WO2022150569A1 - Méthodes d'augmentation de lymphocytes nkp30-positifs chez un sujet et leurs utilisations - Google Patents

Méthodes d'augmentation de lymphocytes nkp30-positifs chez un sujet et leurs utilisations Download PDF

Info

Publication number
WO2022150569A1
WO2022150569A1 PCT/US2022/011582 US2022011582W WO2022150569A1 WO 2022150569 A1 WO2022150569 A1 WO 2022150569A1 US 2022011582 W US2022011582 W US 2022011582W WO 2022150569 A1 WO2022150569 A1 WO 2022150569A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
positive
increase
fold
reduction
Prior art date
Application number
PCT/US2022/011582
Other languages
English (en)
Inventor
Christina Marie Coughlin
Laurence Allan TURKA
Anna Carol SALZBERG WALDNER
Original Assignee
Rubius Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Rubius Therapeutics, Inc. filed Critical Rubius Therapeutics, Inc.
Publication of WO2022150569A1 publication Critical patent/WO2022150569A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/18Erythrocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/179Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1793Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2086IL-13 to IL-16
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Definitions

  • the present invention relates generally to methods of administering enucleated erythroid cells to a subject, methods of increasing NKp30-positive lymphocytes in a subject, and methods of treating a B7-H6-positive cancer in a subject.
  • Engineered enucleated erythroid cells are in development as therapeutic agents which carry or present exogenous protein(s) for patients in need thereof.
  • B7 Homolog 6 B7-H6
  • B7-H6 B7 Homolog 6
  • the disclosure is based, at least in part, on the discovery that administering enucleated erythroid cells comprising a first exogenous polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface, and a second exogenous polypeptide comprising 4-1BBL or a functional fragment thereof, on its extracellular surface, to a subject results in an increase in the number of NKp30-positive lymphocytes in the subject.
  • determination of B7-H6 positivity of a cancer in a subject is particularly useful to identify patients likely to respond to therapy with these cells.
  • NKp30-positive lymphocytes in a subject previously identified or diagnosed as having a B7-H6-positive cancer methods of treating a subject previously identified or diagnosed as having a B7-H6-positive cancer, methods of decreasing the number and/or proliferation of B7-H6-positive cancer cells in a subject previously identified or diagnosed as having a B7-H6- positive cancer, methods of killing a B7-H6-positive cancer cell in a subject previously identified or diagnosed as having a B7-H6-positive cancer, and methods of reducing the volume of a solid tumor in a subject previously identified or diagnosed as having a B7-H6-positive cancer.
  • NKp30-positive lymphocytes in a subject previously identified or diagnosed as having a B7-H6- positive cancer in need thereof that include administering to the subject a therapeutically effective amount of a pharmaceutical composition comprising a population of enucleated erythroid cells comprising a first exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface.
  • the administering results in at least a 5% increase in the number of NKp30-positive lymphocytes in the subject as compared to the number of NKp30-positive lymphocytes in the subject prior to the administering. In some embodiments of any of the methods described herein, the administering results in at least a 10% increase in the number of NKp30-positive lymphocytes in the subject as compared to the number of NKp30-positive lymphocytes in the subject prior to the administering. In some embodiments of any of the methods described herein, the NKp30-positive lymphocytes are NKp30-positive NK cells.
  • the method further results in an increase in the number of NKp30-positive/NKG2A-negative lymphocytes in the subject.
  • the administering step results in at least a 5% increase in the number of NKp30-positive/NKG2A-negative lymphocytes in the subject.
  • the administering step results in at least a 10% increase in the number of NKp30-positive/NKG2A-negative lymphocytes in the subject.
  • the NKp30- positive/NKG2A-negative lymphocytes are NKp30-positive/NKG2A-negative NK cells.
  • the administering step results in an increase in the number of NKp30-positive, CD45- positive, CD56-positive lymphocytes in the subject. In some embodiments of any of the methods described herein, the administering step results in at least a 1.2-fold increase in the percentage of CD45-positive, CD56-positive lymphocytes that are NKp30-positive in the subject. In some embodiments of any of the methods described herein, the administering step results in at least a 1.5-fold increase in the percentage of CD45-positive, CD56-positive lymphocytes that are NKp30-positive in the subject.
  • the administering step results in at least a 2.0-fold increase in the percentage of CD45- positive, CD56-positive lymphocytes that are NKp30-positive in the subject.
  • the NKp30-positive, CD45- positive, CD56-positive lymphocytes are NKp30-positive, CD45 -positive, CD56- positive NK cells.
  • the administering step results in an increase in the number of NKp30-positive, CD45- positive, CD 16-positive, CD56-dim lymphocytes in the subject. In some embodiments of any of the methods described herein, the administering step results in at least a 1.2-fold increase in the percentage of CD56-dim, CD 16-positive, CD45- positive lymphocytes that are NKp30-positive in the subject. In some embodiments of any of the methods described herein, the administering step results in at least a 1.5- fold increase in the percentage of CD56-dim, CD 16-positive, CD45-positive lymphocytes that are NKp30-positive in the subject.
  • the administering step results in at least a 1.7-fold increase in the percentage of CD56-dim, CD 16-positive, CD45-positive lymphocytes that are NKp30-positive in the subject.
  • the NKp30-positive, CD45 -positive, CD 16-positive, CD56-dim lymphocytes are NKp30-positive, CD45-positive, CD 16-positive, CD56-dimNK cells.
  • the administering step results in an increase in the number of NKp30-positive, CD45- positive, CD 16-positive, and CD56-positive lymphocytes in the subject. In some embodiments of any of the methods described herein, the administering step results in at least a 1.5 -fold increase in the percentage of CD56-positive, CD 16-positive, CD45- positive lymphocytes that are NKp30-positive in the subject. In some embodiments of any of the methods described herein, the administering step results in at least a 2.0- fold increase in the percentage of CD56-positive, CD 16-positive, CD45-positive lymphocytes that are NKp30-positive in the subject.
  • the administering step results in at least a 2.5-fold increase in the percentage of CD56-positive, CD 16-positive, CD45-positive lymphocytes that are NKp30-positive in the subject. In some embodiments of any of the methods described herein, the administering step results in at least a 3.0-fold increase in the percentage of CD56-positive, CD 16-positive, CD45-positive lymphocytes that are NKp30-positive in the subject. In some embodiments of any of the methods described herein, the administering step results in at least a 3.5-fold increase in the percentage of CD56-positive, CD 16-positive, CD45-positive lymphocytes that are NKp30-positive in the subject.
  • the NKp30-positive, CD56-positive, CD 16-positive, CD45-positive lymphocytes are NKp30-positive, CD56-positive, CD 16-positive, CD45-positive NK cells.
  • the administering step does not result in a significant level of myeloid cell toxicity in the subject.
  • the method further comprises identifying or diagnosing a subject as having a B7-H6-positive cancer.
  • the B7-H6- positive cancer is selected from the group consisting of: adrenocortical carcinoma, bile duct cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, esophageal cancer, eye cancer, head and neck cancer, acute myeloid leukemia, lymphoid neoplasm diffuse large b-cell lymphoma, kidney cancer, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, kidney chromophobe, liver cancer, lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma, ovarian cancer, pancreatic cancer, rectal cancer, skin cancer, stomach cancer, testicular cancer, thymoma, thyroid cancer, uterine cancer, uterine corpus endometrial carcinoma, uterine carcinosarcoma, and uveal melanoma.
  • adrenocortical carcinoma bile duct cancer, bladder cancer, breast cancer, cervical
  • the method further comprises administering to the subject an NKG2A inhibitor.
  • the NKG2A inhibitor is an antagonistic antibody that binds specifically to NKG2A.
  • Also provided herein are methods of treating a subject previously identified or diagnosed as having a B7-H6-positive cancer that include administering to a subject previously identified or diagnosed as having a B7-H6-positive cancer a therapeutically effective amount of a pharmaceutical composition comprising a population of enucleated erythroid cells comprising a first exogenous polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface.
  • the B7-H6-positive cancer is selected from the group consisting of: adrenocortical carcinoma, bile duct cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, esophageal cancer, eye cancer, head and neck cancer, acute myeloid leukemia, lymphoid neoplasm diffuse large b-cell lymphoma, kidney cancer, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, kidney chromophobe, liver cancer, lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma, ovarian cancer, pancreatic cancer, rectal cancer, skin cancer, stomach cancer, testicular cancer, thymoma, thyroid cancer, uterine cancer, uterine corpus endometrial carcinoma, uterine carcinosarcoma, and uveal melanoma.
  • adrenocortical carcinoma bile duct cancer, bladder cancer, breast cancer, cervical
  • the administering step does not result in a significant level of myeloid cell toxicity in the subject.
  • the method further comprises diagnosing or identifying the subject as having the B7-H6-positive cancer.
  • the method further comprises administering to the subject an NKG2A inhibitor.
  • the NKG2A inhibitor is an antagonistic antibody that binds specifically to NKG2A.
  • this disclosure features methods of decreasing the number and/or proliferation of B7-H6-positive cancer cells in a subject previously identified or diagnosed as having a B7-H6- positive cancer, the method comprising administering a therapeutically effective amount of a pharmaceutical composition comprising a population of enucleated erythroid cells comprising a first exogenous polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface.
  • the administering results in a decrease in the number of B7-H6-positive cancer cells in the subject. In some embodiments of any of the methods described herein, the administering results in at least a 5% decrease in the number of B7-H6-positive cancer cells in the subject as compared to the number of B7-H6-positive cancer cells in the subject prior to the administering. In some embodiments of any of the methods described herein, the administering results in at least a 10% decrease in the number of B7-H6-positive cancer cells in the subject as compared to the number of B7-H6- positive cancer cells in the subject prior to the administering.
  • the administering results in a decrease in the number of B7-H6-positive/HLA-E-negative cancer cells in the subject. In some embodiments of any of the methods described herein, the administering results in at least a 5% decrease in the number of B7-H6- positive/HLA-E-negative cancer cells in the subject as compared to the number of B7- H6-positive/HLA-E-negative cancer cells in the subject prior to the administering.
  • the administering results in at least a 10% decrease in the number of B7-H6-positive/HLA-E-negative cancer cells in the subject as compared to the number of B7-H6-positive/HLA-E-negative cancer cells in the subject prior to the administering.
  • the administering results in a decrease in the proliferation of B7-H6-positive cancer cells in the subject. In some embodiments of any of the methods described herein, the administering results in at least a 5% decrease in the proliferation of B7-H6-positive cancer cells in the subject as compared to the proliferation of B7-H6-positive cancer cells in the subject prior to the administering. In some embodiments of any of the methods described herein, the administering results in at least a 10% decrease in the proliferation of B7-H6-positive cancer cells in the subject as compared to the proliferation of B7-H6-positive cancer cells in the subject prior to the administering.
  • the administering results in a decrease in the proliferation of B7-H6-positive/HLA-E- negative cancer cells in the subject. In some embodiments of any of the methods described herein, the administering results in at least a 5% decrease in the proliferation of B7-H6-positive/HLA-E-negative cancer cells in the subject as compared to the proliferation of B7-H6-positive/HLA-E-negative cancer cells in the subject prior to the administering.
  • the administering results in at least a 10% decrease in the proliferation of B7-H6-positive/HLA-E-negative cancer cells in the subject as compared to the proliferation of B7-H6-positive/HLA-E-negative cancer cells in the subject prior to the administering.
  • the administering step does not result in a significant level of myeloid cell toxicity in the subject.
  • the method further comprises identifying or diagnosing a subject as having a B7-H6-positive cancer.
  • the B7-H6- positive cancer is selected from the group consisting of: adrenocortical carcinoma, bile duct cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, esophageal cancer, eye cancer, head and neck cancer, acute myeloid leukemia, lymphoid neoplasm diffuse large b-cell lymphoma, kidney cancer, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, kidney chromophobe, liver cancer, lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma, ovarian cancer, pancreatic cancer, rectal cancer, skin cancer, stomach cancer, testicular cancer, thymoma, thyroid cancer, uterine cancer, uterine corpus endo
  • the method further comprises administering to the subject an NKG2A inhibitor.
  • the NKG2A inhibitor is an antagonistic antibody that binds specifically to NKG2A.
  • Also provided herein are methods of inducing killing a B7-H6-positive cancer cell in a subject previously identified or diagnosed as having a B7-H6-positive cancer that include administering a therapeutically effective amount of a pharmaceutical composition comprising a population of enucleated erythroid cells comprising an exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface.
  • the killing comprises necrosis. In some embodiments, the killing comprises apoptosis. In some embodiments of any of the methods described herein, the killing is mediated via NK- cell mediated cytolysis.
  • the B7-H6- positive cancer cell is a cancer cell selected from the group consisting of: adrenocortical carcinoma, bile duct cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, esophageal cancer, eye cancer, head and neck cancer, acute myeloid leukemia, lymphoid neoplasm diffuse large b-cell lymphoma, kidney cancer, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, kidney chromophobe, liver cancer, lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma, ovarian cancer, pancreatic cancer, rectal cancer, skin cancer, stomach cancer, testicular cancer, thymoma, thyroid cancer, uterine cancer, uterine corpus endometrial carcinoma, uterine carcinosarcoma, and uveal melanoma.
  • adrenocortical carcinoma bile duct cancer, bladder cancer
  • the administering step does not result in a significant level of myeloid cell toxicity in the subject.
  • the subject has been previously identified or diagnosed as having a B7-H6-positive cancer.
  • the B7-H6-positive cancer is selected from the group consisting of: adrenocortical carcinoma, bile duct cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, esophageal cancer, eye cancer, head and neck cancer, acute myeloid leukemia, lymphoid neoplasm diffuse large b-cell lymphoma, kidney cancer, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, kidney chromophobe, liver cancer, lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma, ovarian cancer, pancreatic cancer, rectal cancer, skin cancer, stomach cancer, testicular cancer, thymoma, thyroid cancer, uterine cancer, uterine corpus endometrial carcinoma, uterine carcinosarcoma, and uveal melanoma.
  • adrenocortical carcinoma bile duct cancer, bladder cancer, breast cancer, cervical
  • the method further comprises administering to the subject an NKG2A inhibitor.
  • the NKG2A inhibitor is an antagonistic antibody that binds specifically to NKG2A.
  • Also provided herein are methods of reducing the volume of a solid tumor in a subject previously identified or diagnosed as having a B7-H6-positive cancer that include administering a therapeutically effective amount of a population of enucleated erythroid cells comprising a first exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface.
  • the administering results in at least a 5% decrease in the volume of the solid tumor in the subject as compared to the volume of the solid tumor prior to the administering. In some embodiments of any of the methods described herein, the administering results in at least a 10% decrease in the volume of the solid tumor in the subject as compared to the volume of the solid tumor prior to the administering. In some embodiments of any of the methods described herein, the administering step does not result in a significant level of myeloid cell toxicity in the subject.
  • the B7-H6- positive cancer is selected from the group consisting of: adrenocortical carcinoma, bile duct cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, esophageal cancer, eye cancer, head and neck cancer, acute myeloid leukemia, lymphoid neoplasm diffuse large b-cell lymphoma, kidney cancer, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, kidney chromophobe, liver cancer, lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma, ovarian cancer, pancreatic cancer, rectal cancer, skin cancer, stomach cancer, testicular cancer, thymoma, thyroid cancer, uterine cancer, uterine corpus endometrial carcinoma, uterine carcinosarcoma, and uveal melanoma.
  • adrenocortical carcinoma bile duct cancer, bladder cancer, breast cancer, cervical
  • the administering step does not result in a significant level of myeloid cell toxicity in the subject.
  • the method further comprises diagnosing or identifying the subject as having the B7-H6-positive cancer.
  • the method further comprises administering to the subject an NKG2A inhibitor.
  • the NKG2A inhibitor is an antagonistic antibody that binds specifically to NKG2A.
  • the enucleated erythroid cell comprises at least 1,000 copies of the exogenous fusion polypeptide. In some embodiments of any of the methods described herein, the enucleated erythroid cell is made by a process comprising: introducing into a nucleated erythroid cell precursor a nucleic acid encoding the first exogenous polypeptide; and culturing the nucleated erythroid cell precursor under conditions sufficient for expression of the the exogenous polypeptide and enucleation of the nucleated erythroid cell precursor.
  • the enucleated erythroid cell further comprises a second exogenous polypeptide comprising 4-1BBL or a functional fragment thereof, wherein the second exogenous polypeptide is present on the extracellular surface of the enucleated erythroid cell.
  • the enucleated erythroid cell comprises at least 1,000 copies of the second exogenous polypeptide.
  • the enucleated erythroid cell is made by a process comprising: introducing into a nucleated erythroid cell precursor a nucleic acid encoding the first exogenous polypeptide comprising 4- 1BBL or a functional fragment thereof, and a nucleic acid encoding the second exogenous polypeptide; and culturing the nucleated erythroid cell precursor under conditions sufficient for expression of the first exogenous polypeptide and the second exogenous polypeptide, and enucleation of the nucleated erythroid cell precursor.
  • the enucleated erythroid cell is not a hypotonically-dialyzed cell. In some embodiments of any of the methods described herein, the enucleated erythroid cell does not comprise a sortase- transfer signature. In some embodiments of any of the methods described herein, the subject is a human and the enucleated erythroid cell is a human cell.
  • kits that include: a pharmaceutical composition comprising an enucleated erythroid cell comprising a first exogenous polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface; and instructions for performing any of the methods described herein.
  • the enucleated erythroid cell comprises at least 1,000 copies of the first exogenous polypeptide.
  • the enucleated erythroid cell is made by a process comprising: introducing into a nucleated erythroid cell precursor a nucleic acid encoding the first exogenous polypeptide; and culturing the nucleated erythroid cell precursor under conditions sufficient for expression of the first exogenous polypeptide and enucleation of the nucleated erythroid cell precursor.
  • the enucleated erythroid cell further comprises a second exogenous polypeptide comprising 4-1BBL or a functional fragment thereof, wherein the second exogenous polypeptide is present on the extracellular surface of the enucleated erythroid cell.
  • the enucleated erythroid cell comprises at least 1,000 copies of the second exogenous polypeptide.
  • the enucleated erythroid cell is made by a process comprising: introducing into a nucleated erythroid cell precursor a nucleic acid encoding the first exogenous polypeptide and a nucleic acid encoding the second exogenous polypeptide; and culturing the nucleated erythroid cell precursor under conditions sufficient for expression of the first exogenous polypeptide and the second exogenous polypeptide, and enucleation of the nucleated erythroid cell precursor.
  • the enucleated erythroid cell is not a hypotonically-dialyzed cell. In some embodiments of any of the kits described herein, the enucleated erythroid cell does not comprise a sortase- transfer signature. In some embodiments of any of the kits described herein, the enucleated erythroid cell is a human cell.
  • engineered enucleated erythroid cell means an enucleated erythroid cell (e.g., a human enucleated erythroid cell) that comprises one or more (e.g., two, three, four, five, or six) exogenous protein(s) (e.g., any combination of the exemplary exogenous proteins described herein or known in the art).
  • an engineered enucleated erythroid cell can have one or more exogenous protein(s) present in its cytosol.
  • an engineered enucleated erythroid cell can have one or more exogenous protein(s) present on its extracellular surface.
  • an engineered enucleated erythroid cell can have (i) one or more exogenous protein(s) present in its cytosol and (ii) one or more exogenous proteins present on its extracellular surface.
  • engineered enucleated erythroid cells include click-conjugated enucleated erythroid cells, enucleated erythroid cells that have been hypotonically loaded, and enucleated erythroid cells that have been loaded by physical manipulation (e.g., any of the exemplary types of physical manipulation described herein or known in the art). Additional non-limiting aspects of engineered enucleated erythroid cells are described herein.
  • conjugated enucleated erythroid cell means an engineered enucleated erythroid cell that has at least one exogenous protein conjugated to another protein (e.g., an endogenous protein of an enucleated red blood cell or different exogenous protein) present on the extracellular surface of an engineered enucleated erythroid cells through the catalytic activity of an enzyme(s) and/or peptide sequence(s), and/or a chemical reaction.
  • another protein e.g., an endogenous protein of an enucleated red blood cell or different exogenous protein
  • hypotonically-loaded enucleated erythroid cell means an engineered enucleated erythroid cell that was generated, at least in part, by exposing an enucleated erythroid cell or an erythroid cell precursor to a low ionic strength buffer (e.g., any of the exemplary low ionic strength buffers described herein) comprising one or more exogenous protein(s).
  • a low ionic strength buffer e.g., any of the exemplary low ionic strength buffers described herein
  • Non-limiting examples of methods that can be used to generate a hypotonically-loaded enucleated erythroid cell are described herein. Additional methods for generating a hypotonically-loaded enucleated erythroid cell are known in the art.
  • nucleated erythroid cell loaded by physical manipulation means an enucleated erythroid cell that was generated, at least in part, by physically manipulating an erythroid cell precursor in a manner that results in the introduction of a nucleic acid encoding one or more exogenous protein(s) (e.g., any of the exemplary exogenous proteins described herein or known in the art) and/or exogenous polypeptides into the erythroid cell precursor.
  • Non-limiting examples of physical manipulation that can be used to introduce a nucleic acid encoding one or more exogenous protein(s) into an erythroid cell precursor include electroporation and particle-mediated transfection. Additional examples of physical manipulation that can be used to introduce a nucleic acid encoding one or more exogenous protein(s) into an erythroid cell precursor are known in the art.
  • exogenous protein refers to a protein that is introduced into or onto a cell, or is caused to be expressed by the cell by introducing an exogenous nucleic acid encoding the protein into the cell or into a precursor of the cell.
  • an exogenous protein is a protein encoded by an exogenous nucleic acid that was introduced into the cell or a precursor of the cell, which nucleic acid is optionally not retained by the cell.
  • an exogenous protein is a protein conjugated to the surface of the cell by chemical or enzymatic means.
  • Non limiting classes of exogenous proteins include enzymes, interleukins, cytokine receptors, Fc-binding molecules, T-cell activating ligands, T-cell receptors, immune inhibitory molecules, MHC molecules, APC-binding molecules, autoantigens, allergens, toxins, targeting agents, receptor ligands (e.g., receptor agonists or receptor antagonists), and antibodies or antibody fragments.
  • an exogenous polypeptide on the extracellular surface
  • an exogenous polypeptide that is physically attached to or at least partially embedded in the membrane of an enucleated erythroid cell e.g., a transmembrane protein, a peripheral membrane protein, a lipid-anchored protein (e.g., a GPI-anchor, an N-myristoylated protein, or a S-palmitoylated protein)
  • an exogenous protein that is stably bound to its cognate receptor where the cognate receptor is physically attached to the membrane of an enucleated erythroid cell (e.g., a ligand bound to its cognate receptor, where the cognate receptor is physically attached to the membrane of the enucleated erythroid cell).
  • Non-limiting methods for determining the presence of an exogenous protein on the extracellular surface of an enucleated erythroid cell include fluorescence-activated cell sorting (FACS), immunohistochemistry, cell-fractionation assays, and Western blotting.
  • FACS fluorescence-activated cell sorting
  • immunohistochemistry cell-fractionation assays
  • Western blotting Western blotting
  • the term “erythroid cell precursor” means a mammalian cell that is capable of eventually differentiating/devel oping into an enucleated erythroid cell.
  • the erythroid cell precursor is a cord blood stem cell, a CD34 + cell, a hematopoietic stem/progenitor cell (HSC, HSPC), a spleen colony forming (CFU-S) cell, a common myeloid progenitor (CMP) cell, a blastocyte colony-forming cell, a burst forming unit-erythroid/erythrocyte (BFU-E), a megakaryocyte-erythroid progenitor (MEP) cell, an erythroid colony-forming unit, or colony-forming unit erythrocyte (CFU-E), an induced pluripotent stem cell (iPSC), a mesenchymal stem cell (MSC), or a combination thereof.
  • HSC hematopoietic stem/pro
  • the subject or “subject in need of treatment” may be a primate (e.g., a human, a simian (e.g., a monkey (e.g., marmoset or baboon), or an ape (e.g., a gorilla, chimpanzee, orangutan, or gibbon)), a rodent (e.g., a mouse, a guinea pig, a hamster, or a rat), a rabbit, a dog, a cat, a horse, a sheep, a cow, a pig, or a goat.
  • a primate e.g., a human, a simian (e.g., a monkey (e.g., marmoset or baboon), or an ape (e.g., a gorilla, chimpanzee, orangutan, or gibbon)
  • a rodent e.g., a mouse,
  • the subject or “subject suitable for treatment” may be a non-human mammal, especially mammals that are conventionally used as models for demonstrating therapeutic efficacy in humans (e.g., a mouse, a pig, a rat, or a non-human primate) may be employed.
  • a subject can be previously diagnosed or identified as being in need of treatment by a medical professional (e.g., a physician, a laboratory technician, a physician’s assistant, a nurse, or a clinical laboratory technician) (e.g., previously diagnosed or identified as having a B7-H6-positive caner or previously diagnosed or identified as having a B7-H6-positive and HLA-E-negative cancer).
  • adult human subject refers to a human aged 18 years or older (e.g., aged 20 years or older, aged 25 years or older, aged 30 years or older, aged 35 years or older, aged 40 years or older, aged 45 years or older, aged 50 years or older, aged 55 years or older, aged 60 years or older, aged 65 years or older, aged 70 years or older, aged 75 years or older, aged 80 years or older, aged 85 years or older, aged 90 years or older, aged 95 years or older, or aged 100 years or older).
