US20200368285A1 - Platform for generating safe cell therapeutics - Google Patents

Platform for generating safe cell therapeutics Download PDF

Info

Publication number
US20200368285A1
US20200368285A1 US16/636,249 US201816636249A US2020368285A1 US 20200368285 A1 US20200368285 A1 US 20200368285A1 US 201816636249 A US201816636249 A US 201816636249A US 2020368285 A1 US2020368285 A1 US 2020368285A1
Authority
US
United States
Prior art keywords
therapeutic
cell
cytoplasts
cytoplast
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US16/636,249
Other languages
English (en)
Inventor
Richard Klemke
Huawei Wang
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.)
University of California
Original Assignee
University of California
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of California filed Critical University of California
Priority to US16/636,249 priority Critical patent/US20200368285A1/en
Assigned to THE REGENTS OF THE UNIVERSITY OF CALIFORNIA reassignment THE REGENTS OF THE UNIVERSITY OF CALIFORNIA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KLEMKE, RICHARD, WANG, HUAWEI
Priority to US16/902,420 priority patent/US10960071B2/en
Publication of US20200368285A1 publication Critical patent/US20200368285A1/en
Pending 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
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • 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
    • 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
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • 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
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2066IL-10
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/208IL-12
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/215Coronaviridae, e.g. avian infectious bronchitis virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0091Purification or manufacturing processes for gene therapy compositions
    • 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
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/88Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
    • 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
    • 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
    • C12N5/0646Natural killers cells [NK], NKT cells
    • 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
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0667Adipose-derived stem cells [ADSC]; Adipose stromal stem cells
    • 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
    • C12N5/10Cells modified by introduction of foreign genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/999Small molecules not provided for elsewhere
    • 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
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • C12N2509/10Mechanical dissociation
    • 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
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • C12N2510/02Cells for production
    • 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
    • C12N2513/003D culture

