately 1:1000, approximately 1:500, approximately 1:250, approximately 1:100, approximately 1:50, approximately 1:25, approximately 1:10, approximately 1:9, approximately 1:8, approximately 1:7, approximately 1:6, approximately 1:5, approximately 1:4, approximately 1:3, approximately 1:2, approximately 1:1, approximately 2:1, approximately 3:1, approximately 4:1, approximately 5:1, approximately 6:1, approximately 7:1, approximately 8:1, approximately 9:1, approximately 10:1, approximately 25:1, approximately 50:1, approximately 100:1, approximately 250:1, approximately 500:1, approximately 1000:1, or greater than approximately 1000:1.
  • the synthetic membrane-receiver polypeptide complex comprises molecules from at least one, two, three, four, five, six, or seven of the following classes of molecules, including, but not limited to, phosphatidylcholine, sphingomyelin, lysophosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, or phosphatidic acid.
  • the lipid composition of the synthetic membrane-receiver polypeptide complex can be experimentally measured using methods known in the art including, e.g., gas-liquid chromatography or thin layer chromatography, see for example Dodge & Phillips, J Lipid Res 1967 8:667.
  • the synthetic membrane-receiver polypeptide complex comprises a lipid bilayer composed of an inner leaflet and an outer leaflet.
  • the composition of the inner and outer leaflet can be determined by transbilayer distribution assays known in the art, see e.g., Kuypers et al. Biohim Biophys Acta 1985 819:170.
  • the composition of the outer leaflet is between approximately 70-90% choline phospholipids, between approximately 0-15% acidic phospholipids, and between approximately 5-30% phosphatidylethanolamine.
  • the composition of the inner leaflet is between approximately 15-40% choline phospholipids, between approximately 10-50% acidic phospholipids, and between approximately 30-60% phosphatidylethanolamine.
  • the synthetic membrane-receiver polypeptide complex comprises cholesterol.
  • the cholesterol content is between approximately 3.0-5.5 nmol cholesterol per 10 ⁇ 7 complexes.
  • the cholesterol content is between approximately 1.8-3.5 nmol cholesterol per 10 ⁇ 7 complexes.
  • the molar ratio of cholesterol to phospholipids in the complex is between approximately 0.5-1.5.
  • the molar ratio of cholesterol to phospholipids is between approximately 0.8-1.2.
  • the molar ratio of cholesterol to phospholipids is between approximately 0.84-0.9.
  • the molar ratio of cholesterol to phospholipids is between approximately 0.5-0.75.
  • the molar ratio of cholesterol to phospholipids is between approximately 0.55-0.6.
  • the synthetic membrane-receiver polypeptide complex comprises polypeptides other than the receiver polypeptide.
  • approximately 52% of the membrane mass is protein, approximately 40% is lipid, and approximately 8% is carbohydrate.
  • approximately 7% of the carbohydrate content is comprised of glycosphingolipids and approximately 93% of the carbohydrate content is comprised of O-linked and N-linked oligosaccharides on membrane-associated polypeptides.
  • the mass ratio of lipid to protein in the synthetic membrane-receiver polypeptide complex is less than 1:1000, approximately 1:1000, approximately 1:500, approximately 1:250, approximately 1:100, approximately 1:50, approximately 1:25, approximately 1:10, approximately 1:9, approximately 1:8, approximately 1:7, approximately 1:6, approximately 1:5, approximately 1:4, approximately 1:3, approximately 1:2, approximately 1:1, approximately 2:1, approximately 3:1, approximately 4:1, approximately 5:1, approximately 6:1, approximately 7:1, approximately 8:1, approximately 9:1, approximately 10:1, approximately 25:1, approximately 50:1, approximately 100:1, approximately 250:1, approximately 500:1, approximately 1000:1, or greater than approximately 1000:1.
  • the mass ratio of lipid to carbohydrate in the synthetic membrane-receiver polypeptide complex is less than 1:1000, approximately 1:1000, approximately 1:500, approximately 1:250, approximately 1:100, approximately 1:50, approximately 1:25, approximately 1:10, approximately 1:9, approximately 1:8, approximately 1:7, approximately 1:6, approximately 1:5, approximately 1:4, approximately 1:3, approximately 1:2, approximately 1:1, approximately 2:1, approximately 3:1, approximately 4:1, approximately 5:1, approximately 6:1, approximately 7:1, approximately 8:1, approximately 9:1, approximately 10:1, approximately 25:1, approximately 50:1, approximately 100:1, approximately 250:1, approximately 500:1, approximately 1000:1, or greater than approximately 1000:1.
  • the mass ratio of carbohydrate to protein in the synthetic membrane-receiver polypeptide complex is less than 1:1000, approximately 1:1000, approximately 1:500, approximately 1:250, approximately 1:100, approximately 1:50, approximately 1:25, approximately 1:10, approximately 1:9, approximately 1:8, approximately 1:7, approximately 1:6, approximately 1:5, approximately 1:4, approximately 1:3, approximately 1:2, approximately 1:1, approximately 2:1, approximately 3:1, approximately 4:1, approximately 5:1, approximately 6:1, approximately 7:1, approximately 8:1, approximately 9:1, approximately 10:1, approximately 25:1, approximately 50:1, approximately 100:1, approximately 250:1, approximately 500:1, approximately 1000:1, or greater than approximately 1000:1.
  • the area occupancy of protein in the synthetic membrane-receiver polypeptide complex is approximately 23% and the area occupancy of lipid in the synthetic membrane-receiver polypeptide complex is approximately 77%.
  • the synthetic membrane-receiver polypeptide complex comprises a polypeptide selected from the following list, including but not limited to, spectrin, myosin-like polypeptide, band 3, SLC4A1, actin, actin-like polypeptide, glyceraldehyde 3-P dehydrogenase (G3PD).
  • a polypeptide selected from the following list, including but not limited to, spectrin, myosin-like polypeptide, band 3, SLC4A1, actin, actin-like polypeptide, glyceraldehyde 3-P dehydrogenase (G3PD).
  • the synthetic membrane-receiver polypeptide complex comprises at least one, two, three, four, five, six, or seven of the polypeptides selected from the following list, including but not limited to, spectrin, myosin-like polypeptide, band 3, SLC4A1, actin, actin-like polypeptide, glyceraldehyde 3-P dehydrogenase (G3PD).
  • the synthetic membrane-receiver complex comprises at least one polypeptide that is not the receiver. In some embodiments, the synthetic membrane-receiver complex comprises at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine or at least ten polypeptides that are not the receiver. In certain instances, the polypeptide is capable of an enzymatic or catalytic function independent of the receiver. The non-receiver polypeptide may be associated with the membrane of the synthetic membrane-receiver complex.
  • the non-receiver polypeptide may, e.g., stabilize the synthetic membrane-receiver complex, target the synthetic membrane-receiver complex to particular cells and tissues, engage the reticulo-endothelial system, protect the synthetic membrane-receiver complex from macrophages and other phagocytic cells, and/or evade other components of the innate immune system.
  • Suitable polypeptides include, e.g., complement regulatory polypeptides, inhibitors of cell-mediated degradation (e.g., CD47, CD55, and CD59), and anti-inflammatory polypeptides.
  • non-receiver polypeptides may shorten or control the half-life of the complex, including targeting to macrophages or other phagocytic cells. Suitable non-receiver polypeptides may promote apoptosis or otherwise trigger opsonization.
  • non-receiver polypeptides include polypeptide carriers, pumps, and channels; Glut1, Band3, aquaporin 1, RhAH, NA/K ATPase, Ca ATPase, Na—H exchanger, KCa3.1, KCl cotransporter, and coenzyme Q10.
  • the synthetic membrane-receiver complexes described herein can be modified to increase or decrease their half-life in circulation.
  • the half-life of the receiver and optionally the payload in circulation may be modified by altering the half-life of the synthetic membrane-receiver complex.
  • the half-life is increased and the increase may be, for instance from about 1.5-fold to 20-fold increase in serum half-life.
  • receivers may reside in circulation and may remain functional and active for substantially the duration of the synthetic membrane-receiver complex in circulation. In some embodiments, receivers may reside in circulation and may remain functional and active for more than 21 days in circulation. In some instances, synthetic membrane-receiver complexes and receivers may reside in circulation for 30 days, 45 days, 60 days, 100 days, 120 days, or longer. In other embodiments, the synthetic membrane-receiver complexes and receivers may reside in circulation for several hours to several days, such as 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 days. Residency in the circulatory system, in certain embodiments, is determined by the presence or absence of certain polypeptides on the synthetic membrane-receiver complex. For example, the synthetic membrane-receiver complex may comprise a CD47, CD55, or CD59 polypeptide or a functional fragment thereof.
  • CD47 is a membrane protein that interacts with the myeloid inhibitory immunoreceptor SIRP ⁇ (also termed CD172a or SHPS-1) that is present, e.g., on macrophages. Engagement of SIRP ⁇ by CD47 provides a down-regulatory signal that inhibits host cell phagocytosis. For example, high levels of CD47 allow cancer cells to avoid phagocytosis despite the presence pro-phagocytic signals, such as high levels of calreticulin. CD47 also has further roles in cell adhesion, e.g., by acting as an adhesion receptor for THBS1 on platelets and in the modulation of integrins.
  • SIRP ⁇ also termed CD172a or SHPS-1
  • CD47 interaction with SIRP ⁇ further prevents maturation of immature dendritic cells, inhibits cytokine production by mature dendritic cells.
  • CD47 interaction with SIRP ⁇ mediates cell-cell adhesion, enhances superantigen-dependent T-cell-mediated proliferation and co-stimulates T-cell activation.
  • CD47 is a 50 kDa membrane receptor that has extracellular N-terminal IgV domain, five transmembrane domains, and a short C-terminal intracellular tail. There are four alternatively spliced isoforms of CD47 that differ only in the length of their cytoplasmic tail.
  • the synthetic membrane-receiver complex may comprise a CD47 or a functional fragment thereof comprising one or more of: the extracellular N-terminal IgV domain, one, two, three, four, or five transmembrane domains, and/or the short C-terminal intracellular tail.
  • the cytoplasmic tail can be found as four different splice isoforms ranging from 4 to 36 amino acids.
  • the 16 amino acid form 2 is expressed in all cells of hematopoietic origin and in endothelial and epithelial cells.
  • the 36 amino acid form 4 is expressed primarily in neurons, intestine, and testis.
  • the 4 amino acid form 1 is found in epithelial and endothelial cells.
  • the expression pattern of the 23 amino acid form 3 resembles that of form 4.
  • the synthetic membrane-receiver complex comprises CD47 or a functional fragment thereof that is of one of form 1, from 2, form 3, or from 4.
  • the synthetic membrane-receiver complex does not comprise form 2.
  • the synthetic membrane-receiver complex comprises CD47 polypeptide or a functional polypeptide fragment thereof in an amount or copy number sufficient to reside in circulation for 15 days, 21 days, 30 days, 45 days, 60 days, 100 days, 120 days, or longer.
  • the synthetic membrane-receiver complex comprises a modified CD47, such as a conformational change.
  • a conformational change in CD47 is introduced so that the modified CD47 is capable of interacting with TSP-1.
  • the modified CD47 comprising the conformational change creates a different binding site for SIRP ⁇ .
  • the synthetic membrane-receiver complex comprises a modified CD47 polypeptide or a functional polypeptide fragment thereof comprising a conformational change in an amount or copy number sufficient to reside in circulation for less than 10, 9, 8, 7, 6, 5, 4, 3, 2, or less than 1 day.
  • the synthetic membrane-receiver complex comprises a fusion of a CD47 isoform to the extracellular domain of a native erythroid polypeptide.
  • the N terminus of glycophorin A may be fused to the CD47 polypeptide or functional fragment thereof, which may lead to a reduction of the SIRP ⁇ -mediated signal to macrophages to phagocytose the synthetic membrane-receiver complex.
  • generating synthetic membrane-receiver complexes includes the step of contacting a receiver (e.g., a polypeptide) with a cell, such as an erythroid cell or a platelet.
  • a receiver e.g., a polypeptide
  • CD47 is expressed in erythrocytes and platelets to mediate phagocytosis.
  • the natural levels of CD47 are altered in erythrocytes or platelets, e.g., by over-expression or inhibition of CD47 expression using any suitable method, such as the introduction of exogenous nucleic acids (e.g., expression vectors, CD47 mRNA, CD47 siRNA, and the like).
  • the natural levels of CD47 are altered such that the synthetic membrane-receiver complex resides in circulation for 15 days, 21 days, 30 days, 45 days, 60 days, 100 days, 120 days, or longer. In some embodiments, the natural levels of CD47 are altered such that the synthetic membrane-receiver complex resides in circulation for less than 10, 9, 8, 7, 6, 5, 4, 3, 2, or less than 1 day.
  • synthetic membrane-receiver complexes that are administered to a subject may comprise elevated CD47 levels when compared to native levels of a suitable control. Elevated CD47 levels may be achieved, e.g., by exogenous expression by the synthetic membrane-receiver complex of CD47 from an exogenous nucleic acid, by loading of CD47 mRNA into the complex, or by conjugating CD47 polypeptide to the surface of the complex. Elevated CD47 levels are useful to increase the half-life of the population of synthetic membrane-receiver complexes in the circulatory system of the subject.
  • the synthetic membrane-receiver complexes comprise a receiver and optionally a payload, such as a therapeutic agent.
  • a population of 10 11 synthetic membrane-receiver polypeptide complexes comprises an adenosine deaminase receiver and an exogenous CD47 polypeptide on its surface.
  • an enzyme deficiency such as ADA-SCID
  • the half-life of the synthetic membrane-receiver polypeptide complex is extended beyond that of a complex not comprising exogenous CD47 polypeptide and the subject requires less frequent dosing.
  • Half-life extension is a particular advantage when compared to current enzyme therapies not involving synthetic membrane-receiver polypeptide complexes.
  • CD47 is altered by heparin and/or chondroitin sulfate glycosaminoglycan (GAG) chains.
  • the synthetic membrane-receiver complex expresses CD47 as a proteoglycan.
  • the synthetic membrane-receiver complex comprises a CD47 proteoglycan that is conjugated to the complex.
  • the CD47 proteoglycan comprises heparin and/or chondroitin sulfate glycosaminoglycan (GAG) chains.
  • that CD47 proteoglycan has a size of greater than 150 kDa, 200 kDa, or greater than 250 kDa.
  • CD47 comprises one or more GAG chains at Ser64.
  • the residency of a synthetic membrane-receiver complex e.g., generated using erythroid cells or platelets can be further modulated by changing the amount or number of oxidized lipids on the membrane of the synthetic membrane-receiver complex.
  • the synthetic membrane-receiver complex comprises oxidized lipids in an amount effective to reside in circulation for less than 10, 9, 8, 7, 6, 5, 4, 3, 2, or less than 1 day.
  • the synthetic membrane-receiver complex comprises oxidized lipids in an amount effective to reside in circulation for 15 days, 21 days, 30 days, 45 days, 60 days, 100 days, 120 days, or longer.
  • the amount of oxidized lipids in the membrane are altered such that mobility of CD47 is increased or decreased, thereby aiding or hindering, respectively the ability of CD47 to cluster on the membrane.
  • CD55 also known as complement decay-accelerating factor or DAF, is a 70 kDa membrane protein. CD55 recognizes C4b and C3b fragments of the complement system that are created during C4 (classical complement pathway and lectin pathway) and C3 (alternate complement pathway) activation. It is thought that interaction of CD55 with cell-associated C4b and C3b proteins interferes with their ability to catalyze the conversion of C2 and factor B to active C2a and Bb and thereby prevents the formation of C4b2a and C3bBb, the amplification convertases of the complement cascade. CD55 is thought to block the formation of membrane attack complexes. CD55 may prevent lysis by the complement cascade.
  • DAF complement decay-accelerating factor
  • the synthetic membrane-receiver complex comprises CD55 polypeptide or a functional polypeptide fragment thereof in an amount or copy number sufficient to reside in circulation for 15 days, 21 days, 30 days, 45 days, 60 days, 100 days, 120 days, or longer.
  • the synthetic membrane-receiver complex comprises an exogenous CD55 polypeptide and an exogenous CD47 polypeptide or functional polypeptide fragments thereof in an amount, copy number and/or ratio sufficient to reside in circulation for 15 days, 21 days, 30 days, 45 days, 60 days, 100 days, 120 days, or longer.
  • CD59 glycoprotein also known as MAC-inhibitory protein (MAC-IP), membrane inhibitor of reactive lysis (MIRL), protectin, or HRF is a protein that attaches to host cells via a glycophosphatidylinositol (GPI) anchor.
  • MAC-IP MAC-inhibitory protein
  • MIRL membrane inhibitor of reactive lysis
  • HRF is a protein that attaches to host cells via a glycophosphatidylinositol (GPI) anchor.
  • GPI glycophosphatidylinositol
  • the synthetic membrane-receiver complex comprises CD59 polypeptide or a functional polypeptide fragment thereof in an amount or copy number sufficient to reside in circulation for 15 days, 21 days, 30 days, 45 days, 60 days, 100 days, 120 days, or longer.
  • the synthetic membrane-receiver complex comprises an exogenous CD59 polypeptide and an exogenous CD47 polypeptide or functional polypeptide fragments thereof in an amount, copy number and/or ratio sufficient to reside in circulation for 15 days, 21 days, 30 days, 45 days, 60 days, 100 days, 120 days, or longer.
  • the synthetic membrane-receiver complex comprises one or more of an exogenous CD55 polypeptide, an exogenous CD59 polypeptide and/or an exogenous CD47 polypeptide or functional polypeptide fragments thereof in an amount, copy number and/or ratio sufficient to reside in circulation for 15 days, 21 days, 30 days, 45 days, 60 days, 100 days, 120 days, or longer.
  • Effective amounts of CD47, CD55, and CD59 include 10 2 , 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , 10 9 polypeptides per synthetic membrane-receiver complex.
  • an effective amount is the amount capable of extending the synthetic membrane-receiver polypeptide complex's half-life by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, 400%, 800%, 1,000%, or 10,000% relative to the half-life that the synthetic membrane-receiver polypeptide complex would exhibit without the polypeptides.
  • a receiver is capable of interacting with a target, e.g., to associate with or bind to a target.
  • a receiver can comprise or may consist essentially of a polypeptide.
  • the receiver comprises a polypeptide, a carbohydrate, a nucleic acid, a lipid, a small molecule, or a combination thereof.
  • receivers do not interact with a target but act as payloads to be delivered by the synthetic membrane-receiver complex to a cell, tissue or other site in the body of a subject.
  • receivers comprise polypeptides.
  • Receiver polypeptides may range in size from 6 amino acids to 3000 amino acids and may exceed 6, 10, 15, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 150, 200, 300, 400 or may exceed 500 amino acids.
  • Receiver polypeptides may range in size from about 20 amino acids to about 500 amino acids, from about 30 amino acids to about 500 amino acids or from about 40 amino acids to about 500 amino acids.
  • the receiver polypeptide comprises a chimeric or fusion protein which may comprise two or more distinct protein domains.
  • These chimeric receivers are heterologous or exogenous in the sense that the various domains are derived from different sources, and as such, are not found together in nature and can be encoded e.g., by exogenous nucleic acids.
  • Receiver polypeptides can be produced by a number of methods, many of which are well known in the art and also described herein. For example, receiver polypeptides can be obtained by extraction (e.g., from isolated cells), by expression of an exogenous nucleic acid encoding the receiver polypeptide, or by chemical synthesis.
  • Receiver polypeptides can be produced by, for example, recombinant technology, and expression vectors encoding the polypeptide introduced into host cells (e.g., by transformation or transfection) for expression of the encoded receiver polypeptide.
  • conservative changes that can generally be made to an amino acid sequence without altering activity. These changes are termed conservative substitutions or mutations; that is, an amino acid belonging to a grouping of amino acids having a particular size, charge or other characteristic can be substituted for another amino acid. Substitutions for an amino acid sequence may be selected from other members of the class to which the amino acid belongs.
  • the nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, methionine, and tyrosine.
  • the polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine and glutamine.
  • the positively charged (basic) amino acids include arginine, lysine and histidine.
  • the negatively charged (acidic) amino acids include aspartic acid and glutamic acid. Such alterations are not expected to substantially affect apparent molecular weight as determined by polyacrylamide gel electrophoresis or isoelectric point. Conservative substitutions also include substituting optical isomers of the sequences for other optical isomers, specifically D amino acids for L amino acids for one or more residues of a sequence. Moreover, all of the amino acids in a sequence may undergo a D to L isomer substitution.
  • Exemplary conservative substitutions include, but are not limited to, Lys for Arg and vice versa to maintain a positive charge; Glu for Asp and vice versa to maintain a negative charge; Ser for Thr so that a free ⁇ OH is maintained; and Gln for Asn to maintain a free NH 2 .
  • point mutations, deletions, and insertions of the polypeptide sequences or corresponding nucleic acid sequences may in some cases be made without a loss of function of the polypeptide or nucleic acid fragment. Substitutions may include, e.g., 1, 2, 3, or more residues.
  • Any teaching of a specific amino acid sequence or an exogenous nucleic acid encoding the polypeptide or teaching of the name of the name thereof includes any conservative substitution point mutations, deletions, and insertions of those polypeptide sequences or corresponding nucleic acid sequences and any sequence deposited for the protein or gene in a database that can be made without a loss of function of the polypeptide or nucleic acid fragment.
  • the receiver polypeptide is associated with the membrane of the synthetic membrane-receiver polypeptide complex. In other embodiments, the receiver polypeptide is not associated with the membrane of the synthetic membrane-receiver polypeptide complex.
  • the mass ratio of lipid to receiver in the synthetic membrane-receiver polypeptide complex is less than 1:1000, approximately 1:1000, approximately 1:500, approximately 1:250, approximately 1:100, approximately 1:50, approximately 1:25, approximately 1:10, approximately 1:9, approximately 1:8, approximately 1:7, approximately 1:6, approximately 1:5, approximately 1:4, approximately 1:3, approximately 1:2, approximately 1:1, approximately 2:1, approximately 3:1, approximately 4:1, approximately 5:1, approximately 6:1, approximately 7:1, approximately 8:1, approximately 9:1, approximately 10:1, approximately 25:1, approximately 50:1, approximately 100:1, approximately 250:1, approximately 500:1, approximately 1000:1, approximately 10,000:1, approximately 100,000:1, approximately 1,000,000:1, approximately 10,000,000:1, approximately 100,000,000:1, approximately 1,000,000,000:1 or greater than approximately 1,000,000,000:1.
  • the mass ratio of non-receiver polypeptide to receiver in the synthetic membrane-receiver polypeptide complex is less than 1:1000, approximately 1:1000, approximately 1:500, approximately 1:250, approximately 1:100, approximately 1:50, approximately 1:25, approximately 1:10, approximately 1:9, approximately 1:8, approximately 1:7, approximately 1:6, approximately 1:5, approximately 1:4, approximately 1:3, approximately 1:2, approximately 1:1, approximately 2:1, approximately 3:1, approximately 4:1, approximately 5:1, approximately 6:1, approximately 7:1, approximately 8:1, approximately 9:1, approximately 10:1, approximately 25:1, approximately 50:1, approximately 100:1, approximately 250:1, approximately 500:1, approximately 1000:1, approximately 10,000:1, approximately 100,000:1, approximately 1,000,000:1, approximately 10,000,000:1, approximately 100,000,000:1, approximately 1,000,000,000:1 or greater than approximately 1,000,000,000:1.
  • the polypeptide receiver is located on the surface and is exposed to the environment around the synthetic membrane-receiver polypeptide complex. In some embodiments, the polypeptide receiver is located inside and faces the unexposed side of the synthetic membrane-receiver polypeptide complex.
  • the polypeptide receiver comprises at least one of the following domains, an S domain (surface), an A domain (anchor), and/or a U domain (unexposed), wherein the S domain is a surface domain exposed to the environment around the synthetic membrane-receiver polypeptide complex, wherein the A domain is an anchor, and wherein the U domain is located within and/or faces the unexposed side of the synthetic membrane-receiver polypeptide complex.
  • the receiver polypeptide comprises i) one or more additional S domains, termed S′ domains, or ii) one or more additional U domains, termed U′ domains.
  • the S domain and the A domain form part of the same polypeptide chain.
  • the A domain and the U domain form part of the same polypeptide chain.
  • any one or more of the S, A, U domain is added to the synthetic membrane-receiver polypeptide complex externally.
  • any one or more of the S, A, U domain is produced within the synthetic membrane-receiver polypeptide complex.
  • any one or more of the S, A, U domain is a polypeptide.
  • any one or more of the S, A, U domain is not a polypeptide.
  • FIGS. 14A , 14 B, and 14 C Schematics of exemplary conformations of receivers within or on synthetic membrane-receiver complexes are shown in FIGS. 14A , 14 B, and 14 C.
  • the A domain is a membrane polypeptide.
  • the A domain can be, e.g., an integral membrane polypeptide or a membrane associated polypeptide.
  • the A domain may be selected from one of the following classes, including but not limited to, for example, alpha-helical bitopic, alpha-helical polytopic, beta-barrel transmembrane, all alpha monotopic/peripheral, all beta monotopic/peripheral, alpha/beta monotopic/peripheral, alpha+beta monotopic/peripheral, alpha helical peptides, beta-hairpin peptides, beta-helical peptides, type 1 transmembrane protein (N-terminus extracellular), type 2 transmembrane protein (N-terminus intracellular), type 3 transmembrane protein, type 4A transmembrane protein, type 4B transmembrane protein, lipid-anchored protein, glycosylphosphatidylinositol (GPI) anchored protein, prenyl chain anchored protein, or peptides of nonregular structure.
  • alpha-helical bitopic alpha-helical polytopic
  • the A domain is endogenous, e.g., endogenous to an erythroid cell, a platelet, or a hematopoietic cell. In some embodiments, the A domain is endogenous to a mammalian cell.
  • the A domain is exogenous, e.g., exogenous to an erythroid cell, a platelet, or a hematopoietic cell. In some embodiments, the A domain is exogenous to a mammalian cell.
  • the A domain may be selected from the following molecules or fragments thereof, including but not limited to, CD1, CD2, CD3, CD4, CD5, CD6, CD7, CD8, CD9, CD10, CD11a, CD11b, CD11c, CD12w, CD13, CD14, CD15, CD16, CDw17, CD18, CD19, CD20, CD21, CD22, CD23, CD24, CD25, CD26, CD27, CD28, CD29, CD30, CD31, CD32, CD33, CD34, CD35, CD36, CD37, CD38, CD39, CD40, CD41, CD42, CD43, CD44, CD45, CD46, CD47, CD48, CD49a, CD49b, CD49c, CD49d, CD49e, CD49f, CD53, CD54, CD55, CD56, CD57, CD58, CD59, CD61, CD62E, CD62L, CD62P, CD63, CD68, CD69, CD71, CD72, CD73, CD74, CD80, CD
  • the S domain is a protein or a polypeptide. In other embodiments, the S domain is a nucleic acid. In some embodiments, the S domain is a chemical. In certain embodiment the S domain is a small molecule.
  • the S domain is a polypeptide selected from or derived from one or more of the following classes, including but not limited to, a flexible linker, an epitope tag, an enzyme, a protease, a nuclease, a receiver, an antibody-like molecule, a ligand of an antibody, a growth factor, a cytokine, a chemokine, a growth factor receptor, a cytokine receptor, a chemokine receptor, an enzymatic recognition sequence, a transpeptidase recognition sequence, a protease recognition sequence, a cleavable domain, an intein, a DNA binding protein, and RNA binding protein, a complement regulatory molecule, a complement cascade molecule, a clotting cascade molecule, a chelator, a complement regulatory domain, an SCR domain, a CCP domain, an immunoglobulin or immunogloblulin-like domain, an armadillo repeat, a leucine zipper
  • the S domain is a non-polypeptide molecule, for example a nucleic acid, a carbohydrate, or a small molecule.
  • the S domain is a nucleic acid selected from one or more of the following classes, including but not limited to, a DNA aptamer, an RNA aptamer, an siRNA, a shRNA, a single-strand RNA probe, a single strand DNA probe, an mRNA, a chemically modified oligonucleotide.
  • the S domain is a small molecule selected from one or more of the following classes, including but not limited to, a chelator, DOTA, a radionuclide, an isotope, an imaging agent, a fluorescent molecule, a chemiluminescent molecule, a gas.
  • the U domain is a protein or a polypeptide. In other embodiments, the U domain is a nucleic acid. In some embodiments, the U domain is a chemical. In certain embodiment the U domain is a small molecule.
  • the U domain is a polypeptide selected from or derived from one or more of the following classes, including but not limited to, a flexible linker, an epitope tag, an enzyme, a protease, a nuclease, a receiver, an antibody-like molecule, a ligand of an antibody, a growth factor, a cytokine, a chemokine, a growth factor receptor, a cytokine receptor, a chemokine receptor, an enzymatic recognition sequence, a transpeptidase recognition sequence, a protease recognition sequence, a cleavable domain, an intein, a DNA binding protein, and RNA binding protein, a complement regulatory molecule, a complement cascade molecule, a clotting cascade molecule, a chelator, a complement regulatory domain, an SCR domain, a CCP domain, an immunoglobulin or immunogloblulin-like domain, an armadillo repeat, a leucine zipper
  • the U domain is a non-polypeptide molecule, for example a nucleic acid, a carbohydrate, or a small molecule.
  • the U domain is a nucleic acid selected from one or more of the following classes, including but not limited to, a DNA aptamer, an RNA aptamer, an siRNA, a shRNA, a single-strand RNA probe, a single strand DNA probe, an mRNA, a chemically modified oligonucleotide.
  • the U domain is a small molecule selected from one or more of the following classes, including but not limited to, a chelator, DOTA, a radionuclide, an isotope, an imaging agent, a fluorescent molecule, a chemiluminescent molecule, a gas.
  • receiver polypeptides include: the polypeptide receiver comprises glycophorin A with HA epitope tag at the N terminus; the polypeptide receiver comprises the leader sequence of glycophorin A, HA epitope tag, and the body sequence of glycophorin A; the polypeptide receiver comprises complement receptor 1 (CR1); the polypeptide receiver comprises the leader sequence of CR1, HA epitope tag, the body sequence of CR1; the polypeptide receiver comprises the leader sequence of CR1, HA epitope tag, six SCR domains of LHR-A and LHR-B of CR1, the membrane proximal two SCR domains of CR1, the transmembrane region of CR1, and the intracellular region of CR1; the polypeptide receiver comprises the leader sequence of CR1, HA epitope tag, nine SCR domains of LHR-A and LHR-B and LHR-C of CR1, the membrane proximal two SCR domains of CR1, the transmembrane region of CR1, and the intracellular region of
  • the polypeptide receiver comprises the leader sequence of CD59, scFv, an HA epitope tag, and the body of CD59; the polypeptide receiver comprises the leader sequence of CD59, and HA epitope tag, and the body of CD59; the polypeptide receiver comprises adenosine deaminase and an HA epitope tag; the polypeptide receiver comprises phenylalanine hydroxylase and an HA epitope tag; the polypeptide receiver comprises adenosine deaminase, a (Gly3Ser)2 (SEQ ID NO: 23) flexible linker, phenylalanine hydroxylase, and an HA epitope tag; the polypeptide receiver comprises glycophorin A, adenosine deaminase at the cytoplasmic C terminus, and an HA epitope tag; the polypeptide receiver comprises glycophorin A, phenylalanine hydroxylase at the cytoplasmic C terminus, and an HA epitope tag;
  • the receiver is capable or interacting with a macrophage.
  • the receiver polypeptide may comprise one or more of: the complement receptor (Rieu et al., J. Cell Biol. 127:2081-2091 (1994)), the scavenger receptor (Brasseur et al., Photochem. Photobiol. 69:345-352 (1999)), the transferrin receptor (Dreier et al., Bioconjug. Chem. 9:482-489 (1998); Hamblin et al., J. Photochem. Photobiol. 26:4556 (1994)); the Fc receptor (Rojanasakul et al., Pharm. Res.
  • receivers capable or interacting with a macrophages include: low density lipoproteins (Mankertz et al., Biochem. Biophys. Res. Commun. 240:112-115 (1997); von Baeyer et al., Int. J. Clin. Pharmacol. Ther. Toxicol. 31:382-386 (1993)), very low density lipoproteins (Tabas et al., J. Cell Biol. 115:1547-1560 (1991)), mannose residues and other carbohydrate moieties (Pittet et al., Nucl. Med. Biol.
  • poly-cationic molecules such as poly-L-lysine (Hamblin et al., J. Photochem. Photobiol. 26:45-56 (1994)), liposomes (Bakker-Woudenberg et al., J. Drug Target. 2:363-371 (1994); Betageri et al., J. Pharm. Pharmacol. 45:48-53 (1993)) and 2-macroglobulin (Chu et al., J. Immunol. 152:1538-1545 (1994)).
  • the synthetic membrane-receiver complex does not comprise a receiver comprising an extracellular domain of an HIV coreceptor. In some embodiments, the synthetic membrane-receiver complex does not comprise a receiver capable of binding to a virus. In some embodiments, the synthetic membrane-receiver complex does not comprise a receiver comprising CD4. In some embodiments, the synthetic membrane-receiver complex does not comprise a receiver comprising an HIV coreceptor.
  • the synthetic membrane-receiver complex does not comprise a receiver comprising CXCR4, CCR5, CCR1, CCR2, CCR3, CCR4, CCR8, CXCR1, CXCR2, CXCR3, CXCR6, GPR15, APJ, CMKLR1, or CX3CR1 or a combination thereof.
  • the synthetic membrane-receiver complex does not contain an exogenous nucleic acid encoding an adenosine deaminase receiver. In some embodiments, the synthetic membrane-receiver complex does not comprise a receiver comprising adenosine deaminase (ADA).
  • ADA adenosine deaminase
  • the synthetic membrane-receiver complex does not comprise an exogenous nucleic acid encoding an oncogene. In some embodiments, the synthetic membrane-receiver complex does not comprise a receiver comprising oncogene.
  • the synthetic membrane-receiver complex does not contain an exogenous nucleic acid encoding cdx1, cdx2, or cdx4. In some embodiments, the synthetic membrane-receiver complex does not comprise a receiver comprising cdx1, cdx2, or cdx4, or a combination thereof.
  • the synthetic membrane-receiver complex does not comprise a receiver comprising a chimeric polypeptide comprising a ligand binding domain. In some embodiments, the synthetic membrane-receiver complex does not comprise a receiver comprising an S domain that is capable of binding a ligand.
  • the synthetic membrane-receiver complex does not comprise a receiver comprising CD3 ⁇ , CD3 ⁇ , an IL-2 receptor, an IL-3 receptor, an IL-4 receptor, an IL-7 receptor, an IL-11 receptor, an IL-13 receptor, a GM-CSF receptor, a LIF receptor, a CNTF receptor, an oncostatin M receptor, a TGF- ⁇ receptor, an EGF receptor, ATR2/neu, a HER2/neu, a HER3/c-erbB-3, Xmrk, an insulin receptor, an IGF-1 receptor, IRR, PDGF receptor, a CSF-1 receptor, c-kit, STK-1/flk-2, an FGF receptor, flg, bek, an NGF receptor, Ror1 and Ror2.
  • a receiver comprising CD3 ⁇ , CD3 ⁇ , an IL-2 receptor, an IL-3 receptor, an IL-4 receptor, an IL-7 receptor, an IL-11 receptor, an IL-13 receptor, a GM-
  • the synthetic membrane-receiver complex does not comprise a receiver comprising E6 or E7 genes of human papillomavirus.
  • the synthetic membrane-receiver complex does not comprise a receiver comprising a tumor antigen.
  • the synthetic membrane-receiver complex does not comprise a receiver comprising glucocerebrosidase.
  • the synthetic membrane-receiver complex does not comprise a receiver comprising asparaginase.
  • the synthetic membrane-receiver complex does not comprise a receiver comprising arginine deiminase.
  • compositions containing functional erythroid cells comprising a receiver having functional activities that are either i) not present in native erythroid cells of the same lineage, or ii) present in native erythroid cells of the same lineage in reduced levels or reduced activity levels as compared to the erythroid cells comprising the receiver.
  • Such functional activities include complement inhibition, immune complex clearance, artificial antigen presentation, modulation of the coagulation cascade, oxygen transfer, drug delivery, cytotoxin adsorption, avoidance of phagocytosis, and extension of circulation time.
  • functional erythroid cells have higher levels of a complement receptor polypeptide, such as CR1, than native erythroid cells of the same lineage by virtue of comprising a CR-1 receiver.
  • the functional erythroid cells comprising a receiver have higher levels of a complement receptor agonist polypeptide or complement associated polypeptide than native erythroid cells of the same lineage, including but not limited to, the polypeptides listed in table 7 and table 10.
  • the complement receptor receiver polypeptide comprises a human Complement Receptor-1 (CR1) polypeptide, variant, or functional fragment thereof.
  • the CR1 receiver polypeptide may be derived from one or more than one of the native alleles of CR1, e.g., the A allele (also termed the F allele or CR1*1 allele), the B allele (also termed the S allele or CR1*2 allele), the C allele (also termed the F′ allele or CR1*3 allele), or the D allele (also termed the CR1*4 allele).
  • the sequences and database accession numbers for these native forms are provided in table 4.
  • the CR1 receiver polypeptide contains a domain of a CR1 polypeptide.
  • the CR1 polypeptide may comprise one or more short consensus repeat (SCR) domains, also termed complement control protein (CCP) modules or Sushi domains, e.g., Genbank accession number AAV65577.1.
  • the CR1 receiver polypeptide comprises one or more Short Consensus Repeats (SCRs), e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44 or greater than 44 SCRs.
  • SCRs Short Consensus Repeats
  • the CR1 receiver polypeptide comprises one or more long homologous repeat (LHR) units of CR1, e.g., LHR-A, LHR-B, LHR-C, or LHR-D, e.g., 1, 2, 3, 4, 5, 6 or greater than 6 LHR domains.
  • the CR1 receiver polypeptide may comprise one or more than one extracellular domains of CR1 fused to another cell membrane protein, e.g., glycophorin A, glycophorin B, glycophorin C, glycophorin D, kell, band 3, aquaporin 1, glut 1, kidd antigen protein, rhesus antigen, including, but not limited to the cell surface moieties listed in table 1 and table 7.
  • a functional erythroid cell contains an exogenous nucleic acid encoding a complement receptor receiver polypeptide, or alternatively or in combination, a complement receptor agonist receiver polypeptide or complement associated receiver polypeptide including but not limited to, the polypeptides, and agonists to the polypeptides, listed in table 10.
  • the functional erythroid cells further contain an exogenous decay-accelerating factor (CD59, GenBank: CAG46523.1) polypeptide, or an exogenous membrane cofactor (CD46, GenBank: BAA12224.1) polypeptide, or a variant or functional fragment thereof, or a combination thereof.
  • CR1 activities include binding to C3b-containing immune complexes and shuttling of these immune complexes from circulation to liver and spleen macrophages of the reticuloendothelial system.
  • the immune complex Upon encounter with cells of the reticuloendothelial system, the immune complex is endocytosed by the phyagocytic cell but the red blood cell is spared to continue its circulation. The removal of the immune complex sometimes results in proteolytic cleavage of CR1 from the surface of the red blood cell.
  • To measure binding activity one can perform an in vitro binding assay between erythroid cells and immune complexes.
  • an in vitro phagocytosis assay with phagocytic cells and immune complex-loaded erythroid cells.
  • an in vivo clearance of circulating immune complexes to the liver one can perform a clearance and biodistribution assay using radiolabeled immune complexes.
  • compositions containing functional erythroid cells containing a receiver comprising a native polypeptide at a level greater than that of a hematopoietic cell of the same lineage not comprising the receiver polypeptide are provided.
  • populations of functional erythroid cells contain receivers, such as complement receptor 1 levels at least about 1.1, e.g., 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, 10000, or more than 10000 times greater than corresponding hematopoietic cells of the same lineage that lack the CR1 receiver polypeptide.
  • compositions that contain populations of functional erythroid cells with CR1 levels of at least about 2500, 5000, 6000, 7000, 8000, 9000, 10000, 15000, 20000, 25000, 30000, 40000, 50000, 100000, 200000, 300000, 400000, 500000, 600000, 700000, 800000, 900000, 1000000, or more than 1000000 molecules per cell.
  • CR1 levels in wild-type and synthetic membrane-receiver polypeptide complexes can be measured and quantified by, for example, flow cytometry with antibodies specific for CR1.
  • the receiver interacts with a circulating pathogen, such as a virus or a bacterium.
  • the functional erythroid cell expresses an exogenous gene encoding an antibody, scFv, or nanobody specific for the circulating pathogen.
  • the antibody, scFv, or nanobody may be expressed as a fusion protein.
  • the antibody, scFv, or nanobody receiver or another receiver with affinity to circulating pathogens is loaded into or onto the erythroid cell.
  • the antibody, scFv, or nanobody receiver or the other receiver with affinity to circulating pathogens may be localized intracellularly or extracellularly.
  • the receiver is specific for a viral or bacterial antigen, such as a surface, envelope or capsid antigen.
  • the receiver interacts with a toxin, preferably a foreign toxin, such as derived from a pathogen or otherwise from the environment.
  • the functional erythroid cell expresses a exogenous gene encoding a receiver comprising an amino acid sequence derived from lipopolysaccharide-binding protein (LBP), bactericidal/permeability-increasing protein (BPI), amyloid P component, or a cationic protein.
  • LBP lipopolysaccharide-binding protein
  • BPI bactericidal/permeability-increasing protein
  • amyloid P component or a cationic protein.
  • Toxin-binding receivers may be expressed as a fusion protein.
  • toxin-binding receivers may be loaded into or onto the erythroid cell.
  • Toxin-binding receivers may be localized intracellularly or extracellularly.
  • the toxin binding receiver is specific for a bacterial toxin such as botul
  • synthetic membrane-receiver complexes may express a receiver capable of enhancing its ability to sequester a target.
  • Potential sequestration enhancement receivers include the polypeptide transporters including, but not limited to, those in table 1.
  • the receiver comprises a polypeptide that comprises an amino acid sequence derived from Duffy Antigen Receptor for Chemokines (DARC).
  • the functional erythroid cell expresses a exogenous gene encoding an amino acid sequence derived from Duffy Antigen Receptor for Chemokines (DARC).
  • the DARC receiver may be expressed as a full-length protein or a fragment thereof. DARC may be expressed as a fusion protein.
  • DARC protein is loaded into or onto the erythroid cell. In some embodiments, the loaded DARC is additionally functionalized or otherwise modified.
  • the DARC receiver molecule may be localized intracellularly or extracellularly.
  • DARC was identified as a potent multi-ligand chemokine receptor.
  • DARC belongs to the family of rhodopsin-like seven-helix transmembrane proteins. Besides erythrocytes DARC is expressed in post capillary venular endothelial cells, which are the primary site of leukocyte transmigration in most tissues. DARC provides a highly specific binding site for both CC and CXC chemokines. DARC is thought to possess a higher affinity for ELR motif CXC chemokines. CXC chemokines are neutrophil chemoattractants and may potentially be pro-angiogenic.
  • DARC dissociation constant