  • aged 18 years or older e.g., aged 20 years or older, aged 25 years or older, aged 30 years or older, aged 35 years or older, aged 40 years or older, aged 45 years or older, aged 50 years or older, aged 55 years or older, aged 60 years or older, aged 65 years or older, aged 70 years or older, aged 75 years or older, aged 80 years or older, aged 85 years or older, aged 90 years or older, aged 95 years or older, or aged 100 years or older).
  • the term “pediatric human subject” refers to a human aged under 18 years old (e.g., 17 years old and younger, 16 years old and younger, 15 years old and younger,
  • treating means a reduction in the number, severity, frequency, and/or duration of one or more symptoms of a medical disease or condition in a subject.
  • B7-H6 means a B7-H6 polypeptide or a B7-H6 mRNA including wild type human B7-H6 polypeptides and wild type human B7-H6 mRNAs, and variants thereof (e.g., truncated or mutated forms).
  • Non-limiting examples of methods of detecting a level of B7-H6 are described herein.
  • B7-H6-positive cancer means a cancer comprising a B7-H6-positive cancer cell.
  • B7-H6-positive cancers are described herein.
  • B7-H6 positive cancer cell means a cancer cell having a level of B7-H6 (B7-H6 protein or B7-H6 mRNA transcript) that is greater than a reference level of B7-H6.
  • B7-H6 B7-H6 protein or B7-H6 mRNA transcript
  • reference levels of B7-H6 are described herein.
  • B7-H6-positive cancer cells are also described herein.
  • HLA-E means a human leukocyte antigen-E (HLA- E) polypeptide or a HLA-E mRNA including wild type human HLA-E polypeptides and wild type human HLA-E mRNAs, and variants thereof (e.g., truncated or mutated forms).
  • HLA-E-negative cancer means a cancer having a level of HLA-E (HLA-E protein or HLA-E mRNA transcript) that is less than a reference level of HLA-E expression.
  • reference levels of HLA-E are described herein.
  • HLA-E-negative cancers are also described herein.
  • HLA-E-negative cancer cell means a cancer comprising a HLA-E-negative cancer cell.
  • Non-limiting examples of cancers that may be HLA-E-negative cancers are also described herein.
  • NKp30 means a NKp30 polypeptide or a NKp30 mRNA including wild type human NKp30 polypeptides and wild type human NKp30 mRNAs, and variants thereof (e.g., truncated or mutated forms).
  • the term “NKp30” also includes all known isoforms of NKp30 protein and NKp30 mRNA. Non-limiting examples of methods of detecting a level of NKp30 are described herein.
  • NKp30-positive lymphocyte means a lymphocyte having a level of NKp30 (NKp30 protein or NKp30 mRNA transcript) that is greater than a reference level of NKp30.
  • NKp30 NKp30 protein or NKp30 mRNA transcript
  • Non-limiting examples of reference levels of NKp30 are described herein.
  • NKG2A means a NK group 2 member A (NKG2A) polypeptide or a NKG2A mRNA including wild type human NKG2A polypeptides and wild type human NKG2A mRNAs, and variants thereof (e.g., truncated or mutated forms).
  • NKG2A NK group 2 member A
  • NKG2A mRNA including wild type human NKG2A polypeptides and wild type human NKG2A mRNAs, and variants thereof (e.g., truncated or mutated forms).
  • Non-limiting examples of methods of detecting a level of NKG2A are also described herein.
  • the nkg2a gene encodes two isoforms, NKG2A and NKG2B, with the latter lacking the stem region.
  • the term “NKG2A” as used herein does not include NKG2B proteins or mRNA transcripts.
  • NKG2A-negative lymphocyte means a lymphocyte having a level of NKG2A (NKG2A protein or NKG2A mRNA) that is less than a reference level of NKG2A.
  • NKG2A NKG2A protein or NKG2A mRNA
  • reference levels of NKG2A are described herein.
  • NK cell-mediated cytotoxicity means an NK cell inducing killing of other cells.
  • NK cell-mediated cytotoxicity is a mechanism used by an NK cell to induce killing of a cancer cell.
  • NK cell-mediated cytolysis means an NK cell having the ability to release lytic granules, where the lytic granules are used to induce killing of other cells.
  • the lytic granules include, at least, perforin and granzymes.
  • sortase transfer signature means an exogenous protein or polypeptide(s) that includes a sequence that can be created by a sortase reaction.
  • proteins and polypeptides that lack a sortase transfer signature are as described in WO 2017/123646, which is incorporated by reference in its entirety.
  • FIG. 1 is a graph showing that in vitro treatment of PBMCs with enucleated erythroid cells comprising a first exogenous polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, and a second exogenous polypeptide comprising 4-1BBL, on their extracellular surface (“RBC-IL15/IL15RA-4-1BBL”) results in increased proportions of NKp30-positive lymphocytes as compared to media control and treatment with rhIL-15 and anti-4-lBB agonistic antibody.
  • a first exogenous polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, and a second exogenous polypeptide comprising 4-1BBL, on their extracellular surface (“RBC-IL15/IL15RA-4-1BBL”) results in increased proportions of NKp30-positive lymphocytes as compared to media control and treatment
  • FIGs. 2A-2C are graphs showing maximum fold change relative to baseline in patients dosed with >1 x 10 10 erythroid cells comprising IL-15/IL-15RA and 4-1BBL prepared generally as described in Example 3 in the percentage of CD56 + lymphocytes that are positive for NKp30 (FIG. 2A), the percentage of
  • CD56dimCD16 + lymphocytes that are positive for NKp30 (FIG. 2B), and the percentage of CD56 + CD16 + lymphocytes that are positive for NKp30 (FIG. 2C).
  • FIGs. 3A-3B are graphs showing the maximum fold change relative to baseline in patients dosed with >1 x 10 10 erythroid cells comprising IL-15/IL-15RA and 4-1BBL prepared generally as described in Example 3 in absolute numbers of CD3 CD16/56 + NK cells/microliter (FIG. 3A) and the percentage of CD8 + Memory T cells expressing granzyme B (%GrB + of CD3 + CD8 + CD45RA) (FIG. 3B).
  • Cell surface markers in live lymphocytes (CD45 + ) within fresh whole blood were evaluated by flow cytometry at multiple timepoints at baseline (pre-treatment) and post-treatment. Post-treatment measurements were conducted across multiple dosing cycles.
  • FIGS. 4A-4B are graphs showing maximum fold change relative to baseline in patients dosed with >1 x 10 10 erythroid cells comprising IL-15/IL-15RA and 4-1BBL prepared generally as described in Example 3 in the percentage of CD3 + lymphocytes that are CD4-positive (FIG. 4A) and the percentage CD3 + lymphocytes that are CD8- positive (FIG. 4B).
  • Cell surface markers in live lymphocytes (CD45 + ) within fresh whole blood were evaluated by flow cytometry at multiple timepoints at baseline (pre treatment) and post-treatment. Post-treatment measurements were conducted across multiple dosing cycles.
  • NKp30-positive lymphocytes e.g., NKp30-positive NK cells
  • methods of increasing the number of NKp30-positive lymphocytes comprising administering to the subject previously identified or diagnosed as having a B7-H6- positive cancer a therapeutically effective amount of a pharmaceutical composition comprising a population of enucleated erythroid cells including a first exogenous polypeptide including (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface.
  • NKp30-positive lymphocytes e.g., NKp30-positive NK cells
  • methods of increasing the number of NKp30-positive lymphocytes comprising administering to a subject a therapeutically effective amount of a pharmaceutical composition comprising a population of enucleated erythroid cells including a first exogenous fusion polypeptide including (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface; and a second exogenous polypeptide including 4-1BBL or a functional fragment thereof, on its extracellular surface.
  • a pharmaceutical composition comprising a population of enucleated erythroid cells including a first exogenous fusion polypeptide including (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface; and a second exogenous polypeptide including 4-1BBL or a functional fragment thereof, on its extracellular surface.
  • Some embodiments of these methods result in at least a 1% increase, at least a 2% increase, at least a 3% increase, at least a 4% increase, at least a 5% increase, at least a 6% increase, at least a 7% increase, at least a 8% increase, at least a 9% increase, at least a 10% increase, at least a 15% increase, at least a 20% increase, at least a 25% increase, at least a 30% increase, at least a 35% increase, at least a 40% increase, at least a 45% increase, at least a 50% increase, at least a 55% increase, at least a 60% increase, at least a 65% increase, at least a 70% increase, at least a 75% increase, at least a 80% increase, at least a 85% increase, at least a 90% increase, at least a 95% increase, or at least a 100% increase, in the number of NKp30-positive lymphocytes (e.g., NKp30-positive NK cells) in the subject
  • Some embodiments of these methods result in about a 1% increase to about a 50% increase, about a 1% increase to about a 45% increase, about a 1% increase to about a 40% increase, about a 1% increase to about a 35% increase, about a 1% increase to about a 30% increase, about a 1% increase to about a 25% increase, about a 1% increase to about a 20% increase, about a 1% increase to about a 15% increase, about a 1% increase to about a 10% increase, about a 1% increase to about a 5% increase, about a 5% increase to about a 50% increase, about a 5% increase to about a 45% increase, about a 5% increase to about a 40% increase, about a 5% increase to about a 35% increase, about a 5% increase to about a 30% increase, about a 5% increase to about a 25% increase, about a 5% increase to about a 20% increase, about a 5% increase to about a 15% increase, about a 5% increase to
  • Some embodiments of these methods result in at least a 1% increase, at least a 2% increase, at least a 3% increase, at least a 4% increase, at least a 5% increase, at least a 6% increase, at least a 7% increase, at least a 8% increase, at least a 9% increase, at least a 10% increase, at least a 15% increase, at least a 20% increase, at least a 25% increase, at least a 30% increase, at least a 35% increase, at least a 40% increase, at least a 45% increase, at least a 50% increase, at least a 55% increase, at least a 60% increase, at least a 65% increase, at least a 70% increase, at least a 75% increase, at least a 80% increase, at least a 85% increase, at least a 90% increase, at least a 95% increase, or at least a 100% increase, in the number of NKp30- positive/NKG2A-negative lymphocytes (e.g., NKp30-positive
  • Some embodiments of these methods result in about a 1% increase to about a 50% increase, about a 1% increase to about a 45% increase, about a 1% increase to about a 40% increase, about a 1% increase to about a 35% increase, about a 1% increase to about a 30% increase, about a 1% increase to about a 25% increase, about a 1% increase to about a 20% increase, about a 1% increase to about a 15% increase, about a 1% increase to about a 10% increase, about a 1% increase to about a 5% increase, about a 5% increase to about a 50% increase, about a 5% increase to about a 45% increase, about a 5% increase to about a 40% increase, about a 5% increase to about a 35% increase, about a 5% increase to about a 30% increase, about a 5% increase to about a 25% increase, about a 5% increase to about a 20% increase, about a 5% increase to about a 15% increase, about a 5% increase to
  • Some embodiments of the methods described herein result in at least a 1% increase, at least a 2% increase, at least a 3% increase, at least a 4% increase, at least a 5% increase, at least a 6% increase, at least a 7% increase, at least a 8% increase, at least a 9% increase, at least a 10% increase, at least a 15% increase, at least a 20% increase, at least a 25% increase, at least a 30% increase, at least a 35% increase, at least a 40% increase, at least a 45% increase, at least a 50% increase, at least a 55% increase, at least a 60% increase, at least a 65% increase, at least a 70% increase, at least a 75% increase, at least a 80% increase, at least a 85% increase, at least a 90% increase, at least a 95% increase, or at least a 100% increase, in the trafficking of NKp30-positive lymphocytes (e.g., NKp30-positive NK cells
  • Some embodiments of any of the methods described herein result in about a 1% increase to about a 50% increase, about a 1% increase to about a 45% increase, about a 1% increase to about a 40% increase, about a 1% increase to about a 35% increase, about a 1% increase to about a 30% increase, about a 1% increase to about a 25% increase, about a 1% increase to about a 20% increase, about a 1% increase to about a 15% increase, about a 1% increase to about a 10% increase, about a 1% increase to about a 5% increase, about a 5% increase to about a 50% increase, about a 5% increase to about a 45% increase, about a 5% increase to about a 40% increase, about a 5% increase to about a 35% increase, about a 5% increase to about a 30% increase, about a 5% increase to about a 25% increase, about a 5% increase to about a 20% increase, about a 5% increase to about a 15% increase, about
  • Some embodiments of the methods described herein result in at least a 1% increase, at least a 2% increase, at least a 3% increase, at least a 4% increase, at least a 5% increase, at least a 6% increase, at least a 7% increase, at least a 8% increase, at least a 9% increase, at least a 10% increase, at least a 15% increase, at least a 20% increase, at least a 25% increase, at least a 30% increase, at least a 35% increase, at least a 40% increase, at least a 45% increase, at least a 50% increase, at least a 55% increase, at least a 60% increase, at least a 65% increase, at least a 70% increase, at least a 75% increase, at least a 80% increase, at least a 85% increase, at least a 90% increase, at least a 95% increase, or at least a 100% increase, in the trafficking of NKp30-positive/NKG2A-negative lymphocytes (e.g., NK
  • Some embodiments of any of the methods described herein result in about a 1% increase to about a 50% increase, about a 1% increase to about a 45% increase, about a 1% increase to about a 40% increase, about a 1% increase to about a 35% increase, about a 1% increase to about a 30% increase, about a 1% increase to about a 25% increase, about a 1% increase to about a 20% increase, about a 1% increase to about a 15% increase, about a 1% increase to about a 10% increase, about a 1% increase to about a 5% increase, about a 5% increase to about a 50% increase, about a 5% increase to about a 45% increase, about a 5% increase to about a 40% increase, about a 5% increase to about a 35% increase, about a 5% increase to about a 30% increase, about a 5% increase to about a 25% increase, about a 5% increase to about a 20% increase, about a 5% increase to about a 15% increase, about
  • Some embodiments of the methods described herein result in an increase in the number of NKp30-positive, CD45 -positive, CD56-positive lymphocytes (e.g., NKp30-positive, CD45-positive, and CD56-positive NK cells) in the subject.
  • NKp30-positive, CD45-positive, and CD56-positive lymphocytes e.g., NKp30-positive, CD45-positive, and CD56-positive NK cells
  • Some embodiments of these methods result in at least a 1.1-fold increase, at least a 1.2-fold increase, at least a 1.3-fold increase, at least a 1.4-fold increase, at least a 1.5-fold increase, at least a 1.6-fold increase, at least a 1.7-fold increase, at least a 1.8-fold increase, at least a 1.9-fold increase, at least a 2.0-fold increase, at least a 2.2-fold increase, at least a 2.4-fold increase, at least a 2.6-fold increase, at least a 2.8-fold increase, at least a 3.0-fold increase, at least a 3.2-fold increase, at least a 3.4-fold increase, at least a 3.6-fold increase, at least a 3.8-fold increase, at least a 4.0-fold increase, at least a 4.2-fold increase, at least a 4.4-fold increase, at least a 4.6-fold increase, at least a 4.8-fold increase, or at least a 5.0-fold
  • a 1.1-fold to about a 5-fold increase e.g., about a 1.1 -fold to about a 4.5-fold increase, about a 1.1 -fold to about a 4.0-fold increase, about a 1.1-fold to about a 3.5-fold increase, about a 1.1-fold to about a 3.0- fold increase, about a 1.1 -fold to about a 2.5-fold increase, about a 1.1 -fold to about a 2.4-fold increase, about a 1.1 -fold to about a 2.2-fold increase, about a 1.1 -fold to about a 2.0-fold increase, about a 1.1 -fold to about a 1.8-fold increase, about a 1.1- fold to about a 1.6-fold increase, about a 1.1 -fold to about a 1.4-fold increase, about a
  • CD45 -positive, CD56-positive lymphocytes are NKp30-positive, CD45-positive, CD56-positive NK cells.
  • Some embodiments of the methods described herein result in an increase in the number of NKp30-positive, CD45 -positive, CD 16-positive, CD56-dim lymphocytes (e.g., NKp30-positive, CD45-positive, CD 16-positive, CD56-dimNK cells) in the subject.
  • NKp30-positive, CD45-positive, CD 16-positive, CD56-dim lymphocytes e.g., NKp30-positive, CD45-positive, CD 16-positive, CD56-dimNK cells
  • Some embodiments of these methods result in at least a 1.1 -fold increase, at least a 1.2-fold increase, at least a 1.4-fold increase, at least a 1.6-fold increase, at least a 1.8-fold increase, at least a 2.0-fold increase, at least a 2.2-fold increase, at least a 2.4-fold increase, at least a 2.6-fold increase, at least a 2.8-fold increase, at least a 3.0-fold increase, at least a 3.2-fold increase, at least a 3.4-fold increase, at least a 3.6-fold increase, at least a 3.8-fold increase, at least a 4.0-fold increase, at least a 4.2-fold increase, at least a 4.4-fold increase, at least a 4.6-fold increase, at least a 4.8-fold increase, or at least a 5.0-fold increase, in the percentage of CD45 -positive, CD 16-positive, CD56-dim lymphocytes that are NKp30-positive (e.g.
  • a 1.1-fold to about a 5-fold increase e.g., about a 1.1-fold to about a 4.5-fold increase, about a 1.1-fold to about a 4.0-fold increase, about a 1.1-fold to about a 3.5- fold increase, about a 1.1 -fold to about a 3.0-fold increase, about a 1.1 -fold to about a 2.5-fold increase, about a 1.1-fold to about a 2.4-fold increase, about a 1.1-fold to about a 2.2-fold increase, about a 1.1-fold to about a 2.0-fold increase, about a 1.1- fold to about a 1.8-fold increase, about a 1.1 -fold to about a 1.6-fold increase, about a 1.1-fold to about a 1.4-fold increase, about a 1.1-fold to about a 1.2-fold increase, about 1.2-fold to about a 5-fold increase, about a 1.1-fold to about a 5-fold increase, about a
  • 1.4-fold to about a 3.5-fold increase about a 1.4-fold to about a 3.0-fold increase, about a 1.4-fold to about a 2.5-fold increase, about a 1.4-fold to about a 2.4-fold increase, about a 1.4-fold to about a 2.2-fold increase, about a 1.4-fold to about a 2.0- fold increase, about a 1.4-fold to about a 1.8-fold increase, about a 1.4-fold to about a 1.6-fold increase, about 1.6-fold to about a 5-fold increase, about a 1.6-fold to about a
  • 1.6-fold to about a 2.2-fold increase about a 1.6-fold to about a 2.0-fold increase, about a 1.6-fold to about a 1.8-fold increase, about 1.8-fold to about a 5-fold increase, about a 1.8-fold to about a 4.5-fold increase, about a 1.8-fold to about a 4.0-fold increase, about a 1.8-fold to about a 3.5-fold increase, about a 1.8-fold to about a 3.0- fold increase, about a 1.8-fold to about a 2.5-fold increase, about a 1.8-fold to about a
  • 2.4-fold increase about a 1.8-fold to about a 2.2-fold increase, about a 1.8-fold to about a 2.0-fold increase, about 2.0-fold to about a 5 -fold increase, about a 2.0-fold to about a 4.5-fold increase, about a 2.0-fold to about a 4.0-fold increase, about a 2.0- fold to about a 3.5 -fold increase, about a 2.0-fold to about a 3.0-fold increase, about a 2.0-fold to about a 2.5-fold increase, about a 2.0-fold to about a 2.4-fold increase, about a 2.0-fold to about a 2.2-fold increase, about 2.2-fold to about a 5-fold increase, about a 2.2-fold to about a 4.5-fold increase, about a 2.2-fold to about a 4.0-fold increase, about a 2.2-fold to about a 3.5-fold increase, about a 2.2-fold to about a 3.0- fold increase, about a 2.2-fold to about a
  • Some embodiments of the methods described herein result in an increase in the number of NKp30-positive, CD45 -positive, CD 16-positive, CD56-positive lymphocytes (e.g., NKp30-positive, CD45-positive, CD 16-positive, CD56-positive NK cells) in the subject.
  • NKp30-positive, CD45-positive, CD 16-positive, CD56-positive lymphocytes e.g., NKp30-positive, CD45-positive, CD 16-positive, CD56-positive NK cells
  • Some embodiments of these methods result in at least a 1.1- fold increase, at least a 1.2-fold increase, at least a 1.4-fold increase, at least a 1.6-fold increase, at least a 1.8-fold increase, at least a 2.0-fold increase, at least a 2.2-fold increase, at least a 2.4-fold increase, at least a 2.6-fold increase, at least a 2.8-fold increase, at least a 3.0-fold increase, at least a 3.2-fold increase, at least a 3.4-fold increase, at least a 3.6-fold increase, at least a 3.8-fold increase, at least a 4.0-fold increase, at least a 4.2-fold increase, at least a 4.4-fold increase, at least a 4.6-fold increase, at least a 4.8-fold increase, or at least a 5.0-fold increase, in the percentage of CD45 -positive, CD 16-positive, CD56-positive lymphocytes that are NKp30- positive (e.g., CD
  • a 1.1-fold to about a 5-fold increase e.g., about a 1.1-fold to about a 4.5-fold increase, about a 1.1-fold to about a 4.0-fold increase, about a 1.1-fold to about a 3.5-fold increase, about a 1.1-fold to about a 3.0-fold increase, about a 1.1-fold to about a 2.5-fold increase, about a 1.1-fold to about a 2.4- fold increase, about a 1.1 -fold to about a 2.2-fold increase, about a 1.1 -fold to about a 2.0-fold increase, about a 1.1-fold to about a 1.8-fold increase, about a 1.1-fold to about a 1.6-fold increase, about a 1.1-fold to about a 1.4-fold increase, about a 1.1-fold to about a 1.2-fold increase, about 1.2-fold to about a 5-fold increase, about a 1.2
  • 1.2-fold to about a 3.5-fold increase about a 1.2-fold to about a 3.0-fold increase, about a 1.2-fold to about a 2.5-fold increase, about a 1.2-fold to about a 2.4-fold increase, about a 1.2-fold to about a 2.2-fold increase, about a 1.2-fold to about a 2.0- fold increase, about a 1.2-fold to about a 1.8-fold increase, about a 1.2-fold to about a 1.6-fold increase, about a 1.2-fold to about a 1.4-fold increase, about 1.4-fold to about a 5-fold increase, about a 1.4-fold to about a 4.5-fold increase, about a 1.4-fold to about a 4.0-fold increase, about a 1.4-fold to about a 3.5-fold increase, about a 1.4-fold to about a 3.0-fold increase, about a 1.4-fold to about a 2.5-fold increase, about a 1.4-fold to about a
  • the NKp30-positive, CD56-positive, CD 16-positive, CD45- positive lymphocytes are NKp30-positive, CD56-positive, CD 16-positive, CD45- positive NK cells.
  • Some embodiments of the methods result in an increase in the number or concentration of circulating NK cells (CD3 CD16/CD56 + cells).
  • the methods result in at least a 1.1 -fold increase, at least a 1.2-fold increase, at least a 1.4-fold increase, at least a 1.6-fold increase, at least a 1.8-fold increase, at least a 2.0-fold increase, at least a 2.2-fold increase, at least a 2.4-fold increase, at least a 2.6-fold increase, at least a 2.8-fold increase, at least a 3.0-fold increase, at least a 3.2-fold increase, at least a 3.4-fold increase, at least a 3.6-fold increase, at least a 3.8-fold increase, at least a 4.0-fold increase, at least a 4.2-fold increase, at least a 4.4-fold increase, at least a 4.6-fold increase, at least a 4.8-fold increase, or at least a 5.0-fold increase, in the number or
  • the methods result in about a 1.1 -fold to about a 5-fold increase (e.g., about a 1.1-fold to about a 4.5-fold increase, about a 1.1-fold to about a 4.0-fold increase, about a 1.1-fold to about a 3.5-fold increase, about a 1.1- fold to about a 3.0-fold increase, about a 1.1-fold to about a 2.5-fold increase, about a
  • 1.1-fold to about a 2.4-fold increase about a 1.1-fold to about a 2.2-fold increase, about a 1.1-fold to about a 2.0-fold increase, about a 1.1-fold to about a 1.8-fold increase, about a 1.1 -fold to about a 1.6-fold increase, about a 1.1 -fold to about a 1.4- fold increase, about a 1.1-fold to about a 1.2-fold increase, about 1.2-fold to about a 5- fold increase, about a 1.2-fold to about a 4.5-fold increase, about a 1.2-fold to about a 4.0-fold increase, about a 1.2-fold to about a 3.5 -fold increase, about a 1.2-fold to about a 3.0-fold increase, about a 1.2-fold to about a 2.5-fold increase, about a 1.2- fold to about a 2.4-fold increase, about a 1.2-fold to about a 2.2-fold increase, about a 1.1-fold
  • Some embodiments of the methods result in an increase in the number or concentration of CD8-positive memory T cells expressing granzyme B (Granzyme B- positive, CD8-positive, CD45RA-negative cells).
  • the methods result in at least a 1.1-fold increase, at least a 1.2-fold increase, at least a 1.4-fold increase, at least a 1.6-fold increase, at least a 1.8-fold increase, at least a 2.0-fold increase, at least a 2.2-fold increase, at least a 2.4-fold increase, at least a 2.6-fold increase, at least a 2.8-fold increase, at least a 3.0-fold increase, at least a 3.2-fold increase, at least a 3.4-fold increase, at least a 3.6-fold increase, at least a 3.8-fold increase, at least a 4.0-fold increase, at least a 4.2-fold increase, at least a 4.4-fold increase, at least a 4.6-fold increase, at least a 4.8-
  • the methods result in about a 1.1-fold to about a 5-fold increase (e.g., about a 1.1-fold to about a 4.5-fold increase, about a 1.1-fold to about a 4.0-fold increase, about a 1.1- fold to about a 3.5 -fold increase, about a 1.1 -fold to about a 3.0-fold increase, about a
  • 1.2-fold to about a 2.2-fold increase about a 1.2-fold to about a 2.0-fold increase, about a 1.2-fold to about a 1.8-fold increase, about a 1.2-fold to about a 1.6-fold increase, about a 1.2-fold to about a 1.4-fold increase, about 1.4-fold to about a 5-fold increase, about a 1.4-fold to about a 4.5-fold increase, about a 1.4-fold to about a 4.0- fold increase, about a 1.4-fold to about a 3.5-fold increase, about a 1.4-fold to about a 3.0-fold increase, about a 1.4-fold to about a 2.5 -fold increase, about a 1.4-fold to about a 2.4-fold increase, about a 1.4-fold to about a 2.2-fold increase, about a 1.4- fold to about a 2.0-fold increase, about a 1.4-fold to about a 1.8-fold increase, about a 1.4-fold to about
  • 2.4-fold increase about a 2.0-fold to about a 2.2-fold increase, about 2.2-fold to about a 5-fold increase, about a 2.2-fold to about a 4.5-fold increase, about a 2.2-fold to about a 4.0-fold increase, about a 2.2-fold to about a 3.5-fold increase, about a 2.2- fold to about a 3.0-fold increase, about a 2.2-fold to about a 2.5-fold increase, about a 2.2-fold to about a 2.4-fold increase, about 2.4-fold to about a 5-fold increase, about a
  • 2.4-fold to about a 4.5-fold increase about a 2.4-fold to about a 4.0-fold increase, about a 2.4-fold to about a 3.5-fold increase, about a 2.4-fold to about a 3.0-fold increase, about a 2.4-fold to about a 2.5-fold increase, about 2.5-fold to about a 5-fold increase, about a 2.5-fold to about a 4.5-fold increase, about a 2.5-fold to about a 4.0- fold increase, about a 2.5-fold to about a 3.5-fold increase, about a 2.5-fold to about a 3.0-fold increase, about 3.0-fold to about a 5-fold increase, about a 3.0-fold to about a
  • a subject previously identified or diagnosed as having a B7-H6-positive cancer comprising administering to a subject previously identified or diagnosed as having a B7-H6-positive cancer a therapeutically effective amount of a pharmaceutical composition comprising a population of enucleated erythroid cells including a first exogenous polypeptide that includes (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface.
  • Also provided herein are methods of treating a subject having a B7-H6-positive cancer the method comprising administering to a subject a therapeutically effective amount of a pharmaceutical composition comprising a population of enucleated erythroid cells including a first exogenous fusion polypeptide including (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface; and a second exogenous polypeptide including 4-1BBL or a functional fragment thereof, wherein the exogenous polypeptide, on its extracellular surface.
  • a pharmaceutical composition comprising a population of enucleated erythroid cells including a first exogenous fusion polypeptide including (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface; and a second exogenous polypeptide including 4-1BBL or a functional fragment thereof, wherein the exogenous polypeptide, on its extra
  • a pharmaceutical composition comprising a population of enucleated erythroid cells including a first exogenous polypeptide that includes (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface.
  • Also provided herein are methods of decreasing the number and/or proliferation of B7-H6-positive cancer cells in a subject previously identified or diagnosed as having a B7-H6-positive cancer the method comprising administering to a subject a therapeutically effective amount of a pharmaceutical composition comprising a population of enucleated erythroid cells including a first exogenous fusion polypeptide including (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface; and a second exogenous polypeptide including 4-1BBL or a functional fragment thereof, wherein the exogenous polypeptide is on its extracellular surface.
  • a pharmaceutical composition comprising a population of enucleated erythroid cells including a first exogenous fusion polypeptide including (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface; and a second exogenous polypeptide including 4-1
  • Some embodiments of these methods result in at least a 1% reduction, at least a 2% reduction, at least a 3% reduction, at least a 4% reduction, at least a 5% reduction, at least a 6% reduction, at least a 7% reduction, at least a 8% reduction, at least a 9% reduction, at least a 10% reduction, at least a 15% reduction, at least a 20% reduction, at least a 25% reduction, at least a 30% reduction, at least a 35% reduction, at least a 40% reduction, at least a 45% reduction, at least a 50% reduction, at least a 55% reduction, at least a 60% reduction, at least a 65% reduction, at least a 70% reduction, at least a 75% reduction, at least a 80% reduction, at least a 85% reduction, at least a 90% reduction, at least a 95% reduction, or at least a 99% reduction, in the number of B7-H6-positive cancer cells in the subject (e.g., as compared to the number of B7