Definitions

  • the present disclosure relates, at least in part, to the field of biotechnology, and more specifically, to methods and compositions using cytoplasts (e.g., enucleated cells) for treatment, prevention of disease, prophylactic treatment, adjuvant therapy, or immunomodulation in healthy or diseased subjects.
  • cytoplasts e.g., enucleated cells
  • the present disclosure is based, at least in part, on the generation of cytoplasts for use as a safe and controllable therapeutic and/or delivery vehicle.
  • the methods and compositions described herein provide several advantages. Methods for enucleating cells to generate cytoplasts, cytoplasts, compositions, and methods of using cytoplasts described can offer several benefits over previous cell-based therapeutics. First, the cytoplasts, compositions, and methods described herein can be safer than previous cell-based therapies. There can, in some embodiments, be a reduced risk of transferring genetic material, including, e.g., nuclear-encoded DNA gene transfer to host. Further potential safety advantages can include one or more of: not responding to the microenvironment(s) of a subject, not proliferating, and not contributing to disease progression (e.g., compared to nucleated mesenchymal stem cells).
  • the cytoplasts described herein can have a limited or defined (e.g., known, or programmable) life span.
  • the cytoplasts described herein can have a reduced size compared to cells in some other cell-based therapies.
  • cryopreservation includes cooling or freezing, and storing, in the short-term or long-term, biological material (e.g., cells, cytoplasts) at very low temperatures (e.g., ⁇ 80° C. in solid C02, ⁇ 196° C. in liquid nitrogen, etc.).
  • Cryohibernation includes short-term cooling and storing of biological material (e.g., cells, cytoplasts) in suspended animation, at non-freezing temperatures, such as, e.g., at 4° C.
  • Cryohibernation of cytoplasts can be advantageous for one or more of the following reasons: cryohibernation is less labor-intensive than cryopreservation, and cytoplasts that have undergone cryohibernation can be transported (e.g., shipped).
  • the cytoplasts described herein can be extensively engineered.
  • the cytoplasts can be engineered to produce or express a therapeutic entity, or home to specific sites.
  • Other advantages of the presently claimed invention are described herein.
  • cytoplasts cytoplasts, compositions comprising cytoplasts, methods of using cytoplasts, and methods of treating a subject, such as providing benefits to a healthy or unhealthy subject, or treating or diagnosing a disease or condition (e.g., a cancer or a neoplasm, an infection, an inflammatory condition, a neurological disease (e.g., a neurodegenerative disease), a degenerative disease, an autoimmune disease, a cardiovascular disease, an ischemic disease, a genetic or inherited disorder, a developmental disorder, an ophthalmologic disease, a skeletal disease, a metabolic disease, a toxicosis, an idiopathic condition, or two or more thereof, or any disease disclosed herein) in a subject.
  • a disease or condition e.g., a cancer or a neoplasm, an infection, an inflammatory condition, a neurological disease (e.g., a neurodegenerative disease), a degenerative disease, an autoimmune disease, a cardiovascular disease, an
  • methods of treating a subject include: administering to the subject a therapeutically effective amount of a composition comprising a cytoplast (e.g., a recombinant cytoplast, any cytoplast described herein).
  • a cytoplast administered to a subject can produce a therapeutic.
  • a cytoplast administered to a subject can produce one or more of: a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic protein (e.g., an enzyme, an antibody, an antigen, a toxin, cytokine, a protein hormone, a growth factor, a cell surface receptor, or a vaccine), a therapeutic peptide (e.g., a peptide hormone or an antigen), a small molecule therapeutic (e.g., a steroid, a polyketide, an alkaloid, a toxin, an antibiotic, an antiviral, a colchicine, a taxol, a mitomycin, or emtansine), or a therapeutic gene editing factor.
  • a therapeutic protein e.g., an enzyme, an antibody, an antigen, a toxin, cytokine, a protein hormone, a growth factor, a cell surface receptor, or a vaccine
  • a therapeutic peptide e.g., a
  • a cytoplast can be engineered to produce one or more of: a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic protein, a therapeutic peptide, a therapeutic small molecule, or a therapeutic gene editing factor. In some embodiments, a cytoplast does not need to be engineered to produce one or more of: a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic protein, a therapeutic peptide, a therapeutic small molecule, or a therapeutic gene editing factor.
  • one or more of: a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic protein, a therapeutic peptide, a small molecule therapeutic, or a therapeutic gene editing factor can be produced by the cell from which the cytoplast was obtained.
  • a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic protein, a therapeutic peptide, a small molecule therapeutic, or a therapeutic gene editing factor can include a targeting moiety.
  • targeting moieties that can be produced by or contained in a cytoplast include chemokine receptors, adhesion molecules, and antigens.
  • a cytoplast administered to a subject can contain a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic protein (e.g., an enzyme, an antibody, an antigen, a toxin, cytokine, a protein hormone, a growth factor, a cell surface receptor, or a vaccine, or any therapeutic protein that is currently available or in development), a therapeutic peptide (e.g., a peptide hormone or an antigen, or any therapeutic peptide that is currently available or in development), a small molecule therapeutic (e.g., a steroid, a polyketide, an alkaloid, a toxin, an antibiotic, an antiviral, an analgesic, an anticoagulant, an antidepressant, an anticancer drug, an antiepileptic, an antipsychotic, a sedative, a colchicine, a taxol, a mitomycin, emtansine, or any small molecule therapeutic that is currently available or in development
  • Non-limiting examples of oncolytic viruses include Talimogene laherparepvec, Onyx-15, GL-ONC1, CV706, Voyager-V1, and HSV-1716.
  • Some wild-type viruses also show oncolytic behavior, such as Vaccinia virus, Vesicular stomatitis virus, Poliovirus, Reovirus, Senecavirus, ECHO-7, and Semliki Forest virus.
  • a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic, a therapeutic peptide, a small molecule therapeutic, or a therapeutic gene editing factor is not produced by the cytoplast.
  • a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic, a therapeutic peptide, a small molecule therapeutic, or a therapeutic gene editing factor is packaged inside the cytoplast.
  • the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor are recombinantly expressed.
  • the cell from which the cytoplast is derived or obtained is engineered to produce one or more of the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor.
  • the cell from which the cytoplast is derived or obtained is engineered to stably (e.g., permanently) express one or more of the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor.
  • the cell from which the cytoplast is derived or obtained is engineered to transiently express one or more of the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor.
  • the cell from which the cytoplast is derived or obtained is engineered prior to enucleation.
  • the cytoplast is engineered to transiently express one or more of the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor (e.g., engineered following enucleation).
  • DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor are not naturally expressed (i.e., in the absence of engineering) in the cell from which the cytoplast was derived or obtained (i.e., the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor are exogenous to the cytoplast).
  • the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor are not naturally expressed in the subject (i.e., the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor are exogenous to the subject).
  • the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor are not naturally expressed in the subject at the intended site of therapy (e.g., a tumor, or a particular tissue, such as the brain, the intestine, the lungs, the heart, the liver, the spleen, the pancreas, muscles, eyes, and the like) (i.e., the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor are exogenous to the intended site of therapy).
  • the intended site of therapy e.g., a tumor, or a particular tissue, such as the brain, the intestine, the lungs, the heart, the liver, the spleen, the pancreas, muscles, eyes, and the like
  • the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor are naturally expressed (i.e., in the absence of engineering) in the cell from which the cytoplast was derived or obtained (i.e., the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor are innately endogenous to the cytoplast) (e.g., in the absence of engineering of the cell from which the cytoplast was derived or obtained).
  • the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor are naturally expressed in the subject (i.e., the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor are endogenous to the subject).
  • the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor are naturally expressed in the subject at the intended site of therapy (e.g., a tumor, or a particular tissue, such as the brain, the intestine, the lungs, the heart, the liver, the spleen, the pancreas, muscles, eyes, and the like) (i.e., the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor are endogenous to the intended site of therapy).
  • the intended site of therapy e.g., a tumor, or a particular tissue, such as the brain, the intestine, the lungs, the heart, the liver, the spleen, the pancreas, muscles, eyes, and the like
  • therapeutic e.g., the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor, is derived from a synthetic cell and loaded into the cytoplast.
  • the cytoplast expresses a corrected, a truncated, or a non-mutated version and/or copy of the DNA molecule, the RNA molecule, the protein, the peptide, the small molecule therapeutic, and/or the gene-editing factor as compared to the cell from which the cytoplast was derived or obtained.
  • the cytoplast is obtained from any nucleated cell (e.g., a eukaryotic cell, a mammalian cell (e.g., a human cell, or any mammalian cell described herein), a protozoal cell (e.g., an amoeba cell), an algal cell, a plant cell, a fungal cell, an invertebrate cell, a fish cell, an amphibian cell, a reptile cell, or a bird cell).
  • a nucleated cell e.g., a eukaryotic cell, a mammalian cell (e.g., a human cell, or any mammalian cell described herein), a protozoal cell (e.g., an amoeba cell), an algal cell, a plant cell, a fungal cell, an invertebrate cell, a fish cell, an amphibian cell, a reptile cell, or a bird cell).
  • a cytoplast can produce or contain at least 2 (e.g., at least 2, 3, 4, 5, or more) different therapeutic DNA molecules, therapeutic RNA molecules, therapeutic proteins, therapeutic peptides, small molecule therapeutics, or therapeutic gene-editing factors, in any combination.
  • a cytoplast can produce or contain a therapeutic DNA molecule and a small molecule therapeutic.
  • a cytoplast can produce or contain two different small molecule therapeutics.
  • a cytoplast can produce or contain a chemokine receptor (e.g., for targeting) and a small molecule therapeutic.
  • the cytoplast is obtained from an immortalized cell, a cancer cell (e.g., any cancer cell) a primary (e.g., host-derived) cell, or a cell line. Any non-immortal cell can be immortalized using methods known in the art.
  • the cytoplast can be obtained from a cell autologous to the subject.
  • the cytoplast can be obtained from a cell allogenic to the subject.
  • the cytoplast is obtained from an immune cell.
  • the cytoplast is obtained from a natural killer (NK) cell, a neutrophil, a macrophage, a lymphocyte, a fibroblast, an adult stem cell (e.g., hematopoietic stem cell, a mammary stem cell, an intestinal stem cell, mesenchymal stem cell, an endothelial stem cell, a neural stem cell, an olfactory adult stem cell, a neural crest stem cell, a skin stem cell, or a testicular cell), a mast cell, a basophil, an eosinophil, or an inducible pluripotent stem cell.
  • NK natural killer
  • neutrophil e.g., hematopoietic stem cell, a mammary stem cell, an intestinal stem cell, mesenchymal stem cell, an endothelial stem cell, a neural stem cell, an olfactory adult stem cell, a neural crest stem cell, a skin stem cell, or a testicular cell
  • two or more cells prior to enucleation, two or more cells (e.g., any of the cells disclosed herein) are fused by any method disclosed herein or known in the art. Enucleation of the fusion product can result in a cytoplast.
  • a first cytoplast is fused to a cell or second cytoplast.
  • the cell is any nucleated (e.g., a mammalian cell (e.g., a human cell, or any mammalian cell described herein), a protozoal cell (e.g., an amoeba cell), an algal cell, a plant cell, a fungal cell, an invertebrate cell, a fish cell, an amphibian cell, a reptile cell, or a bird cell).
  • the second cell is a synthetic cell. Accordingly, provided are methods of altering the behavior of a cell comprising fusing the cell with any of the cytoplasts described herein. Also provided herein are methods comprising administering to a subject a therapeutically effective amount of a cell to which a cytoplast has been fused.
  • the second cytoplast is derived from the same type of cell as the first cytoplast. In some embodiments, the second cytoplast is derived from a different type of cell as the first cytoplast. In some embodiments, the second cytoplast contains or expresses at least one therapeutic DNA molecule, therapeutic RNA molecule, therapeutic protein, therapeutic peptide, small molecule therapeutic, therapeutic gene editing factor, a therapeutic nanoparticle, or another therapeutic agent that is the same as a therapeutic DNA molecule, therapeutic RNA molecule, therapeutic protein, therapeutic peptide, small molecule therapeutic, therapeutic gene editing factor, a therapeutic nanoparticle contained in or expressed by the first cytoplast.
  • the second cytoplast contains or expresses at least one therapeutic DNA molecule, therapeutic RNA molecule, therapeutic protein, therapeutic peptide, small molecule therapeutic, therapeutic gene editing factor, a therapeutic nanoparticle, or another therapeutic agent that is different from a therapeutic DNA molecule, therapeutic RNA molecule, therapeutic protein, therapeutic peptide, small molecule therapeutic, therapeutic gene editing factor, a therapeutic nanoparticle contained in or expressed by the first cytoplast.
  • a first cytoplast can be fused to a cell or to a second cytoplast using any method known in the art, for example, electrofusion or viral fusion using viral-based cell surface peptides.
  • the therapeutic RNA molecule is messenger RNA (mRNA), short hairpin RNA (shRNA), small interfering RNA (siRNA), microRNA, long non-coding RNA (lncRNA) or a RNA virus.
  • the therapeutic DNA molecule is single-stranded DNA, double-stranded DNA, an oligonucleotide, a plasmid, a bacterial DNA molecule or a DNA virus.
  • the therapeutic protein is a cytokine, a growth factor, a hormone, an antibody, a small-peptide based drug, or an enzyme.
  • the cytoplast transiently expresses the therapeutic DNA molecule, the therapeutic RNA molecule, the therapeutic protein, the therapeutic peptide, the small molecule therapeutic, and/or the therapeutic gene editing factor.
  • the expression of the therapeutic DNA molecule, the therapeutic RNA molecule, the therapeutic protein, the therapeutic peptide, the small molecule therapeutic, and/or the therapeutic gene editing factor is inducible.
  • a nucleated cell is permanently engineered to express the therapeutic DNA molecule, the therapeutic RNA molecule, the therapeutic protein, the therapeutic peptide, the small molecule therapeutic, and/or the therapeutic gene editing factor.
  • the cytoplast comprises a therapeutic agent or a nanoparticle.
  • the therapeutic agent is a small molecule or a bacteria or an exosome.
  • the method further includes administering to the subject one or more additional therapies.
  • the one or more additional therapies is selected from the group consisting of: cell-based therapy, a small molecule, immuno-therapy, chemotherapy, radiation therapy, gene therapy, and surgery.
  • isolated cytoplasts e.g., recombinant cytoplasts, or any cytoplasts described herein
  • cytoplasts comprising a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic protein, a therapeutic peptide, a small molecule therapeutic, a therapeutic gene-editing factor a therapeutic nanoparticle and/or another therapeutic agent.
  • the therapeutic agent is a drug or chemotherapeutic or a gene editing agent.
  • Also provided herein are methods of making a recombinant cytoplast e.g., any cytoplast described herein, the method comprising: enucleating a nucleated cell; and introducing into the enucleated cell an therapeutic DNA molecule, an therapeutic RNA molecule, an therapeutic protein, an therapeutic peptide, a small molecule therapeutic, a therapeutic gene-editing factor, a therapeutic nanoparticle and/or another therapeutic agent.
  • the introducing step precedes the enucleating step.
  • the enucleating step precedes the introducing step.
  • the introducing step results in a transient expression of the therapeutic DNA molecule, the therapeutic RNA molecule, the therapeutic protein, the therapeutic peptide, the small molecule therapeutic, the therapeutic gene editing factor, or the other therapeutic agent.
  • the introducing step results in a permanent expression of the therapeutic DNA molecule, the therapeutic RNA molecule, the therapeutic protein, the therapeutic peptide, the small molecule therapeutic, the therapeutic gene editing factor, or the other therapeutic agent.
  • the therapeutic RNA molecule is messenger RNA (mRNA), short hairpin RNA (shRNA), small interfering RNA (siRNA), microRNA, long non-coding RNA (lncRNA) or a RNA virus.
  • the therapeutic DNA molecule is single-stranded DNA, double-stranded DNA, an oligonucleotide, a plasmid, a bacterial DNA molecule or a DNA virus.
  • the therapeutic DNA or the therapeutic RNA is a gene therapy.
  • the therapeutic protein is an enzyme., an antibody, a toxin, cytokine, a protein hormone, a growth factor, or a vaccine.
  • a nucleated cell can be cultured (e.g., in a suspension, as adherent cells, as adherent cells in 3D (e.g., in semi-suspension or other nonadherent methods)) under various conditions (e.g., in a cytokine bath, or under hypoxic conditions) before enucleation.
  • Also provided herein are methods of making a recombinant cytoplast that include: transfecting a nucleated cell with a vector; and enucleating the transfected cell.
  • the vector is a viral vector (e.g., a retrovirus vector (e.g., a lentivirus vector), an adeno-associated virus (AAV) vector, a vesicular virus vector (e.g., vesicular stomatitis virus (VSV) vector), or a hybrid virus vector).
  • a viral vector can be a cytoplasmic-replicating virus.
  • a viral vector can be a nuclear-replicating virus.
  • enucleating occurs after the vector integrates into the genome of the nucleated cell.
  • the vector is transfected after the cell is enucleated.
  • the order of transfection and enucleation can affect the choice of vector. For example, if transfection occurs before enucleation, either a cytoplasmic-replicating virus or a nuclear-replicating virus can be an acceptable vector. For example, if transfection occurs after enucleation, a cytoplasmic-replicating virus can be a better choice of vector than a nuclear-replicating virus; on the other hand, a nuclear-replicating virus can be packaged in an enucleated cytoplast to be released upon death of the cytoplast.
  • the vector comprises a coding sequence of a therapeutic protein.
  • the therapeutic protein is an enzyme, an antibody, a toxin, cytokine, a protein hormone, a growth factor, or a vaccine.
  • a method can include administering to the subject a therapeutically effective amount of a composition comprising a naturally derived cytoplast or an engineered cytoplast expressing a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic protein, a therapeutic peptide, a small molecule therapeutic, and/or a therapeutic gene-editing factor.
  • Also provided herein are methods of treating a subject that include: administering to the subject a therapeutically effective amount of a composition comprising a cytoplast containing a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic protein, a therapeutic peptide, a small molecule therapeutic, a therapeutic gene-editing factor, a therapeutic nanoparticle, and/or another therapeutic agent.
  • the cytoplast is obtained from a mammalian cell. In some embodiments, the cytoplast is obtained from an immune cell.
  • the composition further includes a targeting moiety.
  • the targeting moiety is a cell surface protein.
  • the targeting moiety is a secreted protein, or a protein that is tethered to the extracellular matrix.
  • FIG. 1 is a schematic diagram of one embodiment of the disclosure in which the starting material is either donor-derived allogenic or autologous cells, or engineered cells with a designed function(s).
  • FIG. 2A is a representative fluorescence microscopy image of cytoplasts produced from human telomerase reverse transcriptase (hTERT) adipose-derived human mesenchymal stem cells (MSC).
  • hTERT human telomerase reverse transcriptase
  • MSC mesenchymal stem cells
  • CTMR red dye
  • nuclei were stained with Vybrant® DyecycleTM green.
  • Scale bar 50 ⁇ m.
  • FIG. 2D is a representative fluorescence microscopy image of cytoplasts produced from human natural killer cell line NKL cells (NK).
  • NK human natural killer cell line
  • CMFDA 5-chloromethylfluorescein diacetate
  • DAPI 4′,6-diamidino-2-phenylindole