  • DARC is thought to be a) multi-specific; b) unable to initiate intracellular signals, and c) chemokines bound to erythrocyte surface are believed to be inaccessible to their normal target inflammatory cells (Neote, J Biol Chem, 1993). Erythrocytes may play a role in the regulation of inflammatory processes through the presence of DARC
  • Inflammatory signaling molecules such as cytokines
  • cytokines can trigger local and systemic tissue damage when present in high concentrations. Bursts of cytokines are implicated in the pathogenesis of bacterial sepsis, rheumatoid arthritis, and several other inflammatory diseases.
  • Functional erythroid cells that exogenously express natural cytokine receptors or synthetic antibody-like receptor mimics can sequester the inflammatory cytokines.
  • An exemplary chemokine receptor is DARC.
  • DARC exemplary chemokine receptor
  • functional erythroid cells expressing DARC receiver may be used to modulate chemokine levels in circulation and/or within the body's peripheral tissues.
  • the functional erythroid cells comprising a DARC receiver can either be marked for destruction or can slowly release the inflammatory mediators back into circulation, but at a low and diffuse concentration.
  • the functional erythroid cell comprising a receiver that comprises a chemokine or cytokine receptor may act as a reservoir for signal transduction peptides.
  • the receiver comprises a polypeptide that comprises an amino acid sequence derived from an antibody.
  • the functional erythroid cell expresses a exogenous gene encoding an amino acid sequence derived from an antibody.
  • the antibody receiver may be expressed as a full-length protein or a fragment thereof.
  • the antibody may be expressed as a fusion protein.
  • the antibody protein is loaded into or onto the erythroid cell.
  • the loaded antibody is additionally functionalized or otherwise modified.
  • the antibody receiver may be localized intracellularly or extracellularly.
  • the receiver comprises an antibody amino acid sequence that is specific for a desired target.
  • the antibody is a scFv.
  • the antibody is a nanobody.
  • the functional erythroid cells comprise a receiver that comprises an antibody or fragment thereof that is specific for a target and is located on the cell surface.
  • a variable fragment (Fv) of an antibody specific for botulinum toxin binding is expressed on the surface of the erythroid cell.
  • Botulinum toxin binding antibodies are known in the art (Amersdorfer, Inf and Immunity, 1997), as is the expression of the Fv portion of an antibody (Hoedemaeker, Journ of Bio Chemistry, 1997).
  • the toxin is retained by the erythroid cell through the Fv region, sequestered and shuttled via the circulatory system to the liver for clearance from the body.
  • the receiver comprises a polypeptide that comprises an amino acid sequence derived from a scFv antibody.
  • the functional erythroid cell expresses a exogenous gene encoding an amino acid sequence derived from a scFv antibody.
  • the scFv antibody receiver may be expressed as a full-length protein or a fragment thereof.
  • the scFv antibody may be expressed as a fusion protein.
  • the scFv protein is loaded into or onto the erythroid cell.
  • Suitable scFv receiver polypeptides that may be expressed by functional erythroid cells include, but are not limited to, those listed in table 7.
  • scFv antibodies have been constructed mainly from hybridoma, spleen cells from immunized mice, and B lymphocytes from human.
  • the variable region of an antibody is formed by the noncovalent heterodimer of the variable domains of the V(H) and V(L) domains, which can then be used in the construction of a recombinant scFv antibody.
  • scFvs The production of scFvs is known in the art and require mRNA to first be isolated from hybridoma (or also from the spleen, lymph cells, and bone morrow) followed by reverse transcription into cDNA to serve as a template for antibody gene amplification (PCR).
  • PCR antibody gene amplification
  • the scFv receiver may be made specific to any target molecule including, but not limited to, those in table 5.
  • a scFv receiver specific for anthrax toxin may be expressed on a functional erythroid cell.
  • an effective dose of a population of erythroid cell comprising a receiver molecule specific for anthrax toxin can be used to capture and sequester the anthrax toxin.
  • the erythroid cell migrates to the liver where clearance occurs.
  • erythrocytes comprise a receiver comprising a camelid-derived nanobody expressed on the surface of the cell.
  • Nanobodies are usually 12-15 kDa. They are considerably smaller than antibodies and scFv. Nanobodies may thus be easier to transfect, and the nanobody receiver will be more easily expressed, translated and or transported to the cell surface in an erythroid cell.
  • nanobody receivers are employed to minimize immunogenic effects caused by a specific receiver. Nanobodies because of their small size will offer reduced immunogenic potential.
  • receiver nanobodies are employed because they limit changes in the mechanical and morphological behavior of the plasma membrane of the functional erythroid cell. This may allow the functional erythroid cell to exhibit normal circulatory red blood cell behavior.
  • receiver nanobodies are employed because they have an increased ability to recognize hidden or uncommon epitopes compared to standard antibodies. For example, they can bind to small enzymatic cavities of a target and modulate the molecular behavior of the target.
  • functional erythroid cells comprise receiver nanobodies with specificity to target epitopes of molecules in the human complement system. Such functional erythroid cells may be administered to a subject in need thereof to selectively deplete one or more over-active factors of the complement system.
  • C5 may be targeted by erythroid cells comprising receiver nanobodies with specificity to target epitopes of C5 and cleared from the system by the erythroid cells upon administration of the cells into a subject.
  • This approach is suitable to provide a therapeutic effect, e.g., for a complement disorder, such as paroxysmal nocturnal hemoglobinuria.
  • functional erythroid cells comprise receiver nanobodies with specificity to target epitopes of molecules including, but not limited to, those listed in table 5.
  • the receiver comprises a polypeptide that comprises an amino acid sequence derived from one of proteases, nucleases, amylase, lyase (sucrase) or hydrolase (DNase, lipase).
  • the functional erythroid cell expresses a exogenous gene encoding an amino acid sequence derived from one of proteases, nucleases, amylase, lyase (sucrase) or hydrolase (DNase, lipase).
  • Receiver proteases, nucleases, amylases, lyases and hydrolases may be expressed as a full-length protein or a fragment thereof.
  • Receiver proteases, nucleases, amylases, lyases and hydrolases may be expressed as a fusion protein.
  • receiver proteases, nucleases, amylases, lyases or hydrolases are loaded into or onto the erythroid cell.
  • the loaded receiver proteases, nucleases, amylases, lyases or hydrolases are additionally functionalized or otherwise modified.
  • the receiver protease, nuclease, amylase, lyase or hydrolase receiver molecule may be localized intracellularly or extracellularly.
  • functional erythroid cells comprise a receiver comprising a protease, a nuclease, an amylase, a lyase or a hydrolase.
  • the functional erythroid cell comprising a protease, a nuclease, an amylase, a lyase or a hydrolase receiver is capable of degrading a target on the erythroid cell independent of circulatory clearance, e.g., by macrophages in the liver.
  • functional erythroid cells comprising a receiver comprising a protease, a nuclease, an amylase, a lyase or a hydrolase may be administered to a subject in need thereof to treat a cancer by selectively degrading metabolites that are essential for cancer cell growth.
  • asparaginase is used to decrease local asparagine levels to treat acute lymphoblastic leukemia and acute myeloid leukemia.
  • Suitable receivers may, e.g., comprise one or both of the two major classes of enzymes capable of degrading target molecules, lyases and hydrolases.
  • functional erythroid cells are provided comprising a receiver comprising a molecule including but not limited to those listed in table 7.
  • erythrocytes comprise a receiver comprising a lyase.
  • the lyase is valine decarboxylase.
  • Valine decarboxylase receiver may be expressed within the intracellular space of the erythroid cell.
  • Functional erythroid cells comprising a valine decarboxylase receiver may be administered to a subject in need thereof to modulate valine levels within the blood.
  • Erythroid cells comprising a valine decarboxylase receiver are suitable to treat valinemia, an inherited disorder that increases levels of the amino acid valine in the blood. Affected individuals typically develop vomiting, failure to thrive, intellectual disability, and fatigue. Valinemia is caused by a deficiency of the valine transaminase enzyme and has an autosomal recessive pattern of inheritance.
  • erythrocytes comprise a receiver comprising a hydrolase.
  • the hydrolase is deoxyribonuclease I (DNase I).
  • DNase I receiver may be expressed on the surface of the erythroid cell.
  • Functional erythroid cells comprising a DNase I receiver may be administered to a subject in need thereof to preferentially cleave circulating DNA at phosphodiester linkages adjacent to a pyrimidine nucleotide, yielding 5′-phosphate-terminated polynucleotides with a free hydroxyl group on position 3′. On average tetra-nucleotides are produced.
  • Erythroid cells comprising a DNase I receiver are suitable to treat conditions exacerbated by high levels of immunogenic DNA in circulation, such as systemic lupus erythematosus (SLE).
  • SLE systemic lupus erythematosus
  • the receiver is capable of responding to an external stimulus, e.g., upon binding to a ligand or contacting the stimulus, wherein responding entails, for example, moving, re-folding, changing conformation, forming a dimer, forming a homodimer, forming a heterodimer, forming a multimer, transducing a signal, emitting energy in a detectable form (e.g., fluorescence), functionally interacting with another receiver, or functionally interacting with a non-receiver polypeptide.
  • a detectable form e.g., fluorescence
  • the synthetic membrane-receiver complex does not comprise a fusion molecule capable of promoting fusion of the synthetic membrane-receiver complex to a target cell that is i) different from and/or ii) acts independent of the receiver, wherein the receiver is capable of interacting with a target.
  • the synthetic membrane-receiver complex does not comprise a receiver comprising Syncytin-1.
  • the synthetic membrane-receiver complex does not comprise a photosensitive synthetic compound, such as, e.g. a compound that can be activated by photons or quenchable compounds. In some embodiments, the synthetic membrane-receiver complex does not comprise an activatable molecular detection agent capable of producing a detectable response. In some embodiments, the synthetic membrane-receiver complex does not comprise a diagnostic compound. In some embodiments, the synthetic membrane-receiver complex does not comprise a virus or bacterium.
  • the polypeptide receiver is expressed within the synthetic membrane-receiver polypeptide complex.
  • the polypeptide receiver may be exhibited on the surface of the synthetic membrane-receiver polypeptide complex or may reside within the synthetic membrane-receiver polypeptide complex.
  • the polypeptide receiver is conjugated to the synthetic membrane-receiver polypeptide complex.
  • the polypeptide receiver usually is conjugated to the surface of the synthetic membrane-receiver polypeptide complex. Conjugation may be achieved chemically or enzymatically, by methods known in the art and described herein.
  • Non-polypeptide receivers may also be conjugated to a synthetic membrane-receiver complex. In some embodiments, the receiver is not conjugated to the synthetic membrane-receiver complex.
  • the polypeptide receiver is loaded into the synthetic membrane-receiver polypeptide complex.
  • Non-polypeptide receivers may also be loaded within a synthetic membrane-receiver complex.
  • the receiver is not loaded into or onto the synthetic membrane-receiver complex.
  • the synthetic membrane-receiver complex comprises a receiver that is optionally expressed from an exogenous nucleic acid, conjugated to the complex, loaded into or onto the complex, and any combination thereof.
  • the synthetic membrane-receiver complex comprises a therapeutic agent or other payload.
  • the synthetic membrane-receiver complex is generated by contacting a suitable isolated cell, e.g., an erythroid cell, a reticulocyte, an erythroid cell precursor, a platelet, or a platelet precursor, with an exogenous nucleic acid encoding a receiver polypeptide.
  • the receiver polypeptide is encoded by a DNA, which is contacted with a nucleated erythroid precursor cell or a nucleated platelet precursor cell.
  • the receiver polypeptide is encoded by an RNA, which is contacted with a platelet, a nucleate erythroid cell, a nucleated platelet precursor cell, or a reticulocyte.
  • the receiver is a polypeptide, which is contacted with a primary platelet, a nucleated erythroid cell, a nucleated platelet precursor cell, a reticulocyte, or an erythrocyte.
  • a receiver polypeptide may be expressed from a transgene introduced into an erythroid cell by electroporation, chemical or polymeric transfection, viral transduction, mechanical membrane disruption, or other method; a receiver polypeptide that is expressed from mRNA that is introduced into a cell by electroporation, chemical or polymeric transfection, viral transduction, mechanical membrane disruption, or other method; a receiver polypeptide that is over-expressed from the native locus by the introduction of an external factor, e.g., a transcriptional activator, transcriptional repressor, or secretory pathway enhancer; and/or a receiver polypeptide that is synthesized, extracted, or produced from a production cell or other external system and incorporated into the erythroid cell.
  • an external factor e.g., a transcriptional activator, transcriptional repressor, or secretory pathway enhancer
  • the polypeptide receiver is expressed within the synthetic membrane-receiver polypeptide complex.
  • the polypeptide receiver may be exhibited on the surface of the synthetic membrane-receiver polypeptide complex or may reside within the synthetic membrane-receiver polypeptide complex.
  • the synthetic membrane-receiver polypeptide complex is a cell, e.g., an erythroid cell or a platelet expressing a receiver polypeptide.
  • Receiver polypeptides can be introduced by transfection of single or multiple copies of genes, transduction with a virus, or electroporation in the presence of DNA or RNA.
  • Methods for expression of exogenous proteins in mammalian cells are well known in the art. For example, expression of exogenous factor IX in hematopoietic cells is induced by viral transduction of CD34+ progenitor cells, see Chang et al., Nat Biotechnol 2006, 24:1017.
  • Nucleic acids such as DNA expression vectors or mRNA for producing the receiver polypeptide may be introduced into progenitor cells (e.g., an erythroid cell progenitor or a platelet progenitor and the like) that are suitable to produce the synthetic membrane-receiver polypeptide complexes described herein.
  • the progenitor cells can be isolated from an original source or obtained from expanded progenitor cell population via routine recombinant technology as provided herein.
  • the expression vectors can be designed such that they can incorporate into the genome of cells by homologous or non-homologous recombination by methods known in the art.
  • a nucleic acid encoding a non-receiver polypeptide that can selectively target and cut the genome for example a CRISPR/Cas9, transcriptional activator-like effector nuclease (TALEN), or zinc finger nuclease
  • TALEN transcriptional activator-like effector nuclease
  • zinc finger nuclease is used to direct the insertion of the exogenous nucleic acid of the expression vector encoding the receiver polypeptide to a particular genomic location, for example the CR1 locus (1q32.2), the hemoglobin locus (11p15.4), or another erythroid-associated protein including, but not limited to, those listed in table 1 and table 3.
  • the exogenous nucleic acid is an RNA molecule, or a DNA molecule that encodes for an RNA molecule, that silences or represses the expression of a target gene.
  • the molecule can be a small interfering RNA (siRNA), an antisense RNA molecule, or a short hairpin RNA (shRNA) molecule.
  • Methods for transferring expression vectors into progenitor cells that are suitable to produce the synthetic membrane-receiver polypeptide complexes described herein include, but are not limited to, viral mediated gene transfer, liposome mediated transfer, transformation, gene guns, transfection and transduction, e.g., viral mediated gene transfer such as the use of vectors based on DNA viruses such as adenovirus, adenoassociated virus and herpes virus, as well as retroviral based vectors.
  • modes of gene transfer include e.g., naked DNA, CaPO 4 precipitation, DEAE dextran, electroporation, protoplast fusion, lipofection, and cell microinjection.
  • a progenitor cell subject to transfer of an exogenous nucleic acid that encodes a polypeptide receiver can be cultured under suitable conditions allowing for differentiation into mature enucleated red blood cells, e.g., the in vitro culturing process described herein.
  • the resulting enucleated red blood cells display proteins associated with mature erythrocytes, e.g., hemoglobin, glycophorin A, and receiver polypeptides which can be validated and quantified by standard methods (e.g., Western blotting or FACS analysis).
  • Isolated mature enucleated red blood cells comprising a receiver and platelets comprising a receiver are two examples of synthetic membrane-receiver polypeptide complexes of the invention.
  • the synthetic membrane-receiver complex is generated by contacting a reticulocyte with an exogenous nucleic acid encoding a receiver polypeptide.
  • the receiver polypeptide is encoded by an RNA which is contacted with a reticulocyte.
  • the receiver is a polypeptide which is contacted with a reticulocyte.
  • Isolated reticulocytes may be transfected with mRNA encoding a receiver polypeptide to generate a synthetic membrane-receiver comple.
  • Messenger RNA may be derived from in vitro transcription of a cDNA plasmid construct containing the coding sequence corresponding to the receiver polypeptide.
  • the cDNA sequence corresponding to the receiver polypeptide may be inserted into a cloning vector containing a promoter sequence compatible with specific RNA polymerases.
  • the cloning vector ZAP Express® pBK-CMV (Stratagene, La Jolla, Calif., USA) contains T3 and T7 promoter sequence compatible with T3 and T7 RNA polymerase, respectively.
  • the plasmid is linearized at a restriction site downstream of the stop codon(s) corresponding to the end of the coding sequence of the receiver polypeptide.
  • the mRNA is transcribed from the linear DNA template using a commercially available kit such as, for example, the RNAMaxx® High Yield Transcription Kit (from Stratagene, La Jolla, Calif., USA). In some instances, it may be desirable to generate 5′-m7GpppG-capped mRNA.
  • transcription of a linearized cDNA template may be carried out using, for example, the mMESSAGE mMACHINE High Yield Capped RNA Transcription Kit from Ambion (Austin, Tex., USA).
  • Transcription may be carried out in a reaction volume of 20-100 ⁇ l at 37° C. for 30 min to 4 h.
  • the transcribed mRNA is purified from the reaction mix by a brief treatment with DNase I to eliminate the linearized DNA template followed by precipitation in 70% ethanol in the presence of lithium chloride, sodium acetate or ammonium acetate.
  • the integrity of the transcribed mRNA may be assessed using electrophoresis with an agarose-formaldehyde gel or commercially available Novex pre-cast TBE gels (e.g., Novex, Invitrogen, Carlsbad, Calif., USA).
  • Messenger RNA encoding the receiver polypeptide may be introduced into reticulocytes using a variety of approaches including, for example, lipofection and electroporation (van Tandeloo et al., Blood 98:49-56 (2001)).
  • lipofection for example, 5 mg of in vitro transcribed mRNA in Opti-MEM (Invitrogen, Carlsbad, Calif., USA) is incubated for 5-15 min at a 1:4 ratio with the cationic lipid DMRIE-C(Invitrogen).
  • lipids or cationic polymers may be used to transfect cells with mRNA including, for example, DOTAP, various forms of polyethylenimine, and polyL-lysine (Sigma-Aldrich, Saint Louis, Mo., USA), and Superfect (Qiagen, Inc., Valencia, Calif., USA; See, e.g., Bettinger et al., Nucleic Acids Res. 29:3882-3891 (2001)).
  • DOTAP various forms of polyethylenimine
  • polyL-lysine Sigma-Aldrich, Saint Louis, Mo., USA
  • Superfect Qiagen, Inc., Valencia, Calif., USA; See, e.g., Bettinger et al., Nucleic Acids Res. 29:3882-3891 (2001)
  • the resulting mRNA/lipid complexes are incubated with cells (1-2 ⁇ 10 6 cells/ml) for 2 h at 37° C., washed and returned to culture.
  • electroporation for example, about 5 to 20 ⁇ 10 6 cells in 500 ⁇ l of Opti-MEM (Invitrogen, Carlsbad, Calif., USA) are mixed with about 20 ⁇ g of in vitro transcribed mRNA and electroporated in a 0.4-cm cuvette using, for example, and Easyject Plus device (EquiBio, Kent, United Kingdom).
  • the electroporation parameters required to efficiently transfect cells with mRNA appear to be less detrimental to cells than those required for electroporation of DNA (van Tandeloo et al., Blood 98:49-56 (2001)).
  • mRNA may be transfected into a reticulocyte using a peptide-mediated RNA delivery strategy (See, e.g., Bettinger et al., Nucleic Acids Res. 29:3882-3891 (2001)).
  • a peptide-mediated RNA delivery strategy See, e.g., Bettinger et al., Nucleic Acids Res. 29:3882-3891 (2001)).
  • the cationic lipid polyethylenimine 2 kDA Sigma-Aldrich, Saint Louis, Mo., USA
  • the melittin peptide Alta Biosciences, Birmingham, UK
  • the mellitin peptide may be conjugated to the PEI using a disulfide cross-linker such as, for example, the hetero-bifunctional cross-linker succinimidyl 3-(2-pyridyldithio) propionate.
  • a disulfide cross-linker such as, for example, the hetero-bifunctional cross-linker succinimidyl 3-(2-pyridyldithio) propionate.
  • RNA/peptide/lipid complex In vitro transcribed mRNA is preincubated for 5 to 15 min with the mellitin-PEI to form an RNA/peptide/lipid complex. This complex is then added to cells in serum-free culture medium for 2 to 4 h at 37° C. in a 5% CO 2 humidified environment and then removed and the transfected cells allowed to continue growing in culture.
  • the synthetic membrane-receiver complex is generated by contacting a suitable isolated erythroid cell precursor or a platelet precursor with an exogenous nucleic acid encoding a receiver polypeptide.
  • the receiver polypeptide is encoded by a DNA, which is contacted with a nucleated erythroid precursor cell or a nucleated platelet precursor cell.
  • the receiver polypeptide is encoded by an RNA, which is contacted with a platelet, a nucleate erythroid cell, or a nucleated platelet precursor cell.
  • Receivers may be genetically introduced into erythroid cell precursors, platelet precursor, or nucleated erythroid cells prior to terminal differentiation using a variety of DNA techniques, including transient or stable transfections and gene therapy approaches.
  • the receiver polypeptides may be expressed on the surface and/or in the cytoplasm of mature red blood cell or platelet.
  • Viral gene transfer may be used to transfect the cells with DNA encoding a receiver polypeptide.
  • viruses may be used as gene transfer vehicles including Moloney murine leukemia virus (MMLV), adenovirus, adeno-associated virus (AAV), herpes simplex virus (HSV), lentiviruses such as human immunodeficiency virus 1 (HIV 1), and spumaviruses such as foamy viruses, for example (See, e.g., Osten et al., HEP 178:177-202 (2007)).
  • Retroviruses for example, efficiently transduce mammalian cells including human cells and integrate into chromosomes, conferring stable gene transfer.
  • a receiver polypeptide may be transfected into an erythroid cell precursor, a platelet precursor, or a nucleated erythroid cell, expressed and subsequently retained and exhibited in a mature red blood cell or platelet.
  • a suitable vector is the Moloney murine leukemia virus (MMLV) vector backbone (Malik et al., Blood 91:2664-2671 (1998)). Vectors based on MMLV, an oncogenic retrovirus, are currently used in gene therapy clinical trials (Hossle et al., News Physiol. Sci. 17:87-92 (2002)).
  • MMLV Moloney murine leukemia virus
  • Vectors based on MMLV, an oncogenic retrovirus are currently used in gene therapy clinical trials (Hossle et al., News Physiol. Sci. 17:87-92 (2002)).
  • a DNA construct containing the cDNA encoding a receiver polypeptide can be generated in the MMLV vector backbone using standard molecular biology
  • the construct is transfected into a packaging cell line such as, for example, PA317 cells and the viral supernatant is used to transfect producer cells such as, for example, PG13 cells.
  • the PG13 viral supernatant is incubated with an erythroid cell precursor, a platelet precursor, or a nucleated erythroid cell that has been isolated and cultured or has been freshly isolated as described herein.
  • the expression of the receiver polypeptide may be monitored using FACS analysis (fluorescence-activated cell sorting), for example, with a fluorescently labeled antibody directed against the receiver polypeptide, if it is located on the surface of the synthetic membrane-receiver polypeptide complex. Similar methods may be used to express a receiver polypeptide that is located in the inside of the synthetic membrane-receiver polypeptide complex.
  • a fluorescent tracking molecule such as, for example, green fluorescent protein (GFP) may be transfected using a viral-based approach (Tao et al., Stem Cells 25:670-678 (2007)).
  • Ecotopic retroviral vectors containing DNA encoding the enhanced green fluorescent protein (EGFP) or a red fluorescent protein (e.g., DsRed-Express) are packaged using a packaging cell such as, for example, the Phoenix-Eco cell line (distributed by Orbigen, San Diego, Calif.).
  • Packaging cell lines stably express viral proteins needed for proper viral packaging including, for example, gag, pol, and env.
  • Supernatants from the Phoenix-Eco cells into which viral particles have been shed are used to transduce e.g., erythroid cell precursors, platelet precursors, or a nucleated erythroid cells.
  • transduction may be performed on a specially coated surface such as, for example, fragments of recombinant fibronectin to improve the efficiency of retroviral mediated gene transfer (e.g., RetroNectin, Takara Bio USA, Madison, Wis.). Cells are incubated in RetroNectin-coated plates with retroviral Phoenix-Eco supernatants plus suitable co-factors. Transduction may be repeated the next day.
  • the percentage of cells expressing EGFP or DsRed-Express may be assessed by FACS.
  • Other reporter genes that may be used to assess transduction efficiency include, for example, beta-galactosidase, chloramphenicol acetyltransferase, and luciferase as well as low-affinity nerve growth factor receptor (LNGFR), and the human cell surface CD24 antigen (Bierhuizen et al., Leukemia 13:605-613 (1999)).
  • Nonviral vectors may be used to introduce genetic material into suitable erythroid cells, platelets or precursors thereof to generate synthetic membrane-receiver polypeptide complexes.
  • Nonviral-mediated gene transfer differs from viral-mediated gene transfer in that the plasmid vectors contain no proteins, are less toxic and easier to scale up, and have no host cell preferences.
  • the “naked DNA” of plasmid vectors is by itself inefficient in delivering genetic material encoding a receiver polypeptide to a cell and therefore is combined with a gene delivery method that enables entry into cells.
  • a number of delivery methods may be used to transfer nonviral vectors into suitable erythroid cells, platelets or precursors thereof including chemical and physical methods.
  • a nonviral vector encoding a receiver polypeptide may be introduced into suitable erythroid cells, platelets or precursors thereof using synthetic macromolecules such as cationic lipids and polymers (Papapetrou et al., Gene Therapy 12:S118-S130 (2005)).
  • Cationic liposomes for example form complexes with DNA through charge interactions.
  • the positively charged DNA/lipid complexes bind to the negative cell surface and are taken up by the cell by endocytosis. This approach may be used, for example, to transfect hematopoietic cells (See, e.g., Keller et al., Gene Therapy 6:931-938 (1999)).
  • plasmid DNA For erythroid cells, platelets or precursors thereof the plasmid DNA (approximately 0.5 ⁇ g in 25-100 ⁇ L of a serum free medium, such as, for example, OptiMEM (Invitrogen, Carlsbad, Calif.)) is mixed with a cationic liposome (approximately 4 ⁇ g in 25 ⁇ L of serum free medium) such as the commercially available transfection reagent LipofectamineTM (Invitrogen, Carlsbad, Calif.) and allowed to incubate for at least 20 min to form complexes.
  • a serum free medium such as, for example, OptiMEM (Invitrogen, Carlsbad, Calif.)
  • a cationic liposome approximately 4 ⁇ g in 25 ⁇ L of serum free medium
  • LipofectamineTM the commercially available transfection reagent LipofectamineTM
  • the DNA/liposome complex is added to suitable erythroid cells, platelets or precursors thereof and allowed to incubate for 5-24 h, after which time transgene expression or the receiver polypeptide may be assayed.
  • liposome tranfection agents e.g., In vivo GeneSHUTTLETM, Qbiogene, Carlsbad, Calif.).
  • a cationic polymer such as, for example, polyethylenimine (PEI) may be used to efficiently transfect erythroid cell progenitor cells, for example hematopoietic and umbilical cord blood-derived CD34+ cells (See, e.g., Shin et al., Biochim. Biophys. Acta 1725:377-384 (2005)).
  • PEI polyethylenimine
  • Human CD34+ cells are isolated from human umbilical cord blood and cultured in Iscove's modified Dulbecco's medium supplemented with 200 ng/ml stem cell factor and 20% heat-inactivated fetal bovine serum.
  • Plasmid DNA encoding the receiver polypeptide is incubated with branched or linear PEIs varying in size from 0.8 K to 750 K (Sigma Aldrich, Saint Louis, Mo., USA; Fermetas, Hanover, Md., USA).
  • PEI is prepared as a stock solution at 4.2 mg/ml distilled water and slightly acidified to pH 5.0 using HCl.
  • the DNA may be combined with the PEI for 30 min at room temperature at various nitrogen/phosphate ratios based on the calculation that 1 ⁇ g of DNA contains 3 nmol phosphate and 1 ⁇ l of PEI stock solution contains 10 nmol amine nitrogen.
  • the isolated CD34+ cells are seeded with the DNA/cationic complex, centrifuged at 280 ⁇ g for 5 min and incubated in culture medium for 4 or more h until gene expression of the receiver polypeptide is assessed.
  • a plasmid vector may be introduced into suitable erythroid cells, platelets or precursors thereof using a physical method such as particle-mediated transfection, “gene gun”, biolistics, or particle bombardment technology (Papapetrou, et al., (2005) Gene Therapy 12:S118-S130).
  • DNA encoding the receiver polypeptide is absorbed onto gold particles and administered to cells by a particle gun.
  • This approach may be used, for example, to transfect erythroid progenitor cells, e.g., hematopoietic stem cells derived from umbilical cord blood (See, e.g., Verma et al., Gene Therapy 5:692-699 (1998)).
  • CD34+ cells are purified using an anti-CD34 monoclonal antibody in combination with magnetic microbeads coated with a secondary antibody and a magnetic isolation system (e.g., Miltenyi MiniMac System, Auburn, Calif., USA).
  • the CD34+ enriched cells may be cultured as described herein.
  • plasmid DNA encoding the receiver polypeptide is precipitated onto a particle, for example gold beads, by treatment with calcium chloride and spermidine.
  • the beads may be delivered into the cultured cells using, for example, a Biolistic PDS-1000/He System (Bio-Rad, Hercules, Calif., USA).
  • a reporter gene such as, for example, beta-galactosidase, chloramphenicol acetyltransferase, luciferase, or green fluorescent protein may be used to assess efficiency of transfection.
  • electroporation methods may be used to introduce a plasmid vector into suitable erythroid cells, platelets or precursors thereof. Electroporation creates transient pores in the cell membrane, allowing for the introduction of various molecules into the cells including, for example, DNA and RNA as well as antibodies and drugs.
  • CD34+ cells are isolated and cultured as described herein Immediately prior to electroporation, the cells are isolated by centrifugation for 10 min at 250 ⁇ g at room temperature and resuspended at 0.2-10 ⁇ 10 ⁇ 6 viable cells/ml in an electroporation buffer such as, for example, X-VIVO 10 supplemented with 1.0% human serum albumin (HSA).
  • an electroporation buffer such as, for example, X-VIVO 10 supplemented with 1.0% human serum albumin (HSA).
  • the plasmid DNA (1-50 ⁇ g) is added to an appropriate electroporation cuvette along with 500 ⁇ l of cell suspension. Electroporation may be done using, for example, an ECM 600 electroporator (Genetronics, San Diego, Calif., USA) with voltages ranging from 200 V to 280 V and pulse lengths ranging from 25 to 70 milliseconds.
  • ECM 600 electroporator Gene Pulser XcellTM, BioRad, Hercules, Calif.; Cellject Duo, Thermo Science, Milford, Mass.
  • efficient electroporation of isolated CD34+ cells may be performed using the following parameters: 4 mm cuvette, 1600 ⁇ F, 550 V/cm, and 10 ⁇ g of DNA per 500 ⁇ l of cells at 1 ⁇ 105 cells/ml (Oldak et al., Acta Biochimica Polonica 49:625-632 (2002)).
  • Nucleofection a form of electroporation, may also be used to transfect suitable erythroid cells, platelets or precursors thereof.
  • transfection is performed using electrical parameters in cell-type specific solutions that enable DNA (or other reagents) to be directly transported to the nucleus thus reducing the risk of possible degradation in the cytoplasm.
  • a Human CD34 Cell NucleofectorTM Kit (from amaxa inc.) may be used to transfect suitable erythroid cells, platelets or precursors thereof.
  • 1-5 ⁇ 10 6 cells in Human CD34 Cell NucleofectorTM Solution are mixed with 1-5 ⁇ g of DNA and transfected in the NucleofectorTM instrument using preprogrammed settings as determined by the manufacturer.
  • Erythroid cells, platelets or precursors thereof may be non-virally transfected with a conventional expression vector which is unable to self-replicate in mammalian cells unless it is integrated in the genome.
  • erythroid cells, platelets or precursors thereof may be transfected with an episomal vector which may persist in the host nucleus as autonomously replicating genetic units without integration into chromosomes (Papapetrou et al., Gene Therapy 12:S118-S130 (2005)).
  • viruses exploit genetic elements derived from viruses that are normally extrachromosomally replicating in cells upon latent infection such as, for example, EBV, human polyomavirus BK, bovine papilloma virus-1 (BPV-1), herpes simplex virus-1 (HSV) and Simian virus 40 (SV40).
  • Mammalian artificial chromosomes may also be used for nonviral gene transfer (Vanderbyl et al., Exp. Hematol. 33:1470-1476 (2005)).
  • Exogenous nucleic acids encoding a polypeptide receiver may be assembled into expression vectors by standard molecular biology methods known in the art, e.g., restriction digestion, overlap-extension PCR, and Gibson assembly.
  • Exogenous nucleic acids may comprise a gene encoding a polypeptide receiver that is not normally expressed on the cell surface, e.g., of an erythroid cell, fused to a gene that encodes an endogenous or native membrane protein, such that the receiver polypeptide is expressed on the cell surface.
  • a exogenous gene encoding a receiver polypeptide can be cloned at the N terminus following the leader sequence of a type 1 membrane protein, at the C terminus of a type 2 membrane protein, or upstream of the GPI attachment site of a GPI-linked membrane protein.
  • the flexible linker is a poly-glycine poly-serine linker such as [Gly4Ser]3_(SEQ ID NO: 24) commonly used in generating single-chain antibody fragments from full-length antibodies (Antibody Engineering: Methods & Protocols, Lo 2004), or ala-gly-ser-thr polypeptides such as those used to generate single-chain Arc repressors (Robinson & Sauer, PNAS 1998).
  • the flexible linker provides the receiver polypeptide with more flexibility and steric freedom than the equivalent construct without the flexible linker. This added flexibility is useful in applications that require binding to a target, e.g., an antibody or protein, or an enzymatic reaction of the receiver for which the active site must be accessible to the substrate (e.g., the target).
  • An epitope tag may be placed between two fused genes, such as, e.g., a nucleic acid sequence encoding an HA epitope tag—amino acids YPYDVPDYA (Seq. ID No. 4), a CMyc tag—amino acids EQKLISEEDL (Seq. ID No. 5), or a Flag tag—amino acids DYKDDDDK (Seq. ID No. 6).
  • the epitope tag may be used for the facile detection and quantification of expression using antibodies against the epitope tag by flow cytometry, western blot, or immunoprecipitation.
  • the synthetic membrane-receiver polypeptide comprises a receiver polypeptide and at least one other heterologous polypeptide.
  • the second polypeptide can be a fluorescent protein.
  • the fluorescent protein can be used as a reporter to assess transduction efficiency.
  • the fluorescent protein is used as a reporter to assess expression levels of the receiver polypeptide if both are made from the same transcript.
  • the at least one other polypeptide is heterologous and provides a function, such as, e.g., multiple antigens, multiple capture targets, enzyme cascade.
  • the recombinant nucleic acid comprises a gene encoding a receiver and a second gene, wherein the second gene is separated from the gene encoding the receiver by a viral-derived T2A sequence (gagggcagaggaagtcttctaacatgcggtgacgtggaggsgsstcccggcct (Seq. ID No. 7)) that is post-translationally cleaved into two mature proteins.
  • a viral-derived T2A sequence gagggcagaggaagtcttctaacatgcggtgacgtggaggaggsgsstcccggccct (Seq. ID No. 7)
  • the receiver polypeptide is complement receptor 1 (CR-1).
  • the gene sequence for complement receptor 1 is amplified using PCR.
  • the exogenous nucleic acid encoding a receiver polypeptide comprises a gene sequence for a scFv against hepatitis B antigen that is fused to the 3′ end of the sequence for Kell and amplified using PCR.
  • the exogenous nucleic acid encoding a receiver polypeptide comprises a gene sequence for a scFv against hepatitis B antigen that is fused to a poly-glycine/serine linker, followed by the 3′ end of the sequence for Kell, and amplified using PCR.
  • the exogenous nucleic acid encoding a receiver polypeptide comprises the 3′ end of a gene sequence for a scFv against hepatitis B antigen that is fused to an epitope tag sequence, of which may be one, or a combination of, an; HA-tag, Green fluorescent protein tag, Myc-tag, chitin binding protein, maltose binding protein, glutathione-S-transferase, poly(His)tag, thioredoxin, poly(NANP), FLAG-tag, V5-tag, AviTag, Calmodulin-tag, polyglutamate-tag, E-tag, S-tag, SBP-tag, Softag-1, Softag-3, Strep-tag, TC-tag, VSV-tag, Xpress-tag, Isopeptag, SpyTag, biotin carboxyl carrier protein, Nus-tag, Fc-tag, or Ty-tag.
  • an epitope tag sequence of which may be one, or a
  • the entire construct is fused to the 3′ end of the sequence for Kell and then amplified using PCR.
  • the exogenous gene constructs encoding the various receiver polypeptides are, for example, subsequently loaded into a lentiviral vector and used to transduce a CD34+ cell population.
  • the gene comprising an adenosine deaminase receiver is placed in the pSP64 vector.
  • the vector is linearized and RNA polymerase generates mRNA coding for the receiver polypeptide.
  • a population of neutrophils is electroporated using an Ingenio electroporation kit such that 10, 100, 1,000, 10,000 TU/ml of mRNA coding for surface expression of GluN1 receiver to generate a synthetic membrane-receiver polypeptide complex.
  • a population of platelet cells is incubated with Trans-IT mRNA and 10, 100, or 1000 TU/ml (transducing units/ml) of mRNA coding for thymidine phosphorylase protein receiver to generate a synthetic membrane-receiver polypeptide complex.
  • a population of erythroid cells is incubated with lentiviral vectors comprising exogenous nucleic acid encoding a receiver polypeptide, specific plasmids of which may include; pLKO.1 puro, PLKO.1—TRC cloning vector, pSico, FUGW, pLVTHM, pLJM1, pLionII, pMD2.G, pCMV-VSV-G, pCI-VSVG, pCMV-dR8.2 dvpr, psPAX2, pRSV-Rev, and pMDLg/pRRE to generate a synthetic membrane-receiver polypeptide complex.
  • the vectors may be administered at 10, 100, 1,000, 10,000 pfu and incubated for 12 hrs.
  • a population of erythroid cells is incubated with Lipofectamine 2000 and 10, 100, or 1000 TU/ml (transducing units/ml) of DNA coding for oxalase receiver.
  • the polypeptide receiver is conjugated to the synthetic membrane-receiver polypeptide complex.
  • the polypeptide receiver usually is conjugated to the surface of the synthetic membrane-receiver polypeptide complex. Conjugation may be achieved chemically or enzymatically. Non-polypeptide receivers may also be conjugated to a synthetic membrane-receiver complex.
  • the synthetic membrane-receiver complex comprises a receiver that is chemically conjugated.
  • Chemical conjugation of a receiver may be accomplished by covalent bonding of the receiver to another molecule, with or without use of a linker.
  • the formation of such conjugates is within the skill of artisans and various techniques are known for accomplishing the conjugation, with the choice of the particular technique being guided by the materials to be conjugated.
  • amino acids to the polypeptide (C- or N-terminal) which contain ionizable side chains, e.g., aspartic acid, glutamic acid, lysine, arginine, cysteine, histidine, or tyrosine, and are not contained in the active portion of the polypeptide sequence, serve in their unprotonated state as a potent nucleophile to engage in various bioconjugation reactions with reactive groups attached to polymers, e.g., homo- or hetero-bi-functional PEG (e.g., Lutolf and Hubbell, Biomacromolecules 2003; 4:713-22, Hermanson, Bioconjugate Techniques, London. Academic Press Ltd; 1996).
  • ionizable side chains e.g., aspartic acid, glutamic acid, lysine, arginine, cysteine, histidine, or tyrosine
  • reactive groups attached to polymers e.g., homo- or hetero-bi-functional P
  • Receiver conjugation is not restricted to polypeptides, e.g., a peptide ligand, an antibody, an antibody fragment, or aptamer but is applicable also for non-polypeptide receivers, e.g., lipids, carbohydrates, nucleic acids, and small molecules.
  • polypeptides e.g., a peptide ligand, an antibody, an antibody fragment, or aptamer
  • non-polypeptide receivers e.g., lipids, carbohydrates, nucleic acids, and small molecules.
  • the receiver may be bound to the surface of a synthetic membrane-receiver complex through a biotin-streptavidin bridge.
  • a biotinylated antibody receiver may be linked to a non-specifically biotinylated surface of the synthetic membrane-receiver complex through a streptavidin bridge.
  • Antibodies can be conjugated to biotin by a number of chemical means (See, e.g., Hirsch et al., Methods Mol. Biol. 295: 135-154 (2004)).
  • Any surface membrane proteins of a synthetic membrane-receiver complex may be biotinylated using an amine reactive biotinylation reagent such as, for example, EZ-Link Sulfo-NHS-SS-Biotin (sulfosuccinimidyl 2-(biotinamido)-ethyl-1,3-dithiopropionate; Pierce-Thermo Scientific, Rockford, Ill., USA; See, e.g., Jaiswal et al., Nature Biotech. 21:47-51 (2003)).
  • isolated erythroid cells may be incubated for 30 min at 4° C.
  • the receiver may be attached to the surface of, e.g., an erythroid cell or platelet with a bispecific antibody to generate the synthetic membrane-receiver complex.
  • a bispecific antibody can have specificity for the erythroid cell or platelet and the receiver.
  • the receiver is attached to, e.g., an erythroid cell or platelet via a covalent attachment to generate a synthetic membrane-receiver complex.
  • the receiver may be derivatized and bound to the erythroid cell or platelet using a coupling compound containing an electrophilic group that will react with nucleophiles on the erythroid cell or platelet to form the interbonded relationship.
  • electrophilic groups are ⁇ , ⁇ unsaturated carbonyls, alkyl halides and thiol reagents such as substituted maleimides.
  • the coupling compound can be coupled to a receiver polypeptide via one or more of the functional groups in the polypeptide such as amino, carboxyl and tryosine groups.
  • the functional groups in the polypeptide such as amino, carboxyl and tryosine groups.
  • coupling compounds should contain free carboxyl groups, free amino groups, aromatic amino groups, and other groups capable of reaction with enzyme functional groups.
  • Highly charged receivers can also be prepared for immobilization on, e.g., erythroid cells or platelets through electrostatic bonding to generate synthetic membrane-receiver complexes. Examples of these derivatives would include polylysyl and polyglutamyl enzymes.
  • the choice of the reactive group embodied in the derivative depends on the reactive conditions employed to couple the electrophile with the nucleophilic groups on the erythroid cell or platelet for immobilization.
  • a controlling factor is the desire not to inactivate the coupling agent prior to coupling of the receiver immobilized by the attachment to the erythroid cell or platelet.