  • Some embodiments of these methods result in about a 1% reduction to about a 99% reduction, about a 1% reduction to about a 90% reduction, about a 1% reduction to about a 80% reduction, about a 1% reduction to about a 70% reduction, about a 1% reduction to about a 60% reduction, about a 1% reduction to about a 50% reduction, about a 1% reduction to about a 40% reduction, about a 1% reduction to about a 30% reduction, about a 1% reduction to about a 20% reduction, about a 1% reduction to about a 10% reduction, about a 5% reduction to about a 99% reduction, about a 5% reduction to about a 90% reduction, about a 5% reduction to about a 80% reduction, about a 5% reduction to about a 70% reduction, about a 5% reduction to about a 60% reduction, about a 5% reduction to about a 50% reduction, about a 5% reduction to about a 40% reduction, about a 5% reduction to about a 30% reduction, about a 5% reduction to about
  • Some embodiments of these methods result in at least a 1% reduction, at least a 2% reduction, at least a 3% reduction, at least a 4% reduction, at least a 5% reduction, at least a 6% reduction, at least a 7% reduction, at least a 8% reduction, at least a 9% reduction, at least a 10% reduction, at least a 15% reduction, at least a 20% reduction, at least a 25% reduction, at least a 30% reduction, at least a 35% reduction, at least a 40% reduction, at least a 45% reduction, at least a 50% reduction, at least a 55% reduction, at least a 60% reduction, at least a 65% reduction, at least a 70% reduction, at least a 75% reduction, at least a 80% reduction, at least a 85% reduction, at least a 90% reduction, at least a 95% reduction, or at least a 99% reduction, in the number of B7-H6-positive/HLA-E-negative cancer cells in the subject (e.g., as
  • Some embodiments of these methods result in about a 1% reduction to about a 99% reduction, about a 1% reduction to about a 90% reduction, about a 1% reduction to about a 80% reduction, about a 1% reduction to about a 70% reduction, about a 1% reduction to about a 60% reduction, about a 1% reduction to about a 50% reduction, about a 1% reduction to about a 40% reduction, about a 1% reduction to about a 30% reduction, about a 1% reduction to about a 20% reduction, about a 1% reduction to about a 10% reduction, about a 5% reduction to about a 99% reduction, about a 5% reduction to about a 90% reduction, about a 5% reduction to about a 80% reduction, about a 5% reduction to about a 70% reduction, about a 5% reduction to about a 60% reduction, about a 5% reduction to about a 50% reduction, about a 5% reduction to about a 40% reduction, about a 5% reduction to about a 30% reduction, about a 5% reduction to about
  • Some embodiments of these methods results in at least a 1% reduction, at least a 2% reduction, at least a 3% reduction, at least a 4% reduction, at least a 5% reduction, at least a 6% reduction, at least a 7% reduction, at least a 8% reduction, at least a 9% reduction, at least a 10% reduction, at least a 15% reduction, at least a 20% reduction, at least a 25% reduction, at least a 30% reduction, at least a 35% reduction, at least a 40% reduction, at least a 45% reduction, at least a 50% reduction, at least a 55% reduction, at least a 60% reduction, at least a 65% reduction, at least a 70% reduction, at least a 75% reduction, at least a 80% reduction, at least a 85% reduction, at least a 90% reduction, at least a 95% reduction, or at least a 99% reduction, in the proliferation of B7-H6-positive cancer cells in the subject (e.g., as compared to the proliferation of B7
  • Some embodiments of these methods results in about a 1% reduction to about a 99% reduction, about a 1% reduction to about a 90% reduction, about a 1% reduction to about a 80% reduction, about a 1% reduction to about a 70% reduction, about a 1% reduction to about a 60% reduction, about a 1% reduction to about a 50% reduction, about a 1% reduction to about a 40% reduction, about a 1% reduction to about a 30% reduction, about a 1% reduction to about a 20% reduction, about a 1% reduction to about a 10% reduction, about a 5% reduction to about a 99% reduction, about a 5% reduction to about a 90% reduction, about a 5% reduction to about a 80% reduction, about a 5% reduction to about a 70% reduction, about a 5% reduction to about a 60% reduction, about a 5% reduction to about a 50% reduction, about a 5% reduction to about a 40% reduction, about a 5% reduction to about a 30% reduction, about a 5% reduction to about
  • Some embodiments of these methods results in at least a 1% reduction, at least a 2% reduction, at least a 3% reduction, at least a 4% reduction, at least a 5% reduction, at least a 6% reduction, at least a 7% reduction, at least a 8% reduction, at least a 9% reduction, at least a 10% reduction, at least a 15% reduction, at least a 20% reduction, at least a 25% reduction, at least a 30% reduction, at least a 35% reduction, at least a 40% reduction, at least a 45% reduction, at least a 50% reduction, at least a 55% reduction, at least a 60% reduction, at least a 65% reduction, at least a 70% reduction, at least a 75% reduction, at least a 80% reduction, at least a 85% reduction, at least a 90% reduction, at least a 95% reduction, or at least a 99% reduction, in the proliferation of B7-H6-positive/HLA-E-negative cancer cells in the subject (e.g., as
  • Some embodiments of these methods results in about a 1% reduction to about a 99% reduction, about a 1% reduction to about a 90% reduction, about a 1% reduction to about a 80% reduction, about a 1% reduction to about a 70% reduction, about a 1% reduction to about a 60% reduction, about a 1% reduction to about a 50% reduction, about a 1% reduction to about a 40% reduction, about a 1% reduction to about a 30% reduction, about a 1% reduction to about a 20% reduction, about a 1% reduction to about a 10% reduction, about a 5% reduction to about a 99% reduction, about a 5% reduction to about a 90% reduction, about a 5% reduction to about a 80% reduction, about a 5% reduction to about a 70% reduction, about a 5% reduction to about a 60% reduction, about a 5% reduction to about a 50% reduction, about a 5% reduction to about a 40% reduction, about a 5% reduction to about a 30% reduction, about a 5% reduction to about
  • a method of inducing killing a B7-H6-positive cancer cell in a subject previously identified or diagnosed as having a B7-H6-positive cancer comprising administering a therapeutically effective amount of a pharmaceutical composition comprising a population of enucleated erythroid cells including an first exogenous polypeptide that includes (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface.
  • Also provided herein are methods of killing a B7-H6-positive cancer cell in a subject previously identified or diagnosed as having a B7-H6-positive cancer the method comprising administering to a subject a therapeutically effective amount of a pharmaceutical composition comprising a population of enucleated erythroid cells including a first exogenous fusion polypeptide including (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface; and a second exogenous polypeptide including 4- 1BBL or a functional fragment thereof, wherein the exogenous polypeptide is on its extracellular surface.
  • a method of reducing the volume of a solid tumor in a subject previously identified or diagnosed as having a B7-H6-positive cancer comprising administering a therapeutically effective amount of a pharmaceutical composition comprising a population of enucleated erythroid cells including an first exogenous polypeptide that includes (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface.
  • a pharmaceutical composition comprising a population of enucleated erythroid cells including a first exogenous fusion polypeptide including (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface; and a second exogenous polypeptide including 4- 1BBL or a functional fragment
  • Some embodiments of these methods result in at least a 1% reduction, at least a 2% reduction, at least a 3% reduction, at least a 4% reduction, at least a 5% reduction, at least a 6% reduction, at least a 7% reduction, at least a 8% reduction, at least a 9% reduction, at least a 10% reduction, at least a 15% reduction, at least a 20% reduction, at least a 25% reduction, at least a 30% reduction, at least a 35% reduction, at least a 40% reduction, at least a 45% reduction, at least a 50% reduction, at least a 55% reduction, at least a 60% reduction, at least a 65% reduction, at least a 70% reduction, at least a 75% reduction, at least a 80% reduction, at least a 85% reduction, at least a 90% reduction, at least a 95% reduction, or at least a 99% reduction, in the volume of the solid tumor in the subject (e.g., as compared to the volume of the solid tumor prior to the administering
  • Some embodiments of these methods result in about a 1% reduction to about a 99% reduction, about a 1% reduction to about a 90% reduction, about a 1% reduction to about a 80% reduction, about a 1% reduction to about a 70% reduction, about a 1% reduction to about a 60% reduction, about a 1% reduction to about a 50% reduction, about a 1% reduction to about a 40% reduction, about a 1% reduction to about a 30% reduction, about a 1% reduction to about a 20% reduction, about a 1% reduction to about a 10% reduction, about a 5% reduction to about a 99% reduction, about a 5% reduction to about a 90% reduction, about a 5% reduction to about a 80% reduction, about a 5% reduction to about a 70% reduction, about a 5% reduction to about a 60% reduction, about a 5% reduction to about a 50% reduction, about a 5% reduction to about a 40% reduction, about a 5% reduction to about a 30% reduction, about a 5% reduction to about
  • the subject is an adult human subject. In some embodiments of any of the methods described herein, the subject is a pediatric human subject.
  • compositions and methods are described below. As can be appreciated by those in the field, the exemplary aspects listed below can be used in any combination, and can be combined with other aspects known in the field.
  • aNKp30 polypeptide refers to the amino acid sequence encoded by the natural cytotoxicity triggering receptor 3 (NCR3) gene.
  • NCR3 natural cytotoxicity triggering receptor 3
  • An exemplary NKp30 polypeptide includes without limitation amino acid sequences corresponding to NCBI reference sequences: NP_001138938.1, NP_001138939.1, and NP_667341.1.
  • aNKp30 polypeptide includes an amino acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to MAWMLLLILIMVHPGSCALWVSOPPEIRTLEGSSAFLPCSFNASOGRLAIGSV TWFRDEVVPGKEVRNGTPEFRGRLAPLASSRFLHDHQAELHIRDVRGHDASI YV CRVEVLGLGV GT GNGTRLVVEKEHPQLGAGTVLLLRAGFY AV SFLS V AV GSTVYYQGKYAKSTLSGFPQL (SEQ ID NO: 1) (with or without the signal sequence).
  • SEQ ID NO: 1 indicates the signal sequence.
  • aNKp30 polypeptide is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • TCTACTCTCTCCGGATTCCCCCAACTCTGA SEQ ID NO: 2.
  • aNKp30 polypeptide includes an amino acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • aNKp30 polypeptide is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • AGAGCCCAGATGTCCCTAG (SEQ ID NO: 4).
  • aNKp30 polypeptide includes an amino acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • aNKp30 polypeptide is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the methods include detecting the number of NKp30- positive lymphocytes in the subject.
  • Any appropriate sample containing NKp30- positive lymphocytes can be obtained from a subject.
  • a whole blood sample, a plasma sample, a serum sample, a sample containing a cell fraction (PBMC) of a whole blood sample, a tissue sample, a biopsy sample, and a laser capture dissected biological sample e.g., tumor or tissue sample
  • PBMC cell fraction
  • detecting the number of NKp30-positive lymphocytes is done prior to, simultaneously with, or after, administering a therapeutically effective amount of a pharmaceutical composition comprising a population of enucleated erythroid cells (e.g., any of the exemplary enucleated erythroid cells described herein).
  • the NKp30-positive cells can be determined using any an antibody that binds specifically to NKp30 or an antigen-binding fragment thereof (e.g., Beckman Coulter, clone Z25).
  • NKp30 protein expression can be assessed using fluorescence-assisted cell sorting (FACS) (e.g., using compensation beads (e.g., Bangs beads)) or assessed by other immunological based methods including, without limitation, Western blot analysis, ELISA, tissue array analysis, in situ hybridization, and immunofluoresence, using, e.g., an antibody that binds specifically to NKp30 or an antigen-binding fragment thereof.
  • FACS fluorescence-assisted cell sorting
  • NKp30-positive cells can be determined by measuring NKp30 mRNA.
  • Non-limiting methods of quantifying RNA include: qRT-PCR, RNA-sequencing, fluorescence in situ hybridization combined with flow cytometry, microfluidic capillary electrophoresis, and in situ hybridization.
  • an NKp30-positive lymphocyte is determined by comparing the level of NKp30 in the lymphocyte as compared to a reference level (e.g., a level of NKp30 in a control non-activated or resting lymphocyte, or a level in a cell that is not a lymphocyte).
  • a reference level e.g., a level of NKp30 in a control non-activated or resting lymphocyte, or a level in a cell that is not a lymphocyte.
  • the reference level of NKp30 can be one transcript per one million transcripts.
  • a B7-H6 polypeptide refers to the amino acid sequence encoded by the natural killer cell cytotoxicity receptor 3 ligand 1 (NCR3LG1) gene.
  • An exemplary B7-H6 polypeptide includes without limitation amino acid sequence corresponding to NCBI reference sequences: NP_001189368.1.
  • B7-H6, a member of the B7 family of immunoreceptors, is as a cell-surface ligand for NKp30.
  • a B7-H6 polypeptide can include a sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • SEQ ID NO: 7 (with or without its signal peptide).
  • the underlined portion of SEQ ID NO: 7 above indicates the signal peptide.
  • a B7-H6 polypeptide is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • TACAGTAA (SEQ ID NO: 8).
  • the methods include detecting the presence of B7-H6- positive cancer cells in the subject.
  • Any appropriate sample containing B7-H6- positive cancer cells can be obtained from a subject.
  • a whole blood sample, a plasma sample, a serum sample, a sample containing a cell fraction (PBMC) of a whole blood sample, a tissue sample, a biopsy sample, and a laser capture dissected biological sample e.g., tumor or tissue sample
  • detecting the B7-H6-positive cancer cells is done prior to, simultaneously with, or after, administering the enucleated erythroid cells.
  • the B7-H6-positive cancer cells can be determined using an antibody that binds specifically to B7-H6 or an antigen-binding fragment thereof (e.g., B7-H6 (Abeam)).
  • B7-H6 protein expression can be assessed using fluorescence-associated cell sorting (FACS) and can also be assessed by other immunological based methods including, without limitation, Western blotting, ELISA, and immunofluoresence.
  • FACS fluorescence-associated cell sorting
  • B7-H6-positive cells can be determined by measuring B7-H6 mRNA.
  • Non-limiting methods of quantifying RNA include: qRT-PCR, RNA- sequencing, fluorescence in situ hybridization combined with flow cytometry, microfluidic capillary electrophoresis, and in situ hybridization.
  • Exemplary reference levels of B7-H6 useful for identifying a B7-H6-positive cancer cell or a B7-H6-positive cancer can be a level of B7-H6 in a non-cancerous cell, a median level of B7-H6 in multiple tumor biopsy samples, one standard of deviation higher than a median level of B7-H6 in multiple tumor biopsy samples of the same type of cancer or multiple different cancers, quarter of a standard of deviation higher than a median level of B7-H6 in multiple tumor biopsy samples of the same type of cancer or multiple different cancers, half a standard of deviation higher than a median level of B7-H6 in multiple tumor biopsy samples of the same type of cancer or multiple different cancers, three quarters of a standard deviation higher than a median level of B7-H6 in multiple tumor biopsy samples of the same type of cancer or multiple different cancers, one and half standard deviation higher than a median level of B7-H6 in multiple tumor biopsy samples of the same type of cancer or multiple different cancers, or two standard deviations higher
  • a reference level of B7-H6 is a level of B7-H6 in normal (non-cancerous) tissue of the same type. In some embodiments, a reference level of B7-H6 is a level of B7-H6 in a health tissue that is proximal to a solid tumor. In some embodiments, the reference level is a level that is 50% greater than the level of B7-H6 in the neighboring tissue or a corresponding healthy tissue.
  • a NKG2A polypeptide refers to the amino acid sequence encoded by the killer cell lectin like receptor Cl (KLRC1) gene. NKG2A forms a complex with KLRD1/CD94.
  • KLRC1 polypeptide includes without limitation amino acid sequence corresponding to NCBI reference sequences: NP_001291377.1, NP_002250.2 and NP_998823.1.
  • a NKG2A polypeptide can include a sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • a NKG2A polypeptide is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • a NKG2A polypeptide can include a sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • a NKG2A polypeptide is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the methods include detecting the level of NKG2A in lymphocytes in the subject.
  • Any appropriate sample containing NKp30- positive/NKG2A-negative lymphocytes can be obtained from a subject.
  • a whole blood sample, a plasma sample, a serum sample, a sample containing a cell fraction (PBMC) of a whole blood sample, a tissue sample, a biopsy sample, and a laser capture dissected biological sample e.g., tumor or tissue sample
  • PBMC cell fraction
  • a laser capture dissected biological sample e.g., tumor or tissue sample
  • detecting the NKG2A level in lymphocytes is done prior to, simultaneously with, or after, administering the enucleated erythroid cells.
  • NKG2A-negative lymphocytes can be determined using an antibody that binds specifically to NKG2A or an antigen binding fragment thereof (e.g., Beckman Coulter, clone Z199.1).
  • the level of NKG2A in a lymphocyte can be determined using fluorescence-assisted cell sorting.
  • the level of NKG2A in a lymphocyte can also be assessed by other immunological based methods including, without limitation, Western blotting, ELISA, and immunofluoresence.
  • NKG2A- positive cells can be determined by measuring NKG2A mRNA.
  • Non-limiting methods of quantifying RNA include: qRT-PCR, RNA-sequencing, fluorescence in situ hybridization combined with flow cytometry, microfluidic capillary electrophoresis, and in situ hybridization.
  • a reference level ofNKG2A useful for identifying aNKG2A- negative lymphocyte are known in the art.
  • a reference level can be a level of NKG2A in an activated or proliferating lymphocyte.
  • the methods include modulating the activity of an NKG2A polypeptide.
  • the method further includes administering to the subject an NKG2A inhibitor (e.g., antagonistic antibodies).
  • the method can include administering monalizumab (see, Andre et ak, Cell 175(7): 1731-1743; 2018) or dasatinib (Chang et ak, Front. Immunol. 9:31521; 2019) to the subject, before, simultaneously with, or after administering any of the enucleated erythroid cells described herein.
  • the method further includes administering S095029 to the subject, before, simultaneously with, or after administering any of the enucleated erythroid cells described herein.
  • an HLA-E polypeptide refers to an amino acid sequence encoded by the major histocompatibility complex, class I, E (HLA-E) gene.
  • An exemplary HLA-E polypeptide includes without limitation amino acid sequence corresponding to NCBI reference sequences: NP_005507.3.
  • an HLA-E polypeptide can include an amino acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • a HLA-E polypeptide is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the methods include detecting the level of HLA-E in cancer cells or in a cancer in the subject.
  • Any appropriate sample containing HLA-E cancer cells can be obtained from a subject.
  • whole blood, blood plasma, cell fraction (PBMC) of whole blood, a tissue sample, a biopsy sample, and laser capture microdissection of a tissue sample or biopsy sample can be obtained and assessed to identify the level of HLA-E in cancer cells in a subject.
  • detecting the HLA-E level in cancer cells is done prior to, simultaneously with, or after, administering the enucleated erythroid cells.
  • HLA-E- negative cancer cells can be determined using an antibody that binds specifically to HLA-E or an antigen-binding fragment thereof (e.g., Beckman Coulter, clone Z199.1).
  • the level of HLA-E in a cancer cell can be determined using fluorescence-assisted cell sorting.
  • the level of HLA-E in a cancer cell can also be assessed by other immunological based methods including, without limitation, Western blotting, ELISA, and immunofluoresence.
  • HLA-negative cancer cells can be determined by measuring HLA-E mRNA.
  • Non limiting methods of quantifying RNA include: qRT-PCR, RNA-sequencing, fluorescence in situ hybridization combined with flow cytometry, microfluidic capillary electrophoresis, and in situ hybridization.
  • a HLA-E- negative cancer cell is a cancer cell that has loss of heterozygosity at the HLA-E gene locus.
  • Exemplary reference levels of HLA-E useful for identifying a HLA-E- negative cancer cell can be a level of HLA-E present in a non-cancerous cell or a cancer cell not having a loss of heterozygosity at the HLA-E gene locus. Exemplary levels of HLA-E can be determined using the methods described in Seliger et al., Oncotarget 7(41):67360-67372, 2016).
  • a reference level of B7-H6 is a level of B7-H6 in a health tissue that is proximal to a solid tumor.
  • the reference level is a level that is 50% less than the level of HLA-E in the neighboring tissue or a corresponding healthy tissue.
  • this disclosure features an enucleated erythroid cell that includes a first exogenous polypeptide comprising (i) interleukin- 15 (IL-15) or a functional fragment thereof, and (ii) interleukin- 15 receptor alpha (IL-15RA) or a functional fragment thereof, on its extracellular surface.
  • a first exogenous polypeptide comprising (i) interleukin- 15 (IL-15) or a functional fragment thereof, and (ii) interleukin- 15 receptor alpha (IL-15RA) or a functional fragment thereof, on its extracellular surface.
  • the IL-15 includes the immature form of wild-type human IL-15.
  • the IL-15 includes an amino acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the IL-15 includes a sequence of SEQ ID NO: 15.
  • the IL-15 includes the mature form of wild type human IL-15, or a functional fragment thereof.
  • the IL-15 includes an amino acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the IL-15 includes a sequence of SEQ ID NO: 16.
  • the IL-15 is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the IL-15 is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the functional fragment of IL-15 comprises at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, or 160 amino acids. In some embodiments, the functional fragment of IL-15 comprises fewer than 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, or 160 amino acids.
  • a functional fragment of IL-15 retains at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% of the ability of wild type human IL- 15 polypeptide to bind IL-15RA polypeptide, as measured by assays well known in the art, e.g., ELISA, and surface plasmon resonance (SPR) binding analysis, or co- immunoprecipitation.
  • assays well known in the art, e.g., ELISA, and surface plasmon resonance (SPR) binding analysis, or co- immunoprecipitation.
  • a functional fragment of IL-15 polypeptide retains at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% of the ability of wild type human IL-15 polypeptide to induce IL-15- mediated signal transduction, as measured by assays well-known in the art, e.g., electromobility shift assays, ELISAs, and other immunoassays.
  • a functional fragment of an IL-15 polypeptide can be an IL-15 receptor-binding fragment of the IL-15 polypeptide.
  • the interleukin- 15 receptor alpha (IL-15RA) or a functional fragment thereof can include the immature form of wild-type human IL- 15RA.
  • the IL-15RA can include a sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the IL-15RA polypeptide includes a sequence of SEQ ID NO: 19.
  • the IL-15RA includes the mature form of wild-type human IL-15RA.
  • the IL-15RA includes an amino acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the IL-15RA includes a sequence of SEQ ID NO: 20.
  • the IL-15RA includes an extracellular portion of an IL- 15RA polypeptide.
  • the IL-15RA polypeptide may lack the transmembrane domain of wild type IL-15RA, and optionally, the intracellular domain of wild type IL-15RA.
  • the IL-15RA includes the immature form of an extracellular wild type human IL-15RA.
  • the IL-15RA polypeptide can include a sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to MAPRRARGCRTLGLPALLLLLLLRPPATRGITCPPPMSVEHADIWVKSYSLYS RERYICNSGFKRKAGTSSLTECVLNKATNVAHWTTPSLKCIRDPALVHQRPAP PSTVTTAGVTPQPESLSPSGKEPAASSPSSNNTAATTAAIVPGSQLMPSKSPST GTTEISSHESSHGTPSQTTAKNWELTASASHQPPGVYPQGHSDTT (SEQ ID NO: 21), or a functional fragment thereof.
  • an IL-15RA polypeptide includes a sequence of SEQ ID NO: 21.
  • the IL-15RA includes the mature form of an extracellular wild-type human IL-15RA.
  • the IL-15RA can include a sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • an IL-15RA polypeptide includes a sequence of SEQ ID NO: 22.
  • the IL-15RA is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the IL-15RA or functional fragment thereof includes the “sushi domain” in exon 2 of the extracellular domain of the receptor (Wei et al., J Immunol. 2001; 167:277-282).
  • the IL-15RA or functional fragment thereof that includes the sushi domain of wild type human IL-15RA includes an amino acid sequence that it is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the IL-15RA or functional fragment thereof includes the sushi domain of wild-type human IL-15RA that includes a sequence of SEQ ID NO: 24.
  • the IL-15RA includes the sushi domain of wild type human IL15RA or a functional fragment thereof and at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90 or 100 additional amino acids of wild type human IL-15RA.
  • the IL-15RA or a functional fragment thereof includes the sushi domain of wild type human IL-15RA and 13 additional amino acids of wild type human IL-15RA.
  • the IL-15RA includes an amino acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • an IL-15RA polypeptide includes a sequence of SEQ ID NO: 25.
  • the IL-15RA is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the functional fragment of the IL-15RA comprises at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200 amino acids. In some embodiments, the functional fragment of the IL- 15RA comprises at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200 amino acids, and comprises the IL-15RA sushi domain.
  • the IL-15RA fragments or variants retain at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% of the ability of wild type human IL-15RA to bind an IL-15, as measured by assays well known in the art, e.g., ELISA, surface plasmon resonance (SPR) binding analysis, and co- immunoprecipitation.
  • assays well known in the art, e.g., ELISA, surface plasmon resonance (SPR) binding analysis, and co- immunoprecipitation.
  • IL-15RA variants or fragments retain at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% of the ability of wild type human IL-15RA polypeptide to induce IL-15 -mediated signal transduction, as measured by assays well-known in the art, e.g., electromobility shift assays, ELISAs and other immunoassays.
  • a functional fragment of the IL-15RA polypeptide includes one or both of the sushi domain and the transmembane domain of IL-15RA polypeptide.
  • a functional fragment of the IL-15-RA polypeptide includes the sushi domain.
  • a functional fragment of the IL-15-RA polypeptide includes the transmembrane domain.
  • the first exogenous polypeptide further includes a signal peptide.
  • the first exogenous polypeptide includes a signal peptide that includes an amino acid sequence set forth in Table 3.
  • the first exogenous polypeptide includes a signal peptide that includes a GPA signal peptide.
  • the first exogenous polypeptide comprises a signal peptide having an amino acid sequence of SEQ ID NO: 27.
  • the first exogenous polypeptide includes a leader (signal) sequence that includes the amino acid sequence of SEQ ID NO: 27 an IL-15 polypeptide, and an IL-15RA polypeptide.
  • the first exogenous polypeptide includes a leader (signal) sequence that includes the amino acid sequence of SEQ ID NO: 27, a mature human IL-15 polypeptide that includes the amino acid sequence of SEQ ID NO: 16, and an IL-15RA polypeptide that includes the amino acid sequence of SEQ ID NO: 22.
  • the mature human IL-15 polypeptide and the IL-15RA polypeptide are connected by a flexible linker having an amino acid sequence of SEQ ID NO: 29.
  • one or more linkers are disposed between the IL-15 or the functional fragment thereof, and IL-15RA or the functional fragment thereof. Any of the linkers provided herein may be used.
  • the linker is a peptide that is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more amino acids long.
  • the linker is long enough to preserve the ability of IL-15 to bind to the IL-15RA.