  • FIG. 3A is a representative graph showing the relative fold change in viable cells or cytoplasts over time.
  • FIG. 3B is a representative graph showing the viable cells and cytoplasts after recovery from frozen storage (cryopreservation).
  • FIG. 4 are representative flow cytometry graphs showing the number of events counted over the signal strength of the indicated fluorescent antibody/marker (CD90, CD44, CD146, CD166, CD45, and isotype control).
  • Bone marrow MSCs or MSC-derived cytoplasts were stained 24 hours after enucleation and then analyzed by flow cytometry with FlowJo software. Green (light gray) represents nucleated MSCs and red (dark gray) represents enucleated cytoplasts.
  • AIF mitochondrial marker anti-apoptosis-inducing factor
  • FIG. 6C ′ is an enlarged image of FIG. 6C .
  • FIG. 6D ′ is an enlarged image of FIG. 6D .
  • Scale bar 50 ⁇ m.
  • ER endoplasmic reticulum
  • DAPI dark gray ovals
  • FIG. 8A is a representative bright field microscopy images of MSCs or cytoplasts in Boyden chamber assays that successfully migrated to the undersurface of 8.0 ⁇ m porous filters in 3 hours and were then stained with Crystal Violet.
  • cells and cytoplasts migrated in culture media containing 2% FBS in both the upper and lower chambers.
  • the bottom surface of the upper chamber was coated with fibronectin, and 100 ng/mL of stromal cell-derived factor 1 alpha (SDF-1 ⁇ ) was added to the lower chamber.
  • Scale bar 50 ⁇ m.
  • FIG. 9A-D is a representative epifluorescence microscopy image of MSCs incubated with 100 sM of the cell-permeable peptide (Arg)9-FAM (Fluorescein amidite, light gray) and stained with Hoechst 33342 (nuclei, dark gray ovals). Arrows indicate Hoechst-stained nuclei; arrowheads indicate positive (Arg)9-FAM signal.
  • Arg cell-permeable peptide
  • FIG. 9B is a representative epifluorescence microscopy image of MSC-derived cytoplasts incubated with 100 sM of the cell-permeable fluorescent peptide (Arg)9-FM (light gray) and stained with Hoechst 33342 (nuclei, dark gray ovals). Arrows indicate Hoechst-stained nuclei; arrowheads indicate positive (Arg)9-FAM fluorescence.
  • Arg cell-permeable fluorescent peptide
  • FIG. 9C is a representative epifluorescence microscopy image of MSCs incubated with 100 ⁇ M of the chemotherapeutic drug doxorubicin (light gray) and stained with Hoechst 33342 (faint dark gray ovals). Arrows indicate Hoechst-stained nuclei.
  • FIG. 9D is a representative epifluorescence microscopy image of MSC-derived cytoplasts 100 sM of the chemotherapeutic drug doxorubicin (light gray) and stained with Hoechst 33342 (faint dark gray ovals). Arrowheads indicate positive doxorubicin fluorescence.
  • FIG. 9E is a representative bar graph showing the cell and cytoplast average corrected total cell fluorescence per area, which models the relative fluorescence while accounting for the size difference between cells and cytoplasts.
  • FITC fluorescein isothiocyanate
  • FIG. 11A are representative merged and unmerged epifluorescence microscopy images of MSC and MSC-derived cytoplasts 20 hours after transfection with purified enhanced green fluorescent protein messenger RNA (EGFP-mRNA) and stained with Hoechst 33342.
  • EGFP-mRNA enhanced green fluorescent protein messenger RNA
  • FIG. 12A is a representative Western blot of cells treated for 10 minutes with either control medium, MSC-conditioned medium (MSC CM), MSC-derived cytoplast-conditioned medium (Cytoplast CM), or 50 ng/mL of vascular endothelial growth factor (VEGF).
  • Immunoblotting was performed for protein kinase B (Akt), phosphorylated Akt (p-Akt), extracellular signal-regulated kinase (Erk), phosphorylated Erk (p-Erk).
  • Akt protein kinase B
  • p-Akt phosphorylated Akt
  • Erk extracellular signal-regulated kinase
  • p-Erk phosphorylated Erk
  • Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was a loading control.
  • FIG. 12B is a representative bar graph showing the relative Gaussia luciferase (Gluc) activity in media 48 hours after plating MSCs transfected with Gluc mRNA (MSC-Gluc), non-transfected MSC control cell (MSC), MSC-derived cytoplasts transfected with Gluc mRNA (Cytoplast-Gluc), and non-transfected MSC-derived cytoplasts (Cytoplast).
  • MSC-Gluc Gaussia luciferase
  • FIG. 12C is a representative bar graph showing the ratio of RAW 264.7 macrophages migrating towards a gradient of the indicated conditioned media for 4 hours in a Boyden chamber assay.
  • Migratory cells on the lower membrane surface were stained with Crystal Violet and the number of cells counted per field and normalized to the loading control (cells directly attached to fibronectin-coated plates).
  • Colony stimulating factor 1 (CSF-1), 40 ng/mL of mouse CSF-1 as a positive control; MSC-media, conditioned media from MSCs; Cytoplast media, conditioned media from MSC-derived cytoplasts. Mean ⁇ SEM; n 10.
  • FIG. 13A is a schematic representation of an interleukin 10 (IL-10) mRNA transfected into MSC and cytoplasts.
  • IL-10 interleukin 10
  • CDS IL-10 mRNA coding region
  • 5′UTR and 3′UTR of human beta globin (HBB) mRNA were added respectively to the 5′ and 3′ end of IL-10 CDS.
  • An artificial 5′Cap was added to the 5′ end of the IL-10 mRNA and the pseudouridine modification was engineered to increase mRNA stability.
  • FIG. 13B is a bar graph showing IL-10 concentration in the culture medium of transfected (++) or non-transfected ( ⁇ ) MSC or MSC-derived cytoplasts.
  • MSC-derived cytoplasts were transfected with IL-10 mRNA, then seeded in a 24 well plate at 2.5 ⁇ 10 4 cells/well.
  • Conditioned medium (CM) was collected 24 hours after transfection and the IL-10 concentration determined by ELISA.
  • FIG. 13C is an immunoblot showing protein expression of Stat3 and phosphorylated Stat3 (P-Stat3, a marker of L-10 activation) in serum-starved RAW macrophage cells 10 treated with the indicated conditioned media (CM) from MSCs or cytoplasts treated as in FIG. 13B for 1 hour.
  • Untreated no CM treated control.
  • Complete medium RAW cells treated with MSC complete culture medium.
  • MSC Ctrl RAW cells treated with CM from non-transfected MSCs.
  • MSC IL-10 RAW cells treated with CM from IL-10 mRNA transfected MSCs.
  • Cytoplast Ctrl RAW cells treated with CM from non-transfected cytoplasts.
  • Cytoplasts IL-10 RAW cells treated with CM from IL-10 mRNA transfected cytoplasts.
  • FIG. 13D is a bar graph showing the concentration of secreted IL-10 cytokine in the mouse blood as determined by ELISA.
  • FIG. 15C is a representative bar graph showing the detected Vybrant® DiD-labeled MSCs or cytoplasts present in lung.
  • FIG. 16A is a schematic of a representative lentivirus vector engineered to express CXCR4 on MSCs and cytoplasts (SEQ ID NOs. 1-15).
  • FIG. 16B is a representative flow cytometry graphs showing the number of events counted over the signal strength of the cell surface CXCR4 expression by fluorescent antibody on engineered cytoplasts and engineered parental MSCs as analyzed by FlowJo.
  • FIG. 17A is a schematic of the lentivirus vector engineered to express PSGL1 (P-Selectin Glycoprotein Ligand 1) and Fut7 (Fucosyltransferase, glycosylates/activates PSGL1) on MSCs and cytoplasts.
  • PSGL1 P-Selectin Glycoprotein Ligand 1
  • Fut7 Fucosyltransferase, glycosylates/activates PSGL1
  • FIG. 17B is a representative flow cytometry graph showing the number of events counted over the signal strength of the cell surface PSGL1 expression by fluorescent 30 antibody on engineered cytoplasts and engineered parental MSCs as analyzed by FlowJo.
  • FIG. 17C is a representative graph showing cell surface binding of P-Selectin with engineered MSCs and MSC-derived cytoplasts as determined by flow cytometry.
  • MSC control parental MSCs.
  • Engineered MSC PSGL1/Fut7 engineered MSC.
  • Engineered cytoplast PSGL1/Fut7 engineered MSC-derived cytoplasts.
  • FIG. 18A is a schematic of a lentivirus vector engineered to express mCD47 (SEQ ID NO: 19) on MSCs and cytoplasts.
  • FIG. 18B is a representative flow cytometry graph showing the number of events counted over the signal strength of the cell surface of mCD47 expression on engineered cytoplasts and MSCs as analyzed by FlowJo.
  • FIG. 19A is schematics of IL-12 mRNA design.
  • Kozak sequence was added in front of the start codon of IL-12 mRNA coding region (CDS).
  • 5′UTR and 3′UTR of human beta globin (HBB) mRNA were added respectively to the 5′ and 3′ end of IL-12 CDS.
  • Artificial 5′Cap was added to the 5′ end of the IL-12 mRNA and the pseudouridine modification was implemented to increase mRNA stability.
  • FIG. 19C is an immunoblot showing the activation of the phosphorylated/activated form of Stat4 (P-Stat4).
  • Mouse splenocyte cells were treated with full media, purified IL-12 protein standard or the indicated CM collected from MSCs or cytoplasts engineered as in FIG. 19B for 30 minutes.
  • MSC full medium mouse splenocytes treated with MSC complete culture medium.
  • MSC IL-12 treated with CM from IL-12 mRNA transfected MSCs.
  • Cytoplast IL-12 treated with CM from IL-12 mRNA-transfected cytoplasts.
  • FIG. 19D is a representative scatter plot showing the concentration of secreted IL-12 cytokine per mg of tumor protein.
  • IL-12 engineered or control cytoplasts treated as in FIG. 19B were injected into established E0771 (mouse medullary breast carcinoma) tumors growing in syngeneic C57BL/6 mice. Forty-eight hours after tumor injection, animals were euthanized and tumor samples were collected, lysed and analyzed by ELISA.
  • PBS samples from mice injected with PBS.
  • Cytoplasts samples from mice injected with non-engineered cytoplasts.
  • Cytoplasts IL-12 samples from mice injected with cytoplasts engineered to express IL-12 cytokine.
  • FIG. 20A is a scatter plot showing the fold change of expression for interferon-7 mRNAs.
  • MSC-derived cytoplasts engineered to express IL-12 or control cytoplasts without IL-12 were injected into established E0771 tumors growing in syngeneic C57BL/6 mice. Forty-eight hours after injection, animals were euthanized and tumor samples were collected, lysed and analyzed by Real-time RT-PCR.
  • PBS samples from mice injected with PBS.
  • Cytoplasts samples from mice injected with non-engineered cytoplasts.
  • FIG. 20B is a scatter plot showing the fold change of expression for PD-L1 mRNAs.
  • MSC-derived cytoplasts engineered to express IL-12 or control cytoplasts without IL-12 were injected into established E0771 tumors growing in syngeneic C57BL/6 mice. Forty-eight hours after injection, animals were euthanized and tumor samples were collected, lysed and analyzed by Real-time RT-PCR.
  • PBS samples from mice injected with PBS.
  • Cytoplasts samples from mice injected with non-engineered cytoplasts.
  • FIG. 20C is a scatter plot showing the fold change of expression for CXCL9 mRNAs.
  • MSC-derived cytoplasts engineered to express IL-12 or control cytoplasts without IL-12 were injected into established E0771 tumors growing in syngeneic C57BL/6 mice. Forty-eight hours after injection, animals were euthanized and tumor samples were collected, lysed and analyzed by Real-time RT-PCR.
  • PBS samples from mice injected with PBS.
  • Cytoplasts samples from mice injected with non-engineered cytoplasts.
  • FIG. 20D is a bar graph showing the fold change of E0771 subcutaneous tumor sizein C57Bl/6 mice that were injected intratumorally with 3 ⁇ 10 6 IL-12 engineered cytoplasts (Cytoplasts I12 group) or PBS (PBS group) on day 11, day 14, and day 18 after tumor cell inoculation.
  • FIG. 21B is a bar graph showing percentage of GFP-positive tumor area for tumors treated as in FIG. 21A , which represents the portion of tumor cells infected by MSCs or cytoplasts carrying the oHSV-GFP virus.
  • FIG. 21C is a scatter plot showing the ratio of CD8 + effector T lymphocyte cells out of total CD45+ (immune cells) present in tumors injected with engineered cytoplasts, as analyzed by flow cytometry.
  • Established subcutaneous E0771 tumors in C57Bl/6 mice were injected intratumorally with oHSV-transfected and IL-12 engineered cytoplasts or PBS only injection (negative control).
  • FIG. 22A are representative epifluorescence microscopy images showing RFP expression in nucleated MSC cells successfully fused with Cre-engineered cytoplasts.
  • FIG. 22B is a bar graph showing the percentage of RFP+(fused) cells out of total cells.
  • Loxp MSC only single culture of Loxp-GFP-stop-Loxp-RFP-hTERT-MSCs.
  • Cre Cytoplasts only single culture of cytoplasts engineered to express Cre recombinase.
  • Co-culture Co-culturing of Loxp MSCs and Cre cytoplasts for 48 hours.
  • FIG. 23 is a schematic of the lentivirus vector engineered to express mCCR2 (SEQ ID NO: 20) on MSCs and cytoplasts.
  • FIG. 24A is a representative scatter plot showing the number of DiD-labeled MSCs or cytoplasts detected in the lung.
  • MSCs were cultured under standard adherent conditions (2D) or in suspension by the handing drop method (3D) to generate 3D cytoplasts.
  • FIG. 24B is a representative scatter plot showing the number of DiD-labeled MSCs or cytoplasts detected in the liver.
  • MSCs were cultured under standard adherent conditions (2D) or in suspension by the handing drop method (3D) to generate 3D cytoplasts.
  • FIG. 24C is a representative scatter plot showing the number of Vybrant® DiD-labeled MSCs or cytoplasts detected in the spleen.
  • MSCs were cultured under standard adherent conditions (2D) or in suspension by the handing drop method (3D) to generate 3D cytoplasts.
  • FIG. 25A is a representative line graph showing the viability of MSC and MSC-derived cytoplasts immediately after recovery from cryohibernation at 4 degrees Celsius for the indicated amounts of time. Viability was assessed in an automated cell count (Cell Countess) using Trypan blue dye exclusion and displayed as a ratio to the number of input cells.
  • Cell Countess Automated Cell count
  • FIG. 25B is a representative bar graph comparing the migrated MSC and MSC-derived cytoplasts in a Boyden chamber assay immediately after recovery from cryohibernation at 4 degrees Celsius for the indicated amounts of time.
  • Cells and cytoplasts were allowed to migrate for 3 hours with either no serum (negative control) or 10% premium FBS (P-FBS) as a chemoattractant in the bottom chamber, and counts were normalized to loading controls.
  • P-FBS premium FBS
  • cells from which the nucleus has been removed e.g., cytoplasts
  • cytoplasts e.g., cytoplasts
  • cells can be treated with cytochalasin B to soften the cortical actin cytoskeleton.
  • the nucleus can then be physically extracted from the cell body by high-speed centrifugation in gradients of Ficoll to generate a nucleus-free (enucleated) cytoplast. Because cytoplast and intact nucleated cells sediment to different layers in the Ficoll gradient, cytoplasts can, in some embodiments, be easily isolated and prepared for therapeutic purposes or fusion to other cells (nucleated or enucleated).
  • cytoplasts can be used as a homing vehicle to deliver clinically relevant cargos/payloads to treat healthy individuals (e.g., to improve energy, recovery from exercise, or to deliver natural products) or various diseases (e.g., any of the diseases described herein).
  • cytoplasts may be used to deliver supplements, anti-aging factors, preventative treatments, and the like to healthy individuals, e.g., individuals who have not been diagnosed with a specific disorder for which the delivered therapeutic is effective.
  • Cytoplasts possess significant therapeutic value because they can have one or more of the following properties: remain viable for up to 14 days, do not differentiate into other cell types, secrete bioactive proteins, can physically migrate/home, can be extensively engineered ex vivo to perform specific therapeutic functions, and can be fused to the same or other cell types to transfer desirable cell functions, natural or engineered. Therefore, cytoplasts may have wide utility as a new cellular vehicle to deliver therapeutically important biomolecules, gene editing factors, and disease-targeting cargos including chemotherapeutic drugs (e.g., doxorubicin), genes, viruses, bacteria, mRNAs, shRNAs, siRNA, peptides, plasmids and nanoparticles.
  • chemotherapeutic drugs e.g., doxorubicin
  • genes viruses, bacteria, mRNAs, shRNAs, siRNA, peptides, plasmids and nanoparticles.
  • the present disclosure advantageously enables, in some embodiments, the generation of a safe therapeutic, as it is believed that no unwanted DNA is transferred to the subject using the cytoplasts described herein.
  • the present disclosure advantageously enables controllable therapeutics, as cell death of the cytoplasts can, in some embodiments, occur in a precise amount of time, e.g., 3-4 days.
  • the cytoplasts described herein can act as a cell-based carrier that can be genetically engineered to deliver specific gene editing, disease-fighting, and health promoting cargos to humans or animals.
  • manufacturing significant numbers of therapeutic cells for clinical applications can be limited and expensive, thereby limiting the application of many cell-based therapies, especially in the stem cell field.
  • immortalized cells may cause cancer, and thus can be too dangerous for therapeutic applications.
  • the present disclosure allows for the use of type of nucleated cell (an immortalized cell, a cancer cell (e.g., any cancer cell) a primary (e.g., host-derived) cell, or a cell line) for large-scale manufacturing in culture for therapeutic use, because they can be rendered safe by enucleation prior to administration or use.
  • the present disclosure provides methods to produce cell-based therapeutics that are safe and controllable in a subject, from any nucleated cell type that maintains a nucleus throughout it lifespan or does not naturally enucleate.
  • the disclosure provides methods for the removal of the cell nucleus (also called enucleation) from any nucleated cell derived (e.g., obtained) from either normal or cancer cell lines or any primary cell removed from the body including, but not limited to, commonly used therapeutic cells derived (e.g., obtained) from the immune system (e.g., natural killer (NK) cells, neutrophils, macrophages, lymphocytes, mast cells, basophils, eosinophils), stem cells (including, for example, iPSC (induced pluripotent stem cells), adult stem cells (e.g., mesenchymal stem cells), and embryonic stem cells), and fibroblasts.
  • NK natural killer
  • stem cells including, for example, iPSC (induced pluripotent stem cells), adult stem cells
  • Cell enucleation can create a therapeutic cytoplast which is viable for a limited period of time, for example, up to 3-4 days. Therefore, the present disclosure, in some aspects, provides a new use for cytoplasts as a safe therapeutic vehicle that cannot perform one or more of the following actions: proliferate, differentiate, permanently engraft into the subject, become cancerous, or transfer nuclear-encoded DNA/genes to the subject (e.g., transfer of dangerous nuclear-encoded DNA/genes to the subject).
  • cell-based therapeutics generally use normal or engineered nucleated cells.
  • Some cell-based therapies irradiate cells prior to subject administration in order to prevent cell proliferation and induced lethal DNA-damage.
  • this approach induces mutations and produces significant amounts of reactive oxygen species that can irreversibly damage cellular proteins and DNA, which can release large amounts of damaged/mutated DNA into the body of a subject.
  • Such products can be dangerous if they integrate into other cells and/or induce an unwanted anti-DNA immune response.
  • Irradiated cells can also be dangerous because they can transfer their mutated DNA and genes to host cells by cell-cell fusion. Removing the entire nucleus from a cell is a less damaging and significantly safer method for limiting cellular lifespan that can preclude any introduction of nuclear DNA into a subject.
  • stem cells such as mesenchymal stem cells (MSCs)
  • MSCs mesenchymal stem cells
  • therapeutic cells have been engineered with a drug-inducible suicide switch to limit cellular lifespan.
  • activation of the switch in vivo can require administering a subject with potent and potentially harmful drugs with unwanted side effects. While this method can induce suicide in culture cells (e.g., greater than 95%), it is expected to be inefficient when translated into the clinic.
  • a drug-inducible suicide switch could be an insufficient safety measure for clinical practice, since not all cells in the subject may undergo drug-induced death.
  • a drug-induced suicide switch could be considered dangerous or insufficient for clinical practice.
  • the death of a therapeutic cell can release large amounts of DNA (normal or genetically altered), which can integrate into host cells or induce a dangerous systemic anti-DNA immune response. If the cell mutates and/or loses or inactivates the suicide switch, it can become an uncontrollable mutant cell.
  • these cells can fuse with host cells in the subject, and therefore transfer DNA (e.g., mutant DNA).
  • Such fused cells can be dangerous because not all host cells inherit the suicide gene, but can inherit some of the therapeutic cell's genes/DNA during chromosomal reorganization and cell hybridization.
  • nucleated cell therapies and even some cells inactivated by the methods described above can still transfer DNA to the subject since they retain their nucleus and genetic material. Numerous chemicals inhibit cell proliferation and/or cause cell death prior to therapeutic use, including chemotherapeutic drugs and mitomycin C, etc. However, such drugs can have significant off-target effects that significantly damage the cell, which are unwanted for clinical applications due to high toxicities.
  • the present disclosure provides methods for producing therapeutic cytoplasts with either natural or inducible expression and/or uptake of biomolecules with therapeutic functions including, but not limited to, DNA/genes (e.g., plasmids) RNA (e.g., mRNA, shRNA, siRNA, miRNA), proteins, peptides, small molecule therapeutics (e.g., small molecule drugs), gene editing components, nanoparticles, and other therapeutic agents (e.g., bacteria, bacterial spores, bacteriophages, bacterial components, viruses (e.g., oncolytic viruses), exosomes, lipids, or ions).