  • Such coupling immobilization reactions can proceed in a number of ways.
  • a coupling agent can be used to form a bridge between the receiver and the erythroid cell or platelet.
  • the coupling agent should possess a functional group such as a carboxyl group which can be caused to react with the receiver.
  • One way of preparing the receiver for conjugation includes the utilization of carboxyl groups in the coupling agent to form mixed anhydrides which react with the receiver, in which use is made of an activator which is capable of forming the mixed anhydride.
  • activators are isobutylchloroformate or other chloroformates which give a mixed anhydride with coupling agents such as 5,5′-(dithiobis(2-nitrobenzoic acid) (DTNB), p-chloromercuribenzoate (CMB), or m-maleimidobenzoic acid (MBA).
  • DTNB 5,5′-(dithiobis(2-nitrobenzoic acid)
  • CMB p-chloromercuribenzoate
  • MSA m-maleimidobenzoic acid
  • Functional groups on a receiver polypeptide such as carboxyl groups can be activated with carbodiimides and the like activators. Subsequently, functional groups on the bridging reagent, such as amino groups, will react with the activated group on the receiver polypeptide to form the reactive derivative.
  • the coupling agent should possess a second reactive group which will react with appropriate nucleophilic groups on the erythroid cell or platelet to form the bridge.
  • Typical of such reactive groups are alkylating agents such as iodoacetic acid, ⁇ , ⁇ unsaturated carbonyl compounds, such as acrylic acid and the like, thiol reagents, such as mercurials, substituted maleimides and the like.
  • functional groups on the receiver can be activated so as to react directly with nucleophiles on, e.g., erythroid cells or platelets to obviate the need for a bridge-forming compound.
  • an activator such as Woodward's Reagent K or the like reagent which brings about the formation of carboxyl groups in the receiver into enol esters, as distinguished from mixed anhydrides.
  • the enol ester derivatives of receivers subsequently react with nucleophilic groups on, e.g., an erythroid cell or platelet to effect immobilization of the receiver, thereby creating a synthetic membrane-receiver complex.
  • the synthetic membrane-receiver complex is generated by contacting an erythroid cell with a receiver and optionally a payload, wherein contacting does not include conjugating the receiver to the erythroid cell using an attachment site comprising Band 3 (CD233), aquaporin-1, Glut-1, Kidd antigen, RhAg/Rli50 (CD241), Rli (CD240), Rh30CE (CD240CE), Rh30D (CD240D), Kx, glycophorin B (CD235b), glycophorin C (CD235c), glycophorin D (CD235d), Kell (CD238), Duffy/DARCi (CD234), CR1 (CD35), DAF (CD55), Globoside, CD44, ICAM-4 (CD242), Lu/B-CAM (CD239), XG1/XG2 (CD99), EMMPRIN/neurothelin (CD147), JMH, Glycosyltransferase, Cartwright, Dombrock, C4A/
  • the synthetic membrane-receiver complex comprises a receiver that is enzymatically conjugated.
  • the receiver can be conjugated to the surface of, e.g., an erythroid cell or platelet by various chemical and enzymatic means, including but not limited to those listed in table 9 to generate a synthetic membrane-receiver complex.
  • these methods include chemical conjugation with bifunctional cross-linking agents such as, e.g., an NHS ester-maleimide heterobifunctional crosslinker to connect a primary amine group with a reduced thiol group.
  • These methods also include enzymatic strategies such as, e.g., transpeptidase reaction mediated by a sortase enzyme to connect one polypeptide containing the acceptor sequence LPXTG (SEQ ID NO: 25) or LPXTA (SEQ ID NO: 26) with a polypeptide containing the N-terminal donor sequence GGG, see e.g., Swee et al., PNAS 2013.
  • the methods also include combination methods, such as e.g., sortase-mediated conjugation of Click Chemistry handles (an azide and an alkyne) on the antigen and the cell, respectively, followed by a cyclo-addition reaction to chemically bond the antigen to the cell, see e.g., Neves et al., Bioconjugate Chemistry, 2013.
  • a catalytic bond-forming polypeptide domain can be expressed on or in e.g., an erythroid cell or platelet, either intracellularly or extracellularly.
  • SpyTag and SpyCatcher undergo isopeptide bond formation between Asp117 on SpyTag and Lys31 on SpyCatcher.
  • the reaction is compatible with the cellular environment and highly specific for protein/peptide conjugation (Zakeri, B.; Fierer, J. O.; Celik, E.; Chittock, E.
  • SpyTag and SpyCatcher has been shown to direct post-translational topological modification in elastin-like protein. For example, placement of SpyTag at the N-terminus and SpyCatcher at the C-terminus directs formation of circular elastin-like proteins (Zhang et al, Journal of the American Chemical Society, 2013).
  • the components SpyTag and SpyCatcher can be interchanged such that a system in which molecule A is fused to SpyTag and molecule B is fused to SpyCatcher is functionally equivalent to a system in which molecule A is fused to SpyCatcher and molecule B is fused to SpyTag.
  • a system in which molecule A is fused to SpyTag and molecule B is fused to SpyCatcher is functionally equivalent to a system in which molecule A is fused to SpyCatcher and molecule B is fused to SpyTag.
  • SpyTag and SpyCatcher when used, it is to be understood that the complementary molecule could be substituted in its place.
  • a catalytic bond-forming polypeptide such as a SpyTag/SpyCatcher system, can be used to attach the receiver to the surface of, e.g., an erythroid cell, to generate a synthetic membrane-receiver complex.
  • the SpyTag polypeptide sequence can be expressed on the extracellular surface of the erythroid cell.
  • the SpyTag polypeptide can be, for example, fused to the N terminus of a type-1 or type-3 transmembrane protein, e.g., glycophorin A, fused to the C terminus of a type-2 transmembrane protein, e.g., Kell, inserted in-frame at the extracellular terminus or in an extracellular loop of a multi-pass transmembrane protein, e.g., Band 3, fused to a GPI-acceptor polypeptide, e.g., CD55 or CD59, fused to a lipid-chain-anchored polypeptide, or fused to a peripheral membrane protein.
  • a type-1 or type-3 transmembrane protein e.g., glycophorin A
  • a type-2 transmembrane protein e.g., Kell
  • a GPI-acceptor polypeptide e.g., CD55 or CD59
  • the nucleic acid sequence encoding the SpyTag fusion can be expressed within a synthetic membrane-receiver complex.
  • a receiver polypeptide can be fused to SpyCatcher.
  • the nucleic acid sequence encoding the SpyCatcher fusion can be expressed and secreted from the same erythroid cell that expresses the SpyTag fusion.
  • the nucleic acid sequence encoding the SpyCatcher fusion can be produced exogenously, for example in a bacterial, fungal, insect, mammalian, or cell-free production system.
  • a covalent bond will be formed that attaches the receiver to the surface of the erythroid cell to form a synthetic membrane-receiver complex.
  • An erythroid cell comprising the receiver polypeptide fusion is an example of a synthetic membrane-receiver polypeptide complex that comprises a conjugated receiver.
  • the SpyTag polypeptide may be expressed as a fusion to the N terminus of glycophorin A under the control of the Gata1 promoter in an erythroid cell.
  • a receiver polypeptide for example complement receptor 1 and the receivers listed in table 7, fused to the SpyCatcher polypeptide sequence can be expressed under the control of the Gata1 promoter in the same erythroid cell.
  • an isopeptide bond will be formed between the SpyTag and SpyCatcher polypeptides, forming a covalent bond between the erythroid cell surface and the receiver polypeptide.
  • An erythroid cell comprising the receiver polypeptide fusion is an example of a synthetic membrane-receiver polypeptide complex that comprises a conjugated receiver.
  • the SpyTag polypeptide may be expressed as a fusion to the N terminus of glycophorin A under the control of the Gata1 promoter in an erythroid cell.
  • a receiver polypeptide for example complement receptor 1 fused to the SpyCatcher polypeptide sequence can be expressed in a suitable mammalian cell expression system, for example HEK293 cells.
  • the SpyCatcher fusion polypeptide can be brought in contact with the cell. Under suitable reaction conditions, an isopeptide bond will be formed between the SpyTag and SpyCatcher polypeptides, forming a covalent bond between the erythroid cell surface and the receiver polypeptide.
  • An erythroid cell comprising the receiver polypeptide fusion is an example of a synthetic membrane-receiver polypeptide complex that comprises a conjugated receiver.
  • a catalytic bond-forming polypeptide such as a SpyTag/SpyCatcher system
  • the SpyTag polypeptide sequence can be expressed in the intracellular space of the erythroid cell by a number of methods, including direct expression of the transgene, fusion to an endogenous intracellular protein such as, e.g., hemoglobin, fusion to the intracellular domain of endogenous cell surface proteins such as, e.g., Band 3, glycophorin A, Kell, or fusion to a structural component of the erythroid cytoskeleton.
  • the SpyTag sequence is not limited to a polypeptide terminus and may be integrated within the interior sequence of an endogenous polypeptide such that polypeptide translation and localization is not perturbed.
  • a receiver polypeptide can be fused to SpyCatcher.
  • the nucleic acid sequence encoding the SpyCatcher fusion can be expressed within the same erythroid cell that expresses the SpyTag fusion.
  • a covalent bond will be formed that acts to anchor the receiver polypeptide in the intracellular space of the erythroid cell.
  • An erythroid cell comprising the receiver polypeptide fusion is an example of a synthetic membrane-receiver polypeptide complex that comprises a conjugated receiver.
  • an erythroid cell may express SpyTag fused to hemoglobin beta intracellularly.
  • the erythroid cell may be genetically modified with a gene sequence that includes a hemoglobin promoter, beta globin gene and a SpyTag sequence such that upon translation, intracellular beta globin is fused to SpyTag at is C terminus.
  • the erythroid cell expresses a Gata1 promoter-led gene that codes for SpyCatcher driving phenylalanine hydroxylase (PAH) expression such that upon translation, intracellular PAH is fused to SpyCatcher at its N terminus Upon expression of both fusion proteins the SpyTag bound beta globin is linked through an isopeptide bond to the SpyCatcher bound PAH in the intracellular space, allowing PAH to be anchored to beta globin and retained during maturation.
  • An erythroid cell comprising the receiver polypeptide fusion is an example of a synthetic membrane-receiver polypeptide complex that comprises a conjugated receiver.
  • the SpyTag polypeptide can be expressed as a fusion to the receiver polypeptide within an erythroid cell.
  • the SpyCatcher polypeptide can be expressed as a fusion to the C terminus (intracellular) of glycophorin A within the same erythroid cell.
  • an isopeptide bond will be formed between the SpyTag and SpyCatcher polypeptides, forming a covalent bond between the membrane-anchored endogenous erythroid polypeptide and the receiver molecule.
  • An erythroid cell comprising the receiver polypeptide fusion is an example of a synthetic membrane-receiver polypeptide complex that comprises a conjugated receiver.
  • the polypeptides may be directly conjugated to each other or indirectly through a linker.
  • the linker may be a peptide, a polymer, an aptamer, or a nucleic acid.
  • the polymer may be, e.g., natural, synthetic, linear, or branched.
  • Receiver polypeptides can comprise a heterologous fusion protein that comprises a first polypeptide and a second polypeptide with the fusion protein comprising the polypeptides directly joined to each other or with intervening linker sequences and/or further sequences at one or both ends.
  • the conjugation to the linker may be through covalent bonds or ionic bonds.
  • the polypeptide receiver is loaded into the synthetic membrane-receiver polypeptide complex.
  • Non-polypeptide receivers may also be loaded within a synthetic membrane-receiver complex.
  • synthetic membrane-receiver complexes are generated by loading, e.g., erythroid cells or platelets with one or more receivers, such that the one or more receivers are internalized within the erythroid cells or platelets.
  • the erythroid cells or platelets may additionally be loaded with a payload, such as, e.g., a therapeutic agent.
  • a number of methods may be used to load, e.g., erythroid cells or platelets with a receiver and optionally a payload (e.g., a therapeutic agent).
  • Suitable methods include, for example, hypotonic lysis, hypotonic dialysis, osmosis, osmotic pulsing, osmotic shock, ionophoresis, electroporation, sonication, microinjection, calcium precipitation, membrane intercalation, lipid mediated transfection, detergent treatment, viral infection, diffusion, receptor mediated endocytosis, use of protein transduction domains, particle firing, membrane fusion, freeze-thawing, mechanical disruption, and filtration. Any one such method or a combination thereof may be used to generate the synthetic membrane-receiver complexes described herein.
  • Erythroid cell specifically red blood cells may be hypotonically lysed by adding 30-50 fold volume excess of 5 mM phosphate buffer (pH 8) to a pellet of isolated red blood cells. The resulting lysed cell membranes are isolated by centrifugation. The pellet of lysed red blood cell membranes is resuspended and incubated in the presence of the receiver and/or therapeutic agent in a low ionic strength buffer, e.g., for 30 min.
  • a low ionic strength buffer e.g., for 30 min.
  • the lysed red blood cell membranes may be incubated with the receiver or the payload (e.g., a therapeutic agent) for as little as one minute or as long as several days, depending upon the best conditions determined to efficiently load the erythroid cells.
  • the payload e.g., a therapeutic agent
  • erythroid cells specifically red blood cells may be loaded with a receiver and optionally a payload (e.g., a therapeutic agent) using controlled dialysis against a hypotonic solution to swell the cells and create pores in the cell membrane (See, e.g., U.S. Pat. Nos. 4,327,710; 5,753,221; and 6,495,351).
  • a payload e.g., a therapeutic agent
  • a pellet of isolated red blood cells is resuspended in 10 mM HEPES, 140 mM NaCl, 5 mM glucose pH 7.4 and dialyzed against a low ionic strength buffer containing 10 mM NaH 2 PO 4 , 10 mM NaHCO 3 , 20 mM glucose, and 4 mM MgCl 2 , pH 7.4.
  • the red blood cells are further dialyzed against 16 mM NaH 2 PO 4 , pH 7.4 solution containing the receiver or the payload (e.g., a therapeutic agent) for an additional 30-60 min. All of these procedures may be advantageously performed at a temperature of 4° C.
  • erythroid cells specifically red blood cells with a therapeutic agent
  • a dialysis approach and a specific apparatus designed for this purpose may be used (See, e.g., U.S. Pat. Nos. 4,327,710, 6,139,836 and 6,495,351 B2).
  • the loaded erythroid cells can be resealed by gentle heating in the presence of a physiological solution such as, for example, 0.9% saline, phosphate buffered saline, Ringer's solution, cell culture medium, blood plasma or lymphatic fluid.
  • a physiological solution such as, for example, 0.9% saline, phosphate buffered saline, Ringer's solution, cell culture medium, blood plasma or lymphatic fluid.
  • well-sealed membranes may be generated by treating the disrupted erythroid cells, specifically red blood cells for 1-2 min in 150 mM salt solution of, for example, 100 mM phosphate (pH 8.0) and 150 mM sodium chloride at a temperature of 60° C.
  • the cells may be incubated at a temperature of 25-50° C. for 30 min to 4 h (See, e.g., U.S.
  • the disrupted red blood cells may be resealed by incubation in 5 mM adenine, 100 mM inosine, 2 mM ATP, 100 mM glucose, 100 mM Na-pyruvate, 4 mM MgCl2, 194 mM NaCl, 1.6 M KCl, and 35 mM NaH 2 PO 4 , pH 7.4 at a temperature of 37° C. for 20-30 min (See, e.g., U.S. Pat. No. 5,753,221).
  • erythroid cells or platelets are exposed to an electrical field which causes transient holes in the cell membrane, allowing the receiver and optional payload (e.g., therapeutic agent) to diffuse into the cell (See, e.g., U.S. Pat. No. 4,935,223).
  • Erythroid cells specifically red blood cells, for example, are suspended in a physiological and electrically conductive media such as platelet-free plasma to which the receiver and optional payload (e.g., therapeutic agent) is added.
  • the mixture in a volume ranging from 0.2 to 1.0 ml is placed in an electroporation cuvette and cooled on ice for 10 min.
  • the cuvette is placed in an electroporation apparatus such as, for example, an ECM 830 (from BTX Instrument Division, Harvard Apparatus, Holliston, Mass.).
  • ECM 830 from BTX Instrument Division, Harvard Apparatus, Holliston, Mass.
  • the cells are electroporated with a single pulse of approximately 2.4 milliseconds in length and a field strength of approximately 2.0 kV/cm.
  • electroporation of erythroid cells, specifically red blood cells may be carried out using double pulses of 2.2 kV delivered at 0.25 ⁇ F using a Bio-Rad Gene Pulsar apparatus (Bio-Rad, Hercules, Calif., USA) to achieve a loading capacity of over 60% (Flynn et al., Cancer Lett. 82:225-229 (1994)).
  • the cuvette is returned to the ice bath for 10-60 min and then placed in a 37° C. water bath to induce resealing of the cell membrane. Any suitable electroporation method may be used to generate the synthetic membrane-receiver complexes described herein.
  • erythroid cells are, for example, exposed to high intensity sound waves, causing transient disruption of the cell membrane allowing the receiver and optional payload (e.g., therapeutic agent) to diffuse into the cell.
  • payload e.g., therapeutic agent
  • Any suitable sonication method may be used to generate the synthetic membrane-receiver complexes described herein.
  • erythroid cells for example, are treated with a mild detergent which transiently compromises the cell membrane by creating holes through which the receiver and optional payload (e.g., therapeutic agent) may diffuse.
  • the detergent is washed from the cells.
  • the detergent may be saponin. Any suitable detergent treatment method may be used to generate the synthetic membrane-receiver complexes described herein.
  • erythroid cells may have a surface receptor which upon binding of the receiver or payload (e.g., therapeutic agent) induces internalization of the receptor and the associated receiver or payload (e.g., therapeutic agent).
  • the receiver or payload e.g., therapeutic agent
  • Any suitable endocytosis method may be used to generate the synthetic membrane-receiver complexes described herein.
  • the receiver and optional payload may be loaded, e.g., into an erythroid cell or platelet by fusing or conjugating the receiver or payload to proteins and/or polypeptides capable of crossing or translocating the plasma membrane (See, e.g., U.S. Patent Application 2002/0151004 A1).
  • protein domains and sequences that are capable of translocating a cell membrane include, for example, sequences from the HIV-1-transactivating protein (TAT), the Drosophila Antennapedia homeodomain protein, the herpes simplex-1 virus VP22 protein, and transportin, a fusion between the neuropeptide galanin and the wasp venom peptide mastoparan.
  • a payload may be fused or conjugated to all or part of the TAT peptide.
  • a receiver fusion protein containing all or part of the TAT peptide and/or a fusion protein containing all or part of the TAT peptide and the payload e.g., a therapeutic agent, such as an antibody, enzyme, or peptide
  • the payload e.g., a therapeutic agent, such as an antibody, enzyme, or peptide
  • all or part of the TAT peptide may be chemically coupled to a functional group associated with the payload (e.g., therapeutic agent) such as, for example, a hydroxyl, carboxyl or amino group.
  • the link between the TAT peptide and the payload may be pH sensitive such that once the conjugate or fusion has entered the intracellular environment, the therapeutic agent is separated from the TAT peptide.
  • the synthetic membrane-receiver complex is generated by contacting an erythroid cell with a receiver and optionally a payload without lysing and resealing the cells to incorporate the receiver and/or payload. In some embodiments, the synthetic membrane-receiver complex is generated by contacting an erythroid cell with a receiver and optionally a payload, wherein contacting does not comprise hypotonic dialysis.
  • the synthetic membrane-receiver complex is generated by contacting an erythroid cell with a receiver and optionally a payload, wherein contacting does not include loading the receiver and/or payload into or onto the erythroid cell.
  • the receiver is generated in an entity that is not the erythroid cell to be contacted and/or the receiver is isolated from a sample that does not comprise the erythroid cell to be contacted.
  • suitable entities include a cell line, an in vitro expression system, a bacterial expression system, etc.
  • erythroid cells may be bombarded with the receiver and optional payload (e.g., therapeutic agent) attached to a heavy or charged particle such as, for example, gold microcarriers and are mechanically or electrically accelerated such that they traverse the cell membrane.
  • payload e.g., therapeutic agent
  • a heavy or charged particle such as, for example, gold microcarriers
  • Microparticle bombardment may be achieved using, for example, the Helios Gene Gun (from, e.g., Bio-Rad, Hercules, Calif., USA). Any suitable microparticle bombardment method may be used to generate the synthetic membrane-receiver complexes described herein.
  • erythroid cells or platelets may be loaded with a receiver and optional payload (e.g., therapeutic agent) by fusion with a synthetic vesicle such as, for example, a liposome.
  • a receiver and optional payload e.g., therapeutic agent
  • the vesicles themselves are loaded with the receiver and optional payload using one or more of the methods described herein or known in the art.
  • the receiver and optional payload e.g., therapeutic agent
  • the loaded vesicles are then fused with the erythroid cells or platelets under conditions that enhance cell fusion.
  • Fusion of a liposome, for example, with a cell may be facilitated using various inducing agents such as, for example, proteins, peptides, polyethylene glycol (PEG), and viral envelope proteins or by changes in medium conditions such as pH (See, e.g., U.S. Pat. No. 5,677,176). Any suitable liposomal fusion method may be used to generate the synthetic membrane-receiver complexes described herein.
  • various inducing agents such as, for example, proteins, peptides, polyethylene glycol (PEG), and viral envelope proteins or by changes in medium conditions such as pH (See, e.g., U.S. Pat. No. 5,677,176).
  • Any suitable liposomal fusion method may be used to generate the synthetic membrane-receiver complexes described herein.
  • erythroid cells or platelets and the receiver and optional payload may be forced through a filter of pore size smaller than the cell causing transient disruption of the cell membrane and allowing the receiver and optional therapeutic agent to enter the cell.
  • Any suitable filtration method may be used to generate the synthetic membrane-receiver complexes described herein.
  • erythroid cells are subjected to several freeze thaw cycles, resulting in cell membrane disruption (See, e.g., U.S. Patent Application 2007/0243137 A1).
  • a pellet of packed red blood cells 0.1-1.0 ml
  • an isotonic solution e.g., phosphate buffered saline
  • the receiver and optional payload e.g., therapeutic agent.
  • the red blood cells are frozen by immersing the tube containing the cells and receiver and optional payload into liquid nitrogen.
  • the cells may be frozen by placing the tube in a freezer at ⁇ 20° C. or ⁇ 80° C.
  • the cells are then thawed in, e.g., a 23° C. water bath and the cycle repeated if necessary to increase loading. Any suitable freeze-thaw method may be used to generate the synthetic membrane-receiver complexes described herein.
  • the receiver and optional payload may be loaded into a cell, e.g., an erythroid cell or platelet in a solubilized form, e.g., solubilized in an appropriate buffer prior to loading into erythroid cells or platelets.
  • a cell e.g., an erythroid cell or platelet
  • solubilized form e.g., solubilized in an appropriate buffer prior to loading into erythroid cells or platelets.
  • the receiver and optional payload may be loaded into a cell, e.g., an erythroid cell or platelet in a particulate form as a solid microparticulate (See, e.g., U.S. Patent Applications 2005/0276861 A1 and U.S. 2006/0270030 A1).
  • the receiver or payload may be poorly water-soluble with a solubility of less than 1-10 mg/ml.
  • Microparticles of poorly water-soluble receivers or payloads can be made of less than 10 ⁇ m using a variety of techniques such as, for example, energy addition techniques such as milling (e.g., pearl milling, ball milling, hammer milling, fluid energy milling, jet milling), wet grinding, cavitation or shearing with a microfluidizer, and sonication; precipitation techniques such as, for example, microprecipitation, emulsion precipitation, solvent-antisolvent precipitation, phase inversion precipitation, pH shift precipitation, infusion precipitation, temperature shift precipitation, solvent evaporation precipitation, reaction precipitation, compressed fluid precipitation, protein microsphere precipitation; and other techniques such as spraying into cryogenic fluids (See, e.g., U.S.
  • energy addition techniques such as milling (e.g., pearl milling, ball milling, hammer milling, fluid energy milling, jet milling), wet grinding, cavitation or shearing with a microfluid
  • Water soluble receivers or payloads may also be used to form solid microparticles in the presence of various polymers such as, for example, polylactate-polyglycolate copolymer (PLGA), polycyanoacrylate, albumin, and/or starch (See, e.g., U.S. Patent Application 2005/0276861 A1).
  • PLGA polylactate-polyglycolate copolymer
  • a water soluble receivers or payloads may be encapsulated in a vesicle to form a microparticle.
  • the microparticles composed of the receiver and optional payload e.g., therapeutic agent
  • a cell such as an erythroid cell or platelet using the methods described herein.
  • synthetic membrane-receiver complexes are generated from erythrocytes.
  • erythrocytes may be loaded with a receiver polypeptide or mRNA encoding a receiver polypeptide by controlled cell injury.
  • the cell injury can be caused by, for example, pressure induced by mechanical strain or shear forces, subjecting the cell to deformation, constriction, rapid stretching, rapid compression, or pulse of high shear rate.
  • the controlled cell injury leads to uptake of material, e.g., a receiver and optionally a payload into the cytoplasm of the cell from the surrounding cell medium. Any suitable controlled injury method may be used to generate the synthetic membrane-receiver complexes described herein.
  • controlled cell injury based on controlled cell deformation (e.g., mechanical deformation of the cell as it passes through the constriction) leads to uptake of material, e.g., a receiver and optionally a payload by diffusion rather than endocytosis.
  • material e.g., a receiver and optionally a payload by diffusion rather than endocytosis.
  • the material e.g., a receiver and optionally a payload is present in the cytoplasm rather than in endosomes following cellular uptake upon the controlled injury thereby making the material readily available to the cell.
  • Controlled cell injury e.g., by controlled deformation, preserves cell viability (e.g., greater than 50%, 70%, or greater than 90%).
  • controlled cell injury e.g., by controlled deformation, preserves the state of cellular differentiation and activity.
  • a combination treatment is used, e.g., controlled injury by deformation followed by or preceded by, e.g., electroporation or another cell membrane
  • Mechanical deformation methods are particularly suitable for cells that do not tolerate other membrane permeability increasing methods well, e.g., show decreased viability or a different state of differentiation after performing such methods.
  • Mechanical deformation methods are also suitable for material, e.g., a receiver and optionally a payload that does not tolerate other membrane permeability increasing methods well.
  • the receiver or payload may not be sufficiently introduced into the cell using alternative methods, e.g., because of e.g., charge, hydrophobicity, or size of the payload.
  • a population of reticulocytes is provided that has been subjected to controlled cell injury by controlled deformation to introduce a receiver, thereby generating a synthetic membrane-receiver complex.
  • the cells can, e.g., be compressed and deformed by passage through a micro-channel having a diameter less than that of an individual reticulocyte, thereby causing perturbations in the cell membrane such that the membrane becomes porous.
  • Cells are moved, e.g., pushed, through the channels or conduits by application of pressure.
  • the compression and deformation occurs in a delivery medium comprising, e.g., receiver polypeptide or oligonucleotide (e.g., DNA, RNA, such as mRNA) and optionally a payload.
  • a delivery medium comprising, e.g., receiver polypeptide or oligonucleotide (e.g., DNA, RNA, such as mRNA) and optionally a payload.
  • the delivery medium may comprise a receiver including but not limited to those listed in table 7 or coding mRNA thereof.
  • the reticulocyte takes up and retains the exogenous material.
  • the cells are optionally incubated in a delivery medium that contains the material, e.g., a receiver and optionally a payload.
  • the cells may be maintained in the delivery medium for a few minutes to recover, e.g., to close the injury caused by passing through the constriction. This may occur at room temperature.
  • Controlled cell injury as used herein includes: i) virus-mediated transfection (e.g., Herpes simplex virus, Adeno virus, Adeno-associated virus, Vaccinia virus, or Sindbis virus), ii) chemically-mediated transfection, e.g., cationic polymer, calcium phosphate, cationic lipid, polymers, and nanoparticles, such as cyclodextrin, liposomes, cationic liposomes, DEAE-dextran, polyethyleneimine, dendrimer, polybrene, calcium phosphate, lipofectin, DOTAP, lipofectamine, CTAB/DOPE, DOTMA; and iii) physically-mediated transfection, including direct injection, biolistic particle delivery, electroporation, laser-irradiation, sonoporation, magnetic nanoparticles, and controlled deformation (e.g., cell squeezing), as exemplified by micro-needle, nano-needle,
  • RNA e.g., mRNA encoding a receiver polypeptide
  • a payload e.g., a therapeutic agent
  • Polypeptide receivers can be detected on the synthetic membrane-receiver complex.
  • the presence of the receiver polypeptide can be validated and quantified using standard molecular biology methods, e.g., Western blotting or FACS analysis.
  • Receiver polypeptides present in the intracellular environment may be quantified upon cell lysis or using fluorescent detection.
  • a population of erythroid cells is loaded with adenosine deaminase (ADA) using the Pro-Ject protein transfection reagent kit to generate a synthetic membrane-ADA receiver complex.
  • the population of synthetic membrane-ADA receiver complexes is then characterized for active enzyme loading using LCMS to quantify adenosine and inosine.
  • the population of erythroid cells is incubated in a solution of 10 mM, 100 mM, 500 mM chlorpromazine and 0.01, 0.1, 1.0, 10, 100 mg/ml of adenosine deaminase (ADA).
  • the population of synthetic membrane-ADA receiver complexes are then washed and fluorescent imaging is used to quantify ADA loading.
  • a population of erythrocytes is incubated in a hypotonic salt solution containing a concentration of 0.01, 0.1, 1.0, 10 mg/ml of asparaginase to generate a synthetic membrane-asparaginase receiver complex.
  • the cell population is incubated for 1 hr and then resealed by incubation in a hypertonic solution for 10 min.
  • the population of synthetic membrane-asparaginase receiver complexes is then incubated in an asparagine solution for 1 hr and the asparagine and aspartate concentrations are quantified using LCMS.
  • a population of erythrocytes is incubated in a PBS solution containing a concentration of 0.01, 0.1, 1.0, 10 mg/ml of thymidine phosphorylase that has been fused via both the C and N termini to one or more cell penetrating peptides, including; Penetratin, Antenapedia, TAT, SynB1, SynB3, PTD-4, PTD-5, FHV Coat-(35-49), BMV Gag-(7-25), HTLV-II Rex-(4-16), D-TAT, R9-Tat, Transportan, MAP, SBP, FBP, MPG ac, MPG(NLS), Pep-1, Pep-2, polyarginines, polylysines, (RAca)6R, (RAbu)6R, (RG)6R, (RM)6R, R10, (RA)6R, R7. Following incubation, synthetic membrane
  • Cells may be loaded using a microfluidic device that transiently porates the cells, allowing a payload to enter when the cells are pressured through the system.
  • a population of erythrocytes is pressured through a system of microfluidic channels in a buffer solution containing 0.01, 0.1, 1.0, 10 mg/ml of phenylalanine ammonia hydroxylase.
  • the cell suspension is then characterized for enzymatic activity using LCMS to quantify phenylalanine and trans-cinnamic acid.
  • a synthetic cell membrane-receiver complexes are incubated in a hypotonic solution containing 1 mM of adenosine deaminase for 1 hr.
  • the synthetic membrane-receiver complexes are then transferred to an isotonic solution and allowed to equilibrate and seal in the soluble protein.
  • Synthetic membrane-receiver complexes may optionally be loaded with payloads such as peptides, proteins, DNA, RNA, siRNA, and other macromolecules and small therapeutic molecules.
  • the payload is transferred to a cell, e.g., an erythroid cell or platelet by applying controlled injury to the cell for a predetermined amount of time in order to cause perturbations in the cell membrane such that the payload can be delivered to the inside of the cell (e.g., cytoplasm).
  • the payload may be a therapeutic agent selected from a variety of known small molecule pharmaceuticals.
  • the payload may be may be a therapeutic agent selected from a variety of macromolecules, such as, e.g., an inactivating peptide nuclei acid (PNA), an RNA or DNA oligonucleotide aptamer, an interfering RNA (iRNA), a peptide, or a protein.
  • PNA inactivating peptide nuclei acid
  • iRNA interfering RNA
  • the synthetic membrane-receiver complex is generated from a reticulocyte.
  • reticulocytes may be loaded with an mRNA encoding for a therapeutic exogenous polypeptide by controlled cell injury.
  • the mRNA may be naked or modified, as desired.
  • mRNA modification that improve mRNA stability and/or decrease immunogenicity include, e.g., ARCA: anti-reverse cap analog (m 2 7.3′-O GP 3 G), GP 3 G (Unmethylated Cap Analog), m 7 GP 3 G (Monomethylated Cap Analog), m 3 2.2.7 GP 3 G (Trimethylated Cap Analog), m5CTP (5′-methyl-cytidine triphosphate), m6ATP (N6-methyl-adenosine-5′-triphosphate), s2UTP (2-thio-uridine triphosphate), and ⁇ (pseudouridine triphosphate).
  • ARCA anti-reverse cap analog
  • GP 3 G Unmethylated Cap Analog
  • m 7 GP 3 G Monomethylated Cap Analog
  • m 3 2.2.7 GP 3 G Trimethylated Cap Analog
  • m5CTP (5′-methyl-cytidine triphosphate)
  • m6ATP N6-methyl-adenosine
  • Synthetic membrane-receiver complexes may comprise two or more payloads, including mixtures, fusions, combinations and conjugates, of atoms, molecules, etc. as disclosed herein, for example including but not limited to, a nucleic acid combined with a polypeptide; two or more polypeptides conjugated to each other; a protein conjugated to a biologically active molecule (which may be a small molecule such as a prodrug); and the like.
  • the pharmaceutical composition comprises one or more therapeutic agents and the synthetic membrane-receiver complex described herein.
  • the synthetic membrane-receiver complexes are co-administered with of one or more separate therapeutic agents, wherein co-administration includes administration of the separate therapeutic agent before, after or concurrent with administration of the synthetic membrane-receiver complex.
  • Suitable payloads include, without limitation, pharmacologically active drugs and genetically active molecules, including antineoplastic agents, anti-inflammatory agents, hormones or hormone antagonists, ion channel modifiers, and neuroactive agents.
  • suitable payloads of therapeutic agents include those described in, “The Pharmacological Basis of Therapeutics,” Goodman and Gilman, McGraw-Hill, New York, N.Y., (1996), Ninth edition, under the sections: Drugs Acting at Synaptic and Neuroeffector Junctional Sites; Drugs Acting on the Central Nervous System; Autacoids: Drug Therapy of Inflammation; Water, Salts and Ions; Drugs Affecting Renal Function and Electrolyte Metabolism; Cardiovascular Drugs; Drugs Affecting Gastrointestinal Function; Drugs Affecting Uterine Motility; Chemotherapy of Parasitic Infections; Chemotherapy of Microbial Diseases; Chemotherapy of Neoplastic Diseases; Drugs Used for Immunosuppression; Drugs Acting
  • the synthetic membrane-receiver complex does not comprise a payload comprising a synthetic triphosphorylated nucleoside analog. In some embodiments, the synthetic membrane-receiver complex does not comprise a payload comprising 2′,3′-dideoxycytidine-5′-triphosphate (ddCTP) and/or 3′-azido-3′-deoxythymidine-5′-triphosphate (AZT-TP).
  • ddCTP 2′,3′-dideoxycytidine-5′-triphosphate
  • AZT-TP 3′-azido-3′-deoxythymidine-5′-triphosphate
  • the synthetic membrane-receiver complex does not comprise a payload comprising a bisphosphonate.
  • the payload is a therapeutic agent, such as a small molecule drug or a large molecule biologic.
  • Large molecule biologics include, but are not limited to, a protein, polypeptide, or peptide, including, but not limited to, a structural protein, an enzyme, a cytokine (such as an interferon and/or an interleukin), a polyclonal or monoclonal antibody, or an effective part thereof, such as an Fv fragment, which antibody or part thereof, may be natural, synthetic or humanized, a peptide hormone, a receptor, or a signaling molecule.
  • a therapeutic agent such as a small molecule drug or a large molecule biologic.
  • Large molecule biologics include, but are not limited to, a protein, polypeptide, or peptide, including, but not limited to, a structural protein, an enzyme, a cytokine (such as an interferon and/or an interleukin), a polyclonal or monoclonal antibody, or an effective part
  • Intrabodies Large molecule biologics are immunoglobulins, antibodies, Fv fragments, etc., that are capable of binding to antigens in an intracellular environment. These types of molecules are known as “intrabodies” or “intracellular antibodies.”
  • An “intracellular antibody” or an “intrabody” includes an antibody that is capable of binding to its target or cognate antigen within the environment of a cell, or in an environment that mimics an environment within the cell. Selection methods for directly identifying such “intrabodies” include the use of an in vivo two-hybrid system for selecting antibodies with the ability to bind to antigens inside mammalian cells. Such methods are described in PCT/GB00/00876, incorporated herein by reference.
  • Large molecule biologics include but is not limited to, at least one of a protein, a polypeptide, a peptide, a nucleic acid, a virus, a virus-like particle, an amino acid, an amino acid analogue, a modified amino acid, a modified amino acid analogue, a steroid, a proteoglycan, a lipid and a carbohydrate or a combination thereof (e.g., chromosomal material comprising both protein and DNA components or a pair or set of effectors, wherein one or more convert another to active form, for example catalytically).
  • chromosomal material comprising both protein and DNA components or a pair or set of effectors, wherein one or more convert another to active form, for example catalytically.
  • a Large molecule biologic may include a nucleic acid, including, but not limited to, an oligonucleotide or modified oligonucleotide, an antisense oligonucleotide or modified antisense oligonucleotide, an aptamer, a cDNA, genomic DNA, an artificial or natural chromosome (e.g., a yeast artificial chromosome) or a part thereof, RNA, including an siRNA, a shRNA, mRNA, tRNA, rRNA or a ribozyme, or a peptide nucleic acid (PNA); a virus or virus-like particles; a nucleotide or ribonucleotide or synthetic analogue thereof, which may be modified or unmodified.
  • a nucleic acid including, but not limited to, an oligonucleotide or modified oligonucleotide, an antisense oligonucleotide or modified antisense oligon
  • the large molecule biologic can also be an amino acid or analogue thereof, which may be modified or unmodified or a non-peptide (e.g., steroid) hormone; a proteoglycan; a lipid; or a carbohydrate. If the large molecule biologic is a polypeptide, it can be loaded directly into, e.g., an erythroid cell or a platelet according to the methods described herein. Alternatively, an exogenous nucleic acid encoding a polypeptide, which sequence is operatively linked to transcriptional and translational regulatory elements active in a cell at a target site, may be loaded.
  • a non-peptide e.g., steroid
  • a non-peptide e.g., steroid
  • a non-peptide e.g., steroid
  • a non-peptide e.g., steroid
  • a non-peptide e.g., steroid
  • a non-peptide e
  • Small molecules including inorganic and organic chemicals, may also be used as payloads of the synthetic membrane-receiver complexes described herein.
  • the small molecule is a pharmaceutically active agent.
  • pharmaceutically active agents include, but are not limited to, antibiotics, anti-inflammatory drugs, angiogenic or vasoactive agents, growth factors and chemotherapeutic (anti-neoplastic) agents (e.g., tumour suppressers).
  • the synthetic membrane-receiver complex further comprises a receiver such as an activating polypeptide which converts the inactive prodrug to active drug form.
  • activating receiver polypeptides include, but are not limited to, viral thymidine kinase (encoded by Genbank Accession No. J02224), carboxypeptidase A (encoded by Genbank Accession No. M27717), ⁇ -galactosidase (encoded by Genbank Accession No. M13571), ⁇ -gluucuronidase (encoded by Genbank Accession No.
  • alkaline phosphatase encoded by Genbank Accession No. J03252 J03512
  • cytochrome P-450 encoded by Genbank Accession No. D00003 N00003
  • plasmin carboxypeptidase G2
  • cytosine deaminase glucose oxidase, xanthine oxidase, ⁇ -glucosidase, azoreductase, t-gutamyl transferase, ⁇ -lactamase, and penicillin amidase.
  • Either the receiver polypeptide or the exogenous gene encoding it may be loaded into, e.g., an erythroid cell or platelet, to generate a synthetic membrane-receiver complex.
  • Both the prodrug and the activating receiver polypeptide may be encoded by genes on the same exogenous nucleic acid.
  • either the prodrug or the activating receiver polypeptide of the prodrug may be transgenically expressed in a synthetic membrane-receiver complex.
  • the synthetic membrane-receiver complexes may also be labeled with one or more positive markers that can be used to monitor over time the number or concentration of synthetic membrane-receiver complexes in the blood circulation of an individual.
  • the overall number of synthetic membrane-receiver complexes will decay over time following initial transfusion.
  • the signal from one or more positive markers are correlated with that of an activated molecular marker, generating a proportionality of signal that is independent of the number of synthetic membrane-receiver complexes remaining in the circulation.
  • Suitable fluorescent compounds include those that are approved by the Food & Drug Administration for human use including but not limited to fluorescein, indocyanin green, and rhodamine B.
  • synthetic membrane-receiver complexes may be non-specifically labeled with fluorescein isothiocyanate (FITC; Bratosin et al., Cytometry 46:351-356 (2001)).
  • FITC fluorescein isothiocyanate
  • PMSF phenylmethysulfonyl fluoride
  • erythroid cells or platelets may be labeled with PKH26 Red (See, e.g., Bratosin, et al., (1997) Cytometry 30:269-274).
  • Erythroid cells or platelets (1-3 ⁇ 10 7 cells) are suspended in 1 ml of diluent and rapidly added to 1 ml or 2 ⁇ M PKH26 dissolved in the same diluent. The mixture is mixed by gentle pipetting and incubated at 25° C. for 2-5 min with constant stirring.
  • the labeling may be stopped by adding an equal volume of human serum or compatible protein solution (e.g., 1% bovine serum albumin). After an additional minute, an equal volume of cell culture medium is added and the cells are isolated by centrifugation at 2000 ⁇ g for 5 min. Cells are washed three times by repeated suspension in cell culture medium and centrifugation. PHK26-labeled synthetic membrane-receiver complexes may be monitored with a maximum excitation wavelength of 551 nm and a maximum emission wavelength of 567 nm.
  • human serum or compatible protein solution e.g., 1% bovine serum albumin
  • Synthetic membrane-receiver complexes may be tracked in vivo using VivoTag 680 (VT680; VisEn Medical, Woburn, Mass., USA), a near-infrared fluorochrome with a peak excitation wavelength of 670 ⁇ 5 nm and a peak emission wavelength of 688 ⁇ 5 nm.
  • VT680 also contains an amine reactive NHS ester which enables it to cross-link with proteins and peptides.
  • the surface of cells e.g., erythroid cells or platelets may be labeled with VT680 (See, e.g., Swirski, et al., (2007) PloS ONE 10:e1075).