  • the linker is long enough to preserve the ability of the IL-15/IL-15RA complex to bind to the bg IL-15 receptor complex and to act as an agonist to mediate IL-15 signal transduction.
  • the linker includes an amino acid sequence listed in Table 2.
  • the linker comprises the amino acid sequence (GGGGS) n (SEQ ID NO: 30), wherein n is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10. In some embodiments, the linker consists of the (GGGGS) n linker (SEQ ID NO: 30), wherein n is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10. In some embodiments, the linker includes the amino acid sequence GGGGS GGGGS GGGGS (SEQ ID NO: 29).
  • linker can be between 5 and 25 amino acids in length, 5-20 amino acids in length, 10-25 amino acids in length, or 10-20 amino acids in length. In some embodiments, a linker can be 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acids in length. In some embodiments, the linker is non-immunogenic.
  • the first exogenous polypeptide includes an IL-15 polypeptide or a functional fragment thereof and an extracellular region of the IL- 15Ra polypeptide. In some embodiments, the first exogenous polypeptide includes the amino acid sequence of SEQ ID NO: 1. In some embodiments, the first exogenous polypeptide includes as sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • an first exogenous polypeptide is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • GATACTACT SEQ ID NO: 32.
  • the first exogenous polypeptide comprises an IL-15 polypeptide and the sushi domain of the IL-15RA polypeptide.
  • the first exogenous polypeptide includes the amino acid sequence of SEQ ID NO: 2.
  • the exogenous polypeptide includes as sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • an first exogenous polypeptide is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the first exogenous polypeptide includes an IL-15 polypeptide or a functional fragment thereof and an IL-15RA polypeptide or a functional fragment thereof (e.g., an IL-15 binding fragment). Any of the IL-15 polypeptides described herein may be combined with any of the IL-15RA polypeptides described herein to form the first exogenous polypeptide. In some embodiments, the IL-15 polypeptide or a functional fragment thereof and the extracellular portion of an IL-15RA polypeptide or a functional fragment thereof are present as a complex.
  • the IL-15 polypeptide and the extracellular portion of an IL-15RA polypeptide are present as a fusion polypeptide (e.g., a first exogenous fusion polypeptide).
  • the IL-15 polypeptide is linked to the extracellular portion of the IL-15RA polypeptide by a linker.
  • the IL-15 polypeptide and the IL-15RA polypeptide are present as a complex.
  • the components of an IL-15/IL-15RA complex may be directly fused, using either non-covalent bonds or covalent bonds (e.g., by combining amino acid sequences via peptide bonds).
  • the first exogenous polypeptide, the IL-15 polypeptide, IL-15RA polypeptide, or IL-15/IL-15RA complex are not released from the erythroid cell (e.g., the enucleated erythroid cell).
  • the IL-15 polypeptide, IL-15RA polypeptide, or IL-15/IL-15RA complex or fusion polypeptide are attached to the erythroid cell membrane (e.g., the membrane of an enucleated erythroid cell).
  • the first exogenous polypeptide further includes a polypeptide sequence (e.g. a transmembrane region) that anchors the polypeptide to the erythroid cell membrane (referred to herein as an anchor or transmembrane domain).
  • the polypeptide sequence that anchors the first exogenous polypeptide to the erythroid cell membrane is heterologous to another polypeptide in the first exogenous polypeptide.
  • the polypeptide sequence that anchors the polypeptide to the erythroid cell membrane is heterologous to the IL-15 polypeptide and/or the IL-15RA polypeptide.
  • the polypeptide sequence that anchors the polypeptide to the erythroid cell membrane is a GPA sequence.
  • polypeptides useful for anchoring the first exogenous polypeptides to the erythroid cell membrane are known to the skilled person and are contemplated for inclusion in the exogenous polypeptides comprising IL-15, IL-15RA, or IL-15/IL- 15RA fusion.
  • Non-limiting examples include small integral membrane protein 1 (SMIM1), transferrin receptor, Fas ligand (FasL), Kell, and Band 3.
  • the anchor or transmembrane domain can include a type 1 membrane protein or a transmembrane portion thereof.
  • the anchor or transmembrane domain comprises a type 1 membrane protein or a transmembrane portion thereof selected from the group consisting of glycophorin A (GPA); glycophorin B (GPB); basigin (also known as CD147); CD44; CD58 (also known as LFA3); Intercellular Adhesion Molecule 4 (ICAM4); Basal Cell Adhesion Molecule (BCAM); CR1;
  • the anchor or transmembrane domain comprises or consists of a type 2 membrane protein or a transmembrane portion thereof.
  • the anchor or transmembrane domain comprises a type 2 membrane protein or a transmembrane portion thereof selected from the group consisting of small integral membrane protein 1 (SMIM1), transferrin receptor (CD71); Fas ligand (FasL) transmembrane; and Kell.
  • SMIM1 small integral membrane protein 1
  • CD71 transferrin receptor
  • Kell small integral membrane protein 1
  • the anchor is a GPI-linked membrane protein.
  • the GPI-linked membrane protein anchor is selected from the group consisting of CD59; CD55; and Semaphorin 7A (SEMA7A).
  • the anchor or transmembrane domain can include small integral membrane protein 1 (SMIM1) or a transmembrane portion thereof.
  • SMIM1 small integral membrane protein 1
  • the anchor or transmembrane domain includes glycophorin A (GPA), or a fragment thereof (e.g., a transmembrane portion thereof).
  • GPA glycophorin A
  • the anchor or transmembrane domain include an amino acid sequence provided in Table 1.
  • the first exogenous polypeptide includes an IL-15 polypeptide or a functional fragment thereof and a transmembrane region of the wild- type human IL-15RA. In some embodiments, the first exogenous polypeptide includes an IL-15 polypeptide or a functional fragment thereof and a transmembrane region (e.g., any of the exemplary transmembrane regions or transmembrane domains described herein).
  • a linker is disposed between the anchor or transmembrane domain and an IL-15 polypeptide, an IL-15RA polypeptide, or an IL- 15/IL-15RA polypeptide.
  • Suitable linkers include, without limitation, any linker amino acid sequence provided in Table 2.
  • the linker between the anchor or transmembrane domain, e.g., GPA, and an IL-15 polypeptide, an IL- 15RA polypeptide, or an IL-15/IL-15RA fusion polypeptide comprises or consists of an HA linker.
  • the linker comprises or consists of the amino acid sequence of SEQ ID NO: 33.
  • the first exogenous polypeptide can further include an anchor.
  • the first exogenous polypeptide can comprise the amino acid sequence of SEQ ID NO: 36 (which is encoded by nucleic acid sequence SEQ ID NO: 37), an interleukin- 15 (IL-15) polypeptide, and an extracellular portion of an interleukin- 15 receptor alpha (IL-15RA) polypeptide.
  • the first exogenous polypeptide can comprise an anchor that includes the amino acid sequence of SEQ ID NO: 36, mature human IL-15 that includes the amino acid sequence of SEQ ID NO: 16, and mature human extracellular IL-15RA that includes the amino acid sequence of SEQ ID NO: 22 whereby the mature human IL-15 amino acid sequence and the mature human extracellular IL-15 RA amino acid sequence are connected by a flexible linker that includes the amino acid sequence of SEQ ID NO: 29.
  • the exogenous fusion polypeptide comprises: a signal peptide (e.g., a GPA signal peptide) that includes the amino acid sequence of SEQ ID NO: 27, a mature human IL-15 that includes an amino acid sequence of SEQ ID NO: 16, a flexible linker (e.g., connecting the mature human IL-15 and the mature human extracellular IL-15RA) that includes the amino acid sequence of SEQ ID NO: 29, a mature human extracellular IL-15RA that includes the amino acid sequence of SEQ ID NO: 22, a linker that includes the amino acid sequence of SEQ ID NO: 35, and an anchor that includes an amino acid sequence of SEQ ID NO: 36.
  • a signal peptide e.g., a GPA signal peptide
  • a mature human IL-15 that includes an amino acid sequence of SEQ ID NO: 16
  • a flexible linker e.g., connecting the mature human IL-15 and the mature human extracellular IL-15RA
  • a linker that includes the amino acid sequence of SEQ
  • the exogenous fusion polypeptide comprises (e.g., from N-terminus to C-terminus): a mature human IL-15 that includes an amino acid sequence of SEQ ID NO: 16, a flexible linker (e.g., connecting the mature human IL-15 and the mature human extracellular IL-15RA) that includes the amino acid sequence of SEQ ID NO: 29, a mature human extracellular IL-15RA that includes the amino acid sequence of SEQ ID NO: 22, a linker that includes the amino acid sequence of SEQ ID NO: 35, and an anchor that includes an amino acid sequence of SEQ ID NO: 36.
  • the exogenous fusion polypeptide includes a sequence of SEQ ID NO: 46.
  • an first exogenous polypeptide includes amino acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • an first exogenous polypeptide is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • CAAGTGATCAA SEQ ID NO: 39.
  • an first exogenous polypeptide includes amino acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • an first exogenous polypeptide is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • an first exogenous polypeptide includes amino acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • an first exogenous polypeptide is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the first exogenous polypeptide includes amino acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • DVPLSSVEIENPETSDQ (SEQ ID NO: 44), or a functional fragment thereof.
  • an first exogenous polypeptide is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the first exogenous polypeptide includes amino acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • an first exogenous polypeptide is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the exogenous fusion polypeptide includes an amino acid sequence of SEQ ID NO: 38. In some embodiments, the exogenous fusion polypeptides includes an amino acid sequence of SEQ ID NO: 40. In some embodiments, the exogenous fusion polypeptide includes an amino acid sequence of SEQ ID NO: 42. In some embodiments, the exogenous fusion polypeptide includes an amino acid sequence of SEQ ID NO: 44. In some embodiments, the exogenous fusion polypeptide includes an amino acid sequence of SEQ ID NO: 46.
  • the first exogenous polypeptide is as described in US Patent Application Publication No. 2019/0298769, which is herein incorporated by reference in its entirety.
  • an enucleated erythroid cell can further include an exogenous polypeptide on its extracellular surface.
  • an enucleated erythroid cell includes a first exogenous polypeptide including (i) interleukin- 15 (IL-15) or a functional fragment thereof, and (ii) interleukin- 15 receptor alpha (IL-15RA) or a functional fragment thereof, on its extracellular surface, and a second exogenous polypeptide that includes a 4-1BBL polypeptide or a functional fragment thereof, on its extracellular surface.
  • IL-15 interleukin- 15
  • IL-15RA interleukin- 15 receptor alpha
  • 4-1BBL polypeptide refers the amino acid sequence encoded by the Tumor Necrosis Factor superfamily member 9 (TNFSF9 or CD137L) gene.
  • 4- 1BBL is the ligand for 4-1BB (also known as Tumor Necrosis Factor Receptor Superfamily, Member 9 (TNFRSF9), or CD137), a member of a family of receptors found on the surfaces of cells of the immune system. See Alderson et ak, 1994, Eur.
  • the 4-1BBL is in its natural trimeric form.
  • a 4-1BBL includes an amino acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the 4- 1BBL includes a sequence of SEQ ID NO: 48.
  • the 4-1BBL is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the second exogenous polypeptide includes an amino acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • a 4-1BBL polypeptide includes a sequence of SEQ ID NO: 50.
  • an exogenous fusion polypeptide is encoded by a nucleic acid sequence that is at least 70% identical (e.g., at least 75% identical, at least 80% identical, at least 85% identical, at least 90% identical, at least 95% identical, at least 99% identical, or 100% identical) to
  • the exogenous polypeptide includes a leader (signal) sequence.
  • the 4-1BBL or functional fragment thereof is fused to the leader (signal) sequence.
  • Non-limiting examples of a leader (signal) sequence include the amino acid sequences provided in Table 3.
  • the leader (signal) sequence includes a GPA signal peptide.
  • the leader (signal) sequence includes the amino acid sequence of SEQ ID NO: 27.
  • the exogenous polypeptide comprises a 4-1BBL or a functional fragment thereof and a leader (signal) sequence of the amino acid sequence of SEQ ID NO: 27.
  • the exogenous polypeptide comprises a leader (signal) sequence that includes the amino acid sequence of SEQ ID NO: 27, and a 4- 1BBL having an amino acid sequence of SEQ ID NO: 48.
  • the second exogenous polypeptide is attached to the erythroid cell membrane (e.g., the membrane of an enucleated erythroid cell). In some embodiments, the second exogenous polypeptide is attached to the extracellular surface of the enucleated erythroid cell. In some embodiments, the second exogenous polypeptide further comprises an anchor or transmembrane domain that anchors the second exogenous polypeptide to the erythroid cell membrane (e.g., the membrane of an enucleated erythroid cell). In some embodiments, the anchor or transmembrane domain is heterologous to the second exogenous polypeptide (e.g., 4-1BBL). In some embodiments, the anchor or transmembrane domain includes an endogenous red blood cell transmembrane protein, or a fragment or transmembrane portion thereof.
  • the anchor or transmembrane domain includes GPA or a transmembrane portion thereof. In some embodiments, the anchor or transmembrane domain includes small integral membrane protein 1 (SMIM1), transferrin receptor,
  • the second exogenous polypeptide may comprise any of the anchor or transmembrane domains described herein. In some embodiments, the second exogenous polypeptide comprises an anchor or transmembrane domain set forth in Table 1.
  • the second exogenous polypeptide includes one or more linkers (e.g., any of the exemplary linkers described herein).
  • the second exogenous polypeptide can includes one or more linkers provided in Table 2.
  • a linker is disposed between the 4-1BBL or a functional fragment thereof and an anchor or transmembrane domain in the second exogenous polypeptide.
  • the second exogenous polypeptide can further include a signal peptide (e.g., any of the exemplary signal peptides described herein).
  • the second exogenous polypeptide can include a signal peptide provided in Table 3.
  • the exogenous polypeptide includes a signal peptide, the 4-1BBL or a functional fragment thereof, and an anchor. In some embodiments, the exogenous polypeptide includes a signal peptide, the 4-1BBL or functional fragment thereof, a linker, and an anchor.
  • the exogenous polypeptide comprises (e.g., from N-terminus to C-terminus) a signal peptide that includes the amino acid sequence of SEQ ID NO: 27, the 4-1BBL or a functional fragment thereof that includes the amino acid sequence of SEQ ID NO: 48, a linker that includes the amino acid sequence of SEQ ID NO: 52 (e.g., disposed between the 4-1BBL or the functional fragment thereof and the anchor), and an anchor that includes the amino acid sequence of SEQ ID NO: 36.
  • the exogenous polypeptide comprises the 4-1BBL or a functional fragment thereof, a linker, and an anchor.
  • the exogenous polypeptide comprises (e.g., from N-terminus to C-terminus) the 4-1BBL or functional fragment thereof that includes the amino acid sequence of SEQ ID NO: 48, a linker that includes the amino acid sequence of SEQ ID NO: 52 (e.g., disposed between the 4-1BBL or the functional fragment thereof and the anchor), and an anchor that includes the amino acid sequence of SEQ ID NO: 36.
  • the exogenous polypeptide comprises or consists of the amino acid sequence of SEQ ID NO: 50.
  • the first and/or second exogenous polypeptides may have post-translational modifications characteristic of eukaryotic cells, e.g., mammalian cells, e.g., human cells.
  • one or more (e.g., 2, 3, 4, 5, or more) of the exogenous proteins are glycosylated, phosphorylated, or both.
  • PPS Periodic acid-Schiff
  • Post-translation modifications also include conjugation to a hydrophobic group (e.g., myristoylation, palmitoylation, isoprenylation, prenylation, or glypiation), conjugation to a cofactor (e.g., lipoylation, flavin moiety (e.g., FMN or FAD), heme C attachment, phosphopantetheinylation, or retinylidene Schiff base formation), diphthamide formation, ethanolamine phosphoglycerol attachment, hypusine formation, acylation (e.g.
  • O-acylation, N-acylation, or S-acylation formylation, acetylation, alkylation (e.g., methylation or ethylation), amidation, butyrylation, gamma-carboxylation, malonylation, hydroxylation, iodination, nucleotide addition such as ADP-ribosylation, oxidation, phosphate ester (O-linked) or phosphoramidate (N-linked) formation, (e.g., phosphorylation or adenylylation), propionylation, pyroglutamate formation, S-glutathionylation, S-nitrosylation, succinylation, sulfation, ISGylation, SUMOylation, ubiquitination, Neddylation, or a chemical modification of an amino acid (e.g., citrullination, deamidation, eliminylation, or carbamylation), formation of a disulfide bridge, racemization (e.g.
  • glycosylation includes the addition of a glycosyl group to arginine, asparagine, cysteine, hydroxylysine, serine, threonine, tyrosine, or tryptophan, resulting in a glycoprotein.
  • the glycosylation comprises, e.g., O-linked glycosylation or N-linked glycosylation.
  • the first and second exogenous polypeptides are fusion proteins, e.g., is a fusion with an endogenous red blood cell protein or fragment thereof, e.g., a transmembrane protein, e.g., GPA or a transmembrane fragment thereof.
  • one or more of the exogenous polypeptides is fused with a domain that promotes dimerization or multimerization, e.g., with a second fusion exogenous polypeptide, which optionally comprises a dimerization domain.
  • the dimerization domain comprises a portion of an antibody molecule, e.g., an Fc domain or CH3 domain.
  • the first and second dimerization domains comprise knob-in-hole mutations (e.g., a T366Y knob and a Y407T hole) to promote heterodimerization.
  • the transmembrane domain comprises or consists of a transmembrane domain of a type 1 membrane protein.
  • the type 1 membrane protein is selected from the group consisting of glycophorin A (GPA); glycophorin B (GPB); basigin (also known as CD147); CD44; CD58 (also known as LFA3); Intercellular Adhesion Molecule 4 (ICAM4); Basal Cell Adhesion Molecule (BCAM); CR1; CD99; Erythroblast Membrane Associated Protein (ERMAP); junctional adhesion molecule A (JAM- A); neuroplastin (NPTN); AMIG02; and DS Cell Adhesion Molecule Like 1 (DSCAML1).
  • GPA glycophorin A
  • GPB glycophorin B
  • basigin also known as CD147
  • CD44 also known as CD58
  • CD58 also known as LFA3
  • ICM4 Intercellular Adhesion Molecule 4
  • BCAM Basal Cell Adhesion Molecule
  • CR1
  • the transmembrane domain comprises or consists of a transmembrane domain of a type 2 membrane protein.
  • the type 2 membrane protein is selected from the group consisting of small integral membrane protein 1 (SMIM1), transferrin receptor (CD71); Fas ligand (FasL) transmembrane; and Kell.
  • the polypeptide sequence that anchors the exogenous polypeptide to the enucleated erythroid cell membrane comprises, consists of, or is derived from (e.g., a fragment of) a GPI-linked membrane protein.
  • the GPI-linked membrane protein is selected from the group consisting of CD59; CD55; and Semaphorin 7A (SEMA7A).
  • the transmembrane domain comprises GPA or a transmembrane portion thereof.
  • GPA is preferred because it has a cytoplasmic domain that interacts with the reticulocyte cytoskeleton that has a role in retaining the GPA as the cell differentiates and matures.
  • the transmembrane domain comprises small integral membrane protein 1 (SMIM1) or a transmembrane portion thereof.
  • the anchor is selected from an amino acid sequence listed in Table 1.
  • the enucleated erythroid cells include a first and second exogenous polyepptide
  • the first exogenous polypeptide e.g., any of the exogenous fusion polypeptides described herein
  • the second exogenous polypeptide e.g., any of the exogenous polypeptides described herein
  • the exogenous fusion polypeptide or one or more of the exogenous polypeptides is fused with a domain that promotes dimerization or multimerization, e.g., with a second exogenous polypeptide, which optionally comprises a dimerization domain.
  • the dimerization domain comprises a portion of an antibody molecule, e.g., an Fc domain or CH3 domain.
  • the first and second dimerization domains comprise knob-in-hole mutations (e.g. , a T366Y knob and a Y407T hole) to promote heterodimerization.
  • the first exogenous fusion polypeptide e.g., any of the fusion polypeptides described herein
  • the second exogenous polypeptide e.g., any of the exemplary polypeptides described herein
  • a linker may be disposed between a cytokine polypeptide sequence (e.g., IL-15 or a functional fragment thereof) and a transmembrane domain sequence, or between IL-15 or a functional fragment thereof, and IL-15RA or a functional fragment thereof).
  • a linker may be disposed between a 4-1BBL polypeptide, of a functional fragment thereof, and a transmembrane domain sequence.
  • the linker comprises or consists of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more amino acids in length. In some embodiments, the linker comprises or consists of between about 5 and about 25 amino acids in length, between about 5 and about 20 amino acids in length, between about 10 and about 25 amino acids in length, or between about 10 and about 20 amino acids in length. In some embodiments, the linker useful in the disclosure comprises or consists of 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acids in length. In a preferred embodiment, the linker is non-immunogenic.
  • the linker is selected from an amino acid sequence presented in Table 2.
  • the linker comprises the amino acid sequence (GGGGS) n (SEQ ID NO: 30), wherein n is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10. In some embodiments, the linker consists of the (GGGGS) n linker (SEQ ID NO: 30), wherein n is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10. In some embodiments, the linker comprises the amino acid sequence GGGGS GGGGS GGGGS (SEQ ID NO: 29). In some embodiments, the linker consists of the amino acid sequence of SEQ ID NO: 29. In some embodiments, the linker comprises the amino acid sequence SEQ ID NO: 57.
  • the linker consists of the amino acid sequence of SEQ ID NO: 57. In some embodiments, the linker comprises the amino acid sequence SEQ ID NO: 57.
  • the linker consists of the amino acid sequence of SEQ ID NO: 35. In some embodiments, the linker comprises the amino acid sequence SEQ ID NO: 52. In some embodiments, the linker consists of the amino acid sequence of SEQ ID NO: 52.
  • Other suitable linkers that can be used in the exogenous fusion polypeptide or the exogenous polypeptide (e.g., any of the exogenous polypeptides described herein) are known in the art.
  • the first exogenous fusion polypeptide or the exogenous polypeptide comprises a leader (signal) sequence.
  • the leader sequence is selected from the sequences set forth in Table 3.
  • the enucleated erythroid cell comprises a combination of: a first exogenous fusion polypeptide comprising IL-15, or a fragment thereof, linked to an extracellular portion of IL-15 receptor alpha (IL-15Ra), or a fragment thereof (e.g., an IL-15Ra sushi-binding domain), linked to a transmembrane protein (e.g., GPA, or a transmembrane fragment thereof), and a second exogenous polypeptide comprising 4-1BBL, or a fragment thereof, linked to a transmembrane protein (e.g., GPA, or a transmembrane fragment thereof); e.g., as described in U.S. Patent Application Publication No.
  • the enucleated erythroid cells are negative for (i.e. , do not include) one or more minor blood group antigens, e.g., Le(a b ) (for Lewis antigen system), Fy(ab ) (for Duffy system), Jk(ab ) (for Kidd system), MN (for MNS system), Kk (for Kell system), Lu(ab ) (for Lutheran system), and H-antigen negative (Bombay phenotype), or any combination thereof.
  • minor blood group antigens e.g., Le(a b ) (for Lewis antigen system), Fy(ab ) (for Duffy system), Jk(ab ) (for Kidd system), MN (for MNS system), Kk (for Kell system), Lu(ab ) (for Lutheran system), and H-antigen negative (Bombay phenotype), or any combination thereof.
  • the enucleated erythroid cells are also Type O and/or Rh .
  • Minor blood groups are described, e.g., in Agarwal et al., “Blood group phenotype frequencies in blood donors from a tertiary care hospital in north India,” Blood Res. 48(l):51-54, 2013, and Mitra et al., “Blood groups systems,” Indian J. Anaesth. 58(5):524-528, 2014, the description of which is incorporated herein by reference.
  • the enucleated erythroid cells e.g., human enucleated erythroid cells
  • the population of enucleated erythroid cells has an osmotic fragility of less than 50% cell lysis at 0.3%, 0.35%, 0.4%, 0.45%, or 0.5% NaCl. Osmotic fragility is determined, in some embodiments, using the method described in Example 59 of WO 2015/073587 (the description of which is incorporated herein by reference).
  • the enucleated erythroid cells (e.g., human enucleated erythroid cells) have approximately the same diameter or volume as a wild-type, untreated enucleated erythroid cell.
  • the population of enucleated erythroid cells (e.g., human enucleated erythroid cells) have an average diameter of about 4, 5, 6, 7, 8, 9, 10, 11 or 12 microns, or about 4.0 to about 12.0 microns, about 4.0 to about 11.5 microns, about 4.0 to about 11.0 microns, about 4.0 to about 10.5 microns, about 4.0 to about 10 microns, about 4.0 to about 9.5 microns, about 4.0 to about 9.0 microns, about 4.0 to about 8.5 microns, about 4.0 to about 8.0 microns, about 4.0 to about 7.5 microns, about 4.0 to about 7.0 microns, about 4.0 to about 6.5 microns, about 4.0 to about
  • the volume of the mean corpuscular volume of the enucleated erythroid cell is about 10 fL to about 175 fL, about 10 fL to about 160 fL, about 10 fL to about 140 fL, about 10 fL to about 120 fL, about 10 fL to about 100 fL, about 10 fL to about 90 fL, about 10 fL to about 80 fL, about 10 fL to about 70 fL, about 10 fL to about 60 fL, about 10 fL to about 50 fL, about 10 fL to about 40 fL, about 10 fL to about 30 fL, about 10 fL to about 20 fL, about 20 fL to about 175 fL, about 20 fL to about 160 fL, about 20 fL to about 140 fL, about 20 fL to about 120 fL, about 20 fL to about 100 fL, about 20 fL to about 90 fL,
  • the mean corpuscular volume can be measured, e.g., using a hematological analysis instrument, e.g., a Coulter counter, a Moxi Z cell counter (Orflo), or a Sysmex Hematology analyzer.
  • a hematological analysis instrument e.g., a Coulter counter, a Moxi Z cell counter (Orflo), or a Sysmex Hematology analyzer.
  • the enucleated erythroid cells described herein have one or more (e.g., 2, 3, 4, or more) physical characteristics described herein, e.g., osmotic fragility, cell size, hemoglobin concentration, or phosphatidylserine content. While not wishing to be bound by theory, in some embodiments an enucleated erythroid cell that expresses an exogenous protein has physical characteristics that resemble a wild-type, untreated enucleated erythroid cell.
  • a hypotonically loaded enucleated erythroid cell sometimes displays aberrant physical characteristics such as increased osmotic fragility, altered cell size, reduced hemoglobin concentration, or increased phosphatidylserine levels on the outer leaflet of the cell membrane.
  • the enucleated erythroid cell comprises an exogenous protein that was encoded by an exogenous nucleic acid that was not retained by the cell, has not been purified, or has not existed fully outside an enucleated erythroid cell. In some embodiments, the enucleated erythroid cell is in a composition that lacks a stabilizer.
  • the enucleated erythroid cells has a hemoglobin content similar to a wild-type, untreated enucleated erythroid cell. In some embodiments, the enucleated erythroid cells comprise greater than 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9% or greater than 10% fetal hemoglobin. In some embodiments, the enucleated erythroid cells comprise at least about 20, 22, 24, 26, 28, or 30 pg, and optionally up to about 30 pg, of total hemoglobin. Hemoglobin levels are determined, in some embodiments, using the Drabkin’s reagent method of Example 33 of WO2015/073587.
  • the enucleated enucleated erythroid cells has approximately the same phosphatidylserine content on the outer leaflet of its cell membrane as a wild-type, untreated enucleated erythroid cell.
  • Phosphatidylserine is predominantly on the inner leaflet of the cell membrane of wild-type, untreated enucleated erythroid cells, and hypotonic loading can cause the phosphatidylserine to distribute to the outer leaflet where it can trigger an immune response.
  • the population of enucleated erythroid cells comprises less than about 30, 25, 20, 15, 10, 9, 8, 6, 5, 4, 3, 2, or 1% of cells that are positive for Annexin V staining.
  • Phosphatidylserine exposure is assessed, in some embodiments, by staining for Annexin-V-FITC, which binds preferentially to PS, and measuring FITC fluorescence by flow cytometry, e.g., using the method of Example 54 of WO2015/073587.
  • the population of enucleated erythroid cells comprises at least about 50%, 60%, 70%, 80%, 90%, or 95% (and optionally up to 90 or 100%) of cells that are positive for GPA.
  • the presence of GPA is detected, in some embodiments, using FACS.