  • DNA/genes e.g., plasmids
  • RNA e.g., mRNA, shRNA, siRNA, miRNA
  • proteins e.g., proteins
  • peptides e.g., small molecule therapeutics
  • small molecule therapeutics e.g., small molecule drugs
  • gene editing components e
  • cytoplasts as a vehicle to deliver therapeutic cargos to subjects including, but not limited to, DNA/genes (e.g., plasmids), RNA (e.g., mRNA, shRNA, siRNA, miRNA), proteins, peptides, small molecule therapeutics (e.g., small molecule drugs), gene editing components, nanoparticles, and other therapeutic agents (e.g., bacteria, bacterial spores, bacteriophages, bacterial components, viruses (e.g., oncolytic viruses), exosomes, lipids, or ions).
  • DNA/genes e.g., plasmids
  • RNA e.g., mRNA, shRNA, siRNA, miRNA
  • proteins e.g., proteins, peptides, small molecule therapeutics (e.g., small molecule drugs), gene editing components, nanoparticles, and other therapeutic agents (e.g., bacteria, bacterial spores, bacteriophages, bacterial
  • cytoplasts to produce ions, molecules, compounds, complexes, or biomolecules (which can be, for example, secreted, intracellular, inducible, or a combination thereof) including, but not limited to, DNA/genes (e.g., plasmids), RNA (e.g., mRNA, shRNA, siRNA, miRNA), proteins, peptides, small molecule therapeutics (e.g., small molecule drugs), gene editing components, nanoparticles, and other therapeutic agents (e.g., bacteria, bacterial spores, bacteriophages, bacterial components, viruses, exosomes, or lipids).
  • DNA/genes e.g., plasmids
  • RNA e.g., mRNA, shRNA, siRNA, miRNA
  • proteins e.g., proteins
  • peptides e.g., small molecule therapeutics (e.g., small molecule drugs)
  • gene editing components e.g., bacteria
  • the present disclosure provides methods for the largescale in vitro production of therapeutic cytoplasts derived (e.g., obtained) from any nucleated cell type (e.g., a mammalian cell (e.g., a human cell, or any mammalian cell described herein), a protozoal cell (e.g., an amoeba cell), an algal cell, a plant cell, a fungal cell, an invertebrate cell, a fish cell, an amphibian cell, a reptile cell, or a bird cell).
  • the cell can have been immortalized and/or oncogenically transformed naturally or by genetic engineering.
  • the present disclosure provides methods for the use of therapeutic cytoplasts (natural or engineered) as fusion partners to other cells or cytoplasts (therapeutic or natural) to enhance and/or transfer organelles and biomolecules (secreted, intracellular, and natural and inducible) including, but not limited to, mitochondria, ribosomes, endosomes, lysosomes, Golgi, DNA/genes (e.g., plasmids), RNA (e.g., mRNA, shRNA, siRNA, miRNA), proteins (e.g., cytokines, growth factors, and protein hormones), peptides, small molecule therapeutics (e.g., small molecule drugs), gene editing components, nanoparticles, and other therapeutic agents (e.g., bacteria, bacterial spores, bacteriophages, bacterial components, viruses (e.g., oncolytic viruses), exosomes, lipids, or ions).
  • therapeutic agents e.g., bacteria, bacterial
  • the present disclosure provides methods for the cryopreservation, cryohibernation, storage, and recovery of therapeutic cytoplasts in vitro.
  • the present disclosure provides methods for the use of cytoplasts as biosensors and signal transduction indicators of biological processes and healthy or disease states.
  • the present disclosure enables the generation of a novel nucleus-free, cell-based product that can be used as a therapeutic and/or can be modified or genetically engineered to deliver specific disease-fighting and health-promoting cargos to human or animal subjects in a safe and controllable manner.
  • the present disclosure allows for generation of a new platform of safe, nuclear-free cell therapeutics derived from any nucleated cell type, either normal or engineered, including, but not limited to, iPSC (induced pluripotent stem cells), any immortalized cell, stem cells, primary cells (e.g., host-derived cells), cell lines, any immune cell, or cancerous cells.
  • iPSC induced pluripotent stem cells
  • any immortalized cell stem cells
  • primary cells e.g., host-derived cells
  • cell lines any immune cell, or cancerous cells.
  • a significant problem with many existing cell-based therapeutics is that after delivery to the body, the cells proliferate uncontrollably and can permanently engraft into the body, which can be life-threatening.
  • cytoplasts can perform many of the same biological/therapeutic functions as their nucleated counterpart, but do not proliferate or engraft permanently in the subject, since they can have a defined life span (e.g., of 1 hour to 14 days).
  • a defined life span e.g., of 1 hour to 14 days.
  • cytoplasts can be administered to a subject beyond their defined life span (e.g. “dead” cytoplasts).
  • the death process of the administered cytoplasts can have an immunostimulatory effect on the subject.
  • cytoplasts Prior to patient or subject delivery, traditional cell-based therapeutics are commonly modified or genetically altered ex vivo to generate desirable cellular and therapeutic functions. However, when these cells are introduced into the subject, the new host environment can significantly reprogram and negatively alter, or otherwise render them ineffective. Since cytoplasts are devoid of a nucleus, there is no new gene transcription, meaning that, in some embodiments, cytoplasts cannot respond to reprogramming and detrimental external signals. Therefore, cytoplasts can retain their differentiated phenotype and ex vivo engineered therapeutic functions when introduced into the subject, making them a more controllable and predictable therapeutic vehicle. Likewise, normal donor cells that are immediately enucleated can retain their in vivo programs/attributes when transplanted into the subject.
  • cytoplasts can be a more controllable and predictable therapeutic vehicle than traditional cell-based therapeutics because they can retain their in vitro and in vivo phenotypes and biological functions. Such properties are critically important for many therapeutic applications that rely on freshly-derived (e.g., freshly-obtained) donor cells or cells engineered ex vivo to perform specific therapeutic functions
  • nuclear-free cytoplasts can, in some embodiments, be loaded with high doses of DNA-damaging/gene targeting agents for delivery to subjects as a therapeutic against cancer or other diseases.
  • DNA-damaging chemotherapeutic drugs include DNA-integrating viruses, oncolytic viruses, and gene therapy applications/delivery including, but not limited to, cluster regularly interspaced short palindromic repeats (CRISPR), small clusters of cas (CRISPR/Cas system), and plasmids.
  • CRISPR cluster regularly interspaced short palindromic repeats
  • Cas system small clusters of cas
  • plasmids plasmids.
  • cytoplasts can also innately produce a therapeutic effect upon administration to a subject, without the loading of any cargo into the cytoplasts.
  • cytoplasts can be therapeutic without being engineered to produce one or more therapeutics.
  • cytoplasts can be therapeutic with neither the loading of cargo into the cytoplast nor being engineered to produce one or more therapeutics.
  • an unmanipulated cytoplast itself can have therapeutic properties when delivered into a patient or subject.
  • an unmanipulated cytoplast can produce one or more of: a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic protein, a therapeutic peptide, a small molecule therapeutic, and/or a gene-editing factor.
  • unmanipulated cytoplasts e.g., derived from autologous or allogenic sources
  • a dead cytoplast can innately produce a therapeutic effect.
  • cytoplasts can be applied to or cultured with cells (e.g, xenocultured cells) to alter their properties.
  • cytoplasts e.g., unmanipulated cytoplasts or engineered cytoplasts
  • cytoplasts Unlike nucleated cells, cytoplasts cannot undergo DNA damage-induced apoptotic death, and therefore can be used in combination with apoptotic-inducing and/or DNA toxic/targeting agents for treatment of cancer and other diseases.
  • Cytoplasts are smaller than their nucleated counterparts and for this reason can migrate better through small openings in the vasculature and tissue parenchyma. In addition, removing the large dense nucleus alleviates a major physical barrier allowing the cell to move freely through small openings in the vessels and tissue parenchyma. Therefore, cytoplasts can have improved bio-distribution in the body and movement into target tissues.
  • cytoplasts Unlike nucleated cells, the fusion of cytoplasts to the same or another cell type of similar or different origin generates a unique cell hybrid that lacks problematic nuclear transfer, while maintaining desirable therapeutic attributes including, but not limited to, cell surface proteins, signal transduction molecules, secreted proteins, lipids, and epigenetic changes.
  • Exosomes and small cellular membrane vesicles derived from therapeutic cells have been shown, in some instances, to possess therapeutic efficacy alone or as delivery vessels, but are markedly different than and can be limited as compared to cytoplasts.
  • red blood cells RBCs, erythrocytes
  • RBCs red blood cells
  • cytoplasts can be viable cell-like entities that can retain many active biological processes and all cellular organelles (e.g., ER/Golgi, mitochondrial, endosome, lysosome, cytoskeleton, etc.).
  • cytoplasts can function like nucleated cells and exhibit critical biological functions such as adhesion, tunneling nanotube formation, actin-mediated spreading (2D and 3D), migration, chemoattractant gradient sensing, mitochondrial transfer, mRNA translation, protein synthesis, and secretion of exosomes and other bioactive molecules.
  • critical biological functions such as adhesion, tunneling nanotube formation, actin-mediated spreading (2D and 3D), migration, chemoattractant gradient sensing, mitochondrial transfer, mRNA translation, protein synthesis, and secretion of exosomes and other bioactive molecules.
  • 2D and 3D actin-mediated spreading
  • a cytoplast can be derived from any type of nucleated cell, including, but not limited to iPSC (induced pluripotent stem cells), any immortalized cell, stem cells, primary cells (e.g., host-derived cells), cell lines, any immune cell, cancerous cells, or from any eukaryotic cell.
  • iPSC induced pluripotent stem cells
  • stem cells stem cells
  • primary cells e.g., host-derived cells
  • cell lines e.g., any immune cell, cancerous cells, or from any eukaryotic cell.
  • a limitation to development of cell-based therapeutics for clinical use can be the inability and inefficiency of producing large enough quantities of therapeutic cells, especially stem cells.
  • immortalized cells hTERT, viruses, and oncogenes
  • FDA Food and Drug Administration
  • the present disclosure can allow for the use of immortalized cells, cancer cells (e.g., any cancer cell), primary (e.g., host-derived) cells, or a cell line for large-scale production of therapeutic cells, because such cells are enucleated prior to delivery to render them a safe therapeutic.
  • the present disclosure also allows for generating more cells, cell lines, and/or immortalized cell from individual subjects for large-scale manufacturing and bio-banking for use in autologous or allogenic therapies (see, e.g., FIG. 1 ). This can greatly increase the consistency and quality control of cell-based therapeutics, which can be offered as an off-the-shelf product.
  • cytoplasts can be superior therapeutic vehicles compared to manmade synthetic nanoparticles and liposome formulations because they are derived (e.g., obtained) from cells, and thus are fully functioning cell entities (minus the nucleus), therefore exhibiting crucial physiological functions, cellular attributes/organelles, and physiological capabilities to produce bioactive molecules with reduced subject toxicity.
  • a nucleated cell e.g., an eukaryotic cell, a mammalian cell (e.g., a human cell, a canine cell, a feline cell, an equine cell, a porcine cell, a primate cell, a bovine cell, an ovine cell, a rodent cell (e.g., a mouse cell, a guinea pig cell, a hamster cell, or a mouse cell)), an immune cell, or any nucleated cell described herein), is treated with cytochalasin B to soften the cortical actin cytoskeleton.
  • cytochalasin B e.g., an eukaryotic cell, a mammalian cell (e.g., a human cell, a canine cell, a feline cell, an equine cell, a porcine cell, a primate cell, a bovine cell, an ovine cell, a rodent cell (e.g.,
  • cytoplast or “recombinant cytoplast” are used interchangeably and refer to a nucleus-free cell that was obtained from a previously nucleated cell (e.g., any cell described herein) that consists of the inner mass of a cell and the cell organelles.
  • a cytoplast can express a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic protein, a therapeutic peptide, a small molecule therapeutic, and/or a gene-editing factor.
  • a cytoplast can contain a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic protein, a therapeutic peptide, a small molecule therapeutic, and/or a gene-editing factor, a nanoparticle, or another therapeutic agent.
  • an empty cytoplast e.g., a cytoplast with no exogenous components is used as a negative control.
  • cytoplasts can be engineered to express, for example, chemokine receptors, adhesion molecules, antigens, or other markers that can improve the homing of the cytoplasts to sites in a subject, or stimulate and/or modulate desired immune reactions.
  • a cytoplast can be engineered to express an anti-PD-L1 antibody.
  • a nucleated cell can be cultured (e.g., in a suspension, as adherent cells, as adherent cells in 3D (e.g., in semi-suspension or other nonadherent methods)) or clonally selected/expanded before enucleation.
  • a cytoplast has a defined life span of less than 1 hour to 14 days (e.g., less than 1 hour to 1 hour, less than 1 hour to 6 hours, 6 hours to 12 hours, 12 hours to 1 day, 1 day, 2 days, 3 days, 4 days, 5, days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 13 days, 14 days, 1 to 14 days, 1 to 12 days, 1 to 10 days, 1 to 9 days, 1 to 8 days, 1 to 7 days, 1 to 6 days, 1 to 5 days, 1 to 4 days, 1 to 3 days, 1 to 2 days, 2 to 14 days, 2 to 12 days, 2 to 10 days, 2 to 8 days, 2 to 7 days, 2 to 6 days, 2 to 5 days, 2 to 4 days, 2 to 3 days, 3 to 14 days, 3 to 12 days, 3 to 10 days, 3 to 8 days, 3 to 7 days, 3 to 6 days, 3 to 14 days, 4 to 12 days, 4 to 12 days, 4 to 12 days, 4 to 12 days, 4 to 12 days, 3 to
  • the lifespan of a population of cytoplasts can be evaluated by determining the average time at which a portion of the cytoplast population (e.g., at least 50%, at least 60% at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 98% of the population) is determined to be dead.
  • Cell death can be determined by any method known in the art.
  • the viability of cytoplasts, e.g., at one or more time points can be evaluated by determining whether morphometric or functional parameters are intact (e.g.
  • the life span of a cytoplast may be related to the life span of the cell from which it was obtained. For example, in some embodiments, a cytoplast obtained from a macrophage may live 12 to 24 hours.
  • a cytoplast is not a naturally occurring enucleated cell. In some embodiments, a cytoplast is not obtained from a cell that naturally undergoes enucleation. In some embodiments, a cytoplast is not a cell that has been enucleated by in the body of a subject. In some embodiments, a cytoplast is not obtained from a cell that would be enucleated by in the body of a subject. In some embodiments, a cytoplast is not obtained from an erythroblast. In some embodiments, a cytoplast is obtained from a cell that maintains a nucleus over its lifespan (e.g., in the absence of manipulations such as enucleation as described herein).
  • a cytoplast is not a cell that is found in a subject as an anucleate cell (e.g., a red blood cell (erythrocyte), a platelet, a lens cell, or an immediate nucleated precursor thereof).
  • a cytoplast includes one or more components selected from the group consisting of an endoplasmic reticulum, a Golgi apparatus, mitochondria, ribosomes, proteasomes, or spliceosomes.
  • a cytoplast is characterized by one or more of the following features: adhesion, tunneling nanotube formation, actin-mediated spreading (2D and/or 3D), migration, chemoattractant gradient sensing, mitochondrial transfer, mRNA translation, protein synthesis, and secretion of exosomes and/or other bioactive molecules.
  • a cytoplast is characterized by an ability to secrete proteins (e.g., using exosomes).
  • a cytoplast has been enucleated ex vivo.
  • a cytoplast has been enucleated in vitro.
  • a cytoplast has been physically enucleated (e.g., by centrifugation).
  • a cytoplast is an engineered enucleated cell. In some embodiments, a cytoplast is not a red blood cell. In some embodiments, a cytoplast does not contain hemoglobin. In some embodiments, a cytoplast does not have a bi-concave shape.
  • a cytoplast is not obtained from an erythroblast. In some embodiments, a cytoplast is obtained from a cell that would not become a red blood cell (erythrocyte). In some embodiments, a cytoplast is obtained from a lymphoid progenitor cell. In some embodiments, a cytoplast is obtained from a lymphocyte. In some embodiments, a cytoplast is obtained from a mesenchymal stem cell (e.g., from bone marrow). In some embodiments, a cytoplast is obtained from an endothelial stem cell. In some embodiments, a cytoplast is obtained from a neural stem cell. In some embodiments, a cytoplast is obtained from a skin stem cell.
  • a cytoplast is at least 1 ⁇ m in diameter. In some embodiments, a cytoplast is greater than 1 ⁇ m in diameter. In some embodiments, a cytoplast is 1-100 ⁇ m in diameter (e.g., 1-90 ⁇ m, 1-80 ⁇ m, 1-70 ⁇ m, 1-60 ⁇ m, 1-50 ⁇ m, 1-40 ⁇ m, 1-30 ⁇ m, 1-20 ⁇ m, 1-10 ⁇ m, 1-5 ⁇ m, 5-90 ⁇ m, 5-80 ⁇ m, 5-70 ⁇ m, 5-60 ⁇ m, 5-50 ⁇ m, 5-40 ⁇ m, 5-30 ⁇ m, 5-20 ⁇ m, 5-10 ⁇ m, 10-90 ⁇ m, 10-80 ⁇ m, 10-70 ⁇ m, 10-60 ⁇ m, 10-50 ⁇ m, 10-40 ⁇ m, 10-30 ⁇ m, 10-20 ⁇ m, 10-15 ⁇ m 15-90 ⁇ m, 15-80 ⁇ m, 15-70 ⁇ m, 15-60 ⁇
  • a cytoplast is 10-30 ⁇ m in diameter. In some embodiments, the diameter of a cytoplast is between 5-25 ⁇ m (e.g., 5-20 ⁇ m, 5-15 ⁇ m. 5-10 ⁇ m, 10-25 ⁇ m, 10-20 ⁇ m, 10-15 ⁇ m, 15-25 ⁇ m, 15-20 ⁇ m, or 20-25 ⁇ m). In some embodiments, a cytoplast is not an exosome. Without being bound by any particular theory, it is believed that, in some cases, some cytoplasts can advantageously be small enough to allow for better biodistribution or to be less likely to be trapped in the lungs of a subject.
  • the term “eukaryotic cell” refers to a cell having a distinct, membrane-bound nucleus. Such cells may include, for example, mammalian (e.g., rodent, non-human primate, or human), non-mammalian animal (e.g., fish, bird, reptile, or amphibian), invertebrate, insect, fungal, or plant cells.
  • the eukaryotic cell is a yeast cell, such as Saccharomyces cerevisiae .
  • the eukaryotic cell is a higher eukaryote, such as mammalian, avian, plant, or insect cells.
  • the nucleated cell is a primary cell.
  • the nucleated cell is an immune cell (e.g., a lymphocyte (e.g., a T cell, a B cell), a macrophage, a natural killer cell, a neutrophil, a mast cell, a basophil, a dendritic cell, a monocyte, a myeloid-derived suppressor cell, an eosinophil).
  • the nucleated cell is a phagocyte or a leukocyte.
  • the nucleated cell is a stem cell (e.g., an adult stem cell (e.g., a hematopoietic stem cell, a mammary stem cell, an intestinal stem cell, mesenchymal stem cell, an endothelial stem cell, a neural stem cell, an olfactory adult stem cell, a neural crest stem cell, a testicular cell), an embryonic stem cell, an inducible pluripotent stem cell (iPS)).
  • the nucleated cell is a progenitor cell.
  • the nucleated cell is from a cell line.
  • the nucleated cell is a suspension cell.
  • the nucleated cell is an adherent cell. In some embodiments, the nucleated cell is a cell that has been immortalized by expression of an oncogene. In some embodiments, the nucleated cell is immortalized by the expression of human telomerase reverse transcriptase (hTERT) or any oncogene. In some embodiments, the nucleated cell is a patient or subject derived cell (e.g., an autologous patient-derived cell, or an allogenic patient-derived cell).
  • hTERT human telomerase reverse transcriptase
  • the nucleated cell is a patient or subject derived cell (e.g., an autologous patient-derived cell, or an allogenic patient-derived cell).
  • the nucleated cell is transfected with a vector (e.g., a viral vector (e.g., a retrovirus vector (e.g., a lentivirus vector), an adeno-associated virus (AAV) vector, a vesicular virus vector (e.g., vesicular stomatitis virus (VSV) vector), or a hybrid virus vector), a plasmid) before the nucleated cell is enucleated using any of the enucleation techniques described herein and known in the art.
  • a viral vector e.g., a retrovirus vector (e.g., a lentivirus vector), an adeno-associated virus (AAV) vector, a vesicular virus vector (e.g., vesicular stomatitis virus (VSV) vector), or a hybrid virus vector
  • a vector e.g., a viral vector (e.g., a retrovirus vector (e.g
  • Cells can be maintained in vitro under conditions that favor growth, proliferation, viability, differentiation and/or induction of specific biological functions with therapeutic capabilities/benefits including, but not limited to, 3-dimensional culturing, hypoxic environments, culturing on defined extracellular matrix components, treatment with chemical agents, cytokines, growth factors or exposure to any exogenous agent natural or synthetic that induces a specific desirable cell response.
  • cell therapies already used or in development can be enucleated (e.g., using any of the methods disclosed herein) to form cytoplasts.