  • VT680 4 ⁇ 10 6 cells/ml are incubated with VT680 diluted in complete culture medium at a final concentration of 0.3 to 300 ⁇ g/ml for 30 min at 37° C. The cells are washed twice with complete culture medium after labeling. Cells may be non-specifically labeled based on proteins expressed on the surface of the synthetic membrane-receiver complex. Alternatively, a specific protein, such as a receiver may be labeled with VT680. In some embodiments, a protein or peptide may be directly labeled with VT680 ex vivo and subsequently either attached to the surface of the cell or incorporated into the interior of the cell using methods described herein.
  • In vivo monitoring may, for example, be performed using the dorsal skin fold.
  • Laser scanning microscopy may be performed using, for example, an Olympus IV 100 in which VT680 is excited with a red laser diode of 637 nm and detected with a 660/LP filter.
  • multiphoton microscopy may be performed using, for example, a BioRad Radiance 2100 MP centered around an Olympus BX51 equipped with a 20 ⁇ /0.95 NA objective lens and a pulsed Ti:Sapphire laser tuned to 820 nm. The latter wavelength is chosen because VT680 has a peak in its two-photon cross-section at 820 nm.
  • a synthetic membrane-receiver complex may be labeled with other red and/or near-infrared dyes including, for example, cyanine dyes such as Cy5, Cy5.5, and Cy7 (Amersham Biosciences, Piscataway, N.J., USA) and/or a variety of Alexa Fluor dyes including Alexa Fluor 633, Alexa Fluor 635, Alexa Fluor 647, Alexa Fluor 660, Alexa Fluor 680, Alexa Fluor 700 and Alexa Fluor 750 (Molecular Probes-Invitrogen, Carlsbad, Calif., USA).
  • cyanine dyes such as Cy5, Cy5.5, and Cy7
  • Alexa Fluor dyes including Alexa Fluor 633, Alexa Fluor 635, Alexa Fluor 647, Alexa Fluor 660, Alexa Fluor 680, Alexa Fluor 700 and Alexa Fluor 750 (Molecular Probes-Invitrogen, Carlsbad, Calif., USA).
  • fluorophores include IRD41 and IRD700 (LI-COR, Lincoln, Nebr., USA), NIR-1 and 1C5-OSu (Dejindo, Kumamotot, Japan), LaJolla Blue (Diatron, Miami, Fla., USA), FAR-Blue, FAR-Green One, and FAR-Green Two (Innosense, Giacosa, Italy), ADS 790-NS and ADS 821-NS (American Dye Source, Montreal, Calif.). Quantum dots (Qdots) of various emission/excitation properties may also be used for labeling synthetic membrane-receiver complexes (See, e.g., Jaiswal et al., Nature Biotech. 21:47-51 (2003)).
  • fluorophores are available from commercial sources either attached to primary or secondary antibodies or as amine-reactive succinimidyl or monosuccinimidyl esters, for example, ready for conjugation to a protein or proteins either on the surface or inside the synthetic membrane-receiver complex.
  • Magnetic nanoparticles may be used to track synthetic membrane-receiver complexes in vivo using high resolution MRI (Montet-Abou et al., Molecular Imaging 4:165-171 (2005)). Magnetic particles may be internalized by several mechanisms. Magnetic particles may be taken up by a cell, e.g., an erythroid cell or a platelet through fluid-phase pinocytosis or phagocytosis. Alternatively, the magnetic particles may be modified to contain a surface agent such as, for example, a membrane translocating HIV TAT peptide which promotes internalization.
  • a surface agent such as, for example, a membrane translocating HIV TAT peptide which promotes internalization.
  • a magnetic nanoparticle such as, for example, Feridex IV®, an FDA approved magnetic resonance contrast reagent
  • a transfection agent such as, for example, protamine sulfate (PRO), polylysine (PLL), and lipofectamine (LFA).
  • PRO protamine sulfate
  • PLA polylysine
  • LFA lipofectamine
  • the synthetic membrane-receiver polypeptide complexes are generated comprising contacting an erythroid cell with a receiver, such as a polypeptide.
  • the receiver polypeptide is encoded by an exogenous nucleic acid and is expressed by the erythroid cell.
  • a naturally occurring erythroid cell does not comprise the receiver.
  • a naturally occurring erythroid cell does not express an endogenous polypeptide that is structurally and functionally the same as the receiver polypeptide.
  • the erythroid cell comprises a receiver that is over-expressed.
  • the receiver is present in substantially higher copy numbers than it would be if it were endogenously expressed by a naturally occurring erythroid cell.
  • the synthetic membrane-receiver polypeptide complexes are generated by differentiating and maturing the erythroid cells in vitro or in vivo after contacting the cells with a receiver. It is known in the art that erythrocytes undergo a complex process of maturation as they differentiate from precursor cells. The maturation process includes a substantial cytoskeleton and membrane rearrangement and degradation or expulsion of non-essential polypeptides, see e.g., Liu J et al. (2010) Blood 115(10):2021-2027; and Lodish H F et al. (1975) Developmental Biology 47(1):59).
  • the synthetic membrane-receiver polypeptide complexes generated from erythroid cells retain their receivers during the maturation process, in vitro or in vivo and the receivers are not lost.
  • the synthetic membrane-receiver polypeptide complexes generated from erythroid cells retain their receivers after maturation.
  • fully matured synthetic membrane-receiver polypeptide complexes generated from erythroid cells retain their receiver.
  • the receiver may be retained in vitro, e.g., in culture and/or may be retained in vivo, e.g., after administration to the circulatory system of the subject.
  • the receiver may be retained by the synthetic membrane-receiver polypeptide complexes for the life of the complex in circulation.
  • culturing of eythroid cells comprising a receiver provides a method of producing a substantially more homogeneous and/or substantially more scalable population of therapeutic synthetic membrane-receiver complexes than is achievable by methods relying upon isolation and modification of non-cultured erythrocytes.
  • a great need for human erythroid cell-based treatment and preventive methods and recognition for its value in the art no systems derived from modified cultured cells have previously been generated or shown to retain receiver activity in circulation, and the art suggested that such systems would not be achievable.
  • cultured human erythrocytes have been experimentally administered to a human subject previously they were unmodified (Giarratana et al., Blood 2011, 118:5071).
  • synthetic membrane-receiver polypeptide complexes comprising a receiver polypeptide capable of interacting with a target.
  • synthetic membrane-receiver complexes comprising a non-polypeptide receiver capable of interacting with a target.
  • the synthetic membrane-receiver complexes may be administered to a subject in need thereof to modulate the amount or concentration of a target residing in the circulatory system of the subject.
  • a suitable receiver may be chosen to interact with a specific target.
  • Suitable targets include entities that are associated with a specific disease, disorder, or condition. However, targets may also be chosen independent of a specific disease, disorder, or condition.
  • the target is an antibody or antibody-like molecule, for example an autoimmune or a self-antibody, or a foreign antibody, or a therapeutic antibody, including but not limited to, e.g., an antibody against beta-2 glycoprotein 1, an antibody against I/i antigen, an antibody against the NC1 domain of collagen a3(IV), an antibody against platelet glycoprotein, an antibody against phospholipase A2 receptor, an antibody against erythrocyte glycophorin A, B, or C, or an antibody against erythrocyte Rh antigen.
  • an antibody against beta-2 glycoprotein 1 an antibody against I/i antigen
  • an antibody against platelet glycoprotein an antibody against phospholipase A2 receptor
  • an antibody against erythrocyte glycophorin A, B, or C or an antibody against erythrocyte Rh antigen.
  • the target is a molecule of the complement cascade, for example C1, C1r, C1s, C1q, C2, C2a, C2b, C3, C3a, C3b, C4, C4b, C4a, C3bBb, C3bBb3b, C4b2b, C4b2b3b, C5, CSa, CSb, C6, C7, C8, C9, poly-C9, membrane attack complex.
  • Factor B Factor D, Properdin, C3, C3a, C3b, iC3b, C3c, C3dg, C3dk, C3e, Bb, Factor I, C1q, C1r, C1s, C4, C4a, C4b, C2, C4 bp, Mannose-Binding Lectin (MBL), MBL-Associated Serine Protease 1 (MASP1), MBL-Associated Serine Protease 2 (MASP2), C5, CSa, C6, C7, C8, C9, CR1, CR2, CR3, CR4, C3aR, C3eR, Decay-accelerating factor (DAF), Membrane cofactor protein (MCP), CD59, C3 Beta chain Receptor, C1 inhibitor, C4 binding protein, Factor I, Factor H.
  • MBL Mannose-Binding Lectin
  • MASP1 MBL-Associated Serine Protease 1
  • MASP2 MBL-Associated Serine
  • the target is an immune complex, for example an IgG immune complex, an IgA immune complex, an IgM immune complex.
  • the target is an amyloid placque, for example a placque comprised of beta amyloid, IAPP (Amylin), alpha-synuclein, PrPSc, huntingtin, calcitonin, atrial natriuretic factor, apolipoprotein AI, serum amyloid A, medin, prolactin, transthyretin, lysozyme, beta 2 microglobulin, gelsolin, keratoepithelin, cystatin, immunoglobulin light chain AL, S-IBM.
  • beta amyloid for example a placque comprised of beta amyloid, IAPP (Amylin), alpha-synuclein, PrPSc, huntingtin, calcitonin, atrial natriuretic factor, apolipoprotein AI, serum amyloid A, medin, prolactin, transthyretin, lysozyme, beta 2 microglobulin, gelsolin, keratoepithelin
  • the target is a bacterium, for example Enterococcus, Streptococcus , or Mycobacteria, Rickettsia, Mycoplasma, Neisseria meningitides, Neisseria gonorrheoeae, Legionella, Vibrio cholerae, Streptococci, Staphylococcus aureus, Staphylococcus epidermidis, Pseudomonas aeruginosa, Corynobacteria diphtheriae, Clostridium spp., enterotoxigenic Eschericia coli , and Bacillus anthracis .
  • Enterococcus Streptococcus
  • Mycobacteria Rickettsia
  • Mycoplasma Neisseria meningitides
  • Neisseria gonorrheoeae Legionella
  • Vibrio cholerae Streptococci
  • Staphylococcus aureus Staphylococc
  • pathogens for which bacteremia has been reported at some level include the following: Rickettsia, Bartonella henselae, Bartonella quintana, Coxiella burnetii, chlamydia, Mycobacterium leprae, Salmonella; shigella; Yersinia enterocolitica; Yersinia pseudotuberculosis; Legionella pneumophila; Mycobacterium tuberculosis; Listeria monocytogenes; Mycoplasma spp.; Pseudomonas fluorescens; Vibrio cholerae; Haemophilus influenzae; Bacillus anthracis; Treponema pallidum; Leptospira; Borrelia; Corynebacterium diphtheriae; Francisella; Brucella melitensis; Campylobacter jejuni; Enterobacter; Proteus mirabilis; Proteus ; and Klebsiella
  • the target is a virus, including but limited to, those whose infection involves injection of genetic materials into host cells upon binding to cell surface receptors, viruses whose infection is mediated by cell surface receptors.
  • viruses can be selected from Paramyxoviridae (e.g., pneumovirus, morbillivirus, metapneumovirus, respirovirus or rubulavirus), Adenoviridae (e.g., adenovirus), Arenaviridae (e.g., arenavirus such as lymphocytic choriomeningitis virus), Arteriviridae (e.g., porcine respiratory and reproductive syndrome virus or equine arteritis virus), Bunyaviridae (e.g., phlebovirus or hantavirus), Caliciviridae (e.g., Norwalk virus), Coronaviridae (e.g., coronavirus or torovirus), Filoviridae (e.g., Ebol
  • the target is a parasite, including but not limited to, for example, intestinal or blood-borne parasites, protozoa, trypanosomes; haemoprotozoa and parasites capable of causing malaria; enteric and systemic cestodes including taeniid cestodes; enteric coccidians; enteric flagellate protozoa; filarial nematodes; gastrointestinal and systemic nematodes and hookworms.
  • a parasite including but not limited to, for example, intestinal or blood-borne parasites, protozoa, trypanosomes; haemoprotozoa and parasites capable of causing malaria; enteric and systemic cestodes including taeniid cestodes; enteric coccidians; enteric flagellate protozoa; filarial nematodes; gastrointestinal and systemic nematodes and hookworms.
  • the target is a fungus, including but not limited to, for example, Candida albicans, Candida glabrata, Aspergillus, T. glabrata, Candida tropicalis, C. krusei , and C. parapsilosis.
  • the target is a bacterial toxin, including but not limited to, for example, AB toxin, alpha toxin, anthrax toxin, bacteriocin, botunlinum toxin, cholesterol-dependent cytolysin, Clostridium botulinum C3 toxin, Clostridium difficile toxin A, Clostridium difficile toxin B, Clostridium enterotoxin, Clostridium perfringens alpha toxin, Clostridium perfringens beta toxin, Cord factor, Cry1Ac, Cryptophycin, Delta endotoxin, Diphtheria toxin, Enterotoxin type B, erythrogenic toxin, exfoliatin, haemolysin E, heat-labile enterotoxin, heat-stable enterotoxin, hemolysin, leukocidin, lipopolysaccharide, Listeriolysin O, microcin, Panton-Valen
  • the target is a prion protein, including but not limited to, for example, PRP, PRPc, PRPsc, PRPres.
  • the target is a cytokine or a chemokine or a growth factor, including but not limited to, for example, acylation stimulating protein, adipokine, albinterferon, CCL1, CCL11, CCL12, CCL13, CCL14, CCL15, CCL16, CCL17, CCL18, CCL19, CCL2, CCL20, CCL21, CCL22, CCL23, CCL24, CCL25, CCL26, CCL27, CCL28, CCL3, CCL5, CCL6, CCL7, CCL8, CCL9, colony-stimulating factor, CX3CL1, CX3CR1, CXCL1, CXCL10, CXCL11, CXCL13, CXCL14, CXCL15, CXCL16, CXCL17, CXCL2, CXCL3, CXCL5, CXCL6, CXCL7, CXCL9, erythropoietin, Gc-MAF, granulocyte colon
  • the target is a small molecule, for example a chemical, an amino acid, an atom, an element, an organic acid, ⁇ 2000 Da, ⁇ 1000 Da, ⁇ 500 Da, including but not limited to, for example, iron, copper, calcium, potassium, ethanol, methanol, glycine, alanine, valine, leucine, isoleucine, serine, cysteine, selenocysteine, threonine, methionine, proline, phenylalanine, tyrosine, tryptophan, histidine, lysine, arginine, aspartate, glutamate, asparagine, glutamine.
  • a small molecule for example a chemical, an amino acid, an atom, an element, an organic acid, ⁇ 2000 Da, ⁇ 1000 Da, ⁇ 500 Da, including but not limited to, for example, iron, copper, calcium, potassium, ethanol, methanol, glycine, alanine, valine, leucine, is
  • the target is a lipid, lipid complex, proteolipid complex, or cholesterol, including but not limited to for example, LDL, VLDL, HDL, HDL2B, triglycerides, LP(a), cholesterol.
  • the target is a mammalian cell, including but not limited to, for example, a human cell, a circulating cell, an immune cell, a neutrophil, an eosinophil, a basophil, a lymphocyte, a monocyte, a B cell, a T cell, a CD4+ T cell, a CD8+ T cell, a gamma-delta T cell, a regulatory T cell, a natural killer cell, a natural killer T cell, a macrophage, a Kupffer cell, a dendritic cell, a cancer cell, a cancer stem cell, a circulating tumor cell, a cancer cell from one of the following cancers including, but not limited to, ACUTE lymphoblastic leukaemia (ALL), ACUTE myeloid leukaemia (AML), anal cancer, bile duct cancer, bladder cancer, bone cancer, bowel cancer, brain tumours, breast cancer, cancer of unknown primary, cancer spread to bone, cancer spread to brain, cancer spread to liver, cancer spread to
  • Synthetic membrane-receiver complexes can be generated by any method described herein.
  • the steps comprise contacting isolated optionally cultured cells derived from hematopoietic stem cells with a receiver.
  • Hematopoietic stem cells give rise to all of the blood cell types found in mammalian blood including myeloid (monocytes and macrophages, neutorphils, basophils, eosinophils, erythrocytes, megakaryocytes/platelets, dendritic cells) and lymphoid lineages (T-cells, B-cells, NK-cells).
  • Hematopoietic stem cells may be isolated from the bone marrow of adult bones including, for example, femur, hip, rib, or sternum bones. Cells may be obtained directly from the hip, for example, by removal of cells from the bone marrow using aspiration with a needle and syringe. Alternatively, hematopoietic stem cells may be isolated from normal peripheral blood following pre-treatment with cytokines such as, for example, granulocyte colony stimulating factor (G-CSF). G-CSF mobilizes the release of cells from the bone marrow compartment into the peripheral circulation. Other sources of hematopoietic stem cells include umbilical cord blood and placenta.
  • cytokines such as, for example, granulocyte colony stimulating factor (G-CSF).
  • G-CSF granulocyte colony stimulating factor
  • Other sources of hematopoietic stem cells include umbilical cord blood and placenta.
  • the synthetic membrane-receiver complex is generated from megakaryocytes or platelets. In some embodiments, the synthetic membrane-receiver complex is generated from an erythroid cell, such as, e.g. an erythrocyte or a reticulocyte. In some embodiments, the synthetic membrane-receiver complex is not generated from a neutrophil, an eosinophil, or a basophil. In some embodiments, the synthetic membrane-receiver complex is not generated from a monocyte or a macrophage.
  • the synthetic membrane-receiver complex is not generated from a CD34 + Thy-1 + hematopoietic stem cell or cell populations enriched in CD34 + Lin ⁇ or CD34 + Thy-1 + Lin ⁇ cells.
  • the synthetic membrane-receiver complex is not generated from or does not comprise an autologous CD34+ cell.
  • Isolated hematopoietic stem cells may be cultured, expanded and differentiated ex vivo to provide a variety of source material to generate synthetic membrane-receiver complexes.
  • hematopoietic stem cells isolated from bone marrow, cytokine-stimulated peripheral blood or umbilical cord blood may be expanded and differentiated ex vivo into mature erythrocytes (Giarratana et al., Nature Biotech. 23:69-74 (2005); U.S. Patent Application 2007/0218552).
  • CD34+ cells are isolated from bone marrow or peripheral or cord blood using, for example, magnetic microbead selection and Mini-MACS columns (Miltenyi Biotech).
  • the cells are subsequently cultured in modified serum-free medium supplemented with 1% bovine serum albumin (BSA), 120 mg/ml iron-saturated human transferrin, 900 ng/ml ferrous sulfate, 90 ng/ml ferric nitrate and 10 mg/ml insulin and maintained at 37° C. in 5% carbon dioxide in air.
  • BSA bovine serum albumin
  • the cells may be expanded in the medium described herein in the presence of multiple growth factors including, for example, hydrocortisone, stem cell factor, IL-3, and erythropoietin.
  • the cells may optionally be co-cultured, for example, on an adherent stromal layer in the presence of erythropoietin.
  • the cells may be cultured on an adherent stromal layer in culture medium in the absence of exogenous factors.
  • the adherent stromal layer may be murine MS-5 stromal cells, for example.
  • the adherent stromal layer may be mesenchymal stromal cells derived from adult bone marrow.
  • the adherent stromal cells may be maintained in RPMI supplemented with 10% fetal calf serum, for example.
  • the erythroid precursor cells and cell populations derived therefrom are not co-cultured with non-erythroid cells, e.g., with an adherent stromal layer, i.e. they are cultured in the absence of non-erythroid cells.
  • erythroid cells comprising a receiver are cultured in the absence of non-erythroid cells and are differentiated so that greater than 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or greater than 98% of erythroid cells are enucleated and the population of enucleated cells is obtained without an enrichment step, such as gravitational separation, magnetic or fluorescent sorting, irradiation, poisoning of nucleated cells, and the like to select for enucleated cells.
  • an enrichment step such as gravitational separation, magnetic or fluorescent sorting, irradiation, poisoning of nucleated cells, and the like to select for enucleated cells.
  • CD34+ hematopoietic stem cells may be expanded in vitro in the absence of the adherent stromal cell layer in medium containing various factors including, for example, Flt3 ligand, stem cell factor, thrombopoietin, erythropoietin, and insulin growth factor.
  • the resulting erythroid precursor cells may be characterized by the surface expression of CD36 and GPA, and may be transfused into a subject where terminal differentiation to mature erythrocytes is allowed to occur.
  • the erythroid cell population comprises a plurality of enucleated functional erythroid cells that comprise a receiver polypeptide that is retained during enucleation.
  • the resulting isolated enucleated functional erythroid cell comprising a receiver polypeptide exhibits substantially the same osmotic membrane fragility as a corresponding isolated, unmodified, uncultured erythroid cell.
  • the erythroid cell population comprises a plurality of erythrocyte precursor cells in substantially the same stage of differentiation and/or cell cycle stage, wherein the precursor cells comprise an exogenous nucleic acid encoding a receiver.
  • the majority of erythrocyte precursor cells that comprise an exogenous nucleic acid encoding a receiver are capable of differentiating into mature functional erythrocytes that retain the receiver without retaining the exogenous nucleic acid.
  • the primary cells may be collected through venipuncture, capillary puncture, or arterial puncture. From the collected whole blood erythrocytes, platelets or other cells may then be isolated using one, or a combination of techniques including plasma depletion, density gradient, Hetastarch, PrepaCyte-CB, and centrifugation.
  • generating a synthetic membrane-receiver complex comprises contacting isolated optionally cultured cells that are autologous and/or allogeneic to the subject with a receiver.
  • erythrocytes allogeneic to the subject include one or more of blood type specific erythrocytes or one or more universal donor erythrocytes.
  • synthetic membrane-receiver complexes may be generated through fusion of erythrocytes, e.g., between erythrocytes autologous to the subject and one or more allogeneic erythrocytes, liposomes, and/or artificial vesicles.
  • autologous transfusion of synthetic membrane-receiver complexes includes isolating erythrocytes, reticulocytes or hematopoietic stem cells from a subject, generating a suitable synthetic membrane-receiver complex by contacting the cell with a receiver by methods described herein and administering (e.g., by transfusion) the synthetic membrane-receiver complex into the same subject.
  • allogeneic transfusion of synthetic membrane-receiver complexes includes isolating erythrocytes, reticulocytes or hematopoietic stem cells from a donor, generating a suitable synthetic membrane-receiver complex by contacting the cell with a receiver by methods described herein and administering (e.g., by transfusion) the synthetic membrane-receiver complex into a subject that is different from the donor.
  • allogeneic cells are used for transfusion, care needs to be taken to use a compatible ABO blood group to prevent an acute intravascular hemolytic transfusion reaction which is characterized by complement activation and lysis of incompatible erythrocytes.
  • the ABO blood types are defined based on the presence or absence of the blood type antigens A and B, monosaccharide carbohydrate structures that are found at the termini of oligosaccharide chains associated with glycoproteins and glycolipids on the surface of the erythrocytes (reviewed in Liu et al., Nat. Biotech. 25:454-464 (2007)).
  • Group O erythrocytes lack either of these antigenic monosaccharide structures. Subjects with group A erythrocytes have naturally occurring antibodies to group B erythrocytes whereas subjects with group B erythrocytes have antibodies to group A erythrocytes. Blood group AB subjects have neither antibody and blood group O individuals have both.
  • group O erythrocytes contain neither A nor B antigens, they can be safely transfused into recipients of any ABO blood group, e.g., group A, B, AB, or O recipients.
  • Group O erythrocytes are considered universal and may be used in all blood transfusions.
  • group A erythrocytes may be given to group A and AB recipients
  • group B erythrocytes may be given to group B and AB recipients
  • group AB erythrocytes may only be given to AB recipients.
  • synthetic membrane-receiver complexes are generated by connecting erythrocytes or their precursors with a receiver the sourced erythrocytes or their precursors are matched for compatibility with the recipient.
  • a synthetic membrane-receiver complex comprising a non-group O erythrocyte to a universal blood type. Enzymatic removal of the immunodominant monosaccharides on the surface of group A and group B erythrocytes may be used to generate a population of group O-like synthetic membrane-receiver complexes (See, e.g., Liu et al., Nat. Biotech. 25:454-464 (2007)). Group B synthetic membrane-receiver complexes may be converted using an ⁇ -galactosidase derived from green coffee beans.
  • ⁇ -N-acetylgalactosaminidase and ⁇ -galactosidase enzymatic activities derived from E. meningosepticum bacteria may be used to respectively remove the immunodominant A and B antigens (Liu et al., Nat. Biotech. 25:454-464 (2007)), if present on the synthetic membrane-receiver complexes.
  • packed red blood cells isolated as described herein are incubated in 200 mM glycine (pH 6.8) and 3 mM NaCl in the presence of either ⁇ -N-acetylgalactosaminidase and ⁇ -galactosidase (about 300 ⁇ g/ml packed red blood cells) for 60 min at 26° C. After treatment, the red blood cells are washed by 3-4 rinses in saline with centrifugation and ABO-typed according to standard blood banking techniques.
  • the synthetic membrane-receiver complexes described herein may be generated in the following way.
  • erythroid precursor cells are isolated. These cells may alternatively be autologous to the patient or from substantially universal donor blood.
  • the cells may be ABO type O, rhesus factor Rh r/r, Duffy ⁇ / ⁇ , and large Kell antigen K1 negative.
  • an exogenous nucleic acid encoding the receiver is introduced.
  • the exogenous nucleic acid encoding the receiver can be under the control of an erythroid-specific promoter, such as a GATA-1 promoter (see e.g., Repik et al., Clin Exp Immunol 2005, 140:230).
  • the exogenous nucleic acid encoding the receiver can be introduced in any way known in the art, for example, as plasmid DNA, virus, or mRNA. Nucleic acid introduction can be achieved by a variety of standard methods, e.g., transfection, transduction, or electroporation.
  • the synthetic membrane-receiver complexes described herein may be generated by contacting platelets with a receiver.
  • a receiver Each day an adult human produces 2 ⁇ 10 11 red blood cells, and about one-half as many white cells and platelets. In humans, nearly all blood cell production occurs in the red bone marrow that represents a hierarchical developmental system composed of hematopoietic stem cells, intermediate level progenitors and maturing cells committed to each lineage.
  • megakaryocytes cells committed to platelet production
  • blast cell level of differentiation growing to a size 10 times the diameter of most other bone marrow and blood cells, and containing up to 128 times the normal chromosomal complement
  • these cells give rise to blood platelets.
  • the developing megakaryocyte precursor enters a unique cell cycle characterized by a brief (about 1 h) G1 phase, a typical (7 h) S phase, a very brief ( ⁇ 45 min) G2 phase, followed by the endomitotic phase (an aborted M phase).
  • the cell develops a highly polyploid nucleus, it also develops demarcation membranes necessary for cytoplasmic fragmentation. This event is accompanied by expression of glycoprotein GPIIbIIIa (platelet fibrinogen receptor; Papayannopoulou et al., Exp. Hematol., 24: 660-9, 1996) and GPIb (von Willibrand factor receptor; Kaushansky et al., Nature, 369: 568-571, 1994), the granules that contain ADP, serotonin, -thromboglobulin, and other substances critical for mature platelet function.
  • GPIIbIIIa platelet fibrinogen receptor
  • GPIb von Willibrand factor receptor
  • megakaryocytes are derived from pluripotent marrow stem cells that retain the capacity to extensively self-renew, or to differentiate into all of the elements of the blood. Platelet production is in part regulated by signaling mechanisms induced by interaction between thrombopoietin (TPO) and its cellular receptor TPOR/MPUc-MPL.
  • TPO thrombopoietin
  • Thrombopoietin is a hematopoietic growth factor involved in stimulation of megakaryocytopoiesis and platelet production.
  • TPO is expressed in liver and kidney, and, in response to platelet demand, its expression may be also upregulated in the bone marrow microenvironment (Kato et al., Stem Cells, 16: 322-328, 1998; McCarty et al., Blood, 86:3668-3675, 1995).
  • TPO expression is mostly constitutive, the TPO levels are believed to be regulated by sequestering by platelets (Fielder et al., Blood 87: 2154, 1996).
  • TPO The gene encoding TPO has been cloned and characterized (Kuter et al., Proc. Natl. Acad. Sci. USA, 91:11104-11108, 1994; Bartley et al., Cell, 77:1117-1124, 1994; Kaushansky et al., Nature, 369:568-571, 1994; Wendling et al., Nature, 369:571-574, 1994, and de Sauvage et al., Nature, 369:533-538, 1994).
  • Human TPO (hTPO) cDNA encodes a 353 amino acid-long polypeptide.
  • the full-length hTPO secreted from mammalian cells after cleavage of the signal peptide consists of 332 amino acids. Although the predicted molecular mass of this protein is 38 kD, the molecular masses reported from measurements of material in serum or in culture fluid from recombinant cells vary from 18 to 85 kD (glycosylation, and post-translational proteolytic processing).
  • the cell surface receptor for TPO (TPOR/MPL/c-MPL) is a product of the protooncogene c-mpl, a homologue of v-mpl, an envelope protein of the myeloproliferative leukaemia virus (MPLV) shown to induce a pan-myeloid disorder (Wendling, Virol., 149:242-246, 1986).
  • the human c-mpl gene codes for a protein of 635 aa having a predicted molecular weight of 71 kD (Vigon et al., Proc. Natl. Acad. Sci. USA, 89:5640-44, 1992; Mignotte et al., Genomics, 20: 5-12, 1994).
  • mice rendered null for the expression of either TPO or its receptor manifest a severe thrombocytopenic phenotype (Gurney et al., Science, 265: 1445, 1994; Kaushansky et al., J. Clin. Invest., 96: 1683, 1995; de Sauvage et al., J. Exp. Med., 183: 651, 1996).
  • cytokines e.g., stem cell factor [SCF], IL-1, IL-3, IL-6, IL-11, leukaemia inhibiting factor [LIF], G-CSF, GM-CSF, M-CSF, erythropoietin (EPO), kit ligand, and -interferon
  • SCF stem cell factor
  • IL-1 IL-1
  • IL-3 IL-3
  • IL-6 IL-11
  • LIF leukaemia inhibiting factor
  • G-CSF GM-CSF
  • M-CSF erythropoietin
  • EPO erythropoietin
  • the resulting platelets are small disc-shaped cell fragments which undergo a rapid transformation when they encounter sites of vascular damage. They become more spherical and extrude pseudopodia, their fibrinogen receptors are activated leading to aggregation, and they release their granule contents and eventually they form a plug which is responsible for primary hemostasis (Siess, W., Physiol. Rev. 69: 58-178, 1989). Activation of platelets is also implicated in the pathogenesis of unstable angina, myocardial infarction and stroke (Packham, M. A., Can J. Physiol Pharmacol. 72: 278-284).
  • thrombin thromboxane A2
  • ADP thromboxane A2
  • Collagen binds to several platelet membrane proteins including integrin ⁇ 2 ⁇ 1 leading to platelet activation through the release of TXA 2 and ADP (Shattil, S. J., et al., Curr. Opin. Cell Biol. 6: 695-704, 1994).
  • thrombin In contrast, thrombin, TXA 2 , and ADP, activate G-protein coupled receptors directly and induce platelet aggregation and granule release (Hourani, S. M, and Cusack, N. J., Pharmacol. Rev. 43: 243-298, 1991).
  • the major events involved in platelet activation are believed to be the result of the activation of 3-isoforms of phospholipase C (PLC) leading to the generation of inositol 1,4,5 triphosphate and diacylglycerol.
  • PLC phospholipase C
  • Platelets mainly contain two isoforms, PLC-32 and PLC-33.
  • Platelet receptors which mediate platelet adhesion and aggregation are located on the two major platelet surface glycoprotein complexes. These complexes are the glycoprotein Ib-IX complex which facilitates platelet adhesion by binding von Willebrand factor (vWF), and the glycoprotein IIb-IIIa complex which links platelets into aggregates by binding to fibrinogen.
  • vWF von Willebrand factor
  • vWF von Willebrand factor
  • glycoprotein IIb-IIIa complex which links platelets into aggregates by binding to fibrinogen.
  • Patients with the Bernard-Soulier syndrome, a congenital bleeding disorder show deficient platelet adhesion due to a deficiency in the glycoprotein Ib-IX complex which binds vWF, mild thrombocytopenia, and large lymphocoid platelets.
  • Glycoprotein V is a major ( ⁇ 12,000 molecules/platelet), heavily glycosylated platelet membrane protein (Mr 82,000). Exposure of platelets to thrombin liberates a 69 kDa soluble fragment termed GPVfl.
  • GPV can interact non-covalently with the GPIb-IX complex a complex formed by the non-covalent association of GPIb (consisting of GPIba, a 145 kDa protein, disulfide linked to GPIb ⁇ 3, a 24 kDa protein) with GPIX (a 22 kDa protein).
  • the binding sites for von Willebrand factor and for thrombin on the GPIb-IX complex have been localized on GPIba.
  • thrombin Since thrombin is now known to activate platelets by cleaving the thrombin receptor (Vu et. al., Cell 64:1057-1068 (1990)), a G-protein coupled receptor, it is unknown whether thrombin cleaves GPV incidently as a consequence of thrombin binding to GPIba, or whether this cleavage has a physiological role.
  • GPIB ⁇ , GPIB ⁇ , and GPIX contain one or more homologous 24 amino acid leucine-rich domains. These domains are also found in a large family of leucine-rich glycoproteins (LRG).
  • GPV is a marker for the megakaryocytic cell lineage.
  • a monoclonal antibody specific for GPV does not bind to red cells, leukocytese endothelial cells, or cell lines such as HEL or MEG-01 which are known to express platelet megakaryocyte markers.
  • Mature GPV is composed of 543 amino acids which contain a single transmembrane domain, a short cytoplasmic domain (16 residues) and a large extracellular domain with 8 potential N-glycosylation sites. Analysis of the extracellular domain revealed the presence of 15 tandem Leu-rich repeats of 24 amino acids with homology to GPIba, and identified a cleavage site for thrombin near the C-terminus with homology to the Aa chain of fibrinogen.
  • Sources for generating synthetic membrane-receiver complexes described herein include circulating cells such as erythroid cells.
  • a suitable cell source may be isolated from a subject as described herein from patient-derived hematopoietic or erythroid progenitor cells, derived from immortalized erythroid cell lines, or derived from induced pluripotent stem cells, optionally cultured and differentiated.
  • Methods for generating erythrocytes using cell culture techniques are well known in the art, e.g., Giarratana et al., Blood 2011, 118:5071, Huang et al., Mol Ther 2013, epub ahead of print September 3, or Kurita et al., PLOS One 2013, 8:e59890.
  • Protocols vary according to growth factors, starting cell lines, culture period, and morphological traits by which the resulting cells are characterized. Culture systems have also been established for blood production that may substitute for donor transfusions (Fibach et al. 1989 Blood 73:100). Recently, CD34+ cells were differentiated to the reticulocyte stage, followed by successful transfusion into a human subject (Giarratana et al., Blood 2011, 118:5071).
  • Erythroid cells can be cultured from hematopoietic progenitor cells, including, for example, CD34+ hematopoietic progenitor cells (Giarratana et al., Blood 2011, 118:5071), induced pluripotent stem cells (Kurita et al., PLOS One 2013, 8:e59890), and embryonic stem cells (Hirose et al. 2013 Stem Cell Reports 1:499). Cocktails of growth and differentiation factors that are suitable to expand and differentiate progenitor cells are known in the art.
  • suitable expansion and differentiation factors include, but are not limited to, stem cell factor (SCF), an interleukin (IL) such as IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-11, IL-12, CSF, G-CSF, thrombopoietin (TPO), GM-CSF, erythropoietin (EPO), Flt3, Flt2, PIXY 321, and leukemia inhibitory factor (LIF).
  • SCF stem cell factor
  • IL interleukin
  • Erythroid cells can be cultured from hematopoietic progenitors, such as CD34+ cells, by contacting the progenitor cells with defined factors in a multi-step culture process.
  • erythroid cells can be cultured from hematopoietic progenitors in a three-step process.
  • the first step may comprise contacting the cells in culture with stem cell factor (SCF) at 1-1000 ng/mL, erythropoietin (EPO) at 1-100 U/mL, and interleukin-3 (IL-3) at 0.1-100 ng/mL.
  • SCF stem cell factor
  • EPO erythropoietin
  • IL-3 interleukin-3
  • the first step optionally comprises contacting the cells in culture with a ligand that binds and activates a nuclear hormone receptor, such as e.g., the glucocorticoid receptor, the estrogen receptor, the progesterone receptor, the androgen receptor, or the pregnane ⁇ receptor.
  • a nuclear hormone receptor such as e.g., the glucocorticoid receptor, the estrogen receptor, the progesterone receptor, the androgen receptor, or the pregnane ⁇ receptor.
  • the ligands for these receptors include, for example, a corticosteroid, such as, e.g., dexamethasone at 10 nM-100 ⁇ M or hydrocortisone at 10 nM-100 ⁇ M; an estrogen, such as, e.g., beta-estradiol at 10 nM-100 ⁇ M; a progestogen, such as, e.g., progesterone at 10 nM-100 ⁇ M, hydroxyprogesterone at 10 nM-100 ⁇ M, 5a-dihydroprogesterone at 10 nM-100 ⁇ M, 11-deoxycorticosterone at 10 nM-100 ⁇ M, or a synthetic progestin, such as, e.g., chlormadinone acetate at 10 nM-100 ⁇ M; an androgen, such as, e.g., testosterone at 10 nM-100 ⁇ M, dihydrotestosterone at 10
  • the first step may also optionally comprise contacting the cells in culture with an insulin-like molecule, such as, e.g., insulin at 1-50 ⁇ g/mL, insulin-like growth factor 1 (IGF-1) at 1-50 ⁇ g/mL, insulin-like growth factor 2 (IGF-2) at 1-50 ⁇ g/mL, or mechano-growth factor at 1-50 ⁇ g/mL.
  • the first step further may optionally comprise contacting the cells in culture with transferrin at 0.1-5 mg/mL.
  • the first step may optionally comprise contacting the cells in culture with one or more interleukins (IL) or growth factors such as, e.g., IL-1, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-11, IL-12, granulocyte colony-stimulating factor (G-CSF), macrophage colony-stimulating factor (M-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), thrombopoietin, fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), transforming growth factor beta (TGF-B), tumor necrosis factor alpha (TNF-A), megakaryocyte growth and development factor (MGDF), leukemia inhibitory factor (LIF), and Flt3 ligand.
  • IL interleukins
  • growth factors such as, e.g., IL-1, IL-2, IL-4, IL-5, IL
  • the first step may also optionally comprise contacting the cells in culture with serum proteins or non-protein molecules such as, e.g., fetal bovine serum (1-20%), human plasma (1-20%), plasmanate (1-20%), human serum (1-20%), albumin (0.1-100 mg/mL), or heparin (0.1-10 U/mL).
  • serum proteins or non-protein molecules such as, e.g., fetal bovine serum (1-20%), human plasma (1-20%), plasmanate (1-20%), human serum (1-20%), albumin (0.1-100 mg/mL), or heparin (0.1-10 U/mL).
  • the second step may comprise contacting the cells in culture with stem cell factor (SCF) at 1-1000 ng/mL and erythropoietin (EPO) at 1-100 U/mL.
  • SCF stem cell factor
  • EPO erythropoietin
  • the second step may also optionally comprise contacting the cells in culture with an insulin-like molecule, such as e.g., insulin at 1-50 ⁇ g/mL, insulin-like growth factor 1 (IGF-1) at 1-50 ⁇ g/mL, insulin-like growth factor 2 (IGF-2) at 1-50 ⁇ g/mL, or mechano-growth factor at 1-50 ⁇ g/mL.
  • IGF-1 insulin-like growth factor 1
  • IGF-2 insulin-like growth factor 2
  • mechano-growth factor at 1-50 ⁇ g/mL.
  • the second step may further optionally comprise contacting the cells in culture with transferrin at 0.1-5 mg/mL.
  • the second may also optionally comprise contacting the cells in culture with serum proteins or non-protein molecules such as, e.g., fetal bovine serum (1-20%), human plasma (1-20%), plasmanate (1-20%), human serum (1-20%), albumin (0.1-100 mg/mL), or heparin (0.1-10 U/mL).
  • serum proteins or non-protein molecules such as, e.g., fetal bovine serum (1-20%), human plasma (1-20%), plasmanate (1-20%), human serum (1-20%), albumin (0.1-100 mg/mL), or heparin (0.1-10 U/mL).
  • the third step may comprise contacting the cells in culture with erythropoietin (EPO) at 1-100 U/mL.
  • the third step may optionally comprise contacting the cells in culture with stem cell factor (SCF) at 1-1000 ng/mL.
  • SCF stem cell factor
  • the third step may further optionally comprise contacting the cells in culture with an insulin-like molecule, such as e.g., insulin at 1-50 ⁇ g/mL, insulin-like growth factor 1 (IGF-1) at 1-50 ⁇ g/mL, insulin-like growth factor 2 (IGF-2) at 1-50 ⁇ g/mL, or mechano-growth factor at 1-50 ⁇ g/mL.
  • the third step may also optionally comprise contacting the cells in culture with transferrin at 0.1-5 mg/mL.
  • the third step may also optionally comprise contacting the cells in culture with serum proteins or non-protein molecules such as, e.g., fetal bovine serum (1-20%), human plasma (1-20%), plasmanate (1-20%), human serum (1-20%), albumin (0.1-100 mg/mL), or heparin (0.1-10 U/mL).
  • serum proteins or non-protein molecules such as, e.g., fetal bovine serum (1-20%), human plasma (1-20%), plasmanate (1-20%), human serum (1-20%), albumin (0.1-100 mg/mL), or heparin (0.1-10 U/mL).
  • methods of expansion and differentiation of the synthetic membrane-receiver complexes do not include culturing the synthetic membrane-receiver complexes in a medium comprising a myeloproliferative receptor (mpl) ligand.
  • mpl myeloproliferative receptor
  • the culture process may optionally comprise contacting cells by a method known in the art with a molecule, e.g., a DNA molecule, an RNA molecule, a mRNA, an siRNA, a microRNA, a lncRNA, a shRNA, a hormone, or a small molecule, that activates or knocks down one or more genes.
  • a molecule e.g., a DNA molecule, an RNA molecule, a mRNA, an siRNA, a microRNA, a lncRNA, a shRNA, a hormone, or a small molecule
  • Target genes can include, for example, genes that encode a transcription factor, a growth factor, or a growth factor receptor, including but not limited to, e.g., GATA1, GATA2, CMyc, hTERT, p53, EPO, SCF, insulin, EPO-R, SCF-R, transferrin-R, insulin-R.
  • CD34+ cells are placed in a culture containing varying amounts of IMDM, FBS, glutamine, BSA, holotransferrin, insulin, dexamethasone, ⁇ -estradiol, IL-3, SCF, and erythropoietin, in three separate differentiation stages for a total of 22 days.
  • CD34+ cells are placed in a culture containing varying amounts of IMDM, FBS, glutamine, BSA, holotransferrin, insulin, dexamethasone, ⁇ -estradiol, IL-3, SCF, and thrombopoietin, in three separate differentiation stages for a total of 14 days.
  • CD34+ cells are placed in a culture containing varying amounts of IMDM, FBS, glutamine, BSA, holotransferrin, insulin, dexamethasone, ⁇ -estradiol, IL-3, SCF, and GCSF, in three separate differentiation stages for a total of 15 days.
  • compositions comprising synthetic membrane-receiver complexes that are suitable for administration to a subject.
  • the pharmaceutical compositions generally comprise a population of synthetic membrane-receiver complexes and a pharmaceutically-acceptable carrier in a form suitable for administration to a subject.
  • Pharmaceutically-acceptable carriers are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of pharmaceutical compositions comprising a population of synthetic membrane-receiver complexes. (See, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa. 18th ed. (1990)).
  • the pharmaceutical compositions are generally formulated as sterile, substantially isotonic and in full compliance with all Good Manufacturing Practice (GMP) regulations of the U.S. Food and Drug Administration.
  • GMP Good Manufacturing Practice
  • excipients include excipients that are generally safe, non-toxic, and desirable, including excipients that are acceptable for veterinary use as well as for human pharmaceutical use.
  • excipients can be solid, liquid, semisolid, or, in the case of an aerosol composition, gaseous.
  • Examples of carriers or diluents include, but are not limited to, water, saline, Ringer's solutions, dextrose solution, and 5% human serum albumin.
  • the use of such media and compounds for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or compound is incompatible with the synthetic membrane-receiver complexes described herein, use thereof in the compositions is contemplated. Supplementary therapeutic agents may also be incorporated into the compositions.
  • a pharmaceutical composition is formulated to be compatible with its intended route of administration.
  • the synthetic membrane-receiver complexes can be administered by parenteral, topical, intravenous, oral, subcutaneous, intraarterial, intradermal, transdermal, rectal, intracranial, intraperitoneal, intranasal; intramuscular route or as inhalants.
  • the synthetic membrane-receiver complexes can optionally be administered in combination with other therapeutic agents that are at least partly effective in treating the disease, disorder or condition for which the synthetic membrane-receiver complexes are intended.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial compounds such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating compounds such as ethylenediaminetetraacetic acid (EDTA); buffers such as acetates, citrates or phosphates, and compounds for the adjustment of tonicity such as sodium chloride or dextrose.
  • the pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS).