  • the enucleated erythroid cells have a half-life of at least 30, 45, or 90 days in a subject.
  • a population of cells comprising enucleated erythroid cells comprises less than about 10, 5, 4, 3, 2, or 1% echinocytes.
  • a composition including a plurality of enucleated erythroid cells can be administered to a subject (e.g., any of the subjects described herein. In such embodiments, greater than 50%, greater than 60%, greater than 70%, greater than 80%, or greater than 90% of the cells in the composition can be enucleated.
  • a cell e.g., an enucleated erythroid cell, contains a nucleus that is non-functional, e.g., has been inactivated.
  • the enucleated erythroid cells are human (e.g., derived from a human donor erythroid cell precursor) enucleated erythroid cells.
  • the enucleated erythroid cells are engineered human enucleated erythroid cells.
  • the engineered enucleated erythroid cells comprise a single exogenous protein.
  • the engineered enucleated erythroid cells comprise two or more exogenous proteins (e.g., any of the exemplary exogenous proteins described herein).
  • an exogenous protein present on the membrane of the engineered enucleated erythroid cell can be a product of a click chemistry reaction (e.g., the exogenous protein may be conjugated to a protein present on the membrane of the cell (e.g., a second exogenous protein or an endogenous protein) using any of the methods described herein).
  • an exogenous protein present on the membrane of the engineered enucleated erythroid cell can the a product of a conjugation reaction using a sortase enzyme (e.g., the exogenous protein may be conjugated to a protein present on the membrane of the cell (e.g., a second exogenous protein or an endogenous protein) using any of the methods described herein).
  • a conjugation reaction using a sortase enzyme can be found in U.S. Pat. No. 10,260,038 and U.S. Pat. Pub. No. 2016/0082046 Al.
  • an exogenous protein present on the membrane of the engineered enucleated erythroid cell can be a lipid-anchored protein, e.g., a GPI-anchor, an N- myristolyated protein, or a S-palmitoylated protein.
  • an exogenous protein present on the membrane of the engineered enucleated erythroid cell can be a transmembrane protein (e.g., a single-pass or multi-pass transmembrane protein) or a peripheral membrane protein.
  • an exogenous protein present on the membrane of the engineered enucleated erythroid cell can be a fusion protein comprising a transmembrane domain (e.g., a fusion protein comprising the transmembrane domain of small integral membrane protein 1 (SMIM1) or glycophorin A (GPA)).
  • SMIM1 small integral membrane protein 1
  • GPA glycophorin A
  • an exogenous protein present on the membrane of the engineered enucleated erythroid cell does not have any amino acids protruding into the extracellular space.
  • an exogenous protein present on the membrane of the engineered enucleated erythroid cell does not have any amino acids protruding into the cytosol of the engineered enucleated erythroid cell.
  • an exogenous protein present on the membrane of the engineered enucleated erythroid cell has amino acids protruding into the extracellular space and amino acids protruding into the cytosol of the engineered enucleated erythroid cells.
  • Exemplary methods of producing enucleated erythroid cells using sortagging are described in W02014/183071 or WO2014/183066, each of which is incorporated by reference in its entirety.
  • the engineered enucleated erythroid cells can be produced by introducing one or more nucleic acids (e.g., DNA expression vectors or mRNA) encoding one or more exogenous proteins (e.g., any of the exogenous proteins described herein or known in the art) into an erythroid cell precursor (e.g., any of the erythroid cell precursors described herein or known in the art).
  • nucleic acids e.g., DNA expression vectors or mRNA
  • exogenous proteins e.g., any of the exogenous proteins described herein or known in the art
  • Exemplary methods for introducing DNA expression vectors into erthyroid cell precursor include, but are not limited to, liposome-mediated transfer, transformation, gene guns, transfection, and transduction, e.g., viral -mediated gene transfer (e.g., performed using viral vectors including adenovirus vectors, adeno-associated viral vectors, lentiviral vectors, herpes viral vectors, and retroviral-based vectors).
  • Additional exemplary methods for introducing DNA expression vectors into erythroid cell precursor include the use of, e.g., naked DNA, CaPCri precipitation, DEAE dextran, electroporation, protoplast fusion, lipofection, and cell microinjection.
  • An erythroid cell precursor can optionally be cultured, e.g., before and/or after introduction of one or more nucleic acids encoding one or more exogenous proteins, under suitable conditions allowing for differentiation into engineered enucleated erythroid cells.
  • the resulting engineered enucleated erythroid cells comprise proteins associated with mature erythrocytes, e.g., hemoglobin (e.g., adult hemoglobin and/or fetal hemoglobin), glycophorin A, and exogenous proteins which can be validated and quantified by standard methods (e.g. Western blotting or FACS analysis).
  • the two or more exogenous polypeptides are encoded in a single nucleic acid, e.g. a single vector.
  • the single vector has a separate promoter for each gene, has two proteins that are initially transcribed into a single polypeptide having a protease cleavage site in the middle, so that subsequent proteolytic processing yields two exogenous proteins, or any other suitable configuration.
  • the two or more polypeptides are encoded in two or more nucleic acids, e.g., each vector encodes one of the exogenous polypeptides.
  • the nucleic acid may be, e.g., DNA or RNA.
  • viruses may be used as gene transfer vehicles including retroviruses, Moloney murine leukemia virus (MMLV), adenovirus, adeno-associated virus (AAV), herpes simplex virus (HSV), lentiviruses such as human immunodeficiency virus 1 (HIV 1), and spumaviruses such as foamy viruses, for example.
  • the enucleated erythroid cells are expanded at least 1000, 2000, 5000, 10,000, 20,000, 50,000, or 100,000 fold (and optionally up to 200,000 or 500,000 fold). Number of cells is measured, in some embodiments, using an automated cell counter.
  • enucleated erythroid cells or erythroid cell precursors can be transfected with mRNA encoding an exogenous protein to generate engineered enucleated erythroid cells.
  • Messenger RNA can be derived from in vitro transcription of a cDNA plasmid construct containing a sequence encoding an exogenous protein.
  • the cDNA sequence encoding an exogenous protein may be inserted into a cloning vector containing a promoter sequence compatible with specific RNA polymerases.
  • the cloning vector ZAP Express® pBK-CMV contains T3 and T7 promoter sequences compatible with the T3 and T7 RNA polymerases, respectively.
  • the plasmid is linearized at a restriction site downstream of the stop codon(s) corresponding to the end of the sequence encoding the exogenous protein.
  • the mRNA is transcribed from the linear DNA template using a commercially available kit such as, for example, the RNAMaxx® High Yield Transcription Kit (from Stratagene, La Jolla, Calif., USA).
  • transcription of a linearized cDNA template may be carried out using, for example, the mMESSAGE mMACHINE High Yield Capped RNA Transcription Kit from Ambion (Austin, Tex., USA). Transcription may be carried out in a reaction volume of 20-100 pL at 37 °C for 30 min to 4 h.
  • the transcribed mRNA is purified from the reaction mix by a brief treatment with DNase I to eliminate the linearized DNA template followed by precipitation in 70% ethanol in the presence of lithium chloride, sodium acetate, or ammonium acetate.
  • the integrity of the transcribed mRNA may be assessed using electrophoresis with an agarose- formaldehyde gel or commercially available Novex pre-cast TBE gels (Novex, Invitrogen, Carlsbad, Calif., USA).
  • Messenger RNA encoding an exogenous protein may be introduced into enucleated erythroid cells or erythroid cell precursors using a variety of approaches including, for example, lipofection and electroporation (van Tandeloo et al., Blood 98:49-56, 2001).
  • lipofection for example, 5 pg of in vitro transcribed mRNA in Opti-MEM (Invitrogen, Carlsbad, Calif., USA) is incubated for 5-15 min at a 1:4 ratio with the cationic lipid DMRIE-C (Invitrogen).
  • lipids or cationic polymers may be used to transfect erythroid cell precursors with mRNA including, for example, DOTAP, various forms of polyethylenimine, and polyL-lysine (Sigma-Aldrich, Saint Louis, Mo., USA), and Superfect (Qiagen, Inc., Valencia, Calif., USA; See, e.g., Bettinger et al., Nucleic Acids Res. 29:3882-3891, 2001).
  • the resulting mRNA/lipid complexes are incubated with cells (1 - 2 x 10 6 cells/mL) for 2 hours at 37° C, washed, and returned to culture.
  • electroporation for example, about 5 to 20 x 10 6 cells in 500 pL of Opti-MEM (Invitrogen, Carlsbad, Calif., USA) are mixed with about 20 pg of in vitro transcribed mRNA and electroporated in a 0.4-cm cuvette using, for example, an Easyject Plus device (EquiBio, Kent, United Kingdom).
  • Opti-MEM Invitrogen, Carlsbad, Calif., USA
  • the electroporation parameters required to efficiently transfect cells with mRNA appear to be less detrimental to cells than those required for electroporation of DNA (van Tandeloo et al., Blood 98:49-56, 2001).
  • mRNA may be transfected into enucleated erythroid cells or erythroid cell precursors using a peptide-mediated RNA delivery strategy (See, e.g., Bettinger et al., Nucleic Acids Res. 29:3882-3891, 2001).
  • a peptide-mediated RNA delivery strategy See, e.g., Bettinger et al., Nucleic Acids Res. 29:3882-3891, 2001.
  • the cationic lipid polyethylenimine 2 kDA Sigma-Aldrich, Saint Louis, Mo., USA
  • the melittin peptide Alta Biosciences, Birmingham, UK
  • the mellitin peptide may be conjugated to the PEI using a disulfide cross-linker such as, for example, the hetero-bifunctional cross-linker succinimidyl 3-(2-pyridyldithio) propionate.
  • a disulfide cross-linker such as, for example, the hetero-bifunctional cross-linker succinimidyl 3-(2-pyridyldithio) propionate.
  • RNA/peptide/lipid complex In vitro transcribed mRNA is preincubated for 5 to 15 min with the mellitin-PEI to form an RNA/peptide/lipid complex. This complex is then added to cells in serum-free culture medium for 2 to 4 h at 37 °C in a 5% CO2 humidified environment, then removed, and the transfected cells further cultured.
  • the engineered enucleated erythroid cells are generated by introducing a nucleic acid (e.g., any of the exemplary nucleic acids described herein) encoding one or more exogenous protein(s) (e.g., any exogenous protein or any combination of exogenous proteins described herein) into an erythroid cell precursor.
  • a nucleic acid e.g., any of the exemplary nucleic acids described herein
  • exogenous protein(s) e.g., any exogenous protein or any combination of exogenous proteins described herein
  • the exogenous protein is encoded by a DNA, which is introduced into an erythroid cell precursor.
  • the exogenous protein is encoded by an RNA, which is introduced into an erythroid cell precursor.
  • Nucleic acid encoding one or more exogenous protein(s) may be introduced into an erythroid cell precursor prior to terminal differentiation into an enucleated erythroid cell using a variety of DNA techniques, including, e.g., transient or stable transfections and gene therapy approaches.
  • Viral gene transfer may be used to transfect the cells with a nucleic acid encoding one or more exogenous protein(s).
  • viruses may be used as gene transfer vehicles including Moloney murine leukemia virus (MMLV), adenovirus, adeno-associated virus (AAV), herpes simplex virus (HSV), lentiviruses such as human immunodeficiency virus 1 (HIV1), and spumaviruses such as foamy viruses (see, e.g., Osten et al., HEP 178:177-202, 2007).
  • Retroviruses for example, efficiently transduce mammalian cells including human cells and integrate into chromosomes, conferring stable gene transfer.
  • a nucleic acid encoding one or more exogenous protein(s) can be transfected into an erythroid cell precursor.
  • a suitable vector is the Moloney murine leukemia virus (MMLV) vector (Malik et al., Blood 91:2664-2671, 1998). Vectors based on MMLV, an oncogenic retrovirus, are currently used in gene therapy clinical trials (Hassle et al., News Physiol. Sci. 17:87-92, 2002).
  • MMLV Moloney murine leukemia virus
  • a DNA construct containing the cDNA encoding an exogenous protein can be generated in the MMLV vector backbone using standard molecular biology techniques.
  • the construct is transfected into a packaging cell line such as, for example, PA317 cells and the viral supernatant is used to transfect producer cells such as, for example, PG13 cells.
  • the PG13 viral supernatant is incubated with an erythroid cell precursor.
  • the expression of the exogenous protein may be monitored using FACS analysis (fluorescence- activated cell sorting), for example, with a fluorescently labeled antibody directed against the exogenous protein, if it is present on the membrane of the engineered human enucleated erythroid cell. Similar methods may be used such that an exogenous protein is present in the cytosol of an engineered human enucleated erythroid cell.
  • a nucleic acid encoding a fluorescent tracking molecule such as, for example, green fluorescent protein (GFP)
  • GFP green fluorescent protein
  • Ecotopic retroviral vectors containing DNA encoding the enhanced green fluorescent protein (EGFP) or a red fluorescent protein (e.g., DsRed-Express) are packaged using a packaging cell such as, for example, the Phoenix-Eco cell line (distributed by Orbigen, San Diego, Calif.).
  • Packaging cell lines stably express viral proteins needed for proper viral packaging including, for example, gag, pol, and env.
  • Supernatants from the Phoenix-Eco cells into which viral particles have been shed are used to transduce erythroid cell precursors.
  • transduction may be performed on a specially coated surface such as, for example, fragments of recombinant fibronectin to improve the efficiency of retroviral mediated gene transfer (e.g., RetroNectin, Takara Bio USA, Madison, Wis.).
  • RetroNectin recombinant fibronectin
  • Cells are incubated in RetroNectin-coated plates with retroviral Phoenix-Eco supernatants plus suitable co factors. Transduction may be repeated the next day. In this instance, the percentage of erythroid precursor cells expressing EGFP or DsRed-Express may be assessed by FACS.
  • reporter genes that may be used to assess transduction efficiency include, for example, beta-galactosidase, chloramphenicol acetyltransferase, and luciferase, as well as low-affinity nerve growth factor receptor (LNGFR), and the human cell surface CD24 antigen (Bierhuizen et al., Leukemia 13:605-613, 1999).
  • LNGFR low-affinity nerve growth factor receptor
  • Nonviral vectors may be used to introduce a nucleic acid encoding one or more exogenous protein(s) into an erythroid precursor cell to generate engineered enucleated erythroid cells.
  • a number of delivery methods can be used to introduce nonviral vectors into erythroid cell precursors including chemical and physical methods.
  • a nonviral vector encoding an exogenous protein may be introduced into an erythroid cell precursor using synthetic macromolecules, such as cationic lipids and polymers (Papapetrou et al., Gene Therapy 12:S118-S130, 2005).
  • Cationic liposomes for example form complexes with DNA through charge interactions.
  • the positively charged DNA/lipid complexes bind to the negative cell surface and are taken up by the cell by endocytosis. This approach may be used, for example, to transfect hematopoietic cells (see, e.g., Keller et al., Gene Therapy 6:931-938, 1999).
  • the plasmid DNA in a serum-free medium, such as, for example, OptiMEM (Invitrogen, Carlsbad, Calif.)
  • a cationic liposome in serum free medium
  • LipofectamineTM the commercially available transfection reagent LipofectamineTM
  • the DNA/liposome complex is added to erythroid cell precursors and allowed to incubate for 5-24 h, after which time transgene expression of the exogenous protein(s) may be assayed.
  • other commercially available liposome transfection agents may be used (e.g., In vivo GeneSHUTTLETM, Qbiogene, Carlsbad, Calif.).
  • a cationic polymer such as, for example, polyethylenimine (PEI) may be used to efficiently transfect erythroid cell precursors, for example hematopoietic and umbilical cord blood-derived CD34 + cells (see, e.g., Shin et al., Biochim. Biophys. Acta 1725:377-384, 2005).
  • PEI polyethylenimine
  • Human CD34 + cells are isolated from human umbilical cord blood and cultured in Iscove's modified Dulbecco's medium supplemented with 200 ng/ml stem cell factor and 20% heat-inactivated serum.
  • Plasmid DNA encoding the exogenous protein(s) is incubated with branched or linear PEIs varying in size from 0.8 K to 750 K (Sigma Aldrich, Saint Louis, Mo., USA; Fermetas, Hanover, Md., USA).
  • PEI is prepared as a stock solution at 4.2 mg/mL distilled water and slightly acidified to pH 5.0 using HC1.
  • the DNA may be combined with the PEI for 30 min at room temperature at various nitrogen/phosphate ratios based on the calculation that 1 pg of DNA contains 3 nmol phosphate and 1 pL of PEI stock solution contains 10 nmol amine nitrogen.
  • the isolated CD34 + cells are seeded with the DNA/cationic complex, centrifuged at 280 xg for 5 minutes and incubated in culture medium for 4 or more hours until expression of the exogenous protein(s) is/are assessed.
  • a plasmid vector may be introduced into suitable erythroid cell precursors using a physical method such as particle-mediated transfection, “gene gun,” biolistics, or particle bombardment technology (Papapetrou, et al., Gene Therapy 12:S118-S130, 2005).
  • DNA encoding the exogenous protein is absorbed onto gold particles and administered to cells by a particle gun.
  • This approach may be used, for example, to transfect erythroid cell precursors, e.g., hematopoietic stem cells derived from umbilical cord blood (See, e.g., Verma et al., Gene Therapy 5:692-699, 1998).
  • CD34 + cells are purified using an anti-CD34 monoclonal antibody in combination with magnetic microbeads coated with a secondary antibody and a magnetic isolation system (e.g., Miltenyi MiniMac System, Auburn, Calif., USA).
  • a magnetic isolation system e.g., Miltenyi MiniMac System, Auburn, Calif., USA.
  • the CD34 + enriched cells may be cultured as described herein.
  • plasmid DNA encoding the exogenous protein(s) is precipitated onto a particle, e.g., gold beads, by treatment with calcium chloride and spermidine.
  • the beads may be delivered into the cultured cells using, for example, a Biolistic PDS- 1000/He System (Bio-Rad, Hercules, Calif., USA).
  • a reporter gene such as, for example, beta-galactosidase, chloramphenicol acetyltransferase, luciferase, or green fluorescent protein may be used to assess efficiency of transfection.
  • electroporation methods may be used to introduce a plasmid vector into erythroid cell precursors. Electroporation creates transient pores in the cell membrane, allowing for the introduction of various molecules into the cells including, for example, DNA and RNA.
  • CD34 + cells are isolated and cultured as described herein. Immediately prior to electroporation, the cells are isolated by centrifugation for 10 min at 250xg at room temperature and resuspended at 0.2- lOxlO 6 viable cells/ml in an electroporation buffer such as, for example, X-VIVO 10 supplemented with 1.0% human serum albumin (HSA).
  • HSA human serum albumin
  • Electroporation may be done using, for example, an ECM 600 electroporator (Genetronics, San Diego, Calif., USA) with voltages ranging from 200 V to 280 V and pulse lengths ranging from 25 to 70 milliseconds.
  • ECM 600 electroporator Gene Pulser XcellTM, BioRad, Hercules, Calif.; Cellject Duo, Thermo Science, Milford, Mass.
  • efficient electroporation of isolated CD34 + cells may be performed using the following parameters: 4 mm cuvette, 1600 pE, 550 V/cm, and 10 pg of DNA per 500 pL of cells at lxlO 5 cells/mL (Oldak et al., Acta Biochim. Polonica 49:625-632, 2002).
  • Nucleofection a form of electroporation, may also be used to transfect erythroid cell precursors.
  • transfection is performed using electrical parameters in cell-type specific solutions that enable DNA (or other reagents) to be directly transported to the nucleus, thus reducing the risk of possible degradation in the cytoplasm.
  • a Human CD34 Cell NucleofectorTM Kit (from Amaxa Inc.) may be used to transfect erythroid cell precursors.
  • l-5xl0 6 cells in Human CD34 Cell NucleofectorTM Solution are mixed with 1-5 pg of DNA and transfected in the NucleofectorTM instrument using preprogrammed settings as determined by the manufacturer.
  • Erythroid cell precursors may be non-virally transfected with a conventional expression vector which is unable to self-replicate in mammalian cells unless it is integrated in the genome.
  • erythroid cell precursors may be transfected with an episomal vector which may persist in the host nucleus as autonomously replicating genetic units without integration into chromosomes (Papapetrou et al., Gene Therapy 12:S118-S130, 2005).
  • viruses exploit genetic elements derived from viruses that are normally extrachromosomally replicating in cells upon latent infection such as, for example, EBV, human polyomavirus BK, bovine papilloma virus-1 (BPV-1), herpes simplex virus- 1 (HSV), and Simian virus 40 (SV40).
  • Mammalian artificial chromosomes may also be used for nonviral gene transfer (Vanderbyl et al., Exp. Hematol. 33:1470-1476, 2005).
  • Exogenous nucleic acid encoding one or more exogenous protein(s) can be assembled into expression vectors by standard molecular biology methods known in the art, e.g., restriction digestion, overlap-extension PCR, and Gibson assembly.
  • Exogenous nucleic acids can comprise a gene encoding an exogenous protein that is not normally present on the cell surface, e.g., of an enucleated erythroid cell, fused to a gene that encodes an endogenous or native membrane protein, such that the exogenous protein is expressed on the cell surface.
  • an exogenous gene encoding an exogenous protein can be cloned at the N terminus following the leader sequence of a type 1 membrane protein, at the C terminus of a type 2 membrane protein, or upstream of the GPI attachment site of a GPI-linked membrane protein.
  • the flexible linker is a poly-glycine poly serine linker such as [Gly4Ser]3 (SEQ ID NO: 29) commonly used in generating single-chain antibody fragments from full-length antibodies (Antibody Engineering: Methods & Protocols, B. Lo, ed., Humana Press, 2004, 576 pp.), or Ala-Gly-Ser-Thr (SEQ ID NO: 68) polypeptides such as those used to generate single-chain Arc repressors (Robinson & Sauer, Proc. Nat’l. Acad. Sci. U.S.A. 95: 5929-34, 1998).
  • [Gly4Ser]3 SEQ ID NO: 29
  • Ala-Gly-Ser-Thr SEQ ID NO: 68
  • the flexible linker provides the exogenous protein with more flexibility and steric freedom than the equivalent construct without the flexible linker. This added flexibility is useful in applications that require binding to a target, e.g., an antibody or protein, or an enzymatic reaction of the protein for which the active site must be accessible to the substrate (e.g., the target).
  • a target e.g., an antibody or protein
  • an enzymatic reaction of the protein for which the active site must be accessible to the substrate e.g., the target.
  • the methods provided include the delivery of large nucleic acids (specifically RNAs, such as mRNA) into erythroid cell precursors by contacting the erythroid cell precursor with the nucleic acid and introducing the nucleic acid by electroporation under conditions effective for delivery of the nucleic acid to the cell, such as those described herein.
  • Suitable electroporators include, but are not limited to, the Bio-Rad GENE PULSER and GENE PULSER II; the Life Technologies NEON; BTX GEMINI system; and MAXCYTE electroporator. These methods do not require viral delivery or the use of viral vectors.
  • Suitable nucleic acids include RNAs, such as mRNAs.
  • Suitable nucleic acids also include DNAs, including transposable elements, stable episomes, plasmid DNA, or linear DNA.
  • Suitable electroporation conditions for the methods described herein include for a Life Technologies Neon Transfection System: a pulse voltage ranging from about 500 to about 2000 V, from about 800 to about 1800 V, or from about 850 to about 1700 V; a pulse width ranging from about 5 to about 50 msec, or from about 10 to about 40 msec; and a pulse number ranging from 1 to 2 pulses, 1 to 3 pulses, 1 to 4 pulses, or 1 to 5 pulses.
  • Particularly suitable conditions for electroporation of erythroid cell precursors include, e.g., for 4 days: a) pulse voltage 1300-1400, pulse width: 10-20 msec, number of pulses: 1-3; b) pulse voltage 1400, pulse width: 10 msec, number of pulses: 3; c) pulse voltage 1400, pulse width: 20 msec, number of pulses: 1 ; and d) pulse voltage 1300, pulse width: 10 msec, number of pulses: 3.
  • Particularly suitable conditions for electroporation of erythroid cell precursors include, e.g., for 8-9 days: a) pulse voltage: 1400-1600, pulse width: 20, number of pulses: 1 ; b) pulse voltage: 1100-1300, pulse width: 30, number of pulses: 1; c) pulse voltage: 1000-1200, pulse width: 40, number of pulses: 1 ; d) pulse voltage: 1100- 1400, pulse width: 20, number of pulses: 2; e) pulse voltage: 950-1150, pulse width: 30, number of pulses: 2; f) pulse voltage: 1300-1600, pulse width: 10, number of pulses: 3.
  • Particularly suitable conditions for electroporation of erythroid cell precursors in culture under differentiation conditions include, e.g. for 12-13 days: a) pulse voltage: 1500-1700, pulse width: 20, number of pulses: 1; and b) pulse voltage: 1500- 1600, pulse width: 10, number of pulses: 3.
  • pulse voltage 1500-1700, pulse width: 20, number of pulses: 1
  • pulse voltage 1500- 1600, pulse width: 10, number of pulses: 3.
  • These conditions generally lead to transfections efficiencies of at least about 50% or more (e.g. at least about 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or at least about 97%, or more), and cell viability of at least about 70% or more (e.g. at least about 75%, 80%, 85%, 90%, 95% or at least about 97%, or more).
  • cultured erythroid cell precursors are electroporated for a first time, then cultured for a desired period of time (optionally under differentiation conditions) and then re electroporated a second time.
  • cultured erythroid cell precursors are electroporated for a first time, then cultured for a desired period of time (optionally under differentiation conditions) and then re-electroporated a second, third, fourth, fifth, or sixth time.
  • the culturing period in between the first and second, the second and third, etc. electroporation can be varied. For example, the period in between electroporations may be adjusted as desired, e.g.
  • the period may be 30 minutes, 1 hour, 6 hours, 12, hours, 18 hours, 24 hours, 30 hours, 36 hours, 48 hours, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days 12 days, 13 days 14 days, or 21 days.
  • erythroid cell precursors may be electroporated on day 1 and 2, 1 and 3, 1 and 4, 1 and 5, 1 and 6, 1 and 7, 1 and 8, 1 and 9, 1 and 10, 1 and 11, 1 and 12, 1 and 13, 1 and 14, 1 and 15, or 1 and 16.
  • cells may be electroporated on day 2 and 3, 2 and 4, 2 and 5, 2 and 6, 2 and 7, 2 and 8, 2 and 9, 2 and 10, 2 and 11, 2 and 12, 2 and 13, 2 and 14, 2 and
  • erythroid cell precursors may be electroporated on day 3 and 4, 3 and 5, 3 and 6, 3 and 7, 3 and 8, 3 and 9, 3 and 10, 3 and 11, 3 and 12, 3 and 13, 3 and 14, 3 and 15, or 3 and 16.
  • cells may be electroporated on day 4 and 5, 4 and 6, 4 and 7, 4 and 8, 4 and 9, 4 and 10, 4 and 11, 4 and 12, 4 and 13, 4 and 14, 4 and 15, or 4 and 16.
  • cells may be electroporated on day 5 and 6, 5 and 7, 5 and 8, 5 and 9, 5 and
  • erythroid cell precursors may be electroporated on day 6 and 7, 6 and 8, 6 and 9, 6 and 10, 6 and 11, 6 and 12, 6 and 13, 6 and 14, 6 and 15, or 6 and 16.
  • erythroid cell precursors may be electroporated on day 7 and 8, 7 and 9, 7 and 10, 7 and 11, 7 and 12, 7 and 13, 7 and 14, 7 and 15, or 7 and 16.
  • erythroid cell precursors may be electroporated on day 8 and 9, 8 and 10, 8 and 11, 8 and 12, 8 and 13, 8 and 14, 8 and 15, or 8 and 16.
  • erythroid cell precursors may be electroporated on day 9 10, 9 and 11, 9 and 12, 9 and 13, 9 and 14, 9 and 15, or 9 and 16. In yet another example, erythroid cell precursors may be electroporated on day 10 and 11, 10 and 12, 10 and 13, 10 and 14, 10 and 15, or 10 and 16. In yet another example, erythroid cell precursors may be electroporated on day 11 and 12, 11 and 13, 11 and 14, 11 and 15, or 11 and 16. In yet another example, erythroid cell precursors may be electroporated on day 12 and 13,
  • erythroid cell precursors may be electroporated on day 13 and 14, 13 and 15, or 13 and 16. In yet another example, erythroid cell precursors may be electroporated on day 14 and 15, or 14 and
  • the erythroid cell precursors may be electroporated more than twice, e.g., three times, four times, five times, or six times and the interval may be selected as desired at any points of the differentiation process of the cells.
  • cultured erythroid cell precursors are electroporated on day 1, 2, 3, 4, 5, 6, 7, 8, 9, 10,
  • the engineered enucleated erythroid cells can be click- conjugated engineered enucleated erythroid cells.
  • a catalytic bond-forming polypeptide domain can be expressed on or in, e.g., an erythroid cell precursor, present in the cytosol or present on the membrane.
  • Spy Tag and SpyCatcher undergo isopeptide bond formation between Aspl 17 on Spy Tag and Lys31 on SpyCatcher.
  • the reaction is compatible with the cellular environment and highly specific for protein/peptide conjugation (Zakeri et ak, Proc. Natl. Acad. Sci. U.S.A. 109:E690-E697, 2012).