  • Non-limiting examples of cell therapies already used or in development include: treatment of cancer using chimeric antigen receptor engineered T cells (CAR-T), NK or macrophages; treatment of inflammatory diseases including cancer, autoimmune (Crohn's, rheumatoid arthritis, all types of arthritis, and the like), pancreatitis; regenerative medicine applications, wound healing, bone or cartilage repair, and the like; treatment of cognitive diseases such as Alzheimer's, Parkinson's, and the like; treatments of graft-vs-host disease; gene therapy (e.g., for sickle cell anemia, Severe Combined Immunodeficiency (ADA-SCID/X-SCID), cystic fibrosis, hemophilia, Duchenne's muscular dystrophy, Huntington's disease, Parkinson's, hypercholesterolemia, Alpha-1 antitryps
  • cytoplasts can be engineered using any of the methods described herein.
  • such cytoplasts can be engineered to express therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic protein, a therapeutic peptide, a small molecule therapeutic, and/or a gene-editing factor.
  • a cytoplast can contain a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic protein, a therapeutic peptide, a small molecule therapeutic, and/or a gene-editing factor, a nanoparticle, or another therapeutic agent.
  • a cytoplast can be engineered to express, for example, chemokine receptors, adhesion molecules, antigens, or other markers that can improve the homing of the cytoplasts to sites in a subject, or stimulate and/or modulate desired immune reactions.
  • a cytoplast can be engineered to express an anti-PD-L1 antibody.
  • the cytoplast is cooled or frozen for later use.
  • Various methods of preserving cells are known in the art, including, but not limited to, the use of a serum (e.g., Fetal Bovine Serum) and dimethyl sulfoxide (DMSO) at ultralow temperatures (frozen cryopreservation) or hibernation media for storage at 4 degrees Celsius (cryohibernation).
  • a serum e.g., Fetal Bovine Serum
  • DMSO dimethyl sulfoxide
  • the cytoplast is thawed prior to use.
  • RNA molecules e.g., mRNA, miRNA, siRNA, shRNA, lncRNA
  • DNA molecule e.g., a plasmid
  • a protein e.g., a protein
  • a gene-editing factor e.g., a CRISPR/Cas9 gene-editing factor
  • a peptide e.g., a plasmid
  • cytoplast e.g., a cytoplast derived from any cell described herein.
  • Non-limiting examples of methods that can be used to introduce a biomolecule into a cytoplast include: electroporation, microinjection, lipofection, transfection, calcium phosphate transfection, dendrimer-based transfection, cationic polymer transfection, cell squeezing, sonoporation, optical transfection, impalection, hydrodynamic delivery, magnetofection, and nanoparticle transfection.
  • introducing further includes expressing the biomolecule in a cytoplast.
  • Various expression vectors are known in the art and can be used herein. Non-limiting examples of expression vectors are provided herein. In some embodiments, the expression vector is the vector shown in any one of FIG. 16A , FIG. 17A , FIG. 18A or FIG. 23 .
  • Various gene-editing factors are known in the art. Non-limiting examples of gene-editing factors include: CRISPR/Cas9 gene-editing, transcription activator-like effector nuclease (TALEN), and zinc finger nucleases.
  • a therapeutic agent, a virus, an antibody, drug, or a nanoparticle is introduced into the cytoplasts.
  • a therapeutic DNA, a therapeutic RNA, a therapeutic protein e.g., an enzyme, an antibody, an antigen, a toxin, cytokine, a protein hormone, a growth factor, a cell surface receptor, or a vaccine, or any therapeutic protein that is currently available or in development
  • a therapeutic peptide e.g., a peptide hormone or an antigen, or any therapeutic peptide that is currently available or in development
  • a small molecule therapeutic e.g., steroid, a polyketide, an alkaloid, a toxin, an antibiotic, an antiviral, an analgesic, an anticoagulant, an antidepressant, an anticancer drug, an antiepileptic, an antipsychotic, a sedative, a colchicine, a taxol
  • the cytoplasts can be treated (e.g. stimulated with or loaded) with exosomes.
  • treatment with exosomes can be used to introduce a biomolecule, a therapeutic, a therapeutic peptide, a small molecule therapeutic, a therapeutic gene editing factor, a therapeutic nanoparticle, or another therapeutic agent (e.g., bacteria, bacterial spores, bacteriophages, bacterial components, viruses (e.g., oncolytic viruses), exosomes, lipids, or ions into the cytoplasts.
  • treatment with exosomes can be used to alter the behavior, signaling, secreted factors, or other characteristics of the cytoplasts.
  • the present methods include the use of cytoplasts for treating a disease (e.g., a cancer/neoplasm, an infection, an inflammatory condition, a neurological disease (e.g., a neurodegenerative disease), a degenerative disease, an autoimmune disease, a cardiovascular disease, an ischemic disease, a genetic or inherited disorder, a developmental disorder, an ophthalmologic disease, a skeletal disease, a metabolic disease, a toxicosis, an idiopathic condition, or two or more thereof), in a subject.
  • a disease e.g., a cancer/neoplasm, an infection, an inflammatory condition, a neurological disease (e.g., a neurodegenerative disease), a degenerative disease, an autoimmune disease, a cardiovascular disease, an ischemic disease, a genetic or inherited disorder, a developmental disorder, an ophthalmologic disease, a skeletal disease, a metabolic disease, a toxicosis, an idiopathic condition, or two or more
  • Non-limiting examples of cancers include: acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), cancer in adolescents, adrenocortical carcinoma, anal cancer, appendix cancer, astrocytoma, atypical teratoid/rhabdoid tumor, basal cell carcinoma, bile duct cancer, bladder cancer, bone cancer, brain stem glioma, brain tumor, breast cancer, bronchial tumor, Burkitt lymphoma, carcinoid tumor, unknown primary carcinoma, cardiac tumors, cervical cancer, childhood cancers, chordoma, chronic lymphocytic leukemia (CLL), chronic myelogenous leukemia (CML), chronic myeloproliferative neoplasms, colon cancer, colorectal cancer, craniopharyngioma, cutaneous T-cell lymphoma, bile duct cancer, ductal carcinoma in situ, embryonal tumors, endometrial cancer, ependymoma,
  • Non-limiting types of infections include viral infections, bacterial infections, fungal infections, parasitic infections, and protozoal infections.
  • infections include Acinetobacter infections, Actinomycosis, African sleeping sickness (African trypanosomiasis), AIDS (Acquired immunodeficiency syndrome), Amebiasis, Anaplasmosis, Angiostrongyliasis, Anisakiasis, Anthrax, Arcanobacterium haemolyticum infection, Argentine hemorrhagic fever, Ascariasis, Aspergillosis, Astrovirus infection, Babesiosis, Bacillus cereus infection, Bacterial pneumonia, Bacterial vaginosis, Bacteroides infection, Balantidiasis, Bartonellosis, Baylisascaris infection, BK virus infection, Black piedra , Blastocystosis, Blastomycosis, Venezuelan hemorrhagic fever, Botulism (and Infant bot
  • Non-limiting examples of neurological diseases include Amyotrophic lateral sclerosis (ALS), Alzheimer's disease, Bell's palsy, brain aneurysm, brain injury, brain tumor, cerebral palsy, chronic fatigue syndrome, concussion, dementia, epilepsy, Guillain-Barre syndrome, headache, Huntington's disease migraine, multiple sclerosis, muscular dystrophy, Neuralgia, neuropathy, neuromuscular and related diseases, Parkinson's disease, psychiatric conditions (e.g., depression, obsessive-compulsive disorder), scoliosis, seizures, spinal cord injury, spinal deformity, spinal disorder (e.g., subacute combined degeneration), spine tumor, stroke, and vertigo.
  • ALS Amyotrophic lateral sclerosis
  • Alzheimer's disease Bell's palsy
  • brain aneurysm brain injury
  • brain tumor e.g., cerebral palsy
  • chronic fatigue syndrome concussion
  • dementia epilepsy
  • Non-limiting examples of autoimmune diseases include Achalasia, Addison's disease, Adult Still's disease, Agammaglobulinemia, Alopecia areata, Amyloidosis, Ankylosing spondylitis, Anti-GBM/Anti-TBM nephritis, Antiphospholipid syndrome, Autoimmune angioedema, Autoimmune dysautonomia, Autoimmune encephalomyelitis, Autoimmune hepatitis, Autoimmune inner ear disease (AIED), Autoimmune myocarditis, Autoimmune oophoritis, Autoimmune orchitis, Autoimmune pancreatitis, Autoimmune retinopathy, Axonal & neuronal neuropathy (AMAN), Balo disease, Behcet's disease, Benign mucosal pemphigoid, Bullous pemphigoid, Castleman disease (CD), Celiac disease, Chagas disease, Chronic inflammatory demyelinating polyn
  • Non-limiting examples of cardiovascular diseases include acute myocardial infarction, heart failure, refractory angina, coronary artery disease, rheumatic heart disease, congenital heart disease, stroke, aortic aneurism and/or dissection, peripheral arterial disease, deep vein thrombosis, pulmonary embolism, tumors of the heart, vascular tumors of the brain, cardiomyopathy, heart valve diseases, and pericardial disease.
  • Non-limiting examples of ophthalmologic diseases include glaucoma, cataract, macular degeneration, diabetic retinopathy, strabismus, retinal detachment, uveitis, amblyopia, dry eye syndrome, keratitis, macular edema, corneal ulcer, optic neuropathy, cytomegalovirus retinitis, corneal dystrophy, hyphema, trachoma, central serous retinopathy, retinopathy of prematurity, endophthalmitis, Leber's congenital amaurosis, central retinal artery occlusion, trichiasis, papilledema, Graves' ophthalmopathy, uveal melanoma, branch retinal vein occlusion, choroideremia, and maculopathy.
  • Non-limiting examples of skeletal diseases include osteochondrodysplasia, achondroplasia, hypophospatasia, achondrogenesis, thanatrophoric dysplasia, osteomalacia, rickets, osteopenia, osteoporosis, Paget's disease, osteomyelitis, osteolysis, Haju-Cheney syndrome, hypertrophic pulmonary osteoarthropathy, nonossifying fibroma, pseudarthrosis, fibrous dysplasia, hyperostosis, osteocsclerosis, and pycnodysostosis.
  • Non-limiting examples of metabolic diseases include cystinuria, Fabry disease, galactosemia, Gaucher disease (type I), Hartnup disease, homocystinuria, Hunter syndrome, Hurler syndrome, Lesch-Nyhan syndrome, maple syrup urine disease, Maroteaux-Lamy syndrome, Morquio syndrome, Niemann-Pick disease (type A), phenylketonuria, Pompe disease, porphyria, Scheie syndrome, Tay-Sachs disease, tyrosinemia (hepatorenal), von Gierke disease (glycogen storage deficiency type IA), and Wilson's disease.
  • cystinuria cystinuria
  • Fabry disease type I
  • Hartnup disease homocystinuria
  • Hunter syndrome Hurler syndrome
  • Lesch-Nyhan syndrome maple syrup urine disease
  • Maroteaux-Lamy syndrome Morquio syndrome
  • Niemann-Pick disease type A
  • phenylketonuria Pompe disease
  • porphyria porphyri
  • the subject is in need of, has been determined to be in need of, or is suspected to be in need of a cytoplast treatment.
  • the cancer can be, e.g., acute myeloid leukemia, bladder cancer, breast cancer, kidney cancer, melanoma, small cell lung cancer, non-small cell lung cancer, pancreatic cancer, or prostate cancer.
  • a cytoplast can be used for diagnosis of a disease or condition (e.g., a cancer or a neoplasm, an infection, an inflammatory condition, a neurological disease (e.g., a neurodegenerative disease), a degenerative disease, an autoimmune disease, a cardiovascular disease, an ischemic disease, a genetic or inherited disorder, a developmental disorder, an ophthalmologic disease, a skeletal disease, a metabolic disease, a toxicosis, an idiopathic condition, or two or more thereof).
  • a disease or condition e.g., a cancer or a neoplasm, an infection, an inflammatory condition, a neurological disease (e.g., a neurodegenerative disease), a degenerative disease, an autoimmune disease, a cardiovascular disease, an ischemic disease, a genetic or inherited disorder, a developmental disorder, an ophthalmologic disease, a skeletal disease, a metabolic disease, a toxicosis, an idiopathic condition, or
  • cytoplasts as described herein (e.g., unmanipulated cytoplasts, or cytoplasts genetically engineered or loaded exogenously with bioreporter molecules, or with inducible bioreporter molecules) that can signify a subject's particular health, disease state, condition, or toxin level.
  • the cytoplasts and/or a molecule expressed or secreted by, or contained within, the cytoplast can operate as a bioreporter.
  • bioreporters can be used in subjects or ex vivo.
  • a sample can be obtained from a subject (e.g., blood, urine, stool, or tissue (e.g., a biopsy)).
  • the cytoplasts can express or contain, e.g., colorimetric, fluorescent, luminescent, chemiluminescent or electrochemical molecules that report a measurable clinical signal.
  • the signal can be proportional to the concentration of a chemical, physical agent, or a biomolecule (e.g. growth factors, insulin, cancer antigens, immune factors), or proportional to gene transcriptional activity, or protein translational activity in a subject or a sample from a subject.
  • a subject refers to any organism.
  • a subject can be a mammal, amphibian, fish, reptile, invertebrate, bird, plant, archaea, fungus, or bacteria.
  • the subject is a mammal.
  • the subject may be a rodent (e.g., a mouse, a rat, a hamster, a guinea pig), a canine (e.g., a dog), a feline (e.g., a cat), an equine (e.g., a horse), an ovine, a bovine, a porcine, a non-human primate, e.g., a simian (e.g., a monkey), an ape (e.g., a gorilla, a chimpanzee, an orangutan, a gibbon), or a human.
  • a rodent e.g., a mouse, a rat, a hamster, a guinea pig
  • a canine e.g., a dog
  • a feline e.g., a cat
  • an equine e.g., a horse
  • the subject is between 0 and 120 years old (e.g., between birth and one month (e.g., a neonate), between one month and two years (e.g., an infant), between 2 years and 12 years (e.g., a child), between twelve years and sixteen years (e.g., an adolescent), between 1 and 120 years old, between 1 and 115 years old, between 1 and 110 years old, between 1 and 105 years old, between 1 and 100 years old, between 1 and 95 years old, between 1 and 90 years old between 1 and 85 years old, between 1 and 80 years old, between 1 and 75 years old, between 1 and 70 years old, between 1 and 65 years old, between 1 and 60 years old, between 1 and 50 years old, between 1 and 40 years old, between 1 and 30 years old, between 1 and 25 years old, between 1 and 20 years old, between 1 and 15 years old, between 1 and 10 years old, between 5 and 120 years old, between 5 and 110 years old, between 5 and 100 years old (e.g., between birth and one
  • the subject is not yet born, e.g., in utero. In some embodiments of any of the methods described herein, the subject is at least 1 month old (e.g., at least 2 years old, at least 12 years old, at least 16 years old, or at least 18 years old). Any of the methods described herein can be used to treat a subject, e.g., a diseased subject (i.e., a subject with a disease, e.g., who has been diagnosed with a disease), or an asymptomatic subject (i.e., a subject who clinically presents as healthy, or who has not been diagnosed with a disease).
  • a diseased subject i.e., a subject with a disease, e.g., who has been diagnosed with a disease
  • an asymptomatic subject i.e., a subject who clinically presents as healthy, or who has not been diagnosed with a disease.
  • treating includes “prophylactic treatment” which means reducing the incidence of or preventing (or reducing risk of) a sign or symptom of a disease in a subject at risk for the disease, and “therapeutic treatment”, which means reducing signs or symptoms of a disease, reducing progression of a disease, reducing severity of a disease, re-occurrence in a subject diagnosed with the disease.
  • therapeutic treatment means to ameliorate at least one clinical parameter of the disease, and/or to provide benefits (e.g., anti-aging, anti-scarring, wound healing, anti-depressant, anti-inflammatory, weight loss).
  • disease refers to an abnormality in a subject or any deviation from a healthy state in a subject.
  • diseases and/or conditions include a cancer or a neoplasm, an infection, an inflammatory condition, a neurological disease (e.g., a neurodegenerative disease), a degenerative disease, an autoimmune disease, a cardiovascular disease, an ischemic disease, a genetic or inherited disorder, a developmental disorder, an ophthalmologic disease, a skeletal disease, a metabolic disease, a toxicosis, or an idiopathic condition.
  • the composition is administered at least once (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, 40, 50, 60, 70, 80, 90, 100 times) during a period of time (e.g., every day, every 2 days, twice a week, once a week, every week, three times per month, two times per month, one time per month, every 2 months, every 3 months, every 4 months, every 5 months, every 6 months, every 7 months, every 8 months, every 9 months, every 10 months, every 11 months, once a year).
  • a period of time e.g., every day, every 2 days, twice a week, once a week, every week, three times per month, two times per month, one time per month, every 2 months, every 3 months, every 4 months, every 5 months, every 6 months, every 7 months, every 8 months, every 9 months, every 10 months, every 11 months, once a year.
  • monthly treatments e.g., administering at least once per month for at least 1 month (e.g., at least two, at least three, at least four, at least five, at least six or more months, e.g., 12 or more months), and yearly treatments (e.g., administration once a year for one or more years).
  • Administration can be via any route known in the art, e.g., subcutaneous, intravenous, arterial, ocular, oral, intramuscular, intranasal (e.g., inhalation), intraperitoneal, topical, mucosal, epidural, sublingual, epicutaneous, extra-amniotic, inter-articular, intradermal, intraosseous, intrathecal, intrauterine, intravaginal, intravesical, intravitreal, perivascular, and/or rectal administration, or any combination of known administration methods.
  • route known in the art e.g., subcutaneous, intravenous, arterial, ocular, oral, intramuscular, intranasal (e.g., inhalation), intraperitoneal, topical, mucosal, epidural, sublingual, epicutaneous, extra-amniotic, inter-articular, intradermal, intraosseous, intrathecal, intrauterine, intravaginal, intravesical, intravitreal, perivascular, and/or rectal
  • the death process of cytoplasts can have a therapeutic effect on a subject.
  • the death process of cytoplasts can be immunostimulatory.
  • methods of administering cytoplasts to a subject wherein the death of the cytoplasts has a therapeutic effect on the subject.
  • the cytoplasts administered to the subject are dead.
  • the cytoplasts administered to the subject when administered, have a remaining life span of less than 5 days (e.g., less than 4 days, less than 3 days, less than 2 days, less than 36 hours, less than 1 day, less than 18 hours, less than 12 hours, less than 6 hours, less than 2 hours, or less than 1 hour).
  • cells can be removed from a subject and enucleated.
  • the cells are engineered (e.g., to produce or contain a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic protein, a therapeutic peptide, a small molecule therapeutic, a therapeutic gene-editing factor a therapeutic nanoparticle and/or another therapeutic agent) before being enucleated.
  • cells from a subject are enucleated, and then engineered (e.g., to produce or contain a therapeutic DNA molecule, a therapeutic RNA molecule, a therapeutic protein, a therapeutic peptide, a small molecule therapeutic, a therapeutic gene-editing factor a therapeutic nanoparticle and/or another therapeutic agent).
  • the cytoplasts are administered to the subject from which the cells were removed.
  • the media in which the cytoplasts were cultured and/or stored can have a therapeutic benefit.
  • the media in which cytoplasts were co-cultured and/or stored (e.g., after enucleation) with cells can have a therapeutic benefit.
  • the media in which cytoplasts fused with cells were cultured and/or stored with cells can have a therapeutic benefit.
  • the therapeutic benefit of cultured media can be due to the presence in the media of exosomes (e.g., containing therapeutic protein) secreted by the cytoplasts.
  • the composition is administered with one or more additional therapies (e.g., any drug (e.g., antibiotics, antivirals, anti-inflammatory medications) or chemotherapy (e.g., a chemotherapeutic agent (e.g., doxorubicin, paclitaxel, cyclophosphamide), or any of the small molecule therapeutics described herein), cell-based therapy, radiation therapy, immunotherapy, a small molecule, an inhibitory nucleic acid (e.g., antisense RNA, antisense DNA, miRNA, siRNA, lncRNA), an exosome-based therapy, gene therapy or surgery).
  • additional therapies e.g., any drug (e.g., antibiotics, antivirals, anti-inflammatory medications) or chemotherapy (e.g., a chemotherapeutic agent (e.g., doxorubicin, paclitaxel, cyclophosphamide), or any of the small molecule therapeutics described herein), cell-based therapy, radiation therapy,
  • the composition further includes one or more additional therapies (e.g., any drug (e.g., antibiotics, antivirals) or chemotherapy (e.g., a chemotherapeutic agent (e.g., doxorubicin, paclitaxel, cyclophosphamide)), cell-based therapy, radiation therapy, immunotherapy, a small molecule, an inhibitory nucleic acid (e.g., antisense RNA, antisense DNA, miRNA, siRNA, lncRNA) or surgery).
  • additional therapies e.g., any drug (e.g., antibiotics, antivirals) or chemotherapy (e.g., a chemotherapeutic agent (e.g., doxorubicin, paclitaxel, cyclophosphamide)
  • cell-based therapy e.g., radiation therapy, immunotherapy, a small molecule
  • an inhibitory nucleic acid e.g., antisense RNA, antisense DNA, miRNA, siRNA
  • compositions e.g., pharmaceutical compositions
  • a cytoplast e.g., a cytoplast obtained from any cell described herein.
  • the compositions are formulated for different routes of administration (e.g., intravenous, subcutaneous, intramuscular, retro-orbital, intraperitoneal).
  • the compositions can include a pharmaceutically acceptable carrier (e.g., phosphate buffered saline).
  • cytoplasts are, in some embodiments, much smaller than their parental cells (e.g., about 60% of the diameter of parental cells and 1/8 the volume) and do not have the rigid nucleus, therefore, cytoplasts can pass better through small capillaries and vessels than their parental cells.