  • the composition must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, e.g., water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, e.g., by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal compounds, e.g., parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic compounds e.g., sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition a compound which delays absorption, e.g., aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the synthetic membrane-receiver complexes in an effective amount and in an appropriate solvent with one or a combination of ingredients enumerated herein, as desired.
  • dispersions are prepared by incorporating the synthetic membrane-receiver complexes into a sterile vehicle that contains a basic dispersion medium and any desired other ingredients.
  • methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the synthetic membrane-receiver complexes can be administered in the form of a depot injection or implant preparation which can be formulated in such a manner to permit a sustained or pulsatile release of the synthetic membrane-receiver complexes, their receiver(s) and/or their oprional payload(s).
  • Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the synthetic membrane-receiver complexes can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding compounds, and/or adjuvant materials can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating compound such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening compound such as sucrose or saccharin; or a flavoring compound such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating compound such as alginic acid, Primogel, or corn starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • the synthetic membrane-receiver complexes are delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
  • a suitable propellant e.g., a gas such as carbon dioxide, or a nebulizer.
  • compositions comprising synthetic membrane-receiver complexes can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, e.g., for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • the modified red blood cells are formulated into ointments, salves, gels, or creams as generally known in the art.
  • the synthetic membrane-receiver complexes can also be prepared as pharmaceutical compositions in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
  • suppositories e.g., with conventional suppository bases such as cocoa butter and other glycerides
  • retention enemas for rectal delivery.
  • the synthetic membrane-receiver complexes are prepared with carriers that will decrease the rate with which synthetic membrane-receiver complexes are eliminated from the body of a subject.
  • controlled release formulation are suitable, including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc.
  • the pharmaceutical composition comprising synthetic membrane-receiver polypeptide complexes is administered intravenously into a subject that would benefit from the pharmaceutical composition.
  • the composition is administered to the lymphatic system, e.g., by intralymphatic injection or by intranodal injection (see e.g., Senti et al., 2008 PNAS 105(46):17908), or by intramuscular injection, by subcutaneous administration, by direct injection into the thymus, or into the liver.
  • the pharmaceutical composition comprising synthetic membrane-receiver polypeptide complexes is administered as a liquid suspension.
  • the pharmaceutical composition is administered as a coagulated formulation that is capable of forming a depot following administration, and in a preferred embodiment slowly release synthetic membrane-receiver polypeptide complexes into circulation, or in a preferred embodiment remain in depot form.
  • the pharmaceutical composition comprising synthetic membrane-receiver complexes is stored using methods and buffer compositions that are capable of maintaining viability of the synthetic membrane-receiver complexes. For example, deoxygenation prior to storage to maintain an anaerobic state, manipulation of pH, supplementation of metabolic precursors, manipulation of osmotic balance, increasing of the volume of the suspending medium, and/or reduction of oxidative stress by adding protective molecules can be used to maintain the viability of the synthetic membrane-receiver complexes.
  • deoxygenation prior to storage to maintain an anaerobic state manipulation of pH, supplementation of metabolic precursors, manipulation of osmotic balance, increasing of the volume of the suspending medium, and/or reduction of oxidative stress by adding protective molecules can be used to maintain the viability of the synthetic membrane-receiver complexes.
  • Several studies employing a combination of these strategies have reported maintenance of viability of erythrocytes allowing an extension of storage beyond 6 weeks (see e.g., Yos
  • Pharmaceutically acceptable carriers or excipients may be used to deliver the synthetic membrane-receiver polypeptides described herein.
  • Excipient refers to an inert substance used as a diluent or vehicle.
  • Pharmaceutically acceptable carriers are used, in general, with a compound so as to make the compound useful for a therapy or as a product.
  • a pharmaceutically acceptable carrier is a material that is combined with the substance for delivery to a subject.
  • Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.
  • the carrier is essential for delivery, e.g., to solubilize an insoluble compound for liquid delivery; a buffer for control of the pH of the substance to preserve its activity; or a diluent to prevent loss of the substance in the storage vessel.
  • the carrier is for convenience, e.g., a liquid for more convenient administration.
  • Pharmaceutically acceptable salts of the compounds described herein may be synthesized according to methods known to those skilled in the arts.
  • compositions are highly purified to be free of contaminants, are biocompatible and not toxic, and are suited to administration to a subject. If water is a constituent of the carrier, the water is highly purified and processed to be free of contaminants, e.g., endotoxins.
  • the pharmaceutically acceptable carrier may be lactose, dextrose, sucrose, sorbitol, mannitol, starch, gum acacia, calcium phosphate, alginates, gelatin, calcium silicate, micro-crystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, methyl cellulose, methylhydroxy benzoate, propylhydroxy benzoate, talc, magnesium stearate, and/or mineral oil, but is not limited thereto.
  • the pharmaceutical composition may further include a lubricant, a wetting agent, a sweetener, a flavor enhancer, an emulsifying agent, a suspension agent, and/or a preservative.
  • compositions containing synthetic membrane-receiver complexes having effective levels of receivers contain a plurality of synthetic membrane-receiver complexes, e.g., 1 ⁇ 10 3 complexes, or 1 ⁇ 10 4 , 1 ⁇ 10 5 , 1 ⁇ 10 6 , 1 ⁇ 10 7 , 1 ⁇ 10 8 , 1 ⁇ 10 9 , 1 ⁇ 10 10 , 1 ⁇ 10 11 , 1 ⁇ 10 12 , or greater than 1 ⁇ 10 12 complexes.
  • synthetic membrane-receiver complexes generated from erythroid cells may be administered as packed red blood cells in a saline solution at a concentration of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or greater than 90% mass to volume ratio (% m/v).
  • the time of administration to a patient may range from 10 minutes to four hours, or more.
  • synthetic membrane-receiver complexes generated from erythroid cells can be stored in an appropriate buffer, e.g., an FDA-approved anticoagulant preservative solution such as anticoagulant citrate-dextrose A (ACD-A), citrate-phosphate dextrose (CPD), Citratephosphate-dextrose-dextrose (CP2D), or citrate-phosphate-dextrose-adenine (CPDA-1).
  • ACD-A anticoagulant citrate-dextrose A
  • CPD citrate-phosphate dextrose
  • CP2D Citratephosphate-dextrose-dextrose
  • CPDA-1 citrate-phosphate-dextrose-adenine
  • synthetic membrane-receiver complexes generated from erythroid cells can be stored in an approved additive solution, e.g., AS-1 (Adsol), AS-3 (Nutricel), AS-5 (Optisol), or AS-7 (SOLX).
  • AS-1 Adsol
  • AS-3 Nutricel
  • AS-5 Optisol
  • SOLX SOLX
  • synthetic membrane-receiver complexes generated from erythroid cells can stored in a glycerol cryoprotective solution.
  • the compositions may be frozen and stored for up to 10 years. Frozen cells may be thawed and deglycerolized by successive washing steps, for example with 0.9% sodium chloride before use.
  • compositions and pharmaceutical compositions comprising a plurality of cultured functional erythroid cells that comprise a receiver.
  • the compositions and pharmaceutical compositions may comprise a solution of appropriate storage buffer such as, e.g., anticoagulant citrate-dextrose A.
  • the compositions and pharmaceutical compositions comprising the plurality of cultured functional erythroid cells that comprise a receiver may additionally comprise an approved additive such as, e.g., Adsol.
  • the compositions and pharmaceutical compositions comprising the plurality of cultured functional erythroid cells that comprise receiver may additionally comprise a glycerol cryoprotective solution for frozen storage.
  • the synthetic membrane-receiver polypeptide complex is able to form a multi-complex aggregate, e.g., a dimer, a trimer, a multimer, with another synthetic membrane-receiver polypeptide complex.
  • the synthetic membrane-receiver polypeptide complex is able to form a multi-complex aggregate, e.g., a dimer, a trimer, a multimer, with component of the circulatory system, e.g an erythrocyte, a reticulocyte, a platelet, a macrophage, a lymphocyte, a T cell, a B cell, a mast cell.
  • a multi-complex aggregate e.g., a dimer, a trimer, a multimer
  • component of the circulatory system e.g an erythrocyte, a reticulocyte, a platelet, a macrophage, a lymphocyte, a T cell, a B cell, a mast cell.
  • the dosing and frequency of the administration of the synthetic membrane-receiver complexes and pharmaceutical compositions thereof can be determined by the attending physician based on various factors such as the severity of disease, the patient's age, sex and diet, the severity of any inflammation, time of administration, and other clinical factors.
  • an intravenous administration is initiated at a dose which is minimally effective, and the dose is increased over a pre-selected time course until a positive effect is observed. Subsequently, incremental increases in dosage are made limiting to levels that produce a corresponding increase in effect while taking into account any adverse affects that may appear.
  • Non-limited examples of suitable dosages can range, for example, from 1 ⁇ 10 10 to 1 ⁇ 10 14 , from 1 ⁇ 10 11 to 1 ⁇ 10 13 , or from 5 ⁇ 10 11 to 5 ⁇ 10 12 synthetic membrane-receiver complexes. Specific examples include about 5 ⁇ 10 10 , 6 ⁇ 10 10 , 8 ⁇ 10 10 , 7 ⁇ 10 10 , 9 ⁇ 10 10 1 ⁇ 10 11 , 2 ⁇ 10 11 , 3 ⁇ 10 11 , 4 ⁇ 10 11 , 5 ⁇ 10 11 , 6 ⁇ 10 11 , 7 ⁇ 10 11 , 8 ⁇ 10 11 , 9 ⁇ 10 11 , 1 ⁇ 10 12 , or more synthetic membrane-receiver complexes. Each dose of synthetic membrane-receiver complexes can be administered at intervals such as once daily, once weekly, twice weekly, once monthly, or twice monthly.
  • Complex-based proportional dosage is the number of synthetic membrane-receiver complexes administered as a dose relative to a naturally occurring quantity of circulating entities.
  • the circulating entities may be cells, e.g., erythrocytes, reticulocytes, or lymphocytes, or targets, e.g., antigens, antibodies, viruses, toxins, cytokines, etc.
  • the units are defined as synthetic membrane-receiver complex per circulating entity, ie SCMRC/CE.
  • This dosage unit may include 10 ⁇ 7 , 10 ⁇ 6 , 10 ⁇ 5 , 10 ⁇ 4 , 10 ⁇ 3 , 10 ⁇ 2 , 10 ⁇ 1 , 1, 10, 10 2 , 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , 10 8 , 10 9 .
  • compositions described herein comprise a synthetic membrane-receiver complex and optionally a pharmaceutically active or therapeutic agent.
  • the therapeutic agent can be a biological agent, a small molecule agent, or a nucleic acid agent.
  • Dosage forms are provided that comprise a pharmaceutical composition comprising a synthetic membrane-receiver complex described herein.
  • the dosage form is formulated as a liquid suspension for intravenous injection.
  • Medical devices comprise a container holding a pharmaceutical composition comprising a synthetic membrane-receiver complex described herein and an applicator for intravenous injection of the pharmaceutical composition to a subject.
  • Medical kits comprise a pharmaceutical composition comprising a synthetic membrane-receiver complex described herein and a medical device for intravenous injection of the pharmaceutical composition to a subject.
  • a pharmaceutically acceptable suspension of synthetic membrane-receiver complexes is preferably packaged in a volume of approximately 10 to approximately 250 ml.
  • the packaging can be a syringe or an IV bag suitable for transfusions.
  • Administration of the suspension is carried out, e.g., by intravenous or intra-arterial injection, optionally using a drip from an IV bag or the like.
  • the administration is typically carried out intravenously in the arm or via a central catheter. For administrations exceeding 50 ml use of a drip is preferred.
  • the membrane-receiver complex is generated using a precursor hematopoietic cell, e.g., a CD34+ cell, an erythrocyte, a platelet, a megakaryocyte, or a neutrophil as a source.
  • the precursor hematopoietic cell is isolated from a human donor by a GMP-compliant process.
  • the starting cells are sourced from an autologous donor.
  • the starting cells are sourced from an allogeneic donor.
  • the donor may be typed for blood cell antigen polymorphisms and/or the donor is genotyped for blood cell antigens.
  • the donor can be a universal blood donor.
  • the donor has the Bombay phenotype, .ie. does not express the H antigen.
  • the donor has ABO blood type 0 and is Rh-negative.
  • the membrane-receiver complex is generated using CD34+ hematopoietic progenitor cells, mobilized peripheral CD34+ cells, or bone marrow-derived CD34+ cells as a source for the starting material.
  • the starting cells are derived from umbilical cord blood, are induced pluripotent stem cells or are embryonic stem cells.
  • the synthetic membrane-receiver complex may be cultured. Cultured complexes can be scaled up from bench-top scale to bioreactor scale. For example, the complexes are cultured until they reach saturation density, e.g., 1 ⁇ 10 5 , 1 ⁇ 10 6 , 1 ⁇ 10 7 , or greater than 1 ⁇ 10 7 complexes per ml. Optionally, upon reaching saturation density, the complexes can be transferred to a larger volume of fresh medium.
  • the membrane-receiver complexes may be cultured in a bioreactor, such as, e.g., a Wave-type bioreactor, a stirred-tank bioreactor.
  • bioreactors Various configurations of bioreactors are known in the art and a suitable configuarion may be chosen as desired. Configurations suitable for culturing and/or expanding populations of synthetic membrane-receiver complexes can easily be determined by one of skill in the art without undue experimentation.
  • the bioreactor can be oxygenated.
  • the bioreactor may optionally contain one or more impellers, a recycle stream, a media inlet stream, and control components to regulate the influx of media and nutrients or to regulate the outflux of media, nutrients, and waste products.
  • the bioreactor may contain a population of human functional erythroid cells comprising a receiver that shed their intracellular DNA over the course of the culture process.
  • the bioreactor may contain a population of human erythroid cells, enucleated erythroid cells, and pyrenocytes after culture.
  • the human erythroid cells and enucleated erythroid cells comprise a receiver and the receiver is retained by the enucleated erythroid cell, whereas the exogenous nucleic acid encoding the receiver is not retained by the enucleated cell.
  • the enucleated functional erythroid cell comprising the receiver exhibits substantially the same osmotic membrane fragility as a corresponding isolated unmodified, uncultured erythroid cell.
  • the population of synthetic membrane-receiver complexes generated from erythroid cells or erythroid cell precursors in the bioreactor undergo a total expansion of greater than 20,000-fold in 14 days or greater.
  • the receiver is introduced into a cultured or freshly isolated erythroid cell precursor and after introduction of an exogenous nucleic acid encoding the receiver the population of synthetic membrane-receiver complexes generated from the erythroid cell precursors in the bioreactor expands in the bioreactor from the precursor cells by more than 20,000-fold.
  • the bioreactor is a Wave bioreactor or a impeller-driven agitator.
  • the bioreactor may be aerated by means of a sparger.
  • the bioreactor is disposable.
  • the bioreactor is CIP (cleaned in place).
  • the final complexes number of synthetic membrane-receiver complexes that may be obtained in a bioreactor setting as described herein can be greater than 10 9 , 10 10 , 10 11 , 10 12 , 10 13 or greater than 10 13 complexes.
  • the density of synthetic membrane-receiver complexes may be monitored during culture by measuring cell density by hemacytometer counting or by optical density reading at 600 nm.
  • the culture process is monitored for pH levels, oxygenation, agitation rate, and/or recycle rate.
  • the identity of the membrane-receiver complexes can be assessed by in vitro assays. For example, the identity of the membrane-receiver complexes is assessed by counting the number of complexes in a population, e.g., by microscopy, by flow cytometry, or by hemacytometry. Alternatively or in addition, the identity of the membrane-receiver complexes is assessed by analysis of protein content of the complex, e.g., by flow cytometry, Western blot, immunoprecipitation, fluorescence spectroscopy, chemiluminescence, mass spectrometry, or absorbance spectroscopy.
  • the protein content assayed is a non-surface protein, e.g., an integral membrane protein, hemoglobin, adult hemoglobin, fetal hemoglobin, embryonic hemoglobin, a cytoskeletal protein.
  • the protein content assayed is a surface protein, e.g., a differentiation marker, a receptor, a co-receptor, a transporter, a glycoprotein.
  • the surface protein is selected from the list including, but not limited to, glycophorin A, CKIT, transferrin receptor, Band3, Kell, CD45, CD46, CD47, CD55, CD59, CR1.
  • the identity of the membrane-receiver complexes is assessed by analysis of the receiver content of the complex, e.g., by flow cytometry, Western blot, immunoprecipitation, fluorescence spectroscopy, chemiluminescence, mass spectrometry, or absorbance spectroscopy.
  • the identity of the membrane-receiver complexes can be assessed by the mRNA content of the complexes, e.g., by RT-PCR, flow cytometry, or northern blot.
  • the identity of the membrane-receiver complexes can be assessed by nuclear material content, e.g., by flow cytometry, microscopy, or southern blot, using, e.g., a nuclear stain or a nucleic acid probe.
  • the identity of the membrane-receiver complexes is assessed by lipid content of the complexes, e.g by flow cytometry, liquid chromatography, or by mass spectrometry.
  • the identity of the membrane-receiver complexes is assessed by metabolic activity of the complexes, e.g by mass spectrometry, chemiluminescence, fluorescence spectroscopy, absorbance spectroscopy.
  • Metabolic activity can be assessed by ATP consumption rate and/or the metabolic activity is assessed measuring 2,3-diphosphoglycerate (2,3-DPG) level in the synthetic membrane-receiver complex.
  • the metabolic activity can be assessed as the rate of metabolism of one of the following, including but not limited to, Acetylsalicylic acid, N-Acetylcystein, 4-Aminophenol, Azathioprine, Bunolol, Captopril, Chlorpromazine, Dapsone, Daunorubicin, Dehydroepiandrosterone, Didanosin, Dopamine, Epinephrine, Esmolol, Estradiol, Estrone, Etoposide, Haloperidol, Heroin, Insulin, Isoproterenol, Isosorbide dinitrate, LY 217896,6-mercaptopurine, Misonidazole, Nitroglycerin, Norepinephrine, Para-aminobenzoic acid.
  • the identity of the membrane-receiver complexes is assessed by partitioning of a substrate by the complexes, e.g by mass spectrometry, chemiluminescence, fluorescence spectroscopy, or absorbance spectroscopy.
  • the substrate can be one of the following, including but not limited to, Acetazolamide, Arbutine, Bumetamide, Creatinine, Darstine, Desethyldorzolamide, Digoxigenin digitoxoside, Digoxin-16′-glucuronide, Epinephrine, Gentamycin, Hippuric acid, Metformin, Norepinephrine, p-Aminohippuric acid, Papaverine, Penicillin G, Phenol red, Serotonin, Sulfosalicylic acid, Tacrolimus, Tetracycline, Tucaresol, and Vancomycin.
  • the population of synthetic membrane-receiver complexes is differentiated from a precursor cell or complex.
  • the differentiation state of the population of synthetic membrane-receiver complexes is assessed by an in vitro assay.
  • the in vitro assays include those described herein for assessing the identity of the complexes, including but not limited to expansion rate, number, protein content or expression level, mRNA content or expression level, lipid content, partition of a substrate, catalytic activity, or metabolic activity.
  • the membrane-receiver complexes are cultured and the differentiation state of the complexes is assessed at multiple time points over the course of the culture process.
  • Synthetic membrane-receiver complexes may be generated using reticulocytes as a source for starting material.
  • the purity of isolated reticulocytes may be assessed using microscopy in that reticulocytes are characterized by a reticular (mesh-like) network of ribosomal RNA that becomes visible under a microscope with certain stains such as new methylene blue or brilliant cresyl blue.
  • Surface expression of transferrin receptor (CD71) is also higher on reticulocytes and decreases and they mature to erythrocytes, allowing for enrichment and analysis of reticulocyte populations using anti-CD71 antibodies (See, e.g., Miltenyi CD71 microbeads product insert No. 130-046-201).
  • analysis of creatine and hemoglobin A1C content and pyruvate kinase, aspartate aminotransferase, and porphobilinogen deaminase enzyme activity may be used to assess properties of the isolated reticulocytes relative to mature erythrocytes (See, e.g., Brun et al., Blood 76:2397-2403 (1990)).
  • the activity of porphobilinogen deaminase is nearly 9 fold higher whereas the hemoglobin A1C content is nearly 10 fold less in reticulocytes relative to mature erythrocytes.
  • cells suitable for generating synthetic membrane-receiver complexes are differentiated ex vivo and/or in vivo from one or more stem cells.
  • the one or more stem cells are one or more hematopoietic stem cells.
  • Various assays may be performed to confirm the ex vivo differentiation of cultured hematopoietic stem cells into reticulocytes and erythrocytes, including, for example, microscopy, hematology, flow cytometry, deformability measurements, enzyme activities, and hemoglobin analysis and functional properties (Giarratana et al., Nature Biotech. 23:69-74 (2005)).
  • the phenotype of cultured hematopoietic stem cells may be assessed using microscopy of cells stained, for example, with Cresyl Brilliant blue.
  • Reticulocytes for example, exhibit a reticular network of ribosomal RNA under these staining conditions whereas erythrocytes are devoid of staining.
  • Enucleated cells may also be monitored for standard hematological variables including mean corpuscular volume (MCV; femtoliters (fL)), mean corpuscular hemoglobin concentration (MCHC; %) and mean corpuscular hemoglobin (MCH; pg/cell) using, for example, an XE2100 automat (Sysmex, Roche Diagnostics).
  • the synthetic membrane-receiver complexes are assessed for their basic physical properties, e.g., size, mass, volume, diameter, buoyancy, density, and membrane properties, e.g., viscosity, deformability fluctuation, and fluidity.
  • the diameter of the synthetic membrane-receiver complexes is measured by microscopy or by automated instrumentation, e.g., a hematological analysis instrument. In one embodiment the diameter of the synthetic membrane-receiver complexes is between about 1-20 microns. In one embodiment, the diameter of the synthetic membrane-receiver complexes is at least in one dimension between about 1-20 microns. In one embodiment, the diameter of the synthetic membrane-receiver complexes is less than about 1 micron. In one embodiment, the diameter of the complexes in one dimension is larger than about 20 microns.
  • the diameter of the synthetic membrane-receiver complexes is between about 1 micron and about 20 microns, between about 2 microns and about 20 microns between about 3 microns and about 20 microns between about 4 microns and about 20 microns between about 5 microns and about 20 microns between about 6 microns and about 20 microns, between about 5 microns and about 15 microns or between about 10 microns and about 30 microns.
  • the mean corpuscular volume of the synthetic membrane-receiver complexes is measured using a hematological analysis instrument. In one embodiment the volume of the mean corpuscular volume of the complexes is greater than 10 fL, 20 fL, 30 fL, 40 fL, 50 fL, 60 fL, 70 fL, 80 fL, 90 fL, 100 fL, 110 fL, 120 fL, 130 fL, 140 fL, 150 fL, or greater than 150 fL.
  • the mean corpuscular volume of the complexes is less than 30 fL, 40 fL, 50 fL, 60 fL, 70 fL, 80 fL, 90 fL, 100 fL, 110 fL, 120 fL, 130 fL, 140 fL, 150 fL, 160 fL, 170 fL, 180 fL, 190 fL, 200 fL, or less than 200 fL. In one embodiment the mean corpuscular volume of the complexes is between 80-100 femtoliters (fL).
  • the average buoyant mass of the synthetic membrane-receiver complexes is measured using a suspended microchannel resonatory or a double suspended microchannel resonatory (see e.g., Byun et al PNAS 2013 110(19):7580 and Bryan et al. Lab Chip 2014 14(3):569).
  • the dry density of the synthetic membrane-receiver complexes is measured by buoyant mass in an H2O-D2O exchange assay (see e.g., Feijo Delgado et al., PLOS One 2013 8(7):e67590).
  • the synthetic membrane-receiver complexes have an average membrane deformability fluctuation of standard deviation greater than 10, 20, 30, 40, 50, 60, 70, 80, 90, 100 or greater than 100 mrad as measured by spatial light interference microscopy (SLIM) (see e.g., Bhaduri et al., Sci Reports 2014, 4:6211).
  • SLIM spatial light interference microscopy
  • the average membrane viscosity of a population of synthetic membrane-receiver complexes is measured by detecting the average fluorescence upon incubation with viscosity-dependent quantum yield fluorophores (see e.g., Haidekker et al. Chem & Biol 2001 8(2):123).
  • the membrane fluidity of the synthetic membrane-receiver complexes is measured by fluorescence polarization, e.g., with BMG Labtech POLARstar Omega microplate reader.
  • reticulocytes may be separated from nucleated cells on day 15 of culture, for example, by passage through a deleukocyting filter (e.g., Leucolab LCG2, Macopharma) and subsequently assayed using ektacytometry.
  • the enucleated cells are suspended in 4% polyvinylpyrrolidone solution and then exposed to an increasing osmotic gradient from 60 to 450 mosM. Changes in the laser diffraction pattern (deformability index) of the cells are recorded as a function of osmolarity, to assess the dynamic deformability of the cell membrane.
  • the maximum deformability index achieved at a physiologically relevant osmolarity is related to the mean surface area of erythrocytes.
  • the synthetic membrane-receiver complexes are analyzed for hemoglobin contents.
  • Assays of hemoglobin may be used to assess the phenotype of differentiated cells (Giarratana et al., Nature Biotech. 23:69-74 (2005)).
  • HPLC high performance liquid chromatography
  • Bio-Rad Variant II Hb analyzer Bio-Rad Laboratories
  • Oxygen equilibrium may be measured using a continuous method with a double-wavelength spectrophotometer (e.g., Hemox analyzer, TCS).
  • the binding properties of hemoglobin may be assessed using flash photolysis. In this method, the rebinding of CO to intracellular hemoglobin tetramers are analyzed at 436 nm after photolysis with a 10 nanosecond pulse at 532 nm.
  • the synthetic membrane-receiver complexes described herein can be purified following manufacture if desired. Many suitable methods of purification are known in the art.
  • the synthetic membrane-receiver complexes are purified by centrifugation, magnetophoresis, irradiation, acoustophoresis, and chemical or physical enucleation.
  • synthetic membrane-receiver complexes are purified by ex vivo maturation with, e.g., a stromal cell co-culture.
  • synthetic membrane-receiver complexes are purified by chemical or enzymatic treatment of complexes, e.g by treatment with a deglycosylation enzyme.
  • the synthetic membrane-receiver polypeptide complexes are purified by disabling any residual replicative potential of the membrane-receiver polypeptide complexes.
  • the synthetic membrane-receiver polypeptide complexes are subjected to radiation, e.g., X rays, gamma rays, beta particles, alpha particles, neutrons, protons, elemental nuclei, UV rays in order to damage residual replication-competent nucleic acids.
  • Ionizing radiation is energy transmitted via X rays, gamma rays, beta particles (high-speed electrons), alpha particles (the nucleus of the helium atom), neutrons, protons, and other heavy ions such as the nuclei of argon, nitrogen, carbon, and other elements.
  • X rays and gamma rays are electromagnetic waves like light, but their energy is much higher than that of light (their wavelengths are much shorter).
  • Ultraviolet (UV) light is a radiation of intermediate energy that can damage cells but UV light differs from the forms of electromagnetic radiation mentioned above in that it does not cause ionization (loss of an electron) in atoms or molecules, but rather excitation (change in energy level of an electron).
  • Radio-induced ionizations may act directly on the cellular component molecules or indirectly on water molecules, causing water-derived radicals. Radicals react with nearby molecules in a very short time, resulting in breakage of chemical bonds or oxidation (addition of oxygen atoms) of the affected molecules.
  • the major effect in cells is DNA breaks. Since DNA consists of a pair of complementary double strands, breaks of either a single strand or both strands can occur. However, the latter is believed to be much more important biologically. Most single-strand breaks can be repaired normally thanks to the double-stranded nature of the DNA molecule (the two strands complement each other, so that an intact strand can serve as a template for repair of its damaged, opposite strand). In the case of double-strand breaks, however, repair is more difficult and erroneous rejoining of broken ends may occur. These so-called misrepairs result in induction of mutations, chromosome aberrations, or cell death.
  • Deletion of DNA segments is the predominant form of radiation damage in cells that survive irradiation. It may be caused by (1) misrepair of two separate double-strand breaks in a DNA molecule with joining of the two outer ends and loss of the fragment between the breaks or (2) the process of cleaning (enzyme digestion of nucleotides—the component molecules of DNA) of the broken ends before rejoining to repair one double-strand break.
  • Radiations differ not only by their constituents (electrons, protons, neutrons, etc.) but also by their energy. Radiations that cause dense ionization along their track (such as neutrons) are called high-linear-energy-transfer (high-LET) radiation, a physical parameter to describe average energy released per unit length of the track. (See the accompanying figure.) Low-LET radiations produce ionizations only sparsely along their track and, hence, almost homogeneously within a cell. Radiation dose is the amount of energy per unit of biological material (e.g., number of ionizations per cell).
  • high-LET radiations are more destructive to biological material than low-LET radiations—such as X and gamma rays—because at the same dose, the low-LET radiations induce the same number of radicals more sparsely within a cell, whereas the high-LET radiations—such as neutrons and alpha particles—transfer most of their energy to a small region of the cell.
  • the localized DNA damage caused by dense ionizations from high-LET radiations is more difficult to repair than the diffuse DNA damage caused by the sparse ionizations from low-LET radiations.
  • a population of synthetic membrane-receiver polypeptide complexes are subjected to gamma irradiation using an irradiation dose of more than 1, 5, 10, 15, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, or more than 100 kGy.
  • a population of synthetic membrane-receiver polypeptide complexes are subjected to X-ray irradiation using an irradiation dose of more than 0.1, 0.5, 1, 5, 10, 15, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000, 10000, or greater than 10000 mSv.
  • the purity of a population of synthetic membrane-receiver complexes can be assessed by measuring the homogeneity of the population.
  • the average distribution width of the synthetic membrane-receiver complexes is measured by a hematological analysis instrument.
  • the population of synthetic membrane-receiver complexes has a reticulocyte to non-reticulocyte ratio greater than 10, 100, 1000, 10 4 , 10 5 , 10 6 , or greater than 10 6 .
  • the homogeneity of the population of synthetic membrane-receiver complexes may be assessed by measuring the stromal cell content of the population.
  • the population of synthetic membrane-receiver complexes has less than 1 ppb of stromal cells.
  • the homogeneity of the population of synthetic membrane-receiver complexes is assessed by measuring the viral titer and/or a bacterial colony forming potential of the population.
  • the homogeneity of a population of synthetic membrane-receiver complexes is assessed by an in vitro assay.
  • the in vitro assays include those described herein for assessing the identity of the complexes, including but not limited to expansion rate, number, protein content or expression level, mRNA content or expression level, lipid content, partition of a substrate, catalytic activity, or metabolic activity.
  • Mature erythrocytes for use in generating the synthetic membrane-receiver complexes may be isolated using various methods such as, for example, a cell washer, a continuous flow cell separator, density gradient separation, fluorescence-activated cell sorting (FACS), Miltenyi immunomagnetic depletion (MACS), or a combination of these methods (See, e.g., van der Berg et al., Clin. Chem. 33:1081-1082 (1987); Bar-Zvi et al., J. Biol. Chem. 262:17719-17723 (1987); Goodman et al., Exp. Biol. Med. 232:1470-1476 (2007)).
  • FACS fluorescence-activated cell sorting
  • MCS Miltenyi immunomagnetic depletion
  • Erythrocytes may be isolated from whole blood by simple centrifugation (See, e.g., van der Berg et al., Clin. Chem. 33:1081-1082 (1987)).
  • EDTA-anticoagulated whole blood may be centrifuged at 800 ⁇ g for 10 min at 4° C.
  • the platelet-rich plasma and buffy coat are removed and the red blood cells are washed three times with isotonic saline solution (NaCl, 9 g/L).
  • erythrocytes may be isolated using density gradient centrifugation with various separation mediums such as, for example, Ficoll, Hypaque, Histopaque, Percoll, Sigmacell, or combinations thereof.
  • various separation mediums such as, for example, Ficoll, Hypaque, Histopaque, Percoll, Sigmacell, or combinations thereof.
  • a volume of Histopaque-1077 is layered on top of an equal volume of Histopaque-1119.
  • EDTA-anticoagulated whole blood diluted 1:1 in an equal volume of isotonic saline solution (NaCl, 9 g/L) is layered on top of the Histopaque and the sample is centrifuged at 700 ⁇ g for 30 min at room temperature.
  • granulocytes migrate to the 1077/1119 interface, lymphocytes, other mononuclear cells and platelets remain at the plasma/1077 interface, and the red blood cells are pelleted.
  • the red blood cells are washed twice with isotonic saline solution.
  • erythrocytes may be isolated by centrifugation using a Percoll step gradient (See, e.g., Bar-Zvi et al., J. Biol. Chem. 262:17719-17723 (1987)).
  • a Percoll step gradient See, e.g., Bar-Zvi et al., J. Biol. Chem. 262:17719-17723 (1987)).
  • fresh blood is mixed with an anticoagulant solution containing 75 mM sodium citrate and 38 mM citric acid and the cells washed briefly in Hepes-buffered saline.
  • Leukocytes and platelets are removed by adsorption with a mixture of ⁇ -cellulose and Sigmacell (1:1).
  • the erythrocytes are further isolated from reticulocytes and residual white blood cells by centrifugation through a 45/75% Percoll step gradient for 10 min at 2500 rpm in a Sorvall SS34 rotor.
  • the erythrocytes are recovered in the pellet while reticulocytes band at the 45/75% interface and the remaining white blood cells band at the 0/45% interface.
  • the Percoll is removed from the erythrocytes by several washes in Hepes-buffered saline.
  • Other materials that may be used to generate density gradients for isolation of erythrocytes include OptiPrepTM, a 60% solution of iodixanol in water (from Axis-Shield, Dundee, Scotland).
  • Erythrocytes may be separated from reticulocytes, for example, using flow cytometry (See, e.g., Goodman el al., Exp. Biol. Med. 232:1470-1476 (2007)).
  • whole blood is centrifuged (550 ⁇ g, 20 min, 25° C.) to separate cells from plasma.
  • the cell pellet is resuspended in phosphate buffered saline solution and further fractionated on Ficoll-Paque (1.077 density), for example, by centrifugation (400 ⁇ g, 30 min, 25° C.) to separate the erythrocytes from the white blood cells.
  • the resulting cell pellet is resuspended in RPMI supplemented with 10% fetal bovine serum and sorted on a FACS instrument such as, for example, a Becton Dickinson FACSCalibur (BD Biosciences, Franklin Lakes, N.J., USA) based on size and granularity.
  • a FACS instrument such as, for example, a Becton Dickinson FACSCalibur (BD Biosciences, Franklin Lakes, N.J., USA) based on size and granularity.
  • Erythrocytes may be isolated by immunomagnetic depletion (See, e.g., Goodman, el al., (2007) Exp. Biol. Med. 232:1470-1476).
  • magnetic beads with cell-type specific antibodies are used to eliminate non-erythrocytes.
  • erythrocytes are isolated from the majority of other blood components using a density gradient as described herein followed by immunomagnetic depletion of any residual reticulocytes.
  • the cells are pre-treated with human antibody serum for 20 min at 25° C. and then treated with antibodies against reticulocyte specific antigens such as, for example, CD71 and CD36.
  • the antibodies may be directly attached to magnetic beads or conjugated to PE, for example, to which magnetic beads with anti-PE antibody will react.
  • the antibody-magnetic bead complex is able to selectively extract residual reticulocytes, for example, from the erythrocyte population.
  • Erythrocytes may also be isolated using apheresis.
  • the process of apheresis involves removal of whole blood from a patient or donor, separation of blood components using centrifugation or cell sorting, withdrawal of one or more of the separated portions, and transfusion of remaining components back into the patient or donor.
  • a number of instruments are currently in use for this purpose such as for example the Amicus and Alyx instruments from Baxter (Deerfield, Ill., USA), the Trima Accel instrument from Gambro BCT (Lakewood, Colo., USA), and the MCS+9000 instrument from Haemonetics (Braintree, Mass., USA). Additional purification methods may be necessary to achieve the appropriate degree of cell purity.
  • the synthetic membrane-receiver complexes are differentiated ex vivo and/or in vivo from one or more reticulocytes.
  • Reticulocytes may be used to generate synthetic membrane-receiver complexes.
  • Reticulocytes are immature red blood cells and compose approximately 1% of the red blood cells in the human body. Reticulocytes develop and mature in the bone marrow. Once released into circulation, reticulocytes rapidly undergo terminal differentiation to mature erythrocytes. Like mature erythrocytes, reticulocytes do not have a cell nucleus. Unlike mature erythrocytes, reticulocytes maintain the ability to perform protein synthesis.
  • exogenous nucleic acid such as mRNA
  • encoding a receiver is introduced into reticulocytes to generate synthetic membrane-receiver complexes.
  • Reticulocytes of varying age may be isolated from peripheral blood based on the differences in cell density as the reticulocytes mature. Reticulocytes may be isolated from peripheral blood using differential centrifugation through various density gradients. For example, Percoll gradients may be used to isolate reticulocytes (See, e.g., Noble el al., Blood 74:475-481 (1989)).
  • Sterile isotonic Percoll solutions of density 1.096 and 1.058 g/ml are made by diluting Percoll (Sigma-Aldrich, Saint Louis, Mo., USA) to a final concentration of 10 mM triethanolamine, 117 mM NaCl, 5 mM glucose, and 1.5 mg/ml bovine serum albumin (BSA). These solutions have an osmolarity between 295 and 310 mOsm. Five milliliters, for example, of the first Percoll solution (density 1.096) is added to a sterile 15 ml conical centrifuge tube.
  • Two milliliters, for example, of the second Percoll solution (density 1.058) is layered over the higher density first Percoll solution.
  • Two to four milliliters of whole blood are layered on top of the tube.
  • the tube is centrifuged at 250 ⁇ g for 30 min in a refrigerated centrifuge with swing-out tube holders. Reticulocytes and some white cells migrate to the interface between the two Percoll layers.
  • the cells at the interface are transferred to a new tube and washed twice with phosphate buffered saline (PBS) with 5 mM glucose, 0.03 mM sodium azide and 1 mg/ml BSA. Residual white blood cells are removed by chromatography in PBS over a size exclusion column.
  • PBS phosphate buffered saline
  • reticulocytes may be isolated by positive selection using an immunomagnetic separation approach (See, e.g., Brun et al., Blood 76:2397-2403 (1990)).
  • This approach takes advantage of the large number of transferrin receptors that are expressed on the surface of reticulocytes relative to erythrocytes prior to maturation.
  • Magnetic beads coated with an antibody to the transferrin receptor may be used to selectively isolate reticulocytes from a mixed blood cell population.
  • Antibodies to the transferrin receptor of a variety of mammalian species, including human, are available from commercial sources (e.g., Affinity BioReagents, Golden, Colo., USA; Sigma-Aldrich, Saint Louis, Mo., USA).
  • the transferrin antibody may be directly linked to the magnetic beads.
  • the transferrin antibody may be indirectly linked to the magnetic beads via a secondary antibody.
  • mouse monoclonal antibody 10D2 Affinity BioReagents, Golden, Colo., USA
  • human transferrin may be mixed with immunomagnetic beads coated with a sheep anti-mouse immunoglobulin G (Dynal/Invitrogen, Carlsbad, Calif., USA).
  • the immunomagnetic beads are then incubated with a leukocyte-depleted red blood cell fraction.
  • the beads and red blood cells are incubated at 22° C. with gentle mixing for 60-90 min followed by isolation of the beads with attached reticulocytes using a magnetic field.
  • the isolated reticulocytes may be removed from the magnetic beads using, for example, DETACHaBEAD® solution (from Invitrogen, Carlsbad, Calif., USA).
  • reticulocytes may be isolated from in vitro growth and maturation of CD34+ hematopoietic stem cells using the methods described herein.
  • Terminally-differentiated, enucleated erythrocytes can be separated from other cells based on their DNA content.
  • cells are first labeled with a vital DNA dye, such as Hoechst 33342 (Invitrogen Corp.).
  • Hoechst 33342 is a cell-permeant nuclear counterstain that emits blue fluorescence when bound to double-stranded DNA.
  • Undifferentiated precursor cells, macrophages or other nucleated cells in the culture are stained by Hoechst 33342, while enucleated erythrocytes are Hoechst-negative.
  • the Hoechst-positive cells can be separated from enucleated erythrocytes by using fluorescence activated cell sorters or other cell sorting techniques.
  • the Hoechst dye can be removed from the isolated erythrocytes by dialysis or other suitable methods.
  • a population of synthetic membrane-receiver complexes can be purified by reducing the nuclear material content of the population of complexes. For example, the enucleation rate of the population of complexes is increased, and/or the number of enucleated synthetic membrane-receiver complexes is increased or enriched.
  • Populations of synthetic membrane-receiver complexes can be incubated with a small molecule, e.g., an actin inhibitor, e.g., cytochalasin A, B, C, D, E, F, H, J, and then centrifuged to remove nuclear material.
  • a population of synthetic membrane-receiver complexes can be mechanically manipulated by passing through progressively smaller size-restrictive filters to remove nuclear material.
  • the population of synthetic membrane-receiver complexes may also be incubated on a fibronectin-coated plastic surface to increase the removal of nuclear material.
  • the population of synthetic membrane-receiver complexes is incubated in co-culture with stromal cells, e.g., macrophages, to increase the removal of nuclear material.
  • the population of synthetic membrane-receiver complexes is greater than 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, 99.5%, 99.9%, or greater than 99.9% enucleated.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Genetics & Genomics (AREA)
  • Cell Biology (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • Hematology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Virology (AREA)
  • Microbiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Engineering & Computer Science (AREA)
  • Diabetes (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Dermatology (AREA)
  • Mycology (AREA)
  • Transplantation (AREA)
  • Communicable Diseases (AREA)
  • Botany (AREA)
  • Developmental Biology & Embryology (AREA)
  • Cardiology (AREA)
US14/581,486 2013-11-18 2014-12-23 Synthetic membrane-receiver complexes Abandoned US20150182588A1 (en)