  • SpyTag and SpyCatcher have been shown to direct post-translational topological modification in elastin-like protein. For example, placement of SpyTag at the N-terminus and SpyCatcher at the C-terminus directs formation of circular elastin-like proteins (Zhang et al, J. Am. Chem. Soc. 2013).
  • the components SpyTag and SpyCatcher can be interchanged such that a system in which molecule A is fused to SpyTag and molecule B is fused to SpyCatcher is functionally equivalent to a system in which molecule A is fused to SpyCatcher and molecule B is fused to SpyTag.
  • a system in which molecule A is fused to SpyTag and molecule B is fused to SpyCatcher is functionally equivalent to a system in which molecule A is fused to SpyCatcher and molecule B is fused to SpyTag.
  • the complementary molecule could be substituted in its place.
  • a catalytic bond-forming polypeptide such as a SpyTag/SpyCatcher system, can be used to attach the exogenous protein to the surface of, e.g., an erythroid cell precursor or an enucleated erythroid cell.
  • the SpyTag polypeptide sequence can be expressed on the extracellular surface of the erytroid cell precursor or the enucleated erythroid cell.
  • the SpyTag polypeptide can be, for example, fused to the N terminus of a type-1 or type-3 transmembrane protein, e.g., glycophorin A, fused to the C terminus of a type-2 transmembrane protein, e.g., Kell, inserted in-frame at the extracellular terminus or in an extracellular loop of a multi-pass transmembrane protein, e.g., Band 3, fused to a GPI-acceptor polypeptide, e.g., CD55 or CD59, fused to a lipid-chain-anchored polypeptide, or fused to a peripheral membrane protein.
  • An exogenous protein can be fused to SpyCatcher.
  • the nucleic acid encoding the SpyCatcher fusion can be expressed and secreted from the same erythroid cell precursor or enucleated erythroid cell that expresses the SpyTag fusion.
  • the nucleic acid sequence encoding the SpyCatcher fusion can be produced exogenously, for example in a bacterial, fungal, insect, mammalian, or cell-free production system.
  • a covalent bond will be formed that attaches the exogenous protein to the surface of the erythroid cell precursor or the enucleated erythroid cell.
  • the SpyTag polypeptide may be expressed as a fusion to the N terminus of glycophorin A under the control of the Gatal promoter in an erythroid cell.
  • An exogenous protein fused to the SpyCatcher polypeptide sequence can be expressed under the control of the Gatal promoter in the same erythroid cell.
  • an isopeptide bond will be formed between the SpyTag and SpyCatcher polypeptides, forming a covalent bond between the erythroid cell surface and the exogenous protein.
  • the SpyTag polypeptide may be expressed as a fusion to the N terminus of glycophorin A under the control of the Gatal promoter in an erythroid cell precursor or an enucleated erythroid cell.
  • An exogenous protein fused to the SpyCatcher polypeptide sequence can be expressed in a suitable mammalian cell expression system, for example HEK293 cells.
  • the SpyCatcher fusion polypeptide can be brought in contact with the cell.
  • an isopeptide bond will be formed between the SpyTag and SpyCatcher polypeptides, forming a covalent bond between the erythroid cell precursor surface or enucleated erythroid cell surface and the exogenous protein.
  • a catalytic bond-forming polypeptide such as a SpyTag/SpyCatcher system, can be used to anchor an exogenous protein to the intracellular space of an erythroid cell precursor or enucleated erythroid cell.
  • the SpyTag polypeptide sequence can be expressed in the intracellular space of the erythroid cell precursor or enucleated erythroid cell by a number of methods, including direct expression of the transgene, fusion to an endogenous intracellular protein such as, e.g., hemoglobin, fusion to the intracellular domain of endogenous cell surface proteins such as, e.g., Band 3, glycophorin A, Kell, or fusion to a structural component of the cytoskeleton.
  • the SpyTag sequence is not limited to a polypeptide terminus and may be integrated within the interior sequence of an endogenous polypeptide such that polypeptide translation and localization is not perturbed.
  • An exogenous protein can be fused to SpyCatcher.
  • the nucleic acid sequence encoding the SpyCatcher fusion can be expressed within the same erythroid cell precursor or enucleated erythroid cell that expresses the Spy Tag fusion.
  • a covalent bond will be formed that acts to anchor the exogenous protein in the intracellular space of the erythroid cell precursor or enucleated erythroid cell.
  • an erythroid cell precursor or an enucleated erythroid cell may express SpyTag fused to hemoglobin beta intracellularly.
  • the erythroid cell precursor or enucleated erythroid cell may be genetically modified with a gene sequence that includes a hemoglobin promoter, beta globin gene, and a SpyTag sequence such that upon translation, intracellular beta globin is fused to SpyTag at is C terminus.
  • the erythroid cell precursor or enucleated erythroid cell expresses a Gatal promoter-led gene that codes for SpyCatcher driving protein expression (e.g., phenylalanine hydroxylase (PAH) expression) such that upon translation, intracellular protein (e.g., PAH) is fused to SpyCatcher at its N terminus.
  • SpyCatcher driving protein expression e.g., phenylalanine hydroxylase (PAH) expression
  • PAH phenylalanine hydroxylase
  • the SpyTag polypeptide can be expressed as a fusion to the exogenous protein within an erythroid cell precursor or an enucleated erythroid cell.
  • the SpyCatcher polypeptide can be expressed as a fusion to the C terminus (intracellular) of glycophorin A within the same erythroid cell precursor or enucleated erythroid cell.
  • an isopeptide bond will be formed between the SpyTag and SpyCatcher polypeptides, forming a covalent bond between the membrane-anchored endogenous erythroid polypeptide and the exogenous protein.
  • polypeptides may be directly conjugated to each other or indirectly through a linker.
  • the linker may be a peptide, a polymer, an aptamer, or a nucleic acid.
  • the polymer may be, e.g., natural, synthetic, linear, or branched.
  • Exogenous proteins can comprise a heterologous fusion protein that comprises a first polypeptide and a second polypeptide with the fusion protein comprising the polypeptides directly j oined to each other or with intervening linker sequences and/or further sequences at one or both ends.
  • the conjugation to the linker may be through covalent bonds or ionic bonds.
  • the engineered enucleated erythroid cells are human enucleated erythroid cells that have been hypotonically loaded.
  • erythroid cell precursors or enucleated erythroid cells are exposed to low ionic strength buffer, causing them to burst.
  • the exogenous protein distributes within the cells.
  • Enucleated erythroid cells or erythroid cell precursors may be hypotonically lysed by adding 30-50 fold volume excess of 5 mM phosphate buffer (pH 8) to a pellet of isolated enucleated erythroid cells. The resulting lysed cell membranes are isolated by centrifugation.
  • the pellet of lysed cell membranes is resuspended and incubated in the presence of the exogenous protein in a low ionic strength buffer, e.g., for 30 min.
  • the lysed cell membranes may be incubated with the exogenous protein for as little as one minute or as long as several days, depending upon the best conditions determined to efficiently load the enucleated erythroid cells or erythroid cell precursors.
  • a nucleic acid For hypotonic loading of a nucleic acid encoding one or more exogenous protein(s) (e.g., any of the exemplary exogenous proteins described herein or known in the art), a nucleic acid can be suspended in a hypotonic Tris-HCl solution (pH 7.0) and injected into erythroid cell precursors.
  • concentration of Tris-HCl can be from about 20 mmol/1 to about 150 mmol/1, depending upon the best conditions determined to efficiently load the enucleated erythroid cells.
  • erythroid cell precursors or enucleated erythroid cells may be loaded with an exogenous protein using controlled dialysis against a hypotonic solution to swell the cells and create pores in the cell membrane (See, e.g., U.S. Pat. Nos. 4,327,710; 5,753,221; 6,495,351, and 10,046,009).
  • a pellet of cells is resuspended in 10 mM HEPES, 140 mM NaCl, 5 mM glucose pH 7.4 and dialyzed against a low ionic strength buffer containing 10 mM Na3 ⁇ 4P04, 10 mM NaHCCh, 20 mM glucose, and 4 mM MgCh. pH 7.4.
  • the cells are further dialyzed against 16 mM NaH2P04, pH 7.4 solution containing the exogenous protein for an additional 30-60 min. All of these procedures may be advantageously performed at a temperature of 4 °C. In some instances, it may be beneficial to load a large quantity of erythroid cell precursors or enucleated erythroid cells by a dialysis approach and a specific apparatus designed for this purpose may be used (See, e.g., U.S. Pat. Nos. 4,327,710, 6,139,836 and 6,495,351).
  • enucleated erythroid cells e.g., reticulocytes or erythrocytes
  • exogenous polypeptides are described, e.g., in WO2015/073587, W02015/153102, W02020/243006, and W02020/219909 each of which is incorporated by reference in its entirety.
  • a population of enucleated erythroid cells contains less than 1% live enucleated cells, e.g., contains no detectable live enucleated cells. Enucleation is measured, in some embodiments, by FACS using a nuclear stain. In some embodiments, at least 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, or 80% (and optionally up to about 70, 80, 90, or 100%) of enucleated erythroid cells in the population comprise one or more (e.g., 2, 3, 4 or more) of the exogenous polypeptides. Expression of the exogenous polypeptide can be measured, in some embodiments, by FACS using labeled antibodies against the polypeptides.
  • At least 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, or 80% (and optionally up to about 70, 80, 90, or 100%) of cells in the population are enucleated and comprise one or more (e.g., 2, 3, 4, or more) of the exogenous polypeptides.
  • the population of enucleated erythroid cells comprises about lxl 0 9 - 2x10 9 , 2xl0 9 - 5xl0 9 , 5xl0 9 - lxlO 10 , lxlO 10 - 2xl0 10 , 2xl0 10 - 5xl0 10 , 5xl0 10 - lxlO 11 , lxlO 11 - 2xlO n , 2xlO n - 5xl0 n , 5xl0 n - lxlO 12 , lxlO 12 - 2xl0 12 , 2xl0 12 - 5xl0 12 , or 5xl0 12 - lxlO 13 cells.
  • any of the enucleated erythroid cells described herein can be formulated as described in, e.g., WO 2020/219909 (incorporated herein by reference).
  • NKp30-positive lymphocytes e.g., NKp30-positive NK cells
  • methods of increasing the number of NKp30-positive lymphocytes e.g., NKp30-positive NK cells
  • a therapeutically effective number of enucleated erythroid cells comprising an exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface (e.g., any of the exemplary enucleated erythroid cells described herein).
  • NKp30-positive lymphocytes e.g., NKp30-positive NK cells
  • methods of increasing the number of NKp30-positive lymphocytes comprising administering to the subject a therapeutically effective number of enucleated erythroid cells comprising an exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface; and an exogenous polypeptide comprising a 4-1BBL polypeptide or functional fragment thereof (e.g., any of the exemplary enucleated erythroid cells described herein).
  • Some embodiments of these methods further include: administering to the subject an NKG2A inhibitor (e.g., any of the exemplary NKG2A inhibitors described herein).
  • an NKG2A inhibitor e.g., any of the exemplary NKG2A inhibitors described herein.
  • the administration comprises intravenous administration to the subject.
  • the one or more additional therapeutic agents is a cancer therapeutic agent.
  • the cancer therapeutic agent is selected from the group of: an inhibitor of an immune checkpoint molecule (e.g., any of the exemplary inhibitors of an immune checkpoint molecule described herein), a chemotherapeutic agent, a therapeutic antibody, a chimeric antigen receptor-T cell, a kinase inhibitor, or a soluble cytokine.
  • the one or more additional therapeutic agents can be administered to the subject at the substantially the same time as any of the compositions provided herein.
  • the one or more additional therapeutic agents can be administered to the subject before or after the administration of any of the compositions described herein to the subject.
  • Some embodiments of these methods can further include identifying or diagnosing a subject as having a B7-H6-positive cancer. Some embodiments of these methods can further include identifying or diagnosing a subject as having a B7-H6- positive and HLA-E-negative cancer.
  • the one or more additional therapeutic agents include an agent that blocks, reduces and/or inhibits PD-f and either PD-Lf or PD-L2, and/or blocks, reduces and/or inhibits the binding of PD-f with PD-Lf or PD- L2 (by way of non-limiting example, one or more of nivolumab (ONO-4538/BMS- 936558, MDX1106, OPDIVO, BRISTOL MYERS SQUIBB), pembrolizumab (KEYTRUDA, MERCK), pidilizumab (CT-011, CURE TECH), MK-3475 (MERCK), BMS 936559 (BRISTOL MYERS SQUIBB), and MPDL3280A (ROCHE)).
  • an agent that blocks, reduces and/or inhibits PD-f and either PD-Lf or PD-L2, and/or blocks, reduces and/or inhibits the binding of PD-f with PD-Lf or PD- L2 by
  • the one or more additional therapeutic agents include an agent that blocks, reduces and/or inhibits the activity of CTLA-4 and/or the binding of CTLA-4 with one or more of its receptors (e.g., CD80, CD86, AP2M1, SHP-2, and PPP2R5A).
  • the agent that inhibits the activity of CTLA-4 is an antibody such as ipilimumab (MDX-010, MDX-101, Yervoy, BRISTOL MYERS SQUIBB) and/or tremelimumab (PFIZER).
  • the one or more additional therapeutic agents include blocking antibodies targeted to an immune checkpoint molecule such as, for example, BTLA, HVEM, TIM3, GALS, LAG3, VISTA, KIR, 2B4, CD160 (also referred to as BY55), CGEN-15049, CHK 1 and CHK2 kinases, A2aR, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), GITR, GITRL, galectin-9, CD244, CD 160, TIGIT, SIRPa, ICOS, CD172a, TMIGD2 and various B-7 family ligands (including, but are not limited to, B7-1, B7-2, B7-DC, B7-H1, B7-H2, B7-H3, B7-H4, B7-H5, B7-H6 and B7-H7).
  • an immune checkpoint molecule such as, for example, BTLA, HVEM, TIM3, GALS, LAG3, VISTA,
  • the methods result in an increase (e.g., at least a 5% increase, at least 10%, at least a 15% increase, at least a 20% increase, at least a 25% increase, at least a 30% increase, at least a 35% increase, at least a 40% increase, at least a 45% increase, at least a 50% increase, at least a 55% increase, at least a 60% increase, at least a 65% increase, at least a 70% increase, at least a 75% increase, at least a 80% increase, at least a 85% increase, at least a 90% increase, at least a 95% increase, at least a 100% increase, at least a 120% increase, at least a 140% increase, at least a 160% increase, at least a 180% increase, at least a 200% increase, at least a 220% increase, at least a 240% increase, at least a 260% increase, at least a 280% increase, or at least a 300% increase) in the number of NKp30
  • an increase e.
  • the methods result in an increase (e.g., at least a 5% increase, at least 10%, at least a 15% increase, at least a 20% increase, at least a 25% increase, at least a 30% increase, at least a 35% increase, at least a 40% increase, at least a 45% increase, at least a 50% increase, at least a 55% increase, at least a 60% increase, at least a 65% increase, at least a 70% increase, at least a 75% increase, at least a 80% increase, at least a 85% increase, at least a 90% increase, at least a 95% increase, at least a 100% increase, at least a 120% increase, at least a 140% increase, at least a 160% increase, at least a 180% increase, at least a 200% increase, at least a 220% increase, at least a 240% increase, at least a 260% increase, at least a 280% increase, or at least a 300% increase) in the number of NKp30
  • an increase e.
  • the methods result in an increase (e.g., at least a 5% increase, at least 10%, at least a 15% increase, at least a 20% increase, at least a 25% increase, at least a 30% increase, at least a 35% increase, at least a 40% increase, at least a 45% increase, at least a 50% increase, at least a 55% increase, at least a 60% increase, at least a 65% increase, at least a 70% increase, at least a 75% increase, at least a 80% increase, at least a 85% increase, at least a 90% increase, at least a 95% increase, at least a 100% increase, at least a 120% increase, at least a 140% increase, at least a 160% increase, at least a 180% increase, at least a 200% increase, at least a 220% increase, at least a 240% increase, at least a 260% increase, at least a 280% increase, or at least a 300% increase) in the number of trafficking NK
  • the methods result in an increase (e.g., at least a 5% increase, at least 10%, at least a 15% increase, at least a 20% increase, at least a 25% increase, at least a 30% increase, at least a 35% increase, at least a 40% increase, at least a 45% increase, at least a 50% increase, at least a 55% increase, at least a 60% increase, at least a 65% increase, at least a 70% increase, at least a 75% increase, at least a 80% increase, at least a 85% increase, at least a 90% increase, at least a 95% increase, at least a 100% increase, at least a 120% increase, at least a 140% increase, at least a 160% increase, at least a 180% increase, at least a 200% increase, at least a 220% increase, at least a 240% increase, at least a 260% increase, at least a 280% increase, or at least a 300% increase) in the number of trafficking NK
  • the subject has previously been diagnosed or identified as having a B7-H6-positive cancer (e.g., any of the exemplary B7-H6-positive cancers described herein). In some embodiments, the subject has previously been diagnosed or identified as having a B7-H6-positive and HLA-E-negative cancer. Some embodiments of these methods can further include identifying or diagnosing a subject as having a B7-H6-positive cancer. Some embodiments of these methods can further include identifying or diagnosing a subject as having a B7-H6-positive and HLA-E- negative cancer.
  • Non-limiting examples of B7-H6-positive cancers include: adrenocortical carcinoma, bile duct cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, esophageal cancer, eye cancer, head and neck cancer, acute myeloid leukemia, lymphoid neoplasm diffuse large b-cell lymphoma, kidney cancer, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, kidney chromophobe, liver cancer, lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma, ovarian cancer, pancreatic cancer, rectal cancer, skin cancer, stomach cancer, testicular cancer, thymoma, thyroid cancer, uterine cancer, uterine corpus endometrial carcinoma, uterine carcinosarcoma, and uveal melanoma.
  • the subject is an adult human subject. In some embodiments, the subject is a pediatric human subject.
  • the enucleated erythroid cells can be administered to the subject using any of the doses and at any of the frequencies described herein.
  • a therapeutically effective number of enucleated erythroid cells comprising an exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface (e.g., any of the exemplary enucleated
  • a therapeutically effective number of enucleated erythroid cells comprising an exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on
  • the administration is intravenous administration to the subject.
  • Some embodiments of these methods further include administering to the subject an NKG2A inhibitor (e.g., any of the exemplary NKG2A inhibitors described herein).
  • an NKG2A inhibitor e.g., any of the exemplary NKG2A inhibitors described herein.
  • the one or more additional therapeutic agents is a cancer therapeutic agent.
  • the cancer therapeutic agent is selected from the group of: an inhibitor of an immune checkpoint molecule (e.g., any of the exemplary inhibitors of an immune checkpoint molecule described herein), a chemotherapeutic agent, a therapeutic antibody, a chimeric antigen receptor-T cell, a kinase inhibitor, or a soluble cytokine.
  • the one or more additional therapeutic agents can be administered to the subject at the substantially the same time as any of the compositions provided herein.
  • the one or more additional therapeutic agents can be administered to the subject before or after the administration of any of the compositions described herein to the subject.
  • the one or more additional therapeutic agents include an agent that blocks, reduces and/or inhibits PD-1 and either PD-L1 or PD-L2, and/or blocks, reduces and/or inhibits the binding of PD-1 with PD-L1 or PD- L2 (by way of non-limiting example, one or more of nivolumab (ONO-4538/BMS- 936558, MDX1106, OPDIVO, BRISTOL MYERS SQUIBB), pembrolizumab (KEYTRUDA, MERCK), pidilizumab (CT-011, CURE TECH), MK-3475 (MERCK), BMS 936559 (BRISTOL MYERS SQUIBB), and MPDL3280A (ROCHE)).
  • an agent that blocks, reduces and/or inhibits PD-1 and either PD-L1 or PD-L2, and/or blocks, reduces and/or inhibits the binding of PD-1 with PD-L1 or PD- L2 by way of non-
  • the one or more additional therapeutic agents include an agent that blocks, reduces and/or inhibits the activity of CTLA-4 and/or the binding of CTLA-4 with one or more of its receptors (e.g., CD80, CD86, AP2M1, SHP-2, and PPP2R5A).
  • the agent that inhibits the activity of CTLA-4 is an antibody such as ipilimumab (MDX-010, MDX-101, Yervoy, BRISTOL MYERS SQUIBB) and/or tremelimumab (PFIZER).
  • the one or more additional therapeutic agents include blocking antibodies targeted to an immune checkpoint molecule such as, for example, BTLA, HVEM, TIM3, GALS, LAG3, VISTA, KIR, 2B4, CD160 (also referred to as BY55), CGEN-15049, CHK 1 and CHK2 kinases, A2aR, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), GITR, GITRL, galectin-9, CD244, CD 160, TIGIT, SIRPa, ICOS, CD172a, TMIGD2 and various B-7 family ligands (including, but are not limited to, B7-1, B7-2, B7-DC, B7-H1, B7-H2, B7-H3, B7-H4, B7-H5, B7-H6 and B7-H7).
  • an immune checkpoint molecule such as, for example, BTLA, HVEM, TIM3, GALS, LAG3, VISTA,
  • Some embodiments of these methods can further include identifying or diagnosing a subject as having a B7-H6-positive cancer. Some embodiments of these methods can further include identifying or diagnosing a subject as having a B7-H6- positive and HLA-E-negative cancer.
  • Non-limiting examples of B7-H6-positive cancers include: adrenocortical carcinoma, bile duct cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, esophageal cancer, eye cancer, head and neck cancer, acute myeloid leukemia, lymphoid neoplasm diffuse large b-cell lymphoma, kidney cancer, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, kidney chromophobe, liver cancer, lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma, ovarian cancer, pancreatic cancer, rectal cancer, skin cancer, stomach cancer, testicular cancer, thymoma, thyroid cancer, uterine cancer, uterine corpus endometrial carcinoma, uterine carcinosarcoma, and uveal melanoma.
  • the subject is an adult human subject. In some embodiments, the subject is a pediatric human subject.
  • the enucleated erythroid cells can be administered to the subject using any of the doses and at any of the frequencies described herein.
  • B7-H6-positive cancer cells e.g., any of the exemplary B7-H6- positive cancer cells described herein or known in the art
  • the method comprising administering a therapeutically effective number of enucleated erythroid cells comprising an exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface (e.g., any of the exemplary enucleated erythroid cells described herein).
  • B7-H6-positive cancer cells e.g., any of the exemplary B7-H6-positive cancer cells described herein or known in the art
  • the method comprising administering a therapeutically effective number of enucleated erythroid cells comprising an exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface; and an exogenous polypeptide comprising a 4-1BBL polypeptide or a functional fragment thereof (e.g., any of the exemplary enucleated erythroid cells described herein).
  • the administration is intravenous administration to the subject.
  • Some embodiments of these methods further include administering to the subject an NKG2A inhibitor (e.g., any of the exemplary NKG2A inhibitors described herein).
  • an NKG2A inhibitor e.g., any of the exemplary NKG2A inhibitors described herein.
  • the B7-H6-positive cancer cell is a B7-H6-positive and HLA-E-negative cancer cell.
  • Some embodiments of any of the methods described herein further include administering one or more additional therapeutic agents to the subject.
  • the one or more additional therapeutic agents is a cancer therapeutic agent.
  • the cancer therapeutic agent is selected from the group of: an inhibitor of an immune checkpoint molecule (e.g., any of the exemplary inhibitors of an immune checkpoint molecule described herein), a chemotherapeutic agent, a therapeutic antibody, a chimeric antigen receptor-T cell, a kinase inhibitor, or a soluble cytokine.
  • the one or more additional therapeutic agents can be administered to the subject at the substantially the same time as any of the compositions provided herein. In some embodiments, the one or more additional therapeutic agents can be administered to the subject before or after the administration of any of the compositions described herein to the subject.
  • the one or more additional therapeutic agents include an agent that blocks, reduces and/or inhibits PD-1 and either PD-L1 or PD-L2, and/or blocks, reduces and/or inhibits the binding of PD-1 with PD-L1 or PD- L2 (by way of non-limiting example, one or more of nivolumab (ONO-4538/BMS- 936558, MDX1106, OPDIVO, BRISTOL MYERS SQUIBB), pembrolizumab (KEYTRUDA, MERCK), pidilizumab (CT-011, CURE TECH), MK-3475 (MERCK), BMS 936559 (BRISTOL MYERS SQUIBB), and MPDL3280A (ROCHE)).
  • an agent that blocks, reduces and/or inhibits PD-1 and either PD-L1 or PD-L2, and/or blocks, reduces and/or inhibits the binding of PD-1 with PD-L1 or PD- L2 by way of non-
  • the one or more additional therapeutic agents include an agent that blocks, reduces and/or inhibits the activity of CTLA-4 and/or the binding of CTLA-4 with one or more of its receptors (e.g., CD80, CD86, AP2M1, SHP-2, and PPP2R5A).
  • the agent that inhibits the activity of CTLA-4 is an antibody such as ipilimumab (MDX-010, MDX-101, Yervoy, BRISTOL MYERS SQUIBB) and/or tremelimumab (PFIZER).
  • the one or more additional therapeutic agents include blocking antibodies targeted to an immune checkpoint molecule such as, for example, BTLA, HVEM, TIM3, GALS, LAG3, VISTA, KIR, 2B4, CD160 (also referred to as BY55), CGEN-15049, CHK 1 and CHK2 kinases, A2aR, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), GITR, GITRL, galectin-9, CD244, CD 160, TIGIT, SIRPa, ICOS, CD172a, TMIGD2 and various B-7 family ligands (including, but are not limited to, B7-1, B7-2, B7-DC, B7-H1, B7-H2, B7-H3, B7-H4, B7-H5, B7-H6 and B7-H7).
  • an immune checkpoint molecule such as, for example, BTLA, HVEM, TIM3, GALS, LAG3, VISTA,
  • Some embodiments of these methods can further include identifying or diagnosing a subject as having a B7-H6-positive cancer. Some embodiments of these methods can further include identifying or diagnosing a subject as having a B7-H6- positive and HLA-E-negative cancer.
  • the method results in a decrease (e.g., at least a 5% decrease, at least a 10% decrease, at least a 15% decrease, at least a 20% decrease, at least a 25% decrease, at least a 30% decrease, at least a 35% decrease, at least a 40% decrease, at least a 45% decrease, at least a 50% decrease, at least a 55% decrease, at least a 60% decrease, at least a 65% decrease, at least a 70% decrease, at least a 75% decrease, at least a 80% decrease, at least a 85% decrease, at least a 90% decrease, or at least a 95% decrease, or about a 5% decrease to about a 99% decrease (or any of the subranges of this range described herein)) in the number of B7-H6- positive cancer cells in the subject (e.g., as compared to the number of B7-H6-positive cancer cells in the subject prior to the administering).
  • a decrease e.g., at least a 5% decrease, at least
  • the method results in a decrease (e.g., at least a 5% decrease, at least a 10% decrease, at least a 15% decrease, at least a 20% decrease, at least a 25% decrease, at least a 30% decrease, at least a 35% decrease, at least a 40% decrease, at least a 45% decrease, at least a 50% decrease, at least a 55% decrease, at least a 60% decrease, at least a 65% decrease, at least a 70% decrease, at least a 75% decrease, at least a 80% decrease, at least a 85% decrease, at least a 90% decrease, or at least a 95% decrease, or about a 5% decrease to about a 99% decrease (or any of the subranges of this range described herein)) in the number of B7-H6- positive and HLA-E-negative cancer cells in the subject (e.g., as compared to the number of B7-H6-positive and HLA-E-negative cancer cells in the subject prior to the administering).
  • a decrease e
  • the method results in a decrease (e.g., at least a 5% decrease, at least a 10% decrease, at least a 15% decrease, at least a 20% decrease, at least a 25% decrease, at least a 30% decrease, at least a 35% decrease, at least a 40% decrease, at least a 45% decrease, at least a 50% decrease, at least a 55% decrease, at least a 60% decrease, at least a 65% decrease, at least a 70% decrease, at least a 75% decrease, at least a 80% decrease, at least a 85% decrease, at least a 90% decrease, or at least a 95% decrease, or about a 5% decrease to about a 99% decrease (or any of the subranges of this range described herein)) in the proliferation of B7-H6- positive cancer cells in the subject (e.g., as compared to the proliferation of B7-H6- positive cancer cells in the subject prior to the administering).
  • a decrease e.g., at least a 5% decrease, at least