  • the specific homing of cells to the diseased tissues can depend on the chemokine receptor signaling such as SDF-1 ⁇ /CXCR4, CCL2/CCR2, and the adhesion molecules such as PSGL-1.
  • cytoplasts can be engineered to specifically express functional CXCR4, CCR2 as well as glycosylated PSGL-1, which can greatly promote the specific homing of the engineered cytoplasts.
  • the cytoplasts can further include (e.g. by engineering or from the cell from which they were obtained) a targeting moiety that is expressed on the cell surface of the cytoplast, e.g., CXCR4, CCR2 or PSGL-1.
  • a targeting moiety that is expressed on the cell surface of the cytoplast, e.g., CXCR4, CCR2 or PSGL-1.
  • cell surface proteins that may be expressed on the cell surface of the cytoplastinclude: chemokines such as CXCR4, CCR2, CCR1, CCR5, CXCR7, CXCR2, and CXCR1.
  • the cytoplasts can further include (e.g.
  • the cytoplasts can further include (e.g. by engineering or from the cell from which they were obtained) a surface marker that aids in their evasion of the subject immune system.
  • the cytoplasts can include a CD47 marker.
  • Non-limiting examples of cell-matrix receptors and cell-cell adhesion molecules include integrins, cadherins, glycoproteins, and heparin sulfate proteoglycans.
  • Non-limiting examples of therapeutic molecules include tumor antigens and immunomodulatory peptides, polyamines, and ATP.
  • the cytoplasts can be stored at a temperature between about ⁇ 80° C. and about 16° C. (e.g., about ⁇ 80° C. and about 12° C., ⁇ 80° C. and about 10° C., about ⁇ 80° C. and about 8° C., about ⁇ 80° C. and about 6° C., about ⁇ 80° C. and about 4° C., about ⁇ 80° C. and about 2° C., about ⁇ 80° C. and about 0° C., about ⁇ 80° C. and about ⁇ 4° C., about ⁇ 80° C. and about ⁇ 10° C., about ⁇ 80° C. and about ⁇ 16° C., about ⁇ 80° C.
  • first day to about 7 days e.g., about 1 day to about 6 days, about 1 day to about 5 days, about 1 day to about 4 days, about 1 day to about 3 days, about 1 day to about 2 days, about 2 days to about 7 days, about 2 days to about 6 days, about 2 days to about 5 days, about 2 days to about 4 days, about 2 days to about 3 days, about 3 days to about 7 days, about 3 days to about 6 days, about 3 days to about 5 days, about 3 days to about 4 days, about 4 days to about 7 days, about 4 days to about 6 days, about 4 days to about 5 days, about 5 days to about 7 days, about 5 days to about 6 days, or about 6 days to about 7 days).
  • kits that include any composition described herein.
  • a kit can include instructions for using any of the compositions or methods described herein.
  • the kits can include at least one dose of any of the compositions described herein.
  • therapeutic cytoplasts can be generated from allogenic or autologous donor-derived cells, and can be used for disease treatment as well as for diagnostics.
  • mammalian cells e.g., mesenchymal stem cells, neutrophils, fibroblast, and natural killer cells
  • FIGS. 2A-D the mammalian cells were enucleated by density gradient centrifugation using discontinuous Ficoll gradients, high-speed centrifugation. Table 1 summarizes the results of enucleation using asuspension protocol.
  • Enucleation efficiency and cell viability was the highest in both hTERT transformed and primary mesenchymal stem cells (MSCs), as well as in fibroblasts and neutrophils.
  • Table 2 summarizes the results of enucleation using an adherent protocol. Enucleation efficiency was greater than 70% in both mesenchymal stem cells and macrophages. This experiment showed that various types of mammalian cells could undergo enucleation using any of the methods described herein.
  • cytoplasts did not. Instead, the relative fold change in viable cytoplasts remained fairly constant for 72 hours before declining at 96 hours. Thus, cytoplast survival spanned 34 days. As most cell-based therapies are not used immediately, the viability of cytoplasts after cryopreservation was determined. Surprisingly, the viability of cytoplast after cryopreservation was greater than the viability of MSC following cryopreservation ( FIG. 3B ). Cytoplasts plated immediately after enucleation and cytoplasts recovered from cryopreservation displayed similar relative cell viability after 24 hours ( FIG. 3C ). This experiment showed that cytoplasts survival was not affected by cryopreservation.
  • cytoplasts after cryohibernation was similar to the viability of MSC following cryohibernation ( FIG. 25A ). Cytoplasts recovered after cryohibernation for various lengths of time were able to undergo induced migration in a Boyden chamber assay similar to MSCs recovered after cryohibernation, ( FIG. 25B ).
  • the therapeutic cytoplast is loaded with therapeutic cargo (e.g., mRNA, drugs, peptides, etc. . . . ) for disease treatment.
  • therapeutic cargo e.g., mRNA, drugs, peptides, etc. . . .
  • the therapeutic cytoplast is prepared for immediate use (e.g., for intravenous injection (IV), intraperitoneal injection (IP), tissue, or in vitro applications) for diagnostic use.
  • Example 2 Cytoplasts Retain Intact Organelles, the Ability to Interact with the Extracellular Matrix, Perform Cell-Biological Functions, and can Serve as Delivery Vehicles with Therapeutic Value
  • FIG. 4 After determining whether cytoplasts could retain viability after cryopreservation, flow cytometry analysis were performed in order to determine whether the cell surface marker profile of MSC-derived cytoplasts differed from bone-marrow derived MSC ( FIG. 4 ). As depicted in FIG. 4 , both MSC-derived cytoplasts and bone-marrow derived MSCs maintained cell surface expression of CD45, CD90, CD44, CD146, and CD166.
  • FIGS. 5A-F ′ and FIG. 6A-D ′ showed that cytoplasts attached, reorganized the cytoskeleton, spread on matrix proteins in 2D and 3D culture systems, and formed tunneling nanotubes, which can transfer bioproducts between cells of the same or different origin.
  • FIGS. 7A-E ′ Organelle-staining indicated that Golgi, ER, F-actin cytoskeleton, lysosomes, endosomes, microtubules, and mitochondria remain intact in cytoplasts ( FIGS. 7A-E ′). Furthermore, cytoplasts exhibited homing potential in vitro. Cytoplasts readily migrated on extracellular matrix proteins and migrated directionally towards soluble chemokine gradients (i.e. via chemosensing) ( FIGS. 8A and 8B ). Notably, cytoplasts transfected exogenously with purified mRNAs produced functional intracellular proteins, which could mimic therapeutic mRNA applications being developed for a variety of clinical uses and disease-states. This also demonstrates that the machineries for mRNA translation and protein synthesis operate normally in cytoplasts in the absence of a nucleus, and thus can be used to produce bioactive molecules with therapeutic value.
  • Cytoplasts transfected exogenously with purified mRNA encoding known secreted proteins produce functional extracellular proteins in conditioned culture media, indicating that the ER/Golgi and secretory pathways operate normally in cytoplasts in the absence of a nucleus ( FIG. 11 ).
  • treatment of macrophages and endothelial cells with cytoplast-conditioned media containing secreted proteins activated key signal transduction responses in these cells ( FIG. 12 ).
  • Cytoplasts can be loaded with various cargo including, but not limited to, siRNA, shRNA, mRNA, DNA plasmids, peptides, and chemotherapeutic agents (see, e.g., FIGS. 9 and 10 ).
  • Example 3 Engineered Cytoplasts can Function Both In Vitro and In Vivo
  • FIGS. 13B and 13C show that MSC-derived cytoplasts can be engineered to produce and secrete therapeutic levels of a functional anti-inflammatory cytokine interleukin 10 (IL-10) in vitro and in a preclinical mouse model following intravenous injection.
  • FIG. 13B shows that cytoplasts transfected with IL-10 mRNA can secrete high levels of IL-10.
  • IL-10 functional anti-inflammatory cytokine interleukin 10
  • CM conditioned medium
  • mice were injected retro-orbitally with MSC or MSC-derived cytoplasts expressing IL-10. Two hours post-injection, blood was collected and the levels of IL-10 were determined. Little to no IL-10 was detected in the blood of mice that were injected with untreated MSC ( FIG. 13D ). As shown in FIG. 13D , higher levels of IL-10 were detected in mice injected with MSC-derived cytoplasts expressing IL-10 as compared to the level in mice injected with untreated MSC.
  • MSC-derived cytoplasts were allowed to invade through the basement membrane towards 10% FBS for 24 hours. As shown in FIGS. 14A and 14B , MSC-derived cytoplasts were just was efficient at invading the basement membrane as untreated MSCs in the presence of 10% FBS. Noteworthy, while untreated MSCs were able to invade the basement membrane in the absence of a chemoattractant, MSC-treated cytoplasts were far less able to invade the basement membrane in the absence of a chemoattractant. These data illustrate that MSC-derived cytoplasts can digest and invade through the basement membrane. These data illustrate the innate potential of cytoplast-based cell therapies to penetrate and migrate through complex extracellular matrix barriers to deliver their cargo(s) within tissues.
  • MSC-derived cytoplasts have an average diameter of 12 ⁇ m, while MSC have an average diameter of 20 ⁇ m.
  • mice were retro-orbitally injected with MSC or MSC-derived cytoplasts.
  • FIGS. 15C and 15D more MSC-derived cytoplasts were detected in the liver than the number of MSC detected in the liver.
  • engineered MSCs expressing CXCR4 and engineered MSC-derived cytoplasts expressing CXCR4 express comparable levels of CXCR4, as determined by flow cytometry.
  • MSCs and MSC-derived cytoplasts expressing CXCR4 receptors were allowed to migrate towards various concentrations of SDF-1 ⁇ .
  • MSC-derived cytoplasts engineered to express functional CXCR4 can migrate towards SDF-1 ⁇ , and cell migration increases with increasing concentrations of SDF-1 ⁇ .
  • the number of migrating MSC-derived cytoplasts was greater than the number of migrating MSCs expressing CXCR4 ( FIG. 16C ).
  • FIGS. 17A-C show that MSC-derived cytoplasts can be engineered to express functional cell adhesion proteins known to mediate cell adhesion to the inflamed vasculature.
  • FIGS. 18A-D show that MSC-derived cytoplasts can be engineered to express cell proteins known to modulate macrophage interactions and phagocytosis of therapeutic cells.
  • Example 5 Cytoplasts can be Engineered to Secrete Functional IL-12, and can Induce the Expression of Inflammatory Genes and Suppress Tumor Growth in a Syngeneic Mouse Model of Breast Cancer
  • MSCs and MSC-derived cytoplasts were transfected with IL-12 mRNA.
  • Conditioned medium (CM) was collected 24 hours, 48 hours and 72 hours post-transfection.
  • MSC-derived cytoplasts secrete IL-12.
  • mouse splenocytes were treated with full media, CM from MSC expressing IL-12, CM from MSC-derived cytoplasts expressing IL-12, and purified IL-12.
  • MSC-derived cytoplasts expressing IL-12 and MSC expressing IL-12 secrete functional IL-12 that can cause phosphorylation of STAT4 in mouse splenocytes ( FIG. 19C ).
  • mice were injected either by retro-orbital or intratumoral administration. The number of deaths was recorded and classified according to injection method and cause of death (Table 3). As shown in Table below, intratumoral administration of cytoplasts was well-tolerated with an excellent safety profile.
  • MSC-derived cytoplasts expressing IL-12 and empty MSC-derived cytoplasts were injected into established E0771 subcutaneous tumors. Forty-eight hours after injection, all mice were euthanized, and tumor samples were collected. As shown in FIG. 19D , tumor IL-12 was detected in tumors isolated from mice that were injected with MSC-derived cytoplasts expressing IL-12, whereas little to no tumor IL-12 was detected in tumors isolated from mice that were injected with empty MSC-derived cytoplasts. Taken together, these results indicate that MSC-derived cytoplasts can produce, secrete and deliver clinically relevant levels of therapeutic cytokines to a diseased tissue in a preclinical mouse model.
  • FIGS. 20A-C show that samples taken from mice injected with cytoplasts engineered to express IL-12 cytokine express interferon gamma (IFN ⁇ ), PD-L1 and CXCL9, whereas samples taken from mice that received only PBS or empty cytoplasts expressed low levels of IFN ⁇ , PD-L1 and CXCL9.
  • IFN ⁇ interferon gamma
  • FIG. 20D shows a decrease in tumor size following injection of MSC-derived cytoplasts engineered to express IL-12.
  • FIGS. 21A-C show that MSC-derived cytoplasts can be loaded with oncolytic viruses and can deliver such viruses to tumors growing in immunocompromised and immunocompetent mice, which in combination with IL-12 secretion promotes infiltration of cytotoxic CD8+ T cells into the tumor.
  • FIG. 21 A it is notable that very few cytoplasts can be detected in the tumor after 7 days, whereas a large number of MSCs are present in the center (injection site) and at the outer edge of the growing tumor.
  • FIGS. 22A-B show that genetically engineered MSC-derived cytoplasts can deliver gene editing proteins to regulate gene function in host cells following cytoplast-host cell fusion. These data illustrate the potential of cytoplast-based cell therapies to deliver gene editing components to modify normal or mutant genes in cells.
  • MSCs were seeded at 2.5 M per 15 cm plate (Olympus 25-203) in 20 mL MSC medium [MEM 1 ⁇ (Gibco 12561-056); 16.5% premium FBS (Atlanta Biologics S1150); 1% HEPES 1M (Gibco 15630-80); 1% Anti-Anti 100 ⁇ (Gibco 15240-062); 1% Glutamax 100 ⁇ (Gibco 35050-061)].
  • MSC medium MSC medium
  • Cells were counted, pelleted, and re-suspended with 12.5% Ficoll.
  • the cell-Ficoll mixture was dropwise passed through a 40 um cell strainer (Falcon 352340) into a new 50 mL tube.
  • a 40 um cell strainer Falcon 352340
  • 3.2 mL of cell suspension was slowly loaded onto the pre-made gradients.
  • One mL of 1 ⁇ MEM buffer was added at the final (top) layer with syringe.
  • the tubes were then loaded into rotor buckets, balanced, and run in the ultracentrifuge (Beckman, L8M) for 60 minutes, 26,000 rpm, 31° C., Accel 7, Deccel 7.
  • 1 M cytoplasts were suspended with warm 1 ml amino acid-free ⁇ -MEM full medium (ThermoFisher 12561056; 16.5% Premium fetal bovine serum (FBS), 1% Glutamax (Gibco 35050061), 1% HEPES (Gibco 15630080)).
  • 1 ⁇ g mRNA was diluted with warm opti-MEM and mixed with pipet at least 20 times.
  • 4 ⁇ l lipofectamine-3000 (ThermoFisher L300015) was added to 46 ⁇ l warm opti-MEM (ThermoFisher 31985062) and mixed with pipet for at least 20 times.
  • the ratio of mRNA and lipofectamine-3000 was 1:4 (w/v).
  • the mRNA and lipofectamine-3000 dilutions were mixed with pipet for at least 20 times and incubated at room temperature for 15 minutes.
  • the mRNA and lipofectamine-3000 mixture was added to the cytoplast suspension, mixed well and incubated at 37° C. for 30 minutes. The suspension was shaken every 5 minutes to prevent cell clumping. After incubation, the cells were centrifuged, and re-suspended in normal ⁇ -MEM full medium (16.5% Premium FBS, 1% Antibiotic-Antimycotic, 1% Glutamax, 1% HEPES) or PBS.
  • cytoplasts were suspended with warm 1 ml A/A free ⁇ -MEM full medium (16.5% Premium FBS, 1% Glutamax, 1% HEPES).
  • Two l siRNA was diluted with warm opti-MEM and mixed with pipet at least 20 times.
  • Eight p lipofectamine-3000 was diluted with 92 ⁇ l warm opti-MEM and mixed with pipet at least 20 times.
  • the ratio of siRNA and lipofectamine-3000 was 1:4 (v/v).
  • the siRNA and lipofectamine-3000 dilutions were mixed with pipet at least 20 times and incubated at room temperature for 15 minutes.
  • the siRNA and lipofectamine-3000 mixture was added to the cytoplast suspension, mixed well and incubated at 37° C.
  • Target cells were plated in one well of 6-well plate at density of 1-2 ⁇ 10 5 cells/well, or 10 cm plate with 0.5-1 M MSCs. The next day, the concentrated recombinant lentivirus was thawed in a 37° C. water bath and removed from the bath immediately once thawed. The cells were then washed with PBS 3 times. 200sL serum free medium or 2 mL serum free medium (1:1250 SureENTRY) was added. The target cells were infected in a 6-well plate with MOI 10:1. The next day, the viral supernatant was removed and the appropriate complete growth medium was added to the cells. After 72 hours incubation, the cells were subcultured into 2 ⁇ 100 mm dishes.
  • the appropriate amount of selection drug i.e. puromycin was added for stable cell-line generation. 10-15 days after selection, clones were picked for expansion and were screened for positive ones. The selected positive clones were expanded for enucleation.
  • Engineered cytoplasts were prepared as outlined above. The target protein expression on cytoplasts was determined by ordinary biochemical methods or functional assays, e.g., fluorescent activated cell sorting (FACS), western blot, or Boyden chamber assay.
  • FACS fluorescent activated cell sorting
  • western blot or Boyden chamber assay.
  • Arg9(FAM) (10 mM, Anaspec, AS-61207) was diluted in full media to a total concentration of 1:100 (100 uM). Cytoplasts were then incubated for 1 to 2 hours, and rinsed 3 times with PBS. Hoechst 33342 (Invitrogen) was added at a 1:5000 dilution in full media for at least 10 minutes. Cells were then washed with PBS and imaged by epifluorescent microscopy.
  • Engineered cytoplasts i.e. loaded with IL-12 mRNA
  • PBS pH 7.2
  • desired concentration e.g., 3M/50 uL
  • Animal weight and tumor dimensions were measured.
  • Engineered cytoplasts were injected into the center of the tumor. Mice were monitored; tumors and body weight were measured every 2-3 days.
  • Engineered cytoplasts i.e. loaded with IL-12 mRNA were resuspended in PBS at desired concentration, e.g., 3M/50 ⁇ L. The maximum recommended injection volume was 100 sL. Injections were performed with a lmL tuberculin syringe and 27 G 1 ⁇ 2′′ or 28 G 1 ⁇ 2′′ needle. Institutional Animal Care and Use Committee (IACUC) protocols were followed for intravenous (IV) injection. Retro-orbital injections were performed under anesthesia with ketamine/xylazine intraperitoneal (IP) injection or isofluorane inhalation. Tail vein injections were performed using a restraint device.
  • IACUC Institutional Animal Care and Use Committee
  • Example 6-3D-Cultured MSC can be Enucleated and 3D-Derived Cytoplasts Show Better Biodistribution In Vivo
  • MSCs were cultured in 3D-hanging drops (3D MSCs) then enucleated to generate 3D cytoplasts.
  • 3D MSCs 3D-hanging drops
  • the 3D culture protocol of MSC by hanging drops is modified from Curr Protoc Stem Cell Biol. 2014 Feb. 6; 28: Unit-2B.6. (Thomas J. BartoshI and Joni H. Ylostalo).
  • Healthy MSCs were harvested from 2D-cultured plates by Trypsin and resuspended in fresh ⁇ -MEM (ThermoFisher 12561056) full medium (16.5% Premium FBS, 1% Antibiotic-Antimycotic, 1% Glutamax, 1% HEPES) at 1.43 million cells/ml.
  • the lid of a 15 cm plate was opened completely and 20 ml PBS was added to the plate.
  • a multichannel pipette was used to make droplets on the lid of the plate at 35 ⁇ l per droplet (approx. 50,000 cells/droplet). About 100-120 droplets were placed on each lid. The lid was closed and the plate was placed back into the incubator.
  • Droplets were cultured for 2 days, then harvested by cell lifter and collected into 15 ml tubes (approx. 300 droplets per tube). The tubes were centrifuged for 5 minutes at 1,200 rpm. The supernatant was removed and the tubes were washed twice with PBS. All P BS was then removed and 7.5 ml of freshly thawed 0.25% Trypsin-EDTA (ThermoFisher 25200114) was added to each tube. The tubes were incubated in a water bath for 4 minutes. The droplets were gently pipetted with 1 ml pipettes with low-retention tips about 10-20 times and incubated in the water bath for another 4 minutes.
  • the droplets were again gently pipetted with 1 ml pipettes with low-retention tips about 10-20 times until most of the droplets were dissociated.
  • 7.5 ml of full serum medium (GlutaMAX Supplement (Gibco 35050061); Fetal Bovine Serum—Premium Select (Atlanta Biologicals S11550); HEPES (1 M) (Gibco 15630080); antibiotic-Antimycotic (100 ⁇ ) (Gibco 15240062)) was added to each tube and the tubes were centrifuged for 10 minutes at 1,200 rpm.
  • the dissociated cells were washed with 10 ml of full serum medium and the cells were resuspended with 5 ml full serum medium.
  • the cells were passed through a 70 ⁇ m cell filter and then the filter was washed with 5 ml full serum medium. The cells were counted and resuspended with pre-treated 12.5% Ficoll at more than 10 M/ml. 30-40M cells were used for each enucleation tube. Subsequently, the protocol for enucleation described above was followed.
  • DiD labeled normal 2D-cultured MSCs (2D MSC), 3D MSCs or 3D cytoplasts were retro-orbitally injected into BalB/C mice respectively. Indicated tissues were harvested 24 hours after injection and DiD labeled cells analyzed by FACS.
  • FIG. 24 shows the successful generation of 3D-derived cytoplasts from 3D-cultured MSCs and also shows the 3D-derived cytoplasts have less lung trapping and better biodistribution to peripheral organs than 2D-cultured cells after injection into the circulation. This is expected to greatly improve their therapeutic ability to locate and deliver cargo to tissues.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • Wood Science & Technology (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Developmental Biology & Embryology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Virology (AREA)
  • Hematology (AREA)
  • Mycology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Oncology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Rheumatology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Dermatology (AREA)
  • Communicable Diseases (AREA)
US16/636,249 2017-08-07 2018-08-07 Platform for generating safe cell therapeutics Pending US20200368285A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US16/636,249 US20200368285A1 (en) 2017-08-07 2018-08-07 Platform for generating safe cell therapeutics
US16/902,420 US10960071B2 (en) 2017-08-07 2020-06-16 Platform for generating safe cell therapeutics