Priority Applications (19)

Application Number Priority Date Filing Date Title
US14/581,486 US20150182588A1 (en) 2013-11-18 2014-12-23 Synthetic membrane-receiver complexes
US14/738,414 US9644180B2 (en) 2013-11-18 2015-06-12 Synthetic membrane-receiver complexes
US15/473,421 US10344263B2 (en) 2013-11-18 2017-03-29 Synthetic membrane-receiver complexes
US15/783,921 US10329531B2 (en) 2013-11-18 2017-10-13 Synthetic membrane-receiver complexes
US15/824,877 US10253296B2 (en) 2013-11-18 2017-11-28 Synthetic membrane-receiver complexes
US15/899,895 US20180187153A1 (en) 2013-11-18 2018-02-20 Synthetic membrane-receiver complexes
US15/900,029 US20180187154A1 (en) 2013-11-18 2018-02-20 Synthetic membrane-receiver complexes
US15/925,212 US10301593B2 (en) 2013-11-18 2018-03-19 Synthetic membrane-receiver complexes
US15/940,069 US20180265847A1 (en) 2013-11-18 2018-03-29 Synthetic membrane-receiver complexes
US15/940,215 US20180216067A1 (en) 2013-11-18 2018-03-29 Synthetic membrane-receiver complexes
US16/195,811 US10301594B1 (en) 2013-11-18 2018-11-19 Synthetic membrane-receiver complexes
US16/409,573 US20190264177A1 (en) 2013-11-18 2019-05-10 Synthetic membrane-receiver complexes
US16/409,576 US10557119B2 (en) 2013-11-18 2019-05-10 Erythroid cells comprising phenylalanine ammonia lyase
US16/426,880 US20190316090A1 (en) 2013-11-18 2019-05-30 Erythroid cells comprising arginine deiminase
US16/426,905 US20190316091A1 (en) 2013-11-18 2019-05-30 Erythroid cells comprising arginase
US16/431,236 US20190309261A1 (en) 2013-11-18 2019-06-04 Erythroid cells comprising lysine oxidase
US16/431,270 US20190309262A1 (en) 2013-11-18 2019-06-04 Erythroid cells comprising serine dehydratase
US16/549,840 US20190376034A1 (en) 2013-11-18 2019-08-23 Platelets comprising exogenous polypeptides and uses thereof
US16/572,129 US20200002674A1 (en) 2013-11-18 2019-09-16 Erythroid cells comprising phenylalanine hydroxylase