  • the method results in a decrease (e.g., at least a 5% decrease, at least a 10% decrease, at least a 15% decrease, at least a 20% decrease, at least a 25% decrease, at least a 30% decrease, at least a 35% decrease, at least a 40% decrease, at least a 45% decrease, at least a 50% decrease, at least a 55% decrease, at least a 60% decrease, at least a 65% decrease, at least a 70% decrease, at least a 75% decrease, at least a 80% decrease, at least a 85% decrease, at least a 90% decrease, or at least a 95% decrease, or about a 5% decrease to about a 99% decrease (or any of the subranges of this range described herein)) in the proliferation of B7-H6- positive and HLA-E-negative cancer cells in the subject (e.g., as compared to the proliferation of B7-H6-positive and HLA-E-negative cancer cells in the subject prior to the administering).
  • a decrease e
  • Some embodiments of these methods can further include identifying or diagnosing a subject as having a B7-H6-positive cancer. Some embodiments of these methods can further include identifying or diagnosing a subject as having a B7-H6- positive and HLA-E-negative cancer.
  • Non-limiting examples of B7-H6-positive cancer cells include: adrenocortical carcinoma, bile duct cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, esophageal cancer, eye cancer, head and neck cancer, acute myeloid leukemia, lymphoid neoplasm diffuse large b-cell lymphoma, kidney cancer, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, kidney chromophobe, liver cancer, lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma, ovarian cancer, pancreatic cancer, rectal cancer, skin cancer, stomach cancer, testicular cancer, thymoma, thyroid cancer, uterine cancer, uterine corpus endometrial carcinoma, uterine carcinosarcoma, and uveal melanoma.
  • the subject is an adult human subject. In some embodiments, the subject is a pediatric human subject.
  • the enucleated erythroid cells can be administered to the subject using any of the doses and at any of the frequencies described herein.
  • Methods of treating a subject by inducing killing a B7-H6-positive cancer cell can be administered to the subject using any of the doses and at any of the frequencies described herein.
  • Also provided herein are methods of inducing killing a B7-H6-positive cancer cell (e.g., any of the exemplary B7-H6-positive cancer cells described herein) in a subject previously identified or diagnosed as having a B7-H6-positive cancer that include administering a therapeutically effective number of enucleated erythroid cells comprising an exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface (e.g., any of the exemplary enucleated erythroid cells described herein).
  • Also provided herein are methods of killing a B7-H6-positive cancer cell (e.g., any of the exemplary B7-H6-positive cancer cells described herein) in a subject previously identified or diagnosed as having a B7-H6-positive cancer that include administering a therapeutically effective number of enucleated erythroid cells comprising an exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface; and an exogenous polypeptide comprising a 4-1BBL polypeptide of a functional fragment thereof (e.g., any of the exemplary enucleated erythroid cells described herein).
  • a therapeutically effective number of enucleated erythroid cells comprising an exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface; and
  • the killing of the B7-H6-positive cancer cell is necrosis. In some embodiments, the killing of the B7-H6-positive cancer cell is apoptosis. In some embodiments, the kill is NK cell-mediated cytolysis or NK cell- mediated cytoxicity.
  • Some embodiments of these methods further include: administering to the subject an NKG2A inhibitor (e.g., any of the exemplary NKG2A inhibitors described herein).
  • an NKG2A inhibitor e.g., any of the exemplary NKG2A inhibitors described herein.
  • the administering includes intravenous administration to the subject.
  • the one or more additional therapeutic agents is a cancer therapeutic agent.
  • the cancer therapeutic agent is selected from the group of: an inhibitor of an immune checkpoint molecule (e.g., any of the exemplary inhibitors of an immune checkpoint molecule described herein), a chemotherapeutic agent, a therapeutic antibody, a chimeric antigen receptor-T cell, a kinase inhibitor, or a soluble cytokine.
  • the one or more additional therapeutic agents can be administered to the subject at the substantially the same time as any of the compositions provided herein.
  • the one or more additional therapeutic agents can be administered to the subject before or after the administration of any of the compositions described herein to the subject.
  • the one or more additional therapeutic agents include an agent that blocks, reduces and/or inhibits PD-1 and either PD-L1 or PD-L2, and/or blocks, reduces and/or inhibits the binding of PD-1 with PD-L1 or PD- L2 (by way of non-limiting example, one or more of nivolumab (ONO-4538/BMS- 936558, MDX1106, OPDIVO, BRISTOL MYERS SQUIBB), pembrolizumab (KEYTRUDA, MERCK), pidilizumab (CT-011, CURE TECH), MK-3475 (MERCK), BMS 936559 (BRISTOL MYERS SQUIBB), and MPDL3280A (ROCHE)).
  • an agent that blocks, reduces and/or inhibits PD-1 and either PD-L1 or PD-L2, and/or blocks, reduces and/or inhibits the binding of PD-1 with PD-L1 or PD- L2 by way of non-
  • the one or more additional therapeutic agents include an agent that blocks, reduces and/or inhibits the activity of CTLA-4 and/or the binding of CTLA-4 with one or more of its receptors (e.g., CD80, CD86, AP2M1, SHP-2, and PPP2R5A).
  • the agent that inhibits the activity of CTLA-4 is an antibody such as ipilimumab (MDX-010, MDX-101, Yervoy, BRISTOL MYERS SQUIBB) and/or tremelimumab (PFIZER).
  • the one or more additional therapeutic agents include blocking antibodies targeted to an immune checkpoint molecule such as, for example, BTLA, HVEM, TIM3, GALS, LAG3, VISTA, KIR, 2B4, CD160 (also referred to as BY55), CGEN-15049, CHK 1 and CHK2 kinases, A2aR, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), GITR, GITRL, galectin-9, CD244, CD 160, TIGIT, SIRPa, ICOS, CD172a, TMIGD2 and various B-7 family ligands (including, but are not limited to, B7-1, B7-2, B7-DC, B7-H1, B7-H2, B7-H3, B7-H4, B7-H5, B7-H6 and B7-H7).
  • an immune checkpoint molecule such as, for example, BTLA, HVEM, TIM3, GALS, LAG3, VISTA,
  • Some embodiments of these methods can further include identifying or diagnosing a subject as having a B7-H6-positive cancer. Some embodiments of these methods can further include identifying or diagnosing a subject as having a B7-H6- positive and HLA-E-negative cancer.
  • Non-limiting examples of B7-H6-positive cancer cells include: adrenocortical carcinoma, bile duct cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, esophageal cancer, eye cancer, head and neck cancer, acute myeloid leukemia, lymphoid neoplasm diffuse large b-cell lymphoma, kidney cancer, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, kidney chromophobe, liver cancer, lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma, ovarian cancer, pancreatic cancer, rectal cancer, skin cancer, stomach cancer, testicular cancer, thymoma, thyroid cancer, uterine cancer, uterine corpus endometrial carcinoma, uterine carcinosarcoma, and uveal melanoma.
  • the subject is an adult human subject. In some embodiments, the subject is a pediatric human subject.
  • the enucleated erythroid cells can be administered to the subject using any of the doses and at any of the frequencies described herein.
  • methods of reducing the volume of a solid tumor in a subject previously identified or diagnosed as having a B7-H6-positive cancer that (e.g., any of the exemplary B7-H6 cancers described herein or known in the art) include administering a therapeutically effective number of enucleated erythroid cells comprising an exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface (e.g., any of the exemplary enucleated erythroid cells described herein).
  • Also provided herein are methods of reducing the volume of a solid tumor in a subject previously identified or diagnosed as having a B7-H6-positive cancer e.g., any of the exemplary B7-H6 positive cancers described herein
  • a therapeutically effective number of enucleated erythroid cells comprising an exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface; and an exogenous polypeptide comprising a 4-1BBL polypeptide, or a functional fragment thereof (e.g., any of the exemplary enucleated erythroid cells described herein).
  • Some embodiments of these methods further include administering to the subject an NKG2A inhibitor (e.g., any of the exemplary NKG2A inhibitors described herein).
  • the administering includes intravenous administration to the subject.
  • the one or more additional therapeutic agents is a cancer therapeutic agent.
  • the cancer therapeutic agent is selected from the group of: an inhibitor of an immune checkpoint molecule (e.g., any of the exemplary inhibitors of an immune checkpoint molecule described herein), a chemotherapeutic agent, a therapeutic antibody, a chimeric antigen receptor-T cell, a kinase inhibitor, or a soluble cytokine.
  • the one or more additional therapeutic agents can be administered to the subject at the substantially the same time as any of the compositions provided herein.
  • the one or more additional therapeutic agents can be administered to the subject before or after the administration of any of the compositions described herein to the subject.
  • the one or more additional therapeutic agents include an agent that blocks, reduces and/or inhibits PD-1 and either PD-L1 or PD-L2, and/or blocks, reduces and/or inhibits the binding of PD-1 with PD-L1 or PD- L2 (by way of non-limiting example, one or more of nivolumab (ONO-4538/BMS- 936558, MDX1106, OPDIVO, BRISTOL MYERS SQUIBB), pembrolizumab (KEYTRUDA, MERCK), pidilizumab (CT-011, CURE TECH), MK-3475 (MERCK), BMS 936559 (BRISTOL MYERS SQUIBB), and MPDL3280A (ROCHE)).
  • an agent that blocks, reduces and/or inhibits PD-1 and either PD-L1 or PD-L2, and/or blocks, reduces and/or inhibits the binding of PD-1 with PD-L1 or PD- L2 by way of non-
  • the one or more additional therapeutic agents include an agent that blocks, reduces and/or inhibits the activity of CTLA-4 and/or the binding of CTLA-4 with one or more of its receptors (e.g., CD80, CD86, AP2M1, SHP-2, and PPP2R5A).
  • the agent that inhibits the activity of CTLA-4 is an antibody such as ipilimumab (MDX-010, MDX-101, Yervoy, BRISTOL MYERS SQUIBB) and/or tremelimumab (PFIZER).
  • the one or more additional therapeutic agents include blocking antibodies targeted to an immune checkpoint molecule such as, for example, BTLA, HVEM, TIM3, GALS, LAG3, VISTA, KIR, 2B4, CD160 (also referred to as BY55), CGEN-15049, CHK 1 and CHK2 kinases, A2aR, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), GITR, GITRL, galectin-9, CD244, CD 160, TIGIT, SIRPa, ICOS, CD172a, TMIGD2 and various B-7 family ligands (including, but are not limited to, B7-1, B7-2, B7-DC, B7-H1, B7-H2, B7-H3, B7-H4, B7-H5, B7-H6 and B7-H7).
  • an immune checkpoint molecule such as, for example, BTLA, HVEM, TIM3, GALS, LAG3, VISTA,
  • Some embodiments of these methods can further include identifying or diagnosing a subject as having a B7-H6-positive cancer. Some embodiments of these methods can further include identifying or diagnosing a subject as having a B7-H6- positive and HLA-E-negative cancer.
  • Non-limiting examples of B7-H6-positive cancers include: adrenocortical carcinoma, bile duct cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, esophageal cancer, eye cancer, head and neck cancer, acute myeloid leukemia, lymphoid neoplasm diffuse large b-cell lymphoma, kidney cancer, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, kidney chromophobe, liver cancer, lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma, ovarian cancer, pancreatic cancer, rectal cancer, skin cancer, stomach cancer, testicular cancer, thymoma, thyroid cancer, uterine cancer, uterine corpus endometrial carcinoma, uterine carcinosarcoma, and uveal melanoma.
  • the method results in a decrease (e.g., at least a 5% decrease, at least a 10% decrease, at least a 15% decrease, at least a 20% decrease, at least a 25% decrease, at least a 30% decrease, at least a 35% decrease, at least a 40% decrease, at least a 45% decrease, at least a 50% decrease, at least a 55% decrease, at least a 60% decrease, at least a 65% decrease, at least a 70% decrease, at least a 75% decrease, at least a 80% decrease, at least a 85% decrease, at least a 90% decrease, or at least a 95% decrease, or about a 5% decrease to about a 99% decrease (or any of the subranges of this range described herein)) in the volume of the solid tumor in the subject (e.g., as compared to the volume of the solid tumor prior to the administering).
  • a decrease e.g., at least a 5% decrease, at least a 10% decrease, at least a 15% decrease, at least
  • the subject is an adult human subject. In some embodiments, the subject is a pediatric human subject.
  • the enucleated erythroid cells can be administered to the subject using any of the doses and at any of the frequencies described herein. Doses and Administration
  • enucleated red blood cells e.g., reticulocytes
  • exogenous agent e.g., polypeptides
  • the enucleated red blood cells described herein are administered to a subject, e.g., a mammal, e.g., a human.
  • a subject e.g., a mammal, e.g., a human.
  • mammals that can be treated include without limitation, humans, domestic animals (e.g., dogs, cats and the like), farm animals (e.g., cows, sheep, pigs, horses and the like) and laboratory animals (e.g., monkey, rats, mice, rabbits, guinea pigs and the like).
  • farm animals e.g., cows, sheep, pigs, horses and the like
  • laboratory animals e.g., monkey, rats, mice, rabbits, guinea pigs and the like.
  • the methods described herein are applicable to both human therapy and veterinary applications.
  • the enucleated erythroid cells are administered to a patient every 1, 2, 3, 4, 5, or 6 months.
  • a dose of enucleated erythroid cells comprises about lxlO 8 - lxlO 13 , about lxlO 8 - lxlO 9 , about lxlO 9 - 2xl0 9 , 2xl0 9 - 5xl0 9 , 5xl0 9 - lxlO 10 , lxlO 10 - 2xl0 10 , 2x10 10 - 5xl0 10 , 5xl0 10 - lxlO 11 , lxlO 11 - 2xlO n , 2xlO n - 5xl0 n , 5xl0 n - lxlO 12 , lxlO 12 - 2xl0 12 , 2xl0 12 - 5xl0 12 , or 5xl0 12 - lxlO 13 cells.
  • the engineered erythroid cell can be administered to the subject in a formulation suitable for parenteral, intral-lesional, buccal, ophthalmic, intravenous, intra-organ, or another route of administration.
  • the engineered erythroid cell can be administered to a subject as frequently as several times daily, or it can be administered less frequently, such as once a day, once a week, once every two weeks, once a month, or even less frequently, such as one every several months or even once a year or less.
  • the frequency of the dose will be readily apparent to the skilled artisan and will depend upon any number of factors, such as, but not limited to, the type and severity of the disease being treated, the type and age of the subject, etc.
  • the B7-H6-positive cancer is a cancer selected from the group consisting of: adrenocortical carcinoma, bile duct cancer, bladder cancer, breast cancer, cervical cancer, colon cancer, colorectal cancer, esophageal cancer, eye cancer, head and neck cancer, acute myeloid leukemia, lymphoid neoplasm diffuse large b-cell lymphoma, kidney cancer, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, kidney chromophobe, liver cancer, lung cancer, lung adenocarcinoma, lung squamous cell carcinoma, mesothelioma, ovarian cancer, pancreatic cancer, rectal cancer, skin cancer, stomach cancer, testicular cancer, thymoma, thyroid cancer, uterine cancer, uterine corpus endometrial carcinoma, uterine carcinosarcoma, and uveal melanoma.
  • adrenocortical carcinoma bile duct cancer, bladder cancer, breast
  • kits that include any of the compositions provided herein.
  • a kit that includes a pharmaceutical composition comprising an enucleated erythroid cell comprising an exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface (e.g., any of the exemplary enucleated erythroid cells described herein); and instructions for performing the any of the methods described herein.
  • the enucleated erythroid cell comprises at least 1,000 copies, at least 10,000 copies, or at least 15,000 copies of the exogenous fusion polypeptide.
  • the enucleated erythroid cell comprises at least 1,000 copies, at least 10,000 copies, at least 15,000 copies, at least 20,000 copies, at least 25,000 copies, at least 30,000 copies, at least 40,000 copies, at least 50,000 copies, at least 60,000 copies, at least 80,000 copies, at least 100,000 copies, at least 200,000 copies, at least 300,000 copies, at least 400,000 copies, at least 500,000 copies, or at least 600,000 copies of the exogenous polypeptide.
  • kits that includes a pharmaceutical composition comprising an enucleated erythroid cell comprising an exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface; and an exogenous polypeptide comprising a 4-1BBL polypeptide or a functional fragment thereof, where the exogenous polypeptide is present on the surface of the enucleated erythroid cell (e.g., any of the exemplary enucleated erythroid cells described herein); and instructions for performing the any of the methods described herein.
  • a pharmaceutical composition comprising an enucleated erythroid cell comprising an exogenous fusion polypeptide comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, on its extracellular surface; and an exogenous polypeptide comprising a 4-1BBL polypeptide or a functional fragment thereof,
  • kits that include one or more sterile vessels containing any of the compositions described herein (e.g., a sterile conical tube, a sterile petri dish, a sterile vial (e.g., a borosilicate glass vial), and sterile plastic bags (a di-2-ethylhexyl phthalate (DEHP)-plasticized polyvinyl chloride (PVC) bag, or n- butyryl-tri(n-hexyl)-citrate (BTHC)-plasticized PVC bag).
  • any of the kits provided herein can further include instructions for administration of any of the compositions to a subject in need thereof.
  • kits described herein include a suitable single dosage form of any of the compositions described herein.
  • a single dosage form of any of the compositions described herein can have a volume of, e.g., about 0.5 mL to about 2 L, about 0.5 mL to about 1800 mL, about 0.5 mL to about 1500 mL, about 0.5 mL to about 1200 mL, about 0.5 mL to about 1000 mL, about 0.5 mL to about 800 mL, about 0.5 mL to about 600 mL, about 0.5 mL to about 400 mL, about 0.5 mL to about 200 mL, about 0.5 mL to about 180 mL, about 0.5 mL to about 160 mL, about 0.5 mL to about 140 mL, about 0.5 mL to about 120 mL, about 0.5 mL to about 100 mL, about 0.5 mL to about 80 mL, about 0.5 m
  • Example 1 Generation of erythroid cells genetically engineered to express the first fusion polypeptide
  • IL-15 and IL-15/IL-15RA fusion constructs and polypeptides were prepared for expression in erythroid cells as shown in Table 5 below. Table 5.
  • SEQ ID NOs. refer to amino acid sequences.
  • the DNA constructs were cloned into the multiple cloning site of a lentivirus vector under the control of a MSCV promoter sequence for expression in erythroid cells, as described below.
  • Lentivirus was produced in 293T cells by transfecting the cells with pPACKHl (System Biosciences) and pCDH lentivirus vector containing constructs. Cells were placed in fresh culturing medium. The virus supernatant was collected 48 hours post-medium change by centrifugation at 1,500 rpm for 5 minutes. The supernatant was collected, filtered, and frozen in aliquots at -80°C. Expansion and differentiation of erythroid cells
  • Human CD34 + cells derived from mobilized peripheral blood cells from normal human donors were used.
  • the expansion/differentiation procedure comprises 3 stages.
  • thawed CD34 + erythroid cell precursors were cultured in Iscove’s MDM (IMDM) medium comprising recombinant human insulin, human transferrin, recombinant human stem cell factor, and recombinant human interleukin 3.
  • IMDM Iscove’s MDM
  • erythroid cells were cultured in IMDM medium supplemented with recombinant human insulin, human transferrin, human recombinant stem cell factor, human recombinant erythropoietin, and L-glutamine.
  • erythroid cells were cultured in IMDM medium supplemented with human transferrin, recombinant human insulin, human recombinant erythropoietin, and heparin.
  • the cultures were maintained at 37°C in 5% C02 incubator.
  • Erythroid precursor cells were transduced during stage 1 of the culture process described above. Erythroid precursor cells in culturing medium were combined with lentiviral supernatant and polybrene. Infection was achieved by spinoculation, spinning the plate at 2,000 rpm for 90 minutes at room temperature. After spinoculation, the cells were incubated at 37°C overnight.
  • Antibody Binding Binding of a PE-labelled anti-IL-15-RA antibody (e.g., anti-IL-15RA antibody
  • JM7A4 (ab91270), AbCam) was used to validate expression of the the first exogenous polypeptide in the engineered erythroid cells. Binding of the antibody was measured by flow cytometry for APC fluorescence or PE fluorescence. A gate was set based on stained untransduced cells.
  • Example 2 Generation of erythroid cells genetically engineered to express 4- 1BBL.
  • DNA constructs were prepared for expression in erythroid cells as shown in Table 6 below:
  • SEQ ID NOs. refer to amino acid sequences.
  • Lentiviral Vector 4-1BBL gene construct was constructed as shown in Table 6. Genes were cloned into the multiple cloning site of lentivirus vector pCDH with the MSCV promoter sequence from System Biosciences. Lentivirus was produced in 293T cells by transfecting the cells with pPACKHl (System Biosciences) and pCDH lentivirus vector containing 4-1BBL genes. Cells were placed in fresh culturing medium. The virus supernatant was collected 48 hours post-medium change by centrifugation at 1,500 rpm for 5 minutes. The supernatant was collected, filtered, and frozen in aliquots at -80°C. Expansion and differentiation of erythroid cells
  • Human CD34 + cells derived from mobilized peripheral blood cells from normal human donors were purchased frozen from AllCells Inc.
  • the expansion/differentiation procedure comprises 3 stages.
  • thawed CD34 + erythroid precursors were cultured in Iscove’s MDM medium comprising recombinant human insulin, human transferrin, recombinant human recombinant human stem cell factor, and recombinant human interleukin 3.
  • erythroid cells were cultured in Iscove’s MDM medium supplemented with recombinant human insulin, human transferrin, human recombinant stem cell factor, human recombinant erythropoietin, and L-glutamine.
  • erythroid cells were cultured in Iscove’s MDM medium supplemented with human transferrin, recombinant human insulin, human recombinant erythropoietin, and heparin.
  • the cultures were maintained at 37°C in 5% C02 incubator.
  • Erythroid precursor cells were transduced during step 1 of the culture process described above. Erythroid cells in culturing medium were combined with lentiviral supernatant and polybrene. Infection was achieved by spinoculation, spinning the plate at 2000 rpm for 90 minutes at room temperature. After spinoculation, the cells were incubated at 37°C overnight.
  • Binding of a PE-labelled anti-4-lBBL antibody e.g., purified anti-human 4- 1BB Ligand (CD137L) antibody, BioLegend
  • a PE-labelled anti-4-lBBL antibody e.g., purified anti-human 4- 1BB Ligand (CD137L) antibody, BioLegend
  • Binding of the antibody was measured by flow cytometry for PE fluorescence. A gate was set based on stained untransduced cells.
  • Example 3 Generation of erythroid cells genetically engineered to express the first and second exogenous polypeptides Production of Lentiviral Vectors
  • IL-15/IL-15-RA fusion protein and 4-1BBL were constructed. Each gene was cloned into the multiple cloning site of lentivirus vector pCDH under the control of the MSCV promoter sequence (System Biosciences), such that one vector comprised the gene for IL-15/IL-15RA and another vector comprised the gene for 4-1BBL.
  • Lentivirus was produced in 293T cells by co-transfecting the cells with pPACKHl (System Biosciences) and pCDH lentivirus vector containing IL-15/IL-15- RA gene and pCDH lentivirus vector containing 4-1BBL gene. Cells were placed in fresh culturing medium. The virus supernatant was collected 48 hours post-medium change by centrifugation at 1,500 rpm for 5 minutes. The supernatant was collected, filtered, and frozen in aliquots at -80°C.
  • IL-15/IL-15-RA fusion protein and 4-1BBL genes were constructed and cloned into the multiple cloning site of lentivirus vector pCDH, under the control of the MSCV promoter sequence (System Biosciences), such that a single vector comprised the genes for IL-15/IL-15RA and the gene for 4-1BBL.
  • Lentivirus was produced in 293T cells by co-transfecting the cells with pPACKHl (System Biosciences) and pCDH lentivirus vector containing both IL-15/IL-15-RA gene and 4-1BBL gene. Cells were placed in fresh culturing medium. The virus supernatant was collected 48 hours post-medium change by centrifugation at 1,500 rpm for 5 minutes. The supernatant was collected, filtered, and frozen in aliquots at -80°C.
  • Human CD34+ cells derived from mobilized peripheral blood cells from normal human donors were purchased frozen from AllCells Inc.
  • the expansion/differentiation procedure comprises 3 stages.
  • thawed CD34+ erythroid precursors were cultured in Iscove’s MDM medium comprising recombinant human insulin, human transferrin, recombinant human recombinant human stem cell factor, and recombinant human interleukin 3.
  • erythroid cells were cultured in Iscove’s MDM medium supplemented with recombinant human insulin, human transferrin, human recombinant stem cell factor, human recombinant erythropoietin, and L-glutamine.
  • erythroid cells were cultured in Iscove’s MDM medium supplemented with human transferrin, recombinant human insulin, human recombinant erythropoietin, and heparin.
  • the cultures were maintained at 37°C in 5% C02 incubator.
  • Erythroid precursor cells were transduced during step 1 of the culture process described above. Erythroid cells in culturing medium were combined with lentiviral supernatant and polybrene. Infection was achieved by spinoculation, spinning the plate at 2000 rpm for 90 minutes at room temperature. After spinoculation, the cells were incubated at 37°C overnight.
  • Binding of a PE-labelled anti-IL-15-RA antibody (e.g., anti-IL-15RA antibody (JM7A4) (ab91270), AbCam) was used to validate expression of the IL-15/IL-15-RA in the engineered erythroid cells.
  • Binding of a PE-labelled anti-4-lBBL antibody e.g., purified anti-human 4-1BB Ligand (CD137L) antibody, BioLegend
  • Binding of the antibody was measured by flow cytometry for PE fluorescence. A gate was set based on stained untransduced cells.
  • Example 4 Enucleated erythroid cells genetically engineered to express IL- 15/IL-15RA and 4-1BBL result in an increase in lymphocytes, NKp30-positive lymphocytes, and CD8 + T-cells in vitro
  • Erythroid cells comprising IL-15/IL-15RA and 4-1BBL were prepared generally as described in Example 3.
  • PBMCs peripheral blood mononuclear cells
  • PBMCs were co-cultured with either: (a) enucleated erythroid cells comprising a first exogenous protein comprising (i) IL-15 or a functional fragment thereof, and (ii) IL-15 receptor alpha or a functional fragment thereof, and a second exogenous polypeptide comprising 4-1BBL, on their extracellular surface (200,000 cells per well); (b) recombinant human IL-15 (“rhIL15”; 0.1 ng/mL) and an anti-4-lBB agonistic antibody (1 nM) cross linked with a secondary antibody (2.5 nM), or (c) media only (control).
  • Example 5 Cell surface markers in fresh whole blood at multiple timepoints in patients prior to treatment and during treatment with enucleated erythroid cells genetically engineered to express IL-15/IL-15RA and 4-1BBL
  • Cell surface markers in fresh whole blood were evaluated by flow cytometry at multiple timepoints in patients prior to treatment and during treatment with erythroid cells comprising IL-15/IL-15RA and 4-1BBL prepared generally as described in Example 3.
  • Whole blood was collected from patients at baseline prior to treatment and post-treatment at 4 hours, 1 day, 2 days, 7 days, and 14 days for the first dosing cycle and 2 days, 7 days, and 14 days post-treatment after subsequent dosing cycles.
  • Whole blood was stained with fluorophore-conjugated antibodies using standard techniques for analysis by flow cytometry. Multiple antibody panels were used, with separate tubes for each panel and flow cytometric analysis. The panels included markers to identify lymphocyte, NK cell, and T cell populations along with markers of activation state.
  • Panel 1 The described data were obtained with the following panels: Panel 1 - CD3, CD8, CD16/CD56, CD45, CD45RA, CCR7, HLA-DR, and live/dead stain; Panel 2 - CD3, CD4, Foxp3, CD25, CD127, Ki67, live/dead stain; Panel 3 - CD45, CD16, CD56, NKG2D, NKp30, live/dead stain).
  • the data were then analyzed for frequency of population as a percent of the established parent gate (e.g., %NKp30+ of CD45+CD56+ live lymphocytes).
  • data were also analyzed for quantitative cell population numbers per volume of blood on the basis of an included quantitative count control. Data are reported as maximum-fold change observed relative to baseline for each parameter of interest (cells per pL whole blood or % of parent population).
  • CD45 + live lymphocytes within fresh whole blood were evaluated by flow cytometry at multiple timepoints at baseline (pre treatment) and post-treatment.
  • NKp30 (FIG. 2A), an increase in the percentage of CD45 + CD56dimCD16 + lymphocytes that are positive for NKp30 (FIG. 2B), and an increase in the percentage of CD45 + CD56 + CD16 + lymphocytes that are positive for NKp30 (FIG. 2C).
  • Patients dosed with >1 x 10 10 erythroid cells comprising IL-15/IL-15RA and 4-1BBL prepared generally as described in Example 3 had an increase in the number of circulating CD3 CD16/56 + cells (FIG. 3A) and an increase in CD8 + memory T cells expressing granzyme B (GrB + CD8 + CD45RA)(FIG. 3B).

Landscapes

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

Abstract

La présente invention concerne des méthodes d'augmentation du nombre de lymphocytes NKp30-positifs chez un sujet dont l'état le nécessite, ainsi que leurs utilisations.
PCT/US2022/011582 2021-01-08 2022-01-07 Méthodes d'augmentation de lymphocytes nkp30-positifs chez un sujet et leurs utilisations WO2022150569A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163135559P 2021-01-08 2021-01-08
US63/135,559 2021-01-08

Publications (1)

Publication Number Publication Date
WO2022150569A1 true WO2022150569A1 (fr) 2022-07-14

Family

ID=80448430

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/011582 WO2022150569A1 (fr) 2021-01-08 2022-01-07 Méthodes d'augmentation de lymphocytes nkp30-positifs chez un sujet et leurs utilisations

Country Status (2)

Country Link
TW (1) TW202241471A (fr)
WO (1) WO2022150569A1 (fr)

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4327710A (en) 1980-06-18 1982-05-04 The United States Of America As Represented By The Secretary Of Agriculture Process for encapsulating additives in resealed erythrocytes for disseminating chemicals via the circulatory system
US5753221A (en) 1991-06-14 1998-05-19 Communaute Economique Europeene Transformed erythrocytes, process for preparing the same, and their use in pharmaceutical compositions
US6139836A (en) 1997-05-05 2000-10-31 Dideco, S.P.A. Method of encapsulating biologically active agents within erythrocytes, and apparatus therefor
US6495351B2 (en) 2000-02-08 2002-12-17 Gendel Limited Loading system and method for using the same
WO2014183071A2 (fr) 2013-05-10 2014-11-13 Whitehead Institute For Biomedical Research Production in vitro de globules rouges avec des protéines pouvant être médiées par une sortase
WO2014183066A2 (fr) 2013-05-10 2014-11-13 Whitehead Institute For Biomedical Research Modification protéique de cellules vivantes à l'aide de sortase
WO2015073587A2 (fr) 2013-11-18 2015-05-21 Rubius Therapeutics, Inc. Complexes membrane synthétique- récepteur
WO2015153102A1 (fr) 2014-04-01 2015-10-08 Rubius Therapeutics, Inc. Méthodes et compositions d'immunomodulation
WO2017123646A1 (fr) 2016-01-11 2017-07-20 Rubius Therapeutics, Inc. Compositions et méthodes associées à des systèmes cellulaires thérapeutiques multimodaux pour des indications du cancer
US10046009B2 (en) 2014-02-12 2018-08-14 Erytech Pharma Method of treatment using a pharmaceutical composition comprising erythrocytes encapsulating a PLP-dependent enzyme and its cofactor
WO2019173798A1 (fr) 2018-03-08 2019-09-12 Rubius Therapeutics, Inc. Systèmes cellulaires thérapeutiques et méthodes de traitement du cancer et de maladies infectieuses
WO2020219909A1 (fr) 2019-04-26 2020-10-29 Rubius Therapeutics, Inc. Compositions tamponnées comprenant des cellules érythroïdes énucléées
WO2020243006A1 (fr) 2019-05-24 2020-12-03 Rubius Therapeutics, Inc. Procédés de génération de cellules érythroïdes énucléées

Patent Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4327710A (en) 1980-06-18 1982-05-04 The United States Of America As Represented By The Secretary Of Agriculture Process for encapsulating additives in resealed erythrocytes for disseminating chemicals via the circulatory system
US5753221A (en) 1991-06-14 1998-05-19 Communaute Economique Europeene Transformed erythrocytes, process for preparing the same, and their use in pharmaceutical compositions
US6139836A (en) 1997-05-05 2000-10-31 Dideco, S.P.A. Method of encapsulating biologically active agents within erythrocytes, and apparatus therefor
US6495351B2 (en) 2000-02-08 2002-12-17 Gendel Limited Loading system and method for using the same
US20160082046A1 (en) 2013-05-10 2016-03-24 Whitehead Institute For Biomedical Research In vitro production of red blood cells with sortaggable proteins
WO2014183066A2 (fr) 2013-05-10 2014-11-13 Whitehead Institute For Biomedical Research Modification protéique de cellules vivantes à l'aide de sortase
WO2014183071A2 (fr) 2013-05-10 2014-11-13 Whitehead Institute For Biomedical Research Production in vitro de globules rouges avec des protéines pouvant être médiées par une sortase
US10260038B2 (en) 2013-05-10 2019-04-16 Whitehead Institute For Biomedical Research Protein modification of living cells using sortase
WO2015073587A2 (fr) 2013-11-18 2015-05-21 Rubius Therapeutics, Inc. Complexes membrane synthétique- récepteur
US10046009B2 (en) 2014-02-12 2018-08-14 Erytech Pharma Method of treatment using a pharmaceutical composition comprising erythrocytes encapsulating a PLP-dependent enzyme and its cofactor
WO2015153102A1 (fr) 2014-04-01 2015-10-08 Rubius Therapeutics, Inc. Méthodes et compositions d'immunomodulation
WO2017123646A1 (fr) 2016-01-11 2017-07-20 Rubius Therapeutics, Inc. Compositions et méthodes associées à des systèmes cellulaires thérapeutiques multimodaux pour des indications du cancer
WO2019173798A1 (fr) 2018-03-08 2019-09-12 Rubius Therapeutics, Inc. Systèmes cellulaires thérapeutiques et méthodes de traitement du cancer et de maladies infectieuses
US20190298769A1 (en) 2018-03-08 2019-10-03 Rubius Therapeutics, Inc. Therapeutic cell systems and methods for treating cancer and infectious diseases
WO2020219909A1 (fr) 2019-04-26 2020-10-29 Rubius Therapeutics, Inc. Compositions tamponnées comprenant des cellules érythroïdes énucléées
WO2020243006A1 (fr) 2019-05-24 2020-12-03 Rubius Therapeutics, Inc. Procédés de génération de cellules érythroïdes énucléées

Non-Patent Citations (32)

* Cited by examiner, † Cited by third party
Title
"Antibody Engineering: Methods & Protocols", 2004, HUMANA PRESS, pages: 576
"NCBI", Database accession no. NP _001291377.1
AGARWAL ET AL.: "Blood group phenotype frequencies in blood donors from a tertiary care hospital in north India", BLOOD RES, vol. 48, no. 1, 2013, pages 51 - 54
ALDERSON ET AL., EUR. J. IMMUNOL., vol. 24, 1994, pages 2219 - 2227
ANDRE ET AL., CELL, vol. 175, no. 7, 2018, pages 1731 - 1743
ANDRÉ PASCALE ET AL: "Anti-NKG2A mAb Is a Checkpoint Inhibitor that Promotes Anti-tumor Immunity by Unleashing Both T and NK Cells", CELL, vol. 175, no. 7, 2018, pages 1731, XP085560732, ISSN: 0092-8674, DOI: 10.1016/J.CELL.2018.10.014 *
ANNE-SOPHIE DUGAST ET AL: "American Association for Cancer Research /March 29 ? April RTX-240, an Allogeneic Red Cell Therapeutic Expressing 4-1BBL and IL-15TP, Exhibits Potent In Vitro and In Vivo Activity and a Favorable Preclinical Safety Profile", 2 April 2019 (2019-04-02), XP055752602, Retrieved from the Internet <URL:https://www.rubiustx.com/wp-content/uploads/RTX-240-Dugast-AACR-Poster_FINAL.pdf> [retrieved on 20201120] *
BETTINGER ET AL., NUCLEIC ACIDS RES., vol. 29, 2001, pages 3882 - 3891
BIERHUIZEN ET AL., LEUKEMIA, vol. 13, 1999, pages 605 - 613
CHANG ET AL., FRONT. IMMUNOL., vol. 9, 2019, pages 31521
CHEN YING ET AL: "The B7 Family Member B7-H6: a New Bane of Tumor", PATHOLOGY ONCOLOGY RESEARCH, TUD. KIADO, BUDAPEST, HU, vol. 24, no. 4, 31 October 2017 (2017-10-31), pages 717 - 721, XP036586919, ISSN: 1219-4956, [retrieved on 20171031], DOI: 10.1007/S12253-017-0357-5 *
CLINICALTRIALS.GOV: "RTX-240 Monotherapy and in Combination With Pembrolizumab", 4 May 2020 (2020-05-04), XP055915285, Retrieved from the Internet <URL:https://www.clinicaltrials.gov/ct2/show/NCT04372706?term=RTX-240&draw=2&rank=1> [retrieved on 20220425] *
DUGAST ANNE-SOPHIE ET AL: "144?RTX-240, an allogeneic engineered red blood cell expressing 4-1BBL and IL-15TP, promotes NK cell functionality in vitro and in vivo", REGULAR AND YOUNG INVESTIGATOR AWARD ABSTRACTS, 1 November 2020 (2020-11-01), pages A87.1 - A87, XP055915271, DOI: 10.1136/jitc-2020-SITC2020.0144 *
GERRY KWOK ET AL: "Pembrolizumab (Keytruda)", HUMAN VACCINES & IMMUNOTHERAPEUTICS, vol. 12, no. 11, 4 June 2016 (2016-06-04), US, pages 2777 - 2789, XP055609923, ISSN: 2164-5515, DOI: 10.1080/21645515.2016.1199310 *
HASSLE ET AL., NEWS PHYSIOL. SCI., vol. 17, 2002, pages 87 - 92
KELLER ET AL., GENE THERAPY, vol. 6, 1999, pages 931 - 938
MALIK ET AL., BLOOD, vol. 91, 1998, pages 2664 - 2671
MITRA ET AL.: "Blood groups systems", INDIAN J. ANAESTH., vol. 58, no. 5, 2014, pages 524 - 528
OLDAK ET AL., ACTA BIOCHIM. POLONICA, vol. 49, 2002, pages 625 - 632
OSTEN ET AL., HEP, vol. 178, 2007, pages 177 - 202
PAPAPETROU ET AL., GENE THERAPY, vol. 12, 2005, pages S118 - S130
ROBINSONSAUER, PROC. NAT'L. ACAD. SCI. U.S.A., vol. 95, 1998, pages 5929 - 34
SELIGER ET AL., ONCOTARGET, vol. 7, no. 41, 2016, pages 67360 - 67372
SHIN ET AL., BIOCHIM. BIOPHYS. ACTA, vol. 1725, 2005, pages 377 - 384
TAO ET AL., STEM CELLS, vol. 25, 2007, pages 670 - 678
VAN TENDELOO ET AL., BLOOD, vol. 98, no. l, 2001, pages 49 - 56
VANDERBYL ET AL., EXP. HEMATOL., vol. 33, 2005, pages 1470 - 1476
VERMA ET AL., GENE THERAPY, vol. 5, 1998, pages 692 - 699
WEI ET AL., J IMMUNOL., vol. 167, 2001, pages 277 - 282
ZAKERI ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 109, 2012, pages E690 - E697
ZAKERIHOWARTH, JACS, vol. 132, 2010, pages 4526
ZHANG ET AL., J. AM. CHEM. SOC., 2013

Also Published As

Publication number Publication date
TW202241471A (zh) 2022-11-01

Similar Documents

Publication Publication Date Title
US20220169699A1 (en) Pd-1-cd28 fusion proteins and their use in medicine
KR20200140279A (ko) 암 및 감염성 질환을 치료하기 위한 치료 세포 시스템 및 방법
AU2017250769A1 (en) Transgenic T cell and chimeric antigen receptor T cell compositions and related methods
KR20190114966A (ko) 조작된 자연 살해 세포 및 이의 용도
KR20210029707A (ko) 재조합 수용체를 발현하는 세포의 생산 방법 및 관련 조성물
AU2019275479B2 (en) Chimeric antigen receptors with modified linker domains and uses thereof
CA3138137A1 (fr) Compositions tamponnees comprenant des cellules erythroides enucleees
US20200172597A1 (en) Artificial antigen presenting cells including hla-e and hla-g molecules and methods of use
CN115243713A (zh) 用于递送修饰的淋巴细胞聚集体的方法和组合物
US20200255864A1 (en) Methods and compositions for genetically modifying and expanding lymphocytes and regulating the activity thereof
CA3162272A1 (fr) Procedes d&#39;activation et d&#39;expansion de lymphocytes infiltrant les tumeurs
US20210317408A1 (en) Methods and compositions for genetically modifying lymphocytes in blood or in enriched pbmcs
WO2022150569A1 (fr) Méthodes d&#39;augmentation de lymphocytes nkp30-positifs chez un sujet et leurs utilisations
WO2022197548A1 (fr) Méthodes d&#39;augmentation des lymphocytes nkg2d-positifs chez un sujet et utilisations correspondantes
KR20230167063A (ko) T-세포 활성화 방법
US20210293787A1 (en) Combined invasion and cytotoxicity assay using chemokine secreting target cells
WO2022150578A1 (fr) Méthodes de traitement d&#39;une tumeur chez un patient humain
WO2022256646A1 (fr) Méthodes de traitement d&#39;un cancer associé à hpv16 ou positif au hpv16 chez un sujet
US20210147802A1 (en) Methods of generating enucleated erythroid cells using taurine or hypotaurine
US20210130780A1 (en) Methods of generating enucleated erythroid cells using myo-inositol
JP2024073636A (ja) Pd-1-cd28融合タンパク質および医療におけるその使用

Legal Events

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

Ref document number: 22705487

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22705487

Country of ref document: EP

Kind code of ref document: A1