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201762542133P 2017-08-07 2017-08-07
US16/636,249 US20200368285A1 (en) 2017-08-07 2018-08-07 Platform for generating safe cell therapeutics
PCT/US2018/045686 WO2019032628A1 (en) 2017-08-07 2018-08-07 GENERATING PLATFORM FOR SAFE CELLULAR THERAPEUTIC AGENTS

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/045686 A-371-Of-International WO2019032628A1 (en) 2017-08-07 2018-08-07 GENERATING PLATFORM FOR SAFE CELLULAR THERAPEUTIC AGENTS

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US16/902,414 Continuation US10947507B2 (en) 2017-08-07 2020-06-16 Platform for generating safe cell therapeutics
US16/902,420 Continuation US10960071B2 (en) 2017-08-07 2020-06-16 Platform for generating safe cell therapeutics

Publications (1)

Publication Number Publication Date
US20200368285A1 true US20200368285A1 (en) 2020-11-26

Family

ID=65271704

Family Applications (9)

Application Number Title Priority Date Filing Date
US16/636,249 Pending US20200368285A1 (en) 2017-08-07 2018-08-07 Platform for generating safe cell therapeutics
US16/715,859 Active US10927349B2 (en) 2017-08-07 2019-12-16 Platform for generating safe cell therapeutics
US16/902,414 Active US10947507B2 (en) 2017-08-07 2020-06-16 Platform for generating safe cell therapeutics
US17/145,848 Pending US20210189345A1 (en) 2017-08-07 2021-01-11 Platform for generating safe cell therapeutics
US17/159,595 Active US11248213B2 (en) 2017-08-07 2021-01-27 Platform for generating safe cell therapeutics
US17/554,531 Pending US20220177847A1 (en) 2017-08-07 2021-12-17 Platform for generating safe cell therapeutics
US17/665,130 Active US11674121B2 (en) 2017-08-07 2022-02-04 Platform for generating safe cell therapeutics
US17/992,372 Pending US20230159897A1 (en) 2017-08-07 2022-11-22 Platform for generating safe cell therapeutics
US18/066,612 Pending US20230212520A1 (en) 2017-08-07 2022-12-15 Platform for generating safe cell therapeutics

Family Applications After (8)

Application Number Title Priority Date Filing Date
US16/715,859 Active US10927349B2 (en) 2017-08-07 2019-12-16 Platform for generating safe cell therapeutics
US16/902,414 Active US10947507B2 (en) 2017-08-07 2020-06-16 Platform for generating safe cell therapeutics
US17/145,848 Pending US20210189345A1 (en) 2017-08-07 2021-01-11 Platform for generating safe cell therapeutics
US17/159,595 Active US11248213B2 (en) 2017-08-07 2021-01-27 Platform for generating safe cell therapeutics
US17/554,531 Pending US20220177847A1 (en) 2017-08-07 2021-12-17 Platform for generating safe cell therapeutics
US17/665,130 Active US11674121B2 (en) 2017-08-07 2022-02-04 Platform for generating safe cell therapeutics
US17/992,372 Pending US20230159897A1 (en) 2017-08-07 2022-11-22 Platform for generating safe cell therapeutics
US18/066,612 Pending US20230212520A1 (en) 2017-08-07 2022-12-15 Platform for generating safe cell therapeutics

Country Status (9)

Country Link
US (9) US20200368285A1 (ja)
EP (1) EP3664830A4 (ja)
JP (2) JP2020530454A (ja)
KR (2) KR20220124817A (ja)
CN (1) CN111491655A (ja)
AU (1) AU2018316166A1 (ja)
CA (1) CA3072329A1 (ja)
GB (2) GB2578394A (ja)
WO (1) WO2019032628A1 (ja)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2018316166A1 (en) * 2017-08-07 2020-02-06 The Regents Of The University Of California Platform for generating safe cell therapeutics
US10960071B2 (en) * 2017-08-07 2021-03-30 The Regents Of The University Of California Platform for generating safe cell therapeutics
CN112654700B (zh) 2018-07-06 2023-06-16 科罗拉多州立大学董事会法人团体 用于构建和检测生物活性剂的基因编码系统
WO2021158991A1 (en) * 2020-02-07 2021-08-12 The Regents Of The University Of California Methods and use for bioengineering enucleated cells
EP4103722A4 (en) * 2020-02-11 2024-05-22 Cytonus Therapeutics Inc RAPID VACCINE PLATFORM