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
US201361962867P 2013-11-18 2013-11-18
US201361919432P 2013-12-20 2013-12-20
US201461973764P 2014-04-01 2014-04-01
US201461991319P 2014-05-09 2014-05-09
US201462006832P 2014-06-02 2014-06-02
US201462006825P 2014-06-02 2014-06-02
US201462006829P 2014-06-02 2014-06-02
US201462025367P 2014-07-16 2014-07-16
US201462059100P 2014-10-02 2014-10-02
PCT/US2014/065304 WO2015073587A2 (en) 2013-11-18 2014-11-12 Synthetic membrane-receiver complexes
US14/581,486 US20150182588A1 (en) 2013-11-18 2014-12-23 Synthetic membrane-receiver complexes

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/065304 Continuation WO2015073587A2 (en) 2013-11-18 2014-11-12 Synthetic membrane-receiver complexes

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/738,414 Continuation US9644180B2 (en) 2013-11-18 2015-06-12 Synthetic membrane-receiver complexes

Publications (1)

Publication Number Publication Date
US20150182588A1 true US20150182588A1 (en) 2015-07-02

Family

ID=52293163

Family Applications (20)

Application Number Title Priority Date Filing Date
US14/581,486 Abandoned US20150182588A1 (en) 2013-11-18 2014-12-23 Synthetic membrane-receiver complexes
US14/738,414 Active US9644180B2 (en) 2013-11-18 2015-06-12 Synthetic membrane-receiver complexes
US15/157,006 Abandoned US20160257932A1 (en) 2013-11-18 2016-05-17 Genetically engineered enucleated erythroid cells comprising a phenylalanine ammonia lyase receiver polypeptide
US15/473,421 Active US10344263B2 (en) 2013-11-18 2017-03-29 Synthetic membrane-receiver complexes
US15/783,921 Active US10329531B2 (en) 2013-11-18 2017-10-13 Synthetic membrane-receiver complexes
US15/824,877 Active US10253296B2 (en) 2013-11-18 2017-11-28 Synthetic membrane-receiver complexes
US15/900,029 Abandoned US20180187154A1 (en) 2013-11-18 2018-02-20 Synthetic membrane-receiver complexes
US15/899,895 Abandoned US20180187153A1 (en) 2013-11-18 2018-02-20 Synthetic membrane-receiver complexes
US15/925,212 Active US10301593B2 (en) 2013-11-18 2018-03-19 Synthetic membrane-receiver complexes
US15/940,069 Abandoned US20180265847A1 (en) 2013-11-18 2018-03-29 Synthetic membrane-receiver complexes
US15/940,215 Abandoned US20180216067A1 (en) 2013-11-18 2018-03-29 Synthetic membrane-receiver complexes
US16/195,811 Active US10301594B1 (en) 2013-11-18 2018-11-19 Synthetic membrane-receiver complexes
US16/409,573 Abandoned US20190264177A1 (en) 2013-11-18 2019-05-10 Synthetic membrane-receiver complexes
US16/409,576 Expired - Fee Related US10557119B2 (en) 2013-11-18 2019-05-10 Erythroid cells comprising phenylalanine ammonia lyase
US16/426,905 Abandoned US20190316091A1 (en) 2013-11-18 2019-05-30 Erythroid cells comprising arginase
US16/426,880 Abandoned US20190316090A1 (en) 2013-11-18 2019-05-30 Erythroid cells comprising arginine deiminase
US16/431,236 Abandoned US20190309261A1 (en) 2013-11-18 2019-06-04 Erythroid cells comprising lysine oxidase
US16/431,270 Abandoned US20190309262A1 (en) 2013-11-18 2019-06-04 Erythroid cells comprising serine dehydratase
US16/549,840 Abandoned US20190376034A1 (en) 2013-11-18 2019-08-23 Platelets comprising exogenous polypeptides and uses thereof
US16/572,129 Abandoned US20200002674A1 (en) 2013-11-18 2019-09-16 Erythroid cells comprising phenylalanine hydroxylase

Family Applications After (19)

Application Number Title Priority Date Filing Date
US14/738,414 Active US9644180B2 (en) 2013-11-18 2015-06-12 Synthetic membrane-receiver complexes
US15/157,006 Abandoned US20160257932A1 (en) 2013-11-18 2016-05-17 Genetically engineered enucleated erythroid cells comprising a phenylalanine ammonia lyase receiver polypeptide
US15/473,421 Active US10344263B2 (en) 2013-11-18 2017-03-29 Synthetic membrane-receiver complexes
US15/783,921 Active US10329531B2 (en) 2013-11-18 2017-10-13 Synthetic membrane-receiver complexes
US15/824,877 Active US10253296B2 (en) 2013-11-18 2017-11-28 Synthetic membrane-receiver complexes
US15/900,029 Abandoned US20180187154A1 (en) 2013-11-18 2018-02-20 Synthetic membrane-receiver complexes
US15/899,895 Abandoned US20180187153A1 (en) 2013-11-18 2018-02-20 Synthetic membrane-receiver complexes
US15/925,212 Active US10301593B2 (en) 2013-11-18 2018-03-19 Synthetic membrane-receiver complexes
US15/940,069 Abandoned US20180265847A1 (en) 2013-11-18 2018-03-29 Synthetic membrane-receiver complexes
US15/940,215 Abandoned US20180216067A1 (en) 2013-11-18 2018-03-29 Synthetic membrane-receiver complexes
US16/195,811 Active US10301594B1 (en) 2013-11-18 2018-11-19 Synthetic membrane-receiver complexes
US16/409,573 Abandoned US20190264177A1 (en) 2013-11-18 2019-05-10 Synthetic membrane-receiver complexes
US16/409,576 Expired - Fee Related US10557119B2 (en) 2013-11-18 2019-05-10 Erythroid cells comprising phenylalanine ammonia lyase
US16/426,905 Abandoned US20190316091A1 (en) 2013-11-18 2019-05-30 Erythroid cells comprising arginase
US16/426,880 Abandoned US20190316090A1 (en) 2013-11-18 2019-05-30 Erythroid cells comprising arginine deiminase
US16/431,236 Abandoned US20190309261A1 (en) 2013-11-18 2019-06-04 Erythroid cells comprising lysine oxidase
US16/431,270 Abandoned US20190309262A1 (en) 2013-11-18 2019-06-04 Erythroid cells comprising serine dehydratase
US16/549,840 Abandoned US20190376034A1 (en) 2013-11-18 2019-08-23 Platelets comprising exogenous polypeptides and uses thereof
US16/572,129 Abandoned US20200002674A1 (en) 2013-11-18 2019-09-16 Erythroid cells comprising phenylalanine hydroxylase

Country Status (8)

Country Link
US (20) US20150182588A1 (de)
EP (1) EP3071515A2 (de)
JP (3) JP6702866B2 (de)
AU (2) AU2014348683B2 (de)
BR (1) BR112016011195A2 (de)
CA (1) CA2930665A1 (de)
IL (2) IL245671B (de)
WO (1) WO2015073587A2 (de)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170122944A1 (en) * 2015-10-30 2017-05-04 Genentech, Inc. Methods of measuring factor d activity and potency of factor d inhibitors
US20180372758A1 (en) * 2015-12-24 2018-12-27 Koninklijke Philips N.V. Method and a system for determinations of cell suspensions
EP3449935A1 (de) 2017-08-31 2019-03-06 Erytech Pharma In erythrozyten eingekapselte arginin-deiminase und deren verwendung zur behandlung von krebs und arginase-1-mangel
WO2019042628A1 (en) 2017-08-31 2019-03-07 Erytech Pharma ARGININE DEIMINASE ENCAPSULATED WITHIN ERYTHROCYTES AND THEIR USE IN THE TREATMENT OF CANCER AND ARGINASE-1 DEFICIENCY
CN109580990A (zh) * 2017-09-28 2019-04-05 中国医学科学院基础医学研究所 一种采用原子力显微镜检测细胞表面孔洞的方法
US10253296B2 (en) 2013-11-18 2019-04-09 Rubius Therapeutics, Inc. Synthetic membrane-receiver complexes
US10425485B2 (en) * 2016-11-21 2019-09-24 Cisco Technology, Inc. Integrating information centric networking (ICN) over low power and lossy networks (LLNs)
US10456421B2 (en) 2016-01-11 2019-10-29 Rubius Therapeutics, Inc. Compositions and methods related to engineered erythoid cells comprising 4-1BBL
CN110520522A (zh) * 2017-02-17 2019-11-29 鲁比厄斯治疗法股份有限公司 功能化红系细胞
WO2020123662A3 (en) * 2018-12-11 2020-07-23 AdMIRx Inc. Fusion protein constructs for complement associated disease
GB2583560A (en) * 2018-12-11 2020-11-04 Admirx Inc Fusion protein constructs for complement associated disease
US10851343B2 (en) 2015-10-14 2020-12-01 Megakaryon Corporation Method for producing purified platelets
US10869898B2 (en) 2014-04-01 2020-12-22 Rubius Therapeutics, Inc. Methods and compositions for immunomodulation
US20210369858A1 (en) * 2017-08-16 2021-12-02 Board Of Regents, The University Of Texas System Use of exosomes for targeted delivery of therapeutic agents
US11753682B2 (en) 2016-03-07 2023-09-12 Father Flanagan's Boys'Home Noninvasive molecular controls
US20230349073A1 (en) * 2016-03-09 2023-11-02 Molecular Stethoscope, Inc. Methods and systems for detecting tissue conditions
CN117025553A (zh) * 2023-10-09 2023-11-10 深圳市梓健生物科技有限公司 一种在非超低温下保持病毒活力的病毒保存液及其制备方法
US12077795B2 (en) 2020-08-10 2024-09-03 The Research Foundation For The State University Of New York Method for biocatalytic protein-oligonucleotide conjugation