Family Cites Families (136)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1010543A (en) 1911-05-04 1911-12-05 George A Walter Drinking-fount.
US6538121B1 (en) 1994-11-01 2003-03-25 Human Genome Sciences, Inc. Interleukin-1 β converting enzyme like apoptosis protease-3 and 4
GB9514368D0 (en) 1995-07-13 1995-09-13 Medical Res Council Improvements in or relating to binding assays
US5811407A (en) 1997-02-19 1998-09-22 The University Of North Carolina At Chapel Hill System for the in vivo delivery and expression of heterologous genes in the bone marrow
US6511967B1 (en) 1999-04-23 2003-01-28 The General Hospital Corporation Use of an internalizing transferrin receptor to image transgene expression
US20110171185A1 (en) 1999-06-30 2011-07-14 Klimanskaya Irina V Genetically intact induced pluripotent cells or transdifferentiated cells and methods for the production thereof
US20100158871A1 (en) * 1999-08-30 2010-06-24 David Stephen Terman Sickled erythrocytes with antitumor molecules induce tumoricidal effects
US20040214783A1 (en) 2002-05-08 2004-10-28 Terman David S. Compositions and methods for treatment of neoplastic disease
US8071072B2 (en) 1999-10-08 2011-12-06 Hoffmann-La Roche Inc. Cytotoxicity mediation of cells evidencing surface expression of CD44
DE10003241A1 (de) 2000-01-26 2001-08-02 Werner Lubitz Verschließen von Bakterienghosts
RU2301260C2 (ru) 2000-09-22 2007-06-20 Вирэкссис Корпорейшн Вирусные векторы с зависимой от условий репликацией и их применение
US20020142397A1 (en) * 2000-12-22 2002-10-03 Philippe Collas Methods for altering cell fate
DE10157475A1 (de) 2001-11-23 2003-06-18 Hans-Hermann Gerdes Verfahren und Mittel zur Beeinflussung von interzellulärer Kommunikation und interzellulären Organellentransport
US20030113910A1 (en) * 2001-12-18 2003-06-19 Mike Levanduski Pluripotent stem cells derived without the use of embryos or fetal tissue
AU2003301021C1 (en) 2002-12-16 2010-02-18 Globelmmune, Inc. Yeast-based vaccines as immunotherapy
US20060140963A1 (en) 2003-04-14 2006-06-29 Arius Research, Inc. Cytotoxicity mediation of cells evidencing surface expression of CD59
US20040259249A1 (en) * 2003-05-16 2004-12-23 Nikolai Strelchenko Method of making stem cells from differentiated cells
WO2006055024A2 (en) * 2004-04-05 2006-05-26 Vaxiion Therapeutics, Inc. Minicells as vaccines
CA2601187A1 (en) 2005-03-09 2006-09-14 Board Of Regents, The University Of Texas System Novel htmc promoter and vectors for the tumor-selective and high-efficient expression of cancer therapeutic genes
FR2919804B1 (fr) 2007-08-08 2010-08-27 Erytech Pharma Composition et vaccin therapeutique anti-tumoral
FR2928926B1 (fr) 2008-03-18 2015-08-21 Centre Nat Rech Scient Polynucleotides et polypeptides chimeriques permettant la secretion d'un polypeptide d'interet en association avec des exosomes et leur utilisation pour la production de compositions immunogenes
US20120034156A1 (en) 2010-08-03 2012-02-09 Searete Llc, A Limited Liability Corporation Of The State Of Delaware Artificial cells
EP3320915A1 (en) * 2009-04-17 2018-05-16 Globeimmune, Inc. Combination immunotherapy compositions against cancer and methods
GB201020738D0 (en) 2010-12-07 2011-01-19 Affitech Res As Antibodies
FI20115914L (fi) 2011-09-16 2013-03-17 Oncos Therapeutics Ltd Muunnettu onkolyyttinen virus
US20150104428A1 (en) 2012-03-30 2015-04-16 University Of Southern California Compositions and Treatment Methods for Mesenchymal Stem Cell-Induced Immunoregulation
CN104685055A (zh) 2012-05-09 2015-06-03 格兰达利斯有限公司 对单核苷酸KRAS突变特异的双功能短发夹RNA(bi-shRNA)
JP6542197B2 (ja) 2013-04-12 2019-07-10 エヴォックス・セラピューティクス・リミテッド 治療的送達小胞
EP3546485A1 (en) * 2013-05-10 2019-10-02 Whitehead Institute for Biomedical Research In vitro production of red blood cells with sortaggable proteins
WO2014197599A1 (en) 2013-06-04 2014-12-11 The Johns Hopkins University Peg-prom mediated surface expression of avidin/streptavidin
US10077301B2 (en) 2013-06-27 2018-09-18 Monash University IL-21 binding proteins
US20160199413A1 (en) 2013-08-01 2016-07-14 Isletone Ab Mscs in the treatment of inflammatory pulmonary diseases
US20160272707A1 (en) 2013-09-11 2016-09-22 Compugen Ltd. Vstm5 antibodies, and uses thereof for treatment of cancer, infectious diseases and immune related diseases
EP3071515A2 (en) 2013-11-18 2016-09-28 Rubius Therapeutics, Inc. Synthetic membrane-receiver complexes
DK3071697T3 (da) 2013-11-22 2020-01-27 Dnatrix Inc Adenovirus der udtrykker immuncelle-stimulatorisk(e) receptor agonist(er)
WO2015128492A1 (en) 2014-02-28 2015-09-03 Maghazachi Azzam A Monomethyl- and dimethylfumarate for nk cell activation
HUE054471T2 (hu) * 2014-04-01 2021-09-28 Rubius Therapeutics Inc Immunmodulációs módszerek és készítmények
CN106459997A (zh) 2014-05-16 2017-02-22 耶鲁大学 用于疫苗应用的高效价病毒样囊泡的进化
JP6925264B2 (ja) 2014-08-27 2021-08-25 メモリアル スローン ケタリング キャンサー センター 抗体、組成物および使用
AU2015318233B2 (en) 2014-09-15 2020-03-12 Plexxikon Inc. Heterocyclic compounds and uses thereof
US11285194B2 (en) 2014-10-24 2022-03-29 Calidi Biotherapeutics, Inc. Combination immunotherapy approach for treatment of cancer
CA2966234A1 (en) 2014-12-15 2016-06-23 Bellicum Pharmaceuticals, Inc. Methods for controlled elimination of therapeutic cells
JP6917896B2 (ja) 2015-03-05 2021-08-18 フレッド ハッチンソン キャンサー リサーチ センター 免疫調節性融合タンパク質およびその使用
EP3064507A1 (en) 2015-03-06 2016-09-07 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Fusion proteins comprising a binding protein and an interleukin-15 polypeptide having a reduced affinity for IL15ra and therapeutic uses thereof
JP2018510158A (ja) 2015-03-18 2018-04-12 ステムイミューン,インコーポレイテッド 抗体組み合わせを用いるウイルス療法
WO2016183482A1 (en) * 2015-05-13 2016-11-17 Rubius Therapeutics, Inc. Membrane-receiver complex therapeutics
KR101714695B1 (ko) 2015-05-18 2017-03-09 한국과학기술연구원 가교화된 pva-ecm 복합체를 제조하는 방법 및 그에 의하여 제조된 pva-ecm 복합체
CN104877965B (zh) * 2015-05-27 2018-04-20 上海厚东生物科技有限公司 一种制备成熟红细胞的方法
EA201800148A1 (ru) 2015-08-11 2019-01-31 Калиди Биотерапьютикс, Инк. Оспенная вакцина для лечения рака
CA3129180A1 (en) 2015-09-21 2017-03-30 Plexxikon Inc. Heterocyclic compounds and uses thereof
JP2018529340A (ja) 2015-10-01 2018-10-11 ユニバーシティ・オブ・オタワ 核酸のエキソソームパッケージング
PE20181515A1 (es) 2015-12-04 2018-09-21 Boehringer Ingelheim Int Polipeptidos biparatopicos que antagonizan la senalizacion wnt en celulas tumorales
EP3390434A2 (en) 2015-12-14 2018-10-24 Bellicum Pharmaceuticals, Inc. Dual controls for therapeutic cell activation or elimination
KR20180095098A (ko) 2016-01-11 2018-08-24 루비우스 테라퓨틱스, 아이엔씨. 암 적응증에 대한 다중양식 치료 세포 시스템과 관련된 조성물 및 방법
US10940199B2 (en) 2016-03-09 2021-03-09 Wisconsin Alumni Research Foundation Versikine for inducing and potentiating an immune response
CN108884460B (zh) 2016-03-19 2023-04-28 埃克苏马生物技术公司 淋巴细胞转导及其扩增调节的方法与组合物
EP3432888B1 (en) 2016-03-24 2022-10-05 Cothera Bioscience, Inc. Treatment of cancer with tg02
US11458097B2 (en) 2016-03-30 2022-10-04 The University Of North Carolina At Chapel Hill Biological agent-exosome compositions and uses thereof
EP3246410A1 (en) 2016-05-19 2017-11-22 Klinikum rechts der Isar der Technischen Universität München Vsv/ndv hybrid viruses for oncolytic therapy of cancer
GB201609216D0 (en) 2016-05-25 2016-07-06 Evox Therapeutics And Isis Innovation Ltd Exosomes comprising therapeutic polypeptides
WO2017205875A1 (en) 2016-05-27 2017-11-30 Dnatrix, Inc. Adenovirus and immunomodulator combination therapy
US11427625B2 (en) 2016-06-14 2022-08-30 University of Pittsburgh—of the Commonwealth System of Higher Education Expression of NKG2D activating ligand proteins for sensitizing cancer cells to attack by cytotoxic immune cells
CN107586341A (zh) 2016-07-08 2018-01-16 生命序有限公司 重组免疫检查点受体及免疫检查点抑制分子的共表达及应用
GB2552473A (en) 2016-07-21 2018-01-31 Evox Therapeutics Ltd Surface decoration of extracellular vesicles
CN116655790A (zh) 2016-08-26 2023-08-29 百济神州有限公司 抗tim-3抗体及其用途
JP7096598B2 (ja) 2016-09-07 2022-07-06 トラスティーズ オブ タフツ カレッジ イムノdash阻害剤及びpge2アンタゴニストを用いた併用療法
WO2018064513A1 (en) 2016-09-30 2018-04-05 The Brigham And Women's Hospital, Inc. Natural killer cell-evasive oncolytic viruses
US11084859B2 (en) 2016-10-24 2021-08-10 Orionis Biosciences BV Targeted mutant interferon-gamma and uses thereof
EP3532164A4 (en) 2016-10-28 2020-10-07 Children's Hospital Medical Center TREATMENT OF DISEASES RELATED TO ACTIVATED IRAQ
RU2019120400A (ru) 2016-12-02 2021-01-11 Рубиус Терапьютикс, Инк. Композиции и способы, связанные с клеточными системами для проникновения в солидные опухоли
WO2018112032A1 (en) 2016-12-13 2018-06-21 President And Fellows Of Harvard College Methods and compositions for targeting tumor-infiltrating tregs using inhibitors of ccr8 and tnfrsf8
US11617771B2 (en) 2016-12-15 2023-04-04 The Regents Of The University Of California Oral composition comprising lactic acid bacteria for regulating immune responses and methods related thereto
EP3573640A4 (en) 2017-01-30 2021-04-28 The Ohio State Innovation Foundation PASSIVE ANTIBODY DEPENDENT CELLULAR MEDIATION ACTIVATION
WO2018148378A1 (en) 2017-02-08 2018-08-16 Dana-Farber Cancer Institute, Inc. Modulating biomarkers to increase tumor immunity and improve the efficiacy of cancer immunotherapy
WO2018165194A1 (en) 2017-03-06 2018-09-13 University Of Washington Engineered cells and agent compositions for therapeutic agent delivery and treatments using same
CN108623686A (zh) 2017-03-25 2018-10-09 信达生物制药(苏州)有限公司 抗ox40抗体及其用途
CN111148518A (zh) 2017-03-30 2020-05-12 丹娜法伯癌症研究院 使用cdk4/6抑制剂调控调节性t细胞和免疫应答的方法
CA3047857C (en) 2017-04-12 2021-06-22 Shenzhen International Institute For Biomedical Research Pum 1 protein as a target for virus inhibition
JP7329839B2 (ja) 2017-04-21 2023-08-21 コアスター セラピューティクス インク. 膜脂質被覆されたナノ粒子および使用方法
KR20200034668A (ko) * 2017-05-08 2020-03-31 플래그쉽 파이어니어링 이노베이션스 브이, 인크. 막 융합을 촉진시키기 위한 조성물 및 그의 용도
HUE063120T2 (hu) 2017-05-09 2023-12-28 Scholar Rock Inc LRRC33-inhibitorok és alkalmazásuk
US11433143B2 (en) 2017-05-18 2022-09-06 The Regents Of The University Of California Nano-enabled immunotherapy in cancer
CA3064897A1 (en) 2017-05-25 2018-11-29 University Of Central Florida Research Foundation, Inc. Novel oncolytic viruses for sensitizing tumor cells to killing by natural killer cells
MY200744A (en) 2017-05-31 2024-01-13 Boehringer Ingelheim Int Polypeptides antagonizing wnt signaling in tumor cells
US20210139846A1 (en) 2017-06-02 2021-05-13 Board Of Supervisors Of Louisiana State University And Agricultural And Mechanical College Engineered cells, and methods of using the same
EP3634496A4 (en) 2017-06-06 2021-09-08 Dana-Farber Cancer Institute, Inc. METHOD FOR RISING AWARENESS IN CANCER CELLS AGAINST T-CELL-MEDIATED KILLING BY MODULATING MOLECULAR SIGNAL PATHS
TWI704923B (zh) 2017-06-26 2020-09-21 台灣基督長老教會馬偕醫療財團法人馬偕紀念醫院 包含胞外囊泡之製劑、用以製備該製劑之方法及其用途
WO2019014665A1 (en) 2017-07-14 2019-01-17 Dana-Farber Cancer Institute, Inc. MODULATION OF BIOMARKERS TO INCREASE ANTITUMOR IMMUNITY AND IMPROVE THE EFFECTIVENESS OF ANTICANCER IMMUNOTHERAPY
US20200172868A1 (en) * 2017-07-19 2020-06-04 Rubius Therapeutics, Inc. Compositions and methods related to multimodal therapeutic cell systems for infectious disease
AU2018316166A1 (en) * 2017-08-07 2020-02-06 The Regents Of The University Of California Platform for generating safe cell therapeutics
US10960071B2 (en) 2017-08-07 2021-03-30 The Regents Of The University Of California Platform for generating safe cell therapeutics
CN111511762A (zh) 2017-08-21 2020-08-07 天演药业公司 抗cd137分子及其用途
AU2018328223A1 (en) 2017-09-05 2020-03-12 GLAdiator Biosciences, Inc. Delivery of payloads to stem cells
GB201714430D0 (en) 2017-09-07 2017-10-25 Micol Romain Compositions and processes for targeted delivery and expression and modulation of therapeutic components in tissue
US20230160881A1 (en) 2017-09-15 2023-05-25 Nantomics, Llc HMGB1 RNA And Methods Therefor
CA3077107A1 (en) 2017-09-27 2019-04-04 University Of Ottawa Fluorescent enveloped viral particles as standards for nanosale flow cytometry
US20210106632A1 (en) 2017-09-28 2021-04-15 Tandem Co., Ltd. Novel recombinant plasma membrane-based vesicle, for treating cancer
EP3700933A1 (en) 2017-10-25 2020-09-02 Novartis AG Antibodies targeting cd32b and methods of use thereof
WO2019090148A2 (en) 2017-11-03 2019-05-09 Rubius Therapeutics, Inc. Compositions and methods related to therapeutic cell systems for tumor growth inhibition
WO2019090355A1 (en) 2017-11-06 2019-05-09 Children's National Medical Center Cells expressing antibodies and methods of treatment using the same
WO2019099943A1 (en) 2017-11-16 2019-05-23 Astrazeneca Ab Compositions and methods for improving the efficacy of cas9-based knock-in strategies
US20190153409A1 (en) 2017-11-17 2019-05-23 Aviv MedTech Ltd. Compositions comprising particles and methods for treating cancer
WO2019113512A1 (en) 2017-12-07 2019-06-13 Flagship Pioneering Innovations V, Inc. Cytobiologics and therapeutic uses thereof
EP3724232A4 (en) 2017-12-14 2021-12-15 The University of Ottawa EXOSOME PACKAGING AND TARGETED AUTOPHAGY
WO2019126799A1 (en) 2017-12-22 2019-06-27 Distributed Bio, Inc. Major histocompatibility complex (mhc) compositions and methods of use thereof
CN109970857B (zh) 2017-12-27 2022-09-30 信达生物制药(苏州)有限公司 抗pd-l1抗体及其用途
MX2020006672A (es) 2017-12-28 2020-08-31 Codiak Biosciences Inc Exosomas para inmunooncologia y terapia antiinflamatoria.
JP2021513853A (ja) 2018-02-15 2021-06-03 ノース カロライナ ステート ユニバーシティNorth Carolina State University 癌免疫療法のためのチェックポイント遮断としての操作されたナノベシクル
WO2019169141A1 (en) 2018-02-28 2019-09-06 Lester Smith Medical Research Institute Production and use of extracellular vesicles
SG11202008261WA (en) 2018-03-08 2020-09-29 Rubius Therapeutics Inc Therapeutic cell systems and methods for treating cancer and infectious diseases
JP2021515797A (ja) 2018-03-15 2021-06-24 アトッサ・セラピューティクス・インコーポレイテッド 免疫反応誘導のインサイツ方法
US20200362052A1 (en) 2018-04-02 2020-11-19 Souvie Biodelivery, LLC Compositions and methods for treating toll-like receptor-driven inflammatory diseases
CN112135639B (zh) 2018-04-06 2023-10-20 北卡罗莱纳州立大学 细胞组装介导的癌症免疫治疗检查点抑制剂的递送
US11739350B2 (en) 2018-04-10 2023-08-29 Ottawa Hospital Research Institute MicroRNA-based compositions and methods used in disease treatment
US11413315B2 (en) 2018-04-29 2022-08-16 City Of Hope Neural stem cell-mediated cancer treatment
WO2019213706A1 (en) 2018-05-08 2019-11-14 Deakin University Extracellular vesicle-based drug-delivery
CN112469423A (zh) 2018-05-09 2021-03-09 儿童医学中心公司 经间充质基质细胞外排体处理的单核细胞及其用途
US20200054675A1 (en) 2018-06-01 2020-02-20 Washington University Suppression of cytokine release syndrome in chimeric antigen receptor cell therapy
US11505782B2 (en) 2018-06-04 2022-11-22 Calidi Biotherapeutics, Inc. Cell-based vehicles for potentiation of viral therapy
ES2966045T3 (es) 2018-06-04 2024-04-18 Calidi Biotherapeutics Inc Vehículos basados en células para la potenciación de la terapia viral
AU2019288277A1 (en) 2018-06-19 2021-01-28 H. Lee Moffitt Cancer Center And Research Institute, Inc. Oncolytic virus or antigen presenting cell mediated cancer therapy using type I interferon and CD40-ligand
AU2019290212A1 (en) 2018-06-22 2021-02-04 Mayo Foundation For Medical Education And Research Methods of treating cancer using combination therapy
WO2020016900A1 (en) 2018-07-18 2020-01-23 Exostem Biotec Ltd. Msc- and exosome-based immunotherapy
EP3824097A4 (en) 2018-07-22 2022-03-30 Health Research, Inc. CANCER CELL VACCINE EXPRESSING MAJOR HISTOCOMPATIBILITY COMPLEX CLASS II AND METHODS OF USE TO PRODUCE INTEGRATED IMMUNE RESPONSES
US20220340898A1 (en) 2018-08-01 2022-10-27 Yale University Compositions and Methods for Identification of Membrane Targets for Enhancement of T cell Activity Against Cancer
WO2020072126A2 (en) 2018-08-07 2020-04-09 Dana-Farber Cancer Institute, Inc. Modulating ptpn2 to increase immune responses and perturbing gene expression in hematopoietic stem cell lineages
JP2021535209A (ja) 2018-08-24 2021-12-16 トランスフェール プリュ エス.ウ.セ. ソルチリン受容体を標的化し血管擬態を阻害するための方法及び化合物
US20210198642A1 (en) 2018-09-07 2021-07-01 Astrazeneca Ab Compositions and methods for improved nucleases
AU2019351273A1 (en) 2018-09-27 2021-05-20 Genocea Biosciences, Inc. Treatment methods
KR20210090618A (ko) 2018-09-27 2021-07-20 제노세아 바이오사이언스, 인코퍼레이티드 치료 방법
US20200215114A1 (en) 2018-11-19 2020-07-09 Exosome Therapeutics, Inc. Compositions and methods for producing exosome loaded therapeutics for the treatment of multiple oncological disorders
CA3116192A1 (en) 2018-11-06 2020-05-14 Calidi Biotherapeutics, Inc. Enhanced systems for cell-mediated oncolytic viral therapy
WO2020205579A1 (en) 2019-03-29 2020-10-08 University Of Southern California Genetically modified exosomes for immune modulation
AU2020264083A1 (en) 2019-04-25 2021-12-02 Mendus B.V. Methods of tumor vaccination
WO2021041473A1 (en) 2019-08-27 2021-03-04 The Trustees Of Columbia University In The City Of New York Engineered exosomes for targeted delivery
EP4103722A4 (en) * 2020-02-11 2024-05-22 Cytonus Therapeutics Inc RAPID VACCINE PLATFORM

Also Published As

Publication number Publication date
EP3664830A1 (en) 2020-06-17
US11674121B2 (en) 2023-06-13
WO2019032628A1 (en) 2019-02-14
US20230159897A1 (en) 2023-05-25
AU2018316166A1 (en) 2020-02-06
KR20220124817A (ko) 2022-09-14
JP2022184879A (ja) 2022-12-13
JP2020530454A (ja) 2020-10-22
KR20200037353A (ko) 2020-04-08
US20200216814A1 (en) 2020-07-09
US10927349B2 (en) 2021-02-23
US20210162038A1 (en) 2021-06-03
US20230212520A1 (en) 2023-07-06
US20200368287A1 (en) 2020-11-26
GB202115748D0 (en) 2021-12-15
US10947507B2 (en) 2021-03-16
EP3664830A4 (en) 2020-07-01
US20220177847A1 (en) 2022-06-09
GB202000541D0 (en) 2020-02-26
US11248213B2 (en) 2022-02-15
CN111491655A (zh) 2020-08-04
US20210189345A1 (en) 2021-06-24
CA3072329A1 (en) 2019-02-14
US20220204938A1 (en) 2022-06-30
GB2597623A (en) 2022-02-02
GB2578394A (en) 2020-05-06

Similar Documents

Publication Publication Date Title
US11248213B2 (en) Platform for generating safe cell therapeutics
Riazifar et al. Stem cell extracellular vesicles: extended messages of regeneration
Wu et al. Extracellular vesicles from human embryonic stem cell-derived cardiovascular progenitor cells promote cardiac infarct healing through reducing cardiomyocyte death and promoting angiogenesis
US10960071B2 (en) Platform for generating safe cell therapeutics
Ikemoto et al. Autologous transplantation of SM/C-2.6+ satellite cells transduced with micro-dystrophin CS1 cDNA by lentiviral vector into mdx mice
Chang et al. CAR-neutrophil mediated delivery of tumor-microenvironment responsive nanodrugs for glioblastoma chemo-immunotherapy
Wagner et al. Fate of intravenously injected mesenchymal stem cells and significance for clinical application
Cuchiara et al. Bioactive poly (ethylene glycol) hydrogels to recapitulate the HSC niche and facilitate HSC expansion in culture
US20220025403A1 (en) Reduced and minimal manipulation manufacturing of genetically-modified cells
Budgude et al. Mesenchymal stromal cell‐derived extracellular vesicles as cell‐free biologics for the ex vivo expansion of hematopoietic stem cells
Lepperhof et al. Bioluminescent imaging of genetically selected induced pluripotent stem cell-derived cardiomyocytes after transplantation into infarcted heart of syngeneic recipients
US20210363260A1 (en) Methods and compositions for treating cancer by targeting the clec2d-klrb1 pathway
AU2018290250A1 (en) Immunoprivileged bioactive renal cells for the treatment of kidney disease
Kopan et al. Approaches in immunotherapy, regenerative medicine, and bioengineering for type 1 diabetes
US20230015661A1 (en) Methods and use for bioengineering enucleated cells
Schaaf et al. Ex-vivo expansion of muscle-regenerative cells for the treatment of muscle disorders
JP7385230B2 (ja) 移植用培養細胞または培養組織の調製方法
WO2024107723A2 (en) Hypoimmunogenic biomimetic nanovesicle delivery system for hemophagocytic lymphohistiocytosis (hlh) associated diseases
Lee Engineering Macrophages for Cell-Based Therapies

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE REGENTS OF THE UNIVERSITY OF CALIFORNIA, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KLEMKE, RICHARD;WANG, HUAWEI;REEL/FRAME:052574/0061

Effective date: 20190327

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

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

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

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

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

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

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

Free format text: NON FINAL ACTION MAILED

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

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

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

Free format text: FINAL REJECTION MAILED