Families Citing this family (123)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10689609B2 (en) 2012-03-15 2020-06-23 Flodesign Sonics, Inc. Acoustic bioreactor processes
US9752113B2 (en) 2012-03-15 2017-09-05 Flodesign Sonics, Inc. Acoustic perfusion devices
US9950282B2 (en) 2012-03-15 2018-04-24 Flodesign Sonics, Inc. Electronic configuration and control for acoustic standing wave generation
US10322949B2 (en) 2012-03-15 2019-06-18 Flodesign Sonics, Inc. Transducer and reflector configurations for an acoustophoretic device
US9458450B2 (en) 2012-03-15 2016-10-04 Flodesign Sonics, Inc. Acoustophoretic separation technology using multi-dimensional standing waves
US9745548B2 (en) 2012-03-15 2017-08-29 Flodesign Sonics, Inc. Acoustic perfusion devices
US10967298B2 (en) 2012-03-15 2021-04-06 Flodesign Sonics, Inc. Driver and control for variable impedence load
US10704021B2 (en) 2012-03-15 2020-07-07 Flodesign Sonics, Inc. Acoustic perfusion devices
US10737953B2 (en) 2012-04-20 2020-08-11 Flodesign Sonics, Inc. Acoustophoretic method for use in bioreactors
US9745569B2 (en) 2013-09-13 2017-08-29 Flodesign Sonics, Inc. System for generating high concentration factors for low cell density suspensions
EP3092049A1 (de) 2014-01-08 2016-11-16 Flodesign Sonics Inc. Akustophoresevorrichtung mit doppelter akustophoresekammer
US9744483B2 (en) 2014-07-02 2017-08-29 Flodesign Sonics, Inc. Large scale acoustic separation device
US11708572B2 (en) 2015-04-29 2023-07-25 Flodesign Sonics, Inc. Acoustic cell separation techniques and processes
US11021699B2 (en) 2015-04-29 2021-06-01 FioDesign Sonics, Inc. Separation using angled acoustic waves
US11377651B2 (en) 2016-10-19 2022-07-05 Flodesign Sonics, Inc. Cell therapy processes utilizing acoustophoresis
US11474085B2 (en) 2015-07-28 2022-10-18 Flodesign Sonics, Inc. Expanded bed affinity selection
US11459540B2 (en) 2015-07-28 2022-10-04 Flodesign Sonics, Inc. Expanded bed affinity selection
US11360097B2 (en) 2015-11-12 2022-06-14 Edward J Goetzl Platelet biomarkers and diagnostic methods for vascular diseases
US11377481B2 (en) * 2015-12-22 2022-07-05 The Trustees Of The University Of Pennsylvania SpyCatcher and SpyTag: universal immune receptors for T cells
US11083702B2 (en) * 2016-04-19 2021-08-10 University of Pittsburgh—of the Commonwealth System of Higher Education Anaplerotic agents for treatment of disorders of propionate and long chain fat metabolism
WO2017190056A1 (en) * 2016-04-28 2017-11-02 William Marsh Rice University Conversion of 1-carbon compounds to products
US11214789B2 (en) 2016-05-03 2022-01-04 Flodesign Sonics, Inc. Concentration and washing of particles with acoustics
US11085035B2 (en) 2016-05-03 2021-08-10 Flodesign Sonics, Inc. Therapeutic cell washing, concentration, and separation utilizing acoustophoresis
WO2017196798A1 (en) * 2016-05-09 2017-11-16 Chang Jae-Hyung Robert Accelerated method for preparing platelet rich plasma
EP3481943A1 (de) * 2016-07-07 2019-05-15 Rubius Therapeutics, Inc. Zusammensetzungen und verfahren im zusammenhang mit therapeutischen zellsystemen mit expression von exogener rna
US10738338B2 (en) 2016-10-18 2020-08-11 The Research Foundation for the State University Method and composition for biocatalytic protein-oligonucleotide conjugation and protein-oligonucleotide conjugate
WO2018075830A1 (en) 2016-10-19 2018-04-26 Flodesign Sonics, Inc. Affinity cell extraction by acoustics
EP3548050A1 (de) * 2016-12-02 2019-10-09 Rubius Therapeutics, Inc. Zusammensetzungen und verfahren in zusammenhang mit zellsystemen zum durchdringen von soliden tumoren
SG11201906880VA (en) 2017-02-13 2019-08-27 Codexis Inc Engineered phenylalanine ammonia lyase polypeptides
MA47494A (fr) * 2017-02-17 2019-12-25 Ose Immunotherapeutics Nouvelles utilisations d'anticorps anti-sirpg
US20200172868A1 (en) 2017-07-19 2020-06-04 Rubius Therapeutics, Inc. Compositions and methods related to multimodal therapeutic cell systems for infectious disease
WO2019017937A1 (en) 2017-07-19 2019-01-24 Rubius Therapeutics, Inc. COMPOSITIONS AND METHODS ASSOCIATED WITH MULTIMODAL THERAPEUTIC CELLULAR SYSTEMS FOR THE TREATMENT OF CARDIOMETABOLIC DISEASE
US10960071B2 (en) 2017-08-07 2021-03-30 The Regents Of The University Of California Platform for generating safe cell therapeutics
US20200368285A1 (en) * 2017-08-07 2020-11-26 The Regents Of The University Of California Platform for generating safe cell therapeutics
EP3672602A1 (de) 2017-08-22 2020-07-01 Rubius Therapeutics, Inc. Lipidnanopartikelverfahren und zusammensetzungen zur herstellung von gentechnisch veränderten erythroiden zellen
US11708559B2 (en) 2017-10-27 2023-07-25 The Children's Hospital Of Philadelphia Engineered red blood cells having rare antigen phenotypes
EP3703751A2 (de) 2017-11-03 2020-09-09 Rubius Therapeutics, Inc. Zusammensetzungen und verfahren in zusammenhang mit therapeutischen zellsystemen zur hemmung des tumorwachstums
WO2019118921A1 (en) 2017-12-14 2019-06-20 Flodesign Sonics, Inc. Acoustic transducer drive and controller
RU2763798C1 (ru) 2017-12-23 2022-01-11 Рубиус Терапьютикс, Инк. Искусственные антигенпрезентирующие клетки и способы их применения
WO2019133881A1 (en) 2017-12-29 2019-07-04 Rubius Therapeutics, Inc. Gene editing and targeted transcriptional modulation for enginerering erythroid cells
EP3735457A4 (de) 2018-01-05 2021-08-25 Platelet Biogenesis, Inc. Zusammensetzungen und verfahren zur herstellung von megakaryozyten
WO2019140116A2 (en) 2018-01-10 2019-07-18 Rubius Therapeutics, Inc. Amplifiable rnas for therapeutic cell systems
JP7281633B2 (ja) * 2018-01-29 2023-05-26 地方独立行政法人東京都健康長寿医療センター 平均赤血球年齢を決定する方法
CA3092062A1 (en) 2018-02-22 2019-08-29 Entrada Therapeutics, Inc. Compositions and methods for treating mitochondrial neurogastrointestinal encephalopathy
RU2020132924A (ru) 2018-03-08 2022-04-11 Рубиус Терапьютикс, Инк. Терапевтические клеточные системы и способы лечения рака и инфекционных заболеваний
WO2019183292A1 (en) 2018-03-20 2019-09-26 Rubius Therapeutics, Inc. Therapeutic cell systems and methods for treating hyperuricemia and gout
US20190309271A1 (en) 2018-03-20 2019-10-10 Rubius Therapeutics, Inc. Therapeutic cell systems and methods for treating homocystinuria
EP3790890A4 (de) 2018-05-09 2022-03-02 Ohio State Innovation Foundation Zyklische, zellpenetrierende peptide mit einem oder mehreren hydrophoben resten
US11028425B2 (en) 2018-06-08 2021-06-08 Glympse Bio, Inc. Diagnosis and monitoring of liver disease
US11732009B2 (en) 2018-06-08 2023-08-22 Glympse Bio, Inc. Activity sensor with tunable analyte
US20230242640A1 (en) * 2018-08-22 2023-08-03 Ose Immunotherapeutics ANTI-SIRPg Compounds
WO2020092355A2 (en) * 2018-10-29 2020-05-07 President And Fellows Of Harvard College Nanobody-glycan modifying enzyme fusion proteins and uses thereof
EP3887402A4 (de) * 2018-11-30 2022-10-26 Cellphire, Inc. Thrombozyten als freisetzungsmittel
EP3886879A4 (de) * 2018-11-30 2022-12-07 Cellphire Inc. Thrombozyten als freisetzungsmittel
CA3121258A1 (en) 2018-12-03 2020-06-11 Rubius Therapeutics, Inc. Artificial antigen presenting cells including hla-e and hla-g molecules and methods of use
US11166996B2 (en) 2018-12-12 2021-11-09 Flagship Pioneering Innovations V, Inc. Anellovirus compositions and methods of use
CA3125567A1 (en) 2019-01-18 2020-07-23 Flagship Pioneering, Inc. Trem compositions and uses thereof
AU2020224662A1 (en) 2019-02-20 2021-09-09 Rubius Therapeutics, Inc. Engineered erythroid cells including loadable antigen-presenting polypeptides and methods of use
CA3128626A1 (en) 2019-03-04 2020-09-10 Flagship Pioneering Innovations Vi, Llc Circular polyribonucleotides and pharmaceutical compositions thereof
KR20210142678A (ko) 2019-03-25 2021-11-25 플래그쉽 파이어니어링 이노베이션스 브이아이, 엘엘씨 변형된 원형 폴리리보뉴클레오티드를 포함하는 조성물 및 그의 용도
US20200338129A1 (en) 2019-04-26 2020-10-29 Rubius Therapeutics, Inc. Compositions including enucleated erythroid cells
KR20220016092A (ko) 2019-05-03 2022-02-08 셀파이어, 인크. 혈액 생성물을 생산하기 위한 물질 및 방법
US11162079B2 (en) 2019-05-10 2021-11-02 The Regents Of The University Of California Blood type O Rh-hypo-immunogenic pluripotent cells
AU2020283752A1 (en) 2019-05-24 2021-12-23 Rubius Therapeutics, Inc. Methods of generating enucleated erythroid cells
EP3976782A1 (de) 2019-05-31 2022-04-06 Flagship Pioneering, Inc. Verwendung von trem-zusammensetzungen zur modulation von trna-pools
JP2022537154A (ja) 2019-06-14 2022-08-24 フラッグシップ パイオニアリング イノベーションズ シックス,エルエルシー 細胞治療のための環状rna
EP3986469A1 (de) 2019-06-19 2022-04-27 Flagship Pioneering Innovations VI, LLC Zusammensetzungen mit kreisförmigen polyribonukleotiden zur proteinmodulation und deren verwendungen
EP4013496A4 (de) 2019-08-16 2023-10-18 Cellphire Inc. Thrombosomen als mittel zur aufhebung der thrombozytenaggregationshemmenden wirkung
CN110941181B (zh) * 2019-08-29 2023-01-20 广东工业大学 一种压电陶瓷连接的刚柔耦合运动平台控制方法
US20220330541A1 (en) 2019-09-05 2022-10-20 Hemanext Inc. Methods for the Preservation of Reagent Red Blood Cells Using Carbon Monoxide
EP4045648A4 (de) * 2019-10-15 2023-11-22 The Regents of The University of California Transplantierter zellschutz über fc-sequestrierung
EP4055148A1 (de) 2019-11-04 2022-09-14 Rubius Therapeutics, Inc. Verfahren zum erzeugen von enukelierten erythroidenzellen unter verwendung von taurin oder hypotaurin
WO2021092073A1 (en) 2019-11-04 2021-05-14 Flagship Pioneering, Inc. Methods of modifying a nucleic acid sequence
KR20220110192A (ko) 2019-11-04 2022-08-05 루비우스 테라퓨틱스, 아이엔씨. 미오-이노시톨을 사용하여 제혁 적혈구를 생성하는 방법
CN115003810A (zh) 2019-11-04 2022-09-02 旗舰创业股份有限公司 用于con-稀有密码子的trem组合物及相关用途
GB201918586D0 (en) 2019-12-17 2020-01-29 Patterson James Engineered platelets for targeted delivery of a therapeutic agent
TW202142239A (zh) 2020-01-29 2021-11-16 美商旗艦先鋒創新有限責任公司 包含環狀多核糖核苷酸之組合物之遞送
CN115279415A (zh) 2020-01-29 2022-11-01 旗舰创业创新第六有限责任公司 用于蛋白调节的包含线性多核糖核苷酸的组合物及其用途
TW202142689A (zh) 2020-01-29 2021-11-16 美商旗艦先鋒創新有限責任公司 用於轉譯之組合物及其使用方法
WO2021158646A1 (en) 2020-02-04 2021-08-12 Cellphire, Inc. Treatment of von willebrand disease
EP4103696A1 (de) 2020-02-10 2022-12-21 Rubius Therapeutics, Inc. Gentechnisch veränderte erythroide zellen mit hla-g-polypeptiden und verfahren zu deren verwendung
WO2021183818A1 (en) * 2020-03-12 2021-09-16 University Of Florida Research Foundation, Incorporated Recombinant oxalate oxidase and uses thereof
EP4125947A4 (de) * 2020-03-27 2024-06-12 Platelet Biogenesis, Inc. Neuartige anuclierte zellen zur behandlung von krankheiten
US20230203192A1 (en) 2020-05-20 2023-06-29 Flagship Pioneering, Inc. Compositions and methods for producing human polyclonal antibodies
JP2023526422A (ja) 2020-05-20 2023-06-21 フラッグシップ パイオニアリング イノベーションズ シックス,エルエルシー コロナウイルス抗原組成物及びそれらの使用
MX2022014607A (es) 2020-05-20 2023-03-15 Flagship Pioneering Innovations Vi Llc Composiciones inmunogenicas y usos de las mismas.
EP4158032A2 (de) 2020-05-29 2023-04-05 Flagship Pioneering Innovations VI, LLC Trem-zusammensetzungen und damit in zusammenhang stehende verfahren
WO2021243290A1 (en) 2020-05-29 2021-12-02 Flagship Pioneering Innovations Vi, Llc Trem compositions and methods relating thereto
US20230233514A1 (en) 2020-06-23 2023-07-27 Flagship Pioneering, Inc. Antiviral compounds and methods of using the same
MX2023002439A (es) 2020-09-03 2023-05-09 Flagship Pioneering Innovations Vi Llc Composiciones immunogénicas y usos de las mismas.
US20240175020A1 (en) 2020-12-23 2024-05-30 Flagship Pioneering Innovations Vi, Llc Compositions of modified trems and uses thereof
WO2022150569A1 (en) 2021-01-08 2022-07-14 Rubius Therapeutics, Inc. METHODS OF INCREASING NKp30-POSITIVE LYMPHOCYTES IN A SUBJECT AND USES THEREOF
WO2022150578A1 (en) 2021-01-08 2022-07-14 Rubius Therapeutics, Inc. Methods of treating a tumor in a human subject
WO2022197548A1 (en) 2021-03-14 2022-09-22 Rubius Therapeutics, Inc. Methods of increasing nkg2d-positive lymphocytes in a subject and uses thereof
KR20230165276A (ko) 2021-03-31 2023-12-05 플래그쉽 파이어니어링 이노베이션스 브이, 인크. 타노트랜스미션 폴리펩티드 및 암의 치료에서의 이의 용도
TW202317179A (zh) 2021-06-03 2023-05-01 美商盧比亞斯治療公司 在個體中治療hpv16-陽性或hpv16-相關的癌症之方法
EP4377457A1 (de) 2021-07-26 2024-06-05 Flagship Pioneering Innovations VI, LLC Trem-zusammensetzungen und verwendungen davon
KR20240099185A (ko) 2021-09-17 2024-06-28 플래그쉽 파이어니어링 이노베이션스 브이아이, 엘엘씨 원형 폴리리보뉴클레오티드를 생성하기 위한 조성물 및 방법
WO2023044774A1 (en) * 2021-09-24 2023-03-30 Sichuan Clover Biopharmaceuticals, Inc. Tpo mimetic fusion proteins and methods of use submission of sequence listing as ascii text file
EP4419706A1 (de) 2021-10-18 2024-08-28 Flagship Pioneering Innovations VI, LLC Zusammensetzungen und verfahren zur reinigung von polyribonukleotiden
AU2022398478A1 (en) 2021-11-24 2024-06-13 Flagship Pioneering Innovations Vi, Llc Coronavirus immunogen compositions and their uses
IL312799A (en) 2021-11-24 2024-07-01 Flagship Pioneering Innovations Vi Llc Immunogenic compositions of varicella-zoster virus and uses thereof
WO2023097003A2 (en) 2021-11-24 2023-06-01 Flagship Pioneering Innovations Vi, Llc Immunogenic compositions and their uses
KR20240117149A (ko) 2021-12-22 2024-07-31 플래그쉽 파이어니어링 이노베이션스 브이아이, 엘엘씨 폴리리보뉴클레오티드를 정제하기 위한 조성물 및 방법
CA3241026A1 (en) 2021-12-23 2023-06-29 Flagship Pioneering Innovations Vi, Llc Circular polyribonucleotides encoding antifusogenic polypeptides
WO2023220083A1 (en) 2022-05-09 2023-11-16 Flagship Pioneering Innovations Vi, Llc Trem compositions and methods of use for treating proliferative disorders
TW202409283A (zh) 2022-05-13 2024-03-01 美商旗艦先鋒創新有限責任(Vii)公司 雙股dna組合物及相關方法
WO2023230549A2 (en) 2022-05-25 2023-11-30 Flagship Pioneering Innovations Vii, Llc Compositions and methods for modulation of tumor suppressors and oncogenes
WO2023230573A2 (en) 2022-05-25 2023-11-30 Flagship Pioneering Innovations Vii, Llc Compositions and methods for modulation of immune responses
WO2023230570A2 (en) 2022-05-25 2023-11-30 Flagship Pioneering Innovations Vii, Llc Compositions and methods for modulating genetic drivers
WO2023230578A2 (en) 2022-05-25 2023-11-30 Flagship Pioneering Innovations Vii, Llc Compositions and methods for modulating circulating factors
WO2023230566A2 (en) 2022-05-25 2023-11-30 Flagship Pioneering Innovations Vii, Llc Compositions and methods for modulating cytokines
WO2023250112A1 (en) 2022-06-22 2023-12-28 Flagship Pioneering Innovations Vi, Llc Compositions of modified trems and uses thereof
US20240174732A1 (en) 2022-10-05 2024-05-30 Flagship Pioneering Innovations V, Inc. Nucleic acid molecules encoding trif and additional polypeptides and their use in treating cancer
WO2024097664A1 (en) 2022-10-31 2024-05-10 Flagship Pioneering Innovations Vi, Llc Compositions and methods for purifying polyribonucleotides
WO2024102799A1 (en) 2022-11-08 2024-05-16 Flagship Pioneering Innovations Vi, Llc Compositions and methods for producing circular polyribonucleotides
WO2024129988A1 (en) 2022-12-14 2024-06-20 Flagship Pioneering Innovations Vii, Llc Compositions and methods for delivery of therapeutic agents to bone
US20240252520A1 (en) 2023-01-09 2024-08-01 Beth Israel Deaconess Medical Center, Inc. Therapeutic agents and their use for treating chronic wounds
US20240269251A1 (en) 2023-01-09 2024-08-15 Flagship Pioneering Innovations V, Inc. Genetic switches and their use in treating cancer
US20240238473A1 (en) 2023-01-09 2024-07-18 Beth Israel Deaconess Medical Center, Inc. Recombinant nucleic acid molecules and their use in wound healing
WO2024173836A2 (en) 2023-02-17 2024-08-22 Flagship Pioneering Innovations Vii, Llc Dna compositions comprising modified cytosine
US20240285805A1 (en) 2023-02-17 2024-08-29 Flagship Pioneering Innovations Vii, Llc Dna compositions comprising modified uracil
WO2024173945A1 (en) * 2023-02-17 2024-08-22 Abbott Diabetes Care Inc. Methods for single cell analysis of glucose internalization

Family Cites Families (82)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4327710A (en) 1980-06-18 1982-05-04 The United States Of America As Represented By The Secretary Of Agriculture Process for encapsulating additives in resealed erythrocytes for disseminating chemicals via the circulatory system
IL86650A0 (en) 1987-06-30 1988-11-30 Biophor Corp Animal derived cells and liposomes,having an antigenic protein incorporated into their membrane
US5258499A (en) 1988-05-16 1993-11-02 Vestar, Inc. Liposome targeting using receptor specific ligands
US4935223A (en) 1988-08-04 1990-06-19 Board Of Regents, The University Of Texas System Labeled cells for use in imaging
US5676954A (en) 1989-11-03 1997-10-14 Vanderbilt University Method of in vivo delivery of functioning foreign genes
EP0517986B1 (de) 1991-06-14 1997-09-10 Communaute Economique Europeenne (Cee) Transformierte Erythrozyten, Verfahren zu deren Herstellung, und ihre Verwendung in pharmazeutischen Zusammensetzungen
US5830462A (en) 1993-02-12 1998-11-03 President & Fellows Of Harvard College Regulated transcription of targeted genes and other biological events
US5599705A (en) 1993-11-16 1997-02-04 Cameron; Robert B. In vitro method for producing differentiated universally compatible mature human blood cells
US6984379B1 (en) 1994-04-08 2006-01-10 Children's Hospital of LosAngeles Gene therapy by administration of genetically engineered CD34+ cells obtained from cord blood
US6132764A (en) 1994-08-05 2000-10-17 Targesome, Inc. Targeted polymerized liposome diagnostic and treatment agents
US5648248A (en) 1994-12-30 1997-07-15 Boehringer Ingelheim International Gmbh Methods for producing differentiated cells from immature hematopoietic cells
US6060052A (en) 1995-10-30 2000-05-09 Systemix, Inc. Methods for use of Mpl ligands with primitive human hematopoietic stem cells
WO1998016201A1 (en) 1996-10-11 1998-04-23 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method
TW520297B (en) 1996-10-11 2003-02-11 Sequus Pharm Inc Fusogenic liposome composition and method
JP2001505563A (ja) 1996-11-21 2001-04-24 ニューヨーク・ブラッド・センター・インコーポレイテッド 血液型の変換方法
EP0882448B1 (de) 1997-05-05 2005-01-12 DIDECO S.r.l. Verfahren zur Verkapselung von biologisch aktiven Stoffen in Erythrocyten und Gerät dafür
US6361998B1 (en) 1998-06-25 2002-03-26 Hemosol Inc. Efficient culture of stem cells for the production of hemoglobin
US6197526B1 (en) 1999-01-04 2001-03-06 Dyax Corp. Polypeptides for binding human factor VIII and fragments of human factor VIII
US7109170B2 (en) 1999-06-16 2006-09-19 Saint Louis University Region of factor IXa protease domain that interacts with factor VIIIa and methods therefor
US6624289B1 (en) 1999-06-16 2003-09-23 Saint Louis University Region of factor IXa protease domain that interacts with factor VIIIa and methods therefor
EP1196432A2 (de) 1999-07-02 2002-04-17 Genentech, Inc. Antagonisten des faktor viia
AT411997B (de) 1999-09-14 2004-08-26 Baxter Ag Faktor ix/faktor ixa aktivierende antikörper und antikörper-derivate
US6495351B2 (en) 2000-02-08 2002-12-17 Gendel Limited Loading system and method for using the same
US20020151004A1 (en) 2000-07-24 2002-10-17 Roger Craig Delivery vehicles and methods for using the same
US7767202B2 (en) 2001-03-16 2010-08-03 The Johns Hopkins University Modulation of systemic immune responses by transplantation of hematopoietic stem cells transduced with genes encoding antigens and antigen presenting cell regulatory molecules
US20030207247A1 (en) 2001-12-05 2003-11-06 Cerus Corporation Preparation of red blood cells having reduced immunogenicity
US20030133922A1 (en) 2002-01-15 2003-07-17 Kasha John R. Oral tolerance using allogeneic platelets in ITP
US7427603B2 (en) 2002-09-26 2008-09-23 The Children's Medical Center Corporation Method of enhancing proliferation and/or hematopoietic differentiation of stem cells
ES2348868T3 (es) 2002-12-13 2010-12-16 Genetix Pharmaceuticals Inc. Vectores retrovirales terapeuticos para terapia genica.
DE10335833A1 (de) * 2003-08-05 2005-03-03 Curevac Gmbh Transfektion von Blutzellen mit mRNA zur Immunstimulation und Gentherapie
JP5139799B2 (ja) 2004-06-04 2013-02-06 ユニベルシテ ピエール エ マリー キュリー(パリ シズエム) 赤血球細胞の生産方法
EP1763576A2 (de) 2004-06-15 2007-03-21 Baxter International Inc. Ex-vivo anwendung von festen micropartikeln mit therapeutischen mitteln
FR2873925B1 (fr) 2004-08-05 2006-10-13 Erytech Pharma Soc Par Actions Procede et dispositif de lyse-rescellement pour l'incorporation de principe actif notamment asparaginase ou inositol hexaphosphate, dans des erythrocytes
US7343146B2 (en) * 2004-08-13 2008-03-11 Nokia Corporation Single chip LNA and VCO having similar resonant circuit topology and using same calibration signal to compensate for process variations
DE102004054536A1 (de) 2004-11-06 2006-05-11 Capsulution Nanoscience Ag Multimodal veränderte Zellen als zellulare Darreichungsformen für aktive Substanzen und als diagnostische Zellpartikel
FR2884717B1 (fr) 2005-04-25 2009-07-03 Erytech Pharma Soc Par Actions Erythrocytes renfermant de l'arginine deiminase
SI1926813T2 (sl) 2005-09-23 2019-11-29 Tigenix S A U Celična populacija z imunoregulatorno aktivnostjo, postopek za izolacijo in uporabe
FR2891843A1 (fr) 2005-10-06 2007-04-13 Erytech Pharma Soc Par Actions Erythrocytes contenant du 5-fluorouracile
US7462485B2 (en) * 2005-10-07 2008-12-09 Glaser Lawrence F Modified erythrocytes and uses thereof
US20070243137A1 (en) 2006-04-18 2007-10-18 Nanoprobes, Inc. Cell and sub-cell methods for imaging and therapy
US7534595B2 (en) * 2006-06-12 2009-05-19 Biomarin Pharmaceutical Inc. Compositions of prokaryotic phenylalanine ammonia-lyase and methods of using compositions thereof
US7531341B1 (en) 2006-06-12 2009-05-12 Biomarin Pharmaceutical Inc. Compositions of prokaryotic phenylalanine ammonia-lyase and methods of using compositions thereof
FR2919804B1 (fr) 2007-08-08 2010-08-27 Erytech Pharma Composition et vaccin therapeutique anti-tumoral
US7560263B2 (en) 2007-08-17 2009-07-14 Biomarin Pharmaceutical Inc. Compositions of prokaryotic phenylalanine ammonia-lyase and methods of treating cancer using compositions thereof
FR2925339B1 (fr) 2007-12-24 2010-03-05 Erytech Pharma Medicament pour le traitement du cancer du pancreas
CA2715379A1 (en) 2008-02-13 2009-08-20 Erytech Pharma Formulation and method for the prevention and treatment of skeletal manifestation of gaucher's disease
FR2928270B1 (fr) 2008-03-10 2011-01-21 Erytech Pharma Formulation methode pour la prevention ou le traitement des metastases osseuses et autres maladies de l'os
US8231895B2 (en) 2008-05-22 2012-07-31 Universidade De Coimbra Targeted delivery to human diseases and disorders
US8211656B2 (en) 2008-08-13 2012-07-03 The Invention Science Fund I, Llc Biological targeting compositions and methods of using the same
FR2938332B1 (fr) 2008-11-07 2011-11-25 Erytech Pharma Test predictif de la neutralisation de l'activite asparaginase
US20100216804A1 (en) * 2008-12-15 2010-08-26 Zale Stephen E Long Circulating Nanoparticles for Sustained Release of Therapeutic Agents
FR2944106B1 (fr) 2009-04-03 2012-09-28 Erytech Pharma Methode de dosage de l'inositol hexaphosphate (ihp).
CA2756833C (en) 2009-04-09 2019-11-19 Sangamo Biosciences, Inc. Targeted integration into stem cells
US20120066778A1 (en) 2009-05-19 2012-03-15 Christian Kemmer Control of uric acid homeostasis
CN102596208B (zh) 2009-10-27 2017-06-20 爱瑞泰克药物公司 用于诱导特异性免疫耐受的组合物
AU2011224257A1 (en) 2010-03-12 2012-10-11 The Regents Of The University Of California Triggered cargo release from nanoparticle stabilized liposomes
US9517257B2 (en) 2010-08-10 2016-12-13 Ecole Polytechnique Federale De Lausanne (Epfl) Erythrocyte-binding therapeutics
JP6219832B2 (ja) 2011-10-17 2017-10-25 マサチューセッツ インスティテュート オブ テクノロジー 細胞内送達
MX2014009900A (es) 2012-02-15 2015-06-02 Ecole Polytech Agentes terapeuticos de ligacion a eritrocitos.
EP3295945A1 (de) 2012-03-13 2018-03-21 HLI Cellular Therapeutics, LLC Modifizierte erythrozytenvorläuferzellen und verwendungen davon
EP2827878A1 (de) * 2012-03-21 2015-01-28 Erytech Pharma Verfahren zur behandlung von akuter myeloischer leukämie
KR20140011912A (ko) 2012-07-20 2014-01-29 도꾸리쯔교세이호징 리가가쿠 겐큐소 인간 적혈구 전구세포주 및 인간 탈핵 적혈구의 제조방법
EP2914274A4 (de) 2012-11-01 2016-07-20 Sydney Children S Hospital Network Randwick And Westmead Genetisch verändertes probiotikum zur behandlung von phenylketonurie
US9675678B2 (en) 2013-01-29 2017-06-13 The Regents Of The University Of Colorado, A Body Corporate Compositions and methods for treatment of homocystinuria
CN103224957B (zh) * 2013-04-16 2017-08-04 福建三一造血技术有限公司 体外诱导生成含L‑ASPase II的红细胞药物的方法
FR3005420B1 (fr) 2013-05-07 2015-09-18 Erytech Pharma Procede de stabilisation de suspensions d'erythrocytes encapsulant un principe actif, suspensions obtenues.
EP3693398A1 (de) 2013-05-10 2020-08-12 Whitehead Institute for Biomedical Research In-vitro-herstellung von erythrozyten mit sortasefähigen proteinen
SG11201509252QA (en) 2013-05-10 2015-12-30 Erydel Spa Process for the preparation of erythrocytes loaded with one or more substances of pharmaceutical interest and so obtained erythrocytes
AU2014262469B2 (en) 2013-05-10 2019-11-14 Whitehead Institute For Biomedical Research Protein modification of living cells using sortase
EP2813234A1 (de) 2013-06-11 2014-12-17 Erytech Pharma Zusammensetzung basierend auf Erythrocyten, in die Phenylalanin-Hydroxylase verkapselt ist, und therapeutische Verwendungen davon
CA2930665A1 (en) 2013-11-18 2015-05-21 Rubius Therapeutics, Inc. Synthetic membrane-receiver complexes
FR3017299B1 (fr) 2014-02-12 2018-05-18 Erytech Pharma Composition pharmaceutique comprenant des erythrocytes encapsulant une enzyme a plp et son cofacteur
KR20170005801A (ko) 2014-04-01 2017-01-16 루비우스 테라퓨틱스, 아이엔씨. 면역조절을 위한 방법 및 조성물
US20180135012A1 (en) 2015-05-13 2018-05-17 Rubius Therapeutics, Inc. Membrane-receiver complex therapeutics
EP3187190A1 (de) 2015-12-31 2017-07-05 Erytech Pharma Verfahren zur behandlung eines säugetiers, einschliesslich eines menschen, gegen krebs mittels methionin- und asparagindepletion
CN108697733A (zh) 2016-01-11 2018-10-23 鲁比厄斯治疗法股份有限公司 与癌症适应症的多模式治疗性细胞体系有关的组合物和方法
EP3481943A1 (de) 2016-07-07 2019-05-15 Rubius Therapeutics, Inc. Zusammensetzungen und verfahren im zusammenhang mit therapeutischen zellsystemen mit expression von exogener rna
EP3548050A1 (de) 2016-12-02 2019-10-09 Rubius Therapeutics, Inc. Zusammensetzungen und verfahren in zusammenhang mit zellsystemen zum durchdringen von soliden tumoren
CA3052142A1 (en) 2017-02-17 2018-08-23 Rubius Therapeutics, Inc. Functionalized erythroid cells
EP3672602A1 (de) 2017-08-22 2020-07-01 Rubius Therapeutics, Inc. Lipidnanopartikelverfahren und zusammensetzungen zur herstellung von gentechnisch veränderten erythroiden zellen
EP3703751A2 (de) 2017-11-03 2020-09-09 Rubius Therapeutics, Inc. Zusammensetzungen und verfahren in zusammenhang mit therapeutischen zellsystemen zur hemmung des tumorwachstums
WO2019133881A1 (en) 2017-12-29 2019-07-04 Rubius Therapeutics, Inc. Gene editing and targeted transcriptional modulation for enginerering erythroid cells

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10557119B2 (en) 2013-11-18 2020-02-11 Rubius Therapeutics, Inc. Erythroid cells comprising phenylalanine ammonia lyase
US10253296B2 (en) 2013-11-18 2019-04-09 Rubius Therapeutics, Inc. Synthetic membrane-receiver complexes
US10301594B1 (en) 2013-11-18 2019-05-28 Rubius Therapeutics, Inc Synthetic membrane-receiver complexes
US10301593B2 (en) 2013-11-18 2019-05-28 Rubius Therapeutics, Inc. Synthetic membrane-receiver complexes
US10329531B2 (en) 2013-11-18 2019-06-25 Rubius Therapeutics, Inc. Synthetic membrane-receiver complexes
US10344263B2 (en) 2013-11-18 2019-07-09 Rubius Therapeutics, Inc. Synthetic membrane-receiver complexes
US11576934B2 (en) 2014-04-01 2023-02-14 Rubius Therapeutics, Inc. Methods and compositions for immunomodulation
US11554141B2 (en) 2014-04-01 2023-01-17 Rubius Therapeutics, Inc. Methods and compositions for immunomodulation
US10869898B2 (en) 2014-04-01 2020-12-22 Rubius Therapeutics, Inc. Methods and compositions for immunomodulation
US10851343B2 (en) 2015-10-14 2020-12-01 Megakaryon Corporation Method for producing purified platelets
US10591481B2 (en) * 2015-10-30 2020-03-17 Genentech, Inc. Methods of measuring factor D activity and potency of factor D inhibitors
US20170122944A1 (en) * 2015-10-30 2017-05-04 Genentech, Inc. Methods of measuring factor d activity and potency of factor d inhibitors
US10782306B2 (en) * 2015-12-24 2020-09-22 Koninklijke Philips N.V. Method and a system for determinations of cell suspensions
US20180372758A1 (en) * 2015-12-24 2018-12-27 Koninklijke Philips N.V. Method and a system for determinations of cell suspensions
US10517897B1 (en) 2016-01-11 2019-12-31 Rubius Therapeutics, Inc. Methods related to engineered erythoid cells comprising 4-1BBL
US10568910B2 (en) 2016-01-11 2020-02-25 Rubius Therapeutics, Inc. Compositions and methods related to engineered erythroid cells comprising IL-15
US10456421B2 (en) 2016-01-11 2019-10-29 Rubius Therapeutics, Inc. Compositions and methods related to engineered erythoid cells comprising 4-1BBL
US11753682B2 (en) 2016-03-07 2023-09-12 Father Flanagan's Boys'Home Noninvasive molecular controls
US20230349073A1 (en) * 2016-03-09 2023-11-02 Molecular Stethoscope, Inc. Methods and systems for detecting tissue conditions
US10425485B2 (en) * 2016-11-21 2019-09-24 Cisco Technology, Inc. Integrating information centric networking (ICN) over low power and lossy networks (LLNs)
CN110520522A (zh) * 2017-02-17 2019-11-29 鲁比厄斯治疗法股份有限公司 功能化红系细胞
US11020435B2 (en) 2017-02-17 2021-06-01 Rubius Therapeutics, Inc. Functionalized erythroid cells
US20210369858A1 (en) * 2017-08-16 2021-12-02 Board Of Regents, The University Of Texas System Use of exosomes for targeted delivery of therapeutic agents
WO2019042628A1 (en) 2017-08-31 2019-03-07 Erytech Pharma ARGININE DEIMINASE ENCAPSULATED WITHIN ERYTHROCYTES AND THEIR USE IN THE TREATMENT OF CANCER AND ARGINASE-1 DEFICIENCY
EP3449935A1 (de) 2017-08-31 2019-03-06 Erytech Pharma In erythrozyten eingekapselte arginin-deiminase und deren verwendung zur behandlung von krebs und arginase-1-mangel
CN109580990A (zh) * 2017-09-28 2019-04-05 中国医学科学院基础医学研究所 一种采用原子力显微镜检测细胞表面孔洞的方法
US11053305B2 (en) 2018-12-11 2021-07-06 Q32 Bio Inc. Fusion protein constructs comprising anti-C3d antibody and CR1 polypeptide
US11053306B2 (en) 2018-12-11 2021-07-06 Q32 Bio Inc. Fusion protein constructs comprising anti-C3d antibody and factor H
GB2583560A (en) * 2018-12-11 2020-11-04 Admirx Inc Fusion protein constructs for complement associated disease
WO2020123662A3 (en) * 2018-12-11 2020-07-23 AdMIRx Inc. Fusion protein constructs for complement associated disease
US11879008B2 (en) 2018-12-11 2024-01-23 Q32 Bio Inc. Methods of treating complement mediated diseases with fusion protein constructs comprising anti-C3d antibody and a complement modulator
US12077795B2 (en) 2020-08-10 2024-09-03 The Research Foundation For The State University Of New York Method for biocatalytic protein-oligonucleotide conjugation
CN117025553A (zh) * 2023-10-09 2023-11-10 深圳市梓健生物科技有限公司 一种在非超低温下保持病毒活力的病毒保存液及其制备方法

Also Published As

Publication number Publication date
US20190309262A1 (en) 2019-10-10
US20160257932A1 (en) 2016-09-08
US20190309261A1 (en) 2019-10-10
US20190264177A1 (en) 2019-08-29
US20190144827A1 (en) 2019-05-16
IL281666A (en) 2021-05-31
US20180216067A1 (en) 2018-08-02
AU2020260424A1 (en) 2020-11-26
IL245671B (en) 2021-04-29
US20180187154A1 (en) 2018-07-05
BR112016011195A8 (de) 2022-11-08
US10253296B2 (en) 2019-04-09
BR112016011195A2 (pt) 2017-09-19
JP2019142950A (ja) 2019-08-29
JP6702866B2 (ja) 2020-06-03
IL245671A0 (en) 2016-06-30
JP2021130721A (ja) 2021-09-09
WO2015073587A2 (en) 2015-05-21
US9644180B2 (en) 2017-05-09
JP7116705B2 (ja) 2022-08-10
JP2017504568A (ja) 2017-02-09
EP3071515A2 (de) 2016-09-28
US20190316090A1 (en) 2019-10-17
US10329531B2 (en) 2019-06-25
US10301593B2 (en) 2019-05-28
AU2014348683A1 (en) 2016-06-09
US20180030411A1 (en) 2018-02-01
US20180208897A1 (en) 2018-07-26
US20200002674A1 (en) 2020-01-02
US20180119101A1 (en) 2018-05-03
CA2930665A1 (en) 2015-05-21
US10557119B2 (en) 2020-02-11
AU2014348683B2 (en) 2020-11-05
US20190316091A1 (en) 2019-10-17
US20150306212A1 (en) 2015-10-29
US20170369843A1 (en) 2017-12-28
WO2015073587A3 (en) 2015-08-06
US20180265847A1 (en) 2018-09-20
US10344263B2 (en) 2019-07-09
US20190316089A1 (en) 2019-10-17
US20190376034A1 (en) 2019-12-12
US10301594B1 (en) 2019-05-28
US20180187153A1 (en) 2018-07-05

Similar Documents

Publication Publication Date Title
US10557119B2 (en) Erythroid cells comprising phenylalanine ammonia lyase
US20210180015A1 (en) Membrane-Receiver Complex Therapeutics
US11576934B2 (en) Methods and compositions for immunomodulation

Legal Events

Date Code Title Description
AS Assignment

Owner name: FLAGSHIP VENTURES MANAGEMENT, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:BERRY, DAVID ARTHUR;REEL/FRAME:037537/0107

Effective date: 20141112

Owner name: FLAGSHIP VENTURES MANAGEMENT, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KAHVEJIAN, AVAK;MATA-FINK, JORDI;ROUND, JOHN;AND OTHERS;SIGNING DATES FROM 20141020 TO 20141111;REEL/FRAME:037537/0099

Owner name: VL26, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:FLAGSHIP VENTURES MANAGEMENT, INC.;REEL/FRAME:037537/0126

Effective date: 20141111

Owner name: RUBIUS THERAPEUTICS, INC., MASSACHUSETTS

Free format text: CHANGE OF NAME;ASSIGNOR:VL26, INC.;REEL/FRAME:037565/0839

Effective date: 20150127

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION