WO2021113849A1 - Her2+ cancer treatment with populations of natural killer cells comprising a cleavage resistant cd16 - Google Patents

Her2+ cancer treatment with populations of natural killer cells comprising a cleavage resistant cd16 Download PDF

Info

Publication number
WO2021113849A1
WO2021113849A1 PCT/US2020/063670 US2020063670W WO2021113849A1 WO 2021113849 A1 WO2021113849 A1 WO 2021113849A1 US 2020063670 W US2020063670 W US 2020063670W WO 2021113849 A1 WO2021113849 A1 WO 2021113849A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
group
population
alkyl
substituted
Prior art date
Application number
PCT/US2020/063670
Other languages
French (fr)
Inventor
Irene RAITMAN KHUTORSKOY
Salvatore ROTONDO
Tanel MAHLAKÕIV
Xuan GUO
Andrea DIFIGLIA
Hemlata RANA
Qian Ye
Srinivas SOMANCHI
William VAN DER TOUW
Lin KANG
Shuyang He
Joseph Gleason
Valentina ROUSSEVA
Bhavani Stout
James Mccauley
Xiaokui Zhang
Robert J. Hariri
Original Assignee
Celularity 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 Celularity Inc. filed Critical Celularity Inc.
Publication of WO2021113849A1 publication Critical patent/WO2021113849A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464403Receptors for growth factors
    • A61K39/464406Her-2/neu/ErbB2, Her-3/ErbB3 or Her 4/ ErbB4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70535Fc-receptors, e.g. CD16, CD32, CD64 (CD2314/705F)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0646Natural killers cells [NK], NKT cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/48Blood cells, e.g. leukemia or lymphoma
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/51Stomach
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/515CD3, T-cell receptor complex
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/30Coculture with; Conditioned medium produced by tumour cells

Definitions

  • NK cells and/or ILC3 cells are methods of producing populations of natural killer (NK) cells and/or ILC3 cells from a population of hematopoietic stem or progenitor cells in media comprising stem cell mobilizing factors, e.g., three-stage methods of producing NK cells and/or ILC3 cells in media comprising stem cell mobilizing factors starting with hematopoietic stem or progenitor cells from cells of the placenta, for example, from placental perfusate (e.g., human placental perfusate) or other tissues, for example, umbilical cord blood or peripheral blood.
  • placental perfusate e.g., human placental perfusate
  • other tissues for example, umbilical cord blood or peripheral blood.
  • NK cells and/or ILC3 cells and/or NK progenitor cells described herein are methods of using the placental perfusate, the NK cells and/or ILC3 cells and/or NK progenitor cells described herein, to, e.g., suppress the proliferation of tumor cells, including multiple myeloma and acute myeloid leukemia cells.
  • NK cells exhibit innate anti-tumor activity owing to the expression of a multitude of activating and inhibitory receptors that orchestrate NK cell responses. It is thus possible to use NK cells from allogeneic sources without the risk of graft-vs-host disease 1 , making them very attractive for developing “off-the-shelf’ cellular therapies.
  • the anti-tumor responses of NK cells can be further enhanced by expressing Chimeric Antigen Receptors (CARs).
  • CARs Chimeric Antigen Receptors
  • Celularity has developed a GMP process for generating off-the-shelf, allogeneic human Placental Hematopoietic Stem Cell (HSC) derived Natural Killer cells (PNK).
  • HSC Human Placental Hematopoietic Stem Cell
  • PNK Natural Killer cells
  • the present invention provides methods of suppressing the proliferation of tumor cells comprising contacting the tumor cells with a population of placental-derived natural killer cells comprising a cleavage resistant CD 16 and an antibody, wherein the tumor cells are HER2+, and wherein the antibody is an anti-HER2 antibody.
  • the present invention also provides methods of treating a HER2+ cancer in a subject, comprising administering to the subject a population of placental-derived natural killer cells comprising a cleavage resistant CD 16 and an antibody, wherein the antibody is an anti-HER2 antibody.
  • the placental-derived natural killer (NK) cells are CYNK cells. In one or more embodiments of the invention the CYNK cells are placental CD34+ cell-derived natural killer (NK) cells.
  • the CYNK cells are characterized by expression of one or more markers selected from the group consisting of FGFBP2, GZMH, CCL3L3, GZMM, CXCR4, ZEB2, KLF2, LITAF, RORA, LYAR, CNOT1, IFNG, DUSP2, ATG2A, CD7, PMAIP1, PPP2R5C, NR4A2, ZFP36L2, PIK3R1, KLRFl, SNHG9, MT2A, RGS2, CHD1, DUSP1, EML4, ZFP36, ZC3H12A, DNAJB6, SBDS, IRFl, TSC22D3, TSPYL2, PNRC1, ISCA1, JUNB, WHAMM, RICTOR, TNFAIP3, EPC1, MVD, CLK1, ARL4C, REL, KMT2E, YPEL5, AMD1, BTG2, and IDS which is lower than expression of said markers in peripheral blood natural killer cells and
  • the CYNK cells are characterized by expression of one or more markers selected from the group consisting of FGFBP2, GZMH, CCL3L3, GZMM, CXCR4, ZEB2, KLF2, LITAF, RORA, LYAR, CNOT1, IFNG, DUSP2, ATG2A, CD7, PMAIP1, PPP2R5C, NR4A2, ZFP36L2, PIK3R1, KLRFl, SNHG9, MT2A, RGS2, CHD1, DUSP1, EML4, ZFP36, ZC3H12A, DNAJB6, SBDS, IRFl, TSC22D3, TSPYL2, PNRC1, ISCA1, JUNB, WHAMM, RICTOR, TNFAIP3, EPC1, MVD, CLK1, ARL4C, REL, KMT2E, YPEL5, AMD1, BTG2, and IDS which is lower than expression of said markers in peripheral blood natural killer cells.
  • markers selected from the group consisting of FGF
  • the CYNK cells are characterized by expression of one or more markers selected from the group consisting of NDFIP2, LINC00996, MAL, CCL1, MB, SPINK2, C15orf48, CAMK1, KLRC1, TNFSF10, TNFRSF18, IL32, CAPG, AC092580.4, S100A11, TNFRSF4, ENOl, FCER1G, CCND2, KRT81, MRPS6, ANXA2, PTGER2, GLOl, HAVCR2, PYCARD, LAT2, SLC16A3, COTL1, PKM, TALDOl, CD96, NCR3, KRT86, STMN1, LTB, ARPC1B, ARPC5, FKBP1A, TIMP1, GZMK, CD59, PGK1, RGS10, EVL, RAC2, LGALS1, ITGB7, TUBB, PGAM1, PRF1, GZMB, IL2RB, KLRC
  • a nucleic acid encoding the cleavage resistant CD 16 has been introduced into the NK cells by transfection. In one or more embodiments of the invention a nucleic acid encoding the cleavage resistant CD 16 has been introduced into the NK cells by transduction. In one or more embodiments of the invention a nucleic acid encoding the cleavage resistant CD 16 has been introduced into the NK cells by retroviral transduction. In one or more embodiments of the invention a nucleic acid encoding the cleavage resistant CD 16 has been introduced into the NK cells by lentiviral transduction. In one or more embodiments of the invention greater than about 60%, greater than about 70%, greater than about 80%, greater than about 90%, or greater than about 95% of the cells in the population are CD56+ and CD3-.
  • less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% of the cells in the population are CD3+.
  • less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% of the cells in the population are CD19+.
  • the population of cells comprises cells which express one or more surface markers selected from the group consisting of CD226, NKG2D, CD11a, NKp30, NKp44, NKp46, CD94, and combinations thereof
  • the population of cells exhibit greater antibody- dependent cellular cytotoxicity than a population of placental-derived natural killer cells lacking expression of the cleavage resistant CD 16.
  • said contacting is contacting in vitro. In one or more embodiments of the invention said contacting is contacting in vivo. In one or more embodiments of the invention said contacting is in a human.
  • the cleavage resistant CD 16 comprises a valine residue at position 158. In one or more embodiments of the invention the cleavage resistant CD 16 comprises a proline residue at position 197. In one or more embodiments of the invention the cleavage resistant CD 16 is CD16VP. In one or more embodiments of the invention the cleavage resistant CD 16 does not comprise a proline residue at position 197.
  • the tumor cells are tumor cells from a cancer selected from the group consisting of Bladder cancers, Breast cancers, Cervical cancers, Cholangiocarcinomas (extrahepatic), Cholangiocarcinomas (intrahepatic), Colorectal cancers, Esophageal or esophagogastric junction cancers, Gallbladder cancers, Gastric adenocarcinomas, Gastrointestinal stromal tumors, Glioblastoma multiforme, high grade gliomas, Gliomas (low grade), Head and neck carcinomas, Hepatocellular carcinomas,
  • a cancer selected from the group consisting of Bladder cancers, Breast cancers, Cervical cancers, Cholangiocarcinomas (extrahepatic), Cholangiocarcinomas (intrahepatic), Colorectal cancers, Esophageal or esophagogastric junction cancers, Gallbladder cancers, Gastric adenocarcinomas, Gastrointestinal stromal tumors,
  • Intestinal (small) malignancies Kidney cancers, Lung cancers (non small cells), Lung cancers (small cells), Melanomas, Melanomas (uveal), Neuroendocrine tumors, Oligodendrogliomas, Ovarian (epithelial) cancers, Ovarian (non-epithelial) cancers, Pancreatic adenocarcinomas, Penile cancers, Pituitary cancers, Prostate cancers, Sarcomas (peritoneal, retroperitoneal), Sarcomas (soft tissues), Solitary fibrous tumors, Testicular cancers, Thymic cancers, Thyroid cancers, Uterine cancers, and combinations thereof.
  • the tumor cells are gastric cancer cells.
  • the anti-HER2 antibody is N-HER2 antibody
  • the population of placental- derived natural killer cells comprising a cleavage resistant CD 16 and the anti-HER2 antibody are administered sequentially. In one or more embodiments of the invention the population of placental-derived natural killer cells comprising a cleavage resistant CD 16 and the anti-HER2 antibody are administered concurrently.
  • CYNK cells are prepared by the methods presented herein.
  • the present invention also provides a population of human placental-derived natural killer cells comprising a cleavage resistant CD16 for use in the manufacture of a medicament for treatment of a HER2+ cancer in a subject.
  • the present invention also provides the use of a composition comprising a population of human placental-derived natural killer cells comprising a cleavage resistant CD 16 for treatment of a HER2+ cancer in a subject.
  • the population of human placental-derived natural killer cells is a population of cells of the invention.
  • the present invention also provides methods of determining a patient’s reactivity to a given cell therapy treatment, the method comprising, obtaining a population of T cells from said patient, sequencing a portion of the T cell receptor repertoire of said T cells, obtaining a population of DCs from the donor from which said cell therapy was derived, contacting said T cells with said DCs in an MLR to encourage outgrowth of T cells which are reactive to said cell therapy, sequencing a portion of the T cell receptor repertoire of said reactive T cells, comparing the T cell receptor repertoire of said reactive T cells to the T cell receptor repertoire of the T cells obtained from the patient, and identifying the portion of the patients T cell receptor repertoire which is potentially reactive to the cell therapy.
  • CYNK are CD34+ cell-derived NK cells produced by the methods described herein.
  • CYNK cells are placental-deived NK cells.
  • CYNK-001 and CYNK-101 are specific populations / formulations of CYNK cells.
  • immunomodulatory compound and “IMiDTM” do not encompass thalidomide.
  • “lenalidomide” means 3-(4'aminoisoindoline-r-one)-l- piperidine-2,6-dione (Chemical Abstracts Service name) or 2,6-Piperidinedione,3-(4-amino- l,3-dihydro-l-oxo-2H-isoindol-2-yl)- (International Union of Pure and Applied Chemistry (IUPAC) name).
  • “pomalidomide” means 4-amino-2-(2,6-dioxopiperidin-3- yl)isoindole- 1 ,3 -dione.
  • multipotent when referring to a cell, means that the cell has the capacity to differentiate into a cell of another cell type.
  • a multipotent cell is a cell that has the capacity to grow into a subset of the mammalian body's approximately 260 cell types. Unlike a pluripotent cell, a multipotent cell does not have the capacity to form all of the cell types.
  • feeder cells refers to cells of one type that are co-cultured with cells of a second type, to provide an environment in which the cells of the second type can be maintained, and perhaps proliferate.
  • feeder cells can provide, for example, peptides, polypeptides, electrical signals, organic molecules (e.g., steroids), nucleic acid molecules, growth factors (e.g., bFGF), other factors (e.g., cytokines), and metabolic nutrients to target cells.
  • feeder cells grow in a mono- layer.
  • natural killer cells or “NK cells” produced using the methods described herein, without further modification, include natural killer cells from any tissue source.
  • the “ILC3 cells” produced using the methods described herein, without further modification, include ILC3 cells from any tissue source.
  • placental perfusate means perfusion solution that has been passed through at least part of a placenta, e.g., a human placenta, e.g., through the placental vasculature, and includes a plurality of cells collected by the perfusion solution during passage through the placenta.
  • tumor cell suppression includes slowing the growth of a population of tumor cells, e.g., by killing one or more of the tumor cells in said population of tumor cells, for example, by contacting or bringing, e.g., NK cells or an NK cell population produced using a three-stage method described herein into proximity with the population of tumor cells, e.g., contacting the population of tumor cells with NK cells or an NK cell population produced using a three- stage method described herein.
  • said contacting takes place in vitro or ex vivo. In other embodiments, said contacting takes place in vivo.
  • hematopoietic cells includes hematopoietic stem cells and hematopoietic progenitor cells.
  • the “undefined component” is a term of art in the culture medium field that refers to components whose constituents are not generally provided or quantified.
  • examples of an “undefined component” include, without limitation, serum, for example, human serum (e.g., human serum AB) and fetal serum (e.g., fetal bovine serum or fetal calf serum).
  • “+”, when used to indicate the presence of a particular cellular marker, means that the cellular marker is detectably present in fluorescence activated cell sorting over an isotype control; or is detectable above background in quantitative or semi- quantitative RT-PCR.
  • cellular marker when used to indicate the presence of a particular cellular marker, means that the cellular marker is not detectably present in fluorescence activated cell sorting over an isotype control; or is not detectable above background in quantitative or semi- quantitative RT-PCR.
  • FIG. 1 shows expansion of NK cells for compounds CRLl - CRL11.
  • FIG. 2 shows expansion of NK cells for compounds CRL12 - CRL22.
  • FIG. 3 shows expansion of NK cells relative to SRI positive control.
  • FIG. 4 shows expansion of CD34+ cells from which the NK cells were derived.
  • FIGS. 5A - 5B show cytotoxicity of the expanded NK cultures.
  • FIGS. 6 A - 6C show that PNK cells highly express genes encoding the cytotoxic machinery.
  • FIG. 6A CYNK cells were combined with peripheral blood derived NK cells (PB-NK) at 1:1 ratio and gene expression analyzed on single cell level using 10X Genomics Chromium platform and Illumina sequencing. Bioinformatics analysis utilized 10X Genomics Cell Ranger analysis pipeline. Transcript analysis was restricted to Granzyme B (GZMB) expressing cells.
  • FIG. 6B A representative tSNE plot depicting PNK and PB-NK cells as distinct populations.
  • FIG. 6C tSNE plots of selected NK cell-associated genes. The data is representative of two donors.
  • FIG. 7 shows that PNK and PB-NK cells differentially express genes encoding
  • FIG. 8 shows the gating strategy for PB-NK and CYNK cells.
  • PBMC cells were thawed and stained with fluorophore-coupled antibodies targeting NK cell receptors.
  • the figure demonstrates representative dot plots and the gating strategy for the identification of CYNK and PB-NK cells. See FIG. 9 for further characterization of the populations.
  • FIG. 9 shows differential expression of surface proteins on CYNK and PB-NK cells. CYNK and PB-NK cells were pre-gated as indicated in FIG. 8.
  • FIG. 10 shows that CYNK cells form a distinct cell population from PB-NK cells based on surface protein expression.
  • tSNE plots demonstrating differential clustering of CYNK and PB-NK cells based on their surface markers.
  • tSNE plots were generated of flow cytometry data using FlowJo software.
  • FIG. 11 A shows a schema of placental CD34+ cells expanded and differentiated to NK cells.
  • FIG. 1 ID shows examples of phenotypic characterization of CD16VP cells.
  • FIG. 12A shows expression of CD 16 on non transduced cells (NT) and CD 16
  • FIG 13B shows ADCC of CD 16 VP cells before and after PMA/ionomycin treatment against Daratumumab opsonized Daudi cells.
  • FIG. 14A shows a schematic study plan to test in vivo ADCC activities of
  • FIG 14B shows bioluminescence images of mice from each group after 10 days of cell infusion.
  • FIG. 14D shows survival of mice through the course of the study.
  • FIGS. 15A - 15B show expression of cell surface markers on CYNK CD16-
  • FIG. 16 shows expression of CD 16 on CYNK CD16-VP cells.
  • FIG. 17 shows resistance to shedding of CD 16 on activated CYNK CD16-VP cells from multiple donors.
  • FIG. 18 shows a schema of CD 16 mediated lysis of opsonized tumor cell.
  • FIG. 19 shows a schema of CD16 with the high affinity (158V) modification and an exemplary shedding resistance (197P) modification.
  • FIG. 20 shows a study schema of an in vivo efficacy and safety study.
  • FIGS. 21 A and 21 B show that CYNK- 101 cells exert enhanced cytotoxic activity in combination with Herceptin against HER2 overexpressing gastric tumor cell lines.
  • FIG. 22 shows CYNK-101 in combination with Herceptin specifically kill
  • FIG. 22A shows the expression of HER2 on NHDF, PBMC and NCI-N87.
  • FIG. 23 shows that CYNK-101 cells express high expression of CD16.
  • FIGS. 24A - 24B show that CYNK-101 is resistant to CD 16 shedding post
  • FIG. 24 A shows representative flow cytometry graphs showing CD 16 loss following cleavage on NT cells but cleavage resistance on CYNK-101 cells following 2h activation by PMAi.
  • FIGS. 25 A - 25B show that CYNK-101 cells exert enhanced cytotoxic activity in combination with Herceptin against HER2+ breast cancer cell lines.
  • FIGS. 25A - 25B shows the average cytotoxic activity of CYNK-101 cells in combination with Herceptin or a control IgG against the indicated breast cancer cell lines, AU565, BT-474, HCC-1954, SK-BR-3 and ZR-75-30.
  • the error bars represent the SD from the mean calculated from seven different CYNK-101 donors (unless specified). Cytotoxicity from Herceptin or IgG alone is included for reference. * indicate significantly higher activity of CYNK-101 in combination with Herceptin compared to that of IgG (***p ⁇ 0.001, **p ⁇ 0.005, and *p ⁇ 0.05).
  • FIG. 26 shows increased IFN-g production of CYNK-101 in combination with
  • FIG. 26 shows the average of IFN-g production of target cell alone, CYNK-101 alone, or CYNK-101 coculture with target cells in the presence of Herceptin or IgG control after 24h incubation.
  • the error bars represent the SD from the mean calculated from different CYNK-101 donors (*p ⁇ 0.05).
  • FIG. 27 shows the purity of CYNK-101 enriched from mouse liver post injection.
  • FIG. 27 shows percent NK purity of the cells from the livers of NSG mice pre and post mouse cell depletion in ex vivo study. Representative data from total of three studies.
  • FIG. 28 shows ex vivo CYNK-101 phenotype characterization.
  • FIG. 28 shows
  • NK surface marker expression on the in vitro CYNK-101 donor (prior infusion CYNK-101) compared to the ex vivo CYNK-101.
  • FIG. 29 shows ex vivo CYNK-101 is resistant to PMAi stimulated CD 16 shedding.
  • FIG. 29 shows in vitro CYNK-101 and ex vivo CYNK-101 shedding resistance post PMAi stimulation: CD 16 expression on control, in vitro CYNK-101 and ex vivo CYNK- 101 without stimulation (top graphs). CD16 expression on control, in vitro CYNK-101 and ex vivo CYNK-101 stimulated by PMAi for 2h (bottom graphs).
  • FIG. 30 shows ex vivo CYNK-101 displays ADCC activity against NCI-N87 at E:T ratio of 0.5:1.
  • FIG. 30 shows cytotoxicity of ex vivo CYNK-101 or in vitro CYNK- 101 in combination with Herceptin against NCI-N87 at E:T ratio of 0.5:1.
  • CYNK-lOl+IgG, Herceptin alone or IgG alone served as control.
  • FIG. 31 shows cytokine secretion profiling of ex vivo CYNK-101 in combination with Herceptin against NCI-N87.
  • FIG. 31 shows cytokine secretion profiling of ex vivo CYNK-101 or in vitro CYNK-101 in combination with Herceptin against NCI-N87 at E:T ratio of 0.5:1.
  • FIG. 32 shows in vivo efficacy of CYNK-101 in combination with
  • FIG. 32 shows percentage of tumor volume change over Day 7 baseline. Animals were preconditioned with busulfan on Day -3. NCI-N87 cells were subcutaneously injected on Day 0. Trastuzumab was intraperitonially injected on Day 7. Vehicle or CYNK-101 was intravenously administered on Days 7, 14 and 21. The data are presented as the mean ⁇ SEM; Multiple t tests were performed to compare Trastuzumab with Vehicle and Trastuzumab with CYNK-101 + Trastuzumab. ** P ⁇ 0.01, *** P ⁇ 0.001, ****P ⁇ 0.0001.
  • FIG. 33 shows a model of identifying and tracking alloreactive T cell clones in patients.
  • FIG. 34 shows an overview of placental monocyte-derived DC one-way MLR assay.
  • FIG. 35 shows enrichment of CD33+ monocytes from placental blood.
  • CD33+ monocytes were isolated from cryopreserved placental blood mononuclear cells using antihuman CD33+ magnetic selection beads (Miltenyi) according to manufacturer’s instructions. Preselected, CD33+, and CD33- fractions were stained for both viability and CD33 purity.
  • FIGS. 36A - 36C shows that placental monocytes require additional stimulation to mature DCs. To generate antigen-presenting cells, placental CD33+ monocytes were matured to DC in presence of GM-CSF and IL-4. DCs were activated by LPS on day 6, or further matured with cytokines prior to LPS activation on day 7 where indicated.
  • FIG. 36A Upregulation of HLA-ABC and HLA-DR on maturing monocytes derived from placenta (FIG. 36A) or peripheral blood (FIG. 36B) is shown.
  • FIG. 36C Placental monocytes from two donors monitored for HLA-ABC and HLA-DR expression during course of GM- CSF+IL4 maturation (blue), additional cytokine restimulation (orange), and post-LPS stimulations (green).
  • FIG. 37 shows a purity check of responder CD3+ T cells.
  • CD3+ T cells isolated from representative unrelated cryopreserved PBMC donor using positive magnetic bead selection (Miltenyi).
  • FIGS. 38A - 38D show brightfield Microscopy of Expanding T Cell Clones.
  • FIG. 38A Images of PBMC donor 3994 T cell only control and FIG. 38B) placental donor 777 DC + self T cell control. Images show absence of T cell clonal expansion in both controls.
  • FIG. 38C Placental donor 777 DC co-cultured + responding T cell donor 3994 MLR.
  • FIG. 38D Control blood monocyte derived DC + responding T cell donor 3994 MLR.
  • FIGS. 39A - 39C show that MLR using placental DCs yields expected T cell cytokine profiles.
  • Four different placental donor monocyte- derived DCs were used in MLR with same allogeneic responding T cells isolated from one PBMC donor. Supernatants were collected at the end of the MLR. Multiplex analysis of cytokine production in MLRs were evaluated using Luminex FlexMAP3D custom plex kit cat# HCD8-MAG-15K with FIG. 39A) IFN-g, FIG. 39B) IL-2 and FIG. 39C) TNF-a analytes. No cytokine production was observed in T cell controls only.
  • FIGS. 40A - 40B show that MLR generates CD25+CD69+ proliferating T cells. Phenotype of CD4 and CD8 responding T cells prior to MLR (FIG. 40A), and after (FIG. 40B) MLR. 3 MLRs using T cell donor 5858 were used with stimulator DCs. Representative data from one MLR, 5856 DCs + 5858 T cells, is representative. CD4 and CD8 T cells are analyzed for CD25 and CD69 as markers of cell activation, as well as Ki-67 to assess active proliferation.
  • FIGS. 41A - 41B show that conserved T cell clones expand across multiple
  • NK cells and/or ILC3 cells are novel methods of producing and expanding NK cells and/or ILC3 cells from hematopoietic cells, e.g., hematopoietic stem cells or progenitor cells. Also provided herein are methods, e.g., three-stage methods, of producing NK cell populations and/or ILC3 cell populations from hematopoietic cells, e.g., hematopoietic stem cells or progenitor cells.
  • the hematopoietic cells used to produce the NK cells and/or ILC3 cells, and NK cell populations and/or ILC3 cell populations, may be obtained from any source, for example, without limitation, placenta, umbilical cord blood, placental blood, peripheral blood, spleen or liver.
  • the NK cells and/or ILC3 cells or NK cell populations and/or ILC3 cell populations are produced from expanded hematopoietic cells, e.g., hematopoietic stem cells and/or hematopoietic progenitor cells.
  • hematopoietic cells are collected from a source of such cells, e.g., placenta, for example from placental perfusate, umbilical cord blood, placental blood, peripheral blood, spleen, liver (e.g., fetal liver) and/or bone marrow.
  • placenta for example from placental perfusate, umbilical cord blood, placental blood, peripheral blood, spleen, liver (e.g., fetal liver) and/or bone marrow.
  • hematopoietic cells used to produce the NK cells and/or ILC3 cells, and
  • NK cell populations and/or ILC3 cell populations may be obtained from any animal species.
  • the hematopoietic stem or progenitor cells are mammalian cells.
  • said hematopoietic stem or progenitor cells are human cells.
  • said hematopoietic stem or progenitor cells are primate cells.
  • said hematopoietic stem or progenitor cells are canine cells.
  • said hematopoietic stem or progenitor cells are rodent cells.
  • Hematopoietic cells useful in the methods disclosed herein can be any hematopoietic cells able to differentiate into NK cells and/or ILC3 cells, e.g., precursor cells, hematopoietic progenitor cells, hematopoietic stem cells, or the like.
  • Hematopoietic cells can be obtained from tissue sources such as, e.g., bone marrow, cord blood, placental blood, peripheral blood, liver or the like, or combinations thereof.
  • Hematopoietic cells can be obtained from placenta. In a specific embodiment, the hematopoietic cells are obtained from placental perfusate.
  • the hematopoietic cells are not obtained from umbilical cord blood. In one embodiment, the hematopoietic cells are not obtained from peripheral blood. Hematopoietic cells from placental perfusate can comprise a mixture of fetal and maternal hematopoietic cells, e.g., a mixture in which maternal cells comprise greater than 5% of the total number of hematopoietic cells. In certain embodiments, hematopoietic cells from placental perfusate comprise at least about 90%, 95%, 98%, 99% or 99.5% fetal cells.
  • the hematopoietic cells e.g., hematopoietic stem cells or progenitor cells, from which the NK cell populations and/or ILC3 cell populations produced using a three-stage method described herein are produced, are obtained from placental perfusate, umbilical cord blood, fetal liver, mobilized peripheral blood, or bone marrow.
  • the hematopoietic cells e.g., hematopoietic stem cells or progenitor cells, from which the NK cell populations and/or ILC3 cell populations produced using a three-stage method described herein are produced, are combined cells from placental perfusate and cord blood, e.g., cord blood from the same placenta as the perfusate.
  • said umbilical cord blood is isolated from a placenta other than the placenta from which said placental perfusate is obtained.
  • the combined cells can be obtained by pooling or combining the cord blood and placental perfusate.
  • the cord blood and placental perfusate are combined at a ratio of 100:1, 95:5, 90:10, 85:15, 80:20, 75:25, 70:30, 65:35, 60:40, 55:45: 50:50, 45:55, 40:60, 35:65, 30:70, 25:75, 20:80, 15:85, 10:90, 5:95, 100:1, 95:1, 90:1, 85:1, 80:1, 75:1, 70:1, 65:1, 60:1, 55:1, 50:1, 45:1, 40:1, 35:1, 30:1, 25:1, 20:1, 15:1, 10:1, 5:1, 1:1, 1:5, 1:10, 1:15, 1:20, 1:25, 1:30, 1:35, 1:40, 1:45, 1:50, 1:55, 1:60, 1:65, 1:70, 1:75, 1:80, 1:85, 1:90, 1:95, 1 : 100, or the like by volume to obtain the combined cells.
  • the cord blood and placental perfusate are combined at a ratio of from 10:1 to 1:10, from 5:1 to 1:5, or from 3:1 to 1:3. In another specific embodiment, the cord blood and placental perfusate are combined at a ratio of 10: 1, 5:1, 3:1, 1:1, 1:3, 1:5 or 1:10. In amore specific embodiment, the cord blood and placental perfusate are combined at a ratio of 8.5:1.5 (85%: 15%).
  • the cord blood and placental perfusate are combined at a ratio of 100:1, 95:5, 90:10, 85:15, 80:20, 75:25, 70:30, 65:35, 60:40, 55:45: 50:50, 45:55, 40:60, 35:65, 30:70, 25:75, 20:80, 15:85, 10:90, 5:95, 100:1, 95:1, 90:1, 85:1, 80:1, 75:1, 70:1, 65:1, 60:1, 55:1, 50:1, 45:1, 40:1, 35:1, 30:1, 25:1, 20:1, 15:1, 10:1, 5:1, 1:1, 1:5, 1:10, 1:15, 1:20, 1:25, 1:30, 1:35, 1:40, 1:45, 1:50, 1:55, 1:60, 1:65, 1:70, 1:75, 1:80, 1:85, 1:90, 1:95, 1: 100, or the like by total nucleated cells (TNC) content to
  • the cord blood and placental perfusate are combined at a ratio of from 10:1 to 10:1, from 5:1 to 1:5, or from 3:1 to 1: 3. In another specific embodiment, the cord blood and placental perfusate are combined at a ratio of 10:1, 5:1, 3:1, 1:1, 1:3, 1:5 or 1:10.
  • the hematopoietic cells e.g., hematopoietic stem cells or progenitor cells from which said NK cell populations and/or ILC3 cell populations produced using a three-stage method described herein are produced, are from both umbilical cord blood and placental perfusate, but wherein said umbilical cord blood is isolated from a placenta other than the placenta from which said placental perfusate is obtained.
  • the hematopoietic cells are CD34 + cells.
  • the hematopoietic cells useful in the methods disclosed herein are CD34 + cells.
  • the hematopoietic cells are CD34 + CD38 Lin .
  • the hematopoietic cells are one or more of CD2- CD3 , CDllb , CDllc , CD 14-, CD 16 , CD19 , CD24 , CD56 , CD66b and/or glycophorin A “ .
  • the hematopoietic cells are CD2 . CD3 . CDllb , CDllc , CD 14 . CD 16 . CD 19 . CD24 . CD56 . CD66b and glycophorin A “ .
  • the hematopoietic cells are CD34 CD38 CD33 CD1 17 .
  • the hematopoietic cells are CD34 CD38 CD33 CD117 CD235 CD36 .
  • the hematopoietic cells are CD45 + .
  • the hematopoietic cells are CD34 + CD45 + .
  • the hematopoietic cell is Thy-1 + .
  • the hematopoietic cell is CD34 + Thy- 1 + .
  • the hematopoietic cells are CD133 + .
  • the hematopoietic cells are CD34 + CD133 + or CD133 + Thy-1 + .
  • the CD34 + hematopoietic cells are CXCR4 + .
  • the CD34 + hematopoietic cells are CXCR4 .
  • the hematopoietic cells are positive for KDR (vascular growth factor receptor 2).
  • the hematopoietic cells are CD34 + KDR + , CD133 + KDR + or Thy-1 + KDR + .
  • the hematopoietic cells are positive for aldehyde dehydrogenase (ALDH + ), e.g., the cells are CD34 + ALDH + .
  • the CD34 + cells are CD45 .
  • the CD34 + cells e.g., CD34 + , C D45 cells express one or more, or all, of the miRNAs hsa-miR-380, hsa-miR-512, hsa-miR-517, hsa-miR-518c, hsa-miR-519b, hsa-miR- 520a, hsa-miR-337, hsa-miR-422a, hsa-miR-549, and/or hsa-miR-618.
  • the hematopoietic cells are CD34 .
  • the hematopoietic cells can also lack certain markers that indicate lineage commitment, or a lack of developmental naivete.
  • the hematopoietic cells are HLA-DR .
  • the hematopoietic cells are CD34 + HLA-DR , CD133 HLA-DR .
  • Thy-1 + HLA-DR or ALDH 1 HLA-DR In another embodiment, the hematopoietic cells are negative for one or more, or all, of lineage markers CD2, CD3, CDllb, CDllc, CD14, CD16, CD19, CD24, CD56, CD66b and glycophorin A.
  • hematopoietic cells can be selected for use in the methods disclosed herein on the basis of the presence of markers that indicate an undifferentiated state, or on the basis of the absence of lineage markers indicating that at least some lineage differentiation has taken place. Methods of isolating cells, including hematopoietic cells, on the basis of the presence or absence of specific markers is discussed in detail below.
  • Hematopoietic cells used in the methods provided herein can be a substantially homogeneous population, e.g., a population comprising at least about 95%, at least about 98% or at least about 99% hematopoietic cells from a single tissue source, or a population comprising hematopoietic cells exhibiting the same hematopoietic cell-associated cellular markers.
  • the hematopoietic cells can comprise at least about 95%, 98% or 99% hematopoietic cells from bone marrow, cord blood, placental blood, peripheral blood, or placenta, e.g., placenta perfusate.
  • Hematopoietic cells used in the methods provided herein can be obtained from a single individual, e.g., from a single placenta, or from a plurality of individuals, e.g., can be pooled. Where the hematopoietic cells are obtained from a plurality of individuals and pooled, the hematopoietic cells may be obtained from the same tissue source. Thus, in various embodiments, the pooled hematopoietic cells are all from placenta, e.g., placental perfusate, all from placental blood, all from umbilical cord blood, all from peripheral blood, and the like.
  • placenta e.g., placental perfusate, all from placental blood, all from umbilical cord blood, all from peripheral blood, and the like.
  • Hematopoietic cells used in the methods disclosed herein can, in certain embodiments, comprise hematopoietic cells from two or more tissue sources.
  • a plurality of the hematopoietic cells used to produce natural killer cells using a three-stage method described herein comprise hematopoietic cells from placenta, e.g., placenta perfusate.
  • the hematopoietic cells used to produce NK cell populations and/or ILC3 cell populations produced using a three-stage method described herein comprise hematopoietic cells from placenta and from cord blood; from placenta and peripheral blood; from placenta and placental blood, or placenta and bone marrow.
  • the hematopoietic cells comprise hematopoietic cells from placental perfusate in combination with hematopoietic cells from cord blood, wherein the cord blood and placenta are from the same individual, i.e., wherein the perfusate and cord blood are matched.
  • the hematopoietic cells from the sources can be combined in a ratio of, for example, 1:10, 2:9, 3:8, 4:7:, 5:6, 6:5, 7:4, 8:3, 9:2, 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1 or 9:1.
  • the hematopoietic cells used in the methods provided herein are placental hematopoietic cells.
  • placental hematopoietic cells are CD34 + .
  • the placental hematopoietic cells are predominantly (e.g., at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 98%)
  • the placental hematopoietic cells are predominantly (e.g., at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 98%) CD34 + CD38 + cells.
  • Placental hematopoietic cells can be obtained from a postpartum mammalian (e.g., human) placenta by any means known to those of skill in the art, e.g., by perfusion.
  • the placental hematopoietic cell is CD45 .
  • the hematopoietic cell is CD34 CD45 .
  • the placental hematopoietic cells are CD34 + CD45 + .
  • Production of NK cells and/or ILC3 cells and NK cell and/or ILC3 cell populations by the present methods comprises expanding a population of hematopoietic cells. During cell expansion, a plurality of hematopoietic cells within the hematopoietic cell population differentiate into NK cells and/or ILC3 cells.
  • a method of producing NK cells comprising culturing hematopoietic stem cells or progenitor cells, e.g., CD34 + stem cells or progenitor cells, in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells, subsequently culturing said first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells, and subsequently culturing said second population of cells in a third medium comprising IL-2 and IL-15, and lacking a stem cell mobilizing agent and LMWH, to produce a third population of cells, wherein the third population of cells comprises natural killer cells that are CD56+, CD3-, and wherein at least 70%, for example at least 80%, of the natural killer cells are viable.
  • Tpo thrombopoietin
  • such natural killer cells comprise natural killer cells that are CD 16-, In certain embodiments, such natural killer cells comprise natural killer cells that are CD94+. In certain embodiments, such natural killer cells comprise natural killer cells that are CD94+ or CD16+. In certain embodiments, such natural killer cells comprise natural killer cells that are CD94- or CD 16-, In certain embodiments, such natural killer cells comprise natural killer cells that are CD94+ and CD16+. In certain embodiments, such natural killer cells comprise natural killer cells that are CD94- and CD16-. In certain embodiments, said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1 ⁇ ).
  • LIF leukemia inhibiting factor
  • MIP-1 ⁇ macrophage inflammatory protein-1 alpha
  • said third medium lacks LIF, MIP-1 ⁇ , and FMS-like tyrosine kinase-3 ligand (Flt-3L).
  • said first medium and said second medium lack LIF and MIP-1 ⁇
  • said third medium lacks LIF, MIP-1 ⁇ , and Flt3L.
  • none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
  • a method of producing NK cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells; and (c) culturing the second population of cells in a third medium comprising IL-2 and IL-15, and lacking LMWH, to produce a third population of cells; wherein the third population of cells comprises natural killer cells that are CD56+, CD3-, and CD11a+.
  • Tpo thrombopoietin
  • said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1 ⁇ ).
  • said third medium lacks LIF, MIP-1 ⁇ , and FMS-like tyrosine kinase-3 ligand (Flt-3L).
  • said first medium and said second medium lack LIF and MIP-1 ⁇
  • said third medium lacks LIF, MIP-1 ⁇ , and Flt3L.
  • none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
  • a method of producing NK cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells; and (c) culturing the second population of cells in a third medium comprising IL-2 and IL-15, and lacking each of stem cell factor (SCF) and LMWH, to produce a third population of cells; wherein the third population of cells comprises natural killer cells that are CD56+, CD3-, and CD11a+.
  • Tpo thrombopoietin
  • said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1 ⁇ ).
  • said third medium lacks LIF, MIP-1 ⁇ , and FMS-like tyrosine kinase- 3 ligand (Flt-3L).
  • said first medium and said second medium lack LIF, MIP-1 ⁇ , and FMS-like tyrosine kinase- 3 ligand (Flt-3L).
  • none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
  • a method of producing NK cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells; and (c) culturing the second population of cells in a third medium comprising IL-2 and IL-15, and lacking each of SCF, a stem cell mobilizing agent, and LMWH, to produce a third population of cells; wherein the third population of cells comprises natural killer cells that are CD56+, CD3-, and CD11a+.
  • Tpo thrombopoietin
  • said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1 ⁇ ).
  • said third medium lacks LIF, MIP-1 ⁇ , and FMS- like tyrosine kinase-3 ligand (Flt-3L).
  • said first medium and said second medium lack LIF and MIP-1 ⁇
  • said third medium lacks LIF, MIP-1 ⁇ , and Flt3L.
  • none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
  • a method of producing NK cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells;
  • IL-15 interleukin- 15
  • said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1 ⁇ ).
  • LIF leukemia inhibiting factor
  • MIP-1 ⁇ macrophage inflammatory protein-1 alpha
  • said third medium lacks LIF, MIP-1 ⁇ , and FMS-like tyrosine kinase-3 ligand (Flt-3L).
  • said first medium and said second medium lack LIF and MIP-1 ⁇
  • said third medium lacks LIF, MIP-1 ⁇ , and Flt3L.
  • none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
  • said natural killer cells express perforin and EOMES. In certain embodiments, said natural killer cells do not express either RORyt or IL1R1.
  • a method of producing ILC3 cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells; and (c) culturing the second population of cells in a third medium comprising IL-2 and IL-15, and lacking LMWH, to produce a third population of cells; wherein the third population of cells comprises ILC3 cells that are CD56+, CD3-, and CDlla-.
  • said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein- 1 alpha (MIP-1 ⁇ ).
  • said third medium lacks LIF, MIP-1 ⁇ , and FMS-like tyrosine kinase-3 ligand (Flt-3L).
  • said first medium and said second medium lack LIF and MIP-1 ⁇
  • said third medium lacks LIF, MIP-1 ⁇ , and Flt3L.
  • none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
  • a method of producing ILC3 cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells; and (c) culturing the second population of cells in a third medium comprising a stem cell mobilizing agent, IL-2 and IL-15, and lacking LMWH, to produce a third population of cells; wherein the third population of cells comprises ILC3 cells that are CD56+, CD3-, and CD11a-.
  • said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1 ⁇ ).
  • said third medium lacks LIF, MIP-1 ⁇ , and FMS-like tyrosine kinase- 3 ligand (Flt-3L).
  • said first medium and said second medium lack LIF and MIP-1 ⁇
  • said third medium lacks LIF, MIP-1 ⁇ , and Flt3L.
  • none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
  • a method of producing ILC3 cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells; and (c) culturing the second population of cells in a third medium comprising SCF, IL-2 and IL-15, and lacking LMWH, to produce a third population of cells; wherein the third population of cells comprises ILC3 cells that are CD56+, CD3-, and CDlla-.
  • said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1 ⁇ ).
  • said third medium lacks LIF, MIP-1 ⁇ , and FMS-like tyrosine kinase-3 ligand (Flt-3L).
  • said first medium and said second medium lack LIF and MIP-1 ⁇
  • said third medium lacks LIF, MIP-1 ⁇ , and Flt3L.
  • none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
  • a method of producing ILC3 cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells; and (c) culturing the second population of cells in a third medium comprising a stem cell mobilizing agent, SCF, IL-2 and IL-15, and lacking LMWH, to produce a third population of cells; wherein the third population of cells comprises ILC3 cells that are CD56+, CD3-, and CD11a-.
  • said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1 ⁇ ).
  • said third medium lacks LIF, MIP-1 ⁇ , and FMS-like tyrosine kinase- 3 ligand (Flt-3L).
  • said first medium and said second medium lack LIF and MIP-1 ⁇
  • said third medium lacks LIF, MIP-1 ⁇ , and Flt3L.
  • none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
  • a method of producing ILC3 cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells;
  • IL-15 interleukin- 15
  • said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1 ⁇ ).
  • LIF leukemia inhibiting factor
  • MIP-1 ⁇ macrophage inflammatory protein-1 alpha
  • said third medium lacks LIF, MIP-1 ⁇ , and FMS-like tyrosine kinase-3 ligand (Flt-3L).
  • said first medium and said second medium lack LIF and MIP-1 ⁇
  • said third medium lacks LIF, MIP-1 ⁇ , and Flt3L.
  • none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
  • said ILC3 cells express RORyt and IL1R1. In certain embodiments, said ILC3 cells do not express either perforin or EOMES.
  • a three-stage method of producing NK cell and/or ILC3 cell populations comprises maintaining the cell population comprising said hematopoietic cells at between about 2 x 10 4 and about 6 x 10 6 cells per milliliter.
  • said hematopoietic stem or progenitor cells are initially inoculated into said first medium from 1 x 10 4 to 1 x 10 5 cells/mL.
  • said hematopoietic stem or progenitor cells are initially inoculated into said first medium at about 3 x 10 4 cells/mL.
  • said first population of cells are initially inoculated into said second medium from 5 x 10 4 to 5 x 10 5 cells/mL. In a specific aspect, said first population of cells is initially inoculated into said second medium at about 1 x 10 5 cells/mL.
  • said second population of cells is initially inoculated into said third medium from 1 x 10 5 to 5 x 10 6 cells/mL. In certain aspects, said second population of cells is initially inoculated into said third medium from 1 x 10 5 to 1 x 10 6 cells/mL. In a specific aspect, said second population of cells is initially inoculated into said third medium at about 5 x 10 5 cells/mL. In a more specific aspect, said second population of cells is initially inoculated into said third medium at about 5 x 10 5 cells/mL in a spinner flask. In a specific aspect, said second population of cells is initially inoculated into said third medium at about 3 x lO 5 cells/mL.
  • the three-stage method comprises a first stage (“stage 1”) comprising culturing hematopoietic stem cells or progenitor cells, e.g., CD34 + stem cells or progenitor cells, in a first medium for a specified time period, e.g., as described herein, to produce a first population of cells.
  • the first medium comprises a stem cell mobilizing agent and thrombopoietin (Tpo).
  • the first medium comprises in addition to a stem cell mobilizing agent and Tpo, one or more of LMWH, Flt-3L, SCF, IL-6, IL-7, G-CSF, and GM-CSF.
  • the first medium comprises in addition to a stem cell mobilizing agent and Tpo, each of LMWH, Flt- 3L, SCF, IL-6, IL-7, G-CSF, and GM-CSF.
  • the first medium lacks added LMWH.
  • the first medium lacks added desulphated glycosaminoglycans.
  • the first medium lacks LMWH.
  • the first medium lacks desulphated glycosaminoglycans.
  • each of Flt-3L, SCF, IL-6, IL-7, G-CSF, and GM-CSF in addition to a stem cell mobilizing agent and Tpo, each of Flt-3L, SCF, IL-6, IL-7, G-CSF, and GM-CSF.
  • the first medium lacks leukemia inhibiting factor (LIF), macrophage inhibitory protein- 1 alpha (MIP-1 ⁇ ) or both.
  • LIF leukemia inhibiting factor
  • MIP-1 ⁇ macrophage inhibitory protein- 1 alpha
  • the second medium comprises a stem cell mobilizing agent and interleukin- 15 (IL-15) and lacks Tpo.
  • the second medium comprises, in addition to a stem cell mobilizing agent and IL-15, one or more of LMWH, Flt-3, SCF, IL-6, IL-7, G-CSF, and GM-CSF.
  • the second medium comprises, in addition to a stem cell mobilizing agent and IL-15, each of LMWH, Flt-3, SCF, IL-6, IL-7, G-CSF, and GM-CSF.
  • the second medium lacks added LMWH.
  • the second medium lacks added desulphated glycosaminoglycans.
  • the second medium lacks heparin, e.g., LMWH.
  • the second medium lacks desulphated glycosaminoglycans.
  • the second medium comprises, in addition to a stem cell mobilizing agent and IL-15, each of Flt-3, SCF, IL-6, IL-7, G-CSF, and GM-CSF.
  • the second medium lacks leukemia inhibiting factor (LIF), macrophage inhibitory protein- 1 alpha (MIP-1 ⁇ ) or both.
  • LIF leukemia inhibiting factor
  • MIP-1 ⁇ macrophage inhibitory protein- 1 alpha
  • subsequently, in “stage 3” said cells are cultured in a third medium for a specified time period, e.g., as described herein, to produce a third population of cell, e.g., natural killer cells.
  • the third medium comprises IL-2 and IL-15, and lacks a stem cell mobilizing agent and LMWH.
  • the third medium comprises in addition to IL-2 and IL-15, one or more of SCF, IL-6, IL-7, G-CSF, and GM-CSF.
  • the third medium comprises, in addition to IL-2 and IL-15, each of SCF, IL-6, IL-7, G-CSF, and GM-CSF.
  • the first medium lacks one, two, or all three of LIF, MIP-1 ⁇ , and Flt3L.
  • the third medium lacks added desulphated glycosaminoglycans.
  • the third medium lacks desulphated glycosaminoglycans.
  • the third medium lacks heparin, e.g., LMWH.
  • the three-stage method is used to produce NK cell and/or ILC3 cell populations.
  • the three-stage method is conducted in the absence of stromal feeder cell support.
  • the three-stage method is conducted in the absence of exogenously added steroids (e.g., cortisone, hydrocortisone, or derivatives thereof).
  • said first medium used in the three-stage method comprises a stem cell mobilizing agent and thrombopoietin (Tpo).
  • the first medium used in the three-stage method comprises, in addition to a stem cell mobilizing agent and Tpo, one or more of Low Molecular Weight Heparin (LMWH), Flt-3 Ligand (Flt-3L), stem cell factor (SCF), IL-6, IL-7, granulocyte colony-stimulating factor (G-CSF), or granulocyte- macrophage-stimulating factor (GM-CSF).
  • LMWH Low Molecular Weight Heparin
  • Flt-3L Flt-3 Ligand
  • SCF stem cell factor
  • IL-6 IL-6
  • IL-7 granulocyte colony-stimulating factor
  • G-CSF granulocyte colony-stimulating factor
  • GM-CSF granulocyte- macrophage-stimulating factor
  • the first medium used in the three-stage method comprises, in addition to a stem cell mobilizing agent and Tpo, each of LMWH, Flt-3L, SCF, IL-6, IL-7, G-CSF, and GM-CSF. In certain aspects, the first medium used in the three-stage method comprises, in addition to a stem cell mobilizing agent and Tpo, each of Flt-3L, SCF, IL-6, IL-7, G-CSF, and GM-CSF. In a specific aspect, the first medium lacks added LMWH. In a specific aspect, the first medium lacks added desulphated glycosaminoglycans. In a specific aspect, the first medium lacks LMWH.
  • the first medium lacks desulphated glycosaminoglycans.
  • said Tpo is present in the first medium at a concentration of from 1 ng/mL to 100 ng/mL, from 1 ng/mL to 50 ng/mL, from 20 ng/mL to 30 ng/mL, or about 25 ng/mL.
  • said Tpo is present in the first medium at a concentration of from 100 ng/mL to 500 ng/mL, from 200 ng/mL to 300 ng/mL, or about 250 ng/mL.
  • the LMWH when LMWH is present in the first medium, the LMWH is present at a concentration of from lU/mL to lOU/mL; the Flt-3L is present at a concentration of from 1 ng/mL to 50 ng/mL; the SCF is present at a concentration of from 1 ng/mL to 50 ng/mL; the IL-6 is present at a concentration of from 0.01 ng/mL to 0.1 ng/mL; the IL-7 is present at a concentration of from 1 ng/mL to 50 ng/mL; the G-CSF is present at a concentration of from 0.01 ng/mL to 0.50 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.1 ng/mL.
  • the Flt-3L is present at a concentration of from 1 ng/mL to 50 ng/mL;
  • the SCF is present at a concentration of from 1 ng/mL to 50 ng/mL;
  • the IL-6 is present at a concentration of from 0.01 ng/mL to 0.1 ng/mL;
  • the IL-7 is present at a concentration of from 1 ng/mL to 50 ng/mL;
  • the G-CSF is present at a concentration of from 0.01 ng/mL to 0.50 ng/mL;
  • the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.1 ng/mL.
  • the LMWH when LMWH is present in the first medium, the LMWH is present at a concentration of from 4U/mL to 5U/mL; the Flt-3L is present at a concentration of from 20 ng/mL to 30 ng/mL; the SCF is present at a concentration of from 20 ng/mL to 30 ng/mL; the IL-6 is present at a concentration of from 0.04 ng/mL to 0.06 ng/mL; the IL-7 is present at a concentration of from 20 ng/mL to 30 ng/mL; the G-CSF is present at a concentration of from 0.20 ng/mL to 0.30 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.5 ng/mL.
  • the Flt-3L is present at a concentration of from 20 ng/mL to 30 ng/mL;
  • the SCF is present at a concentration of from 20 ng/mL to 30 ng/mL;
  • the IL-6 is present at a concentration of from 0.04 ng/mL to 0.06 ng/mL;
  • the IL-7 is present at a concentration of from 20 ng/mL to 30 ng/mL;
  • the G-CSF is present at a concentration of from 0.20 ng/mL to 0.30 ng/mL;
  • the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.5 ng/mL.
  • the LMWH when LMWH is present in the first medium, the LMWH is present at a concentration of about 4.5U/mL; the Flt-3L is present at a concentration of about 25 ng/mL; the SCF is present at a concentration of about 27 ng/mL; the IL-6 is present at a concentration of about 0.05 ng/mL; the IL-7 is present at a concentration of about 25 ng/mL; the G-CSF is present at a concentration of about .25 ng/mL; and the GM-CSF is present at a concentration of about 0.01 ng/mL.
  • the Flt-3L is present at a concentration of about 25 ng/mL; the SCF is present at a concentration of about 27 ng/mL; the IL-6 is present at a concentration of about 0.05 ng/mL; the IL-7 is present at a concentration of about 25 ng/mL; the G-CSF is present at a concentration of about .25 ng/mL; and the GM-CSF is present at a concentration of about 0.01 ng/mL.
  • said first medium additionally comprises one or more of the following: antibiotics such as gentamycin; antioxidants such as transferrin, insulin, and/or beta-mercaptoethanol; sodium selenite; ascorbic acid; ethanolamine; and glutathione.
  • antibiotics such as gentamycin
  • antioxidants such as transferrin, insulin, and/or beta-mercaptoethanol
  • sodium selenite sodium selenite
  • ascorbic acid ethanolamine
  • glutathione glutathione
  • the medium that provides the base for the first medium is a cell/tissue culture medium known to those of skill in the art, e.g., a commercially available cell/tissue culture medium such as SCGMTM, STEMMACSTM, GBGM®, AIM-V®, X-VIVOTM 10, X-VIVOTM 15, OPTMIZER, STEMSPAN® H3000, CELLGRO COMPLETETM, DMEM:Ham’s F12 (“F12”) (e.g., 2:1 ratio, or high glucose or low glucose DMEM), Advanced DMEM (Gibco), EL08-1D2, MyelocultTM H5100, IMDM, and/or RPMI- 1640; or is a medium that comprises components generally included in known cell/tissue culture media, such as the components included in GBGM®, AIM-V®, X-VIVOTM 10, X- VIVOTM 15, OPTMIZER, STEMSPAN® H3000, CELLGRO COMPLETETM,
  • F12
  • DMEM:Ham s F12 (“F12”) (e.g., 2:1 ratio, or high glucose or low glucose DMEM), Advanced DMEM (Gibco), EL08-1D2, MyelocultTM H5100, IMDM, and/or RPMI-1640.
  • F12 e.g., 2:1 ratio, or high glucose or low glucose DMEM
  • Advanced DMEM Gabco
  • EL08-1D2 elocultTM H5100
  • IMDM RPMI-1640.
  • said first medium is not GBGM®.
  • the first medium lacks LIF, MIP-1 ⁇ , or both.
  • said second medium used in the three-stage method comprises a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacks Tpo.
  • the second medium used in the three-stage method comprises, in addition to a stem cell mobilizing agent and IL-15, one or more of LMWH, Flt-3, SCF, IL-6, IL-7, G-CSF, and GM-CSF.
  • the second medium used in the three-stage method comprises, in addition to a stem cell mobilizing agent and IL-15, each of LMWH, Flt-3, SCF, IL-6, IL-7, G-CSF, and GM-CSF.
  • the second medium used in the three-stage method comprises, in addition to a stem cell mobilizing agent and IL-15, each of Flt-3, SCF, IL-6, IL- 7, G-CSF, and GM-CSF.
  • the second medium lacks added LMWH.
  • the second medium lacks added desulphated glycosaminoglycans.
  • the second medium lacks LMWH.
  • the second medium lacks desulphated glycosaminoglycans.
  • said IL-15 is present in said second medium at a concentration of from 1 ng/mL to 50 ng/mL, from 10 ng/mL to 30 ng/mL, or about 20 ng/mL.
  • the LMWH is present at a concentration of from lU/mL to lOU/mL
  • the Flt-3L is present at a concentration of from 1 ng/mL to 50 ng/mL
  • the SCF is present at a concentration of from 1 ng/mL to 50 ng/mL
  • the IL-6 is present at a concentration of from 0.01 ng/mL to 0.1 ng/mL
  • the IL-7 is present at a concentration of from 1 ng/mL to 50 ng/mL
  • the G-CSF is present at a concentration of from 0.01 ng/mL to 0.50 ng/mL
  • the GM-CSF is present at a concentration of from 0.005 ng/
  • the Flt-3L is present at a concentration of from 1 ng/mL to 50 ng/mL;
  • the SCF is present at a concentration of from 1 ng/mL to 50 ng/mL;
  • the IL-6 is present at a concentration of from 0.01 ng/mL to 0.1 ng/mL;
  • the IL-7 is present at a concentration of from 1 ng/mL to 50 ng/mL;
  • the G-CSF is present at a concentration of from 0.01 ng/mL to 0.50 ng/mL;
  • the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.1 ng/mL.
  • the LMWH when LMWH is present in the second medium, the LMWH is present in the second medium at a concentration of from 4U/mL to 5U/mL; the Flt-3L is present at a concentration of from 20 ng/mL to 30 ng/mL; the SCF is present at a concentration of from 20 ng/mL to 30 ng/mL; the IL-6 is present at a concentration of from 0.04 ng/mL to 0.06 ng/mL; the IL-7 is present at a concentration of from 20 ng/mL to 30 ng/mL; the G-CSF is present at a concentration of from 0.20 ng/mL to 0.30 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.5 ng/mL.
  • the Flt-3L is present at a concentration of from 20 ng/mL to 30 ng/mL;
  • the SCF is present at a concentration of from 20 ng/mL to 30 ng/mL;
  • the IL-6 is present at a concentration of from 0.04 ng/mL to 0.06 ng/mL;
  • the IL-7 is present at a concentration of from 20 ng/mL to 30 ng/mL;
  • the G-CSF is present at a concentration of from 0.20 ng/mL to 0.30 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.5 ng/mL.
  • the LMWH when LMWH is present in the second medium, the LMWH is present in the second medium at a concentration of from 4U/mL to 5U/mL; the Flt-3L is present at a concentration of from 20 ng/mL to 30 ng/mL; the SCF is present at a concentration of from 20 ng/mL to 30 ng/mL; the IL-6 is present at a concentration of from 0.04 ng/mL to 0.06 ng/mL; the IL-7 is present at a concentration of from 20 ng/mL to 30 ng/mL; the G-CSF is present at a concentration of from 0.20 ng/mL to 0.30 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.5 ng/mL.
  • the Flt-3L is present at a concentration of from 20 ng/mL to 30 ng/mL;
  • the SCF is present at a concentration of from 20 ng/mL to 30 ng/mL;
  • the IL-6 is present at a concentration of from 0.04 ng/mL to 0.06 ng/mL;
  • the IL-7 is present at a concentration of from 20 ng/mL to 30 ng/mL;
  • the G-CSF is present at a concentration of from 0.20 ng/mL to 0.30 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.5 ng/mL.
  • the LMWH when LMWH is present in the second medium, the LMWH is present in the second medium at a concentration of about 4.5U/mL; the Flt-3L is present at a concentration of about 25 ng/mL; the SCF is present at a concentration of about 27 ng/mL; the IL-6 is present at a concentration of about 0.05 ng/mL; the IL-7 is present at a concentration of about 25 ng/mL; the G-CSF is present at a concentration of about 0.25 ng/mL; and the GM-CSF is present at a concentration of about
  • the Flt-3L is present at a concentration of about 25 ng/mL; the SCF is present at a concentration of about 27 ng/mL; the IL-6 is present at a concentration of about 0.05 ng/mL; the IL-7 is present at a concentration of about 25 ng/mL; the G-CSF is present at a concentration of about 0.25 ng/mL; and the GM-CSF is present at a concentration of about 0.01 ng/mL.
  • said second medium additionally comprises one or more of the following: antibiotics such as gentamycin; antioxidants such as transferrin, insulin, and/or beta- mercaptoethanol; sodium selenite; ascorbic acid; ethanolamine; and glutathione.
  • antibiotics such as gentamycin
  • antioxidants such as transferrin, insulin, and/or beta- mercaptoethanol
  • sodium selenite sodium selenite
  • ascorbic acid ethanolamine
  • glutathione glutathione
  • the medium that provides the base for the second medium is a cell/tissue culture medium known to those of skill in the art, e.g., a commercially available cell/tissue culture medium such as SCGMTM, STEMMACSTM, GBGM®, AIM-V®, X-VIVOTM 10, X- VIVOTM 15, OPTMIZER, STEMSPAN® H3000, CELLGRO COMPLETETM, DMEM:Ham’s F12 (“F12”) (e.g., 2:1 ratio, or high glucose or low glucose DMEM), Advanced DMEM (Gibco), EL08-1D2, MyelocultTM H5100, IMDM, and/or RPMI-1640; or is a medium that comprises components generally included in known cell/tissue culture media, such as the components included in GBGM®, AIM-V®, X-VIVOTM 10, X-VIVOTM 15, OPTMIZER, STEMSPAN® H3000, CELLGRO COMPLETETM,
  • F12
  • said third medium used in the three-stage method comprises IL-2 and IL-15, and lacks a stem cell mobilizing agent and LMWH. In certain aspects, said third medium used in the three-stage method comprises IL-2 and IL-15, and lacks LMWH.
  • said third medium used in the three-stage method comprises IL-2 and IL- 15, and lacks SCF and LMWH. In certain aspects, said third medium used in the three-stage method comprises IL-2 and IL-15, and lacks SCF, a stem cell mobilizing agent and LMWH. In certain aspects, said third medium used in the three-stage method comprises a stem cell mobilizing agent, IL-2 and IL-15, and lacks LMWH. In certain aspects, said third medium used in the three-stage method comprises SCF, IL-2 and IL-15, and lacks LMWH. In certain aspects, said third medium used in the three-stage method comprises a stem cell mobilizing agent, SCF, IL-2 and IL-15, and lacks LMWH.
  • said third medium used in the three-stage method comprises IL-2 and IL-15, and lacks a stem cell mobilizing agent and LMWH.
  • the third medium used in the three-stage method comprises, in addition to IL-2 and IL-15, one or more of SCF, IL-6, IL-7, G-CSF, or GM-CSF.
  • the third medium used in the three-stage method comprises, in addition to IL-2 and IL-15, each of SCF, IL-6, IL-7, G-CSF, and GM-CSF.
  • said IL-2 is present in said third medium at a concentration of from 10 U/mL to 10,000 U/mL and said IL-15 is present in said third medium at a concentration of from 1 ng/mL to 50 ng/mL. In certain aspects, said IL-2 is present in said third medium at a concentration of from 100 U/mL to 10,000 U/mL and said IL-15 is present in said third medium at a concentration of from 1 ng/mL to 50 ng/mL. In certain aspects, said IL-2 is present in said third medium at a concentration of from 300 U/mL to 3,000 U/mL and said IL-15 is present in said third medium at a concentration of from 10 ng/mL to 30 ng/mL.
  • said IL-2 is present in said third medium at a concentration of about 1,000 U/mL and said IL-15 is present in said third medium at a concentration of about 20 ng/mL.
  • the SCF is present at a concentration of from 1 ng/mL to 50 ng/mL;
  • the IL-6 is present at a concentration of from 0.01 ng/mL to 0.1 ng/mL;
  • the IL-7 is present at a concentration of from 1 ng/mL to 50 ng/mL;
  • the G-CSF is present at a concentration of from 0.01 ng/mL to 0.50 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.1 ng/mL.
  • the SCF is present at a concentration of from 20 ng/mL to 30 ng/mL; the IL-6 is present at a concentration of from 0.04 ng/mL to 0.06 ng/mL; the IL-7 is present at a concentration of from 20 ng/mL to 30 ng/mL; the G-CSF is present at a concentration of from 0.20 ng/mL to 0.30 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.5 ng/mL.
  • the SCF is present at a concentration of about 22 ng/mL; the IL-6 is present at a concentration of about 0.05 ng/mL; the IL-7 is present at a concentration of about 20 ng/mL; the G-CSF is present at a concentration of about 0.25 ng/mL; and the GM-CSF is present at a concentration of about 0.01 ng/mL.
  • the third medium comprises 100 ng/mL IL-7, 1000 ng/mL IL-2, 20 ng/mL IL-15, and 10 stem cell mobilizing agent and lacks SCF.
  • the third medium comprises 20 ng/mL IL-7, 1000 ng/mL IL-2, 20 ng/mL IL-15, and stem cell mobilizing agent and lacks SCF. In certain aspects, the third medium comprises 20 ng/mL IL-7, 20 ng/mL IL-15, and stem cell mobilizing agent and lacks SCF. In certain aspects, the third medium comprises 100 ng/mL IL-7, 22 ng/mL SCF, 1000 ng/mL IL-2, and 20 ng/mL IL-15 and lacks stem cell mobilizing agent. In certain aspects, the third medium comprises 22 ng/mL SCF, 1000 ng/mL IL-2, and 20 ng/mL IL-15 and lacks stem cell mobilizing agent.
  • the third medium comprises 20 ng/mL IL-7, 22 ng/mL SCF, 1000 ng/mL IL-2, and 20 ng/mL IL-15 and lacks stem cell mobilizing agent. In certain aspects, the third medium comprises 20 ng/mL IL-7, 22 ng/mL SCF, and 1000 ng/mL IL-2 and lacks stem cell mobilizing agent. In specific embodiments of any of the above embodiments, the first medium lacks one, two, or all three of LIF, MIP-1 ⁇ , Flt-3L.
  • said third medium additionally comprises one or more of the following: antibiotics such as gentamycin; antioxidants such as transferrin, insulin, and/or beta-mercaptoethanol; sodium selenite; ascorbic acid; ethanolamine; and glutathione.
  • antibiotics such as gentamycin
  • antioxidants such as transferrin, insulin, and/or beta-mercaptoethanol
  • sodium selenite sodium selenite
  • ascorbic acid ethanolamine
  • glutathione glutathione
  • the medium that provides the base for the third medium is a cell/tissue culture medium known to those of skill in the art, e.g., a commercially available cell/tissue culture medium such as SCGMTM, STEMMACSTM, GBGM®, AIM-V®, X-VIVOTM 10, X-VIVOTM 15, OPTMIZER, STEMSPAN® H3000, CELLGRO COMPLETETM, DMEM:Ham’s F12 (“F12”) (e.g., 2:1 ratio, or high glucose or low glucose DMEM), Advanced DMEM (Gibco), EL08-1D2, MyelocultTM H5100, IMDM, and/or RPMI- 1640; or is a medium that comprises components generally included in known cell/tissue culture media, such as the components included in GBGM®, AIM-V®, X-VIVOTM 10, X- VIVOTM 15, OPTMIZER, STEMSPAN® H3000, CELLGRO COMPLETETM,
  • F12
  • DMEM:Ham s F12 (“F12”) (e.g., 2:1 ratio, or high glucose or low glucose DMEM), Advanced DMEM (Gibco), EL08-1D2, MyelocultTM H5100, IMDM, and/or RPMI-1640.
  • F12 e.g., 2:1 ratio, or high glucose or low glucose DMEM
  • Advanced DMEM Gabco
  • EL08-1D2 elocultTM H5100
  • IMDM e.g., RPMI-1640.
  • said third medium is not GBGM®.
  • the particularly recited medium components do not refer to possible constituents in an undefined component of said medium.
  • said Tpo, IL-2, and IL-15 are not comprised within an undefined component of the first medium, second medium or third medium, e.g., said Tpo, IL-2, and IL-15 are not comprised within serum.
  • said LMWH, Flt-3, SCF, IL-6, IL-7, G-CSF, and/or GM-CSF are not comprised within an undefined component of the first medium, second medium or third medium, e.g., said LMWH, Flt-3, SCF, IL-6, IL-7, G-CSF, and/or GM-CSF are not comprised within serum.
  • said first medium, second medium or third medium comprises human serum-AB.
  • any of said first medium, second medium or third medium comprises 1% to 20% human serum-AB, 5% to 15% human serum-AB, or about 2, 5, or 10% human serum-AB.
  • said hematopoietic stem or progenitor cells are cultured in said first medium for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 days.
  • cells are cultured in said second medium for 1, 2, 3, 4, 5, 6,
  • cells are cultured in said third medium for 1, 2, 3, 4, 5,
  • said hematopoietic stem or progenitor cells are cultured in said first medium for 7-13 days to produce a first population of cells, before said culturing in said second medium; said first population of cells are cultured in said second medium for 2-6 days to produce a second population of cells before said culturing in said third medium; and said second population of cells are cultured in said third medium for 10-30 days, i.e., the cells are cultured a total of 19- 49 days.
  • said hematopoietic stem or progenitor cells are cultured in said first medium for 8-12 days to produce a first population of cells, before said culturing in said second medium; said first population of cells are cultured in said second medium for 3-5 days to produce a second population of cells before said culturing in said third medium; and said second population of cells are cultured in said third medium for 15-25 days, i.e., the cells are cultured a total of 26-42 days.
  • said hematopoietic stem or progenitor cells are cultured in said first medium for about 10 days to produce a first population of cells, before said culturing in said second medium; said first population of cells are cultured in said second medium for about 4 days to produce a second population of cells before said culturing in said third medium; and said second population of cells are cultured in said third medium for about 21 days, i.e., the cells are cultured a total of about 35 days.
  • the three-stage method disclosed herein produces at least 5000-fold more natural killer cells as compared to the number of hematopoietic stem cells initially inoculated into said first medium. In certain aspects, said three-stage method produces at least 10,000-fold more natural killer cells as compared to the number of hematopoietic stem cells initially inoculated into said first medium. In certain aspects, said three-stage method produces at least 50,000-fold more natural killer cells as compared to the number of hematopoietic stem cells initially inoculated into said first medium. In certain aspects, said three-stage method produces at least 75,000-fold more natural killer cells as compared to the number of hematopoietic stem cells initially inoculated into said first medium.
  • the viability of said natural killer cells is determined by 7- aminoactinomycin D (7AAD) staining. In certain aspects, the viability of said natural killer cells is determined by annexin-V staining. In specific aspects, the viability of said natural killer cells is determined by both 7-AAD staining and annexin-V staining. In certain aspects, the viability of said natural killer cells is determined by trypan blue staining.
  • the three-stage method disclosed herein produces at least 5000-fold more ILC3 cells as compared to the number of hematopoietic stem cells initially inoculated into said first medium. In certain aspects, said three-stage method produces at least 10,000-fold more ILC3 cells as compared to the number of hematopoietic stem cells initially inoculated into said first medium. In certain aspects, said three-stage method produces at least 50,000-fold more ILC3 cells as compared to the number of hematopoietic stem cells initially inoculated into said first medium. In certain aspects, said three-stage method produces at least 75,000-fold more ILC3 cells as compared to the number of hematopoietic stem cells initially inoculated into said first medium.
  • the three-stage method produces natural killer cells that comprise at least 20% CD56+CD3- natural killer cells. In certain aspects, the three-stage method produces natural killer cells that comprise at least 40% CD56+CD3- natural killer cells. In certain aspects, the three-stage method produces natural killer cells that comprise at least 60% CD56+CD3- natural killer cells. In certain aspects, the three-stage method produces natural killer cells that comprise at least 70% CD56+CD3- natural killer cells. In certain aspects, the three-stage method produces natural killer cells that comprise at least 80% CD56+CD3- natural killer cells.
  • the three-stage method disclosed herein produces natural killer cells that comprise at least 20% CD56+CD3-CDlla+ natural killer cells. In certain aspects, the three-stage method disclosed herein produces natural killer cells that comprise at least 40% CD56+CD3- CD11a+ natural killer cells. In certain aspects, the three-stage method disclosed herein produces natural killer cells that comprise at least 60% CD56+CD3- CDlla+ natural killer cells. In certain aspects, the three-stage method disclosed herein produces natural killer cells that comprise at least 80% CD56+CD3- CDlla+ natural killer cells.
  • the three-stage method disclosed herein produces ILC3 cells that comprise at least 20% CD56+CD3- CD11a- ILC3 cells. In certain aspects, the three- stage method disclosed herein produces ILC3 cells that comprise at least 40% CD56+CD3- CDlla- ILC3 cells. In certain aspects, the three-stage method disclosed herein produces
  • ILC3 cells that comprise at least 60% CD56+CD3- CDlla- ILC3 cells.
  • the three-stage method disclosed herein produces natural killer cells that comprise at least 80% CD56+CD3- CD11a- ILC3 cells.
  • the three-stage method produces natural killer cells that exhibit at least 20% cytotoxicity against K562 cells when said natural killer cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10:1. In certain aspects, the three- stage method produces natural killer cells that exhibit at least 35% cytotoxicity against the K562 cells when said natural killer cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10: 1. In certain aspects, the three-stage method produces natural killer cells that exhibit at least 45% cytotoxicity against the K562 cells when said natural killer cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10: 1.
  • the three-stage method produces natural killer cells that exhibit at least 60% cytotoxicity against the K562 cells when said natural killer cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10: 1. In certain aspects, the three-stage method produces natural killer cells that exhibit at least 75% cytotoxicity against the K562 cells when said natural killer cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10: 1.
  • the three-stage method produces ILC3 cells that exhibit at least 20% cytotoxicity against K562 cells when said ILC3 cells and said K562 cells are co- cultured in vitro or ex vivo at a ratio of 10: 1. In certain aspects, the three-stage method produces ILC3 cells that exhibit at least 35% cytotoxicity against the K562 cells when said ILC3 cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10:1. In certain aspects, the three-stage method produces ILC3 cells that exhibit at least 45% cytotoxicity against the K562 cells when said ILC3 cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10: 1.
  • the three-stage method produces ILC3 cells that exhibit at least 60% cytotoxicity against the K562 cells when said ILC3 cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10:1. In certain aspects, the three-stage method produces ILC3 cells that exhibit at least 75% cytotoxicity against the K562 cells when said ILC3 cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10: 1. [00135] In certain aspects, after said third culturing step, said third population of cells, e.g., said population of natural killer cells and/or ILC3 cells, is cryopreserved. In certain aspects, after said fourth step, said fourth population of cells, e.g., said population of natural killer cells and/or ILC3 cells, is cryopreserved.
  • populations of cells comprising natural killer cells, i.e., natural killers cells produced by a three-stage method described herein.
  • said natural killer cell population comprises at least 20% CD56+CD3- natural killer cells. In a specific embodiment, said natural killer cell population comprises at least 40% CD56+CD3- natural killer cells. In a specific embodiment, said natural killer cell population comprises at least 60% CD56+CD3- natural killer cells. In a specific embodiment, said natural killer cell population comprises at least 80% CD56+CD3- natural killer cells. In a specific embodiment, said natural killer cell population comprises at least 60% CD16- cells. In a specific embodiment, said natural killer cell population comprises at least 80% CD16- cells. In a specific embodiment, said natural killer cell population comprises at least 20% CD94+ cells. In a specific embodiment, said natural killer cell population comprises at least 40% CD94+ cells.
  • a population of natural killer cells that is CD56+CD3- CD117+CDl la+, wherein said natural killer cells express perforin and/or EOMES, and do not express one or more of RORyt, aryl hydrocarbon receptor (AHR), and IL1R1.
  • said natural killer cells express perforin and EOMES, and do not express any of RORyt, aryl hydrocarbon receptor, or IL1R1.
  • said natural killer cells additionally express T-bet, GZMB, NKp46, NKp30, and NKG2D.
  • said natural killer cells express CD94. In certain aspects, said natural killer cells do not express CD94.
  • a population of ILC3 cells that is CD56+CD3- CD117+CDl la-, wherein said ILC3 cells express one or more of RORyt, aryl hydrocarbon receptor, and IL1R1, and do not express one or more of CD94, perforin, and EOMES.
  • said ILC3 cells express RORyt, aryl hydrocarbon receptor, and IL1R1, and do not express any of CD94, perforin, or EOMES.
  • said ILC3 cells additionally express CD226 and/or 2B4.
  • said ILC3 cells additionally express one or more of IL-22, TNFa, and DNAM-1.
  • said ILC3 cells express CD226, 2B4, IL-22, TNFa, and DNAM-1.
  • a method of producing a cell population comprising natural killer cells and ILC3 cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL- 15), and lacking Tpo, to produce a second population of cells; (c) culturing the second population of cells in a third medium comprising IL-2 and IL-15, and lacking each of a stem cell mobilizing agent and LMWH, to produce a third population of cells; and (d) separating CD11a+ cells and CD11a- cells from the third population of cells; and (e) combining the CDlla+ cells with the CDlla- cells in a ratio of 50:1, 40:1, 30:1,
  • said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1 ⁇ ).
  • said third medium lacks LIF, MIP-1 ⁇ , and FMS-like tyrosine kinase-3 ligand (Flt-3L).
  • said first medium and said second medium lack LIF and MIP-1 ⁇
  • said third medium lacks LIF, MIP-1 ⁇ , and Flt3L.
  • none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
  • the CD11a+ cells and CDlla- cells are combined in a ratio of 50:1. In certain aspects, in the fourth population of cells, the CD11a+ cells and CD11a- cells are combined in a ratio of 20: 1. In certain aspects, in the fourth population of cells, the CD11a+ cells and CD11a- cells are combined in a ratio of 10: 1. In certain aspects, in the fourth population of cells, the CD11a+ cells and CD11a- cells are combined in a ratio of 5: 1. In certain aspects, in the fourth population of cells, the CD11a+ cells and CD 11a- cells are combined in a ratio of 1 : 1.
  • the CD11a+ cells and CD11a- cells are combined in a ratio of 1 :5. In certain aspects, in the fourth population of cells, the CD11a+ cells and CD11a- cells are combined in a ratio of 1 : 10. In certain aspects, in the fourth population of cells, the CD11 a+ cells and CD11a- cells are combined in a ratio of 1 :20. In certain aspects, in the fourth population of cells, the CDlla+ cells and CDlla- cells are combined in aratio of 1:50.
  • the term “about” or “approximately” means an acceptable error for a particular value as determined by one of ordinary skill in the art, which depends in part on how the value is measured or determined. In certain embodiments, the term “about” or “approximately” means within 1, 2, 3, or 4 standard deviations. In certain embodiments, the term “about” or “approximately” means within 50%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.05% of a given value or range.
  • any "R" group(s) such as, without limitation, R a , R b , R c , R d ,
  • R e , R f R g , R h , R m , R g , R j , R k , R U , R V R y , and R Z represent substituents that can be attached to the indicated atom.
  • An R group may be substituted or unsubstituted. If two "R" groups are described as being “taken together" the R groups and the atoms they are attached to can form a cycloalkyl, cycloalkenyl, aryl, heteroaryl or heterocycle. For example, without limitation, if R a and R b of an NR a R b group are indicated to be "taken together," it means that they are covalently bonded to one another to form a ring:
  • R groups are described as being “taken together” with the atom(s) to which they are attached to form a ring as an alternative, the R groups are not limited to the variables or substituents defined previously.
  • the indicated “optionally substituted” or “substituted” group may be substituted with one or more group(s) individually and independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, acylalkyl, hydroxy, alkoxy, alkoxyalkyl, aminoalkyl, amino acid, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl), heterocyclyl(alkyl), hydroxyalkyl, acyl, cyano, halogen, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, O-carboxy, isocyanato, thiocyana
  • C a to Cb in which “a” and “b” are integers refer to the number of carbon atoms in an alkyl, alkenyl or alkynyl group, or the number of carbon atoms in the ring of a cycloalkyl, cycloalkenyl, aryl, heteroaryl or heteroalicyclyl group.
  • the alkyl, alkenyl, alkynyl, ring(s) of the cycloalkyl, ring(s) of the cycloalkenyl, ring(s) of the aryl, ring(s) of the heteroaryl or ring(s) of the heteroalicyclyl can contain from “a” to “b”, inclusive, carbon atoms.
  • a “C 1 to C 4 alkyl” group refers to all alkyl groups having from 1 to 4 carbons, that is, CH 3 -, CH 3 CH 2 -, CH 3 CH 2 CH 2 -, ( CH 3 ) 2 CH-, CH 3 CH 2 CH 2 CH 2 -, CH 3 CH 2 CH(CH 3 )- and (CH 3 ) 3 C -. If no “a” and “b” are designated with regard to an alkyl, alkenyl, alkynyl, cycloalkyl cycloalkenyl, aryl, heteroaryl or heteroalicyclyl group, the broadest range described in these definitions is to be assumed.
  • alkyl refers to a straight or branched hydrocarbon chain that comprises a fully saturated (no double or triple bonds) hydrocarbon group.
  • the alkyl group may have 1 to 20 carbon atoms (whenever it appears herein, a numerical range such as “1 to 20” refers to each integer in the given range; e.g., “1 to 20 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 20 carbon atoms, although the present definition also covers the occurrence of the term “alkyl” where no numerical range is designated).
  • the alkyl group may also be a medium size alkyl having 1 to 10 carbon atoms.
  • the alkyl group could also be a lower alkyl having 1 to 6 carbon atoms.
  • the alkyl group of the compounds may be designated as “C 1 -C 4 alkyl” or similar designations.
  • “C 1 -C 4 alkyl” indicates that there are one to four carbon atoms in the alkyl chain, i.e., the alkyl chain is selected from methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl.
  • Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl and hexyl.
  • the alkyl group may be substituted or unsubstituted.
  • alkenyl refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more double bonds. Examples of alkenyl groups include allenyl, vinylmethyl and ethenyl. An alkenyl group may be unsubstituted or substituted.
  • alkynyl refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more triple bonds. Examples of alkynyls include ethynyl and propynyl. An alkynyl group may be unsubstituted or substituted.
  • cycloalkyl refers to a completely saturated (no double or triple bonds) mono- or multi- cyclic hydrocarbon ring system. When composed of two or more rings, the rings may be joined together in a fused fashion. Cycloalkyl groups can contain 3 to 10 atoms in the ring(s) or 3 to 8 atoms in the ring(s). A cycloalkyl group may be unsubstituted or substituted. Typical cycloalkyl groups include, but are in no way limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl.
  • cycloalkenyl refers to a mono- or multi- cyclic hydrocarbon ring system that contains one or more double bonds in at least one ring; although, if there is more than one, the double bonds cannot form a fully delocalized pi-electron system throughout all the rings (otherwise the group would be “aryl,” as defined herein). Cycloalkenyl groups can contain 3 to 10 atoms in the ring(s) or 3 to 8 atoms in the ring(s). When composed of two or more rings, the rings may be connected together in a fused fashion. A cycloalkenyl group may be unsubstituted or substituted.
  • aryl refers to a carbocyclic (all carbon) monocyclic or multicyclic aromatic ring system (including fused ring systems where two carbocyclic rings share a chemical bond) that has a fully delocalized pi-electron system throughout all the rings.
  • the number of carbon atoms in an aryl group can vary.
  • the aryl group can be a C 6 -C 14 aryl group, a C 6 -C 10 aryl group, or a C 6 aryl group.
  • Examples of aryl groups include, but are not limited to, benzene, naphthalene and azulene.
  • An aryl group may be substituted or unsubstituted.
  • heteroaryl refers to a monocyclic or multicyclic aromatic ring system (a ring system with fully delocalized pi-electron system) that contain(s) one, two, three or more heteroatoms, that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur.
  • the number of atoms in the ring(s) of a heteroaryl group can vary.
  • the heteroaryl group can contain 4 to 14 atoms in the ring(s), 5 to 10 atoms in the ring(s) or 5 to 6 atoms in the ring(s).
  • heteroaryl includes fused ring systems where two rings, such as at least one aryl ring and at least one heteroaryl ring, or at least two heteroaryl rings, share at least one chemical bond.
  • heteroaryl rings include, but are not limited to, those described herein and the following: furan, furazan, thiophene, benzothiophene, phthalazine, pyrrole, oxazole, benzoxazole, 1,2,3- oxadiazole, 1,2,4-oxadiazole, thiazole, 1,2,3-thiadiazole, 1,2,4-thiadiazole, benzothiazole, imidazole, benzimidazole, indole, indazole, pyrazole, benzopyrazole, isoxazole, benzoisoxazole, isothiazole, triazole, benzotriazole, thiadiazole, tetrazole, pyridine, pyri
  • heteroaryl group may be substituted or unsubstituted.
  • heterocyclyl or “heteroalicyclyl” refers to three-, four-, five-, six-, seven-, eight-, nine-, ten-, up to 18-membered monocyclic, bicyclic, and tricyclic ring system wherein carbon atoms together with from 1 to 5 heteroatoms constitute said ring system.
  • a heterocycle may optionally contain one or more unsaturated bonds situated in such a way, however, that a fully delocalized pi-electron system does not occur throughout all the rings.
  • the heteroatom(s) is an element other than carbon including, but not limited to, oxygen, sulfur, and nitrogen.
  • a heterocycle may further contain one or more carbonyl or thiocarbonyl functionalities, so as to make the definition include oxo-systems and thio- systems such as lactams, lactones, cyclic imides, cyclic thioimides and cyclic carbamates. When composed of two or more rings, the rings may be joined together in a fused fashion. Additionally, any nitrogens in a heterocyclyl may be quatemized. Heterocyclyl or heteroalicyclic groups may be unsubstituted or substituted.
  • heterocyclyl or “heteroalicyclyl” groups include, but are not limited to, those described herein and the following: 1,3-dioxin, 1,3-dioxane, 1,4-dioxane, 1,2-dioxolane, 1,3-dioxolane, 1,4- dioxolane, 1,3-oxathiane, 1,4-oxathiin, 1,3-oxathiolane, 1,3-dithiole, 1,3-dithiolane, 1,4- oxathiane, tetrahydro-l,4-thiazine, 1,3-thiazinane, 2H-l,2-oxazine, maleimide, succinimide, barbituric acid, thiobarbituric acid, dioxopiperazine, hydantoin, dihydrouracil, trioxane, hexahydro-l,3,5-triazine, imi
  • aralkyl and “aryl(alkyl)” refer to an aryl group connected, as a substituent, via a lower alkylene group.
  • the lower alkylene and aryl group of an aralkyl may be substituted or unsubstituted. Examples include but are not limited to benzyl, 2- phenylalkyl, 3-phenylalkyl and naphthylalkyl.
  • heteroarylkyl and “heteroaryl(alkyl)” refer to a heteroaryl group connected, as a substituent, via a lower alkylene group.
  • the lower alkylene and heteroaryl group of heteroaralkyl may be substituted or unsubstituted. Examples include but are not limited to 2-thienylalkyl, 3-thienylalkyl, furylalkyl, thienylalkyl, pyrrolylalkyl, pyridylalkyl, isoxazolylalkyl, imidazolylalkyl and their benzo-fused analogs.
  • heteroalicyclyl(alkyl) and “heterocyclyl(alkyl)” refer to a heterocyclic or a heteroalicyclylic group connected, as a substituent, via a lower alkylene group.
  • the lower alkylene and heterocyclyl of a heteroalicyclyl(alkyl) may be substituted or unsubstituted. Examples include but are not limited tetrahydro-2H-pyran-4-yl(methyl), piperidin-4- yl(ethyl), piperidin-4-yl(propyl), tetrahydro-2H-thiopyran-4-yl(methyl), and l,3-thiazinan-4- yl(methyl).
  • “Lower alkylene groups” are straight-chained -CH 2 - tethering groups, forming bonds to connect molecular fragments via their terminal carbon atoms. Examples include but are not limited to methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), propylene (-CH 2 CH 2 CH 2 -), and butylene (-CH 2 CH 2 CH 2 CH 2 -).
  • a lower alkylene group can be substituted by replacing one or more hydrogen of the lower alkylene group with a substituent(s) listed under the definition of “substituted.”
  • alkoxy refers to the formula -OR wherein R is an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl) is defined herein.
  • R is an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl) is defined herein.
  • a non-limiting list of alkoxys are methoxy, ethoxy, n-propoxy, 1-methylethoxy (isopropoxy), n- butoxy, iso-butoxy,
  • acyl refers to a hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl) connected, as substituents, via a carbonyl group. Examples include formyl, acetyl, propanoyl, benzoyl and acryl. An acyl may be substituted or unsubstituted.
  • alkoxyalkyl refers to an alkoxy group connected, as a substituent, via a lower alkylene group. Examples include C 1-4 alkyl-O-(CH 2 )n- , wherein n is an integer in the range of 1 to 6.
  • aminoalkyl refers to an optionally substituted amino group connected, as a substituent, via a lower alkylene group.
  • examples include H2N(CH 2 )n- , wherein n is an integer in the range of 1 to 6.
  • hydroxyalkyl refers to an alkyl group in which one or more of the hydrogen atoms are replaced by a hydroxy group.
  • exemplary hydroxyalkyl groups include but are not limited to, 2-hydroxy ethyl, 3-hydroxypropyl, 2-hydroxypropyl, and 2,2- dihydroxyethyl.
  • a hydroxyalkyl may be substituted or unsubstituted.
  • haloalkyl refers to an alkyl group in which one or more of the hydrogen atoms are replaced by a halogen (e.g., mono-haloalkyl, di-haloalkyl and tri- haloalkyl).
  • a halogen e.g., mono-haloalkyl, di-haloalkyl and tri- haloalkyl.
  • groups include but are not limited to, chloromethyl, fluoromethyl, difluoromethyl, trifluoromethyl, chloro-fluoroalkyl, chloro-difluoroalkyl and 2- fluoroisobutyl.
  • a haloalkyl may be substituted or unsubstituted.
  • haloalkoxy refers to an alkoxy group in which one or more of the hydrogen atoms are replaced by a halogen (e.g., mono-haloalkoxy, di- haloalkoxy and tri- haloalkoxy).
  • a halogen e.g., mono-haloalkoxy, di- haloalkoxy and tri- haloalkoxy.
  • groups include but are not limited to, chloromethoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy, chloro-fluoroalkyl, chloro-difluoroalkoxy and 2- fluoroisobutoxy.
  • a haloalkoxy may be substituted or unsubstituted.
  • a “sulfenyl” group refers to an “-SR” group in which R can be hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • a sulfenyl may be substituted or unsubstituted.
  • a sulfmyl may be substituted or unsubstituted.
  • a “sulfonyl” group refers to an “SO2R” group in which R can be the same as defined with respect to sulfenyl.
  • a sulfonyl may be substituted or unsubstituted.
  • An O-carboxy may be substituted or unsubstituted.
  • An ester and C-carboxy may be substituted or unsubstituted.
  • a thiocarbonyl may be substituted or unsubstituted.
  • a “trihalomethanesulfonyl” group refers to an “X 3 CSO 2 -” group wherein each X is a halogen.
  • a “trihalomethanesulfonamido” group refers to an “X 3 CS(O) 2 N(RA)-” group wherein each X is a halogen, and RA hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • amino refers to a -NT 2 group.
  • hydroxy refers to a -OH group.
  • a “cyano” group refers to a “-CN” group.
  • the term “azido” as used herein refers to a -N3 group.
  • An “isocyanato” group refers to a “-NCO” group.
  • a “thiocyanato” group refers to a “-CNS” group.
  • An “isothiocyanato” group refers to an “ -NCS” group.
  • S-sulfonamido refers to a “-SO 2 N(RARB)” group in which RA and RB can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • An S-sulfonamido may be substituted or unsubstituted.
  • N-sulfonamido refers to a “RSO 2 N(RA)-” group in which R and RA can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • R and RA can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • An N-sulfonamido may be substituted or unsubstituted.
  • An O-carbamyl may be substituted or unsubstituted.
  • An N-carbamyl may be substituted or unsubstituted.
  • An O-thiocarbamyl may be substituted or unsubstituted.
  • An N-thiocarbamyl may be substituted or unsubstituted.
  • a C-amido may be substituted or unsubstituted.
  • R and RA can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
  • An N-amido may be substituted or unsubstituted.
  • a urea may be substituted or unsubstituted.
  • halogen atom or “halogen” as used herein, means any one of the radio-stable atoms of column 7 of the Periodic Table of the Elements, such as, fluorine, chlorine, bromine and iodine.
  • substituents there may be one or more substituents present.
  • haloalkyl may include one or more of the same or different halogens.
  • C 1 -C 3 alkoxyphenyl may include one or more of the same or different alkoxy groups containing one, two or three atoms.
  • optically active and “enantiomerically active” refer to a collection of molecules, which has an enantiomeric excess of no less than about 50%, no less than about 70%, no less than about 80%, no less than about 90%, no less than about 91%, no less than about 92%, no less than about 93%, no less than about 94%, no less than about 95%, no less than about 96%, no less than about 97%, no less than about 98%, no less than about 99%, no less than about 99.5%, or no less than about 99.8%.
  • the compound comprises about 95% or more of the desired enantiomer and about 5% or less of the less preferred enantiomer based on the total weight of the two enantiomers in question.
  • R and S are used to denote the absolute configuration of the optically active compound about its chiral center(s).
  • the (+) and (-) are used to denote the optical rotation of an optically active compound, that is, the direction in which a plane of polarized light is rotated by the optically active compound.
  • the (-) prefix indicates that an optically active compound is levorotatory, that is, the compound rotates the plane of polarized light to the left or counterclockwise.
  • (+) prefix indicates that an optically active compound is dextrorotatory, that is, the compound rotates the plane of polarized light to the right or clockwise.
  • sign of optical rotation, (+) and (-) is not related to the absolute configuration of a compound, R and S.
  • an “isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, hydrogen ( 1 H), deuterium ( 2 H), tritium ( 3 H), carbon-11 ( 11 C), carbon-12 ( 12 C), carbon-13 ( 13 C), carbon-14 ( 14 C), nitrogen-13 ( 13 N), nitrogen-14 ( 14 N), nitrogen-15 ( 15 N), oxygen-14 ( 14 O), oxygen-15 ( 15 O), oxygen-16 ( 16 O), oxygen-17 ( 17 O), oxygen-18 ( 18 O), fluorine-17 ( 17 F), fluorine-18 ( 18 F), phosphorus-31 ( 31 P), phosphorus-32 ( 32 P), phosphorus-33 ( 33 P), sulfur-32 ( 32 S), sulfur-33 ( 33 S), sulfur-34 ( 34 S), sulfur-35 ( 35 S), sulfur-36 ( 36 S), chlorine-
  • an “isotopic variant” of a compound is in a stable form, that is, non-radioactive.
  • an “isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, hydrogen ( 1 H), deuterium ( 2 H), carbon- 12 ( 12 C), carbon- 13 ( 13 C), nitrogen- 14 ( 14 N), nitrogen-15 ( 15 N), oxygen-16 ( 16 O), oxygen-17 ( 17 O), oxygen-18 ( 18 O), fluorine-17 ( 17 F), phosphorus-31 ( 31 P), sulfur-32 ( 32 S), sulfur-33 ( 33 S), sulfur-34 ( 34 S), sulfur-36 ( 36 S), chlorine-35 ( 35 C1), chlorine-37 ( 37 C1), bromine-79 ( 79 Br), bromine-81 ( 81 Br), and iodine-127 ( 127 I).
  • an “isotopic variant” of a compound is in an unstable form, that is, radioactive.
  • an “isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, tritium ( 3 H), carbon-11 ( 11 C), carbon-14 ( 14 C), nitrogen-13 ( 13 N), oxygen-14 ( 14 O), oxygen-15 ( 15 O), fluorine-18 ( 18 F), phosphorus-32 ( 32 P), phosphorus-33 ( 33 P), sulfur-35 ( 35 S), chlorine-36 ( 36 C1), iodine-123 ( 123 I), iodine-125 ( 125 I), iodine-129 ( 129 I), and iodine-131 ( 131 I).
  • any hydrogen can be 2 H, for example, or any carbon can be 13 C, for example, or any nitrogen can be 15 N, for example, or any oxygen can be 18 0, for example, where feasible according to the judgment of one of skill.
  • an “isotopic variant” of a compound contains unnatural proportions of deuterium (D).
  • solvate refers to a complex or aggregate formed by one or more molecules of a solute, e.g., a compound provided herein, and one or more molecules of a solvent, which present in a stoichiometric or non-stoichiometric amount.
  • Suitable solvents include, but are not limited to, water, methanol, ethanol, «-propanol, isopropanol, and acetic acid.
  • the solvent is pharmaceutically acceptable.
  • the complex or aggregate is in a crystalline form.
  • the complex or aggregate is in a noncry stalline form.
  • the solvent is water
  • the solvate is a hydrate. Examples of hydrates include, but are not limited to, a hemihydrate, monohydrate, dihydrate, trihydrate, tetrahydrate, and pentahydrate.
  • an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof has the same meaning as the phrase “(i) an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant of the compound referenced therein; (ii) a pharmaceutically acceptable salt, solvate, hydrate, or prodrug of the compound referenced therein; or (iii) a pharmaceutically acceptable salt, solvate, hydrate, or prodrug of an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant of the compound referenced therein.”
  • the stem cell mobilizing factor is a compound having Formula (I), (I- A), (I-B), (I-C), or (I-D), as described below.
  • [00204] can represent a single bond. In other embodiments, can represent a double bond. In some embodiments, joining Y and
  • joining Y and Z can represent a single bond. In other embodiments, joining Y and Z can represent a double bond. In some embodiments, when joining G and J representes a single bond,
  • G can be N and the N is substituted with R G .
  • G when joining G and J represents a double bond, G can be N.
  • joining G and J when joining G and J representes a double bond, then joining J and R J can be a single bond.
  • joining J and R J when . joining G and J representes a double bond, then joining J and R J can not be a double bond.
  • joining J and R J when joining G and J representes a double bond, then joining G and J can be a single bond.
  • joining J and R J when joining J and R J representes a double bond, then . joining G and J can not be a double bond.
  • G and J represents a single bond and G is N and the N is substituted with R G .
  • R a can be hydrogen. In some embodiments, R a can be C 1 -C 4 alkyl. For example, R a can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl.
  • R b can be R c .
  • R b can be -(C 1 - C4 alkyl)-R c .
  • R b can be -CH 2 -R C , -CH 2 CH 2 -R C ,
  • R c can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S.
  • the moiety when a R c moiety is indicated as substituted, the moiety can be substituted with one or more, for example, one, two, three, or four substituents E.
  • E can be -OH.
  • E can be C 1 -C 4 alkyl.
  • E can be C 1 -C 4 haloalkyl.
  • E can be -O(C 1 -C 4 alkyl).
  • E can be -O(C 1 -C 4 haloalkyl).
  • R c when R b is -CH 2 CH 2 -R C , R c can be unsubstituted C 6-10 aryl. In other embodiments, when R b is -CH 2 CH 2 -R C , R c can be substituted C 6-10 aryl. In still other embodiments, when R b is -CH 2 CH 2 -R C , R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S.
  • R b can be -(C 1 -C 4 alkyl)-R c and R c can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S.
  • R c moiety When a R c moiety is indicated as substituted, the moiety can be substituted with one or more, for example, one, two, three, or four substituents E.
  • E can be -OH.
  • E can be C 1 -C 4 alkyl. In still other embodiments, E can be C 1 -C 4 haloalkyl. In still other embodiments, E can be -O(C 1 -C 4 alkyl). In still other embodiments, E can be -O(C 1 -C 4 haloalkyl).
  • R c when R b is -CH 2 CH 2 -R C , R c can be phenyl. In other embodiments, when R b is -CH 2 CH 2 -R C , R c can be naphthyl. In still other embodiments, when R b is -CH 2 CH 2 -R C , R c can be hydroxyphenyl. In still other embodiments, when R b is - CH 2 CH 2 -R c , R C can be indolyl.
  • R K can be hydrogen. In other embodiments, R K can be unsubstituted C 1-6 alkyl.
  • R K can be methyl, ethyl, n- propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, pentyl (branched and straight-chained), or hexyl (branched and straight-chained).
  • R K can be substituted C 1-6 alkyl. In other embodiments, R K can be -NH(C 1-4 alkyl).
  • R K can be -NH(CH 3 ), -NH(CH 2 CH 3 ), -NH(isopropyl), or -NH(sec-butyl).
  • R K can be -N(C 1-4 alky 1)2.
  • R K can be unsubstituted C 6-10 aryl. In other embodiments, R K can be substituted C 6-10 aryl. In other embodiments, R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S. In other embodiments, R K can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S. When a R K moiety is indicated as substituted, the moiety can be substituted with one or more, for example, one, two, three, or four substituents substituents Q. In some embodiments, Q can be -OH.
  • Q can be C 1-4 alkyl. In still other embodiments, Q can be C 1-4 haloalkyl. In still other embodiments, Q can be halo. In still other embodiments, Q can be cyano. In still other embodiments, Q can be -O-(C 1-4 alkyl). In still other embodiments, Q can be -O-(C 1-4 haloalkyl).
  • R K can be phenyl or naphthyl. In other embodiments, R K can be benzothiophenyl. In other embodiments, R K can be benzothiophenyl. In other embodiments, R K can be benzothiophenyl. In still other embodiments, R K can be pyridinyl.
  • R K can be pyridinyl substituted with one or more substituents Q.
  • R K can be methylpyridinyl, ethylpyridinyl cyanopyridinyl, chloropyridinyl, fluoropyridinyl, or bromopyridinyl.
  • R Y and R Z can independently be absent. In other embodiments, R Y and R Z can independently be hydrogen. In other embodiments, R Y and R Z can independently be halo. In other embodiments, R Y and R Z can independently be C 1-6 alkyl. In other embodiments, R Y and R Z can independently be -OH. In still other embodiments, R Y and R Z can independently be -O-(C 1-4 alkyl). In other embodiments, R Y and R Z can independently be -NH(C 1-4 alkyl). For example, R Y and R Z can independently be
  • R Y and R Z can independently be - N(C 1-4 alkyl)2.
  • R Y and R Z taken together with the atoms to which they are attached can be joined together to form a ring. In some embodiments, R Y and R Z taken together with the atoms to which they are attached can be joined together to form In other embodiments, R Y and R Z taken together with the atoms to which they are attached can be joined together to form In other embodiments, R Y and R Z taken together with the atoms to which they are attached can be joined together to form In still other embodiments, R Y and R Z taken together with the atoms to which they are attached can be joined together to form In yet still other embodiments,
  • R Y and R Z taken together with the atoms to which they are attached can be joined together to form in other embodiments, R Y and R Z taken together with the atoms to which they are attached can be joined together to form In yet other embodiments, R Y and R Z taken together with the atoms to which they are attached can be joined together to form In yet still other embodiments, R Y and R Z taken together with the atoms to which they are attached can be joined together to form In other embodiments, R Y and R Z taken together with the atoms to which they are attached can be joined together to form In still other embodiments, R Y and R Z taken together with the atoms to which they are attached can be joined together to form and In some embodiments, when R Y and R Z taken together with the atoms to which they are attached can be joined together to form a ring, the ring can be substituted with one, two, or three groups independently selected from C 1 -C 4 alkyl, -N(C 1 -C 4 alkyl)2, cyan
  • R Y and R Z taken together with the atoms to which they are attached can be joined together to form In other embodiments, R Y and R Z taken together with the atoms to which they are attached can be joined together to form
  • R Y and R Z taken together with the atoms to which they are attached can be joined together to form In other embodiments, R Y and R Z taken together with the atoms to which they are attached can be joined together to form
  • R Y and R Z taken together with the atoms to which they are attached can be joined together to form In other embodiments, R Y and R Z taken together with the atoms to which they are attached can be joined together to form In other embodiments, R Y and R Z taken together with the atoms to which they are attached can be joined together to form In other embodiments, R Y and R Z taken together with the atoms to which they are attached can be joined together to form In other embodiments, R Y and R Z taken together with the atoms to which they are attached can be joined together to form
  • R Y and R Z taken together with the atoms to which they are attached can be joined together to form In other embodiments, R Y and R Z taken together with the atoms to which they are attached can be joined together to form In some embodiments, when R Y and R Z taken together with the atoms to which they are attached can be joined together to form a ring, the ring can be substituted with one, two, or three groups independently selected from C 1 -C 4 alkyl, -N(C 1 -C 4 alkyl)2, cyano, unsubstituted phenyl, and phenyl substituted with 1-5 halo atoms.
  • R Y and R Z taken together with the atoms to which they are attached can be In other embodiments, R Y and R Z taken together with the atoms to which they are attached can be In still other embodiments, R Y and R Z taken together with the atoms to which they are attached can be In yet still other embodiments, R Y and R Z taken together with the atoms to which they are attached can be In other embodiments, R Y and R Z taken together with the atoms to which they are attached can be
  • R d can be hydrogen. In other embodiments, R d can be C 1 -C 4 alkyl. For example R d can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl. In still other embodiments, R d can be halo. In other embodiments, R d can be cyano.
  • R m can be hydrogen. In other embodiments, R m can be C 1 -C 4 alkyl. For example R m can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl. In still other embodiments, R m can be halo. For example, R m can be fluoro, chloro, bromo, or iodo. In other embodiments, R m can be cyano.
  • X, Y, and Z can each be independently N or C, wherein the valency of any carbon atom is filled as needed with hydrogen atoms.
  • X can be N, Y can be N, and Z can be N.
  • X can be N, Y can be N, and Z can be CH.
  • X can be N, Y can be CH, and Z can be N.
  • X can be CH, Y can be N, and Z can be N.
  • X can be CH, Y can be CH, and Z can be N.
  • X can be CH, Y can be CH, and Z can be N.
  • X can be CH, Y can be CH, and Z can be N.
  • X can be CH, Y can be N, and Z can be CH.
  • X can be N, Y can be CH, and Z can be CH. In other embodiments, X can be CH, Y can be CH, and Z can be CH.
  • R a can be hydrogen;
  • R b can be -CH 2 CH 2 -R C ;
  • R K can be selected from the group consisting of: hydrogen, methyl, substituted pyridinyl, unsubstituted benzothiophenyl, and -NH(C 1 -C 4 alkyl);
  • R Y can be -NH(C 1 -C 4 alkyl);
  • R Z can be absent or hydrogen; or R Y and R Z taken together with the atoms to which they are attached can be joined together to form a ring
  • R d can be C 1 -C 4 alkyl
  • R m can be cyano
  • X can be N or CH.
  • R J when R J is -NR a R b ; G can be N; - j pining G and J can be a double bond; R a can hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; or R c can be substituted C 6-10 aryl, substituted with one or more E, wherein E is - OH; R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; or R K can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; substituted with one or more Q, wherein Q can be selected from cyano, halo, or C 1 -C 4 alky
  • R J when R J is -NR a R b ; G can be N; . j pining G and J can be a double bond; R a can hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; or R c can be substituted C 6-10 aryl, substituted with one or more E, wherein E is - OH; R K can be hydrogen, C 1-4 alkyl, or unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and R Y and R Z taken together can be [00226] In some embodiments, when R J is -NR a R b ; G can be N; - j pining G and J can be a double bond; R a can hydrogen
  • R J when R J is -NR a R b ; G can be N; j pining G and J can be a double bond, R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be substituted G,- lo aryl; substituted with one or more E, wherein E can be
  • R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S;
  • R Y can be -NH(C 1-4 alkyl);
  • R Z can be hydrogen; J can be C; X can be N; Y can be C; Z can be C; and joining Y and Z can be a double bond.
  • the compound of Formula (I) can be 4-(2-((2- (benzo[b]thiophen-3-yl)-6-(isopropylamino)pyrimidin-4-yl)amino)ethyl)phenol.
  • R J when R J is -NR a R b ; G can be N; . j pining G and J can be a double bond; R a can be hydrogen; R b can be -CH 2 CH 2 -R C , R c can be substituted G,- lo aryl, substituted with one or more E, wherein E can be
  • R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R Y and R Z taken together is wherein the ring is substituted with C 1 -C 4 alkyl; J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I) can be 4-(2-((2-(benzo[b]thiophen-3-yl)- 7-isopropylthieno[3,2-b]pyrimidin-4-yl)amino)ethyl)phenol.
  • R J when R J is -NR a R b ; G can be N; - joining G and J can be a double bond; R a can be hydrogen; R b can be -CH 2 CH 2 -R C , R c can be substituted G,- 10 aryl, substituted with one or more E, wherein E can be -OH; R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R Y and R Z taken together is R d can be C 1 -C 4 alkyl; J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I) can be 4-(2- ((2-(benzo[b]thiophen-3-yl)-7-isopropyl-6.7-dihydro-5H-pyrrolo[2,3-d]pyri midin-4- yl)amino)ethyl)phenol.
  • R J when R J is -NR a R b ; G can be N; joining G and J can be a double bond; R a can be hydrogen; R b can be -CH 2 CH 2 -R C , R c can be substituted G,- 10 aryl, substituted with one or more E, wherein E can be
  • R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R Y and R Z taken together is R d can be C 1 -C 4 alkyl; J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I) can be 2-(benzo[b]thiophen-3-yl)-4-((4- hydroYyphenethyl)amino)-7-isopropyl-5.7-dihydro-6H-pyrrolo[2,3-d]pyrimidin-6-one.
  • R J when R J is -OR b ; G can be N; joining G and J can be a double bond; R b can be -CH 2 CH 2 -R C ; R c can be
  • R K can unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R Y and R Z taken together can be R d can be C 1 -C 4 alkyl; J can be C; X can be N; Y can be C; and Z is C.
  • the compound of Formula (I) can be 3-((2-(benzo[b]thiophen-3-yl)-9- isopropyl-9H-purin-6-yl)oYy)propanamide.
  • R J when R J is is -NR a R b ; G can be N; - joining G and J can be a double bond; R b can be -CH 2 CH 2 -R C ; R c can be substituted C 6-10 aryl, substituted with one or more E, wherein E is -OH; R K is unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R Y and R Z taken together can ; wherein said ring is substituted with -N(C 1-4 alkyl)2; J can be C; X can be N; Y can be C; and Z is C.
  • the compound of Formula (I) can be 4-(2-((2-(benzo[b]thiophen-3-yl)-8-(dimethylamino)pyrimido[5,4-ri]pyrimidin-4- yl)amino)ethyl)phenol.
  • the compound of Formula (I) can be 5-(2-((2- (1H-indol-3-yl)ethyl)amino)-6-(sec-butylamino)pyrimidin-4-yl)nicotinonitrile.
  • R J when R J is -NR a R b ; G can be N; . j pining G and J can be a double bond; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O,
  • R K can be unsubstituted C 1-6 alkyl
  • R Y and R Z taken together can wherein the ring is substituted with unsubstituted C 6 -C 10 aryl
  • J can be C
  • X can be N
  • Y can be C
  • Z can be C.
  • the compound of Formula (I) can be N-(2-(lH- indol-3-yl)ethyl)-2-methyl-6-phenylthieno[2,3-d]pyrimidin-4-amine
  • R J when R J can be -NR a R b ; G can be N; - joining G and J can be a double bond; R a can be hydrogen; R b can be
  • R C can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S;
  • R K can be hydrogen;
  • R Y and R Z taken together can ; wherein the ring is substituted with substituted C 6 -C 10 aryl; J can be C;
  • X can be N;
  • Y can be C; and
  • Z can be C.
  • the compound of Formula (I) can be N-(2-(1H-indol-3-yl)ethyl)-6-(4-fluorophenyl)thieno[2,3- ⁇ f]pyrimi din-4- amine
  • R J O
  • G can be N substituted with R G ; — joining G and J can be a single bond;
  • R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group i consisting of O, N, and S;
  • R Y and R Z taken together can be R d can be C 1 -C 4 alkyl; J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I) can be 3-(2-(benzo[b]thiophen-3-yl)-9-isopropyl-6-oxo-6,9- dihydro- lif-purin- 1 -yljpropan amide.
  • R J when R J is -NR a R b ; G can be N; - j pining G and J can be a double bond R a can be hydrogen R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R K moiety indicated as substituted is substituted with one or more Q, wherein Q can be halo; R Y and R Z taken together can be J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I) can be N-(2-(1H-indol-3-yl)ethyl)-2-(5-fluorop
  • R J when R J is -NR a R b ; G is N; - joining G and J can be a double bond; R a can be hydrogen R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R K moiety indicated as substituted is substituted with one or more Q, wherein Q can be cyano; R Y and R Z taken together is J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I) can be 5-(4-((2-(1H-indol-3-yl)ethyl)amino)quinazolin-2-yl)nicotinonitrile.
  • R J when R J is -NR a R b ; G can be N; - j pining G and J can be a double bond; R a can be hydrogen R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O,
  • R K can be -NH(C 1-4 alkyl); R Y and R Z taken together can be : J can be
  • the compound of Formula (I) can be N 4 -(2-(1H-indol-3-yl)ethyl)-N 2 -(sec-butyl)quinazoline-2, 4-diamine.
  • R J when R J is -NR a R b ; G can be N; . joining G and
  • J can be a double bond
  • R a can be hydrogen
  • R b can be -CH 2 CH 2 -R C
  • R c can be substituted C 6-10 aryl, substituted with one or more E, wherein E is -OH
  • R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and
  • R Y and R Z taken together can be ; wherein the ring is substituted with cyano;
  • R d can be C 1 -C 4 alkyl; J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I) can be 2-(benzo[b]thiophen-3-yl)-4-((4- hydro ⁇ yphenethyl)amino)-7-isopropyl-7H-pyrrolo[2,3-d]pyrimidine-5-carbonitrile.
  • R J when R J is -NR a R b ; G can be N; . j pining G and J can be a double bond; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R Y and R Z taken together can be ; wherein the ring is substituted with C 1-4 alkyl; J can be C; X can be C; Y can be N; and Z can be C; wherein the valency of any carbon atom is filled as needed with hydrogen atoms.
  • the compound of Formula (I) can be N-(2-(1H-indol-3- yl)ethyl)-6-(benzo[b]thiophen-3-yl)-3-isopropylimidazo[1,5- ⁇ ]pyrazin-8-amine.
  • R J when R J is -NR a R b ; G can be N; - joining G and J can be a double bond; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be substituted C 6 - 10 aryl, substituted with one or more E, wherein E is -OH; R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S;
  • R Y and R Z taken together can be wherein the ring can be substituted with C 1-4 alkyl; J can be C; X can be C; Y can be N; and Z can be C; wherein the valency of any carbon atom is filled as needed with hydrogen atoms.
  • the compound of Formula (I) can be 4-(2-((6-(benzo[b]thiophen-3-yl)-3-isopropylimidazo[1,5-a]pyrazin-8- yl)amino)ethyl)phenol.
  • R J when R J is -NR a R b ; G can be N; . j pining G and J represents a double bond; R a can be hydrogen R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R K moiety indicated as substituted is substituted with one or more Q, wherein Q is cyano; R Y and R Z taken together is ; wherein the ring is substituted with C 1 -C 4 alkyl; J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I) can be 5-(4-((2- (1H-indol-3-yl)ethyl)amino)-7-isopropylthieno[2,3-d] pyrimi din-2 -yl)nicotinonitrile.
  • R J when R J is -NR a R b ; G can be N; j pining G and J represents a double bond; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R K moiety indicated as substituted is substituted with one or more Q, wherein Q is halo; R Y and R Z taken together can be ; wherein the ring is substituted with C 1 -C 4 alkyl; J can be C; X can be N;
  • Y can be C; and Z can be C.
  • the compound of Formula (I) can be N- (2-(1H-indol-3-yl)ethyl)-2-(5-fluoropyridin-3-yl)-7-isopropylthieno[2,3-d]pyrimi din-4- amine.
  • R J when R J is -NR a R b ; G can be N; . j pining G and J can be a double bond; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R K moiety indicated as substituted is substituted with one or more Q, wherein Q is halo; R Y and R Z taken together can be J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I) can beN-(2-(1H-indol-3-yl)ethyl)-2-(5-fluoropyridin-3-yl)furo[3,2- d ⁇ py rimidin-4-amine.
  • R J when R J is -NR a R b ; G can be N; - joining G and J can be a double bond; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R K moiety indicated as substituted is substituted with one or more Q, wherein Q is C 1 -C 4 alkyl; R Y and R Z taken together can be J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I) can be N-(2-(1H-indol-3-yl)ethyl)-2-(5-methylpyridin-3- yl)furo[2,3-d]pyrimidin-4-amine.
  • R J when R J is -NR a R b ; G can be N; - joining G and J can be a double bond; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R K moiety indicated as substituted is substituted with one or more Q, wherein Q is C 1 -C 4 alkyl; R Y and R Z taken together can be wherein the ring is substituted with C 1 -C 4 alkyl J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I) can be N-(2-(lH- indol-3-yl)ethyl)-7-isopropyl-2-(5-methylpyridin-3-yl)thieno[2,3-d]pyrimidin-4-amine.
  • R J when R J is -NR a R b ; G is N; - joining G and J can be a double bond; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R K moiety indicated as substituted is substituted with one or more Q, wherein Q is cyano; R Y and R Z taken together can be J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I) can be 5-(4-((2-(1H-indol-3-yl)ethyl)amino)furo[2,3-d]pyrimidin-2- yljnicotinonitrile.
  • compound of Formula (I) wherein the compound can be selected from:
  • the compound of Formula (I) can have the structure of Formula (I-A): (I-A), including pharmaceutically acceptable salts thereof, wherein: R J can be -NR a R b ; R a can be hydrogen or C 1 -C 4 alkyl; R b can be R c or -(C 1 -C 4 alkyl)-R c ; R c can be selected from the group consisting of: unsubstituted C 6-10 aryl; substituted C 6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R c moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of:
  • R a can be hydrogen. In other embodiments, R a can be C 1 -C 4 alkyl.
  • R b can be -(C 1 -C 4 alkyl)-R c .
  • R b can be -
  • R c can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S.
  • R c moiety when a R c moiety is indicated as substituted, the moiety can be substituted with one or more, for example, one, two, three, or four substituents E.
  • E can be
  • E can be C 1 -C 4 alkyl. In some embodiments, E can be C 1 -C 4 haloalkyl. In some embodiments, E can be -O(C 1 -C 4 alkyl). In some embodiments, E can be -O(C 1 -C 4 haloalkyl). In some embodiments R c can be phenyl. In other embodiments, R c can be hydroxyphenyl. In still other embodiments, R c can be indolyl.
  • R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S.
  • R K can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein the substituted heteroaryl can substituted with one or more substituents Q, wherein each Q can independently selected from the group consisting of: -OH, C 1-4 alkyl, C 1-4 haloalkyl, halo, cyano, -O-(C 1-4 alkyl), and -O- (C 1-4 haloalkyl).
  • R K can be pyridinyl. In other embodiments, R K can be pyridinyl substituted with one or more substituents Q. For example, R K can be methylpyridinyl, ethylpyridinyl cyanopyridinyl, chloropyridinyl, fluoropyridinyl, or bromopyridinyl.
  • R e can be hydrogen. In some embodiments, R e can be C 1 -C 4 alkyl. For example, R e can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl.
  • R a can be hydrogen;
  • R b can be -(C 1 -C 4 alkyl)-R c ;
  • R c can be selected from the group consisting of: unsubstituted C 6-10 aryl; substituted C 6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R c moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C 1 -C 4 alkyl, C 1 -C 4 haloalkyl, - O(C 1 -C 4 alkyl), and -O(C 1 -C 4 haloalkyl);
  • R K can be selected from the group consisting
  • R a can be hydrogen;
  • R b can be -(CH 2 -CH 2 )-R C ;
  • R c can be selected from the group consisting of: substituted phenyl and unsubstituted indolyl; wherein the substituted phenyl is substituted with one substituent E, wherein E can be -OH;
  • R K can be selected from the group consisting of: unsubstituted benzothiophenyl and substituted pyridinyl; wherein the substituted pyridinyl is substituted with one substituent Q, wherein Q can be selected from the group consisting of: C 1-4 alkyl, halo, and cyano; and
  • R e can be isopropyl.
  • R J when W is O, R J can be -NR a R b ; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be selected from the group consisting of: unsubstituted C 6-10 aryl; substituted C 6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R c moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C 1 -C 4 alkyl, and -O(C 1 -C 4 alkyl); R K can be selected from the group consisting of unsubstituted five- to ten-membere
  • R J when W is S, R J can be -NR a R b ; R a can be hydrogen; R b can be -CH 2 CH 2 -R c ; R c can be selected from the group consisting of: unsubstituted C 6-10 aryl; substituted C 6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R c moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C 1 -C 4 alkyl, and -O(C 1 -C 4 alkyl); R K can be selected from the group consisting of unsubstituted five- to ten-member
  • R J when R J is -NR a R b ; G can be N; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R K moiety indicated as substituted is substituted with one or more Q, wherein Q is C 1 -C 4 alkyl; W can be S; R e can be C 1 -C 4 alkyl; J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I-A) can be Y-(2-(1H-indol-3- yl)ethyl)-7-isopropyl-2-(5-methylpyridin-3-yl)thieno[2,3-d] pyrimidin-4-amine.
  • R J when R J is -NR a R b ; G can be N; R a can be hydrogen R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R K moiety indicated as substituted is substituted with one or more Q, wherein Q is cyano; W can be S; R e can be C 1 -C 4 alkyl; J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I- A) can be 5-(4-((2-(1H-indol-3- yl)ethyl)amino)-7-isopropylthieno[2,3-d] pyrimi din-2 -yl)nicotinonitrile.
  • R J when R J is -NR a R b ; G can be N; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R K moiety indicated as substituted is substituted with one or more Q, wherein Q is halo; W can be S; R e can be C 1 -C 4 alkyl; J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I- A) can be N-(2-(1H-indol-3-yl)ethyl)- 2-(5-fluoropyridin-3-yl)-7-isopropylthieno[2,3-d]pyrimidin-4-amine.
  • R J when R J is -NR a R b ; G can be N; R a can be hydrogen; R b can be -CH 2 CH 2 -R C , R c can be substituted C 6-10 aryl, substituted with one or more E, wherein E can be -OH; R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; W can be S; R e can be C 1 -C 4 alkyl;
  • the compound of Formula (I-A) can be 4-(2-((2-(benzo[b]thiophen-3-yl)-7-isopropylthieno[2,3-d] pyrimidin- 4-yl)amino)ethyl)phenol.
  • R J when R J is -NR a R b ; G can be N; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R K moiety indicated as substituted is substituted with one or more Q, wherein Q is halo; W can be O; R e can be hydrogen; J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I-A) can be Y-(2-(1H-indol-3-yl)ethyl)-2- (-5fluoropyridin-3-yl)furo[2,3-d]pyrimidin-4-amine.
  • R J when R J is -NR a R b ; G can be N; . j pining G and J can be a double bond; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R K moiety indicated as substituted is substituted with one or more Q, wherein Q is C 1 -C 4 alkyl; W can be O; R e can be hydrogen;
  • the compound of Formula (I-A) can be N-(2-(1H-indol-3-yl)ethyl)-2-(5-methylpyridin-3-yl)furo [3.2- d] pyrimidin-4-amine.
  • R J when R J is -NR a R b ; G is NR a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R K moiety indicated as substituted is substituted with one or more Q, wherein Q is cyano; W can be O; R e can be hydrogen; J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I- A) can be 5-(4-((2-(1H-indol-3- yl)ethyl)amino)furo[2,3-d] pyrimidin-2-yl)nicotinonitrile.
  • the compound of Formula (I- A), or a pharmaceutically acceptable salt thereof can selected from the group consisting of:
  • R a can be hydrogen. In other embodiments, R a can be C 1 -C 4 alkyl.
  • R b can be -(C 1 -C 4 alkyl)-R c .
  • R b can be - CH 2 -R c , -CH 2 CH 2 -R c , -CH 2 CH 2 CH 2 -R c , or -CH 2 CH 2 CH 2 CH 2 -R C .
  • R b can be -(CH 2 CH 2 )-R C .
  • R b can be
  • R b can be -(CH 2 CH 2 )-(indolyl). In certain embodiments, R b can be -(CH 2 CH 2 )-(hydroxyphenyl).
  • R c can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S.
  • the moiety when a R c moiety is indicated as substituted, the moiety can be substituted with one or more, for example, one, two, three, or four substituents E.
  • E can be -OH.
  • E can be C 1 -C 4 alkyl.
  • E can be C 1 -C 4 haloalkyl.
  • E can be -O(C 1 -C 4 alkyl).
  • E can be -O(C 1 -C 4 haloalkyl).
  • R K can be hydrogen. In other embodiments, R K can be C 1 -C 4 alkyl.
  • R K can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl.
  • R K can be selected from the group consisting of: unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein the substituted heteroaryl can substituted with one or more substituents Q, wherein each Q can independently selected from the group consisting of: -OH, C 1-4 alkyl, C 1-4 haloalkyl, halo, cyano, -O-(C 1-4 alkyl), and -O- ( C 1-4 haloalkyl).
  • R K can be benzothiophenyl.
  • R K can be pyridinyl substituted with one or more substituents Q.
  • R K can be methylpyridinyl, ethylpyridinyl cyanopyridinyl, chloropyridinyl, fluoropyridinyl, or bromopyridinyl.
  • R f can be hydrogen. In other embodiments, R f can be C 1-4 alkyl.
  • R f can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl.
  • R f can be unsubstituted C 6 -C 10 aryl.
  • R f can be C 6 -C 10 aryl substituted with 1-5 halo atoms.
  • R f can be phenyl substituted with 1-5 halo atoms.
  • R f can be fluorophenyl.
  • U can be N. In other embodiments, U can be CR U .
  • V can be S. In other embodiments, V can be NR V .
  • R U can be hydrogen. In some embodiments, R U can be C 1-4 alkyl. In other embodiments R U can be halo. For example, R U can be fluoro, chloro, bromo, or iodo. In still other embodiments, R U can be cyano.
  • R V can be hydrogen. In other embodiments, R V can be C 1-4 alkyl. For example, R V can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl.
  • Y and Z can each be C and X can be N. In other embodiments, Y and Z can each be C and X can be CH.
  • R f can be selected from the group consisting of hydrogen, unsubstituted phenyl, and phenyl substituted with 1-5 halo atoms; Y and Z each can be C; and X can be CH.
  • R a can be hydrogen;
  • R b can be -(CH 2 -CH 2 )-R C ;
  • R K can be selected from the group consisting of: unsubstituted benzothiohenyl and substituted pyridinyl; wherein the substituted pyridinyl is substituted with one substituent Q, wherein Q can be selected from the group consisting of: C 1-4 alkyl, halo, and cyano;
  • R f can be selected from the group consisting of hydrogen, phenyl, and fluorophenyl;
  • Y and Z each can be C; and
  • R J when R J is -OR b ; G can be N; - joining G and J can be a double bond; R b can be -CH 2 CH 2 -R C ; R c can be
  • R K can unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; U can N; V can be NR v ; R v can be C 1 -C 4 alkyl; R f can be hydrogen; J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I-B) can be 3-((2-(benzo[b]thiophen-3-yl)-9- isopropyl-9H-purin-6-yl)o ⁇ y)propan amide.
  • R J O
  • G can be N substituted with R G ; - joining G and J can be a single bond;
  • R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S;
  • U can N;
  • V can be NR v ;
  • R v can be C 1 -C 4 alkyl;
  • R f can be hydrogen; J can be C;
  • X can be N;
  • the compound of Formula (I-B) can be 3-(2-(benzo[b]thiophen-3-yl)-9-isopropyl-6-oxo-6,9- dihydro- lH-purin- 1 -yl)propanamide.
  • R J when R J is -NR a R b ; G can be N; . joining G and
  • J can be a double bond;
  • R a can be hydrogen;
  • R b can be -CH 2 CH 2 -R C ;
  • R c can be substituted C 6-10 aryl, substituted with one or more E, wherein E is -OH;
  • R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S;
  • U can be CR U ;
  • R u can be cyano;
  • V can be NR v ;
  • R v can be C 1 -C 4 alkyl;
  • R f can be hydrogen;
  • J can be C;
  • X can be N;
  • the compound of Formula (I-B) can be 2-(benzo[b]thiophen-3-yl)-4-((4-hydroxyphenethyl)amino)-7- isopropyl-7H-pyrrolo[2,3-d]pyrimidine-5-carbonitrile.
  • R J when R J is -NR a R b ; G can be N; . j pining G and J can be a double bond; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be unsubstituted C 1-6 alkyl; U can be CR U ; R u can be hydrogen; V can be S; R f can be phenyl; J can be C; X can be N; Y can be C; Z can be C.
  • the compound of Formula (I-B) can be N-(2-(1H-indol-3-yl)ethyl)-2-methyl-6- pheny lthieno [2,3 -d ⁇ pyrimidin-4-amine.
  • R J when R J can be -NR a R b ; G can be N; . joining G and J can be a double bond; R a can be hydrogen; R b can be
  • R C can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S;
  • R K can be hydrogen; U can be CR U ; R u can be hydrogen; V can be S; R f can be fluorophenyl; J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I-B) can be N-(2-(1H- indol-3-yl)ethyl)-6-(4-fluorophenyl)thieno[2,3-d]pyrimidin-4-amine.
  • the compound of Formula (I-B), or a pharmaceutically acceptable salt thereof can selected from the group consisting of: 3-((2-(benzo[b]thiophen-3-yl)-9-isopropyl-9H-purin-6-yl)o ⁇ y)propanamide:
  • the compound of Formula (I) can have the structure of Formula (I-C): (I-C), including pharmaceutically acceptable salts thereof, wherein: R J can be -NR a R b ; R a can be hydrogen or C 1 -C 4 alkyl; R b can be R c or -(C 1 -C 4 alkyl)-R c ; R c can be selected from the group consisting of: unsubstituted C 6-10 aryl; substituted C 6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R c moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of
  • R K can be -NH(C 1-4 alkyl).
  • R K can be -NH(CH 3 ), -NH(CH 2 CH 3 ), -NH(isopropyl), or -NH(sec-butyl).
  • R K can be unsubstituted benzothiophenyl.
  • R K can be substituted pyridinyl.
  • R K can be methylpyridinyl, ethylpyridinyl, cyanopyridinyl, chloropyridinyl, fluoropyridinyl, or bromopyridinyl.
  • A can be N and B can be N. In other embodiments, A can be N and B can be CH. In still other embodiments, A can be CH and B can be N. In yet still other embodiments, A can be CH and B can be CH. [00292] In some embodiments, R 8 can be hydrogen. In other embodiments, R 8 can be -
  • R 8 can be
  • R a can be hydrogen;
  • R b can be -(C 1 -C 4 alkyl)-R c ;
  • R c can be selected from the group consisting of: unsubstituted C 6-10 aryl; substituted C 6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R c moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C 1 -C 4 alkyl, C 1 -C 4 haloalkyl, - O(C 1 -C 4 alkyl), and -O(C 1 -C 4 haloalkyl);
  • R K can be selected from the
  • R a can be hydrogen;
  • R b can be -(C 1 -C 4 alkyl)-R c ;
  • R c can be selected from the group consisting of: substituted phenyl and unsubstituted indolyl; wherein the substituted phenyl is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C 1 -C 4 alkyl, C 1 -C 4 haloalkyl, -O(C 1 -C 4 alkyl), and -O(C 1 -C 4 haloalkyl);
  • R K can be selected from the group consisting of: -NH(C 1-4 alkyl); unsubstituted benzothiophenyl; and substituted pyridinyl; wherein the substituted pyridinyl is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: -
  • R a can be hydrogen;
  • R b can be -(CH 2 CH 2 )-R C ;
  • R c can be selected from the group consisting of: substituted phenyl and unsubstituted indolyl; wherein the substituted phenyl is substituted with one substituent E, wherein E can be -OH;
  • R K can be selected from the group consisting of: -NHfsec-butyl): unsubstituted benzothiohenyl, and substituted pyridinyl; wherein the substituted pyridinyl is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: C 1-4 alkyl, halo, and cyano; and
  • R 8 can be hydrogen or -N(CH 3 ) 2 .
  • R J when A is C and B is C, R J can be -NR a R b ; G can be N; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be substituted C 6 - 10 aryl, substituted with one or more E, wherein E is -OH; or unsubstituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R 8 can be hydrogen; J can be C; X can be N; Y can be C; and Z is C.
  • R J when R J is -NR a R b ; G can be N; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be substituted C 6-10 aryl, substituted with one or more E, wherein E is -OH; R K is unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; A can be N; B can be N; R 8 can be -N(C 1-4 alkyl)2; J can be C; X can be N; Y can be C; and Z is C.
  • the compound of Formula (I-C) can be 4-(2-((2-(benzo[b]thiophen-3-yl)-8- (dimethylamino)pyrimido
  • R J when R J is -NR a R b ; G can be N; R a can be hydrogen R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R K moiety indicated as substituted is substituted with one or more Q, wherein Q can be halo; A can be CH; B can be CH; R 8 can be hydrogen; J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I-C) can be N-( 2- (1H-indol-3-yl)ethyl)-2-(5-fluoropyridin-3-yl)
  • R J when R J is -NR a R b ; G is N; - joining G and J can be a double bond; R a can be hydrogen R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R K moiety indicated as substituted is substituted with one or more Q, wherein Q can be cyano; A can be CH; B can be CH; R 8 can be hydrogen; J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I-C) can be 5-(4-((2-(1H-indol-3-yl)ethyl)amino)quinazolin-2- yl)nicotinonitrile.
  • R J when R J is -NR a R b ; G can be N; - joining G and J can be a double bond; R a can be hydrogen R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R K can be -NH(C 1-4 alkyl); A can be CH; B can be CH; R g can be hydrogen; J can be C; X can be N; Y can be C; and Z can be C.
  • the compound of Formula (I-C) can be N 4 -(2-(1H-indol-3-yl)ethyl)-N 2 -(sec-butyl)quinazoline-2, 4-diamine.
  • the compound of Formula (I-C), or a pharmaceutically acceptable salt thereof can selected from the group consisting of:
  • the compound of Formula (I) can have the structure of Formula (I-D): (I-D), including pharmaceutically acceptable salts thereof, wherein: R J can be -NR a R b ; R a can be hydrogen or C 1 -C 4 alkyl; R b can be R c or -(C 1-4 alkyl)-R c ; R c can be selected from the group consisting of: unsubstituted C 6-10 aryl; substituted C 6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R c moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of:
  • R h can be hydrogen. In other embodiments, R h can be C 1-4 alkyl.
  • R h can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl.
  • D can be N. In other embodiments, D can be CH. [00305] In some embodiments, when D is N, Y can be N, Z can be C, and X can be N. In other embodiments, when D is N, Y can be N, Z can be C, and X can be CH. In some embodiments, when D is CH, Y can be N, Z can be C, and X can be N. In other embodiments, when D is CH, Y can be N, Z can be C, and X can be CH.
  • R a can be hydrogen;
  • R b can be -(C 1-4 alkyl)-R c ;
  • R c can be selected from the group consisting of: unsubstituted C 6-10 aryl; substituted C 6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a R c moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C 1 -C 4 alkyl, C 1 -C 4 haloalkyl, - O(C 1 -C 4 alkyl), and -O(C 1 -C 4 haloalkyl);
  • R K can be selected from the group consisting
  • R a can be hydrogen;
  • R b can be -(C 1 -C 4 alkyl)-R c ;
  • R c can be selected from the group consisting of: substituted phenyl and unsubstituted indolyl; wherein the substituted phenyl is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C 1 -C 4 alkyl, C 1 -C 4 haloalkyl, -O(C 1 -C 4 alkyl), and -O(C 1 -C 4 haloalkyl);
  • R K can be unsubstituted benzothiophenyl; and
  • R b can be hydrogen or C 1-4 alkyl.
  • R a can be hydrogen;
  • R b can be -(CH 2 -CH 2 )-R C ;
  • R c can be selected from the group consisting of: substituted phenyl and unsubstituted indolyl; wherein the substituted phenyl is substituted with one substituent E, wherein E can be -OH;
  • R K can be unsubstituted benzothiophenyl; and
  • R b can be hydrogen or C 1-4 alkyl.
  • R J when D is N; R J is -NR a R b ; G can be N; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; or substituted C 6-10 aryl, substituted with one or more E, wherein E is -OH; R K can be unsubstituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R b can be C 1-4 alkyl; J can be C; X can be C; Y can be N; and Z can be C; wherein the valency of any carbon atom is filled as needed with hydrogen atoms.
  • R J when R J is -NR a R b ; G can be N; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S or substituted C 6-10 aryl, substituted with one or more E, wherein E is -OH; R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; D can be N; R b can be C 1-4 alkyl; J can be C; X can be C; Y can be N; and Z can be C; wherein the valency of any carbon atom is filled as needed with hydrogen atoms.
  • the compound of Formula (I-D) can beN-(2-(1H-indol-3-yl)ethyl)-6-(benzo[b]thiophen-3- yl)-3-isopropylimidazo[1,5- ⁇ ]pyrazin-8-amine.
  • R J when R J is -NR a R b ; G can be N; - joining G and J can be a double bond; R a can be hydrogen; R b can be -CH 2 CH 2 -R C ; R c can be substituted G,- lo aryl, substituted with one or more E, wherein E is -OH; R K can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; D can be N; R b can be C 1-4 alkyl; J can be C; X can be C; Y can be N; and Z can be C; wherein the valency of any carbon atom is filled as needed with hydrogen atoms.
  • the compound of Formula (I-D) can be 4-(2-((6-(benzo[b]thiophen-3-yl)-3- isopropybmidazo[1,5- ⁇ ]pyrazin-8-yl)amino)ethyl)phenol.
  • the compound of Formula (I-D), or a pharmaceutically acceptable salt thereof can selected from the group consisting of:
  • the compounds provided herein may be enantiomerically pure, such as a single enantiomer or a single diastereomer, or be stereoisomeric mixtures, such as a mixture of enantiomers, e.g., a racemic mixture of two enantiomers; or a mixture of two or more diastereomers.
  • a compound in its (R) form is equivalent, for compounds that undergo epimerization in vivo, to administration of the compound in its ( S) form.
  • Conventional techniques for the preparation/isolation of individual enantiomers include synthesis from a suitable optically pure precursor, asymmetric synthesis from achiral starting materials, or resolution of an enantiomeric mixture, for example, chiral chromatography, recrystallization, resolution, diastereomeric salt formation, or derivatization into diastereomeric adducts followed by separation.
  • NK cells can be isolated or enriched, for example, by staining cells, in one embodiment, with antibodies to CD56 and CD3, and selecting for CD56 CD3 cells.
  • the NK cells are enriched for CD56 CD3 cells in comparison with total cells produced using the three-stage method, described herein.
  • NK cells e.g., cells produced using the three-stage method, described herein, can be isolated using a commercially available kit, for example, the NK Cell Isolation Kit (Miltenyi Biotec).
  • NK cells e.g., cells produced using the three-stage method, described herein
  • NK cells e.g., cells produced using the three-stage method, described herein
  • Negative isolation can be carried out using a commercially available kit, e.g., the NK Cell Negative Isolation Kit (Dynal Biotech). Cells isolated by these methods may be additionally sorted, e.g., to separate CDlla+ and CDlla- cells, and/or CD117+ and CD117- cells, and/or CD16 + and CD16 cells, and/or CD94 + and CD94+ In certain embodiments, cells, e.g., cells produced by the three-step methods described herein, are sorted to separate CD11a+ and CD11a- cells. In specific embodiments, CD11a+ cells are isolated. In certain embodiments, the cells are enriched for CD11a + cells in comparison with total cells produced using the three-stage method, described herein.
  • a commercially available kit e.g., the NK Cell Negative Isolation Kit (Dynal Biotech). Cells isolated by these methods may be additionally sorted, e.g., to separate CDlla+ and CDlla- cells, and
  • CD11a- cells are isolated. In certain embodiments, the cells are enriched for CD11a- cells in comparison with total cells produced using the three-stage method, described herein. In certain embodiments, cells are sorted to separate CD117+ and CD117- cells. In specific embodiments, CD117+ cells are isolated. In certain embodiments, the cells are enriched for CD117 + cells in comparison with total cells produced using the three-stage method, described herein. In specific embodiments, CD117- cells are isolated. In certain embodiments, the cells are enriched for CD117- cells in comparison with total cells produced using the three- stage method, described herein. In certain embodiments, cells are sorted to separate CD16 + and C D 16 cells. In specific embodiments, CD16 + cells are isolated.
  • the cells are enriched for CD16 + cells in comparison with total cells produced using the three-stage method, described herein.
  • C D 16 cells are isolated.
  • the cells are enriched for CD 16- cells in comparison with total cells produced using the three-stage method, described herein.
  • cells are sorted to separate CD94 + and CD 94 cells.
  • CD94 + cells are isolated.
  • the cells are enriched for CD94 + cells in comparison with total cells produced using the three-stage method, described herein.
  • CD 94 cells are isolated.
  • the cells are enriched for CD94- cells in comparison with total cells produced using the three-stage method, described herein.
  • isolation is performed using magnetic separation.
  • isolation is performed using flow cytometry.
  • ILC3 cells can be isolated or enriched, for example, by staining cells, in one embodiment, with antibodies to CD56, CD3, and CDlla, and selecting for CD56 CD3 C D 1 1 a cells.
  • ILC3 cells e.g., cells produced using the three-stage method, described herein, can also be isolated or enriched by removal of cells other than ILC3 cells in a population of cells that comprise the ILC3 cells, e.g., cells produced using the three-stage method, described herein.
  • ILC3 cells e.g., cells produced using the three-stage method, described herein, may be isolated or enriched by depletion of cells displaying non-ILC3 cell markers using, e.g., antibodies to one or more of CD3, CD4, CDlla, CD14, CD19, CD20, CD36, CD66b, CD94, CD123, HLA DR and/or CD235a (glycophorin A). Cells isolated by these methods may be additionally sorted, e.g., to separate CD117 + and C D 1 17 cells.
  • NK cells can be isolated or enriched, for example, by staining cells, in one embodiment, with antibodies to CD56, CD3, CD94, and CDlla, and selecting for CD56 + CD3-CD94 + CDlla + cells.
  • NK cells e.g., cells produced using the three-stage method, described herein, can also be isolated or enriched by removal of cells other than NK cells in a population of cells that comprise the NK cells, e.g., cells produced using the three-stage method, described herein.
  • the NK cells are enriched for C D56 1 C D3 C D94 1 C D 1 la + cells in comparison with total cells produced using the three-stage method, described herein.
  • ILC3 cells are isolated or enriched by selecting for CD56 CD3-CD1 la ⁇ cells. In certain embodiments, the ILC3 cells are enriched for CD56 CD3-CD1 la ⁇ cells in comparison with total cells produced using the three-stage method, described herein. In one embodiment, ILC3 cells are isolated or enriched by selecting for CD56 CD3-CD1 1 a C D 117+ cells. In certain embodiments, the ILC3 cells are enriched for CD56 + CD3-CD11a CD117+ cells in comparison with total cells produced using the three-stage method, described herein. In one embodiment, ILC3 cells are isolated or enriched by selecting for CD56 + CD3-CD11a CD117 + CDIL1R1 + cells. In certain embodiments, the ILC3 cells are enriched for CD56 + CD3-CDlla CD117 + CDIL1R1 + cells in comparison with total cells produced using the three-stage method, described herein.
  • NK cells are isolated or enriched by selecting for CD56 CD3-CD94 CD1 la + cells. In certain embodiments, the NK cells are enriched for CD56 CD3-CD94 CD1 la + cells in comparison with total cells produced using the three- stage method, described herein. In one embodiment, NK cells are isolated or enriched by selecting for CD56 + CD3-CD94 + CD11a + CDl 17 cells. In certain embodiments, the NK cells are enriched for CD56 + CD3-CD94 + CD11a + CDl 17 “ cells in comparison with total cells produced using the three-stage method, described herein.
  • Cell separation can be accomplished by, e.g., flow cytometry, fluorescence- activated cell sorting (FACS), or, in one embodiment, magnetic cell sorting using microbeads conjugated with specific antibodies.
  • the cells may be isolated, e.g., using a magnetic activated cell sorting (MACS) technique, a method for separating particles based on their ability to bind magnetic beads (e.g., about 0.5-100 pm diameter) that comprise one or more specific antibodies, e.g., anti-CD56 antibodies.
  • Magnetic cell separation can be performed and automated using, e.g., an AUTOMACSTM Separator (Miltenyi).
  • a variety of useful modifications can be performed on the magnetic microspheres, including covalent addition of antibody that specifically recognizes a particular cell surface molecule or hapten.
  • the beads are then mixed with the cells to allow binding.
  • Cells are then passed through a magnetic field to separate out cells having the specific cell surface marker.
  • these cells can then isolated and re-mixed with magnetic beads coupled to an antibody against additional cell surface markers.
  • the cells are again passed through a magnetic field, isolating cells that bound both the antibodies.
  • Such cells can then be diluted into separate dishes, such as microtiter dishes for clonal isolation. 5.5. Placental Perfusate
  • NK cells and/or ILC3 cells may be produced from hematopoietic cells, e.g., hematopoietic stem or progenitors from any source, e.g., placental tissue, placental perfusate, umbilical cord blood, placental blood, peripheral blood, spleen, liver, or the like.
  • the hematopoietic stem cells are combined hematopoietic stem cells from placental perfusate and from cord blood from the same placenta used to generate the placental perfusate.
  • Placental perfusate comprising placental perfusate cells that can be obtained, for example, by the methods disclosed in U.S. Patent Nos. 7,045,148 and 7,468,276 and U.S. Patent Application Publication No. 2009/0104164, the disclosures of which are hereby incorporated in their entireties.
  • the placental perfusate and perfusate cells from which hematopoietic stem or progenitors may be isolated, or useful in tumor suppression or the treatment of an individual having tumor cells, cancer or a viral infection, e.g., in combination with the NK cells and/or ILC3 cells, e.g., NK cell and/or ILC3 cell populations produced according to the three-stage method provided herein, can be collected by perfusion of a mammalian, e.g., human postpartum placenta using a placental cell collection composition.
  • a mammalian e.g., human postpartum placenta
  • Perfusate can be collected from the placenta by perfusion of the placenta with any physiologically-acceptable solution, e.g., a saline solution, culture medium, or a more complex cell collection composition.
  • a physiologically-acceptable solution e.g., a saline solution, culture medium, or a more complex cell collection composition.
  • a cell collection composition suitable for perfusing a placenta, and for the collection and preservation of perfusate cells is described in detail in related U.S. Application Publication No. 2007/0190042, which is incorporated herein by reference in its entirety.
  • the cell collection composition can comprise any physiologically-acceptable solution suitable for the collection and/or culture of stem cells, for example, a saline solution (e.g., phosphate-buffered saline, Kreb’s solution, modified Kreb’s solution, Eagle’s solution, 0.9% NaCl. etc), a culture medium (e.g., DMEM, H.DMEM, etc), and the like.
  • a saline solution e.g., phosphate-buffered saline, Kreb’s solution, modified Kreb’s solution, Eagle’s solution, 0.9% NaCl. etc
  • a culture medium e.g., DMEM, H.DMEM, etc
  • the cell collection composition can comprise one or more components that tend to preserve placental cells, that is, prevent the placental cells from dying, or delay the death of the placental cells, reduce the number of placental cells in a population of cells that die, or the like, from the time of collection to the time of culturing.
  • Such components can be, e.g., an apoptosis inhibitor (e.g., a caspase inhibitor or JNK inhibitor); a vasodilator (e.g., magnesium sulfate, an antihypertensive drug, atrial natriuretic peptide (ANP), adrenocorticotropin, corticotropin-releasing hormone, sodium nitroprusside, hydralazine, adenosine triphosphate, adenosine, indomethacin or magnesium sulfate, a phosphodiesterase inhibitor, etc.),' a necrosis inhibitor (e.g., 2-(lH-Indol-3-yl)-3-pentylamino-maleimide, pyrrolidine dithiocarbamate, or clonazepam); a TNF-a inhibitor; and/or an oxygen-carrying perfluorocarbon (e.g., perfluorooctyl bro
  • the cell collection composition can comprise one or more tissue-degrading enzymes, e.g., a metalloprotease, a serine protease, a neutral protease, a hyaluronidase, an RNase, or a DNase, or the like.
  • tissue-degrading enzymes include, but are not limited to, collagenases (e.g., collagenase I, II, III or IV, a collagenase from Clostridium histolyticum, etc.),' dispase, thermolysin, elastase, trypsin, LIBERASE, hyaluronidase, and the like.
  • the cell collection composition can comprise a bacteriocidally or bacteriostatically effective amount of an antibiotic.
  • the antibiotic is a macrolide (e.g., tobramycin), a cephalosporin (e.g., cephalexin, cephradine, cefuroxime, cefprozil, cefaclor, cefixime or cefadroxil), a clarithromycin, an erythromycin, a penicillin (e.g., penicillin V) or a quinolone (e.g., ofloxacin, ciprofloxacin or norfloxacin), a tetracycline, a streptomycin, etc.
  • the antibiotic is active against Gram(+) and/or Gram(-) bacteria, e.g., Pseudomonas aeruginosa, Staphylococcus aureus, and the like.
  • the cell collection composition can also comprise one or more of the following compounds: adenosine (about 1 mM to about 50 mM); D-glucose (about 20 mM to about 100 mM); magnesium ions (about 1 mM to about 50 mM); a macromolecule of molecular weight greater than 20,000 daltons, in one embodiment, present in an amount sufficient to maintain endothelial integrity and cellular viability (e.g., a synthetic or naturally occurring colloid, a polysaccharide such as dextran or a polyethylene glycol present at about 25 g/1 to about 100 g/1, or about 40 g/1 to about 60 g/1); an antioxidant (e.g., butylated hydroxyanisole, butylated hydroxytoluene, glutathione, vitamin C or vitamin E present at about 25 mM to about 100 mM); a reducing agent (e.g., N-acetylcysteine present at about 0.1 m
  • a human placenta is recovered shortly after its expulsion after birth.
  • the placenta is recovered from a patient after informed consent and after a complete medical history of the patient is taken and is associated with the placenta.
  • the medical history continues after delivery.
  • the umbilical cord blood and placental blood Prior to recovery of perfusate, the umbilical cord blood and placental blood are removed. In certain embodiments, after delivery, the cord blood in the placenta is recovered.
  • the placenta can be subjected to a conventional cord blood recovery process. Typically a needle or cannula is used, with the aid of gravity, to exsanguinate the placenta (see, e.g., Anderson, U.S. Patent No. 5,372,581; Hessel et ctl, U.S. Patent No. 5,415,665).
  • the needle or cannula is usually placed in the umbilical vein and the placenta can be gently massaged to aid in draining cord blood from the placenta.
  • cord blood recovery may be performed commercially, e.g., LifeBank Inc., Cedar Knolls, N.J., ViaCord, Cord Blood Registry and CryoCell.
  • the placenta is gravity drained without further manipulation so as to minimize tissue disruption during cord blood recovery.
  • a placenta is transported from the delivery or birthing room to another location, e.g., a laboratory, for recovery of cord blood and collection of perfusate.
  • the placenta can be transported in a sterile, thermally insulated transport device (maintaining the temperature of the placenta between 20-28 °C), for example, by placing the placenta, with clamped proximal umbilical cord, in a sterile zip-lock plastic bag, which is then placed in an insulated container.
  • the placenta is transported in a cord blood collection kit substantially as described in U.S. Patent No. 7,147,626.
  • the placenta is delivered to the laboratory four to twenty-four hours following delivery.
  • the proximal umbilical cord is clamped, for example within 4-5 cm (centimeter) of the insertion into the placental disc prior to cord blood recovery. In other embodiments, the proximal umbilical cord is clamped after cord blood recovery but prior to further processing of the placenta.
  • the placenta prior to collection of the perfusate, can be stored under sterile conditions and at either room temperature or at a temperature of 5 to 25 °C (centigrade).
  • the placenta may be stored for a period of longer than forty eight hours, or for a period of four to twenty -four hours prior to perfusing the placenta to remove any residual cord blood.
  • the placenta can be stored in an anticoagulant solution at a temperature of 5 °C to 25 °C (centigrade). Suitable anticoagulant solutions are well known in the art. For example, a solution of heparin or warfarin sodium can be used.
  • the anticoagulant solution comprises a solution of heparin (e.g., 1% w/w in 1:1000 solution).
  • the exsanguinated placenta is stored for no more than 36 hours before placental perfusate is collected.
  • Perfusate can be obtained by passage of perfusion solution, e.g., saline solution, culture medium or cell collection compositions described above, through the placental vasculature.
  • perfusion solution e.g., saline solution, culture medium or cell collection compositions described above
  • a mammalian placenta is perfused by passage of perfusion solution through either or both of the umbilical artery and umbilical vein.
  • the flow of perfusion solution through the placenta may be accomplished using, e.g., gravity flow into the placenta.
  • the perfusion solution is forced through the placenta using a pump, e.g., a peristaltic pump.
  • the umbilical vein can be, e.g., cannulated with a cannula, e.g., a TEFLON® or plastic cannula, that is connected to a sterile connection apparatus, such as sterile tubing.
  • the sterile connection apparatus is connected to a perfusion manifold.
  • the placenta can be oriented in such a manner that the umbilical artery and umbilical vein are located at the highest point of the placenta.
  • the placenta can be perfused by passage of a perfusion solution through the placental vasculature, or through the placental vasculature and surrounding tissue.
  • the umbilical artery and the umbilical vein are connected simultaneously to a pipette that is connected via a flexible connector to a reservoir of the perfusion solution.
  • the perfusion solution is passed into the umbilical vein and artery.
  • the perfusion solution exudes from and/or passes through the walls of the blood vessels into the surrounding tissues of the placenta, and is collected in a suitable open vessel from the surface of the placenta that was attached to the uterus of the mother during gestation.
  • the perfusion solution may also be introduced through the umbilical cord opening and allowed to flow or percolate out of openings in the wall of the placenta which interfaced with the maternal uterine wall.
  • the perfusion solution is passed through the umbilical veins and collected from the umbilical artery, or is passed through the umbilical artery and collected from the umbilical veins, that is, is passed through only the placental vasculature (fetal tissue).
  • the umbilical artery and the umbilical vein are connected simultaneously, e.g., to a pipette that is connected via a flexible connector to a reservoir of the perfusion solution.
  • the perfusion solution is passed into the umbilical vein and artery.
  • the perfusion solution exudes from and/or passes through the walls of the blood vessels into the surrounding tissues of the placenta, and is collected in a suitable open vessel from the surface of the placenta that was attached to the uterus of the mother during gestation.
  • the perfusion solution may also be introduced through the umbilical cord opening and allowed to flow or percolate out of openings in the wall of the placenta which interfaced with the maternal uterine wall.
  • Placental cells that are collected by this method which can be referred to as a “pan” method, are typically a mixture of fetal and maternal cells.
  • the perfusion solution is passed through the umbilical veins and collected from the umbilical artery, or is passed through the umbilical artery and collected from the umbilical veins.
  • Placental cells collected by this method which can be referred to as a “closed circuit” method, are typically almost exclusively fetal.
  • the closed circuit perfusion method can, in one embodiment, be performed as follows.
  • a post-partum placenta is obtained within about 48 hours after birth.
  • the umbilical cord is clamped and cut above the clamp.
  • the umbilical cord can be discarded, or can processed to recover, e.g., umbilical cord stem cells, and/or to process the umbilical cord membrane for the production of a biomaterial.
  • the amniotic membrane can be retained during perfusion, or can be separated from the chorion, e.g., using blunt dissection with the fingers.
  • amniotic membrane is separated from the chorion prior to perfusion, it can be, e.g., discarded, or processed, e.g., to obtain stem cells by enzymatic digestion, or to produce, e.g., an amniotic membrane biomaterial, e.g., the biomaterial described in U.S. Application Publication No. 2004/0048796.
  • an amniotic membrane biomaterial e.g., the biomaterial described in U.S. Application Publication No. 2004/0048796.
  • the umbilical cord vessels are exposed, e.g., by partially cutting the umbilical cord membrane to expose a cross-section of the cord.
  • the vessels are identified, and opened, e.g., by advancing a closed alligator clamp through the cut end of each vessel.
  • the apparatus e.g., plastic tubing connected to a perfusion device or peristaltic pump, is then inserted into each of the placental arteries.
  • the pump can be any pump suitable for the purpose, e.g., a peristaltic pump.
  • Plastic tubing, connected to a sterile collection reservoir, e.g., a blood bag such as a 250 mL collection bag, is then inserted into the placental vein.
  • the tubing connected to the pump is inserted into the placental vein, and tubes to a collection reservoir(s) are inserted into one or both of the placental arteries.
  • the placenta is then perfused with a volume of perfusion solution, e.g., about 750 ml of perfusion solution. Cells in the perfusate are then collected, e.g., by centrifugation.
  • the proximal umbilical cord is clamped during perfusion, and, more specifically, can be clamped within 4-5 cm (centimeter) of the cord’s insertion into the placental disc.
  • the first collection of perfusion fluid from a mammalian placenta during the exsanguination process is generally colored with residual red blood cells of the cord blood and/or placental blood.
  • the perfusion fluid becomes more colorless as perfusion proceeds and the residual cord blood cells are washed out of the placenta.
  • Generally from 30 to 100 mL of perfusion fluid is adequate to initially flush blood from the placenta, but more or less perfusion fluid may be used depending on the observed results.
  • cord blood is removed from the placenta prior to perfusion (e.g., by gravity drainage), but the placenta is not flushed (e.g., perfused) with solution to remove residual blood.
  • cord blood is removed from the placenta prior to perfusion (e.g., by gravity drainage), and the placenta is flushed (e.g., perfused) with solution to remove residual blood.
  • the volume of perfusion liquid used to perfuse the placenta may vary depending upon the number of placental cells to be collected, the size of the placenta, the number of collections to be made from a single placenta, etc.
  • the volume of perfusion liquid may be from 50 mL to 5000 mL, 50 mL to 4000 mL, 50 mL to 3000 mL, 100 mL to 2000 mL, 250 mL to 2000 mL, 500 mL to 2000 mL, or 750 mL to 2000 mL.
  • the placenta is perfused with 700-800 mL of perfusion liquid following exsanguination.
  • the placenta can be perfused a plurality of times over the course of several hours or several days. Where the placenta is to be perfused a plurality of times, it may be maintained or cultured under aseptic conditions in a container or other suitable vessel, and perfused with a cell collection composition, or a standard perfusion solution (e.g., a normal saline solution such as phosphate buffered saline (“PBS”) with or without an anticoagulant (e.g., heparin, warfarin sodium, coumarin, bishy droxycoumarin), and/or with or without an antimicrobial agent (e.g., b-mercaptoethanol (0.1 mM); antibiotics such as streptomycin (e.g., at 40-100 mg/ml), penicillin (e.g., at 40 U/ml), amphotericin B (e.g., at 0.5 pg/ml).
  • PBS phosphate buffered saline
  • an isolated placenta is maintained or cultured for a period of time without collecting the perfusate, such that the placenta is maintained or cultured for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours, or 2 or 3 or more days before perfusion and collection of perfusate.
  • the perfused placenta can be maintained for one or more additional time(s), e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or more hours, and perfused a second time with, e.g., 700-800 mL perfusion fluid.
  • the placenta can be perfused 1, 2, 3, 4, 5 or more times, for example, once every 1, 2, 3, 4, 5 or 6 hours.
  • perfusion of the placenta and collection of perfusion solution e.g., placental cell collection composition, is repeated until the number of recovered nucleated cells falls below 100 cells/ml.
  • the perfusates at different time points can be further processed individually to recover time-dependent populations of cells, e.g., total nucleated cells. Perfusates from different time points can also be pooled.
  • placental perfusate and Placental Perfusate Cells typically comprise about 100 million to about 500 million nucleated cells, including hematopoietic cells from which NK cells and/or ILC3 cells, e.g., NK cells and/or ILC3 cells produced according to the three-stage method described herein, may be produced by the method disclosed herein.
  • the placental perfusate or perfusate cells comprise CD34 + cells, e.g., hematopoietic stem or progenitor cells.
  • Such cells can, in a more specific embodiment, comprise CD34 CD45 stem or progenitor cells, CD34 + CD45 + stem or progenitor cells, or the like.
  • the perfusate or perfusate cells are cryopreserved prior to isolation of hematopoietic cells therefrom.
  • the placental perfusate comprises, or the perfusate cells comprise, only fetal cells, or a combination of fetal cells and maternal cells.
  • NK Cells Produced by Three-Stage Method In another embodiment, provided herein is an isolated NK cell population, wherein said NK cells are produced according to the three-stage method described above. [00343] In one embodiment, provided herein is an isolated NK cell population produced by a three-stage method described herein, wherein said NK cell population comprises a greater percentage of CD3-CD56+ cells than an NK progenitor cell population produced by a three-stage method described herein, e.g., an NK progenitor cell population produced by the same three-stage method with the exception that the third culture step used to produce the NK progenitor cell population was of shorter duration than the third culture step used to produce the NK cell population. In a specific embodiment, said NK cell population comprises about 70% or more, in some embodiments, 75%, 80%, 85%, 90%,
  • said NK cell population comprises no less than 80%, 85%, 90%, 95%, 98%, or 99% CD3-CD56+ cells.
  • said NK cell population comprises between 70%-75%, 75%- 80%, 80%-85%, 85%-90%, 90%-95%, or 95%-99% CD3-CD56+ cells.
  • said CD3-CD56 + cells in said NK cell population comprises CD3-CD56 + cells that are additionally NKp46 + .
  • said CD3-CD56 + cells in said NK cell population comprises CD3-CD56 + cells that are additionally CD16-.
  • said CD3-CD56 + cells in said NK cell population comprises CD3-CD56 + cells that are additionally CD16+.
  • said CD3-CD56 + cells in said NK cell population comprises CD3-CD56 + cells that are additionally CD94-.
  • said CD3-CD56 + cells in said NK cell population comprises CD3-CD56 + cells that are additionally CD94+.
  • said CD3-CD56 + cells in said NK cell population comprises CD3-CD56 + cells that are additionally CD11a + . In certain embodiments, said CD3-CD56 + cells in said NK cell population comprises CD3-CD56 + cells that are additionally NKp30 + . In certain embodiments, said CD3-CD56 + cells in said NK cell population comprises CD3-CD56 + cells that are additionally CD161 + . In certain embodiments, said CD3-CD56 + cells in said NK cell population comprises CD3-CD56 + cells that are additionally DNAM-1 + . In certain embodiments, said CD3-CD56 + cells in said NK cell population comprises CD3-CD56 + cells that are additionally T-bet + .
  • an NK cell population produced by a three-stage method described herein comprises cells which are CD117+. In one embodiment, an NK cell population produced by a three-stage method described herein comprises cells which are NKG2D+. In one embodiment, an NK cell population produced by a three-stage method described herein comprises cells which are NKp44+. In one embodiment, an NK cell population produced by a three-stage method described herein comprises cells which are CD244+. In one embodiment, an NK cell population produced by a three-stage method described herein comprises cells which express perforin. In one embodiment, an NK cell population produced by a three-stage method described herein comprises cells which express
  • an NK cell population produced by a three-stage method described herein comprises cells which express granzyme B. In one embodiment, an NK cell population produced by a three-stage method described herein comprises cells which secrete IFNy, GM-CSF and/or TNFa.
  • ILC3 Cells Produced by Three-Stage Method In another embodiment, provided herein is an isolated ILC3 cell population, wherein said ILC3 cells are produced according to the three-stage method described above. [00347] In one embodiment, provided herein is an isolated ILC3 cell population produced by a three-stage method described herein, wherein said ILC3 cell population comprises a greater percentage of CD3-CD56+ cells than an ILC3 progenitor cell population produced by a three-stage method described herein, e.g., an ILC3 progenitor cell population produced by the same three-stage method with the exception that the third culture step used to produce the ILC3 progenitor cell population was of shorter duration than the third culture step used to produce the ILC3 cell population. In a specific embodiment, said ILC3 cell population comprises about 70% or more, in some embodiments, 75%, 80%, 85%, 90%,
  • said ILC3 cell population comprises no less than 80%, 85%, 90%, 95%, 98%, or 99% CD3-CD56+ cells.
  • said ILC3 cell population comprises between 70%-75%, 75%-80%, 80%-85%, 85%-90%, 90%-95%, or 95%-99% CD3-CD56+ cells.
  • said CD3-CD56 + cells in said ILC3 cell population comprises CD3-CD56 + cells that are additionally NKp46 , In certain embodiments, said CD3-CD56 + cells in said ILC3 cell population comprises CD3-CD56 + cells that are additionally CD16-. In certain embodiments, said CD3-CD56 + cells in said ILC3 cell population comprises CD3-CD56 + cells that are additionally IL1R1+. In certain embodiments, said CD3-CD56 + cells in said ILC3 cell population comprises CD3-CD56 + cells that are additionally CD94-. In certain embodiments, said CD3-CD56 + cells in said ILC3 cell population comprises CD3-CD56 + cells that are additionally RORyt+.
  • said CD3-CD56 + cells in said ILC3 cell population comprises CD3-CD56 + cells that are additionally CD 11a-. In certain embodiments, said CD3-CD56 + cells in said ILC3 cell population comprises CD3-CD56 + cells that are additionally T-bet+.
  • an ILC3 cell population produced by a three-stage method described herein comprises cells which are CD117+. In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which are NKG2D . In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which are NKp30 , In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which are CD244+. In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which are DNAM-1+. In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which express AHR.
  • an ILC3 cell population produced by a three-stage method described herein comprises cells which do not express perforin. In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which do not express EOMES. In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which do not express granzyme B. In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which secrete IL-22 and/or IL-8.
  • cell populations produced by the three-stage method described herein comprise CDlla+ cells and CDlla- cells in a ratio of 50:1, 40:1, 30:1, 20:1, 10:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:10, 1:20, 1:30, 1:40, or 1:50.
  • a population of cells described herein comprises CD11a+ cells and CD11a- cells in a ratio of 50: 1.
  • a population of cells described herein comprises CD11a+ cells and CDlla- cells in a ratio of 20:1.
  • a population of cells described herein comprises CDlla+ cells and CDlla- cells in a ratio of 10:1.
  • a population of cells described herein comprises CD11a+ cells and CD11a- cells in a ratio of 5:1. In certain aspects, a population of cells described herein comprises CDlla+ cells and CD 11a- cells in a ratio of 1 : 1. In certain aspects, a population of cells described herein comprises CD11a+ cells and CD11a- cells in a ratio of 1 :5. In certain aspects, a population of cells described herein comprises CD11a+ cells and CD11a- cells in a ratio of 1 : 10. In certain aspects, a population of cells described herein comprises CD11a+ cells and CD11a- cells in a ratio of 1 :20. In certain aspects, a population of cells described herein comprises CDlla+ cells and CDlla- cells in a ratio of 1:50.
  • cell populations described herein are produced by combining the CDlla+ cells with the CDlla- cells in a ratio of 50:1, 40:1, 30:1, 20:1, 10:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:10, 1:20, 1:30, 1:40, or 1:50 to produce a combined population of cells.
  • a combined population of cells described herein comprises CD11a+ cells and CD11a- cells combined in a ratio of 50: 1.
  • a combined population of cells described herein comprises CDlla+ cells and CD11a- cells combined in a ratio of 20: 1.
  • a combined population of cells described herein comprises CD11a+ cells and CD11a- cells combined in a ratio of 10:1. In certain aspects, a combined population of cells described herein comprises CD11a+ cells and CD11a- cells combined in a ratio of 5:1. In certain aspects, a combined population of cells described herein comprises CDlla+ cells and CD11a- cells combined in a ratio of 1:1. In certain aspects, a combined population of cells described herein comprises CD11a+ cells and CDlla- cells combined in a ratio of 1:5. In certain aspects, a combined population of cells described herein comprises CD11a+ cells and CD 11a- cells combined in a ratio of 1 : 10.
  • a combined population of cells described herein comprises CD11a+ cells and CDlla- cells combined in a ratio of 1:20. In certain aspects, a combined population of cells described herein comprises CDlla+ cells and CDlla- cells combined in a ratio of 1:50.
  • cell populations produced by the three-stage method described herein comprise NK cells and ILC3 cells in a ratio of 50:1, 40:1, 30:1, 20:1, 10:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:10, 1:20, 1:30, 1:40, or 1:50. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 50: 1.
  • a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 20: 1. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 10:1. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 5:1. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 1:1. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 1 :5. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 1 : 10. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 1 :20. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 1:50.
  • cell populations described herein are produced by combining the NK cells with the ILC3 cells in a ratio of 50:1, 40:1, 30:1, 20:1, 10:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:10, 1:20, 1:30, 1:40, or 1:50 to produce a combined population of cells.
  • a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 50: 1.
  • a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 20: 1.
  • a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 10: 1.
  • a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 5:1. In certain aspects, a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 1: 1. In certain aspects, a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 1 :5. In certain aspects, a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 1 : 10. In certain aspects, a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 1:20. In certain aspects, a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 1:50.
  • compositions Comprising NK Cells and/or ILC3 Cells
  • a composition e.g., a pharmaceutical composition, comprising an isolated NK cell and/or ILC3 cell population produced using the three-stage method described herein.
  • said isolated NK cell and/or ILC3 cell population is produced from hematopoietic cells, e.g., hematopoietic stem or progenitor cells isolated from placental perfusate, umbilical cord blood, and/or peripheral blood.
  • said isolated NK cell and/or ILC3 cell population comprises at least 50% of cells in the composition.
  • said isolated NK cell and/or ILC3 cell population e.g., CD3-CD56 + cells, comprises at least 80%, 85%, 90%. 95%, 98% or 99% of cells in the composition. In certain embodiments, no more than 5%, 10%, 15%, 20%, 25%, 30%, 35%, or 40% of the cells in said isolated NK cell and/or ILC3 cell population are CD3-CD56 + cells. In certain embodiments, said CD3-CD56 + cells are CD 16-.
  • NK cell and/or ILC3 cell populations produced using the three-stage method described herein can be formulated into pharmaceutical compositions for use in vivo.
  • Such pharmaceutical compositions comprise a population of NK cells and/or ILC3 cells in a pharmaceutically-acceptable carrier, e.g., a saline solution or other accepted physiologically- acceptable solution for in vivo administration.
  • Pharmaceutical compositions of the invention can comprise any of the NK cell and/or ILC3 cell populations described elsewhere herein.
  • the pharmaceutical compositions of the invention comprise populations of cells that comprise 50% viable cells or more (that is, at least 50% of the cells in the population are functional or living). Preferably, at least 60% of the cells in the population are viable. More preferably, at least 70%, 80%, 90%, 95%, or 99% of the cells in the population in the pharmaceutical composition are viable.
  • compositions of the invention can comprise one or more compounds that, e.g., facilitate engraftment; stabilizers such as albumin, dextran 40, gelatin, hydroxyethyl starch, and the like.
  • the pharmaceutical composition of the invention comprises about 1.25% HSA and about 2.5% dextran.
  • Other injectable formulations, suitable for the administration of cellular products, may be used.
  • the compositions, e.g., pharmaceutical compositions, provided herein are suitable for systemic or local administration.
  • the compositions, e.g., pharmaceutical compositions, provided herein are suitable for parenteral administration.
  • the compositions, e.g., pharmaceutical compositions, provided herein are suitable for injection, infusion, intravenous (IV) administration, intrafemoral administration, or intratumor administration.
  • the compositions, e.g., pharmaceutical compositions, provided herein are suitable for administration via a device, a matrix, or a scaffold.
  • the compositions, e.g., pharmaceutical compositions provided herein are suitable for injection.
  • the compositions, e.g., pharmaceutical compositions, provided herein are suitable for administration via a catheter.
  • the compositions, e.g., pharmaceutical compositions, provided herein are suitable for local injection.
  • the compositions, e.g., pharmaceutical compositions, provided herein are suitable for local injection directly into a solid tumor (e.g., a sarcoma).
  • the compositions, e.g., pharmaceutical compositions, provided herein are suitable for injection by syringe.
  • the compositions, e.g., pharmaceutical compositions, provided herein are suitable for administration via guided delivery.
  • the compositions, e.g., pharmaceutical compositions, provided herein are suitable for injection aided by laparoscopy, endoscopy, ultrasound, computed tomography, magnetic resonance, or radiology.
  • compositions e.g., pharmaceutical compositions provided herein, comprising NK cells and/or ILC3 cells produced using the methods described herein, are provided as pharmaceutical grade administrable units.
  • Such units can be provided in discrete volumes, e.g., 15 mL, 20 mL, 25 mL, 30 nL. 35 mL, 40 mL, 45 mL,
  • Such units can be provided so as to contain a specified number of cells, e.g., NK cells and/or ILC3 cells, e.g., 1 x 10 4 , 5 x 10 4 , 1 x 10 5 , 5 x 10 5 , 1 x 10 6 , 5 x 10 6 , 1 x 10 7 , 5 x 10 7 , 1 x 10 8 , 5 x 10 8 or more cells per milliliter, or 1 x 10 4 , 5 x 10 4 , 1 x 10 5 , 5 x 10 5 , 1 x 10 6 , 5 x 10 6 , 1 x 10 7 , 5 x 10 7 , 1 x 10 8 , 5 x 10 8 , 1 x 10 9 , 5 x 10 9 , 1 x 10 10 , 5 x 10 10 , 1 x 10 11 or more cells per unit.
  • NK cells and/or ILC3 cells e.g., 1 x 10 4 , 5 x 10
  • the units can comprise about, at least about, or at most about 1 x 10 4 , 5 x 10 4 , 1 x 10 5 , 5 x 10 5 , 1 x 10 6 , 5 x 10 6 or more NK cells and/or ILC3 cells per milliliter, or 1 x 10 4 , 5 x 10 4 , 1 x 10 5 , 5 x 10 5 , 1 x 10 6 , 5 x 10 6 , 1 x 10 7 , 5 x 10 7 , 1 x 10 8 , 5 x 10 8 , 1 x 10 9 , 5 x 10 9 , 1 x 10 10 , 5 x 10 10 , 1 x 10 11 or more cells per unit.
  • Such units can be provided to contain specified numbers of NK cells and/or ILC3 cells or NK cell and/or ILC3 cell populations and/or any of the other cells.
  • the NK cells and ILC3 cells are present in ratios provided herein.
  • said isolated NK cells and/or ILC3 cells in said composition are from a single individual.
  • said isolated NK cells and/or ILC3 cells comprise NK cells and/or ILC3 cells from at least two different individuals.
  • said isolated NK cells and/or ILC3 cells in said composition are from a different individual than the individual for whom treatment with the NK cells and/or ILC3 cells is intended.
  • said NK cells have been contacted or brought into proximity with an immunomodulatory compound or thalidomide in an amount and for a time sufficient for said NK cells to express detectably more granzyme B or perforin than an equivalent number of natural killer cells, i.e.
  • said composition additionally comprises an immunomodulatory compound or thalidomide.
  • the immunomodulatory compound is a compound described below. See, e.g., U.S. Patent No. 7,498,171, the disclosure of which is hereby incorporated by reference in its entirety.
  • the immunomodulatory compound is an amino-substituted isoindoline.
  • the immunomodulatory compound is 3 -(4-amino- 1-oxo- 1,3- dihydroisoindol-2-yl)-piperidine-2,6-dione; 3-(4'aminoisolindoline-r-one)-l-piperidine-2,6- dione; 4-(amino)-2-(2,6-dioxo(3-piperidyl))-isoindoline-l,3-dione; or 4-Amino-2-(2,6- dioxopiperidin-3-yl)isoindole-l,3-dione.
  • the immunomodulatory compound is pomalidomide, or lenalidomide.
  • R 1 is H, (C 1 -C 8 )alkyl, (C 3 -C 7 )cycloalkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, benzyl, aryl, (C 0 -C 4 )alkyl-(C 1 -C 6 )heterocycloalkyl, (C 0 -C 4 )alkyl-(C 2 -C5)heteroaryl, C(O)R 3 , C(S)R 3 , C(O)OR 4 , (C 1 -C 8 )alkyl-N(R 6 )2, (C 1 -C 8 )alkyl-OR 5 , (C 1 -C 8 )alkyl-C(O)OR 5 , C(O)NHR 3 , C(S)NHR 3 , C(O)NR 3 R 3’ , C(S)NR 3 R 3’ or (C 1
  • R 2 is H, F, benzyl, (C 1 -C 8 )alkyl, (C 2 -C 8 )alkenyl, or (C 2 -C 8 )alkynyl;
  • R 3 and R 3 are independently (C 1 -C 8 )alkyl, (C3-C7)cycloalkyl, (C 2 -C 8 )alkenyl, (C 2 - C 8 )alkynyl, benzyl, aryl, (C 0 -C 4 )alkyl-(C 1 -C 6 )heterocycloalkyl, (C 0 -C 4 )alkyl-(C 2 - C 5 )heteroaiyl, (C 0 -C 8 )alkyl-N(R 6 )2, (C 1 -C 8 )alkyl-OR 5 , (C 1 -C 8 )alkyl-C(O)OR 5 , (C 1 -C 8 )alkyl- O(CO)R 5 , or C(O)OR 5 ;
  • R 4 is (C 1 -C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, (C 1 -C 4 )alkyl-OR 5 , benzyl, aryl, (C 0 -C 4 )alkyl-(C 1 -C 6 )heterocycloalkyl, or (C 0 -C 4 )alkyl-(C 2 -C5)heteroaryl;
  • R 5 is (C 1 -C 8 )alkyl, (C 2 -C 8 )alkenyl, (C 2 -C 8 )alkynyl, benzyl, aryl, or (C 2 -C5)heteroaryl; each occurrence of R 6 is independently H, (C 1 -C 8 )alkyl, (C 2 -C 8 )alkenyl.
  • (C 2 - C 8 )alkynyl, benzyl, aryl, (C 2 -C 5 )heteroaryl, or (C 0 -C 8 )alkyl-C(O)O-R 5 or the R 6 groups can join to form a heterocycloalkyl group; n is 0 or 1; and
  • R is H or CH 2 OCOR’
  • each of R 1 , R 2 , R 3 , or R 4 independently of the others, is halo, alkyl of 1 to 4 carbon atoms, or alkoxy of 1 to 4 carbon atoms or (ii) one of R 1 , R 2 , R 3 , or R 4 is nitro or -NHR 5 and the remaining of R 1 , R 2 , R 3 , or R 4 are hydrogen;
  • R 5 is hydrogen or alkyl of 1 to 8 carbons
  • R 6 hydrogen, alkyl of 1 to 8 carbon atoms, benzo, chloro, or fluoro;
  • R’ is R 7 -CHR 10 -N(R 8 R 9 );
  • R 7 is m-phenylene or p-phenylene or -(C n H 2n )- in which n has a value of 0 to 4; each of R 8 and R 9 taken independently of the other is hydrogen or alkyl of 1 to 8 carbon atoms, or R 8 and R 9 taken together are tetramethylene, pentamethylene, hexamethylene, or -CH 2 CH 2 X1CH 2 CH 2 - in which Xi is -O-, -S-, or -NH-;
  • R 10 is hydrogen, alkyl of to 8 carbon atoms, or phenyl; and * represents a chiral-carbon center; or a pharmaceutically acceptable salt, hydrate, solvate, clathrate, enantiomer, diastereomer, racemate, or mixture of stereoisomers thereof.
  • the composition additionally comprises one or more anticancer compounds, e.g., one or more of the anticancer compounds described below.
  • the composition comprises NK cells and/or ILC3 cells from another source, or made by another method.
  • said other source is placental blood and/or umbilical cord blood.
  • said other source is peripheral blood.
  • the NK cell and/or ILC3 cell population in said composition is combined with NK cells and/or ILC3 cells from another source, or made by another method in a ratio of about 100: 1, 95:5, 90: 10, 85: 15, 80:20, 75:25, 70:30, 65:35, 60:40, 55:45: 50:50, 45:55, 40:60, 35:65, 30:70, 25:75, 20:80, 15:85, 10:90, 5:95, 100:1, 95:1, 90:1, 85:1, 80:1, 75:1, 70:1, 65:1, 60:1, 55:1, 50:1, 45:1,
  • the composition comprises an NK cell and/or ILC3 cell population produced using the three-stage method described herein and either isolated placental perfusate or isolated placental perfusate cells.
  • said placental perfusate is from the same individual as said NK cell and/or ILC3 cell population.
  • said placental perfusate comprises placental perfusate from a different individual than said NK cell and/or ILC3 cell population.
  • all, or substantially all (e.g ., greater than 90%, 95%, 98% or 99%) of cells in said placental perfusate are fetal cells.
  • the placental perfusate or placental perfusate cells comprise fetal and maternal cells.
  • the fetal cells in said placental perfusate comprise less than about 90%, 80%, 70%, 60% or 50% of the cells in said perfusate.
  • said perfusate is obtained by passage of a 0.9% NaCl solution through the placental vasculature.
  • said perfusate comprises a culture medium. In another specific embodiment, said perfusate has been treated to remove erythrocytes.
  • said composition comprises an immunomodulatory compound, e g., an immunomodulatory compound described below, e.g., an amino-substituted isoindoline compound.
  • the composition additionally comprises one or more anticancer compounds, e.g., one or more of the anti cancer compounds described below.
  • the composition comprises an NK cell and/or ILC3 cell population and placental perfusate cells. In a more specific embodiment, said placental perfusate cells are from the same individual as said NK cell and/or ILC3 cell population.
  • said placental perfusate cells are from a different individual than said NK cell and/or ILC3 cell population.
  • the composition comprises isolated placental perfusate and isolated placental perfusate cells, wherein said isolated perfusate and said isolated placental perfusate cells are from different individuals.
  • said placental perfusate comprises placental perfusate from at least two individuals.
  • said isolated placental perfusate cells are from at least two individuals.
  • said composition comprises an immunomodulatory compound.
  • the composition additionally comprises one or more anticancer compounds, e.g., one or more of the anticancer compounds described below.
  • a pharmaceutical pack or kit comprising one or more containers filled with one or more of the compositions described herein, e.g., a composition comprising NK cells and/or ILC3 cells produced by a method described herein, e.g., NK cell and/or ILC3 cell populations produced using the three-stage method described herein.
  • a composition comprising NK cells and/or ILC3 cells produced by a method described herein, e.g., NK cell and/or ILC3 cell populations produced using the three-stage method described herein.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • kits encompassed herein can be used in accordance with the methods described herein, e.g., methods of suppressing the growth of tumor cells and/or methods of treating cancer, e.g., hematologic cancer, and/or methods of treating viral infection.
  • a kit comprises NK cells and/or ILC3 cells produced by a method described herein or a composition thereof, in one or more containers.
  • a kit comprising an NK cell and/or ILC3 cell population produced by a three-stage method described herein, or a composition thereof.
  • Example 1 Three-stage method of producing natural killer cells from hematopoietic stem or progenitor cells
  • CD34 + cells are cultured in the following medium formulations for the indicated number of days, and aliquots of cells are taken for assessment of cell count, cell viability, characterization of natural killer cell differentiation and functional evaluation.
  • Stage 1 medium 90% Stem Cell Growth Medium (SCGM) (CellGro®), 10% Human Serum-AB, supplemented with 25 ng/mL or 250 ng/mL recombinant human thrombopoietin (TPO), 25 ng/mL recombinant human Flt3L, 27 ng/mL recombinant human stem cell factor (SCF), 25 ng/mL recombinant human IL-7, 0.05 ng/mL or 0.025 ng/mL recombinant human IL-6, 0.25 ng/mL or 0.125 ng/mL recombinant human granulocyte colony-stimulating factor (G-CSF), 0.01 ng/mL or 0.025 ng/m
  • SCGM Stem Cell
  • Stage 2 medium 90% SCGM, 10% Human Serum- AB, supplemented with 25 ng/mL recombinant human Flt3L, 27 ng/mL recombinant human SCF, 25 ng/mL recombinant human IL-7, 20 ng/mL recombinant human IL-15, 0.05 ng/mL or 0.025 ng/mL recombinant human IL-6, 0.25 ng/mL or 0.125 ng/mL recombinant human granulocyte colony-stimulating factor (G-CSF), 0.01 ng/mL or 0.025 ng/mL recombinant human granulocyte-macrophage colony-stimulating factor (GM-CSF), 0.10% gentamicin, and 1 to 10 ⁇ m SRI or other stem cell mobilizing agent.
  • G-CSF granulocyte colony-stimulating factor
  • GM-CSF granulocyte-macrophage colony-
  • Stage 3 medium 90% STEMMACSTM, 10% Human Serum-AB, 0.025 mM 2- mercaptoethanol (55 mM), supplemented with 22 ng/mL recombinant human SCF, 1000 U/mL recombinant human IL-2, 20 ng/mL recombinant human IL-7, 20 ng/mL recombinant human IL-15, 0.05 ng/mL or 0.025 ng/mL recombinant human IL-6, 0.25 ng/mL or 0.125 ng/mL recombinant human granulocyte colony-stimulating factor (G-CSF), 0.01 ng/mL or 0.025 ng/mL recombinant human granulocyte-macrophage colony-stimulating factor (GM- CSF), and 0.10% gentamicin.
  • G-CSF granulocyte colony-stimulating factor
  • GM- CSF granulocyte-macrophage colon
  • Cells are seeded at Day 0 at 3x10 4 cells/mL in Stage 1 media, and cells are tested for purity by a CD34+ and CD45+ count and viability by 7AAD staining.
  • At Day 5 cells are counted and seeded to a concentration of 1 x10 5 cells/mL with Stage 1 medium.
  • At Day 7 cells are counted and seeded to a concentration of 1 x 10 5 cells/mL with Stage 1 medium.
  • the following protocol is used through Day 14: Cells seeded at Day 0 at 7.5 x 10 3 cells/mL in Stage 1 media, and cells are tested for purity by a CD34+ and CD45+ count and viability by 7AAD staining. At Day 7 cells are counted and seeded to a concentration of 3 x 10 5 cells/mL with Stage 1 medium. At Day 9 cells are counted and seeded to a concentration of 3 x 10 5 cells/mL with Stage 2 medium. At Day 12, cells are counted and seeded to a concentration of 3 x 10 5 cells/mL in Stage 2 medium. At Day 14, cells are counted and seeded to a concentration of 3 x 10 5 cells/mL in Stage 2 medium.
  • seeding of cells into at passage is performed either by dilution of the culture with fresh media or by centrifugation of cells and resuspension / addition of fresh media.
  • cells are spun at 400 xg for seven minutes, followed by suspension of the pellet in an equal volume of Plasmalyte A. The suspension is spun at 400*g for seven minutes, and the resulting pellet is suspended in 10% HSA (w/v), 60% Plasmalyte A (v/v) at the target cell concentration. The cells are then strained through a 70 pm mesh, the final container is filled, an aliquot of the cells are tested for viability, cytotoxicity, purity, and cell count, and the remainder is packaged.
  • Example 2 Selection of stem cell mobilizing agents for the expansion of NK cells
  • UCB CD34+ cells were cultivated in presence of cytokines including thrombopoietin, SCF, Flt3 ligand, IL-7, IL-15 and IL-2 for 35 days to produce three-stage NK cells, as described in Example 1. Multi-color flow cytometry was used to determine the phenotypic characteristics of three-stage NK cells.
  • the compounds were provided to culture to evaluate their effects on NK cell expansion and differentiation. Specifically, donors of CD34+ cells (StemCell Technology) were thawed and expanded in vitro following NK culture protocol. During the first 14 days of the culture, each CRL compounds was dissolved in DMSO and added to the culture at 10 mM concentration. SRI (at 10 pM) served as a positive control compound, while DMSO alone without any compound served as a negative control. At the end of the culture on Day 35, cell expansion, natural killer (NK) cell differentiation and cytotoxicity of the cells against K562 tumor cell line were characterized. Due to the large number of the compounds, the testing was performed in two experiments, CRLl-11 and CRL 12-22. The same donors were used for each experiment. Positive and negative controls were also included in both experiments.
  • Cytotoxicity assay was run using compound cultured cells against K562 tumor cells at 10: 1 effector to target ratio (FIGS. 5 A - 5B) to evaluate cell functions. The results showed that the cells cultured with compounds killed 30-60% of K562 cells at 10:1 E:T ratio, indicating that the cells present NK functions. For both donors, cells cultured with CRL 17, 18, 19 and 21 demonstrated similar or greater killing activities compared to those cultured with SRI .
  • PBMC Peripheral blood derived NKs
  • PB-NK Peripheral blood derived NKs
  • CYNK cells were generated from umbilical cord blood-derived CD34+ stem cells (Ref: Zhang et al. J Immunother Cancer. 2015). Briefly, the CD34+ cells were cultivated in the presence of cytokines including thromobopoietin, SCF, Flt3 ligand, IL-7, IL-15 and IL- 2 for 35 days.
  • cytokines including thromobopoietin, SCF, Flt3 ligand, IL-7, IL-15 and IL- 2 for 35 days.
  • PBNK and CYNK cells were cryopreserved until analysis.
  • CYNK cells were combined with PB-NK at 1 : 1 ratio and gene expression analyzed on single cell level using 10X Genomics Chromium platform and Illumina sequencing. Bioinformatics analysis utilized 10X Genomics Cell Ranger analysis pipeline.
  • CYNK cells efficiently kill various tumor cell lines in vitro, however, the mechanisms CYNK cells use to induce cell death remains poorly understood (rel).
  • scRNAseq single-cell RNA sequencing
  • PB-NK peripheral blood NK cells
  • FIG. 6A Unbiased transcriptional clustering revealed two distinct signatures differentiating between CYNK and PB-NK cells (FIG. 6B).
  • Tables 1 and 2 list top 50 upregulated genes per cluster in PB-NK and CYNK cells, respectively.
  • the gene set expressed higher in PB-NK cells included genes associated with NK cell functional roles, including FGFBP2, granzymes (GZMH, GZMM), CXCR4, KLRFl, KLF2, IFNG (Table 1).
  • ⁇ FGFBP2 encoding fibroblast growth factor-binding protein, is known to be secreted by cytotoxic lymphocytes.
  • ⁇ Granzymes are a group of serine proteases which are stored in the cytotoxic granules of NK cells and cytotoxic T lymphocytes (ref). While GzmA and GzmB induce target cell death upon release to their cytoplasm and have been extensively studied, less is known about the functional role of GzmH, GzmK and GzmM.
  • ⁇ CXCR4 regulates NK cell homing to bone marrow.
  • ⁇ KLRFl encodes NKp80, an activating C-type lectin-like immunoreceptor that is activated upon binding to activation-induced C-type lectin (AICL), inducing NK cell cytotoxicity and cytokine secretion.
  • AICL activation-induced C-type lectin
  • ⁇ NK cell-derived IFN-g (IFNG gene) is a key immunoregulatory factor secreted from activated NK cells that promotes adaptive immune response by modulating dendritic cell and T cell responses.
  • Table 1 Top 50 upregulated genes per PB-NK cluster.
  • Top differentially expressed genes in CYNK cluster that are encode factors associated with NK cell functional role include surface receptors and co-receptors (CD96, NCR3, CD59, KLRC1), TNFSF10, immune checkpoint genes (TNFRSF18, TNFRSF4, HAVCR2), NK cell receptor adaptor molecule genes (FCER1G and LAT2) (Table 2).
  • Table 2 Top 50 upregulated genes per CYNK cluster.
  • qRT-PCR demonstrated high expression of CD69, KLRK1 and KLRB1 relative to the housekeeping gene GAPDH in both CYNK and PB-NK cells, whereas, KLRKl and KLRBl, encoding for NKG2D and CD161/KLRB1, respectively, were significantly higher expressed in PB-NK cells.
  • KLRBl inhibitory killer cell lectin-like receptor
  • KLRBl inhibitory killer cell lectin-like receptor
  • KLRDl inhibitory killer cell lectin-like receptor
  • NCR2 cytotoxicity receptor 2 (encoding NKp44) was differentially expressed with high expression in CYNK cells and almost no expression in PB- NK cells.
  • CD244 co-activating NK cell receptor genes
  • DNAM-1 co-activating NK cell receptor genes
  • FCGR3A encoding an Fc receptor CD16 that is required for antibody-dependent cell-mediated cytotoxicity.
  • NK cells express high level of the NK cell marker CD56 and lack the expression of T cell, B cell and myeloid cell markers CD3, CD19 and CD14, respectively (FIG. 8). Whereas a majority of PB-NK cells express CD56 at a low level, a small subset of PB-NK cells express CD56 at a level seen in CYNK cells (FIG. 9).
  • NCR analysis demonstrated a high expression of NKp44 in CYNK cells, whereas, NKp44 was expressed at a low level in PB-NK, corresponding well to our transcriptional analysis (FIG. 7).
  • NKp80 on the other hand, was expressed on PB-NK cell and little on CYNK, also confirming the transcriptional data of KLRFl expression (Table 1 and FIG. 7).
  • CD 16 was virtually not expressed on CYNK cells, whereas the majority of PB-NK cells expressed CD16 at a high level. CD16 protein expression, therefore, also corresponds well to transcriptional analysis (Table 1 and FIG. 7).
  • killer cell lectin-like receptors was comparable between CYNK and PB- NK cells, with CYNK cells demonstrating higher mean fluorescence intensity compared to PB-NK cells for NKG2D, NKG2C, CD94 (NKG2C) and NKG2A.
  • GITR a checkpoint inhibitor molecule, encoded by TNFRSF18, was not expressed on PB-NK cells but highly on all CYNK cells, correlating well to qRT-PCR data.
  • Example 5 Cleavage Resistant CD16 Expressing NK Cells
  • Celularity, Inc. is developing human placental hematopoietic stem cells- derived, cryopreserved, off-the shelf, ex-vivo expanded and allogenic Natural killer (PNK) cells for various hematological malignancies and solid tumors.
  • NK cells play a central role in antibody dependent cell mediated cytotoxicity (ADCC) through Fc receptor CD16 in monoclonal antibody mediated anti -tumor therapies.
  • ADCC antibody dependent cell mediated cytotoxicity
  • Two allelic forms of CD 16 have been identified with the 158Val/Val form has shown to have higher IgG binding affinity comparing with the 158Phe/Phe form.
  • the high IgG binding allele are found in about 10-20% of the normal population.
  • activation of NK cells induces CD16 shedding by matrix metalloprotease ADAM17 at 197Ser, thus limiting ADCC responses.
  • a single mutation (Serl97Pro) prevents CD 16 shedding and increases ADCC activity in NK cells. Since the antibody binding affinity and CD 16 expression of PNK could vary with different donors, we hypothesize that expressing a high affinity (158Val) and proteinase cleavage resistant (197Pro) CD16 variant (CD16VP) augments anti-tumor ADCC activity.
  • Lentivirus expressing CD16VP was used to transduce human placental CD34+ cells. After transduction, the cells were cultured in the presence of cytokines including thrombopoietin, SCF, Flt3 ligand, IL-7, IL-15 and IL-2, for 35 days to generate PNK- CD16VP cells. Non-transduced PNK cells (NT) served as a control. Expression of CD16VP was evaluated by activating cells with PMA/ionomycin to induce CD16 cleavage (CD16 shedding assay) followed by immunostaining with CD 16 antibody and analyzed using flowcytometry.
  • cytokines including thrombopoietin, SCF, Flt3 ligand, IL-7, IL-15 and IL-2
  • ADCC of PNK-CD16VP cells was assessed against Daratumumab (anti- CD38) or Rituximab (anti-CD20) opsonized lymphoma cell lines at various effector to target (E:T) ratios. IgG was used as ADCC control.
  • IgG was used as ADCC control.
  • In vivo anti-tumor activity was assessed in a Daudi disseminated Xenograft model in NSG mice. Luciferase-expressing Daudi cells (3x106) were intravenously (IV) administered at day 0, followed by PNK-CD16VP cells (10x106) IV at day 1 and day 3, and Daratumumab at day 3. Tumor burden in mice was monitored by Bioluminescence Imaging (BLI). Statistical differences between the groups were calculated using paired t-test using Prism.
  • Cell culture Human placental CD34+ cells were isolated and cultured in the presence of cytokines including thrombopoietin, SCF, Flt3 ligand, IL-7, IL-15 and IL-2, for 35 days to generate NK cells.
  • cytokines including thrombopoietin, SCF, Flt3 ligand, IL-7, IL-15 and IL-2
  • CD56+/CD3-, and CD16 using flow cytometry.
  • CD16VP Shedding Assay Expression of CD16VP was evaluated by activating cells with PMA/ionomycin to induce CD 16 cleavage followed by immunostaining with CD 16 antibody and analyzed using flow cytometry.
  • ADCC activity of CD16VP cells was assessed against
  • CD16VP expression was shown to be resistant to shedding after activation.
  • CD16VP cells demonstrated enhanced ADCC in vitro against lymphoma cell lines in combination with Daratumumab or Rituximab.
  • CD16VP resistance to activation induced shedding supported sustained killing in vitro.
  • CD16VP cells showed in vivo anti-tumor activities at early time points in an
  • ADCC lymphoma model ADCC lymphoma model.
  • CD16VP provides a promising approach to augment the anti-tumor activities in combination with monoclonal antibodies. Further investigation is perused to support [00412] development of CD16VP in combination with therapeutic antibodies for various hematological malignancies and solid tumors.
  • PNK-CD16VP were used to test anti-tumor ADCC in vivo using a disseminated Daudi Xenograft model.
  • the preliminary data demonstrated that PNK-CD16VP combined with Daratumumab reduced BLI signal (>50%) compared to vehicle or Daratumumab alone at day 10 after treatment. This observation suggested that PNK-CD16VP demonstrated in vivo ADCC anti-tumor activity.
  • PNK-CD16VP cells demonstrated enhanced ADCC function against lymphoma cell lines in vitro and in vivo. Further development of PNK-CD16VP for immune-oncology therapeutics is warranted.
  • Example 6 Cleavage Resistant CD16 Expressing CYNK Cells
  • CYNK cells were transduced with a CD16VP lentivirus and expanded as set forth above followed by analysis of cell surface marker expression, CD 16 evpression and CD16 shedding. See, FIG. 15A, FIG. 15B, FIG. 16, and FIG. 17.
  • CYNK-101 showed greater than 90% CD56+CD3-, less than 1% CD3 or CD 19, greater than 65% CD 16, and expression of NK surface markers such as CD226, NKG2D, CDlla, NKp30, NKp44, NKp46, and CD94.
  • CYNK-101 was resistant to CD 16 shedding following PMAi stimulation. [00422] CYNK-101 displayed cytotoxicity against K562 cells with a dose dependent manner. In the mixed targets culture system of K562 plus normal PBMCs, CYNK-101 can specifically kill K562 while sparing normal PBMCs even at the E:T ratio up to 100:1.
  • cytokine production such as GM-CSF, TNF-a, IFN-g was shown from CYNK-101 in the presence of K562, or stimulated with PMAi, or IL-12+IL- 18 compared to that of CYNK-101 alone.
  • CYNK-101 is a human placental hematopoietic stem cell derived natural killer (NK) cell product, that is genetically modified to express a variant of CD 16, Fc gamma receptor III (FcgRIII), via lentiviral vector transduction.
  • CD16 plays a central role in antibody dependent cell mediated cytotoxicity (ADCC) in NK cells through binding to the Fc portion of IgG antibodies.
  • FIG. 18 shows the rationale of ADCC by engineering CD 16 on NK cells.
  • CYNK-101 was designed to express a high-affinity proteolytic cleavage-resistant CD16 variant with a Valine at amino acid position 158 and Proline at position 197 as demonstrated in the Construct Design in FIG. 19 [Wu, 1997; Sugita, 1999; Koene, 1997;
  • CYNK-101 To assess the anti-tumor ADCC activity of CYNK-101, extensive in vitro, ex vivo and in vivo studies have been conducted using CYNK-101 in combination with trastuzumab (Herceptin) against HER2 + Gastric/Gastroesophageal Junction (G/GEJ) cancer cell lines, and HER2 + breast cancer cell lines.
  • trastuzumab Herceptin
  • G/GEJ Gastric/Gastroesophageal Junction
  • the in vitro study examined CYNK-101 multiparameter phenotypic characterization, CD16 shedding resistance evaluation of CYNK-101 followed by PMAi stimulation, cytotoxicity evaluation of CYNK-101 in combination with Herceptin against G/GEJ cancer cell lines as NCI-N87 and OE19; and breast cancer cell lines as AU565, BT-474, HCC-1954, SKBR-3, ZR-75-30; and assessment of cytokine secretion such as IFN-g, TNF-a, and GM-CSF from CYNK-101 in combination with Herceptin in the presence of the above tumor cell lines.
  • An ex vivo study was conducted to further evaluate ADCC activity of CYNK-101 administered to NSG mice.
  • CYNK-101 cells were isolated from mouse liver thirteen days post injection and evaluated for phenotypic characterization, CD 16 shedding resistance and anti -tumor ADCC activity. An in vivo study was further performed to evaluate the anti -tumor activity of CYNK-101 in combination with Trastuzumab in a subcutaneous gastric tumor mouse model. In addition, the safety of CYNK- 101 in combination with Trastuzumab was also evaluated in this study. The results summarized here provide a rationale to support clinical development of CYNK-101 in combination with trastuzumab against HER2 overexpressing G/GEJ cancer.
  • CYNK-101 cells (donor IDs: 2000112643, 2000113629, 2000113366, 2000113511, 2000113472, 2000113880, 2000114102) with purity of >85% CD56 + CD3 ' were served as effector cells in this assay.
  • the following HER2 overexpressing tumor cell lines were used as target cells: NCI-N87 (ATCC, Cat# CRL-5822), OE19 (Sigma, Cat#
  • 96071721) HCC-1954 (ATCC, Cat# CRL-2338), SKBR-3 (ATCC, Cat# HTB-30), BT-474 (ATCC, Cat# CRL-3247), AU565 (ATCC, Cat# CRL-2351), and ZR-75-30 (ATCC, Cat# CRL-1504).
  • the anti-HER2 antibody Herceptin Blue Door Pharma, Rockville, MD, Cat# 50242-132-01
  • the Ultra-LEAF purified human IgGi isotype control recombinant antibody Biolegend, Cat# 403501 was used at lpg/mL.
  • the xCELLigence real time cell analysis (RTCA) system (ACEA Biosciences, San Diego, CA) was used for the determination of
  • ADCC activities mediated by the combination of effector cells and antibody 8 x 10 4 /well NCI-N87, 6 x 10 4 /well OE19, 1 x 10 4 /well HCC-1954, 2 x 10 4 /well SKBR-3, 2 x 10 4 /well BT-474, 2 x 10 4 /well AU565, or 2 x 10 4 /well ZR-75-30 target cells were seeded in the E- plates in 100 pL of assay buffer (RPMI 1640 media supplemented with 10% fetal bovine serum (FBS), 1% penicillin and 1% streptomycin) for 24 hours at 37°C in 5% CCh.
  • assay buffer RPMI 1640 media supplemented with 10% fetal bovine serum (FBS), 1% penicillin and 1% streptomycin
  • IgGi or Herceptin was then added 30 minutes prior to the addition of different amounts of CYNK- 101 cells to achieve various E:T ratios (10:1, 5:1, 2.5:1, 1:1 and 0.6:1) for each donor in duplicate.
  • Target cells were incubated with CYNK-101 cells in the presence of the antibodies in 200 pL final volume.
  • the xCELLigence software was then used to determine the percent cytotoxicity at 4h and 24h after effector cells added. The results from different experiments were reported as mean of donors ⁇ standard deviation of the mean (SD).
  • CYNK-101 from seven different donors (donor IDs: 2000112643, 2000113629, 2000113366, 2000113511, 2000113472, 2000113880, 2000114102) used in all the assays were > 85% CD56 + CD3 " cells, > 70% viability by 7-AAD " .
  • Cytotoxicity assays were performed by using CYNK-101 cells as effector cells and the tumor cell lines, such as K562 (ATCC, Cat# CCL-243) as the target cells.
  • Target cell number was fixed at 1 x 10 4 while CYNK-101 cells were used in different amounts to achieve various E:T ratios (10:1, 5:1, 2.5:1, and 1.25:1).
  • target cells were labeled with 7.5 pM PKH26 fluorescent dye (Sigma- Aldrich, St Louis, MO, Cat# PKH26-GL).
  • Target cells were incubated with NK cells in 96-well U- bottom tissue culture plates in 200 pL of assay buffer for 4 hours at 37°C in 5% CO2. After incubation, cells were harvested and TO-PRO-3 (Invitrogen, Carlsbad, CA Cat# T3605), a membrane-impermeable DNA stain, was added to the cultures at 1 pM final concentration in order to identify dead cells (TO-PRO-3 4 ).
  • PKH26-labeled target cells were cultured alone for the duration of the assay.
  • 1 x 10 5 labeled target cells were permeabilized with 350 pL of Cytofix/Cytoperm buffer (BD Biosciences, Cat# 554722) for 20 minutes at 4°C.
  • the data were acquired on a FACSCanto X (BD Biosciences, San Jose, CA) and analyzed by FlowJo (Tree Star, Ashland, OR).
  • the percentage of dead target cells in each sample was calculated as follows: %TO-PRO-3 + PKH26 + cells / (%TO-PRO-3 + PKH26 + + %TO-PRO-3 PKH26 + ) * 100%. Percent cytotoxicity reported was calculated by subtracting the percent of dead target cells in cultures of target cells alone from the percent of dead target cells in co-cultures of effector and target cells. The results are reported as the mean of the donors ⁇ standard deviation of the mean (SD).
  • FACS buffer PBS with 0.5 mM EDTA and 2% FBS
  • human Fc block BD Biosciences, Cat# 564220.
  • the cells were stained with anti- CD16 PE (BD Biosciences, Cat# 556619), anti-CD56 APC (BD Biosciences, Cat# 555518), and anti-CDl la FITC (BD Biosciences, Cat# 555383) for 30 minutes at 4 °C.
  • NK receptors NKG2D (BD Biosciences, Cat# 562364), NKp46 (BD Biosciences, Cat# 563230), NKp44 (BD Biosciences, Cat# 744305), NKp30 (BD Biosciences, Cat# 563385), CD94 (R & D Systems, Cat# FAB1058A), CDlla (BD Biosciences, Cat# 561387), and DNAM-1 (BD Biosciences, Cat# 559788).
  • CYNK-101 cells (donor IDs: 2000112643, 2000113629, 2000113366, 2000113511, 2000113472, 2000113880, 2000114102) were plated in 96-well plates at 2 x 10 5 cells/well in 200 ⁇ L of RPMI 1640 medium with 10% FBS and antibiotics.
  • Stimulating agents were added to the corresponding wells as followings: IX Cell Stimulation Cocktail containing PMAi (eBiosciences, Cat# 00-4970-03), or IL-12 (20 ng/mL) (R&D Systems, Minneapolis, MN, Cat# 219-IL-025) plus IL-18 (100 ng/mL) (R&D Systems, Cat# B003-2). Additionally, 1 x 10 5 K562 tumor cells were added with the CYNK-101 cells at E:T ratio of 2: 1. Control wells were left unstimulated for the duration of stimulation.
  • the protein transport inhibitor Brefeldin A (BD Fastlmmune, San Diego, CA, Cat# 347688) was added to all the wells at 10 ⁇ g/mL final concentration. Stimulation was performed for an additional 4 hours, after which cells were labeled with a viability stain, LIVE/DEAD Aqua, according to the manufacturer’s protocol (Invitrogen, Cat# L34957). Cells were then stained with anti-CD56 PE-Cy7 (Biolegend, Cat# 362510), anti-CD3 APC-H7 (BD Biosciences,
  • Permeabilized cells were stained with anti-IFN-g APC (BD Biosciences, Cat# 554702), anti- TNF-a BV650 (BD Biosciences, Cat# 563418), anti-GM-CSF PE (BD Biosciences, Cat# 554507), anti-Perforin BV421 (Biolegend, Cat# 308122), and anti-Granzyme B AF700 (BD Biosciences, Cat# 560213) antibodies in brilliant stain buffer. Data were acquired on BD LSRFortessaX-20 and analyzed using the FlowJo software. Data were expressed as % positive cells gated under CD56 + CD3-Aqua- single cells.
  • the gate that delineated cells positive for a given cytokine was set by utilizing unstimulated samples, isotypes, and/or separation of stained populations. The data were analyzed and presented using GraphPad Prism (GraphPad Software, La Jolla, CA) and depicted as mean of the donors ⁇ SD.
  • CYNK-101 cells (donor IDs: 2000112643, 2000113629, 2000113366, 2000113511, 2000113472, 2000113880, 2000114102) were incubated with tumor cell lines, or tumor cell lines with either the IgGi or Herceptin antibodies at 1 pg/mL in 96-well U- bottom tissue culture plates at an E:T ratio of 1:1 (1 x 10 5 cells each) in 200 pL of RPMI 1640 supplemented with 10% FBS and antibiotics.
  • cytokine concentrations were determined by Luminex analysis using MILLIPLEX MAP magnetic bead kits (EMD Millipore, Burlington, MA, Cat# HCD8MAG-15K-07 for granulocyte-macrophage colony-stimulating factor (GM-CSF), perforin, TNF-a, IL-10, granzyme A, granzyme B and IFN-g) according to the protocol provided by the manufacturer.
  • the data were analyzed using Milliplex Xponent and Analyst software (EMD Millipore). The results from different experiments were depicted as mean of donors ⁇ SD.
  • NOD-scid IL2Rgamma nu11 immunodeficient (NSG) mice at DayO Recombinant human IL-15 was intraperitoneally injected at Days 0, 2, 4, 6, 8, 10 and 12 to support NK cell proliferation and survival.
  • CYNK-101 cells were isolated from mouse liver and evaluated for phenotype characterization, CD 16 shedding resistance followed by PMAi stimulation and ADCC activity against NCI-N87.
  • the phenotyping panel included the following: AQUA, mCD45, hCD45, CD3, CD56, CD16, CDlla, CD94, CD158a, CD158el/e2, CD158bl/b2, NKG2D, NKp46, andNKp44.
  • NCI-N87 gastric cancer cell line was subcutaneously inoculated at the dose of 3 x 10 6 cells/mouse on Day 0.
  • Trastuzumab (lOmg/kg) was administered intraperitonially (IP) once on Day 7, and CYNK-101 at the dose of 10 x 10 6 cells/mouse was administered intravenously (IV) three times on Days 7, 14 and 21.
  • Enhanced in vitro ADCC activities of CYNK-101 against G/GEJ tumor cell lines [00437] To assess the in vitro ADCC activity of CYNK-101 in combination with the anti-HER2 antibody Herceptin against HER2 overexpressing G/GEJ cancer cell lines, NCI- N87 and OE-19 were used as targets in a xCELLigence real time cell analysis-based assay. To assess NK cell dose dependent effects, varying E:T ratios from 10:1 to 0.6:1 for 7 different CYNK-101 donors were used in the assay. The Herceptin alone or IgGl control alone against the targets were included as controls. In addition, low HER2 expression normal human dermal fibroblasts cells (NHDF) were used as target cells to determine whether cytotoxic effects of CYNK-101 plus Herceptin were specific for HER2 + tumor cells.
  • NHDF normal human dermal fibroblasts cells
  • CYNK-101 when combined with Herceptin, displayed enhanced cytotoxic activity against both NCI-N87 and OE19 G/GEJ tumor cell lines at an E:T ratio as low as 0.6: 1 compared to when the IgG antibody was added. This activity increased with increasing E:T ratios in a dose-dependent manner at the early timepoint of 4h for both tumor lines and at 24h for OE19 (FIGS. 21A - 21B).
  • FIGS. 21 A - 21 B shows the average cytotoxic activity of CYNK-101 cells in combination with Herceptin or a control IgG against the indicated gastric tumor cell lines, NCI-N87 and OE19.
  • the error bars represent the SD from the mean calculated from seven different CYNK-101 donors. Cytotoxicity from Herceptin or IgG alone is included for reference. * indicate significantly higher activity of CYNK-101 in combination with Herceptin compared to that of IgG (***p ⁇ 0.001 and *p ⁇ 0.05).
  • CYNK-101 cells in combination with Herceptin specifically kill NCI-N87, while spare NHDF cells at any of the E:T ratios tested up to 100: 1 (FIG. 22B), indicating that CYNK-101 cells in combination with Herceptin were capable not only of lysing HER2 + tumor cells but also of discriminating between HER2 + normal and tumor targets.
  • IFN -gamma, TNF-alpha and GM-CSF cytokines secretion of IFN -gamma, TNF-alpha and GM-CSF cytokines in response to gastric cancer tumor cell lines
  • NK cell cytokine secretion is implicated in anti-tumor immune responses [Marcus, 20141.
  • CYNK-101 cytokine secretion was tested following co-culture with gastric cancer tumor cell lines. After 24 hours of co-culture, supernatant was analyzed for the presence of IFN-g, TNF-a, and GM-CSF. The results are summarized in Table 5.
  • IFN- ⁇ was significantly induced from CYNK-101 in the presence of both NCI-N87 and OE19 when Herceptin was added (Table 5).
  • CYNK-101 cells secreted 803 ⁇ 559 pg IFN-g with NCI-N87 and 88 ⁇ 72 pg with OE19 when Herceptin was present compared to 15 ⁇ 20 pg and 0 ⁇ 0 pg when IgGi was present, respectively; the secretion in the presence of IgG was not significantly different from that of the baseline for CYNK-101 cells of 13 ⁇ 16 pg.
  • TNF-a and GM-CSF were significantly induced in the presence of both gastric cancer tumor cell lines when Herceptin was added in combination (Table 5).
  • CYNK-101 cells secreted relevant immunomodulatory cytokines in the presence of Herceptin and gastric cancer tumor cell lines.
  • Herceptin included in the CYNK-101 cells in combination with Herceptin.
  • CYNK-101 cells in combination with Herceptin were not only capable of directly lysing gastric cancer tumor cells but also capable of indirectly eliciting anti-tumor responses through their ability to secrete IFN-g, TNF-a, and GM-CSF.
  • Table 5 Summary of cytokine secretion from CYNK-101 in response to gastric cancer tumor cells a) Number of CYNK-101 donors b) Average
  • CYNK-101 was stimulated with PMAi for 2h, followed by surface staining of CD16, non-transduced NK cells were served as control.
  • FIGS. 24 A - 24B followed by PMAi stimulation, the percentage of CD 16 expression on CYNK-101 maintained compared to that of control, while around 70% CD 16 expression decreased from non-transduced NK cells.
  • CYNK-101 demonstrates CD 16 shedding resistant post PMAi stimulation.
  • CYNK-101 cells express molecules required for cytotoxic activity
  • CYNK-101 cells expressed all of the factors required for cytotoxic activity tested: perforin (25.8% ⁇ 23.0% cells), granzyme B (30.4% ⁇ 30.4% cells), CD16 (74.1% ⁇ 5.6% cells), CD226 (49.6% ⁇ 9.7 % cells), NKG2D (40.6% ⁇ 14.0% cells), CD94 (48.9% ⁇ 10.7% cells), , NKp30 (37.4% ⁇ 16.8% cells), NKp44 (93.8% ⁇ 3.1% cells), andNKp46 (15.7% ⁇ 5.0% cells).
  • CYNK-101 cells exerted cytotoxic effects onto their targets by utilizing pathways that have been well described for NK cells involving recognition of tumor targets via activating receptor engagement and secretion of perforin and granzyme B-containing granules.
  • Enhanced in vitro ADCC activities of CYNK-101 against HER2+ breast cancer cell lines [00447] To assess the in vitro ADCC activity of CYNK-101 in combination with Herceptin against HER2 + breast cancer cell lines, AU565, BT-474, HCC-1954, SKBR-3, ZR- 75-30 were used as targets in axCELLigence real time cell analysis-based assay. To assess NK cell dose dependent effects, varying E:T ratios from 10:1 to 0.6:1 for 7 different CYNK- 101 donors (unless specified) were used in the assay.
  • FIGS. 25 A - 25B enhanced anti -tumor ADCC activity of CYNK-101 in combination with Herceptin against the HER2 + breast cancer cell lines was demonstrated at both 4h and 24h with the indicated E:T ratios compared to that of CYNK- 101 plus IgG control.
  • CYNK-101 cytokine secretion was tested following co-culture with breast cancer cell lines in the presence of Herceptin or IgG control. After 24 hours of co-culture, supernatant was analyzed for the production of IFN-g, TNF-a, and GM-CSF.
  • IFN-g was significantly induced from CYNK-101 in the presence of the breast cancer cell lines as indicated compared to that of IgG control.
  • TNF-a and GM-CSF were significantly induced in the presence of the breast cancer cell lines when Herceptin was added in combination.
  • CYNK-101 cells secreted relevant immunomodulatory cytokines in the presence of Herceptin and breast cancer cell lines.
  • CYNK-101 cells in combination with Herceptin were not only capable of directly lysing breast cancer cells but also capable of indirectly eliciting anti-tumor responses through their ability to secrete IFN-g, TNF-a, and GM-CSF.
  • CYNK-101 Ex vivo anti-tumor ADCC activity of CYNK-101 in combination with Herceptin against HER2+ gastric cancer cells
  • An ex vivo model was developed to further evaluate ADCC activity of CYNK- 101 administered to NSG mice.
  • CYNK-101 was IV-injected at the dose of 2 x 10 7 cells/animal into busulfan-pretreated NSG mice at Day 0.
  • Recombinant human IL-15 was intraperitoneally injected at Days 0, 2, 4, 6, 8, 10 and 12 to support CYNK-101 in vivo expansion and persistence.
  • CYNK-101 cells were isolated from mouse livers (ex v/voCYNK-101) and assessed for phenotype and ADCC activity.
  • FIG. 30 demonstrated that enhanced ADCC activity of ex v/voCYNK-101 in combination with Herceptin against NCI-N87 at E:T ratio of 0.5:1 compared to that of IgG, or in vitro CYNK-101 plus Herceptin, in vitro CYNK-101 plus IgG.
  • ex v/vo-CYNK-101 can be enriched from mouse liver followed by 13 days post injection.
  • Ex v/vo-CYNK-101 was CD 16 shedding resistance post PMAi stimulation.
  • Ex vivo-CYNK-101 in combination with Herceptin showed enhanced ADCC activity and enhanced cytokine secretion against NCI- N87 compared to that of ex v/vo-CYNK-101+IgG, in vitro CYNK-101+Herceptin, and in vitro CYNK-lOl+IgG.
  • CYNK-101 in combination with Trastuzumab demonstrated a synergistic anti-tumor activity and significantly suppressed tumor growth compared to Trastuzumab or CYNK-101 alone. More significantly, CYNK-101 plus Trastuzumab treatment continuously reduced tumor volume from Day 7 to Day 28, and approximately 30% tumor volume reduction was achieved on Day 28 compared to Day 7 baseline.
  • CYNK-101 Three repeated weekly IV administration of CYNK-101 at the dose of 10 x 10 6 cells/mouse was safe and well tolerated with or without Trastuzumab combination therapy. No abnormal clinical symptoms, morbidity and mortality occurred throughout the study. No CYNK-101 or CYNK-101 + Trastuzumab related abnormal effects on body weight and gross pathology at necropsy were observed.
  • Fc gammaRIIIa- 158V/F polymorphism influences the binding of IgG by natural killer cell Fc gammaRIIIa, independently of the Fc gammaRIIIa-48L/R/H phenotype. Blood. 1997 Aug 1;90(3): 1109-14.
  • Example 8 Monitoring T cell reactivity to allogeneic placental cell products using multiple lymphocyte reaction and TCR sequencing approaches
  • Allogeneic stem cell-derived therapies offer patients a readily accessible source of off-the-shelf product. However, allogeneic cells may be recognized by the recipient’s immune system thus negatively affecting persistence and efficacy.
  • allelic differences in human leukocyte antigen (HLA) complex genes are a major source of alloantigen which can elicit immunity. While there are established assays for measuring alloantibodies, monitoring T cell alloreactivity is more challenging. Unique clonal populations vary between individuals and functional assays are difficult to scale and standardize if alloreactive populations are rare/infrequent.
  • DC dendritic cell
  • LPS lipopoly saccharide
  • Proliferating alloantigen-specific T cells were observed by bright field microscopy and by flow cytometry.
  • T cell cytokine production was observed by multiplex analysis of supernatants from the MLR.
  • TCR locus sequencing was performed on cDNA libraries generated from unstimulated and post-MLR cultures using the Illumina platform.
  • Immune rejection is a potential limitation of allogeneic cell therapy.
  • monocytes can be cryopreserved from placental and peripheral blood in anticipation of peforming MLR functional assays. This permits the downstream monitoring of T cell alloreactivity to cell therapy products using a treated patient’s blood sample as a source of responding T cells.
  • Our data highlight the opportunity to identify alloreactive T cell clones from patients, followed by clone tracking over time following exposure to allogeneic cell therapy.

Abstract

Provided herein are methods of suppressing the proliferation of HER2+ tumor cells and methods of treating HER2+ cancers in a subject with populations of placental-derived natural killer cells comprising a cleavage resistant CD16. The cells, such as CYNK cells, can be placental CD34+ cell-derived natural killer (NK) cells. The present invention also provides methods of improving cell therapy by determining the presence of T cell clones reactive to the cell therapy.

Description

HER2+ CANCER TREATMENT WITH POPULATIONS OF NATURAL KILLER CELLS COMPRISING A CLEAVAGE RESISTANT CD16
[0001] This application claims priority to U.S. Provisional Patent Application Nos.
62/944,334, filed December 5, 2019, 62/944,341, filed December 5, 2019, 63/035,377, filed June 5, 2020, 63/035,496, filed June 5, 2020, and 63/115,953, filed November 19, 2020, the disclosures of which are incorporated herein by reference in their entireties.
1. FIELD
[0002] Provided herein are methods of producing populations of natural killer (NK) cells and/or ILC3 cells from a population of hematopoietic stem or progenitor cells in media comprising stem cell mobilizing factors, e.g., three-stage methods of producing NK cells and/or ILC3 cells in media comprising stem cell mobilizing factors starting with hematopoietic stem or progenitor cells from cells of the placenta, for example, from placental perfusate (e.g., human placental perfusate) or other tissues, for example, umbilical cord blood or peripheral blood. Further provided herein are methods of using the placental perfusate, the NK cells and/or ILC3 cells and/or NK progenitor cells described herein, to, e.g., suppress the proliferation of tumor cells, including multiple myeloma and acute myeloid leukemia cells.
2. BACKGROUND
[0003] Natural killer (NK) cells exhibit innate anti-tumor activity owing to the expression of a multitude of activating and inhibitory receptors that orchestrate NK cell responses. It is thus possible to use NK cells from allogeneic sources without the risk of graft-vs-host disease1, making them very attractive for developing “off-the-shelf’ cellular therapies. The anti-tumor responses of NK cells can be further enhanced by expressing Chimeric Antigen Receptors (CARs).
[0004] Celularity has developed a GMP process for generating off-the-shelf, allogeneic human Placental Hematopoietic Stem Cell (HSC) derived Natural Killer cells (PNK). The placental HSC source is vast, and Celularity process is streamlined to yield large quantities of differentiated and activated NK cells that have been well characterized. Here, we report the development of natural killer cells expressing a cleavage-resistant CD 16.
3. SUMMARY
[0005] The present invention provides methods of suppressing the proliferation of tumor cells comprising contacting the tumor cells with a population of placental-derived natural killer cells comprising a cleavage resistant CD 16 and an antibody, wherein the tumor cells are HER2+, and wherein the antibody is an anti-HER2 antibody.
[0006] The present invention also provides methods of treating a HER2+ cancer in a subject, comprising administering to the subject a population of placental-derived natural killer cells comprising a cleavage resistant CD 16 and an antibody, wherein the antibody is an anti-HER2 antibody.
[0007] In one or more embodiments of the invention the placental-derived natural killer (NK) cells are CYNK cells. In one or more embodiments of the invention the CYNK cells are placental CD34+ cell-derived natural killer (NK) cells.
[0008] In one or more embodiments of the invention the CYNK cells are characterized by expression of one or more markers selected from the group consisting of FGFBP2, GZMH, CCL3L3, GZMM, CXCR4, ZEB2, KLF2, LITAF, RORA, LYAR, CNOT1, IFNG, DUSP2, ATG2A, CD7, PMAIP1, PPP2R5C, NR4A2, ZFP36L2, PIK3R1, KLRFl, SNHG9, MT2A, RGS2, CHD1, DUSP1, EML4, ZFP36, ZC3H12A, DNAJB6, SBDS, IRFl, TSC22D3, TSPYL2, PNRC1, ISCA1, JUNB, WHAMM, RICTOR, TNFAIP3, EPC1, MVD, CLK1, ARL4C, REL, KMT2E, YPEL5, AMD1, BTG2, and IDS which is lower than expression of said markers in peripheral blood natural killer cells and / or expression of one or more markers selected from the group consisting of NDFIP2, LINC00996, MAL, CCL1, MB, SPINK2, C15orf48, CAMK1, KLRCl, TNFSF10, TNFRSF18, IL32, CAPG, AC092580.4, S100A11, TNFRSF4, ENOl, FCER1G, CCND2, KRT81, MRPS6, ANXA2, PTGER2, GLOl, HAVCR2, PYCARD, LAT2, SLC16A3, COTL1, PKM, TALDOl, CD96, NCR3, KRT86, STMN1, LTB, ARPC1B, ARPC5, FKBPIA, TIMP1, GZMK, CD59, PGK1, RGS10, EVL, RAC2, LGALS1, ITGB7, TUBB, PGAM1, PRF1, GZMB, IL2RB, KLRC2, and KLRBl which is higher than expression of said markers in peripheral blood natural killer cells.
[0009] In one or more embodiments of the invention the CYNK cells are characterized by expression of one or more markers selected from the group consisting of FGFBP2, GZMH, CCL3L3, GZMM, CXCR4, ZEB2, KLF2, LITAF, RORA, LYAR, CNOT1, IFNG, DUSP2, ATG2A, CD7, PMAIP1, PPP2R5C, NR4A2, ZFP36L2, PIK3R1, KLRFl, SNHG9, MT2A, RGS2, CHD1, DUSP1, EML4, ZFP36, ZC3H12A, DNAJB6, SBDS, IRFl, TSC22D3, TSPYL2, PNRC1, ISCA1, JUNB, WHAMM, RICTOR, TNFAIP3, EPC1, MVD, CLK1, ARL4C, REL, KMT2E, YPEL5, AMD1, BTG2, and IDS which is lower than expression of said markers in peripheral blood natural killer cells. [0010] In one or more embodiments of the invention expression of 2, 3, 4, 5, 6, 7, 8,
9, 10, or more markers selected from the group consisting of FGFBP2, GZMH, CCL3L3, GZMM, CXCR4, ZEB2, KLF2, LITAF, RORA, LYAR, CNOT1, IFNG, DUSP2, ATG2A, CD7, PMAIP1, PPP2R5C, NR4A2, ZFP36L2, PIK3R1, KLRFl, SNHG9, MT2A, RGS2, CHD1, DUSP1, EML4, ZFP36, ZC3H12A, DNAJB6, SBDS, IRF1, TSC22D3, TSPYL2, PNRC1, ISCA1, JUNB, WHAMM, RICTOR, TNFAIP3, EPC1, MVD, CLK1, ARL4C, REL, KMT2E, YPEL5, AMD1, BTG2, and IDS is lower than expression of said markers in peripheral blood natural killer cells.
[0011] In one or more embodiments of the invention the CYNK cells are characterized by expression of one or more markers selected from the group consisting of NDFIP2, LINC00996, MAL, CCL1, MB, SPINK2, C15orf48, CAMK1, KLRC1, TNFSF10, TNFRSF18, IL32, CAPG, AC092580.4, S100A11, TNFRSF4, ENOl, FCER1G, CCND2, KRT81, MRPS6, ANXA2, PTGER2, GLOl, HAVCR2, PYCARD, LAT2, SLC16A3, COTL1, PKM, TALDOl, CD96, NCR3, KRT86, STMN1, LTB, ARPC1B, ARPC5, FKBP1A, TIMP1, GZMK, CD59, PGK1, RGS10, EVL, RAC2, LGALS1, ITGB7, TUBB, PGAM1, PRF1, GZMB, IL2RB, KLRC2, and KLRBl which is higher than expression of said markers in peripheral blood natural killer cells.
[0012] In one or more embodiments of the invention the expression of 2, 3, 4, 5, 6, 7,
8, 9, 10, or more markers selected from the group consisting of NDFIP2, LINC00996, MAL, CCL1, MB, SPINK2, C15orf48, CAMK1, KLRC1, TNFSF10, TNFRSF18, IL32, CAPG, AC092580.4, S100A11, TNFRSF4, ENOl, FCER1G, CCND2, KRT81, MRPS6, ANXA2, PTGER2, GLOl, HAVCR2, PYCARD, LAT2, SLC16A3, COTL1, PKM, TALDOl, CD96, NCR3, KRT86, STMN1, LTB, ARPC1B, ARPC5, FKBP1A, TIMP1, GZMK, CD59, PGK1, RGS10, EVL, RAC2, LGALS1, ITGB7, TUBB, PGAM1, PRF1, GZMB, IL2RB, KLRC2, and KLRBl is higher than expression of said markers in peripheral blood natural killer cells. In one or more embodiments of the invention a nucleic acid encoding the cleavage resistant CD 16 has been introduced into the NK cells by transfection. In one or more embodiments of the invention a nucleic acid encoding the cleavage resistant CD 16 has been introduced into the NK cells by transduction. In one or more embodiments of the invention a nucleic acid encoding the cleavage resistant CD 16 has been introduced into the NK cells by retroviral transduction. In one or more embodiments of the invention a nucleic acid encoding the cleavage resistant CD 16 has been introduced into the NK cells by lentiviral transduction. In one or more embodiments of the invention greater than about 60%, greater than about 70%, greater than about 80%, greater than about 90%, or greater than about 95% of the cells in the population are CD56+ and CD3-.
[0013] In one or more embodiments of the invention less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% of the cells in the population are CD3+.
[0014] In one or more embodiments of the invention less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% of the cells in the population are CD19+.
[0015] In one or more embodiments of the invention greater than about 40%, greater than about 50%, greater than about 60%, or greater than about 65% of the cells in the population are greater than about 75% of the cells in the population are CD16+.
[0016] In one or more embodiments of the invention the population of cells comprises cells which express one or more surface markers selected from the group consisting of CD226, NKG2D, CD11a, NKp30, NKp44, NKp46, CD94, and combinations thereof In one or more embodiments of the invention the population of cells exhibit greater antibody- dependent cellular cytotoxicity than a population of placental-derived natural killer cells lacking expression of the cleavage resistant CD 16.
[0017] In one or more embodiments of the invention said contacting is contacting in vitro. In one or more embodiments of the invention said contacting is contacting in vivo. In one or more embodiments of the invention said contacting is in a human.
[0018] In one or more embodiments of the invention the cleavage resistant CD 16 comprises a valine residue at position 158. In one or more embodiments of the invention the cleavage resistant CD 16 comprises a proline residue at position 197. In one or more embodiments of the invention the cleavage resistant CD 16 is CD16VP. In one or more embodiments of the invention the cleavage resistant CD 16 does not comprise a proline residue at position 197.
[0019] In one or more embodiments of the invention the tumor cells are tumor cells from a cancer selected from the group consisting of Bladder cancers, Breast cancers, Cervical cancers, Cholangiocarcinomas (extrahepatic), Cholangiocarcinomas (intrahepatic), Colorectal cancers, Esophageal or esophagogastric junction cancers, Gallbladder cancers, Gastric adenocarcinomas, Gastrointestinal stromal tumors, Glioblastoma multiforme, high grade gliomas, Gliomas (low grade), Head and neck carcinomas, Hepatocellular carcinomas,
Intestinal (small) malignancies, Kidney cancers, Lung cancers (non small cells), Lung cancers (small cells), Melanomas, Melanomas (uveal), Neuroendocrine tumors, Oligodendrogliomas, Ovarian (epithelial) cancers, Ovarian (non-epithelial) cancers, Pancreatic adenocarcinomas, Penile cancers, Pituitary cancers, Prostate cancers, Sarcomas (peritoneal, retroperitoneal), Sarcomas (soft tissues), Solitary fibrous tumors, Testicular cancers, Thymic cancers, Thyroid cancers, Uterine cancers, and combinations thereof.
In preferred embodiments of the invention, the tumor cells are gastric cancer cells.
[0020] In one or more embodiments of the invention the anti-HER2 antibody is
Trastuzumab.
[0021] In one or more embodiments of the invention the population of placental- derived natural killer cells comprising a cleavage resistant CD 16 and the anti-HER2 antibody are administered sequentially. In one or more embodiments of the invention the population of placental-derived natural killer cells comprising a cleavage resistant CD 16 and the anti-HER2 antibody are administered concurrently.
[0022] In one or more embodiments of the invention the CYNK cells are prepared by the methods presented herein.
[0023] The present invention also provides a population of human placental-derived natural killer cells comprising a cleavage resistant CD16 for use in the manufacture of a medicament for treatment of a HER2+ cancer in a subject.
[0024] The present invention also provides the use of a composition comprising a population of human placental-derived natural killer cells comprising a cleavage resistant CD 16 for treatment of a HER2+ cancer in a subject.
[0025] In one or more embodiments of the invention the population of human placental-derived natural killer cells is a population of cells of the invention.
[0026] The present invention also provides methods of determining a patient’s reactivity to a given cell therapy treatment, the method comprising, obtaining a population of T cells from said patient, sequencing a portion of the T cell receptor repertoire of said T cells, obtaining a population of DCs from the donor from which said cell therapy was derived, contacting said T cells with said DCs in an MLR to encourage outgrowth of T cells which are reactive to said cell therapy, sequencing a portion of the T cell receptor repertoire of said reactive T cells, comparing the T cell receptor repertoire of said reactive T cells to the T cell receptor repertoire of the T cells obtained from the patient, and identifying the portion of the patients T cell receptor repertoire which is potentially reactive to the cell therapy. Terminology [0027] As used herein, the term CYNK are CD34+ cell-derived NK cells produced by the methods described herein. In specific embodiments, CYNK cells are placental-deived NK cells. In other specific embodiments, CYNK-001 and CYNK-101 are specific populations / formulations of CYNK cells.
[0028] As used herein, the terms “immunomodulatory compound” and “IMiD™” do not encompass thalidomide.
[0029] As used herein, “lenalidomide” means 3-(4'aminoisoindoline-r-one)-l- piperidine-2,6-dione (Chemical Abstracts Service name) or 2,6-Piperidinedione,3-(4-amino- l,3-dihydro-l-oxo-2H-isoindol-2-yl)- (International Union of Pure and Applied Chemistry (IUPAC) name). As used herein, “pomalidomide” means 4-amino-2-(2,6-dioxopiperidin-3- yl)isoindole- 1 ,3 -dione.
[0030] As used herein, “multipotent,” when referring to a cell, means that the cell has the capacity to differentiate into a cell of another cell type. In certain embodiments, “a multipotent cell” is a cell that has the capacity to grow into a subset of the mammalian body's approximately 260 cell types. Unlike a pluripotent cell, a multipotent cell does not have the capacity to form all of the cell types.
[0031] As used herein, “feeder cells” refers to cells of one type that are co-cultured with cells of a second type, to provide an environment in which the cells of the second type can be maintained, and perhaps proliferate. Without being bound by any theory, feeder cells can provide, for example, peptides, polypeptides, electrical signals, organic molecules (e.g., steroids), nucleic acid molecules, growth factors (e.g., bFGF), other factors (e.g., cytokines), and metabolic nutrients to target cells. In certain embodiments, feeder cells grow in a mono- layer.
[0032] As used herein, the “natural killer cells” or “NK cells” produced using the methods described herein, without further modification, include natural killer cells from any tissue source.
[0033] As used herein, the “ILC3 cells” produced using the methods described herein, without further modification, include ILC3 cells from any tissue source.
[0034] As used herein, “placental perfusate” means perfusion solution that has been passed through at least part of a placenta, e.g., a human placenta, e.g., through the placental vasculature, and includes a plurality of cells collected by the perfusion solution during passage through the placenta.
[0035] As used herein, “placental perfusate cells” means nucleated cells, e.g., total nucleated cells, isolated from, or isolatable from, placental perfusate. [0036] As used herein, “tumor cell suppression,” “suppression of tumor cell proliferation,” and the like, includes slowing the growth of a population of tumor cells, e.g., by killing one or more of the tumor cells in said population of tumor cells, for example, by contacting or bringing, e.g., NK cells or an NK cell population produced using a three-stage method described herein into proximity with the population of tumor cells, e.g., contacting the population of tumor cells with NK cells or an NK cell population produced using a three- stage method described herein. In certain embodiments, said contacting takes place in vitro or ex vivo. In other embodiments, said contacting takes place in vivo.
[0037] As used herein, the term “hematopoietic cells” includes hematopoietic stem cells and hematopoietic progenitor cells.
[0038] As used herein, the “undefined component” is a term of art in the culture medium field that refers to components whose constituents are not generally provided or quantified. Examples of an “undefined component” include, without limitation, serum, for example, human serum (e.g., human serum AB) and fetal serum (e.g., fetal bovine serum or fetal calf serum).
[0039] As used herein, “+”, when used to indicate the presence of a particular cellular marker, means that the cellular marker is detectably present in fluorescence activated cell sorting over an isotype control; or is detectable above background in quantitative or semi- quantitative RT-PCR.
[0040] As used herein,
Figure imgf000009_0001
when used to indicate the presence of a particular cellular marker, means that the cellular marker is not detectably present in fluorescence activated cell sorting over an isotype control; or is not detectable above background in quantitative or semi- quantitative RT-PCR.
4. BRIEF DESCRIPTION OF THE FIGURES
[0041] FIG. 1 shows expansion of NK cells for compounds CRLl - CRL11.
[0042] FIG. 2 shows expansion of NK cells for compounds CRL12 - CRL22.
[0043] FIG. 3 shows expansion of NK cells relative to SRI positive control.
[0044] FIG. 4 shows expansion of CD34+ cells from which the NK cells were derived.
[0045] FIGS. 5A - 5B show cytotoxicity of the expanded NK cultures.
[0046] FIGS. 6 A - 6C show that PNK cells highly express genes encoding the cytotoxic machinery. FIG. 6A CYNK cells were combined with peripheral blood derived NK cells (PB-NK) at 1:1 ratio and gene expression analyzed on single cell level using 10X Genomics Chromium platform and Illumina sequencing. Bioinformatics analysis utilized 10X Genomics Cell Ranger analysis pipeline. Transcript analysis was restricted to Granzyme B (GZMB) expressing cells. FIG. 6B A representative tSNE plot depicting PNK and PB-NK cells as distinct populations. FIG. 6C tSNE plots of selected NK cell-associated genes. The data is representative of two donors.
[0047] FIG. 7 shows that PNK and PB-NK cells differentially express genes encoding
NK cell receptors. The expression of selected NK cell receptor genes analyzed by real-time quantitative PCR in peripheral blood NK cells (PB-NK) and CD11a+ -bead-purified PNK cells. An alternative name indicated above the histogram for selected markers. The data represents mean ± SD of three donors for CYNK and PBNK cells (n=3). * p<0.05, ** p<0.005, *** pO.OOl.
[0048] FIG. 8 shows the gating strategy for PB-NK and CYNK cells. CYNK and
PBMC cells were thawed and stained with fluorophore-coupled antibodies targeting NK cell receptors. The figure demonstrates representative dot plots and the gating strategy for the identification of CYNK and PB-NK cells. See FIG. 9 for further characterization of the populations.
[0049] FIG. 9 shows differential expression of surface proteins on CYNK and PB-NK cells. CYNK and PB-NK cells were pre-gated as indicated in FIG. 8.
[0050] FIG. 10 shows that CYNK cells form a distinct cell population from PB-NK cells based on surface protein expression. tSNE plots demonstrating differential clustering of CYNK and PB-NK cells based on their surface markers. tSNE plots were generated of flow cytometry data using FlowJo software.
[0051] FIG. 11 A shows a schema of placental CD34+ cells expanded and differentiated to NK cells. FIG. 1 IB and FIG. 11C show total fold expansion of 35-day cultures and confirmation of NK phenotype of CD16VP cells (n=8 donors). FIG. 1 ID shows examples of phenotypic characterization of CD16VP cells.
[0052] FIG. 12A shows expression of CD 16 on non transduced cells (NT) and CD 16
VP groups after transduction and on day 35 (n= 8 donors). FIG. 12B and FIG. 12C show quantitative assessment of CD 16 shedding (n= 4) and representative flow cytometry diagrams showing CD 16 shedding of NT and CD 16 VP cells upon activation by PMA/ionomycin.
[0053] FIG. 13A shows representative examples of ADCC activities of CD 16 VP cells compared to NT cells against Daratumumab or Rituximab opsonized lymphoma cells lines (line graphs) and average ADCC activities of CD 16 VP cells compared to NT cells against Daratumumab opsonized lymphoma cells lines at 10 1 E/T ratio (n= 3 to 5) * indicates significant higher activities compared to NT control (bar graphs, p < 005). FIG 13B shows ADCC of CD 16 VP cells before and after PMA/ionomycin treatment against Daratumumab opsonized Daudi cells.
[0054] FIG. 14A shows a schematic study plan to test in vivo ADCC activities of
CD16VP cells using a disseminated Daudi Xenograft model. FIG 14B shows bioluminescence images of mice from each group after 10 days of cell infusion. FIG. 14C shows analysis of BLI after 10 days of cell infusion. * indicates a significant difference in between groups (n=6, p<0.05). FIG. 14D shows survival of mice through the course of the study.
[0055] FIGS. 15A - 15B show expression of cell surface markers on CYNK CD16-
VP cells.
[0056] FIG. 16 shows expression of CD 16 on CYNK CD16-VP cells.
[0057] FIG. 17 shows resistance to shedding of CD 16 on activated CYNK CD16-VP cells from multiple donors.
[0058] FIG. 18 shows a schema of CD 16 mediated lysis of opsonized tumor cell.
[0059] FIG. 19 shows a schema of CD16 with the high affinity (158V) modification and an exemplary shedding resistance (197P) modification.
[0060] FIG. 20 shows a study schema of an in vivo efficacy and safety study.
[0061] FIGS. 21 A and 21 B show that CYNK- 101 cells exert enhanced cytotoxic activity in combination with Herceptin against HER2 overexpressing gastric tumor cell lines. [0062] FIG. 22 shows CYNK-101 in combination with Herceptin specifically kill
HER2 overexpressing gastric tumor cells and spare low HER2 expression normal cells. FIG. 22A shows the expression of HER2 on NHDF, PBMC and NCI-N87. FIG. 22B shows the average cytotoxicity of CYNK-101 (n=3 donors) in combination with Herceptin against mixed targets of NCI-N87 and NHDF. CYNK-101 plus IgG, IgG alone and Herceptin alone served as control.
[0063] FIG. 23 shows that CYNK-101 cells express high expression of CD16. FIG.
23 shows CD16 expression on CD56+/CD3- CYNK-101 cells (mean ± SD, n=7 donors) with a representative CYNK-101 donor CD 16 flow cytometry graph shown on the left.
[0064] FIGS. 24A - 24B show that CYNK-101 is resistant to CD 16 shedding post
PMAi stimulation. FIG. 24 A shows representative flow cytometry graphs showing CD 16 loss following cleavage on NT cells but cleavage resistance on CYNK-101 cells following 2h activation by PMAi. FIG. 24B shows quantification of CD 16 cleavage resistance in CYNK- 101 cells (n=7 donors) and lack of cleavage resistance in a NT (non-transduced) donor.
[0065] FIGS. 25 A - 25B show that CYNK-101 cells exert enhanced cytotoxic activity in combination with Herceptin against HER2+ breast cancer cell lines. FIGS. 25A - 25B shows the average cytotoxic activity of CYNK-101 cells in combination with Herceptin or a control IgG against the indicated breast cancer cell lines, AU565, BT-474, HCC-1954, SK-BR-3 and ZR-75-30. The error bars represent the SD from the mean calculated from seven different CYNK-101 donors (unless specified). Cytotoxicity from Herceptin or IgG alone is included for reference. * indicate significantly higher activity of CYNK-101 in combination with Herceptin compared to that of IgG (***p<0.001, **p<0.005, and *p<0.05).
[0066] FIG. 26 shows increased IFN-g production of CYNK-101 in combination with
Herceptin against HER2+ breast cancer cell lines. FIG. 26 shows the average of IFN-g production of target cell alone, CYNK-101 alone, or CYNK-101 coculture with target cells in the presence of Herceptin or IgG control after 24h incubation. The error bars represent the SD from the mean calculated from different CYNK-101 donors (*p<0.05).
[0067] FIG. 27 shows the purity of CYNK-101 enriched from mouse liver post injection. FIG. 27 shows percent NK purity of the cells from the livers of NSG mice pre and post mouse cell depletion in ex vivo study. Representative data from total of three studies. [0068] FIG. 28 shows ex vivo CYNK-101 phenotype characterization. FIG. 28 shows
NK surface marker expression on the in vitro CYNK-101 donor (prior infusion CYNK-101) compared to the ex vivo CYNK-101.
[0069] FIG. 29 shows ex vivo CYNK-101 is resistant to PMAi stimulated CD 16 shedding. FIG. 29 shows in vitro CYNK-101 and ex vivo CYNK-101 shedding resistance post PMAi stimulation: CD 16 expression on control, in vitro CYNK-101 and ex vivo CYNK- 101 without stimulation (top graphs). CD16 expression on control, in vitro CYNK-101 and ex vivo CYNK-101 stimulated by PMAi for 2h (bottom graphs).
[0070] FIG. 30 shows ex vivo CYNK-101 displays ADCC activity against NCI-N87 at E:T ratio of 0.5:1. FIG. 30 shows cytotoxicity of ex vivo CYNK-101 or in vitro CYNK- 101 in combination with Herceptin against NCI-N87 at E:T ratio of 0.5:1. CYNK-lOl+IgG, Herceptin alone or IgG alone served as control.
[0071] FIG. 31 shows cytokine secretion profiling of ex vivo CYNK-101 in combination with Herceptin against NCI-N87. FIG. 31 shows cytokine secretion profiling of ex vivo CYNK-101 or in vitro CYNK-101 in combination with Herceptin against NCI-N87 at E:T ratio of 0.5:1.
[0072] FIG. 32 shows in vivo efficacy of CYNK-101 in combination with
Trastuzumab inNCI-N87 xenograft NSG-Tg-hIL 15 mouse model. FIG. 32 shows percentage of tumor volume change over Day 7 baseline. Animals were preconditioned with busulfan on Day -3. NCI-N87 cells were subcutaneously injected on Day 0. Trastuzumab was intraperitonially injected on Day 7. Vehicle or CYNK-101 was intravenously administered on Days 7, 14 and 21. The data are presented as the mean ± SEM; Multiple t tests were performed to compare Trastuzumab with Vehicle and Trastuzumab with CYNK-101 + Trastuzumab. ** P<0.01, *** P<0.001, ****P<0.0001.
[0073] FIG. 33 shows a model of identifying and tracking alloreactive T cell clones in patients.
[0074] FIG. 34 shows an overview of placental monocyte-derived DC one-way MLR assay.
[0075] FIG. 35 shows enrichment of CD33+ monocytes from placental blood. CD33+ monocytes were isolated from cryopreserved placental blood mononuclear cells using antihuman CD33+ magnetic selection beads (Miltenyi) according to manufacturer’s instructions. Preselected, CD33+, and CD33- fractions were stained for both viability and CD33 purity. [0076] FIGS. 36A - 36C shows that placental monocytes require additional stimulation to mature DCs. To generate antigen-presenting cells, placental CD33+ monocytes were matured to DC in presence of GM-CSF and IL-4. DCs were activated by LPS on day 6, or further matured with cytokines prior to LPS activation on day 7 where indicated. Upregulation of HLA-ABC and HLA-DR on maturing monocytes derived from placenta (FIG. 36A) or peripheral blood (FIG. 36B) is shown. (FIG. 36C) Placental monocytes from two donors monitored for HLA-ABC and HLA-DR expression during course of GM- CSF+IL4 maturation (blue), additional cytokine restimulation (orange), and post-LPS stimulations (green).
[0077] FIG. 37 shows a purity check of responder CD3+ T cells. CD3+ T cells isolated from representative unrelated cryopreserved PBMC donor using positive magnetic bead selection (Miltenyi).
[0078] FIGS. 38A - 38D show brightfield Microscopy of Expanding T Cell Clones.
Prior to harvest of MLR on day 5, images of MLRs and experimental controls were captured in 48-well plate using EVOS microscopic imaging system. FIG. 38A) Images of PBMC donor 3994 T cell only control and FIG. 38B) placental donor 777 DC + self T cell control. Images show absence of T cell clonal expansion in both controls. FIG. 38C) Placental donor 777 DC co-cultured + responding T cell donor 3994 MLR. FIG. 38D) Control blood monocyte derived DC + responding T cell donor 3994 MLR.
[0079] FIGS. 39A - 39C show that MLR using placental DCs yields expected T cell cytokine profiles. Four different placental donor monocyte- derived DCs were used in MLR with same allogeneic responding T cells isolated from one PBMC donor. Supernatants were collected at the end of the MLR. Multiplex analysis of cytokine production in MLRs were evaluated using Luminex FlexMAP3D custom plex kit cat# HCD8-MAG-15K with FIG. 39A) IFN-g, FIG. 39B) IL-2 and FIG. 39C) TNF-a analytes. No cytokine production was observed in T cell controls only.
[0080] FIGS. 40A - 40B show that MLR generates CD25+CD69+ proliferating T cells. Phenotype of CD4 and CD8 responding T cells prior to MLR (FIG. 40A), and after (FIG. 40B) MLR. 3 MLRs using T cell donor 5858 were used with stimulator DCs. Representative data from one MLR, 5856 DCs + 5858 T cells, is representative. CD4 and CD8 T cells are analyzed for CD25 and CD69 as markers of cell activation, as well as Ki-67 to assess active proliferation.
[0081] FIGS. 41A - 41B show that conserved T cell clones expand across multiple
DC donors. Responding T cells from donor 5858 were sequenced by Illuminanext generation sequencing using 5’ RACE at the V(D)J TCRab locus. Frequencies of TCR clones identified by the CDR3 variable region are shown and used for the basis of clonal frequency. (FIG. 41 A) Responding T cell clonal diversity from PBMC prior to MLR. Indicated TCR clones in color represent dominant clones identified from MLRs. (FIG. 41B) Responding T cell clonal diversity following MLR using 3 unique DC donors.
5. DETAILED DESCRIPTION
[0082] Provided herein are novel methods of producing and expanding NK cells and/or ILC3 cells from hematopoietic cells, e.g., hematopoietic stem cells or progenitor cells. Also provided herein are methods, e.g., three-stage methods, of producing NK cell populations and/or ILC3 cell populations from hematopoietic cells, e.g., hematopoietic stem cells or progenitor cells. The hematopoietic cells (e.g., CD34+ hematopoietic stem cells) used to produce the NK cells and/or ILC3 cells, and NK cell populations and/or ILC3 cell populations, may be obtained from any source, for example, without limitation, placenta, umbilical cord blood, placental blood, peripheral blood, spleen or liver. In certain embodiments, the NK cells and/or ILC3 cells or NK cell populations and/or ILC3 cell populations are produced from expanded hematopoietic cells, e.g., hematopoietic stem cells and/or hematopoietic progenitor cells. In one embodiment, hematopoietic cells are collected from a source of such cells, e.g., placenta, for example from placental perfusate, umbilical cord blood, placental blood, peripheral blood, spleen, liver (e.g., fetal liver) and/or bone marrow.
[0083] The hematopoietic cells used to produce the NK cells and/or ILC3 cells, and
NK cell populations and/or ILC3 cell populations, may be obtained from any animal species. In certain embodiments, the hematopoietic stem or progenitor cells are mammalian cells. In specific embodiments, said hematopoietic stem or progenitor cells are human cells. In specific embodiments, said hematopoietic stem or progenitor cells are primate cells. In specific embodiments, said hematopoietic stem or progenitor cells are canine cells. In specific embodiments, said hematopoietic stem or progenitor cells are rodent cells.
5.1. Hematopoietic Cells
[0084] Hematopoietic cells useful in the methods disclosed herein can be any hematopoietic cells able to differentiate into NK cells and/or ILC3 cells, e.g., precursor cells, hematopoietic progenitor cells, hematopoietic stem cells, or the like. Hematopoietic cells can be obtained from tissue sources such as, e.g., bone marrow, cord blood, placental blood, peripheral blood, liver or the like, or combinations thereof. Hematopoietic cells can be obtained from placenta. In a specific embodiment, the hematopoietic cells are obtained from placental perfusate. In one embodiment, the hematopoietic cells are not obtained from umbilical cord blood. In one embodiment, the hematopoietic cells are not obtained from peripheral blood. Hematopoietic cells from placental perfusate can comprise a mixture of fetal and maternal hematopoietic cells, e.g., a mixture in which maternal cells comprise greater than 5% of the total number of hematopoietic cells. In certain embodiments, hematopoietic cells from placental perfusate comprise at least about 90%, 95%, 98%, 99% or 99.5% fetal cells.
[0085] In another specific embodiment, the hematopoietic cells, e.g., hematopoietic stem cells or progenitor cells, from which the NK cell populations and/or ILC3 cell populations produced using a three-stage method described herein are produced, are obtained from placental perfusate, umbilical cord blood, fetal liver, mobilized peripheral blood, or bone marrow. In another specific embodiment, the hematopoietic cells, e.g., hematopoietic stem cells or progenitor cells, from which the NK cell populations and/or ILC3 cell populations produced using a three-stage method described herein are produced, are combined cells from placental perfusate and cord blood, e.g., cord blood from the same placenta as the perfusate. In another specific embodiment, said umbilical cord blood is isolated from a placenta other than the placenta from which said placental perfusate is obtained. In certain embodiments, the combined cells can be obtained by pooling or combining the cord blood and placental perfusate. In certain embodiments, the cord blood and placental perfusate are combined at a ratio of 100:1, 95:5, 90:10, 85:15, 80:20, 75:25, 70:30, 65:35, 60:40, 55:45: 50:50, 45:55, 40:60, 35:65, 30:70, 25:75, 20:80, 15:85, 10:90, 5:95, 100:1, 95:1, 90:1, 85:1, 80:1, 75:1, 70:1, 65:1, 60:1, 55:1, 50:1, 45:1, 40:1, 35:1, 30:1, 25:1, 20:1, 15:1, 10:1, 5:1, 1:1, 1:5, 1:10, 1:15, 1:20, 1:25, 1:30, 1:35, 1:40, 1:45, 1:50, 1:55, 1:60, 1:65, 1:70, 1:75, 1:80, 1:85, 1:90, 1:95, 1 : 100, or the like by volume to obtain the combined cells. In a specific embodiment, the cord blood and placental perfusate are combined at a ratio of from 10:1 to 1:10, from 5:1 to 1:5, or from 3:1 to 1:3. In another specific embodiment, the cord blood and placental perfusate are combined at a ratio of 10: 1, 5:1, 3:1, 1:1, 1:3, 1:5 or 1:10. In amore specific embodiment, the cord blood and placental perfusate are combined at a ratio of 8.5:1.5 (85%: 15%).
[0086] In certain embodiments, the cord blood and placental perfusate are combined at a ratio of 100:1, 95:5, 90:10, 85:15, 80:20, 75:25, 70:30, 65:35, 60:40, 55:45: 50:50, 45:55, 40:60, 35:65, 30:70, 25:75, 20:80, 15:85, 10:90, 5:95, 100:1, 95:1, 90:1, 85:1, 80:1, 75:1, 70:1, 65:1, 60:1, 55:1, 50:1, 45:1, 40:1, 35:1, 30:1, 25:1, 20:1, 15:1, 10:1, 5:1, 1:1, 1:5, 1:10, 1:15, 1:20, 1:25, 1:30, 1:35, 1:40, 1:45, 1:50, 1:55, 1:60, 1:65, 1:70, 1:75, 1:80, 1:85, 1:90, 1:95, 1: 100, or the like by total nucleated cells (TNC) content to obtain the combined cells. In a specific embodiment, the cord blood and placental perfusate are combined at a ratio of from 10:1 to 10:1, from 5:1 to 1:5, or from 3:1 to 1: 3. In another specific embodiment, the cord blood and placental perfusate are combined at a ratio of 10:1, 5:1, 3:1, 1:1, 1:3, 1:5 or 1:10.
[0087] In another specific embodiment, the hematopoietic cells, e.g., hematopoietic stem cells or progenitor cells from which said NK cell populations and/or ILC3 cell populations produced using a three-stage method described herein are produced, are from both umbilical cord blood and placental perfusate, but wherein said umbilical cord blood is isolated from a placenta other than the placenta from which said placental perfusate is obtained.
[0088] In certain embodiments, the hematopoietic cells are CD34+ cells. In specific embodiments, the hematopoietic cells useful in the methods disclosed herein are
CD34+CD38+ or CD34 CD38 . In a more specific embodiment, the hematopoietic cells are CD34+CD38 Lin . In another specific embodiment, the hematopoietic cells are one or more of CD2- CD3 , CDllb , CDllc , CD 14-, CD 16 , CD19 , CD24 , CD56 , CD66b and/or glycophorin A. In another specific embodiment, the hematopoietic cells are CD2 . CD3 . CDllb , CDllc , CD 14 . CD 16 . CD 19 . CD24 . CD56 . CD66b and glycophorin A. In another more specific embodiment, the hematopoietic cells are CD34 CD38 CD33 CD1 17 . In another more specific embodiment, the hematopoietic cells are CD34 CD38 CD33 CD117 CD235 CD36 .
[0089] In another embodiment, the hematopoietic cells are CD45+. In another specific embodiment, the hematopoietic cells are CD34+CD45+. In another embodiment, the hematopoietic cell is Thy-1+. In a specific embodiment, the hematopoietic cell is CD34+Thy- 1+. In another embodiment, the hematopoietic cells are CD133+. In specific embodiments, the hematopoietic cells are CD34+CD133+ or CD133+Thy-1+. In another specific embodiment, the CD34+ hematopoietic cells are CXCR4+. In another specific embodiment, the CD34+ hematopoietic cells are CXCR4 . In another embodiment, the hematopoietic cells are positive for KDR (vascular growth factor receptor 2). In specific embodiments, the hematopoietic cells are CD34+KDR+, CD133+KDR+ or Thy-1+KDR+. In certain other embodiments, the hematopoietic cells are positive for aldehyde dehydrogenase (ALDH+), e.g., the cells are CD34+ALDH+.
[0090] In certain other embodiments, the CD34+ cells are CD45 . In specific embodiments, the CD34+ cells, e.g., CD34+, C D45 cells express one or more, or all, of the miRNAs hsa-miR-380, hsa-miR-512, hsa-miR-517, hsa-miR-518c, hsa-miR-519b, hsa-miR- 520a, hsa-miR-337, hsa-miR-422a, hsa-miR-549, and/or hsa-miR-618.
[0091] In certain embodiments, the hematopoietic cells are CD34 .
[0092] The hematopoietic cells can also lack certain markers that indicate lineage commitment, or a lack of developmental naivete. For example, in another embodiment, the hematopoietic cells are HLA-DR . In specific embodiments, the hematopoietic cells are CD34+HLA-DR , CD133 HLA-DR . Thy-1+HLA-DR or ALDH1 HLA-DR In another embodiment, the hematopoietic cells are negative for one or more, or all, of lineage markers CD2, CD3, CDllb, CDllc, CD14, CD16, CD19, CD24, CD56, CD66b and glycophorin A. [0093] Thus, hematopoietic cells can be selected for use in the methods disclosed herein on the basis of the presence of markers that indicate an undifferentiated state, or on the basis of the absence of lineage markers indicating that at least some lineage differentiation has taken place. Methods of isolating cells, including hematopoietic cells, on the basis of the presence or absence of specific markers is discussed in detail below. [0094] Hematopoietic cells used in the methods provided herein can be a substantially homogeneous population, e.g., a population comprising at least about 95%, at least about 98% or at least about 99% hematopoietic cells from a single tissue source, or a population comprising hematopoietic cells exhibiting the same hematopoietic cell-associated cellular markers. For example, in various embodiments, the hematopoietic cells can comprise at least about 95%, 98% or 99% hematopoietic cells from bone marrow, cord blood, placental blood, peripheral blood, or placenta, e.g., placenta perfusate.
[0095] Hematopoietic cells used in the methods provided herein can be obtained from a single individual, e.g., from a single placenta, or from a plurality of individuals, e.g., can be pooled. Where the hematopoietic cells are obtained from a plurality of individuals and pooled, the hematopoietic cells may be obtained from the same tissue source. Thus, in various embodiments, the pooled hematopoietic cells are all from placenta, e.g., placental perfusate, all from placental blood, all from umbilical cord blood, all from peripheral blood, and the like.
[0096] Hematopoietic cells used in the methods disclosed herein can, in certain embodiments, comprise hematopoietic cells from two or more tissue sources. For example, in certain embodiments, when hematopoietic cells from two or more sources are combined for use in the methods herein, a plurality of the hematopoietic cells used to produce natural killer cells using a three-stage method described herein comprise hematopoietic cells from placenta, e.g., placenta perfusate. In various embodiments, the hematopoietic cells used to produce NK cell populations and/or ILC3 cell populations produced using a three-stage method described herein, comprise hematopoietic cells from placenta and from cord blood; from placenta and peripheral blood; from placenta and placental blood, or placenta and bone marrow. In one embodiment, the hematopoietic cells comprise hematopoietic cells from placental perfusate in combination with hematopoietic cells from cord blood, wherein the cord blood and placenta are from the same individual, i.e., wherein the perfusate and cord blood are matched. In embodiments in which the hematopoietic cells comprise hematopoietic cells from two tissue sources, the hematopoietic cells from the sources can be combined in a ratio of, for example, 1:10, 2:9, 3:8, 4:7:, 5:6, 6:5, 7:4, 8:3, 9:2, 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1 or 9:1.
5.1.1. Placental Hematopoietic Stem Cells
[0097] In certain embodiments, the hematopoietic cells used in the methods provided herein are placental hematopoietic cells. In one embodiment, placental hematopoietic cells are CD34+. In a specific embodiment, the placental hematopoietic cells are predominantly (e.g., at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 98%)
CD34 CD38 cells. In another specific embodiment, the placental hematopoietic cells are predominantly (e.g., at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 98%) CD34+CD38+ cells. Placental hematopoietic cells can be obtained from a postpartum mammalian (e.g., human) placenta by any means known to those of skill in the art, e.g., by perfusion.
[0098] In another embodiment, the placental hematopoietic cell is CD45 . In a specific embodiment, the hematopoietic cell is CD34 CD45 . In another specific embodiment, the placental hematopoietic cells are CD34+CD45+.
5.2. Production of Natural Killer and/or ILC3 Cells and Natural Killer Cell and/or ILC3 Cell Populations
[0099] Production of NK cells and/or ILC3 cells and NK cell and/or ILC3 cell populations by the present methods comprises expanding a population of hematopoietic cells. During cell expansion, a plurality of hematopoietic cells within the hematopoietic cell population differentiate into NK cells and/or ILC3 cells. In one aspect, provided herein is a method of producing NK cells comprising culturing hematopoietic stem cells or progenitor cells, e.g., CD34+ stem cells or progenitor cells, in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells, subsequently culturing said first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells, and subsequently culturing said second population of cells in a third medium comprising IL-2 and IL-15, and lacking a stem cell mobilizing agent and LMWH, to produce a third population of cells, wherein the third population of cells comprises natural killer cells that are CD56+, CD3-, and wherein at least 70%, for example at least 80%, of the natural killer cells are viable. In certain embodiments, such natural killer cells comprise natural killer cells that are CD 16-, In certain embodiments, such natural killer cells comprise natural killer cells that are CD94+. In certain embodiments, such natural killer cells comprise natural killer cells that are CD94+ or CD16+. In certain embodiments, such natural killer cells comprise natural killer cells that are CD94- or CD 16-, In certain embodiments, such natural killer cells comprise natural killer cells that are CD94+ and CD16+. In certain embodiments, such natural killer cells comprise natural killer cells that are CD94- and CD16-. In certain embodiments, said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1α). In certain embodiments, said third medium lacks LIF, MIP-1α, and FMS-like tyrosine kinase-3 ligand (Flt-3L). In specific embodiments, said first medium and said second medium lack LIF and MIP-1α, and said third medium lacks LIF, MIP-1α, and Flt3L. In certain embodiments, none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
[00100] In one aspect, provided herein is a method of producing NK cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells; and (c) culturing the second population of cells in a third medium comprising IL-2 and IL-15, and lacking LMWH, to produce a third population of cells; wherein the third population of cells comprises natural killer cells that are CD56+, CD3-, and CD11a+. In certain embodiments, said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1α). In certain embodiments, said third medium lacks LIF, MIP-1α, and FMS-like tyrosine kinase-3 ligand (Flt-3L). In specific embodiments, said first medium and said second medium lack LIF and MIP-1α, and said third medium lacks LIF, MIP-1α, and Flt3L. In certain embodiments, none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
[00101] In one aspect, provided herein is a method of producing NK cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells; and (c) culturing the second population of cells in a third medium comprising IL-2 and IL-15, and lacking each of stem cell factor (SCF) and LMWH, to produce a third population of cells; wherein the third population of cells comprises natural killer cells that are CD56+, CD3-, and CD11a+. In certain embodiments, said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1α). In certain embodiments, said third medium lacks LIF, MIP-1α, and FMS-like tyrosine kinase- 3 ligand (Flt-3L). In specific embodiments, said first medium and said second medium lack
LIF and MIP-1α, and said third medium lacks LIF, MIP-1α, and Flt3L. In certain embodiments, none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
[00102] In one aspect, provided herein is a method of producing NK cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells; and (c) culturing the second population of cells in a third medium comprising IL-2 and IL-15, and lacking each of SCF, a stem cell mobilizing agent, and LMWH, to produce a third population of cells; wherein the third population of cells comprises natural killer cells that are CD56+, CD3-, and CD11a+. In certain embodiments, said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1α). In certain embodiments, said third medium lacks LIF, MIP-1α, and FMS- like tyrosine kinase-3 ligand (Flt-3L). In specific embodiments, said first medium and said second medium lack LIF and MIP-1α, and said third medium lacks LIF, MIP-1α, and Flt3L. In certain embodiments, none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
[00103] In one aspect, provided herein is a method of producing NK cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells;
(c) culturing the second population of cells in a third medium comprising IL-2 and IL-15, and lacking each of a stem cell mobilizing agent and LMWH, to produce a third population of cells; and (d) isolating CD11a+ cells from the third population of cells to produce a fourth population of cells; wherein the fourth population of cells comprises natural killer cells that are CD56+, CD3-, and CDlla+. In certain embodiments, said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1α). In certain embodiments, said third medium lacks LIF, MIP-1α, and FMS-like tyrosine kinase-3 ligand (Flt-3L). In specific embodiments, said first medium and said second medium lack LIF and MIP-1α, and said third medium lacks LIF, MIP-1α, and Flt3L. In certain embodiments, none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin. [00104] In certain embodiments, of any of the above embodiments, said natural killer cells express perforin and EOMES. In certain embodiments, said natural killer cells do not express either RORyt or IL1R1.
[00105] In one aspect, provided herein is a method of producing ILC3 cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells; and (c) culturing the second population of cells in a third medium comprising IL-2 and IL-15, and lacking LMWH, to produce a third population of cells; wherein the third population of cells comprises ILC3 cells that are CD56+, CD3-, and CDlla-. In certain embodiments, said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein- 1 alpha (MIP-1α). In certain embodiments, said third medium lacks LIF, MIP-1α, and FMS-like tyrosine kinase-3 ligand (Flt-3L). In specific embodiments, said first medium and said second medium lack LIF and MIP-1α, and said third medium lacks LIF, MIP-1α, and Flt3L. In certain embodiments, none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
[00106] In one aspect, provided herein is a method of producing ILC3 cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells; and (c) culturing the second population of cells in a third medium comprising a stem cell mobilizing agent, IL-2 and IL-15, and lacking LMWH, to produce a third population of cells; wherein the third population of cells comprises ILC3 cells that are CD56+, CD3-, and CD11a-. In certain embodiments, said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1α). In certain embodiments, said third medium lacks LIF, MIP-1α, and FMS-like tyrosine kinase- 3 ligand (Flt-3L). In specific embodiments, said first medium and said second medium lack LIF and MIP-1α, and said third medium lacks LIF, MIP-1α, and Flt3L. In certain embodiments, none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin. [00107] In one aspect, provided herein is a method of producing ILC3 cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells; and (c) culturing the second population of cells in a third medium comprising SCF, IL-2 and IL-15, and lacking LMWH, to produce a third population of cells; wherein the third population of cells comprises ILC3 cells that are CD56+, CD3-, and CDlla-. In certain embodiments, said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1α). In certain embodiments, said third medium lacks LIF, MIP-1α, and FMS-like tyrosine kinase-3 ligand (Flt-3L). In specific embodiments, said first medium and said second medium lack LIF and MIP-1α, and said third medium lacks LIF, MIP-1α, and Flt3L. In certain embodiments, none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
[00108] In one aspect, provided herein is a method of producing ILC3 cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells; and (c) culturing the second population of cells in a third medium comprising a stem cell mobilizing agent, SCF, IL-2 and IL-15, and lacking LMWH, to produce a third population of cells; wherein the third population of cells comprises ILC3 cells that are CD56+, CD3-, and CD11a-. In certain embodiments, said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1α). In certain embodiments, said third medium lacks LIF, MIP-1α, and FMS-like tyrosine kinase- 3 ligand (Flt-3L). In specific embodiments, said first medium and said second medium lack LIF and MIP-1α, and said third medium lacks LIF, MIP-1α, and Flt3L. In certain embodiments, none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
[00109] In one aspect, provided herein is a method of producing ILC3 cells comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacking Tpo, to produce a second population of cells;
(c) culturing the second population of cells in a third medium comprising IL-2 and IL-15, and lacking each of a stem cell mobilizing agent and LMWH, to produce a third population of cells; and (d) isolating CD11a- cells, or removing CD11a+ cells, from the third population of cells to produce a fourth population of cells; wherein the fourth population of cells comprises ILC3 cells that are CD56+, CD3-, and CD11a-. In certain embodiments, said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1α). In certain embodiments, said third medium lacks LIF, MIP-1α, and FMS-like tyrosine kinase-3 ligand (Flt-3L). In specific embodiments, said first medium and said second medium lack LIF and MIP-1α, and said third medium lacks LIF, MIP-1α, and Flt3L. In certain embodiments, none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin.
[00110] In certain embodiments, said ILC3 cells express RORyt and IL1R1. In certain embodiments, said ILC3 cells do not express either perforin or EOMES.
5.2.1. Production of NK Cell and/or ILC3 Cell Populations Using a Three- Stage Method
[00111] In one embodiment, provided herein is a three-stage method of producing NK cell and/or ILC3 cell populations. In certain embodiments, the method of expansion and differentiation of the hematopoietic cells, as described herein, to produce NK cell and/or ILC3 cell populations according to a three-stage method described herein comprises maintaining the cell population comprising said hematopoietic cells at between about 2 x 104 and about 6 x 106 cells per milliliter. In certain aspects, said hematopoietic stem or progenitor cells are initially inoculated into said first medium from 1 x 104 to 1 x 105 cells/mL. In a specific aspect, said hematopoietic stem or progenitor cells are initially inoculated into said first medium at about 3 x 104 cells/mL.
[00112] In certain aspects, said first population of cells are initially inoculated into said second medium from 5 x 104 to 5 x 105 cells/mL. In a specific aspect, said first population of cells is initially inoculated into said second medium at about 1 x 105 cells/mL.
[00113] In certain aspects said second population of cells is initially inoculated into said third medium from 1 x 105 to 5 x 106 cells/mL. In certain aspects, said second population of cells is initially inoculated into said third medium from 1 x 105 to 1 x 106 cells/mL. In a specific aspect, said second population of cells is initially inoculated into said third medium at about 5 x 105 cells/mL. In a more specific aspect, said second population of cells is initially inoculated into said third medium at about 5 x 105 cells/mL in a spinner flask. In a specific aspect, said second population of cells is initially inoculated into said third medium at about 3 x lO5 cells/mL. In a more specific aspect, said second population of cells is initially inoculated into said third medium at about 3 x 105 cells/mL in a static culture. [00114] In a certain embodiment, the three-stage method comprises a first stage (“stage 1”) comprising culturing hematopoietic stem cells or progenitor cells, e.g., CD34+ stem cells or progenitor cells, in a first medium for a specified time period, e.g., as described herein, to produce a first population of cells. In certain embodiments, the first medium comprises a stem cell mobilizing agent and thrombopoietin (Tpo). In certain embodiments, the first medium comprises in addition to a stem cell mobilizing agent and Tpo, one or more of LMWH, Flt-3L, SCF, IL-6, IL-7, G-CSF, and GM-CSF. In a specific embodiment, the first medium comprises in addition to a stem cell mobilizing agent and Tpo, each of LMWH, Flt- 3L, SCF, IL-6, IL-7, G-CSF, and GM-CSF. In a specific embodiment, the first medium lacks added LMWH. In a specific embodiment, the first medium lacks added desulphated glycosaminoglycans. In a specific embodiment, the first medium lacks LMWH. In a specific embodiment, the first medium lacks desulphated glycosaminoglycans. In a specific embodiment, in addition to a stem cell mobilizing agent and Tpo, each of Flt-3L, SCF, IL-6, IL-7, G-CSF, and GM-CSF. In specific embodiments, the first medium lacks leukemia inhibiting factor (LIF), macrophage inhibitory protein- 1 alpha (MIP-1α) or both.
[00115] In certain embodiments, subsequently, in “stage 2” said cells are cultured in a second medium for a specified time period, e.g., as described herein, to produce a second population of cells. In certain embodiments, the second medium comprises a stem cell mobilizing agent and interleukin- 15 (IL-15) and lacks Tpo. In certain embodiments, the second medium comprises, in addition to a stem cell mobilizing agent and IL-15, one or more of LMWH, Flt-3, SCF, IL-6, IL-7, G-CSF, and GM-CSF. In certain embodiments, the second medium comprises, in addition to a stem cell mobilizing agent and IL-15, each of LMWH, Flt-3, SCF, IL-6, IL-7, G-CSF, and GM-CSF. In a specific embodiment, the second medium lacks added LMWH. In a specific embodiment, the second medium lacks added desulphated glycosaminoglycans. In a specific embodiment, the second medium lacks heparin, e.g., LMWH. In a specific embodiment, the second medium lacks desulphated glycosaminoglycans. In certain embodiments, the second medium comprises, in addition to a stem cell mobilizing agent and IL-15, each of Flt-3, SCF, IL-6, IL-7, G-CSF, and GM-CSF.
In specific embodiments, the second medium lacks leukemia inhibiting factor (LIF), macrophage inhibitory protein- 1 alpha (MIP-1α) or both. [00116] In certain embodiments, subsequently, in “stage 3” said cells are cultured in a third medium for a specified time period, e.g., as described herein, to produce a third population of cell, e.g., natural killer cells. In certain embodiments, the third medium comprises IL-2 and IL-15, and lacks a stem cell mobilizing agent and LMWH. In certain embodiments, the third medium comprises in addition to IL-2 and IL-15, one or more of SCF, IL-6, IL-7, G-CSF, and GM-CSF. In certain embodiments, the third medium comprises, in addition to IL-2 and IL-15, each of SCF, IL-6, IL-7, G-CSF, and GM-CSF. In specific embodiments, the first medium lacks one, two, or all three of LIF, MIP-1α, and Flt3L. In specific embodiments, the third medium lacks added desulphated glycosaminoglycans. In specific embodiments, the third medium lacks desulphated glycosaminoglycans. In specific embodiments, the third medium lacks heparin, e.g., LMWH.
[00117] In a specific embodiment, the three-stage method is used to produce NK cell and/or ILC3 cell populations. In certain embodiments, the three-stage method is conducted in the absence of stromal feeder cell support. In certain embodiments, the three-stage method is conducted in the absence of exogenously added steroids (e.g., cortisone, hydrocortisone, or derivatives thereof).
[00118] In certain aspects, said first medium used in the three-stage method comprises a stem cell mobilizing agent and thrombopoietin (Tpo). In certain aspects, the first medium used in the three-stage method comprises, in addition to a stem cell mobilizing agent and Tpo, one or more of Low Molecular Weight Heparin (LMWH), Flt-3 Ligand (Flt-3L), stem cell factor (SCF), IL-6, IL-7, granulocyte colony-stimulating factor (G-CSF), or granulocyte- macrophage-stimulating factor (GM-CSF). In certain aspects, the first medium used in the three-stage method comprises, in addition to a stem cell mobilizing agent and Tpo, each of LMWH, Flt-3L, SCF, IL-6, IL-7, G-CSF, and GM-CSF. In certain aspects, the first medium used in the three-stage method comprises, in addition to a stem cell mobilizing agent and Tpo, each of Flt-3L, SCF, IL-6, IL-7, G-CSF, and GM-CSF. In a specific aspect, the first medium lacks added LMWH. In a specific aspect, the first medium lacks added desulphated glycosaminoglycans. In a specific aspect, the first medium lacks LMWH. In a specific aspect, the first medium lacks desulphated glycosaminoglycans. In certain aspects, said Tpo is present in the first medium at a concentration of from 1 ng/mL to 100 ng/mL, from 1 ng/mL to 50 ng/mL, from 20 ng/mL to 30 ng/mL, or about 25 ng/mL. In other aspects, said Tpo is present in the first medium at a concentration of from 100 ng/mL to 500 ng/mL, from 200 ng/mL to 300 ng/mL, or about 250 ng/mL. In certain aspects, when LMWH is present in the first medium, the LMWH is present at a concentration of from lU/mL to lOU/mL; the Flt-3L is present at a concentration of from 1 ng/mL to 50 ng/mL; the SCF is present at a concentration of from 1 ng/mL to 50 ng/mL; the IL-6 is present at a concentration of from 0.01 ng/mL to 0.1 ng/mL; the IL-7 is present at a concentration of from 1 ng/mL to 50 ng/mL; the G-CSF is present at a concentration of from 0.01 ng/mL to 0.50 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.1 ng/mL. In certain aspects, in the first medium, the Flt-3L is present at a concentration of from 1 ng/mL to 50 ng/mL; the SCF is present at a concentration of from 1 ng/mL to 50 ng/mL; the IL-6 is present at a concentration of from 0.01 ng/mL to 0.1 ng/mL; the IL-7 is present at a concentration of from 1 ng/mL to 50 ng/mL; the G-CSF is present at a concentration of from 0.01 ng/mL to 0.50 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.1 ng/mL. In certain aspects, when LMWH is present in the first medium, the LMWH is present at a concentration of from 4U/mL to 5U/mL; the Flt-3L is present at a concentration of from 20 ng/mL to 30 ng/mL; the SCF is present at a concentration of from 20 ng/mL to 30 ng/mL; the IL-6 is present at a concentration of from 0.04 ng/mL to 0.06 ng/mL; the IL-7 is present at a concentration of from 20 ng/mL to 30 ng/mL; the G-CSF is present at a concentration of from 0.20 ng/mL to 0.30 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.5 ng/mL. In certain aspects, in the first medium, the Flt-3L is present at a concentration of from 20 ng/mL to 30 ng/mL; the SCF is present at a concentration of from 20 ng/mL to 30 ng/mL; the IL-6 is present at a concentration of from 0.04 ng/mL to 0.06 ng/mL; the IL-7 is present at a concentration of from 20 ng/mL to 30 ng/mL; the G-CSF is present at a concentration of from 0.20 ng/mL to 0.30 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.5 ng/mL. In certain aspects, when LMWH is present in the first medium, the LMWH is present at a concentration of about 4.5U/mL; the Flt-3L is present at a concentration of about 25 ng/mL; the SCF is present at a concentration of about 27 ng/mL; the IL-6 is present at a concentration of about 0.05 ng/mL; the IL-7 is present at a concentration of about 25 ng/mL; the G-CSF is present at a concentration of about .25 ng/mL; and the GM-CSF is present at a concentration of about 0.01 ng/mL. In certain aspects, in the first medium, the Flt-3L is present at a concentration of about 25 ng/mL; the SCF is present at a concentration of about 27 ng/mL; the IL-6 is present at a concentration of about 0.05 ng/mL; the IL-7 is present at a concentration of about 25 ng/mL; the G-CSF is present at a concentration of about .25 ng/mL; and the GM-CSF is present at a concentration of about 0.01 ng/mL. In certain embodiments, said first medium additionally comprises one or more of the following: antibiotics such as gentamycin; antioxidants such as transferrin, insulin, and/or beta-mercaptoethanol; sodium selenite; ascorbic acid; ethanolamine; and glutathione. In certain embodiments, the medium that provides the base for the first medium is a cell/tissue culture medium known to those of skill in the art, e.g., a commercially available cell/tissue culture medium such as SCGM™, STEMMACS™, GBGM®, AIM-V®, X-VIVO™ 10, X-VIVO™ 15, OPTMIZER, STEMSPAN® H3000, CELLGRO COMPLETE™, DMEM:Ham’s F12 (“F12”) (e.g., 2:1 ratio, or high glucose or low glucose DMEM), Advanced DMEM (Gibco), EL08-1D2, Myelocult™ H5100, IMDM, and/or RPMI- 1640; or is a medium that comprises components generally included in known cell/tissue culture media, such as the components included in GBGM®, AIM-V®, X-VIVO™ 10, X- VIVO™ 15, OPTMIZER, STEMSPAN® H3000, CELLGRO COMPLETE™,
DMEM:Ham’s F12 (“F12”) (e.g., 2:1 ratio, or high glucose or low glucose DMEM), Advanced DMEM (Gibco), EL08-1D2, Myelocult™ H5100, IMDM, and/or RPMI-1640. In certain embodiments, said first medium is not GBGM®. In specific embodiments of any of the above embodiments, the first medium lacks LIF, MIP-1α, or both.
[00119] In certain aspects, said second medium used in the three-stage method comprises a stem cell mobilizing agent and interleukin- 15 (IL-15), and lacks Tpo. In certain aspects, the second medium used in the three-stage method comprises, in addition to a stem cell mobilizing agent and IL-15, one or more of LMWH, Flt-3, SCF, IL-6, IL-7, G-CSF, and GM-CSF. In certain aspects, the second medium used in the three-stage method comprises, in addition to a stem cell mobilizing agent and IL-15, each of LMWH, Flt-3, SCF, IL-6, IL-7, G-CSF, and GM-CSF. In certain aspects, the second medium used in the three-stage method comprises, in addition to a stem cell mobilizing agent and IL-15, each of Flt-3, SCF, IL-6, IL- 7, G-CSF, and GM-CSF. In a specific aspect, the second medium lacks added LMWH. In a specific aspect, the second medium lacks added desulphated glycosaminoglycans. In a specific aspect, the second medium lacks LMWH. In a specific aspect, the second medium lacks desulphated glycosaminoglycans. In certain aspects, said IL-15 is present in said second medium at a concentration of from 1 ng/mL to 50 ng/mL, from 10 ng/mL to 30 ng/mL, or about 20 ng/mL. In certain aspects, when LMWH is present in said second medium, the LMWH is present at a concentration of from lU/mL to lOU/mL; the Flt-3L is present at a concentration of from 1 ng/mL to 50 ng/mL; the SCF is present at a concentration of from 1 ng/mL to 50 ng/mL; the IL-6 is present at a concentration of from 0.01 ng/mL to 0.1 ng/mL; the IL-7 is present at a concentration of from 1 ng/mL to 50 ng/mL; the G-CSF is present at a concentration of from 0.01 ng/mL to 0.50 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.1 ng/mL. In certain aspects, in said second medium, the Flt-3L is present at a concentration of from 1 ng/mL to 50 ng/mL; the SCF is present at a concentration of from 1 ng/mL to 50 ng/mL; the IL-6 is present at a concentration of from 0.01 ng/mL to 0.1 ng/mL; the IL-7 is present at a concentration of from 1 ng/mL to 50 ng/mL; the G-CSF is present at a concentration of from 0.01 ng/mL to 0.50 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.1 ng/mL. In certain aspects, when LMWH is present in the second medium, the LMWH is present in the second medium at a concentration of from 4U/mL to 5U/mL; the Flt-3L is present at a concentration of from 20 ng/mL to 30 ng/mL; the SCF is present at a concentration of from 20 ng/mL to 30 ng/mL; the IL-6 is present at a concentration of from 0.04 ng/mL to 0.06 ng/mL; the IL-7 is present at a concentration of from 20 ng/mL to 30 ng/mL; the G-CSF is present at a concentration of from 0.20 ng/mL to 0.30 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.5 ng/mL. In certain aspects, in the second medium, the Flt-3L is present at a concentration of from 20 ng/mL to 30 ng/mL; the SCF is present at a concentration of from 20 ng/mL to 30 ng/mL; the IL-6 is present at a concentration of from 0.04 ng/mL to 0.06 ng/mL; the IL-7 is present at a concentration of from 20 ng/mL to 30 ng/mL; the G-CSF is present at a concentration of from 0.20 ng/mL to 0.30 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.5 ng/mL. In certain aspects, when LMWH is present in the second medium, the LMWH is present in the second medium at a concentration of from 4U/mL to 5U/mL; the Flt-3L is present at a concentration of from 20 ng/mL to 30 ng/mL; the SCF is present at a concentration of from 20 ng/mL to 30 ng/mL; the IL-6 is present at a concentration of from 0.04 ng/mL to 0.06 ng/mL; the IL-7 is present at a concentration of from 20 ng/mL to 30 ng/mL; the G-CSF is present at a concentration of from 0.20 ng/mL to 0.30 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.5 ng/mL. In certain aspects, in the second medium, the Flt-3L is present at a concentration of from 20 ng/mL to 30 ng/mL; the SCF is present at a concentration of from 20 ng/mL to 30 ng/mL; the IL-6 is present at a concentration of from 0.04 ng/mL to 0.06 ng/mL; the IL-7 is present at a concentration of from 20 ng/mL to 30 ng/mL; the G-CSF is present at a concentration of from 0.20 ng/mL to 0.30 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.5 ng/mL. In certain aspects, when LMWH is present in the second medium, the LMWH is present in the second medium at a concentration of about 4.5U/mL; the Flt-3L is present at a concentration of about 25 ng/mL; the SCF is present at a concentration of about 27 ng/mL; the IL-6 is present at a concentration of about 0.05 ng/mL; the IL-7 is present at a concentration of about 25 ng/mL; the G-CSF is present at a concentration of about 0.25 ng/mL; and the GM-CSF is present at a concentration of about
0.01 ng/mL. In certain aspects, in the second medium, the Flt-3L is present at a concentration of about 25 ng/mL; the SCF is present at a concentration of about 27 ng/mL; the IL-6 is present at a concentration of about 0.05 ng/mL; the IL-7 is present at a concentration of about 25 ng/mL; the G-CSF is present at a concentration of about 0.25 ng/mL; and the GM-CSF is present at a concentration of about 0.01 ng/mL. In certain embodiments, said second medium additionally comprises one or more of the following: antibiotics such as gentamycin; antioxidants such as transferrin, insulin, and/or beta- mercaptoethanol; sodium selenite; ascorbic acid; ethanolamine; and glutathione. In certain embodiments, the medium that provides the base for the second medium is a cell/tissue culture medium known to those of skill in the art, e.g., a commercially available cell/tissue culture medium such as SCGM™, STEMMACS™, GBGM®, AIM-V®, X-VIVO™ 10, X- VIVO™ 15, OPTMIZER, STEMSPAN® H3000, CELLGRO COMPLETE™, DMEM:Ham’s F12 (“F12”) (e.g., 2:1 ratio, or high glucose or low glucose DMEM), Advanced DMEM (Gibco), EL08-1D2, Myelocult™ H5100, IMDM, and/or RPMI-1640; or is a medium that comprises components generally included in known cell/tissue culture media, such as the components included in GBGM®, AIM-V®, X-VIVO™ 10, X-VIVO™ 15, OPTMIZER, STEMSPAN® H3000, CELLGRO COMPLETE™, DMEM:Ham’s F12 (“FI 2”) (e.g., 2:1 ratio, or high glucose or low glucose DMEM), Advanced DMEM (Gibco), EL08-1D2, Myelocult™ H5100, IMDM, and/or RPMI-1640. In certain embodiments, said second medium is not GBGM®. In specific embodiments of any of the above embodiments, the first medium lacks LIF, MIP-1α, or both.
[00120] In certain aspects, said third medium used in the three-stage method comprises IL-2 and IL-15, and lacks a stem cell mobilizing agent and LMWH. In certain aspects, said third medium used in the three-stage method comprises IL-2 and IL-15, and lacks LMWH.
In certain aspects, said third medium used in the three-stage method comprises IL-2 and IL- 15, and lacks SCF and LMWH. In certain aspects, said third medium used in the three-stage method comprises IL-2 and IL-15, and lacks SCF, a stem cell mobilizing agent and LMWH. In certain aspects, said third medium used in the three-stage method comprises a stem cell mobilizing agent, IL-2 and IL-15, and lacks LMWH. In certain aspects, said third medium used in the three-stage method comprises SCF, IL-2 and IL-15, and lacks LMWH. In certain aspects, said third medium used in the three-stage method comprises a stem cell mobilizing agent, SCF, IL-2 and IL-15, and lacks LMWH. In certain aspects, said third medium used in the three-stage method comprises IL-2 and IL-15, and lacks a stem cell mobilizing agent and LMWH. In certain aspects, the third medium used in the three-stage method comprises, in addition to IL-2 and IL-15, one or more of SCF, IL-6, IL-7, G-CSF, or GM-CSF. In certain aspects, the third medium used in the three-stage method comprises, in addition to IL-2 and IL-15, each of SCF, IL-6, IL-7, G-CSF, and GM-CSF. In certain aspects, said IL-2 is present in said third medium at a concentration of from 10 U/mL to 10,000 U/mL and said IL-15 is present in said third medium at a concentration of from 1 ng/mL to 50 ng/mL. In certain aspects, said IL-2 is present in said third medium at a concentration of from 100 U/mL to 10,000 U/mL and said IL-15 is present in said third medium at a concentration of from 1 ng/mL to 50 ng/mL. In certain aspects, said IL-2 is present in said third medium at a concentration of from 300 U/mL to 3,000 U/mL and said IL-15 is present in said third medium at a concentration of from 10 ng/mL to 30 ng/mL. In certain aspects, said IL-2 is present in said third medium at a concentration of about 1,000 U/mL and said IL-15 is present in said third medium at a concentration of about 20 ng/mL. In certain aspects, in said third medium, the SCF is present at a concentration of from 1 ng/mL to 50 ng/mL; the IL-6 is present at a concentration of from 0.01 ng/mL to 0.1 ng/mL; the IL-7 is present at a concentration of from 1 ng/mL to 50 ng/mL; the G-CSF is present at a concentration of from 0.01 ng/mL to 0.50 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.1 ng/mL. In certain aspects, in said third medium, the SCF is present at a concentration of from 20 ng/mL to 30 ng/mL; the IL-6 is present at a concentration of from 0.04 ng/mL to 0.06 ng/mL; the IL-7 is present at a concentration of from 20 ng/mL to 30 ng/mL; the G-CSF is present at a concentration of from 0.20 ng/mL to 0.30 ng/mL; and the GM-CSF is present at a concentration of from 0.005 ng/mL to 0.5 ng/mL. In certain aspects, in said third medium, the SCF is present at a concentration of about 22 ng/mL; the IL-6 is present at a concentration of about 0.05 ng/mL; the IL-7 is present at a concentration of about 20 ng/mL; the G-CSF is present at a concentration of about 0.25 ng/mL; and the GM-CSF is present at a concentration of about 0.01 ng/mL. In certain aspects, the third medium comprises 100 ng/mL IL-7, 1000 ng/mL IL-2, 20 ng/mL IL-15, and 10 stem cell mobilizing agent and lacks SCF. In certain aspects, the third medium comprises 20 ng/mL IL-7, 1000 ng/mL IL-2, 20 ng/mL IL-15, and stem cell mobilizing agent and lacks SCF. In certain aspects, the third medium comprises 20 ng/mL IL-7, 20 ng/mL IL-15, and stem cell mobilizing agent and lacks SCF. In certain aspects, the third medium comprises 100 ng/mL IL-7, 22 ng/mL SCF, 1000 ng/mL IL-2, and 20 ng/mL IL-15 and lacks stem cell mobilizing agent. In certain aspects, the third medium comprises 22 ng/mL SCF, 1000 ng/mL IL-2, and 20 ng/mL IL-15 and lacks stem cell mobilizing agent. In certain aspects, the third medium comprises 20 ng/mL IL-7, 22 ng/mL SCF, 1000 ng/mL IL-2, and 20 ng/mL IL-15 and lacks stem cell mobilizing agent. In certain aspects, the third medium comprises 20 ng/mL IL-7, 22 ng/mL SCF, and 1000 ng/mL IL-2 and lacks stem cell mobilizing agent. In specific embodiments of any of the above embodiments, the first medium lacks one, two, or all three of LIF, MIP-1α, Flt-3L.
[00121] In certain embodiments, said third medium additionally comprises one or more of the following: antibiotics such as gentamycin; antioxidants such as transferrin, insulin, and/or beta-mercaptoethanol; sodium selenite; ascorbic acid; ethanolamine; and glutathione. In certain embodiments, the medium that provides the base for the third medium is a cell/tissue culture medium known to those of skill in the art, e.g., a commercially available cell/tissue culture medium such as SCGM™, STEMMACS™, GBGM®, AIM-V®, X-VIVO™ 10, X-VIVO™ 15, OPTMIZER, STEMSPAN® H3000, CELLGRO COMPLETE™, DMEM:Ham’s F12 (“F12”) (e.g., 2:1 ratio, or high glucose or low glucose DMEM), Advanced DMEM (Gibco), EL08-1D2, Myelocult™ H5100, IMDM, and/or RPMI- 1640; or is a medium that comprises components generally included in known cell/tissue culture media, such as the components included in GBGM®, AIM-V®, X-VIVO™ 10, X- VIVO™ 15, OPTMIZER, STEMSPAN® H3000, CELLGRO COMPLETE™,
DMEM:Ham’s F12 (“F12”) (e.g., 2:1 ratio, or high glucose or low glucose DMEM), Advanced DMEM (Gibco), EL08-1D2, Myelocult™ H5100, IMDM, and/or RPMI-1640. In certain embodiments, said third medium is not GBGM®.
[00122] Generally, the particularly recited medium components do not refer to possible constituents in an undefined component of said medium. For example, said Tpo, IL-2, and IL-15 are not comprised within an undefined component of the first medium, second medium or third medium, e.g., said Tpo, IL-2, and IL-15 are not comprised within serum. Further, said LMWH, Flt-3, SCF, IL-6, IL-7, G-CSF, and/or GM-CSF are not comprised within an undefined component of the first medium, second medium or third medium, e.g., said LMWH, Flt-3, SCF, IL-6, IL-7, G-CSF, and/or GM-CSF are not comprised within serum. [00123] In certain aspects, said first medium, second medium or third medium comprises human serum-AB. In certain aspects, any of said first medium, second medium or third medium comprises 1% to 20% human serum-AB, 5% to 15% human serum-AB, or about 2, 5, or 10% human serum-AB.
[00124] In certain embodiments, in the three-stage methods described herein, said hematopoietic stem or progenitor cells are cultured in said first medium for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 days. In certain embodiments, in the three- stage methods described herein, cells are cultured in said second medium for 1, 2, 3, 4, 5, 6,
7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 days. In certain embodiments, in the three-stage methods described herein, cells are cultured in said third medium for 1, 2, 3, 4, 5,
6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 days, or for more than 30 days.
[00125] In a specific embodiment, in the three-stage methods described herein, said hematopoietic stem or progenitor cells are cultured in said first medium for 7-13 days to produce a first population of cells, before said culturing in said second medium; said first population of cells are cultured in said second medium for 2-6 days to produce a second population of cells before said culturing in said third medium; and said second population of cells are cultured in said third medium for 10-30 days, i.e., the cells are cultured a total of 19- 49 days.
[00126] In a specific embodiment, in the three-stage methods described herein, in the three-stage methods described herein, said hematopoietic stem or progenitor cells are cultured in said first medium for 8-12 days to produce a first population of cells, before said culturing in said second medium; said first population of cells are cultured in said second medium for 3-5 days to produce a second population of cells before said culturing in said third medium; and said second population of cells are cultured in said third medium for 15-25 days, i.e., the cells are cultured a total of 26-42 days.
[00127] In a specific embodiment, in the three-stage methods described herein, said hematopoietic stem or progenitor cells are cultured in said first medium for about 10 days to produce a first population of cells, before said culturing in said second medium; said first population of cells are cultured in said second medium for about 4 days to produce a second population of cells before said culturing in said third medium; and said second population of cells are cultured in said third medium for about 21 days, i.e., the cells are cultured a total of about 35 days.
[00128] In certain aspects, the three-stage method disclosed herein produces at least 5000-fold more natural killer cells as compared to the number of hematopoietic stem cells initially inoculated into said first medium. In certain aspects, said three-stage method produces at least 10,000-fold more natural killer cells as compared to the number of hematopoietic stem cells initially inoculated into said first medium. In certain aspects, said three-stage method produces at least 50,000-fold more natural killer cells as compared to the number of hematopoietic stem cells initially inoculated into said first medium. In certain aspects, said three-stage method produces at least 75,000-fold more natural killer cells as compared to the number of hematopoietic stem cells initially inoculated into said first medium. In certain aspects, the viability of said natural killer cells is determined by 7- aminoactinomycin D (7AAD) staining. In certain aspects, the viability of said natural killer cells is determined by annexin-V staining. In specific aspects, the viability of said natural killer cells is determined by both 7-AAD staining and annexin-V staining. In certain aspects, the viability of said natural killer cells is determined by trypan blue staining.
[00129] In certain aspects, the three-stage method disclosed herein produces at least 5000-fold more ILC3 cells as compared to the number of hematopoietic stem cells initially inoculated into said first medium. In certain aspects, said three-stage method produces at least 10,000-fold more ILC3 cells as compared to the number of hematopoietic stem cells initially inoculated into said first medium. In certain aspects, said three-stage method produces at least 50,000-fold more ILC3 cells as compared to the number of hematopoietic stem cells initially inoculated into said first medium. In certain aspects, said three-stage method produces at least 75,000-fold more ILC3 cells as compared to the number of hematopoietic stem cells initially inoculated into said first medium.
[00130] In certain aspects, the three-stage method produces natural killer cells that comprise at least 20% CD56+CD3- natural killer cells. In certain aspects, the three-stage method produces natural killer cells that comprise at least 40% CD56+CD3- natural killer cells. In certain aspects, the three-stage method produces natural killer cells that comprise at least 60% CD56+CD3- natural killer cells. In certain aspects, the three-stage method produces natural killer cells that comprise at least 70% CD56+CD3- natural killer cells. In certain aspects, the three-stage method produces natural killer cells that comprise at least 80% CD56+CD3- natural killer cells.
[00131] In certain aspects, the three-stage method disclosed herein produces natural killer cells that comprise at least 20% CD56+CD3-CDlla+ natural killer cells. In certain aspects, the three-stage method disclosed herein produces natural killer cells that comprise at least 40% CD56+CD3- CD11a+ natural killer cells. In certain aspects, the three-stage method disclosed herein produces natural killer cells that comprise at least 60% CD56+CD3- CDlla+ natural killer cells. In certain aspects, the three-stage method disclosed herein produces natural killer cells that comprise at least 80% CD56+CD3- CDlla+ natural killer cells.
[00132] In certain aspects, the three-stage method disclosed herein produces ILC3 cells that comprise at least 20% CD56+CD3- CD11a- ILC3 cells. In certain aspects, the three- stage method disclosed herein produces ILC3 cells that comprise at least 40% CD56+CD3- CDlla- ILC3 cells. In certain aspects, the three-stage method disclosed herein produces
ILC3 cells that comprise at least 60% CD56+CD3- CDlla- ILC3 cells. In certain aspects, the three-stage method disclosed herein produces natural killer cells that comprise at least 80% CD56+CD3- CD11a- ILC3 cells.
[00133] In certain aspects, the three-stage method produces natural killer cells that exhibit at least 20% cytotoxicity against K562 cells when said natural killer cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10:1. In certain aspects, the three- stage method produces natural killer cells that exhibit at least 35% cytotoxicity against the K562 cells when said natural killer cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10: 1. In certain aspects, the three-stage method produces natural killer cells that exhibit at least 45% cytotoxicity against the K562 cells when said natural killer cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10: 1. In certain aspects, the three-stage method produces natural killer cells that exhibit at least 60% cytotoxicity against the K562 cells when said natural killer cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10: 1. In certain aspects, the three-stage method produces natural killer cells that exhibit at least 75% cytotoxicity against the K562 cells when said natural killer cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10: 1.
[00134] In certain aspects, the three-stage method produces ILC3 cells that exhibit at least 20% cytotoxicity against K562 cells when said ILC3 cells and said K562 cells are co- cultured in vitro or ex vivo at a ratio of 10: 1. In certain aspects, the three-stage method produces ILC3 cells that exhibit at least 35% cytotoxicity against the K562 cells when said ILC3 cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10:1. In certain aspects, the three-stage method produces ILC3 cells that exhibit at least 45% cytotoxicity against the K562 cells when said ILC3 cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10: 1. In certain aspects, the three-stage method produces ILC3 cells that exhibit at least 60% cytotoxicity against the K562 cells when said ILC3 cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10:1. In certain aspects, the three-stage method produces ILC3 cells that exhibit at least 75% cytotoxicity against the K562 cells when said ILC3 cells and said K562 cells are co-cultured in vitro or ex vivo at a ratio of 10: 1. [00135] In certain aspects, after said third culturing step, said third population of cells, e.g., said population of natural killer cells and/or ILC3 cells, is cryopreserved. In certain aspects, after said fourth step, said fourth population of cells, e.g., said population of natural killer cells and/or ILC3 cells, is cryopreserved.
[00136] In certain aspects, provided herein are populations of cells comprising natural killer cells, i.e., natural killers cells produced by a three-stage method described herein.
Accordingly, provided herein is an isolated natural killer cell population produced by a three- stage method described herein. In a specific embodiment, said natural killer cell population comprises at least 20% CD56+CD3- natural killer cells. In a specific embodiment, said natural killer cell population comprises at least 40% CD56+CD3- natural killer cells. In a specific embodiment, said natural killer cell population comprises at least 60% CD56+CD3- natural killer cells. In a specific embodiment, said natural killer cell population comprises at least 80% CD56+CD3- natural killer cells. In a specific embodiment, said natural killer cell population comprises at least 60% CD16- cells. In a specific embodiment, said natural killer cell population comprises at least 80% CD16- cells. In a specific embodiment, said natural killer cell population comprises at least 20% CD94+ cells. In a specific embodiment, said natural killer cell population comprises at least 40% CD94+ cells.
[00137] In certain aspects, provided herein is a population of natural killer cells that is CD56+CD3- CD117+CDl la+, wherein said natural killer cells express perforin and/or EOMES, and do not express one or more of RORyt, aryl hydrocarbon receptor (AHR), and IL1R1. In certain aspects, said natural killer cells express perforin and EOMES, and do not express any of RORyt, aryl hydrocarbon receptor, or IL1R1. In certain aspects, said natural killer cells additionally express T-bet, GZMB, NKp46, NKp30, and NKG2D. In certain aspects, said natural killer cells express CD94. In certain aspects, said natural killer cells do not express CD94.
[00138] In certain aspects, provided herein is a population of ILC3 cells that is CD56+CD3- CD117+CDl la-, wherein said ILC3 cells express one or more of RORyt, aryl hydrocarbon receptor, and IL1R1, and do not express one or more of CD94, perforin, and EOMES. In certain aspects, said ILC3 cells express RORyt, aryl hydrocarbon receptor, and IL1R1, and do not express any of CD94, perforin, or EOMES. In certain aspects, said ILC3 cells additionally express CD226 and/or 2B4. In certain aspects, said ILC3 cells additionally express one or more of IL-22, TNFa, and DNAM-1. In certain aspects, said ILC3 cells express CD226, 2B4, IL-22, TNFa, and DNAM-1.
[00139] In certain aspects, provided herein is a method of producing a cell population comprising natural killer cells and ILC3 cells, comprising (a) culturing hematopoietic stem or progenitor cells in a first medium comprising a stem cell mobilizing agent and thrombopoietin (Tpo) to produce a first population of cells; (b) culturing the first population of cells in a second medium comprising a stem cell mobilizing agent and interleukin- 15 (IL- 15), and lacking Tpo, to produce a second population of cells; (c) culturing the second population of cells in a third medium comprising IL-2 and IL-15, and lacking each of a stem cell mobilizing agent and LMWH, to produce a third population of cells; and (d) separating CD11a+ cells and CD11a- cells from the third population of cells; and (e) combining the CDlla+ cells with the CDlla- cells in a ratio of 50:1, 40:1, 30:1, 20:1, 10:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:10, 1:20, 1:30, 1:40, or 1:50 to produce a fourth population of cells. In certain embodiments, said first medium and/or said second medium lack leukemia inhibiting factor (LIF) and/or macrophage inflammatory protein-1 alpha (MIP-1α). In certain embodiments, said third medium lacks LIF, MIP-1α, and FMS-like tyrosine kinase-3 ligand (Flt-3L). In specific embodiments, said first medium and said second medium lack LIF and MIP-1α, and said third medium lacks LIF, MIP-1α, and Flt3L. In certain embodiments, none of the first medium, second medium or third medium comprises heparin, e.g., low-molecular weight heparin. In certain aspects, in the fourth population of cells, the CD11a+ cells and CDlla- cells are combined in a ratio of 50:1. In certain aspects, in the fourth population of cells, the CD11a+ cells and CD11a- cells are combined in a ratio of 20: 1. In certain aspects, in the fourth population of cells, the CD11a+ cells and CD11a- cells are combined in a ratio of 10: 1. In certain aspects, in the fourth population of cells, the CD11a+ cells and CD11a- cells are combined in a ratio of 5: 1. In certain aspects, in the fourth population of cells, the CD11a+ cells and CD 11a- cells are combined in a ratio of 1 : 1. In certain aspects, in the fourth population of cells, the CD11a+ cells and CD11a- cells are combined in a ratio of 1 :5. In certain aspects, in the fourth population of cells, the CD11a+ cells and CD11a- cells are combined in a ratio of 1 : 10. In certain aspects, in the fourth population of cells, the CD11 a+ cells and CD11a- cells are combined in a ratio of 1 :20. In certain aspects, in the fourth population of cells, the CDlla+ cells and CDlla- cells are combined in aratio of 1:50.
5.3. Stem Cell Mobilizing Factors
5.3.1. Chemistry definitions
[00140] To facilitate understanding of the disclosure of stem cell mobilizing factors set forth herein, a number of terms are defined below.
[00141] Generally, the nomenclature used herein and the laboratory procedures in biology, cellular biology, biochemistry, organic chemistry, medicinal chemistry, and pharmacology described herein are those well known and commonly employed in the art. Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs.
[00142] The term “about” or “approximately” means an acceptable error for a particular value as determined by one of ordinary skill in the art, which depends in part on how the value is measured or determined. In certain embodiments, the term “about” or “approximately” means within 1, 2, 3, or 4 standard deviations. In certain embodiments, the term “about” or “approximately” means within 50%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.05% of a given value or range.
[00143] As used herein, any "R" group(s) such as, without limitation, Ra, Rb, Rc, Rd,
Re, Rf Rg, Rh, Rm, Rg, Rj, Rk, RU, RV Ry, and RZ represent substituents that can be attached to the indicated atom. An R group may be substituted or unsubstituted. If two "R" groups are described as being "taken together" the R groups and the atoms they are attached to can form a cycloalkyl, cycloalkenyl, aryl, heteroaryl or heterocycle. For example, without limitation, if Ra and Rb of an NRaRb group are indicated to be "taken together," it means that they are covalently bonded to one another to form a ring:
Figure imgf000038_0001
[00145] In addition, if two “R” groups are described as being “taken together” with the atom(s) to which they are attached to form a ring as an alternative, the R groups are not limited to the variables or substituents defined previously.
[00146] Whenever a group is described as being “optionally substituted” that group may be unsubstituted or substituted with one or more of the indicated substituents. Likewise, when a group is described as being “unsubstituted or substituted” if substituted, the substituent(s) may be selected from one or more the indicated substituents. If no substituents are indicated, it is meant that the indicated “optionally substituted” or “substituted” group may be substituted with one or more group(s) individually and independently selected from alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, acylalkyl, hydroxy, alkoxy, alkoxyalkyl, aminoalkyl, amino acid, aryl, heteroaryl, heterocyclyl, aryl(alkyl), heteroaryl(alkyl), heterocyclyl(alkyl), hydroxyalkyl, acyl, cyano, halogen, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, C-carboxy, O-carboxy, isocyanato, thiocyanato, isothiocyanato, azido, nitro, silyl, sulfenyl, sulfmyl, sulfonyl, haloalkyl, haloalkoxy, trihalomethanesulfonyl, trihalomethanesulfonamido, an amino, a mono-substituted amino group and a di-substituted amino group.
[00147] As used herein, “Ca to Cb” in which “a” and “b” are integers refer to the number of carbon atoms in an alkyl, alkenyl or alkynyl group, or the number of carbon atoms in the ring of a cycloalkyl, cycloalkenyl, aryl, heteroaryl or heteroalicyclyl group. That is, the alkyl, alkenyl, alkynyl, ring(s) of the cycloalkyl, ring(s) of the cycloalkenyl, ring(s) of the aryl, ring(s) of the heteroaryl or ring(s) of the heteroalicyclyl can contain from “a” to “b”, inclusive, carbon atoms. Thus, for example, a “C1 to C4 alkyl” group refers to all alkyl groups having from 1 to 4 carbons, that is, CH3-, CH3CH2-, CH3CH2CH2-, ( CH3)2CH-, CH3CH2CH2CH2-, CH3CH2CH(CH3)- and (CH3)3C -. If no “a” and “b” are designated with regard to an alkyl, alkenyl, alkynyl, cycloalkyl cycloalkenyl, aryl, heteroaryl or heteroalicyclyl group, the broadest range described in these definitions is to be assumed. [00148] As used herein, “alkyl” refers to a straight or branched hydrocarbon chain that comprises a fully saturated (no double or triple bonds) hydrocarbon group. The alkyl group may have 1 to 20 carbon atoms (whenever it appears herein, a numerical range such as “1 to 20” refers to each integer in the given range; e.g., “1 to 20 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 20 carbon atoms, although the present definition also covers the occurrence of the term “alkyl” where no numerical range is designated). The alkyl group may also be a medium size alkyl having 1 to 10 carbon atoms. The alkyl group could also be a lower alkyl having 1 to 6 carbon atoms. The alkyl group of the compounds may be designated as “C1-C4 alkyl” or similar designations. By way of example only, “C1-C4 alkyl” indicates that there are one to four carbon atoms in the alkyl chain, i.e., the alkyl chain is selected from methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, and t-butyl. Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl and hexyl. The alkyl group may be substituted or unsubstituted.
[00149] As used herein, “alkenyl” refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more double bonds. Examples of alkenyl groups include allenyl, vinylmethyl and ethenyl. An alkenyl group may be unsubstituted or substituted.
[00150] As used herein, “alkynyl” refers to an alkyl group that contains in the straight or branched hydrocarbon chain one or more triple bonds. Examples of alkynyls include ethynyl and propynyl. An alkynyl group may be unsubstituted or substituted.
[00151] As used herein, “cycloalkyl” refers to a completely saturated (no double or triple bonds) mono- or multi- cyclic hydrocarbon ring system. When composed of two or more rings, the rings may be joined together in a fused fashion. Cycloalkyl groups can contain 3 to 10 atoms in the ring(s) or 3 to 8 atoms in the ring(s). A cycloalkyl group may be unsubstituted or substituted. Typical cycloalkyl groups include, but are in no way limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and cyclooctyl. [00152] As used herein, “cycloalkenyl” refers to a mono- or multi- cyclic hydrocarbon ring system that contains one or more double bonds in at least one ring; although, if there is more than one, the double bonds cannot form a fully delocalized pi-electron system throughout all the rings (otherwise the group would be “aryl,” as defined herein). Cycloalkenyl groups can contain 3 to 10 atoms in the ring(s) or 3 to 8 atoms in the ring(s). When composed of two or more rings, the rings may be connected together in a fused fashion. A cycloalkenyl group may be unsubstituted or substituted.
[00153] As used herein, “aryl” refers to a carbocyclic (all carbon) monocyclic or multicyclic aromatic ring system (including fused ring systems where two carbocyclic rings share a chemical bond) that has a fully delocalized pi-electron system throughout all the rings. The number of carbon atoms in an aryl group can vary. For example, the aryl group can be a C6-C14 aryl group, a C6-C10 aryl group, or a C6 aryl group. Examples of aryl groups include, but are not limited to, benzene, naphthalene and azulene. An aryl group may be substituted or unsubstituted.
[00154] As used herein, “heteroaryl” refers to a monocyclic or multicyclic aromatic ring system (a ring system with fully delocalized pi-electron system) that contain(s) one, two, three or more heteroatoms, that is, an element other than carbon, including but not limited to, nitrogen, oxygen and sulfur. The number of atoms in the ring(s) of a heteroaryl group can vary. For example, the heteroaryl group can contain 4 to 14 atoms in the ring(s), 5 to 10 atoms in the ring(s) or 5 to 6 atoms in the ring(s). Furthermore, the term “heteroaryl” includes fused ring systems where two rings, such as at least one aryl ring and at least one heteroaryl ring, or at least two heteroaryl rings, share at least one chemical bond. Examples of heteroaryl rings include, but are not limited to, those described herein and the following: furan, furazan, thiophene, benzothiophene, phthalazine, pyrrole, oxazole, benzoxazole, 1,2,3- oxadiazole, 1,2,4-oxadiazole, thiazole, 1,2,3-thiadiazole, 1,2,4-thiadiazole, benzothiazole, imidazole, benzimidazole, indole, indazole, pyrazole, benzopyrazole, isoxazole, benzoisoxazole, isothiazole, triazole, benzotriazole, thiadiazole, tetrazole, pyridine, pyridazine, pyrimidine, pyrazine, purine, pteridine, quinoline, isoquinoline, quinazoline, quinoxaline, cinnoline and triazine. A heteroaryl group may be substituted or unsubstituted. [00155] As used herein, “heterocyclyl” or “heteroalicyclyl” refers to three-, four-, five-, six-, seven-, eight-, nine-, ten-, up to 18-membered monocyclic, bicyclic, and tricyclic ring system wherein carbon atoms together with from 1 to 5 heteroatoms constitute said ring system. A heterocycle may optionally contain one or more unsaturated bonds situated in such a way, however, that a fully delocalized pi-electron system does not occur throughout all the rings. The heteroatom(s) is an element other than carbon including, but not limited to, oxygen, sulfur, and nitrogen. A heterocycle may further contain one or more carbonyl or thiocarbonyl functionalities, so as to make the definition include oxo-systems and thio- systems such as lactams, lactones, cyclic imides, cyclic thioimides and cyclic carbamates. When composed of two or more rings, the rings may be joined together in a fused fashion. Additionally, any nitrogens in a heterocyclyl may be quatemized. Heterocyclyl or heteroalicyclic groups may be unsubstituted or substituted. Examples of such “heterocyclyl” or “heteroalicyclyl” groups include, but are not limited to, those described herein and the following: 1,3-dioxin, 1,3-dioxane, 1,4-dioxane, 1,2-dioxolane, 1,3-dioxolane, 1,4- dioxolane, 1,3-oxathiane, 1,4-oxathiin, 1,3-oxathiolane, 1,3-dithiole, 1,3-dithiolane, 1,4- oxathiane, tetrahydro-l,4-thiazine, 1,3-thiazinane, 2H-l,2-oxazine, maleimide, succinimide, barbituric acid, thiobarbituric acid, dioxopiperazine, hydantoin, dihydrouracil, trioxane, hexahydro-l,3,5-triazine, imidazoline, imidazolidine, isoxazoline, isoxazolidine, oxazoline, oxazolidine, oxazolidinone, thiazoline, thiazolidine, morpholine, oxirane, piperidine N-Oxide, piperidine, piperazine, pyrrolidine, pyrrolidone, pyrrolidione, 4-piperidone, pyrazoline, pyrazolidine, 2-oxopyrrolidine, tetrahydropyran, 4H-pyran, tetrahydrothiopyran, thiamorpholine, thiamorpholine sulfoxide, thiamorpholine sulfone, and their benzo-fused analogs (e.g., benzimidazolidinone, tetrahydroquinoline, and 3,4-methylenedioxyphenyl). [00156] As used herein, “aralkyl” and “aryl(alkyl)” refer to an aryl group connected, as a substituent, via a lower alkylene group. The lower alkylene and aryl group of an aralkyl may be substituted or unsubstituted. Examples include but are not limited to benzyl, 2- phenylalkyl, 3-phenylalkyl and naphthylalkyl.
[00157] As used herein, “heteroaralkyl” and “heteroaryl(alkyl)” refer to a heteroaryl group connected, as a substituent, via a lower alkylene group. The lower alkylene and heteroaryl group of heteroaralkyl may be substituted or unsubstituted. Examples include but are not limited to 2-thienylalkyl, 3-thienylalkyl, furylalkyl, thienylalkyl, pyrrolylalkyl, pyridylalkyl, isoxazolylalkyl, imidazolylalkyl and their benzo-fused analogs.
[00158] A “heteroalicyclyl(alkyl)” and “heterocyclyl(alkyl)” refer to a heterocyclic or a heteroalicyclylic group connected, as a substituent, via a lower alkylene group. The lower alkylene and heterocyclyl of a heteroalicyclyl(alkyl) may be substituted or unsubstituted. Examples include but are not limited tetrahydro-2H-pyran-4-yl(methyl), piperidin-4- yl(ethyl), piperidin-4-yl(propyl), tetrahydro-2H-thiopyran-4-yl(methyl), and l,3-thiazinan-4- yl(methyl). [00159] “Lower alkylene groups” are straight-chained -CH2- tethering groups, forming bonds to connect molecular fragments via their terminal carbon atoms. Examples include but are not limited to methylene (-CH2-), ethylene (-CH2CH2-), propylene (-CH2CH2CH2-), and butylene (-CH2CH2CH2CH2-). A lower alkylene group can be substituted by replacing one or more hydrogen of the lower alkylene group with a substituent(s) listed under the definition of “substituted.”
[00160] As used herein, “alkoxy” refers to the formula -OR wherein R is an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl) is defined herein. A non-limiting list of alkoxys are methoxy, ethoxy, n-propoxy, 1-methylethoxy (isopropoxy), n- butoxy, iso-butoxy, sec-butoxy, tert-butoxy, phenoxy and benzoxy. An alkoxy may be substituted or unsubstituted.
[00161] As used herein, “acyl” refers to a hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl) connected, as substituents, via a carbonyl group. Examples include formyl, acetyl, propanoyl, benzoyl and acryl. An acyl may be substituted or unsubstituted.
[00162] As used herein, “acylalkyl” refers to an acyl connected, as a substituent, via a lower alkylene group. Examples include aryl-C(=O)-(CH2)n- and heteroaryl-C(=O)-(CH2)n-, where n is an integer in the range of 1 to 6.
[00163] As used herein, “alkoxyalkyl” refers to an alkoxy group connected, as a substituent, via a lower alkylene group. Examples include C1-4 alkyl-O-(CH2)n- , wherein n is an integer in the range of 1 to 6.
[00164] As used herein, “aminoalkyl” refers to an optionally substituted amino group connected, as a substituent, via a lower alkylene group. Examples include H2N(CH2)n- , wherein n is an integer in the range of 1 to 6.
[00165] As used herein, “hydroxyalkyl” refers to an alkyl group in which one or more of the hydrogen atoms are replaced by a hydroxy group. Exemplary hydroxyalkyl groups include but are not limited to, 2-hydroxy ethyl, 3-hydroxypropyl, 2-hydroxypropyl, and 2,2- dihydroxyethyl. A hydroxyalkyl may be substituted or unsubstituted.
[00166] As used herein, “haloalkyl” refers to an alkyl group in which one or more of the hydrogen atoms are replaced by a halogen (e.g., mono-haloalkyl, di-haloalkyl and tri- haloalkyl). Such groups include but are not limited to, chloromethyl, fluoromethyl, difluoromethyl, trifluoromethyl, chloro-fluoroalkyl, chloro-difluoroalkyl and 2- fluoroisobutyl. A haloalkyl may be substituted or unsubstituted.
[00167] As used herein, “haloalkoxy” refers to an alkoxy group in which one or more of the hydrogen atoms are replaced by a halogen (e.g., mono-haloalkoxy, di- haloalkoxy and tri- haloalkoxy). Such groups include but are not limited to, chloromethoxy, fluoromethoxy, difluoromethoxy, trifluoromethoxy, chloro-fluoroalkyl, chloro-difluoroalkoxy and 2- fluoroisobutoxy. A haloalkoxy may be substituted or unsubstituted.
[00168] A “sulfenyl” group refers to an “-SR” group in which R can be hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). A sulfenyl may be substituted or unsubstituted.
[00169] A “sulfmyl” group refers to an “-S(=O)-R” group in which R can be the same as defined with respect to sulfenyl. A sulfmyl may be substituted or unsubstituted.
[00170] A “sulfonyl” group refers to an “SO2R” group in which R can be the same as defined with respect to sulfenyl. A sulfonyl may be substituted or unsubstituted.
[00171] An “O-carboxy” group refers to a “RC(=O)O-” group in which R can be hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl), as defined herein. An O-carboxy may be substituted or unsubstituted.
[00172] The terms “ester” and “C-carboxy” refer to a “-C(=O)OR” group in which R can be the same as defined with respect to O-carboxy. An ester and C-carboxy may be substituted or unsubstituted.
[00173] A “thiocarbonyl” group refers to a “-C(=S)R” group in which R can be the same as defined with respect to O-carboxy. A thiocarbonyl may be substituted or unsubstituted.
[00174] A “trihalomethanesulfonyl” group refers to an “X3CSO2-” group wherein each X is a halogen.
[00175] A “trihalomethanesulfonamido” group refers to an “X3CS(O)2N(RA)-” group wherein each X is a halogen, and RA hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl).
[00176] The term “amino” as used herein refers to a -NT2 group.
[00177] As used herein, the term “hydroxy” refers to a -OH group.
[00178] A “cyano” group refers to a “-CN” group. [00179] The term “azido” as used herein refers to a -N3 group.
[00180] An “isocyanato” group refers to a “-NCO” group.
[00181] A “thiocyanato” group refers to a “-CNS” group.
[00182] An “isothiocyanato” group refers to an “ -NCS” group.
[00183] A “carbonyl” group refers to a C=O group.
[00184] An “S-sulfonamido” group refers to a “-SO2N(RARB)” group in which RA and RB can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An S-sulfonamido may be substituted or unsubstituted.
[00185] An “N-sulfonamido” group refers to a “RSO2N(RA)-” group in which R and RA can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An N-sulfonamido may be substituted or unsubstituted.
[00186] An “O-carbamyl” group refers to a “-OC(=O)N(RARB)” group in which RA and RB can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An O-carbamyl may be substituted or unsubstituted.
[00187] An “N-carbamyl” group refers to an “ROC(=O)N(RA)-” group in which R and RA can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An N-carbamyl may be substituted or unsubstituted.
[00188] An “O-thiocarbamyl” group refers to a “-OC(=S)-N(RARB)” group in which RA and RB can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An O-thiocarbamyl may be substituted or unsubstituted.
[00189] An “N-thiocarbamyl” group refers to an “ROC(=S)N(RA)-” group in which R and RA can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An N-thiocarbamyl may be substituted or unsubstituted.
[00190] A “C-amido” group refers to a “-C(=O)N(RARB)” group in which RA and RB can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). A C-amido may be substituted or unsubstituted. [00191] An “N-amido” group refers to a “RC(=O)N(RA)-” group in which R and RA can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). An N-amido may be substituted or unsubstituted.
[00192] A “urea” group refers to “N(R)-C(=O)-NRARB group in which R can be hydrogen or an alkyl, and RA and RB can be independently hydrogen, an alkyl, an alkenyl, an alkynyl, a cycloalkyl, a cycloalkenyl, aryl, heteroaryl, heterocyclyl, cycloalkyl(alkyl), aryl(alkyl), heteroaryl(alkyl) or heterocyclyl(alkyl). A urea may be substituted or unsubstituted.
[00193] The term “halogen atom” or “halogen” as used herein, means any one of the radio-stable atoms of column 7 of the Periodic Table of the Elements, such as, fluorine, chlorine, bromine and iodine.
[00194] As used herein, indicates a single or double bond, unless stated
Figure imgf000045_0001
otherwise.
[00195] Where the numbers of substituents is not specified (e.g. haloalkyl), there may be one or more substituents present. For example “haloalkyl” may include one or more of the same or different halogens. As another example, “C1-C3 alkoxyphenyl” may include one or more of the same or different alkoxy groups containing one, two or three atoms.
[00196] As used herein, the abbreviations for any protective groups, amino acids and other compounds, are, unless indicated otherwise, in accord with their common usage, recognized abbreviations, or the IUPAC-IUB Commission on Biochemical Nomenclature (See, Biochem. 11:942-944 (1972)).
[00197] In certain embodiments, “optically active” and “enantiomerically active” refer to a collection of molecules, which has an enantiomeric excess of no less than about 50%, no less than about 70%, no less than about 80%, no less than about 90%, no less than about 91%, no less than about 92%, no less than about 93%, no less than about 94%, no less than about 95%, no less than about 96%, no less than about 97%, no less than about 98%, no less than about 99%, no less than about 99.5%, or no less than about 99.8%. In certain embodiments, the compound comprises about 95% or more of the desired enantiomer and about 5% or less of the less preferred enantiomer based on the total weight of the two enantiomers in question. [00198] In describing an optically active compound, the prefixes R and S are used to denote the absolute configuration of the optically active compound about its chiral center(s). The (+) and (-) are used to denote the optical rotation of an optically active compound, that is, the direction in which a plane of polarized light is rotated by the optically active compound. The (-) prefix indicates that an optically active compound is levorotatory, that is, the compound rotates the plane of polarized light to the left or counterclockwise. The (+) prefix indicates that an optically active compound is dextrorotatory, that is, the compound rotates the plane of polarized light to the right or clockwise. However, the sign of optical rotation, (+) and (-), is not related to the absolute configuration of a compound, R and S.
[00199] The term “isotopic variant” refers to a compound that contains an unnatural proportion of an isotope at one or more of the atoms that constitute such a compound. In certain embodiments, an “isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, hydrogen (1H), deuterium (2H), tritium (3H), carbon-11 (11C), carbon-12 (12C), carbon-13 (13C), carbon-14 (14C), nitrogen-13 (13N), nitrogen-14 (14N), nitrogen-15 (15N), oxygen-14 (14O), oxygen-15 (15O), oxygen-16 (16O), oxygen-17 (17O), oxygen-18 (18O), fluorine-17 (17F), fluorine-18 (18F), phosphorus-31 (31P), phosphorus-32 (32P), phosphorus-33 (33P), sulfur-32 (32S), sulfur-33 (33S), sulfur-34 (34S), sulfur-35 (35S), sulfur-36 (36S), chlorine-35 (35C1), chlorine-36 (36C1), chlorine-37 (37C1), bromine-79 (79Br), bromine-81 (81Br), iodine-123 (123I), iodine-125 (125I), iodine-127 (127I), iodine-129 (129I), and iodine-131 (131I). In certain embodiments, an “isotopic variant” of a compound is in a stable form, that is, non-radioactive. In certain embodiments, an “isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, hydrogen (1H), deuterium (2H), carbon- 12 (12C), carbon- 13 (13C), nitrogen- 14 (14N), nitrogen-15 (15N), oxygen-16 (16O), oxygen-17 (17O), oxygen-18 (18O), fluorine-17 (17F), phosphorus-31 (31P), sulfur-32 (32S), sulfur-33 (33S), sulfur-34 (34S), sulfur-36 (36S), chlorine-35 (35C1), chlorine-37 (37C1), bromine-79 (79Br), bromine-81 (81Br), and iodine-127 (127I). In certain embodiments, an “isotopic variant” of a compound is in an unstable form, that is, radioactive. In certain embodiments, an “isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, tritium (3H), carbon-11 (11C), carbon-14 (14C), nitrogen-13 (13N), oxygen-14 (14O), oxygen-15 (15O), fluorine-18 (18F), phosphorus-32 (32P), phosphorus-33 (33P), sulfur-35 (35S), chlorine-36 (36C1), iodine-123 (123I), iodine-125 (125I), iodine-129 (129I), and iodine-131 (131I). It will be understood that, in a compound as provided herein, any hydrogen can be 2H, for example, or any carbon can be 13C, for example, or any nitrogen can be 15N, for example, or any oxygen can be 180, for example, where feasible according to the judgment of one of skill. In certain embodiments, an “isotopic variant” of a compound contains unnatural proportions of deuterium (D). [00200] The term “solvate” refers to a complex or aggregate formed by one or more molecules of a solute, e.g., a compound provided herein, and one or more molecules of a solvent, which present in a stoichiometric or non-stoichiometric amount. Suitable solvents include, but are not limited to, water, methanol, ethanol, «-propanol, isopropanol, and acetic acid. In certain embodiments, the solvent is pharmaceutically acceptable. In one embodiment, the complex or aggregate is in a crystalline form. In another embodiment, the complex or aggregate is in a noncry stalline form. Where the solvent is water, the solvate is a hydrate. Examples of hydrates include, but are not limited to, a hemihydrate, monohydrate, dihydrate, trihydrate, tetrahydrate, and pentahydrate.
[00201] The phrase “an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, hydrate, or prodrug thereof’ has the same meaning as the phrase “(i) an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant of the compound referenced therein; (ii) a pharmaceutically acceptable salt, solvate, hydrate, or prodrug of the compound referenced therein; or (iii) a pharmaceutically acceptable salt, solvate, hydrate, or prodrug of an enantiomer, a mixture of enantiomers, a mixture of two or more diastereomers, or an isotopic variant of the compound referenced therein.”
5.3.2. Stem cell mobilizing compounds
[00202] In certain aspects, the stem cell mobilizing factor is a compound having Formula (I), (I- A), (I-B), (I-C), or (I-D), as described below.
Formula (I)
[00203] Some embodiments disclosed herein relate to a compound of Formula (I), or a pharmaceutically acceptable salt thereof, having the structure:
Figure imgf000047_0001
wherein: each can independently represent a single bond or a double bond; RJ can be
Figure imgf000047_0002
selected from the group consisting of-NRaRb, -ORb, and =O; wherein if RJ is =O, then - joining G and J represents a single bond and G is N and the N is substituted with RG; otherwise joining G and J represents a double bond and G is N; Ra can be hydrogen or C1-C4 alkyl; Rb can be Rc or -(C1-C4 alkyl)-Rc; Rc can be selected from the group consisting of: -OH, -O(C1-C4 alkyl), -O(C1-C4 haloalkyl); -C(=O)NH2; unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a Rc moiety indicated as substituted can be substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C1-C4 alkyl, C1-C4 haloalkyl, -O(C1-C4 alkyl), and -O(C1-C4 haloalkyl); RK can be selected from the group consisting of: hydrogen, unsubstituted C1-6 alkyl; substituted C1-6 alkyl; -NH(C1-4 alkyl); - N(C1-4 alkyl)2, unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted can be substituted with one or more substituents Q, wherein each Q is independently selected from the group consisting of: -OH, C1-4 alkyl, C1-4 haloalkyl, halo, cyano, -O-(C1-4 alkyl), and -O-(C1-4 haloalkyl); RG can be selected from the group consisting of hydrogen, C1-4 alkyl, and -(C1-4 alkyl)-C(=O)NH2; RY and RZ can each independently be absent or be selected from the group consisting of: hydrogen, halo, C1-6 alkyl, -OH, -O-(C1-4 alkyl), -NH(C1-4 alkyl), and -N(C1-4 alky 1)2; or RY and RZ taken together with the atoms to which they are attached can joined together to form a ring selected from:
Figure imgf000048_0001
said ring can be optionally substituted with one, two, or three groups independently selected from C1-4 alkyl, C1-4 haloalkyl, halo, cyano, -OH, -O-(C1-4 alkyl), -N(C1-4 alkyl)2, unsubstituted C6-C10 aryl, C6-C10 aryl substituted with 1-5 halo atoms, and -O-(C1-4 haloalkyl); and wherein if RY and RZ taken together forms
Figure imgf000048_0002
then RJ can be -ORb or =O; Rd can be hydrogen or C1-C4 alkyl; Rm can be selected from the group consisting of C1-4 alkyl, halo, and cyano; J can be C; and X, Y, and Z can each be independently N or C, wherein the valency of any carbon atom is filled as needed with hydrogen atoms.
[00204] [0077] In some embodiments, can represent a single bond. In other
Figure imgf000049_0001
embodiments,
Figure imgf000049_0003
can represent a double bond. In some embodiments,
Figure imgf000049_0002
joining Y and
Z can represent a single bond. In other embodiments,
Figure imgf000049_0004
joining Y and Z can represent a double bond. In some embodiments, when
Figure imgf000049_0005
joining G and J representes a single bond,
G can be N and the N is substituted with RG. In other embodiments, when
Figure imgf000049_0007
joining G and J represents a double bond, G can be N. In some embodiments, when
Figure imgf000049_0012
joining G and J representes a double bond, then
Figure imgf000049_0011
joining J and RJ can be a single bond. In some embodiments, when . joining G and J representes a double bond, then
Figure imgf000049_0008
joining J and RJ can not be a double bond. In some embodiments, when
Figure imgf000049_0006
joining J and RJ representes a double bond, then
Figure imgf000049_0010
joining G and J can be a single bond. In some embodiments, when
Figure imgf000049_0009
joining J and RJ representes a double bond, then . joining G and J can not be a double bond.
[00205] In some embodiments, RJ can be -NRaRb. In other embodiments, RJ can be - ORb. In still other embodiments, RJ can be =O. In some embodiments, when RJ is =O, then
- joining G and J represents a single bond and G is N and the N is substituted with RG.
In some embodiments, RG is -CH2CH2-C(=O)NH2.
[00206] In some embodiments, Ra can be hydrogen. In some embodiments, Ra can be C1-C4 alkyl. For example, Ra can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl.
[00207] In some embodiments, Rb can be Rc. In some embodiments, Rb can be -(C1- C4 alkyl)-Rc. For example, Rb can be -CH2-RC, -CH2CH2-RC,
-CH2CH2CH2-Rc, or -CH2CH2CH2CH2-R^ In some embodiments, when Rb is -CH2CH2-Rc, RC can be -O(C1-C4 alkyl). In other embodiments, when Rb is -CH2CH2-Rc, RC can be -O(C1-C4 haloalkyl). In still other embodiments, when Rb is - CH2CH2-Rc, RC can be -C(=O)NH2.
[00208] In some embodiments, Rc can be -OH. In some embodiments, Rc can be -O(C1-C4 alkyl). In some embodiments, Rc can be -O(C1-C4 haloalkyl). In some embodiments, Rc can be -C(=O)NH2. In some embodiments, Rc can be unsubstituted C6-10 aryl. In some embodiments, Rc can be substituted C6-10 aryl. In some embodiments, Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S. In some embodiments, Rc can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S. In some embodiments, when a Rc moiety is indicated as substituted, the moiety can be substituted with one or more, for example, one, two, three, or four substituents E. In some embodiments, E can be -OH. In some embodiments, E can be C1-C4 alkyl. In some embodiments, E can be C1-C4 haloalkyl. In some embodiments, E can be -O(C1-C4 alkyl).
In some embodiments, E can be -O(C1-C4 haloalkyl).
[00209] In some embodiments, when Rb is -CH2CH2-RC, Rc can be unsubstituted C6-10 aryl. In other embodiments, when Rb is -CH2CH2-RC, Rc can be substituted C6-10 aryl. In still other embodiments, when Rb is -CH2CH2-RC, Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S. In yet still other embodiments, Rb can be -(C1-C4 alkyl)-Rc and Rc can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S. When a Rc moiety is indicated as substituted, the moiety can be substituted with one or more, for example, one, two, three, or four substituents E. In some embodiments, E can be -OH.
In other embodiments, E can be C1-C4 alkyl. In still other embodiments, E can be C1-C4 haloalkyl. In still other embodiments, E can be -O(C1-C4 alkyl). In still other embodiments, E can be -O(C1-C4 haloalkyl).
[00210] In some embodiments, when Rb is -CH2CH2-RC, Rc can be phenyl. In other embodiments, when Rb is -CH2CH2-RC, Rc can be naphthyl. In still other embodiments, when Rb is -CH2CH2-RC, Rc can be hydroxyphenyl. In still other embodiments, when Rb is - CH2CH2-Rc, RC can be indolyl.
[00211] In some embodiments, RK can be hydrogen. In other embodiments, RK can be unsubstituted C1-6 alkyl. For example, in some embodiments, RK can be methyl, ethyl, n- propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, pentyl (branched and straight-chained), or hexyl (branched and straight-chained). In other embodiments, RK can be substituted C1-6 alkyl. In other embodiments, RK can be -NH(C1-4 alkyl). For example, in some embodiments, RK can be -NH(CH3), -NH(CH2CH3), -NH(isopropyl), or -NH(sec-butyl). In other embodiments, RK can be -N(C1-4 alky 1)2.
[00212] In some embodiments, RK can be unsubstituted C6-10 aryl. In other embodiments, RK can be substituted C6-10 aryl. In other embodiments, RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S. In other embodiments, RK can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S. When a RK moiety is indicated as substituted, the moiety can be substituted with one or more, for example, one, two, three, or four substituents substituents Q. In some embodiments, Q can be -OH. In other embodiments, Q can be C1-4 alkyl. In still other embodiments, Q can be C1-4 haloalkyl. In still other embodiments, Q can be halo. In still other embodiments, Q can be cyano. In still other embodiments, Q can be -O-(C1-4 alkyl). In still other embodiments, Q can be -O-(C1-4 haloalkyl).
[00213] In some embodiments, RK can be phenyl or naphthyl. In other embodiments, RK can be benzothiophenyl. In other embodiments, RK can be benzothiophenyl. In other embodiments, RK can be benzothiophenyl. In still other embodiments, RK can be pyridinyl.
In yet still other embodiments, RK can be pyridinyl substituted with one or more substituents Q. For example, RK can be methylpyridinyl, ethylpyridinyl cyanopyridinyl, chloropyridinyl, fluoropyridinyl, or bromopyridinyl.
[00214] In some embodiments, RG can be hydrogen. In some embodiments, RG can be C1-4 alkyl. In some embodiments, RG can be -(C1-4 alkyl)-C(=O)NH2.
[00215] In some embodiments, RY and RZ can independently be absent. In other embodiments, RY and RZ can independently be hydrogen. In other embodiments, RY and RZ can independently be halo. In other embodiments, RY and RZ can independently be C1-6 alkyl. In other embodiments, RY and RZ can independently be -OH. In still other embodiments, RY and RZ can independently be -O-(C1-4 alkyl). In other embodiments, RY and RZ can independently be -NH(C1-4 alkyl). For example, RY and RZ can independently be
-NH(CH3), -NH(CH2CH3),
-NH(isopropyl), or -NH (.sec-butyl). In other embodiments, RY and RZ can independently be - N(C1-4 alkyl)2.
[00216] In some embodiments, RY and RZ taken together with the atoms to which they are attached can be joined together to form a ring. In some embodiments, RY and RZ taken together with the atoms to which they are attached can be joined together to form
Figure imgf000051_0001
In other embodiments, RY and RZ taken together with the atoms to which they are attached can be joined together to form In other embodiments, RY and R
Figure imgf000051_0002
Z taken together with the atoms to which they are attached can be joined together to form
Figure imgf000052_0001
In still other embodiments, RY and RZ taken together with the atoms to which they are attached can be joined together to form
Figure imgf000052_0002
In yet still other embodiments,
RY and RZ taken together with the atoms to which they are attached can be joined together to form
Figure imgf000052_0003
in other embodiments, RY and RZ taken together with the atoms to which they are attached can be joined together to form
Figure imgf000052_0004
In yet other embodiments, RY and RZ taken together with the atoms to which they are attached can be joined together to form In yet still other embodiments, RY and RZ taken together with the atoms to
Figure imgf000052_0005
which they are attached can be joined together to form
Figure imgf000052_0006
In other embodiments, RY and RZ taken together with the atoms to which they are attached can be joined together to form In still other embodiments, RY and RZ taken together
Figure imgf000052_0007
with the atoms to which they are attached can be joined together to form and In
Figure imgf000052_0008
some embodiments, when RY and RZ taken together with the atoms to which they are attached can be joined together to form a ring, the ring can be substituted with one, two, or three groups independently selected from C1-C4 alkyl, -N(C1-C4 alkyl)2, cyano, unsubstituted phenyl, and phenyl substituted with 1-5 halo atoms.
[00217] In some embodiments, when RY and RZ taken together forms , then
Figure imgf000053_0001
RJ can be -ORb or =O.
[00218] In some embodiments, RY and RZ taken together with the atoms to which they are attached can be joined together to form
Figure imgf000053_0002
In other embodiments, RY and RZ taken together with the atoms to which they are attached can be joined together to form
. In other embodiments, RY and RZ taken together with the atoms to which they
Figure imgf000053_0003
are attached can be joined together to form
Figure imgf000053_0004
In other embodiments, RY and RZ taken together with the atoms to which they are attached can be joined together to form
. In other embodiments, RY and RZ taken together with the atoms to which they
Figure imgf000053_0005
are attached can be joined together to form
Figure imgf000053_0006
In other embodiments, RY and RZ taken together with the atoms to which they are attached can be joined together to form In other embodiments, RY and RZ taken together with the atoms to which they
Figure imgf000054_0001
are attached can be joined together to form
Figure imgf000054_0002
In other embodiments, RY and RZ taken together with the atoms to which they are attached can be joined together to form
In other embodiments, RY and RZ taken together with the atoms to which
Figure imgf000054_0003
they are attached can be joined together to form In other embodiments, RY and
Figure imgf000054_0004
RZ taken together with the atoms to which they are attached can be joined together to form In some embodiments, when RY and RZ taken together with the atoms to
Figure imgf000054_0005
which they are attached can be joined together to form a ring, the ring can be substituted with one, two, or three groups independently selected from C1-C4 alkyl, -N(C1-C4 alkyl)2, cyano, unsubstituted phenyl, and phenyl substituted with 1-5 halo atoms. In some embodiments, RY and RZ taken together with the atoms to which they are attached can be
Figure imgf000054_0006
In other embodiments, RY and RZ taken together with the atoms to which they are attached can be In still other embodiments, RY and RZ taken together with the
Figure imgf000054_0007
atoms to which they are attached can be In yet still other embodiments, RY
Figure imgf000055_0001
and RZ taken together with the atoms to which they are attached can be
Figure imgf000055_0002
In other embodiments, RY and RZ taken together with the atoms to which they are attached can be
Figure imgf000055_0003
[00219] In some embodiments, Rd can be hydrogen. In other embodiments, Rd can be C1-C4 alkyl. For example Rd can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl. In still other embodiments, Rd can be halo. In other embodiments, Rd can be cyano.
[00220] In some embodiments, Rm can be hydrogen. In other embodiments, Rm can be C1-C4 alkyl. For example Rm can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl. In still other embodiments, Rm can be halo. For example, Rm can be fluoro, chloro, bromo, or iodo. In other embodiments, Rm can be cyano.
[00221] In some embodiments, X, Y, and Z can each be independently N or C, wherein the valency of any carbon atom is filled as needed with hydrogen atoms. In some embodiments, X can be N, Y can be N, and Z can be N. In other embodiments, X can be N, Y can be N, and Z can be CH. In some embodiments, X can be N, Y can be CH, and Z can be N. In still other embodiments, X can be CH, Y can be N, and Z can be N. In yet still other embodiments, X can be CH, Y can be CH, and Z can be N. In other embodiments, X can be CH, Y can be N, and Z can be CH. In yet other embodiments, X can be N, Y can be CH, and Z can be CH. In other embodiments, X can be CH, Y can be CH, and Z can be CH. [00222] In some embodiments, Ra can be hydrogen; Rb can be -(C1-C4 alkyl)-Rc; Rc can be selected from the group consisting of: -C(=O)NH2; unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a Rc moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C1-C4 alkyl, C1-C4 haloalkyl, -O(C1-C4 alkyl), and -O(C1-C4 haloalkyl); RK can be selected from the group consisting of: hydrogen, unsubstituted C1-6 alkyl; -NH(C1-4 alkyl); -N(C1-4 alkyl)2, unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: -OH, C1-4 alkyl, C1-4 haloalkyl, halo, cyano, -O-(C1-4 alkyl), and -O-(C1-4 haloalkyl); RG can be -(C1-4 alkyl)- C(=O)NH2; RY and RZ can each be independently absent or be selected from the group consisting of: hydrogen, C1-6 alkyl, and -NH(C1-4 alkyl); or RY and RZ taken together with the atoms to which they are attached can be joined together to form a ring selected from: ; wherein said ring can be optionally substituted
Figure imgf000056_0001
with one, two, or three groups independently selected from C1-4 alkyl, C1-4 haloalkyl, halo, cyano, -OH, -O-(C1-4 alkyl), -N(C1-4 alky 1)2, unsubstituted C6-C10 aryl, C6-C10 aryl substituted with 1-5 halo atoms, and -O-(C1-4 haloalkyl); Rd can be C1-C4 alkyl; Rm can be cyano; and X, Y, and Z can each be independently N or C, wherein the valency of any carbon atom is filled as needed with hydrogen atoms.
[00223] In some embodiments, Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be selected from the group consisting of: unsubstituted phenyl, substituted phenyl, indolyl, and - C(=O)NH2; RK can be selected from the group consisting of: hydrogen, methyl, substituted pyridinyl, unsubstituted benzothiophenyl, and -NH(C1-C4 alkyl); RG can be -CH2CH2- C(=O)NH2; RY can be -NH(C1-C4 alkyl); RZ can be absent or hydrogen; or RY and RZ taken together with the atoms to which they are attached can be joined together to form a ring
Figure imgf000057_0001
substituted with one, two, or three groups independently selected from C1-C4 alkyl, -N(C1-C4 alky 1)2, cyano, unsubstituted phenyl, and phenyl substituted with 1-5 halo atoms; Rd can be C1-C4 alkyl; Rm can be cyano; and X can be N or CH.
[00224] In some embodiments, when RJ is -NRaRb; G can be N; - j pining G and J can be a double bond; Ra can hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; or Rc can be substituted C6-10 aryl, substituted with one or more E, wherein E is - OH; RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; or RK can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; substituted with one or more Q, wherein Q can be selected from cyano, halo, or C1-C4 alkyl; RY and RZ taken together can be
Figure imgf000057_0002
[00225] In some embodiments, when RJ is -NRaRb; G can be N; . j pining G and J can be a double bond; Ra can hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; or Rc can be substituted C6-10 aryl, substituted with one or more E, wherein E is - OH; RK can be hydrogen, C1-4 alkyl, or unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and RY and RZ taken together can be
Figure imgf000057_0003
[00226] In some embodiments, when RJ is -NRaRb; G can be N; - j pining G and J can be a double bond; Ra can hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; or Rc can be substituted C6-10 aryl, substituted with one or more E, wherein E is - OH; RK can be hydrogen, C1-4 alkyl, or unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and RY and RZ taken
Figure imgf000058_0001
[00227] In some embodiments, when RJ is -NRaRb; G can be N;
Figure imgf000058_0003
j pining G and J can be a double bond, Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be substituted G,- lo aryl; substituted with one or more E, wherein E can be
-OH; RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RY can be -NH(C1-4 alkyl); RZ can be hydrogen; J can be C; X can be N; Y can be C; Z can be C; and
Figure imgf000058_0004
joining Y and Z can be a double bond. In some embodiments, the compound of Formula (I) can be 4-(2-((2- (benzo[b]thiophen-3-yl)-6-(isopropylamino)pyrimidin-4-yl)amino)ethyl)phenol.
[00228] In some embodiments, when RJ is -NRaRb; G can be N; . j pining G and J can be a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC, Rc can be substituted G,- lo aryl, substituted with one or more E, wherein E can be
-OH; RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RY and RZ taken together is
Figure imgf000058_0002
wherein the ring is substituted with C1-C4 alkyl; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I) can be 4-(2-((2-(benzo[b]thiophen-3-yl)- 7-isopropylthieno[3,2-b]pyrimidin-4-yl)amino)ethyl)phenol.
[00229] In some embodiments, when RJ is -NRaRb; G can be N; - joining G and J can be a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC, Rc can be substituted G,- 10 aryl, substituted with one or more E, wherein E can be -OH; RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RY and RZ taken together is Rd can be C1-C4 alkyl; J can be C; X can be N; Y
Figure imgf000059_0001
can be C; and Z can be C. In some embodiments, the compound of Formula (I) can be 4-(2- ((2-(benzo[b]thiophen-3-yl)-7-isopropyl-6.7-dihydro-5H-pyrrolo[2,3-d]pyri midin-4- yl)amino)ethyl)phenol.
[00230] In some embodiments, when RJ is -NRaRb; G can be N; joining G and J
Figure imgf000059_0003
can be a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC, Rc can be substituted G,- 10 aryl, substituted with one or more E, wherein E can be
-OH; RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RY and RZ taken together is Rd can
Figure imgf000059_0002
be C1-C4 alkyl; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I) can be 2-(benzo[b]thiophen-3-yl)-4-((4- hydroYyphenethyl)amino)-7-isopropyl-5.7-dihydro-6H-pyrrolo[2,3-d]pyrimidin-6-one. [00231] In some embodiments, when RJ is -ORb; G can be N;
Figure imgf000059_0005
joining G and J can be a double bond; Rb can be -CH2CH2-RC; Rc can be
-C(=O)NH2; RK can unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RY and RZ taken together can be Rd can be C1-C4 alkyl; J can be C; X can be N; Y can be C; and Z is C. In some
Figure imgf000059_0004
embodiments, the compound of Formula (I) can be 3-((2-(benzo[b]thiophen-3-yl)-9- isopropyl-9H-purin-6-yl)oYy)propanamide.
[00232] In some embodiments, when RJ is is -NRaRb; G can be N; - joining G and J can be a double bond; Rb can be -CH2CH2-RC; Rc can be substituted C6-10 aryl, substituted with one or more E, wherein E is -OH; RK is unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RY and RZ taken together can
Figure imgf000060_0001
; wherein said ring is substituted with -N(C1-4 alkyl)2; J can be C; X can be N; Y can be C; and Z is C. In some embodiments, the compound of Formula (I) can be 4-(2-((2-(benzo[b]thiophen-3-yl)-8-(dimethylamino)pyrimido[5,4-ri]pyrimidin-4- yl)amino)ethyl)phenol.
[00233] In some embodiments, when RJ is is -NRaRb; G can be N; - joining G and J can be a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q is cyano; RY can be -NH(C1-4 alkyl); RZ can be absent; J can be C; X can be C; Y can be C; Z can be N; and == joining Y and Z can be a double bond. In some embodiments, the compound of Formula (I) can be 5-(2-((2- (1H-indol-3-yl)ethyl)amino)-6-(sec-butylamino)pyrimidin-4-yl)nicotinonitrile.
[00234] In some embodiments, when RJ is -NRaRb; G can be N; . j pining G and J can be a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O,
N, and S; RK can be unsubstituted C1-6 alkyl; RY and RZ taken together can
Figure imgf000060_0002
wherein the ring is substituted with unsubstituted C6-C10 aryl; J can be C; X can be N; Y can be C; Z can be C. . In some embodiments, the compound of Formula (I) can be N-(2-(lH- indol-3-yl)ethyl)-2-methyl-6-phenylthieno[2,3-d]pyrimidin-4-amine
[00235] In some embodiments, when RJ can be -NRaRb; G can be N; - joining G and J can be a double bond; Ra can be hydrogen; Rb can be
-CH2CH2-Rc; RC can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be hydrogen; RY and RZ taken together can
Figure imgf000060_0003
; wherein the ring is substituted with substituted C6-C10 aryl; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I) can be N-(2-(1H-indol-3-yl)ethyl)-6-(4-fluorophenyl)thieno[2,3-<f]pyrimi din-4- amine
[00236] In some embodiments, when RJ is =O; G can be N substituted with RG; — joining G and J can be a single bond; RG can be -(C1-4 alkyl)-C(=O)NH2; RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group i consisting of O, N, and S; RY and RZ taken together can be Rd can be C1-C4
Figure imgf000061_0001
alkyl; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I) can be 3-(2-(benzo[b]thiophen-3-yl)-9-isopropyl-6-oxo-6,9- dihydro- lif-purin- 1 -yljpropan amide.
[00237] In some embodiments, when RJ is -NRaRb; G can be N; - j pining G and J can be a double bond Ra can be hydrogen Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q can be halo; RY and RZ taken together can be
Figure imgf000061_0002
J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I) can be N-(2-(1H-indol-3-yl)ethyl)-2-(5-fluorop\Tidin-3- yl)quinazolin-4-amine.
[00238] In some embodiments, when RJ is -NRaRb; G is N; - joining G and J can be a double bond; Ra can be hydrogen Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q can be cyano; RY and RZ taken together is
Figure imgf000061_0003
J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I) can be 5-(4-((2-(1H-indol-3-yl)ethyl)amino)quinazolin-2-yl)nicotinonitrile. [00239] In some embodiments, when RJ is -NRaRb; G can be N; - j pining G and J can be a double bond; Ra can be hydrogen Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O,
N, and S; RK can be -NH(C1-4 alkyl); RY and RZ taken together can be
Figure imgf000062_0001
: J can be
C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I) can be N4-(2-(1H-indol-3-yl)ethyl)-N2-(sec-butyl)quinazoline-2, 4-diamine.
[00240] In some embodiments, when RJ is -NRaRb; G can be N; . joining G and
J can be a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be substituted C6-10 aryl, substituted with one or more E, wherein E is -OH; RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and
S; RY and RZ taken together can be
Figure imgf000062_0002
; wherein the ring is substituted with cyano;
Rd can be C1-C4 alkyl; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I) can be 2-(benzo[b]thiophen-3-yl)-4-((4- hydro\yphenethyl)amino)-7-isopropyl-7H-pyrrolo[2,3-d]pyrimidine-5-carbonitrile.
[00241] In some embodiments, when RJ is -NRaRb; G can be N; . j pining G and J can be a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RY and RZ taken together can be
Figure imgf000062_0003
; wherein the ring is substituted with C1-4 alkyl; J can be C; X can be C; Y can be N; and Z can be C; wherein the valency of any carbon atom is filled as needed with hydrogen atoms. In some embodiments, the compound of Formula (I) can be N-(2-(1H-indol-3- yl)ethyl)-6-(benzo[b]thiophen-3-yl)-3-isopropylimidazo[1,5-α]pyrazin-8-amine.
[00242] In some embodiments, when RJ is -NRaRb; G can be N; - joining G and J can be a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be substituted C6- 10 aryl, substituted with one or more E, wherein E is -OH; RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S;
RY and RZ taken together can be
Figure imgf000063_0001
wherein the ring can be substituted with C1-4 alkyl; J can be C; X can be C; Y can be N; and Z can be C; wherein the valency of any carbon atom is filled as needed with hydrogen atoms. In some embodiments, the compound of Formula (I) can be 4-(2-((6-(benzo[b]thiophen-3-yl)-3-isopropylimidazo[1,5-a]pyrazin-8- yl)amino)ethyl)phenol.
[00243] In some embodiments, when RJ is -NRaRb; G can be N; . j pining G and J represents a double bond; Ra can be hydrogen Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q is cyano; RY and RZ taken together is
Figure imgf000063_0002
; wherein the ring is substituted with C1-C4 alkyl; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I) can be 5-(4-((2- (1H-indol-3-yl)ethyl)amino)-7-isopropylthieno[2,3-d] pyrimi din-2 -yl)nicotinonitrile.
[00244] In some embodiments, when RJ is -NRaRb; G can be N; j pining G and J
Figure imgf000063_0004
represents a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q is halo; RY and RZ taken together can be
Figure imgf000063_0003
; wherein the ring is substituted with C1-C4 alkyl; J can be C; X can be N;
Y can be C; and Z can be C. In some embodiments, the compound of Formula (I) can be N- (2-(1H-indol-3-yl)ethyl)-2-(5-fluoropyridin-3-yl)-7-isopropylthieno[2,3-d]pyrimi din-4- amine.
[00245] In some embodiments, when RJ is -NRaRb; G can be N; . j pining G and J can be a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q is halo; RY and RZ taken together can be
Figure imgf000064_0001
J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I) can beN-(2-(1H-indol-3-yl)ethyl)-2-(5-fluoropyridin-3-yl)furo[3,2- d\ py rimidin-4-amine.
[00246] In some embodiments, when RJ is -NRaRb; G can be N; - joining G and J can be a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q is C1-C4 alkyl; RY and RZ taken together can be
Figure imgf000064_0002
J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I) can be N-(2-(1H-indol-3-yl)ethyl)-2-(5-methylpyridin-3- yl)furo[2,3-d]pyrimidin-4-amine.
[00247] In some embodiments, when RJ is -NRaRb; G can be N; - joining G and J can be a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q is C1-C4 alkyl; RY and RZ taken together can be wherein the ring is substituted with C1-C4 alkyl J can be C; X can be N; Y can be
Figure imgf000065_0001
C; and Z can be C. In some embodiments, the compound of Formula (I) can be N-(2-(lH- indol-3-yl)ethyl)-7-isopropyl-2-(5-methylpyridin-3-yl)thieno[2,3-d]pyrimidin-4-amine.
[00248] In some embodiments, when RJ is -NRaRb; G is N; - joining G and J can be a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q is cyano; RY and RZ taken together can be
Figure imgf000065_0002
J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I) can be 5-(4-((2-(1H-indol-3-yl)ethyl)amino)furo[2,3-d]pyrimidin-2- yljnicotinonitrile.
[00249] In some emdiments, provided herein is compound of Formula (I), wherein the compound can be selected from:
4-(2-((2-(benzo[b]thiophen-3-yl)-6-(isopropylamino)pyrimidin-4-yl)amino)ethyl)phenol; 4-(2-((2-(benzo[b]thiophen-3-yl)-7-isopropylthieno[2,3-d] pyrimidin-4- yl)amino)ethyl)phenol;
4-(2-((2-(benzo[b]thiophen-3-yl)-7-isopropyl-6,7-dihydro-5H-pyrrolo[2,3-d]pyrimidin-4- yl)amino)ethyl)phenol;
2-(benzo[b]thiophen-3-yl)-4-((4-hydroxyphenethyl)amino)-7-isopropyl-5.7-dihydro-6H- pyrrolo[2,3-d]pyrimidin-6-one;
3-((2-(benzo[b]thiophen-3-yl)-9-isopropyl-9H-purin-6-yl)oxy)propanamide:
4-(2-((2-(benzo[b]thiophen-3-yl)-8-(dimethylamino)pyrimido|5.4-6/|pyrimidin-4- yl)amino)ethyl)phenol;
5-(2-((2-(1H-indol-3-yl)ethyl)amino)-6-(sec-butylamino)pyrimidin-4-yl)nicotinonitrile: N-(2-(1H-indol-3-yl)ethyl)-2-methyl-6-phenylthieno[2,3-d] pyrimidin-4-amine: N-(2-(1H-indol-3-yl)ethyl)-6-(4-fluorophenyl)thieno[2,3-d]pyrimidin-4-amine;
3-(2-(benzo[b]thiophen-3-yl)-9-isopropyl- 6-oxo-6.9-dihydro-1H-purin-1-yljpropanamide: N-(2-(1H-indol-3-yl)ethyl)-2-(5-fluoropyridin-3-yl)quinazolin-4-amine;
5-(4-((2-(1H-indol-3-yl)ethyl)amino)quinazolin-2-yl)nicotinonitrile:
N4-(2-(1H-indol-3-yl)ethyl)- N2-(sec-butyl)quinazoline-2, 4-diamine; 2-(benzo[b]thiophen-3-yl)-4-((4-hydroxyphenethyl)amino)-7-isopropyl-7H-pyrrolo[2,3- 6/|pyrimidine-5-carbonitrile:
N-(2-(1H-indol-3-yl)ethyl)-6-(benzo[b]thiophen-3-yl)-3-isopropylimidazo[1,5-α]pyrazin-8- amine;
4-(2-((6-(benzo[b]thiophen-3-yl)-3-isopropylimidazo[1,5-α]pyrazin-8-yl)amino)ethyl)phenol;
5-(4-((2-(1H-indol-3-yl)ethyl)amino)-7-isopropylthieno[2,3-d] pyrimidin-2-yl)nicotinonitrile;
N-(2-(1H-indol-3-yl)ethyl)-2-(5-fluoropyridin-3-yl)-7-isopropylthieno[2,3-d]pyrimi din-4- amine;
A-(2-(1H-indol-3-yl)ethyl)-2-(5-fluoropyridin-3-yl)furo[2,3-d]pyrimidin-4-amine;
A-(2-(1H-indol-3-yl)ethyl)-2-(5-methylpyridin-3-yl)furo[2,3-d]pyrimidin-4-amine;
N-(2-(1H-indol-3-yl)ethyl)-7-isopropyl-2-(5-methylpyridin-3-yl)thieno[2,3-d]pyrimidin-4- amine;
5-(4-((2-(1H-indol-3-yl)ethyl)amino)furo[2,3-d]pyrimidin-2-ly)nicotinonitrile; and pharmaceutically acceptable salts thereof.
Formula (I-A)
[00250] In some embodiments provided herein, the compound of Formula (I) can have the structure of Formula (I-A):
Figure imgf000066_0001
(I-A), including pharmaceutically acceptable salts thereof, wherein: RJ can be -NRaRb; Ra can be hydrogen or C1-C4 alkyl; Rb can be Rc or -(C1-C4 alkyl)-Rc; Rc can be selected from the group consisting of: unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a Rc moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C1-C4 alkyl, C1-C4 haloalkyl, -O(C1-C4 alkyl), and -O(C1-C4 haloalkyl); RK can be selected from the group consisting of: hydrogen, unsubstituted C1-6 alkyl;
-NH(C1-4 alkyl); -N(C1-4 alkyl)2, unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: -OH, C1-4 alkyl, C1-4 haloalkyl, halo, cyano, -O-(C1-4 alkyl), and -O-(C1-4 haloalkyl); Y and Z can each be C; X can be N or CH; W can be O or S; and Re can be hydrogen or C1-C4 alkyl.
[00251] In some embodiments, Ra can be hydrogen. In other embodiments, Ra can be C1-C4 alkyl.
[00252] In some embodiments, Rb can be -(C1-C4 alkyl)-Rc. For example, Rb can be -
CH2-Rc, -CH2CH2-Rc, -CH2CH2CH2-Rc, or
-CH2CH2CH2CH2-Rc.
[00253] In some embodiments, Rc can be -OH. In some embodiments, Rc can be - O(C1-C4 alkyl). In some embodiments, Rc can be -O(C1-C4 haloalkyl). In some embodiments, Rc can be -C(=O)NH2. In some embodiments, Rc can be unsubstituted G,-in aryl. In some embodiments, Rc can be substituted C6-10 aryl. In some embodiments, Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S. In some embodiments, Rc can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S. In some embodiments, when a Rc moiety is indicated as substituted, the moiety can be substituted with one or more, for example, one, two, three, or four substituents E. In some embodiments, E can be
-OH. In some embodiments, E can be C1-C4 alkyl. In some embodiments, E can be C1-C4 haloalkyl. In some embodiments, E can be -O(C1-C4 alkyl). In some embodiments, E can be -O(C1-C4 haloalkyl). In some embodiments Rc can be phenyl. In other embodiments, Rc can be hydroxyphenyl. In still other embodiments, Rc can be indolyl.
[00254] In some embodiments, RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S. In some embodiments, RK can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein the substituted heteroaryl can substituted with one or more substituents Q, wherein each Q can independently selected from the group consisting of: -OH, C1-4 alkyl, C1-4 haloalkyl, halo, cyano, -O-(C1-4 alkyl), and -O- (C1-4haloalkyl). In some embodiments, RK can be pyridinyl. In other embodiments, RK can be pyridinyl substituted with one or more substituents Q. For example, RK can be methylpyridinyl, ethylpyridinyl cyanopyridinyl, chloropyridinyl, fluoropyridinyl, or bromopyridinyl.
[00255] In some embodiments, Re can be hydrogen. In some embodiments, Re can be C1-C4 alkyl. For example, Re can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl.
[00256] In some embodiments, Ra can be hydrogen; Rb can be -(C1-C4 alkyl)-Rc; Rc can be selected from the group consisting of: unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a Rc moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C1-C4 alkyl, C1-C4 haloalkyl, - O(C1-C4 alkyl), and -O(C1-C4 haloalkyl); RK can be selected from the group consisting of: unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein the substituted heteroaryl is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: -OH, C1-4 alkyl, C1-4 haloalkyl, halo, cyano, -O-(C1-4 alkyl), and -O-(C1-4 haloalkyl); and Re can be C1-C4 alkyl.
[00257] In some embodiments, Ra can be hydrogen; Rb can be -(CH2-CH2)-RC; Rc can be selected from the group consisting of: substituted phenyl and unsubstituted indolyl; wherein the substituted phenyl is substituted with one substituent E, wherein E can be -OH; RK can be selected from the group consisting of: unsubstituted benzothiophenyl and substituted pyridinyl; wherein the substituted pyridinyl is substituted with one substituent Q, wherein Q can be selected from the group consisting of: C1-4 alkyl, halo, and cyano; and Re can be isopropyl.
[00258] In some embodiments, when W is O, RJ can be -NRaRb; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be selected from the group consisting of: unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a Rc moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C1-C4 alkyl, and -O(C1-C4 alkyl); RK can be selected from the group consisting of unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: - C1-4 alkyl, halo, cyano, and -O-(C1-4 alkyl); Y and Z can each be C; X can be N or CH; and Re can be hydrogen or C1-C4 alkyl.
[00259] In some embodiments, when W is S, RJ can be -NRaRb; Ra can be hydrogen; Rb can be -CH2CH2-Rc; Rc can be selected from the group consisting of: unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a Rc moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C1-C4 alkyl, and -O(C1-C4 alkyl); RK can be selected from the group consisting of unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: - C1-4 alkyl, halo, cyano, and -O-(C1-4 alkyl); Y and Z can each be C; X can be N or CH; and Re can be hydrogen or C1-C4 alkyl.
[00260] In some embodiments, when RJ is -NRaRb; G can be N; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q is C1-C4 alkyl; W can be S; Re can be C1-C4 alkyl; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I-A) can be Y-(2-(1H-indol-3- yl)ethyl)-7-isopropyl-2-(5-methylpyridin-3-yl)thieno[2,3-d] pyrimidin-4-amine.
[00261] In some embodiments, when RJ is -NRaRb; G can be N; Ra can be hydrogen Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q is cyano; W can be S; Re can be C1-C4 alkyl; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I- A) can be 5-(4-((2-(1H-indol-3- yl)ethyl)amino)-7-isopropylthieno[2,3-d] pyrimi din-2 -yl)nicotinonitrile.
[00262] In some embodiments, when RJ is -NRaRb; G can be N; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q is halo; W can be S; Re can be C1-C4 alkyl; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I- A) can be N-(2-(1H-indol-3-yl)ethyl)- 2-(5-fluoropyridin-3-yl)-7-isopropylthieno[2,3-d]pyrimidin-4-amine.
[00263] In some embodiments, when RJ is -NRaRb; G can be N; Ra can be hydrogen; Rb can be -CH2CH2-RC, Rc can be substituted C6-10 aryl, substituted with one or more E, wherein E can be -OH; RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; W can be S; Re can be C1-C4 alkyl;
J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I-A) can be 4-(2-((2-(benzo[b]thiophen-3-yl)-7-isopropylthieno[2,3-d] pyrimidin- 4-yl)amino)ethyl)phenol.
[00264] In some embodiments, when RJ is -NRaRb; G can be N; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q is halo; W can be O; Re can be hydrogen; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I-A) can be Y-(2-(1H-indol-3-yl)ethyl)-2- (-5fluoropyridin-3-yl)furo[2,3-d]pyrimidin-4-amine.
[00265] In some embodiments, when RJ is -NRaRb; G can be N; . j pining G and J can be a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q is C1-C4 alkyl; W can be O; Re can be hydrogen;
J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I-A) can be N-(2-(1H-indol-3-yl)ethyl)-2-(5-methylpyridin-3-yl)furo [3.2- d] pyrimidin-4-amine.
[00266] In some embodiments, when RJ is -NRaRb; G is NRa can be hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q is cyano; W can be O; Re can be hydrogen; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I- A) can be 5-(4-((2-(1H-indol-3- yl)ethyl)amino)furo[2,3-d] pyrimidin-2-yl)nicotinonitrile.
[00267] [0140] In some embodiments, the compound of Formula (I- A), or a pharmaceutically acceptable salt thereof, can selected from the group consisting of:
N-(2-(1H-indol-3-yl)ethyl)-7-isopropyl-2-(5-methylpyridin-3-yl)thieno[2,3-d]pyrimidin-4- amine;
5-(4-((2-(1H-indol-3-yl)ethyl)amino)-7-isopropylthieno[2,3-d]pyrimidin-2-yl)nicotinonitrile:
N-(2-(1H-indol-3-yl)ethyl)-2-(5-fluoropyridin-3-yl)-7-isopropylthieno[2,3-d]pyrimidin-4- amine;
4-(2-((2-(benzo[b]thiophen-3-yl)-7-isopropylthieno[2,3-d] pyri midin-4- yl)amino)ethyl)phenol;
N-(2-(1H-indol-3-yl)ethyl)-2-(5-fluoropyridin-3-yl)furo[2,3-d]pyrimidin-4-amine; N-(2-(1H-indol-3-yl)ethyl)-2-(5-methylpyridin-3-yl)furo[2,3-d]pyrimidin-4-amine; and
5-(4-((2-(1H-indol-3-yl)ethyl)amino)furo[2,3-d]pyrimi din-2 -yl)nicotinonitrile.
Formula (I-B)
[00268] In other embodiments provided herein, the compound of Formula (I) can have the structure of Formula (I-B): (I-B) including pharmaceutically
Figure imgf000071_0001
acceptable salts thereof, wherein: Ra can be hydrogen or C1-C4 alkyl; Rb can be Rc or -(C1-4 alkyl)-Rc; Rc can be selected from the group consisting of: -OH, -O(C1-C4 alkyl), -O(C1-C4 haloalkyl); -C(=O)NH2; unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a Rc moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C1-C4 alkyl, C1-C4 haloalkyl, -O(C1-C4 alkyl), and -O(C1-C4 haloalkyl); RK can be selected from the group consisting of: hydrogen, unsubstituted C1-6 alkyl; substituted C1-6 alkyl; -NH(C1-4 alkyl); -N(C1-4 alkyl)2, unsubstituted C6-10 aryl; substituted G,- 10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: -OH, C1-4 alkyl, C1-4 haloalkyl, halo, cyano, -O-(C1-4 alkyl), and -O-(C1-4 haloalkyl); RG can be selected from the group consisting of hydrogen, C1-4 alkyl, and -(C1-4 alkyl)-C(=O)NH2; Rf can be selected from the group consisting of hydrogen, C1-4 alkyl, unsubstituted C6-C10 aryl, and C6-C10 aryl substituted with 1-5 halo atoms; U can be N or CRU; V can be S or NRV; RU can be selected from the group consisting of hydrogen, C1-4 alkyl, halo, and cyano; RV can be hydrogen or C1-C4 alkyl; wherein when U is CRU and V is NRV, RU is selected from the group consisting of C1-4 alkyl, halo, and cyano; Y and Z can each be C; and X can be N or CH.
[00269] In some embodiments, Ra can be hydrogen. In other embodiments, Ra can be C1-C4 alkyl.
[00270] In some embodiments, Rb can be -(C1-C4 alkyl)-Rc. For example, Rb can be - CH2-Rc, -CH2CH2-Rc, -CH2CH2CH2-Rc, or -CH2CH2CH2CH2-RC. In certain embodiments, Rb can be -(CH2CH2)-RC. In certain embodiments, Rb can be
-(CH2CH2)-C(=O)NH2. In certain embodiments, Rb can be -(CH2CH2)-(indolyl). In certain embodiments, Rb can be -(CH2CH2)-(hydroxyphenyl).
[00271] In some embodiments, Rc can be -OH. In some embodiments, Rc can be - O(C1-C4 alkyl). In some embodiments, Rc can be -O(C1-C4 haloalkyl). In some embodiments, Rc can be -C(=O)NH2. In some embodiments, Rc can be unsubstituted C6-10 aryl. In some embodiments, Rc can be substituted C6-10 aryl. In some embodiments, Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S. In some embodiments, Rc can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S. In some embodiments, when a Rc moiety is indicated as substituted, the moiety can be substituted with one or more, for example, one, two, three, or four substituents E. In some embodiments, E can be -OH. In some embodiments, E can be C1-C4 alkyl. In some embodiments, E can be C1-C4 haloalkyl. In some embodiments, E can be -O(C1-C4 alkyl). In some embodiments, E can be -O(C1-C4 haloalkyl).
[00272] In some embodiments, RK can be hydrogen. In other embodiments, RK can be C1-C4 alkyl. For example, RK can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl. In some embodiments, RK can be selected from the group consisting of: unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein the substituted heteroaryl can substituted with one or more substituents Q, wherein each Q can independently selected from the group consisting of: -OH, C1-4 alkyl, C1-4 haloalkyl, halo, cyano, -O-(C1-4 alkyl), and -O- ( C1-4 haloalkyl). In certain mbodiments, RK can be benzothiophenyl. In other embodiments, RK can be pyridinyl substituted with one or more substituents Q. For example, RK can be methylpyridinyl, ethylpyridinyl cyanopyridinyl, chloropyridinyl, fluoropyridinyl, or bromopyridinyl.
[00273] In some embodiments, RG can be selected from the group consisting of hydrogen, C1-4 alkyl, and -(C1-4 alkyl)-C(=O)NH2. In certain embodiments, RG can be - (CH2CH2)-C(=O)NH2.
[00274] In some embodiments, Rf can be hydrogen. In other embodiments, Rf can be C1-4 alkyl. For example, Rf can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl. In some embodiments, Rf can be unsubstituted C6-C10 aryl. In other embodiments, Rf can be C6-C10 aryl substituted with 1-5 halo atoms. In certain embodiments, Rf can be phenyl substituted with 1-5 halo atoms. In certain embodiments, Rf can be fluorophenyl.
[00275] In some embodiments, U can be N. In other embodiments, U can be CRU.
[00276] In some embodiments, V can be S. In other embodiments, V can be NRV.
[00277] In some embodiments, RU can be hydrogen. In some embodiments, RU can be C1-4 alkyl. In other embodiments RU can be halo. For example, RU can be fluoro, chloro, bromo, or iodo. In still other embodiments, RU can be cyano.
[00278] In some embodiments, RV can be hydrogen. In other embodiments, RV can be C1-4 alkyl. For example, RVcan be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl. In some embodiments, Y and Z can each be C and X can be N. In other embodiments, Y and Z can each be C and X can be CH. [00279] In some embodiments, Ra can be hydrogen; Rb can be -(C1-4 alkyl)-Rc; Rc can be selected from the group consisting of: -C(=O)NH2, unsubstituted C6-10 aryl; substituted C6- 10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a Rc moiety indicated as substituted can be substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C1-C4 alkyl, C1-C4 haloalkyl, - O(C1-C4 alkyl), and -O(C1-C4 haloalkyl); RK can be selected from the group consisting of: unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein the substituted heteroaryl is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: -OH, C1-4 alkyl, C1-4 haloalkyl, halo, cyano, -O-(C1-4 alkyl), and -O-(C1-4 haloalkyl); RG is C1-4 alkyl or
-(C1-4 alkyl)-C(=O)NH2; Rf can be selected from the group consisting of hydrogen, unsubstituted phenyl, and phenyl substituted with 1-5 halo atoms; Y and Z each can be C; and X can be CH.
[00280] In some embodiments, Ra can be hydrogen; Rb can be -(CH2-CH2)-RC; Rc can be selected from the group consisting of: -C(=O)NH2, substituted phenyl and unsubstituted indolyl; wherein the substituted phenyl is substituted with one substituent E, wherein E can be -OH; RK can be selected from the group consisting of: unsubstituted benzothiohenyl and substituted pyridinyl; wherein the substituted pyridinyl is substituted with one substituent Q, wherein Q can be selected from the group consisting of: C1-4 alkyl, halo, and cyano; RG can be -(CH2CH2)-C(=O)NH2; Rf can be selected from the group consisting of hydrogen, phenyl, and fluorophenyl; Y and Z each can be C; and X can be CH.
[00281] In some embodiments, when V is S, Ra can be hydrogen or C1-C4 alkyl; Rb can be Rc or -(CH2-CH2)-Rc; Rc can be selected from the group consisting of: -C(=O)NH2; unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a Rc moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C1-C4 alkyl, and -O(C1-C4 alkyl); RK can be selected from the group consisting of: hydrogen, unsubstituted C1-6 alkyl; substituted C1-6 alkyl; -NH(C1-4 alkyl); and -N(C1-4 alkyl)2; wherein a RK moiety indicated as substituted is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: -OH, C1-4 alkyl, halo, cyano, and -O-(C1-4 alkyl; RG can be selected from the group consisting of hydrogen, C1-4 alkyl, and -(C1-4 alkyl)-C(=O)NH2; Rf can be selected from the group consisting of hydrogen, C1-4 alkyl, unsubstituted C6-C10 aryl, and C6-C10 aryl substituted with 1-5 halo atoms; U can be CRU; RU can be selected from the group consisting of hydrogen, C1-4 alkyl, halo, and cyano; Y and Z can each be C; and X can be N.
[00282] In some embodiments, when V is NRV, Ra can be hydrogen or C1-C4 alkyl; Rb can be Rc or -(CH2-CH2)-RC; Rc can be selected from the group consisting of: -C(=O)NH2; unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a Rc moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C1-C4 alkyl, C1-C4, and -O(C1-C4 alkyl); RK can be selected from the group consisting of: unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: -OH, C1-4 alkyl, halo, cyano, and -O-(C1-4 alkyl); RG can be selected from the group consisting of hydrogen, C1-4 alkyl, and -(C1-4 alkyl)-C(=O)NH2; Rf can be hydrogen; U can be N or CRU; RU can be selected from the group consisting of C1-4 alkyl, halo, and cyano; RV can be hydrogen or C1- C4 alkyl; Y and Z can each be C; and X can be N or CH.
[00283] In some embodiments, when RJ is -ORb; G can be N; - joining G and J can be a double bond; Rb can be -CH2CH2-RC; Rc can be
-C(=O)NH2; RK can unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; U can N; V can be NRv; Rv can be C1-C4 alkyl; Rf can be hydrogen; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I-B) can be 3-((2-(benzo[b]thiophen-3-yl)-9- isopropyl-9H-purin-6-yl)o\y)propan amide.
[00284] In some embodiments, when RJ is =O; G can be N substituted with RG; - joining G and J can be a single bond; RG can be -(C1-4 alkyl)-C(=O)NH2; RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; U can N; V can be NRv; Rv can be C1-C4 alkyl; Rf can be hydrogen; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I-B) can be 3-(2-(benzo[b]thiophen-3-yl)-9-isopropyl-6-oxo-6,9- dihydro- lH-purin- 1 -yl)propanamide.
[00285] In some embodiments, when RJ is -NRaRb; G can be N; . joining G and
J can be a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be substituted C6-10 aryl, substituted with one or more E, wherein E is -OH; RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; U can be CRU; Ru can be cyano; V can be NRv; Rv can be C1-C4 alkyl; Rf can be hydrogen; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I-B) can be 2-(benzo[b]thiophen-3-yl)-4-((4-hydroxyphenethyl)amino)-7- isopropyl-7H-pyrrolo[2,3-d]pyrimidine-5-carbonitrile.
[00286] In some embodiments, when RJ is -NRaRb; G can be N; . j pining G and J can be a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be unsubstituted C1-6 alkyl; U can be CRU; Ru can be hydrogen; V can be S; Rf can be phenyl; J can be C; X can be N; Y can be C; Z can be C. In some embodiments, the compound of Formula (I-B) can be N-(2-(1H-indol-3-yl)ethyl)-2-methyl-6- pheny lthieno [2,3 -d\ pyrimidin-4-amine.
[00287] In some embodiments, when RJ can be -NRaRb; G can be N; . joining G and J can be a double bond; Ra can be hydrogen; Rb can be
-CH2CH2-Rc; RC can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be hydrogen; U can be CRU; Ru can be hydrogen; V can be S; Rf can be fluorophenyl; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I-B) can be N-(2-(1H- indol-3-yl)ethyl)-6-(4-fluorophenyl)thieno[2,3-d]pyrimidin-4-amine.
[00288] In some embodiments, the compound of Formula (I-B), or a pharmaceutically acceptable salt thereof, can selected from the group consisting of: 3-((2-(benzo[b]thiophen-3-yl)-9-isopropyl-9H-purin-6-yl)o\y)propanamide:
3-(2-(benzo[b]thiophen-3-yl)-9-isopropyl-6-o\o-6.9-dihydro-1H-purin-1-yl)propanamide:
2-(benzo[b]thiophen-3-yl)-4-((4-hydro\yphenethyl)amino)-7-isopropyl-7H-pyrrolo[2.3- d]pyrimidine-5-carbonitrile:
N-(2-(1H-indol-3-yl)ethyl)-2-methyl-6-phenylthieno[2,3-d]pyrimidin-4-amine: and
N-(2-(1H-indol-3-yl)ethyl)-6-(4-riuorophenyl)thieno[2,3-d]pyrimidin-4-amine. Formula (I-C)
[00289] In still other embodiments provided herein, the compound of Formula (I) can have the structure of Formula (I-C):
Figure imgf000077_0001
(I-C), including pharmaceutically acceptable salts thereof, wherein: RJ can be -NRaRb; Ra can be hydrogen or C1-C4 alkyl; Rb can be Rc or -(C1-C4 alkyl)-Rc; Rc can be selected from the group consisting of: unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a Rc moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C1-C4 alkyl, C1-C4 haloalkyl, -O(C1-C4 alkyl), and -O(C1-C4 haloalkyl); RK can be selected from the group consisting of: hydrogen, unsubstituted C1-6 alkyl;-NH(C1-4 alkyl); -N(C1-4 alkyl)2, unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: -OH, C1-4 alkyl, C1-4 haloalkyl, halo, cyano, -O-(C1-4 alkyl), and -O-(C1-4 haloalkyl); A can be N or CH; B can be N or CH; Rgcan be selected from the group consisting of hydrogen, C1-4 alkyl, and -N(C1-4 alkyl)2; Y and Z can each be C; and X can be N or CH.
[00290] In some embodiments, RK can be -NH(C1-4 alkyl). For example, in some embodiments, RK can be -NH(CH3), -NH(CH2CH3), -NH(isopropyl), or -NH(sec-butyl). In some embodiments, RK can be unsubstituted benzothiophenyl. In other embodiments, RK can be substituted pyridinyl. For example, RK can be methylpyridinyl, ethylpyridinyl, cyanopyridinyl, chloropyridinyl, fluoropyridinyl, or bromopyridinyl.
[00291] In some embodiments, A can be N and B can be N. In other embodiments, A can be N and B can be CH. In still other embodiments, A can be CH and B can be N. In yet still other embodiments, A can be CH and B can be CH. [00292] In some embodiments, R8 can be hydrogen. In other embodiments, R8 can be -
N(C1-4 alkyl)2. In certain embodiments, R8can be
-N(CH3)2.
[00293] In some embodiments, Ra can be hydrogen; Rb can be -(C1-C4 alkyl)-Rc; Rc can be selected from the group consisting of: unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a Rc moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C1-C4 alkyl, C1-C4 haloalkyl, - O(C1-C4 alkyl), and -O(C1-C4 haloalkyl); RK can be selected from the group consisting of: - NH(C1-4 alkyl); unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein the substituted heteroaryl is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: -OH, C1-4 alkyl, C1-4 haloalkyl, halo, cyano, -O-(C1-4 alkyl), and -O-(C1-4 haloalkyl); and R8 can be hydrogen or -N(C1-4 alky 1)2. [00294] In some embodiments, Ra can be hydrogen; Rb can be -(C1-C4 alkyl)-Rc; Rc can be selected from the group consisting of: substituted phenyl and unsubstituted indolyl; wherein the substituted phenyl is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C1-C4 alkyl, C1-C4 haloalkyl, -O(C1-C4 alkyl), and -O(C1-C4 haloalkyl); RK can be selected from the group consisting of: -NH(C1-4 alkyl); unsubstituted benzothiophenyl; and substituted pyridinyl; wherein the substituted pyridinyl is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: -OH, C1-4 alkyl, C1-4 haloalkyl, halo, cyano, -O-(C1-4 alkyl), and -O-(C1-4 haloalkyl); and R8can be hydrogen or -N(C1-4 alkyl)2.
[00295] In some embodiments, Ra can be hydrogen; Rb can be -(CH2CH2)-RC; Rc can be selected from the group consisting of: substituted phenyl and unsubstituted indolyl; wherein the substituted phenyl is substituted with one substituent E, wherein E can be -OH; RK can be selected from the group consisting of: -NHfsec-butyl): unsubstituted benzothiohenyl, and substituted pyridinyl; wherein the substituted pyridinyl is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: C1-4 alkyl, halo, and cyano; and R8can be hydrogen or -N(CH3)2. [00296] In some embodiments, when A is C and B is C, RJ can be -NRaRb; G can be N; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be substituted C6- 10 aryl, substituted with one or more E, wherein E is -OH; or unsubstituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; R8 can be hydrogen; J can be C; X can be N; Y can be C; and Z is C.
[00297] In some embodiments, when RJ is -NRaRb; G can be N; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be substituted C6-10 aryl, substituted with one or more E, wherein E is -OH; RK is unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; A can be N; B can be N; R8 can be -N(C1-4 alkyl)2; J can be C; X can be N; Y can be C; and Z is C. In some embodiments, the compound of Formula (I-C) can be 4-(2-((2-(benzo[b]thiophen-3-yl)-8- (dimethylamino)pyrimido|5.4-6/|pyrimidin-4-yl)amino)ethyl)phenol.
[00298] In some embodiments, when RJ is -NRaRb; G can be N; Ra can be hydrogen Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q can be halo; A can be CH; B can be CH; R8 can be hydrogen; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I-C) can be N-( 2- (1H-indol-3-yl)ethyl)-2-(5-fluoropyridin-3-yl)quinazolin-4-amine.
[00299] In some embodiments, when RJ is -NRaRb; G is N; - joining G and J can be a double bond; Ra can be hydrogen Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more Q, wherein Q can be cyano; A can be CH; B can be CH; R8 can be hydrogen; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I-C) can be 5-(4-((2-(1H-indol-3-yl)ethyl)amino)quinazolin-2- yl)nicotinonitrile.
[00300] In some embodiments, when RJ is -NRaRb; G can be N; - joining G and J can be a double bond; Ra can be hydrogen Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; RK can be -NH(C1-4 alkyl); A can be CH; B can be CH; Rg can be hydrogen; J can be C; X can be N; Y can be C; and Z can be C. In some embodiments, the compound of Formula (I-C) can be N4-(2-(1H-indol-3-yl)ethyl)-N2-(sec-butyl)quinazoline-2, 4-diamine. [00301] In some embodiments, the compound of Formula (I-C), or a pharmaceutically acceptable salt thereof, can selected from the group consisting of:
4-(2-((2-(benzo[b]thiophen-3-yl)-8-(dimethylamino)pyrimido [5.4-d]pyrimidin-4- yl)amino)ethyl)phenol;
N-(2-(1H-indol-3-yl)ethyl)-2-(5-fluoropyridin-3-yl)quinazolin-4-amine:
5-(4-((2-(1H-indol-3-yl)ethyl)amino)quinazolin-2-yl)nicotinonitrile: and
N4-(2-(1H-indol-3-yl)ethyl)-N2-(sec-butyl)quinazoline-2, 4-diamine.
Formula (I-D)
[00302] In yet still other embodiments provided herein, the compound of Formula (I) can have the structure of Formula (I-D):
Figure imgf000080_0001
(I-D), including pharmaceutically acceptable salts thereof, wherein: RJ can be -NRaRb; Ra can be hydrogen or C1-C4 alkyl; Rb can be Rc or -(C1-4 alkyl)-Rc; Rc can be selected from the group consisting of: unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a Rc moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C1-C4 alkyl, C1-C4 haloalkyl, -O(C1-C4 alkyl), and -O(C1-C4 haloalkyl); RK can be selected from the group consisting of: unsubstituted C6-10 aryl; substituted G,-in aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: -OH, C1-4 alkyl, C1-4 haloalkyl, halo, cyano, -O-(C1-4 alkyl), and -O-(C1-4 haloalkyl); Rb can be hydrogen or C1-4 alkyl; D can be N or CH; Y can be N; Z can be C; and X can be N or CH. [00303] In some embodiments, Rh can be hydrogen. In other embodiments, Rh can be C1-4 alkyl. For example, Rh can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl or tert-butyl.
[00304] In some embodiments, D can be N. In other embodiments, D can be CH. [00305] In some embodiments, when D is N, Y can be N, Z can be C, and X can be N. In other embodiments, when D is N, Y can be N, Z can be C, and X can be CH. In some embodiments, when D is CH, Y can be N, Z can be C, and X can be N. In other embodiments, when D is CH, Y can be N, Z can be C, and X can be CH.
[00306] In some embodiments, Ra can be hydrogen; Rb can be -(C1-4 alkyl)-Rc; Rc can be selected from the group consisting of: unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a Rc moiety indicated as substituted is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C1-C4 alkyl, C1-C4 haloalkyl, - O(C1-C4 alkyl), and -O(C1-C4 haloalkyl); RK can be selected from the group consisting of: unsubstituted C6-10 aryl; substituted C6-10 aryl; unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; and substituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; wherein a RK moiety indicated as substituted is substituted with one or more substituents Q, wherein each Q can be independently selected from the group consisting of: -OH, C1-4 alkyl, C1-4 haloalkyl, halo, cyano, -O-(C1-4 alkyl), and -O-(C1-4 haloalkyl); and Rh can be hydrogen or C1-4 alkyl.
[00307] In some embodiments, Ra can be hydrogen; Rb can be -(C1-C4 alkyl)-Rc; Rc can be selected from the group consisting of: substituted phenyl and unsubstituted indolyl; wherein the substituted phenyl is substituted with one or more substituents E, wherein each E can be independently selected from the group consisting of: -OH, C1-C4 alkyl, C1-C4 haloalkyl, -O(C1-C4 alkyl), and -O(C1-C4 haloalkyl); RK can be unsubstituted benzothiophenyl; and Rb can be hydrogen or C1-4 alkyl.
[00308] In some embodiments, Ra can be hydrogen; Rb can be -(CH2-CH2)-RC; Rc can be selected from the group consisting of: substituted phenyl and unsubstituted indolyl; wherein the substituted phenyl is substituted with one substituent E, wherein E can be -OH; RK can be unsubstituted benzothiophenyl; and Rb can be hydrogen or C1-4 alkyl. [00309] In some embodiments, when D is N; RJ is -NRaRb; G can be N; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; or substituted C6-10 aryl, substituted with one or more E, wherein E is -OH; RK can be unsubstituted five- to ten- membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; Rb can be C1-4 alkyl; J can be C; X can be C; Y can be N; and Z can be C; wherein the valency of any carbon atom is filled as needed with hydrogen atoms.
[00310] In some embodiments, when RJ is -NRaRb; G can be N; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S or substituted C6-10 aryl, substituted with one or more E, wherein E is -OH; RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; D can be N; Rb can be C1-4 alkyl; J can be C; X can be C; Y can be N; and Z can be C; wherein the valency of any carbon atom is filled as needed with hydrogen atoms. In some embodiments, the compound of Formula (I-D) can beN-(2-(1H-indol-3-yl)ethyl)-6-(benzo[b]thiophen-3- yl)-3-isopropylimidazo[1,5-α]pyrazin-8-amine.
[00311] In some embodiments, when RJ is -NRaRb; G can be N; - joining G and J can be a double bond; Ra can be hydrogen; Rb can be -CH2CH2-RC; Rc can be substituted G,- lo aryl, substituted with one or more E, wherein E is -OH; RK can be unsubstituted five- to ten-membered heteroaryl having 1-4 atoms selected from the group consisting of O, N, and S; D can be N; Rb can be C1-4 alkyl; J can be C; X can be C; Y can be N; and Z can be C; wherein the valency of any carbon atom is filled as needed with hydrogen atoms. In some embodiments, the compound of Formula (I-D) can be 4-(2-((6-(benzo[b]thiophen-3-yl)-3- isopropybmidazo[1,5-α]pyrazin-8-yl)amino)ethyl)phenol.
[00312] In some embodiments, the compound of Formula (I-D), or a pharmaceutically acceptable salt thereof, can selected from the group consisting of:
N-(2-(1H-indol-3-yl)ethyl)-6-(benzo[b]thiophen-3-yl)-3-isopropylimidazo[1,5-α]pyrazin-8- amine; and 4-(2-((6-(benzo[b]thiophen-3-yl)-3-isopropylimidazo[1,5-α]pyrazin-8- yl)amino)ethyl)phenol.
[00313] The compounds provided herein may be enantiomerically pure, such as a single enantiomer or a single diastereomer, or be stereoisomeric mixtures, such as a mixture of enantiomers, e.g., a racemic mixture of two enantiomers; or a mixture of two or more diastereomers. As such, one of skill in the art will recognize that administration of a compound in its (R) form is equivalent, for compounds that undergo epimerization in vivo, to administration of the compound in its ( S) form. Conventional techniques for the preparation/isolation of individual enantiomers include synthesis from a suitable optically pure precursor, asymmetric synthesis from achiral starting materials, or resolution of an enantiomeric mixture, for example, chiral chromatography, recrystallization, resolution, diastereomeric salt formation, or derivatization into diastereomeric adducts followed by separation.
5.4. Isolation of NK Cells
[00314] Methods of isolating natural killer cells are known in the art and can be used to isolate the natural killer cells, e.g., NK cells produced using the three-stage method, described herein. For example, NK cells can be isolated or enriched, for example, by staining cells, in one embodiment, with antibodies to CD56 and CD3, and selecting for CD56 CD3 cells. In certain embodiments, the NK cells are enriched for CD56 CD3 cells in comparison with total cells produced using the three-stage method, described herein. NK cells, e.g., cells produced using the three-stage method, described herein, can be isolated using a commercially available kit, for example, the NK Cell Isolation Kit (Miltenyi Biotec). NK cells, e.g., cells produced using the three-stage method, described herein, can also be isolated or enriched by removal of cells other than NK cells in a population of cells that comprise the NK cells, e.g., cells produced using the three-stage method, described herein. For example, NK cells, e.g., cells produced using the three-stage method, described herein, may be isolated or enriched by depletion of cells displaying non-NK cell markers using, e.g., antibodies to one or more of CD3, CD4, CD14, CD19, CD20, CD36, CD66b, CD123, HLA DR and/or CD235a (glycophorin A). Negative isolation can be carried out using a commercially available kit, e.g., the NK Cell Negative Isolation Kit (Dynal Biotech). Cells isolated by these methods may be additionally sorted, e.g., to separate CDlla+ and CDlla- cells, and/or CD117+ and CD117- cells, and/or CD16+ and CD16 cells, and/or CD94+ and CD94+ In certain embodiments, cells, e.g., cells produced by the three-step methods described herein, are sorted to separate CD11a+ and CD11a- cells. In specific embodiments, CD11a+ cells are isolated. In certain embodiments, the cells are enriched for CD11a+ cells in comparison with total cells produced using the three-stage method, described herein. In specific embodiments, CD11a- cells are isolated. In certain embodiments, the cells are enriched for CD11a- cells in comparison with total cells produced using the three-stage method, described herein. In certain embodiments, cells are sorted to separate CD117+ and CD117- cells. In specific embodiments, CD117+ cells are isolated. In certain embodiments, the cells are enriched for CD117+ cells in comparison with total cells produced using the three-stage method, described herein. In specific embodiments, CD117- cells are isolated. In certain embodiments, the cells are enriched for CD117- cells in comparison with total cells produced using the three- stage method, described herein. In certain embodiments, cells are sorted to separate CD16+ and C D 16 cells. In specific embodiments, CD16+ cells are isolated. In certain embodiments, the cells are enriched for CD16+ cells in comparison with total cells produced using the three-stage method, described herein. In specific embodiments, C D 16 cells are isolated. In certain embodiments, the cells are enriched for CD 16- cells in comparison with total cells produced using the three-stage method, described herein. In certain embodiments, cells are sorted to separate CD94+ and CD 94 cells. In specific embodiments, CD94+ cells are isolated. In certain embodiments, the cells are enriched for CD94+ cells in comparison with total cells produced using the three-stage method, described herein. In specific embodiments, CD 94 cells are isolated. In certain embodiments, the cells are enriched for CD94- cells in comparison with total cells produced using the three-stage method, described herein. In certain embodiments, isolation is performed using magnetic separation. In certain embodiments, isolation is performed using flow cytometry.
[00315] Methods of isolating ILC3 cells are known in the art and can be used to isolate the ILC3 cells, e.g., ILC3 cells produced using the three-stage method, described herein. For example, ILC3 cells can be isolated or enriched, for example, by staining cells, in one embodiment, with antibodies to CD56, CD3, and CDlla, and selecting for CD56 CD3 C D 1 1 a cells. ILC3 cells, e.g., cells produced using the three-stage method, described herein, can also be isolated or enriched by removal of cells other than ILC3 cells in a population of cells that comprise the ILC3 cells, e.g., cells produced using the three-stage method, described herein. For example, ILC3 cells, e.g., cells produced using the three-stage method, described herein, may be isolated or enriched by depletion of cells displaying non-ILC3 cell markers using, e.g., antibodies to one or more of CD3, CD4, CDlla, CD14, CD19, CD20, CD36, CD66b, CD94, CD123, HLA DR and/or CD235a (glycophorin A). Cells isolated by these methods may be additionally sorted, e.g., to separate CD117+ and C D 1 17 cells. NK cells can be isolated or enriched, for example, by staining cells, in one embodiment, with antibodies to CD56, CD3, CD94, and CDlla, and selecting for CD56+CD3-CD94+CDlla+ cells. NK cells, e.g., cells produced using the three-stage method, described herein, can also be isolated or enriched by removal of cells other than NK cells in a population of cells that comprise the NK cells, e.g., cells produced using the three-stage method, described herein. In certain embodiments, the NK cells are enriched for C D561 C D3 C D941 C D 1 la+ cells in comparison with total cells produced using the three-stage method, described herein.
[00316] In one embodiment, ILC3 cells are isolated or enriched by selecting for CD56 CD3-CD1 la~ cells. In certain embodiments, the ILC3 cells are enriched for CD56 CD3-CD1 la~ cells in comparison with total cells produced using the three-stage method, described herein. In one embodiment, ILC3 cells are isolated or enriched by selecting for CD56 CD3-CD1 1 a C D 117+ cells. In certain embodiments, the ILC3 cells are enriched for CD56+CD3-CD11a CD117+ cells in comparison with total cells produced using the three-stage method, described herein. In one embodiment, ILC3 cells are isolated or enriched by selecting for CD56+CD3-CD11a CD117+CDIL1R1+ cells. In certain embodiments, the ILC3 cells are enriched for CD56+CD3-CDlla CD117+CDIL1R1+ cells in comparison with total cells produced using the three-stage method, described herein.
[00317] In one embodiment, NK cells are isolated or enriched by selecting for CD56 CD3-CD94 CD1 la+ cells. In certain embodiments, the NK cells are enriched for CD56 CD3-CD94 CD1 la+ cells in comparison with total cells produced using the three- stage method, described herein. In one embodiment, NK cells are isolated or enriched by selecting for CD56+CD3-CD94+CD11a+CDl 17 cells. In certain embodiments, the NK cells are enriched for CD56+CD3-CD94+CD11a+CDl 17 cells in comparison with total cells produced using the three-stage method, described herein.
[00318] Cell separation can be accomplished by, e.g., flow cytometry, fluorescence- activated cell sorting (FACS), or, in one embodiment, magnetic cell sorting using microbeads conjugated with specific antibodies. The cells may be isolated, e.g., using a magnetic activated cell sorting (MACS) technique, a method for separating particles based on their ability to bind magnetic beads (e.g., about 0.5-100 pm diameter) that comprise one or more specific antibodies, e.g., anti-CD56 antibodies. Magnetic cell separation can be performed and automated using, e.g., an AUTOMACS™ Separator (Miltenyi). A variety of useful modifications can be performed on the magnetic microspheres, including covalent addition of antibody that specifically recognizes a particular cell surface molecule or hapten. The beads are then mixed with the cells to allow binding. Cells are then passed through a magnetic field to separate out cells having the specific cell surface marker. In one embodiment, these cells can then isolated and re-mixed with magnetic beads coupled to an antibody against additional cell surface markers. The cells are again passed through a magnetic field, isolating cells that bound both the antibodies. Such cells can then be diluted into separate dishes, such as microtiter dishes for clonal isolation. 5.5. Placental Perfusate
[00319] NK cells and/or ILC3 cells, e.g., NK cell and/or ILC3 cell populations produced according to the three-stage method described herein may be produced from hematopoietic cells, e.g., hematopoietic stem or progenitors from any source, e.g., placental tissue, placental perfusate, umbilical cord blood, placental blood, peripheral blood, spleen, liver, or the like. In certain embodiments, the hematopoietic stem cells are combined hematopoietic stem cells from placental perfusate and from cord blood from the same placenta used to generate the placental perfusate. Placental perfusate comprising placental perfusate cells that can be obtained, for example, by the methods disclosed in U.S. Patent Nos. 7,045,148 and 7,468,276 and U.S. Patent Application Publication No. 2009/0104164, the disclosures of which are hereby incorporated in their entireties.
5.5.1. Cell Collection Composition
[00320] The placental perfusate and perfusate cells, from which hematopoietic stem or progenitors may be isolated, or useful in tumor suppression or the treatment of an individual having tumor cells, cancer or a viral infection, e.g., in combination with the NK cells and/or ILC3 cells, e.g., NK cell and/or ILC3 cell populations produced according to the three-stage method provided herein, can be collected by perfusion of a mammalian, e.g., human postpartum placenta using a placental cell collection composition. Perfusate can be collected from the placenta by perfusion of the placenta with any physiologically-acceptable solution, e.g., a saline solution, culture medium, or a more complex cell collection composition. A cell collection composition suitable for perfusing a placenta, and for the collection and preservation of perfusate cells is described in detail in related U.S. Application Publication No. 2007/0190042, which is incorporated herein by reference in its entirety.
[00321] The cell collection composition can comprise any physiologically-acceptable solution suitable for the collection and/or culture of stem cells, for example, a saline solution (e.g., phosphate-buffered saline, Kreb’s solution, modified Kreb’s solution, Eagle’s solution, 0.9% NaCl. etc), a culture medium (e.g., DMEM, H.DMEM, etc), and the like.
[00322] The cell collection composition can comprise one or more components that tend to preserve placental cells, that is, prevent the placental cells from dying, or delay the death of the placental cells, reduce the number of placental cells in a population of cells that die, or the like, from the time of collection to the time of culturing. Such components can be, e.g., an apoptosis inhibitor (e.g., a caspase inhibitor or JNK inhibitor); a vasodilator (e.g., magnesium sulfate, an antihypertensive drug, atrial natriuretic peptide (ANP), adrenocorticotropin, corticotropin-releasing hormone, sodium nitroprusside, hydralazine, adenosine triphosphate, adenosine, indomethacin or magnesium sulfate, a phosphodiesterase inhibitor, etc.),' a necrosis inhibitor (e.g., 2-(lH-Indol-3-yl)-3-pentylamino-maleimide, pyrrolidine dithiocarbamate, or clonazepam); a TNF-a inhibitor; and/or an oxygen-carrying perfluorocarbon (e.g., perfluorooctyl bromide, perfluorodecyl bromide, etc.).
[00323] The cell collection composition can comprise one or more tissue-degrading enzymes, e.g., a metalloprotease, a serine protease, a neutral protease, a hyaluronidase, an RNase, or a DNase, or the like. Such enzymes include, but are not limited to, collagenases (e.g., collagenase I, II, III or IV, a collagenase from Clostridium histolyticum, etc.),' dispase, thermolysin, elastase, trypsin, LIBERASE, hyaluronidase, and the like.
[00324] The cell collection composition can comprise a bacteriocidally or bacteriostatically effective amount of an antibiotic. In certain non-limiting embodiments, the antibiotic is a macrolide (e.g., tobramycin), a cephalosporin (e.g., cephalexin, cephradine, cefuroxime, cefprozil, cefaclor, cefixime or cefadroxil), a clarithromycin, an erythromycin, a penicillin (e.g., penicillin V) or a quinolone (e.g., ofloxacin, ciprofloxacin or norfloxacin), a tetracycline, a streptomycin, etc. In a particular embodiment, the antibiotic is active against Gram(+) and/or Gram(-) bacteria, e.g., Pseudomonas aeruginosa, Staphylococcus aureus, and the like.
[00325] The cell collection composition can also comprise one or more of the following compounds: adenosine (about 1 mM to about 50 mM); D-glucose (about 20 mM to about 100 mM); magnesium ions (about 1 mM to about 50 mM); a macromolecule of molecular weight greater than 20,000 daltons, in one embodiment, present in an amount sufficient to maintain endothelial integrity and cellular viability (e.g., a synthetic or naturally occurring colloid, a polysaccharide such as dextran or a polyethylene glycol present at about 25 g/1 to about 100 g/1, or about 40 g/1 to about 60 g/1); an antioxidant (e.g., butylated hydroxyanisole, butylated hydroxytoluene, glutathione, vitamin C or vitamin E present at about 25 mM to about 100 mM); a reducing agent (e.g., N-acetylcysteine present at about 0.1 mM to about 5 mM); an agent that prevents calcium entry into cells (e.g., verapamil present at about 2 mM to about 25 mM); nitroglycerin (e.g., about 0.05 g/L to about 0.2 g/L); an anticoagulant, in one embodiment, present in an amount sufficient to help prevent clotting of residual blood (e.g., heparin or hirudin present at a concentration of about 1000 units/1 to about 100,000 units/1); or an amiloride containing compound (e.g., amiloride, ethyl isopropyl amiloride, hexamethylene amiloride, dimethyl amiloride or isobutyl amiloride present at about 1.0 mM to about 5 mM).
5.5.2. Collection and Handling of Placenta
[00326] Generally, a human placenta is recovered shortly after its expulsion after birth. In one embodiment, the placenta is recovered from a patient after informed consent and after a complete medical history of the patient is taken and is associated with the placenta. In one embodiment, the medical history continues after delivery.
[00327] Prior to recovery of perfusate, the umbilical cord blood and placental blood are removed. In certain embodiments, after delivery, the cord blood in the placenta is recovered. The placenta can be subjected to a conventional cord blood recovery process. Typically a needle or cannula is used, with the aid of gravity, to exsanguinate the placenta (see, e.g., Anderson, U.S. Patent No. 5,372,581; Hessel et ctl, U.S. Patent No. 5,415,665).
The needle or cannula is usually placed in the umbilical vein and the placenta can be gently massaged to aid in draining cord blood from the placenta. Such cord blood recovery may be performed commercially, e.g., LifeBank Inc., Cedar Knolls, N.J., ViaCord, Cord Blood Registry and CryoCell. In one embodiment, the placenta is gravity drained without further manipulation so as to minimize tissue disruption during cord blood recovery.
[00328] Typically, a placenta is transported from the delivery or birthing room to another location, e.g., a laboratory, for recovery of cord blood and collection of perfusate.
The placenta can be transported in a sterile, thermally insulated transport device (maintaining the temperature of the placenta between 20-28 °C), for example, by placing the placenta, with clamped proximal umbilical cord, in a sterile zip-lock plastic bag, which is then placed in an insulated container. In another embodiment, the placenta is transported in a cord blood collection kit substantially as described in U.S. Patent No. 7,147,626. In one embodiment, the placenta is delivered to the laboratory four to twenty-four hours following delivery. In certain embodiments, the proximal umbilical cord is clamped, for example within 4-5 cm (centimeter) of the insertion into the placental disc prior to cord blood recovery. In other embodiments, the proximal umbilical cord is clamped after cord blood recovery but prior to further processing of the placenta.
[00329] The placenta, prior to collection of the perfusate, can be stored under sterile conditions and at either room temperature or at a temperature of 5 to 25 °C (centigrade). The placenta may be stored for a period of longer than forty eight hours, or for a period of four to twenty -four hours prior to perfusing the placenta to remove any residual cord blood. The placenta can be stored in an anticoagulant solution at a temperature of 5 °C to 25 °C (centigrade). Suitable anticoagulant solutions are well known in the art. For example, a solution of heparin or warfarin sodium can be used. In one embodiment, the anticoagulant solution comprises a solution of heparin (e.g., 1% w/w in 1:1000 solution). In some embodiments, the exsanguinated placenta is stored for no more than 36 hours before placental perfusate is collected.
5.5.3. Placental Perfusion
[00330] Methods of perfusing mammalian placentae and obtaining placental perfusate are disclosed, e.g., in Hariri, U.S. Patent Nos. 7,045,148 and 7,255,879, and in U.S. Application Publication Nos. 2009/0104164, 2007/0190042 and 20070275362, issued as U.S. Pat No. 8,057,788, the disclosures of which are hereby incorporated by reference herein in their entireties.
[00331] Perfusate can be obtained by passage of perfusion solution, e.g., saline solution, culture medium or cell collection compositions described above, through the placental vasculature. In one embodiment, a mammalian placenta is perfused by passage of perfusion solution through either or both of the umbilical artery and umbilical vein. The flow of perfusion solution through the placenta may be accomplished using, e.g., gravity flow into the placenta. For example, the perfusion solution is forced through the placenta using a pump, e.g., a peristaltic pump. The umbilical vein can be, e.g., cannulated with a cannula, e.g., a TEFLON® or plastic cannula, that is connected to a sterile connection apparatus, such as sterile tubing. The sterile connection apparatus is connected to a perfusion manifold. [00332] In preparation for perfusion, the placenta can be oriented in such a manner that the umbilical artery and umbilical vein are located at the highest point of the placenta. The placenta can be perfused by passage of a perfusion solution through the placental vasculature, or through the placental vasculature and surrounding tissue. In one embodiment, the umbilical artery and the umbilical vein are connected simultaneously to a pipette that is connected via a flexible connector to a reservoir of the perfusion solution. The perfusion solution is passed into the umbilical vein and artery. The perfusion solution exudes from and/or passes through the walls of the blood vessels into the surrounding tissues of the placenta, and is collected in a suitable open vessel from the surface of the placenta that was attached to the uterus of the mother during gestation. The perfusion solution may also be introduced through the umbilical cord opening and allowed to flow or percolate out of openings in the wall of the placenta which interfaced with the maternal uterine wall. In another embodiment, the perfusion solution is passed through the umbilical veins and collected from the umbilical artery, or is passed through the umbilical artery and collected from the umbilical veins, that is, is passed through only the placental vasculature (fetal tissue).
[00333] In one embodiment, for example, the umbilical artery and the umbilical vein are connected simultaneously, e.g., to a pipette that is connected via a flexible connector to a reservoir of the perfusion solution. The perfusion solution is passed into the umbilical vein and artery. The perfusion solution exudes from and/or passes through the walls of the blood vessels into the surrounding tissues of the placenta, and is collected in a suitable open vessel from the surface of the placenta that was attached to the uterus of the mother during gestation. The perfusion solution may also be introduced through the umbilical cord opening and allowed to flow or percolate out of openings in the wall of the placenta which interfaced with the maternal uterine wall. Placental cells that are collected by this method, which can be referred to as a “pan” method, are typically a mixture of fetal and maternal cells.
[00334] In another embodiment, the perfusion solution is passed through the umbilical veins and collected from the umbilical artery, or is passed through the umbilical artery and collected from the umbilical veins. Placental cells collected by this method, which can be referred to as a “closed circuit” method, are typically almost exclusively fetal.
[00335] The closed circuit perfusion method can, in one embodiment, be performed as follows. A post-partum placenta is obtained within about 48 hours after birth. The umbilical cord is clamped and cut above the clamp. The umbilical cord can be discarded, or can processed to recover, e.g., umbilical cord stem cells, and/or to process the umbilical cord membrane for the production of a biomaterial. The amniotic membrane can be retained during perfusion, or can be separated from the chorion, e.g., using blunt dissection with the fingers. If the amniotic membrane is separated from the chorion prior to perfusion, it can be, e.g., discarded, or processed, e.g., to obtain stem cells by enzymatic digestion, or to produce, e.g., an amniotic membrane biomaterial, e.g., the biomaterial described in U.S. Application Publication No. 2004/0048796. After cleaning the placenta of all visible blood clots and residual blood, e.g., using sterile gauze, the umbilical cord vessels are exposed, e.g., by partially cutting the umbilical cord membrane to expose a cross-section of the cord. The vessels are identified, and opened, e.g., by advancing a closed alligator clamp through the cut end of each vessel. The apparatus, e.g., plastic tubing connected to a perfusion device or peristaltic pump, is then inserted into each of the placental arteries. The pump can be any pump suitable for the purpose, e.g., a peristaltic pump. Plastic tubing, connected to a sterile collection reservoir, e.g., a blood bag such as a 250 mL collection bag, is then inserted into the placental vein. Alternatively, the tubing connected to the pump is inserted into the placental vein, and tubes to a collection reservoir(s) are inserted into one or both of the placental arteries. The placenta is then perfused with a volume of perfusion solution, e.g., about 750 ml of perfusion solution. Cells in the perfusate are then collected, e.g., by centrifugation.
[00336] In one embodiment, the proximal umbilical cord is clamped during perfusion, and, more specifically, can be clamped within 4-5 cm (centimeter) of the cord’s insertion into the placental disc.
[00337] The first collection of perfusion fluid from a mammalian placenta during the exsanguination process is generally colored with residual red blood cells of the cord blood and/or placental blood. The perfusion fluid becomes more colorless as perfusion proceeds and the residual cord blood cells are washed out of the placenta. Generally from 30 to 100 mL of perfusion fluid is adequate to initially flush blood from the placenta, but more or less perfusion fluid may be used depending on the observed results.
[00338] In certain embodiments, cord blood is removed from the placenta prior to perfusion (e.g., by gravity drainage), but the placenta is not flushed (e.g., perfused) with solution to remove residual blood. In certain embodiments, cord blood is removed from the placenta prior to perfusion (e.g., by gravity drainage), and the placenta is flushed (e.g., perfused) with solution to remove residual blood.
[00339] The volume of perfusion liquid used to perfuse the placenta may vary depending upon the number of placental cells to be collected, the size of the placenta, the number of collections to be made from a single placenta, etc. In various embodiments, the volume of perfusion liquid may be from 50 mL to 5000 mL, 50 mL to 4000 mL, 50 mL to 3000 mL, 100 mL to 2000 mL, 250 mL to 2000 mL, 500 mL to 2000 mL, or 750 mL to 2000 mL. Typically, the placenta is perfused with 700-800 mL of perfusion liquid following exsanguination.
[00340] The placenta can be perfused a plurality of times over the course of several hours or several days. Where the placenta is to be perfused a plurality of times, it may be maintained or cultured under aseptic conditions in a container or other suitable vessel, and perfused with a cell collection composition, or a standard perfusion solution (e.g., a normal saline solution such as phosphate buffered saline (“PBS”) with or without an anticoagulant (e.g., heparin, warfarin sodium, coumarin, bishy droxycoumarin), and/or with or without an antimicrobial agent (e.g., b-mercaptoethanol (0.1 mM); antibiotics such as streptomycin (e.g., at 40-100 mg/ml), penicillin (e.g., at 40 U/ml), amphotericin B (e.g., at 0.5 pg/ml). In one embodiment, an isolated placenta is maintained or cultured for a period of time without collecting the perfusate, such that the placenta is maintained or cultured for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, or 24 hours, or 2 or 3 or more days before perfusion and collection of perfusate. The perfused placenta can be maintained for one or more additional time(s), e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or more hours, and perfused a second time with, e.g., 700-800 mL perfusion fluid. The placenta can be perfused 1, 2, 3, 4, 5 or more times, for example, once every 1, 2, 3, 4, 5 or 6 hours. In one embodiment, perfusion of the placenta and collection of perfusion solution, e.g., placental cell collection composition, is repeated until the number of recovered nucleated cells falls below 100 cells/ml. The perfusates at different time points can be further processed individually to recover time-dependent populations of cells, e.g., total nucleated cells. Perfusates from different time points can also be pooled.
5.5.4. Placental Perfusate and Placental Perfusate Cells [00341] Typically, placental perfusate from a single placental perfusion comprises about 100 million to about 500 million nucleated cells, including hematopoietic cells from which NK cells and/or ILC3 cells, e.g., NK cells and/or ILC3 cells produced according to the three-stage method described herein, may be produced by the method disclosed herein. In certain embodiments, the placental perfusate or perfusate cells comprise CD34+ cells, e.g., hematopoietic stem or progenitor cells. Such cells can, in a more specific embodiment, comprise CD34 CD45 stem or progenitor cells, CD34+CD45+ stem or progenitor cells, or the like. In certain embodiments, the perfusate or perfusate cells are cryopreserved prior to isolation of hematopoietic cells therefrom. In certain other embodiments, the placental perfusate comprises, or the perfusate cells comprise, only fetal cells, or a combination of fetal cells and maternal cells.
5.6. NK Cells
5.6.1. NK Cells Produced by Three-Stage Method [00342] In another embodiment, provided herein is an isolated NK cell population, wherein said NK cells are produced according to the three-stage method described above. [00343] In one embodiment, provided herein is an isolated NK cell population produced by a three-stage method described herein, wherein said NK cell population comprises a greater percentage of CD3-CD56+ cells than an NK progenitor cell population produced by a three-stage method described herein, e.g., an NK progenitor cell population produced by the same three-stage method with the exception that the third culture step used to produce the NK progenitor cell population was of shorter duration than the third culture step used to produce the NK cell population. In a specific embodiment, said NK cell population comprises about 70% or more, in some embodiments, 75%, 80%, 85%, 90%,
95%, 98%, or 99% CD3-CD56+ cells. In another specific embodiment, said NK cell population comprises no less than 80%, 85%, 90%, 95%, 98%, or 99% CD3-CD56+ cells.
In another specific embodiment, said NK cell population comprises between 70%-75%, 75%- 80%, 80%-85%, 85%-90%, 90%-95%, or 95%-99% CD3-CD56+ cells.
[00344] In certain embodiments, said CD3-CD56+ cells in said NK cell population comprises CD3-CD56+ cells that are additionally NKp46+. In certain embodiments, said CD3-CD56+ cells in said NK cell population comprises CD3-CD56+ cells that are additionally CD16-. In certain embodiments, said CD3-CD56+ cells in said NK cell population comprises CD3-CD56+ cells that are additionally CD16+. In certain embodiments, said CD3-CD56+ cells in said NK cell population comprises CD3-CD56+ cells that are additionally CD94-. In certain embodiments, said CD3-CD56+ cells in said NK cell population comprises CD3-CD56+ cells that are additionally CD94+. In certain embodiments, said CD3-CD56+ cells in said NK cell population comprises CD3-CD56+ cells that are additionally CD11a+. In certain embodiments, said CD3-CD56+ cells in said NK cell population comprises CD3-CD56+ cells that are additionally NKp30+. In certain embodiments, said CD3-CD56+ cells in said NK cell population comprises CD3-CD56+ cells that are additionally CD161+. In certain embodiments, said CD3-CD56+ cells in said NK cell population comprises CD3-CD56+ cells that are additionally DNAM-1+. In certain embodiments, said CD3-CD56+ cells in said NK cell population comprises CD3-CD56+ cells that are additionally T-bet+.
[00345] In one embodiment, an NK cell population produced by a three-stage method described herein comprises cells which are CD117+. In one embodiment, an NK cell population produced by a three-stage method described herein comprises cells which are NKG2D+. In one embodiment, an NK cell population produced by a three-stage method described herein comprises cells which are NKp44+. In one embodiment, an NK cell population produced by a three-stage method described herein comprises cells which are CD244+. In one embodiment, an NK cell population produced by a three-stage method described herein comprises cells which express perforin. In one embodiment, an NK cell population produced by a three-stage method described herein comprises cells which express
EOMES. In one embodiment, an NK cell population produced by a three-stage method described herein comprises cells which express granzyme B. In one embodiment, an NK cell population produced by a three-stage method described herein comprises cells which secrete IFNy, GM-CSF and/or TNFa.
5.7. ILC3 Cells
5.7.1. ILC3 Cells Produced by Three-Stage Method [00346] In another embodiment, provided herein is an isolated ILC3 cell population, wherein said ILC3 cells are produced according to the three-stage method described above. [00347] In one embodiment, provided herein is an isolated ILC3 cell population produced by a three-stage method described herein, wherein said ILC3 cell population comprises a greater percentage of CD3-CD56+ cells than an ILC3 progenitor cell population produced by a three-stage method described herein, e.g., an ILC3 progenitor cell population produced by the same three-stage method with the exception that the third culture step used to produce the ILC3 progenitor cell population was of shorter duration than the third culture step used to produce the ILC3 cell population. In a specific embodiment, said ILC3 cell population comprises about 70% or more, in some embodiments, 75%, 80%, 85%, 90%,
95%, 98%, or 99% CD3-CD56+ cells. In another specific embodiment, said ILC3 cell population comprises no less than 80%, 85%, 90%, 95%, 98%, or 99% CD3-CD56+ cells.
In another specific embodiment, said ILC3 cell population comprises between 70%-75%, 75%-80%, 80%-85%, 85%-90%, 90%-95%, or 95%-99% CD3-CD56+ cells.
[00348] In certain embodiments, said CD3-CD56+ cells in said ILC3 cell population comprises CD3-CD56+ cells that are additionally NKp46 , In certain embodiments, said CD3-CD56+ cells in said ILC3 cell population comprises CD3-CD56+ cells that are additionally CD16-. In certain embodiments, said CD3-CD56+ cells in said ILC3 cell population comprises CD3-CD56+ cells that are additionally IL1R1+. In certain embodiments, said CD3-CD56+ cells in said ILC3 cell population comprises CD3-CD56+ cells that are additionally CD94-. In certain embodiments, said CD3-CD56+ cells in said ILC3 cell population comprises CD3-CD56+ cells that are additionally RORyt+. In certain embodiments, said CD3-CD56+ cells in said ILC3 cell population comprises CD3-CD56+ cells that are additionally CD 11a-. In certain embodiments, said CD3-CD56+ cells in said ILC3 cell population comprises CD3-CD56+ cells that are additionally T-bet+.
[00349] In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which are CD117+. In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which are NKG2D . In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which are NKp30 , In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which are CD244+. In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which are DNAM-1+. In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which express AHR. In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which do not express perforin. In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which do not express EOMES. In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which do not express granzyme B. In one embodiment, an ILC3 cell population produced by a three-stage method described herein comprises cells which secrete IL-22 and/or IL-8.
[00350] In certain aspects, cell populations produced by the three-stage method described herein comprise CDlla+ cells and CDlla- cells in a ratio of 50:1, 40:1, 30:1, 20:1, 10:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:10, 1:20, 1:30, 1:40, or 1:50. In certain aspects, a population of cells described herein comprises CD11a+ cells and CD11a- cells in a ratio of 50: 1. In certain aspects, a population of cells described herein comprises CD11a+ cells and CDlla- cells in a ratio of 20:1. In certain aspects, a population of cells described herein comprises CDlla+ cells and CDlla- cells in a ratio of 10:1. In certain aspects, a population of cells described herein comprises CD11a+ cells and CD11a- cells in a ratio of 5:1. In certain aspects, a population of cells described herein comprises CDlla+ cells and CD 11a- cells in a ratio of 1 : 1. In certain aspects, a population of cells described herein comprises CD11a+ cells and CD11a- cells in a ratio of 1 :5. In certain aspects, a population of cells described herein comprises CD11a+ cells and CD11a- cells in a ratio of 1 : 10. In certain aspects, a population of cells described herein comprises CD11a+ cells and CD11a- cells in a ratio of 1 :20. In certain aspects, a population of cells described herein comprises CDlla+ cells and CDlla- cells in a ratio of 1:50.
[00351] In certain aspects, cell populations described herein are produced by combining the CDlla+ cells with the CDlla- cells in a ratio of 50:1, 40:1, 30:1, 20:1, 10:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:10, 1:20, 1:30, 1:40, or 1:50 to produce a combined population of cells. In certain aspects, a combined population of cells described herein comprises CD11a+ cells and CD11a- cells combined in a ratio of 50: 1. In certain aspects, a combined population of cells described herein comprises CDlla+ cells and CD11a- cells combined in a ratio of 20: 1. In certain aspects, a combined population of cells described herein comprises CD11a+ cells and CD11a- cells combined in a ratio of 10:1. In certain aspects, a combined population of cells described herein comprises CD11a+ cells and CD11a- cells combined in a ratio of 5:1. In certain aspects, a combined population of cells described herein comprises CDlla+ cells and CD11a- cells combined in a ratio of 1:1. In certain aspects, a combined population of cells described herein comprises CD11a+ cells and CDlla- cells combined in a ratio of 1:5. In certain aspects, a combined population of cells described herein comprises CD11a+ cells and CD 11a- cells combined in a ratio of 1 : 10. In certain aspects, a combined population of cells described herein comprises CD11a+ cells and CDlla- cells combined in a ratio of 1:20. In certain aspects, a combined population of cells described herein comprises CDlla+ cells and CDlla- cells combined in a ratio of 1:50. [00352] In certain aspects, cell populations produced by the three-stage method described herein comprise NK cells and ILC3 cells in a ratio of 50:1, 40:1, 30:1, 20:1, 10:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:10, 1:20, 1:30, 1:40, or 1:50. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 50: 1. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 20: 1. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 10:1. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 5:1. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 1:1. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 1 :5. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 1 : 10. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 1 :20. In certain aspects, a population of cells described herein comprises NK cells and ILC3 cells in a ratio of 1:50.
[00353] In certain aspects, cell populations described herein are produced by combining the NK cells with the ILC3 cells in a ratio of 50:1, 40:1, 30:1, 20:1, 10:1, 5:1, 4:1, 3:1, 2:1, 1:1, 1:2, 1:3, 1:4, 1:5, 1:10, 1:20, 1:30, 1:40, or 1:50 to produce a combined population of cells. In certain aspects, a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 50: 1. In certain aspects, a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 20: 1. In certain aspects, a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 10: 1. In certain aspects, a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 5:1. In certain aspects, a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 1: 1. In certain aspects, a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 1 :5. In certain aspects, a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 1 : 10. In certain aspects, a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 1:20. In certain aspects, a combined population of cells described herein comprises NK cells and ILC3 cells combined in a ratio of 1:50.
5.8. Compositions Comprising NK Cells and/or ILC3 Cells
5.8.1. NK Cells and/or ILC3 Cells Produced Using The Three-Stage Method
[00354] In some embodiments, provided herein is a composition, e.g., a pharmaceutical composition, comprising an isolated NK cell and/or ILC3 cell population produced using the three-stage method described herein. In a specific embodiment, said isolated NK cell and/or ILC3 cell population is produced from hematopoietic cells, e.g., hematopoietic stem or progenitor cells isolated from placental perfusate, umbilical cord blood, and/or peripheral blood. In another specific embodiment, said isolated NK cell and/or ILC3 cell population comprises at least 50% of cells in the composition. In another specific embodiment, said isolated NK cell and/or ILC3 cell population, e.g., CD3-CD56+ cells, comprises at least 80%, 85%, 90%. 95%, 98% or 99% of cells in the composition. In certain embodiments, no more than 5%, 10%, 15%, 20%, 25%, 30%, 35%, or 40% of the cells in said isolated NK cell and/or ILC3 cell population are CD3-CD56+ cells. In certain embodiments, said CD3-CD56+ cells are CD 16-.
[00355] NK cell and/or ILC3 cell populations produced using the three-stage method described herein, can be formulated into pharmaceutical compositions for use in vivo. Such pharmaceutical compositions comprise a population of NK cells and/or ILC3 cells in a pharmaceutically-acceptable carrier, e.g., a saline solution or other accepted physiologically- acceptable solution for in vivo administration. Pharmaceutical compositions of the invention can comprise any of the NK cell and/or ILC3 cell populations described elsewhere herein. [00356] The pharmaceutical compositions of the invention comprise populations of cells that comprise 50% viable cells or more (that is, at least 50% of the cells in the population are functional or living). Preferably, at least 60% of the cells in the population are viable. More preferably, at least 70%, 80%, 90%, 95%, or 99% of the cells in the population in the pharmaceutical composition are viable.
[00357] The pharmaceutical compositions of the invention can comprise one or more compounds that, e.g., facilitate engraftment; stabilizers such as albumin, dextran 40, gelatin, hydroxyethyl starch, and the like.
[00358] When formulated as an injectable solution, in one embodiment, the pharmaceutical composition of the invention comprises about 1.25% HSA and about 2.5% dextran. Other injectable formulations, suitable for the administration of cellular products, may be used.
[00359] In one embodiment, the compositions, e.g., pharmaceutical compositions, provided herein are suitable for systemic or local administration. In specific embodiments, the compositions, e.g., pharmaceutical compositions, provided herein are suitable for parenteral administration. In specific embodiments, the compositions, e.g., pharmaceutical compositions, provided herein are suitable for injection, infusion, intravenous (IV) administration, intrafemoral administration, or intratumor administration. In specific embodiments, the compositions, e.g., pharmaceutical compositions, provided herein are suitable for administration via a device, a matrix, or a scaffold. In specific embodiments, the compositions, e.g., pharmaceutical compositions provided herein are suitable for injection. In specific embodiments, the compositions, e.g., pharmaceutical compositions, provided herein are suitable for administration via a catheter. In specific embodiments, the compositions, e.g., pharmaceutical compositions, provided herein are suitable for local injection. In more specific embodiments, the compositions, e.g., pharmaceutical compositions, provided herein are suitable for local injection directly into a solid tumor (e.g., a sarcoma). In specific embodiments, the compositions, e.g., pharmaceutical compositions, provided herein are suitable for injection by syringe. In specific embodiments, the compositions, e.g., pharmaceutical compositions, provided herein are suitable for administration via guided delivery. In specific embodiments, the compositions, e.g., pharmaceutical compositions, provided herein are suitable for injection aided by laparoscopy, endoscopy, ultrasound, computed tomography, magnetic resonance, or radiology.
[00360] In certain embodiments, the compositions, e.g., pharmaceutical compositions provided herein, comprising NK cells and/or ILC3 cells produced using the methods described herein, are provided as pharmaceutical grade administrable units. Such units can be provided in discrete volumes, e.g., 15 mL, 20 mL, 25 mL, 30 nL. 35 mL, 40 mL, 45 mL,
50 mL, 55 mL, 60 mL, 65 mL, 70 mL, 75 mL, 80 mL, 85 mL, 90 mL, 95 mL, 100 mL, 150 mL, 200 mL, 250 mL, 300 mL, 350 mL, 400 mL, 450 mL, 500 mL, or the like. Such units can be provided so as to contain a specified number of cells, e.g., NK cells and/or ILC3 cells, e.g., 1 x 104, 5 x 104, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108 or more cells per milliliter, or 1 x 104, 5 x 104, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108, 1 x 109, 5 x 109, 1 x 1010, 5 x 1010, 1 x 1011 or more cells per unit. In specific embodiments, the units can comprise about, at least about, or at most about 1 x 104, 5 x 104, 1 x 105, 5 x 105, 1 x 106, 5 x 106 or more NK cells and/or ILC3 cells per milliliter, or 1 x 104, 5 x 104, 1 x 105, 5 x 105, 1 x 106, 5 x 106, 1 x 107, 5 x 107, 1 x 108, 5 x 108, 1 x 109, 5 x 109, 1 x 1010, 5 x 1010, 1 x 1011 or more cells per unit. Such units can be provided to contain specified numbers of NK cells and/or ILC3 cells or NK cell and/or ILC3 cell populations and/or any of the other cells. In specific embodiments, the NK cells and ILC3 cells are present in ratios provided herein.
[00361] In another specific embodiment, said isolated NK cells and/or ILC3 cells in said composition are from a single individual. In a more specific embodiment, said isolated NK cells and/or ILC3 cells comprise NK cells and/or ILC3 cells from at least two different individuals. In another specific embodiment, said isolated NK cells and/or ILC3 cells in said composition are from a different individual than the individual for whom treatment with the NK cells and/or ILC3 cells is intended. In another specific embodiment, said NK cells have been contacted or brought into proximity with an immunomodulatory compound or thalidomide in an amount and for a time sufficient for said NK cells to express detectably more granzyme B or perforin than an equivalent number of natural killer cells, i.e. NK cells not contacted or brought into proximity with said immunomodulatory compound or thalidomide. In another specific embodiment, said composition additionally comprises an immunomodulatory compound or thalidomide. In certain embodiments, the immunomodulatory compound is a compound described below. See, e.g., U.S. Patent No. 7,498,171, the disclosure of which is hereby incorporated by reference in its entirety. In certain embodiments, the immunomodulatory compound is an amino-substituted isoindoline. In one embodiment, the immunomodulatory compound is 3 -(4-amino- 1-oxo- 1,3- dihydroisoindol-2-yl)-piperidine-2,6-dione; 3-(4'aminoisolindoline-r-one)-l-piperidine-2,6- dione; 4-(amino)-2-(2,6-dioxo(3-piperidyl))-isoindoline-l,3-dione; or 4-Amino-2-(2,6- dioxopiperidin-3-yl)isoindole-l,3-dione. In another embodiment, the immunomodulatory compound is pomalidomide, or lenalidomide. In another embodiment, said immunomodulatory compound is a compound having the structure
Figure imgf000100_0001
wherein one of X and Y is C=O, the other of X and Y is C=O or CH2 , and R2 is hydrogen or lower alkyl, or a pharmaceutically acceptable salt, hydrate, solvate, clathrate, enantiomer, diastereomer, racemate, or mixture of stereoisomers thereof. In another embodiment, said immunomodulatory compound is a compound having the structure
Figure imgf000100_0002
wherein one of X and Y is C=O and the other is CH2 or C=O;
R1 is H, (C1-C8 )alkyl, (C3-C7)cycloalkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, benzyl, aryl, (C0-C4)alkyl-(C1-C6)heterocycloalkyl, (C0-C4)alkyl-(C2-C5)heteroaryl, C(O)R3, C(S)R3, C(O)OR4, (C1-C8)alkyl-N(R6)2, (C1-C8)alkyl-OR5, (C1-C8)alkyl-C(O)OR5, C(O)NHR3, C(S)NHR3, C(O)NR3R3’, C(S)NR3R3’ or (C1-C8)alkyl-O(CO)R5;
R2 is H, F, benzyl, (C1-C8)alkyl, (C2-C8)alkenyl, or (C2-C8)alkynyl;
R3 and R3 are independently (C1-C8)alkyl, (C3-C7)cycloalkyl, (C2-C8)alkenyl, (C2- C8)alkynyl, benzyl, aryl, (C0-C4)alkyl-(C1-C6)heterocycloalkyl, (C0-C4)alkyl-(C2- C5)heteroaiyl, (C0-C8)alkyl-N(R6)2, (C1-C8)alkyl-OR5, (C1-C8)alkyl-C(O)OR5, (C1-C8)alkyl- O(CO)R5, or C(O)OR5;
R4 is (C1-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C1-C4)alkyl-OR5, benzyl, aryl, (C0-C4)alkyl-(C1-C6)heterocycloalkyl, or (C0-C4)alkyl-(C2-C5)heteroaryl;
R5 is (C1-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, benzyl, aryl, or (C2-C5)heteroaryl; each occurrence of R6 is independently H, (C1-C8)alkyl, (C2-C8)alkenyl. (C2- C8)alkynyl, benzyl, aryl, (C2-C5)heteroaryl, or (C0-C8)alkyl-C(O)O-R5 or the R6 groups can join to form a heterocycloalkyl group; n is 0 or 1; and
* represents a chiral-carbon center; or a pharmaceutically acceptable salt, hydrate, solvate, clathrate, enantiomer, diastereomer, racemate, or mixture of stereoisomers thereof. In another embodiment, said immunomodulatory compound is a compound having the structure
Figure imgf000101_0001
wherein: one of X and Y is C=O and the other is CH2 or C=O;
R is H or CH2OCOR’;
(i) each of R1, R2, R3, or R4, independently of the others, is halo, alkyl of 1 to 4 carbon atoms, or alkoxy of 1 to 4 carbon atoms or (ii) one of R1, R2, R3, or R4is nitro or -NHR5 and the remaining of R1, R2, R3, or R4 are hydrogen;
R5 is hydrogen or alkyl of 1 to 8 carbons
R6 hydrogen, alkyl of 1 to 8 carbon atoms, benzo, chloro, or fluoro;
R’ is R7-CHR10-N(R8R9);
R7 is m-phenylene or p-phenylene or -(CnH2n)- in which n has a value of 0 to 4; each of R8 and R9 taken independently of the other is hydrogen or alkyl of 1 to 8 carbon atoms, or R8 and R9 taken together are tetramethylene, pentamethylene, hexamethylene, or -CH2CH2X1CH2CH2- in which Xi is -O-, -S-, or -NH-;
R10 is hydrogen, alkyl of to 8 carbon atoms, or phenyl; and * represents a chiral-carbon center; or a pharmaceutically acceptable salt, hydrate, solvate, clathrate, enantiomer, diastereomer, racemate, or mixture of stereoisomers thereof.
[00362] In another specific embodiment, the composition additionally comprises one or more anticancer compounds, e.g., one or more of the anticancer compounds described below.
[00363] In a more specific embodiment, the composition comprises NK cells and/or ILC3 cells from another source, or made by another method. In a specific embodiment, said other source is placental blood and/or umbilical cord blood. In another specific embodiment, said other source is peripheral blood. In more specific embodiments, the NK cell and/or ILC3 cell population in said composition is combined with NK cells and/or ILC3 cells from another source, or made by another method in a ratio of about 100: 1, 95:5, 90: 10, 85: 15, 80:20, 75:25, 70:30, 65:35, 60:40, 55:45: 50:50, 45:55, 40:60, 35:65, 30:70, 25:75, 20:80, 15:85, 10:90, 5:95, 100:1, 95:1, 90:1, 85:1, 80:1, 75:1, 70:1, 65:1, 60:1, 55:1, 50:1, 45:1,
40:1, 35:1, 30:1, 25:1, 20:1, 15:1, 10:1, 5:1, 1:1, 1:5, 1:10, 1:15, 1:20, 1:25, 1:30, 1:35, 1:40, 1:45, 1:50, 1:55, 1:60, 1:65, 1:70, 1:75, 1:80, 1:85, 1:90, 1:95, 1:100, or the like.
[00364] In another specific embodiment, the composition comprises an NK cell and/or ILC3 cell population produced using the three-stage method described herein and either isolated placental perfusate or isolated placental perfusate cells. In a more specific embodiment, said placental perfusate is from the same individual as said NK cell and/or ILC3 cell population. In another more specific embodiment, said placental perfusate comprises placental perfusate from a different individual than said NK cell and/or ILC3 cell population. In another specific embodiment, all, or substantially all ( e.g ., greater than 90%, 95%, 98% or 99%) of cells in said placental perfusate are fetal cells. In another specific embodiment, the placental perfusate or placental perfusate cells, comprise fetal and maternal cells. In a more specific embodiment, the fetal cells in said placental perfusate comprise less than about 90%, 80%, 70%, 60% or 50% of the cells in said perfusate. In another specific embodiment, said perfusate is obtained by passage of a 0.9% NaCl solution through the placental vasculature.
In another specific embodiment, said perfusate comprises a culture medium. In another specific embodiment, said perfusate has been treated to remove erythrocytes. In another specific embodiment, said composition comprises an immunomodulatory compound, e g., an immunomodulatory compound described below, e.g., an amino-substituted isoindoline compound. In another specific embodiment, the composition additionally comprises one or more anticancer compounds, e.g., one or more of the anti cancer compounds described below. [00365] In another specific embodiment, the composition comprises an NK cell and/or ILC3 cell population and placental perfusate cells. In a more specific embodiment, said placental perfusate cells are from the same individual as said NK cell and/or ILC3 cell population. In another more specific embodiment, said placental perfusate cells are from a different individual than said NK cell and/or ILC3 cell population. In another specific embodiment, the composition comprises isolated placental perfusate and isolated placental perfusate cells, wherein said isolated perfusate and said isolated placental perfusate cells are from different individuals. In another more specific embodiment of any of the above embodiments comprising placental perfusate, said placental perfusate comprises placental perfusate from at least two individuals. In another more specific embodiment of any of the above embodiments comprising placental perfusate cells, said isolated placental perfusate cells are from at least two individuals. In another specific embodiment, said composition comprises an immunomodulatory compound. In another specific embodiment, the composition additionally comprises one or more anticancer compounds, e.g., one or more of the anticancer compounds described below.
[00366]
6. KITS
[00367] Provided herein is a pharmaceutical pack or kit comprising one or more containers filled with one or more of the compositions described herein, e.g., a composition comprising NK cells and/or ILC3 cells produced by a method described herein, e.g., NK cell and/or ILC3 cell populations produced using the three-stage method described herein. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
[00368] The kits encompassed herein can be used in accordance with the methods described herein, e.g., methods of suppressing the growth of tumor cells and/or methods of treating cancer, e.g., hematologic cancer, and/or methods of treating viral infection. In one embodiment, a kit comprises NK cells and/or ILC3 cells produced by a method described herein or a composition thereof, in one or more containers. In a specific embodiment, provided herein is a kit comprising an NK cell and/or ILC3 cell population produced by a three-stage method described herein, or a composition thereof.
7. EXAMPLES
7.1. Example 1: Three-stage method of producing natural killer cells from hematopoietic stem or progenitor cells
[00369] CD34+ cells are cultured in the following medium formulations for the indicated number of days, and aliquots of cells are taken for assessment of cell count, cell viability, characterization of natural killer cell differentiation and functional evaluation. [00370] Stage 1 medium: 90% Stem Cell Growth Medium (SCGM) (CellGro®), 10% Human Serum-AB, supplemented with 25 ng/mL or 250 ng/mL recombinant human thrombopoietin (TPO), 25 ng/mL recombinant human Flt3L, 27 ng/mL recombinant human stem cell factor (SCF), 25 ng/mL recombinant human IL-7, 0.05 ng/mL or 0.025 ng/mL recombinant human IL-6, 0.25 ng/mL or 0.125 ng/mL recombinant human granulocyte colony-stimulating factor (G-CSF), 0.01 ng/mL or 0.025 ng/mL recombinant human granulocyte-macrophage colony-stimulating factor (GM-CSF), 0.10% gentamicin, and 1 to 10μm StemRegenin-1 (SR-1) or other stem cell mobilizing agent.
[00371] Stage 2 medium: 90% SCGM, 10% Human Serum- AB, supplemented with 25 ng/mL recombinant human Flt3L, 27 ng/mL recombinant human SCF, 25 ng/mL recombinant human IL-7, 20 ng/mL recombinant human IL-15, 0.05 ng/mL or 0.025 ng/mL recombinant human IL-6, 0.25 ng/mL or 0.125 ng/mL recombinant human granulocyte colony-stimulating factor (G-CSF), 0.01 ng/mL or 0.025 ng/mL recombinant human granulocyte-macrophage colony-stimulating factor (GM-CSF), 0.10% gentamicin, and 1 to 10μm SRI or other stem cell mobilizing agent.
[00372] Stage 3 medium: 90% STEMMACS™, 10% Human Serum-AB, 0.025 mM 2- mercaptoethanol (55 mM), supplemented with 22 ng/mL recombinant human SCF, 1000 U/mL recombinant human IL-2, 20 ng/mL recombinant human IL-7, 20 ng/mL recombinant human IL-15, 0.05 ng/mL or 0.025 ng/mL recombinant human IL-6, 0.25 ng/mL or 0.125 ng/mL recombinant human granulocyte colony-stimulating factor (G-CSF), 0.01 ng/mL or 0.025 ng/mL recombinant human granulocyte-macrophage colony-stimulating factor (GM- CSF), and 0.10% gentamicin.
[00373] Cells are seeded at Day 0 at 3x104 cells/mL in Stage 1 media, and cells are tested for purity by a CD34+ and CD45+ count and viability by 7AAD staining. At Day 5 cells are counted and seeded to a concentration of 1 x105 cells/mL with Stage 1 medium. At Day 7 cells are counted and seeded to a concentration of 1 x 105 cells/mL with Stage 1 medium.
[00374] At Day 10, cells are counted and seeded to a concentration of 1 x 105 cells/mL in Stage 2 medium. At Day 12, cells are counted and seeded to a concentration of 3 x 105 cells/mL in Stage 2 medium. At Day 14, cells are counted and seeded in Stage 3 medium. Cells are maintained in Stage 3 media until day 35.
[00375] Alternatively, the following protocol is used through Day 14: Cells seeded at Day 0 at 7.5 x 103 cells/mL in Stage 1 media, and cells are tested for purity by a CD34+ and CD45+ count and viability by 7AAD staining. At Day 7 cells are counted and seeded to a concentration of 3 x 105 cells/mL with Stage 1 medium. At Day 9 cells are counted and seeded to a concentration of 3 x 105 cells/mL with Stage 2 medium. At Day 12, cells are counted and seeded to a concentration of 3 x 105 cells/mL in Stage 2 medium. At Day 14, cells are counted and seeded to a concentration of 3 x 105 cells/mL in Stage 2 medium.
[00376] Seeding of cells into at passage is performed either by dilution of the culture with fresh media or by centrifugation of cells and resuspension / addition of fresh media. [00377] For harvest, cells are spun at 400 xg for seven minutes, followed by suspension of the pellet in an equal volume of Plasmalyte A. The suspension is spun at 400*g for seven minutes, and the resulting pellet is suspended in 10% HSA (w/v), 60% Plasmalyte A (v/v) at the target cell concentration. The cells are then strained through a 70 pm mesh, the final container is filled, an aliquot of the cells are tested for viability, cytotoxicity, purity, and cell count, and the remainder is packaged.
7.2. Example 2: Selection of stem cell mobilizing agents for the expansion of NK cells
[00378] The following compounds were investigated for their ability to promote the expansion of NK cell populations in vitro:
4-(2-((2-(benzo[b]thiophen-3-yl)-6-(isopropylamino)pyrimidin-4-yl)amino)ethyl)phenol)
(“CRL1”)
Figure imgf000105_0001
Figure imgf000105_0002
4-(2-((2-(benzo[b]thiophen-3-yl)-7-isopropyl-6.7-dihydro-5H-pyrrolo[2,3-d]pyrimidin-4- yl)amino)ethyl)phenol (“CRL3”)
Figure imgf000106_0001
2-(benzo[b]thiophen-3-yl)-4-((4-hydro\yphenethyl)amino)-7-isopropyl-5.7-dihydro-6H- pyrrolo[2,3-d]pyriinidin-6-one (“CRL4”)
Figure imgf000106_0002
3-((2-(benzo[b]thiophen-3-yl)-9-isopropyl-9H-purin-6-yl)oxy)propanamide (“CRL5”)
Figure imgf000106_0003
4-(2-((2-(benzo[b]thiophen-3-yl)-8-(dimethylamino)pyrimido [5.4-d]pyrimidin-4- yl)amino)ethyl)phenol (“CRL6”)
Figure imgf000106_0004
5-(2-((2-(1H-indol-3-yl)ethyl)amino)-6-(sec-butylamino)pyrimidin-4-yl)nicotinonitrile (“CRL7”)
Figure imgf000107_0001
N-(2-(1H-indol-3-yl)ethyl)-2-methyl-6-phenylthieno[2,3-d]pyrimidin-4-amine (“CRL8”)
Figure imgf000107_0002
N-(2-(1H-indol-3-yl)ethyl)-6-(4-fluorophenyl)thieno[2,3-d]pyrimidin-4-amine (“CRL9”)
Figure imgf000107_0003
3-(2-(benzo[b] |thiophen-3-yl)-9-isopropyl-6-oxo-6.9-dihydro-1H-purin-1-yl)propanamide (“CRLIO”)
Figure imgf000107_0004
N-(2-(1H-indol-3-yl)ethyl)-2-(5-fluoropyridin-3-yl)quinazolin-4-amine (“CRLll”)
Figure imgf000108_0001
5-(4-((2-(1H-indol-3-yl)ethyl)amino)quinazolin-2-yl)nicotinonitrile (“CRL12”)
Figure imgf000108_0002
N4-(2-(1H-indol-3-yl)ethyl)-N2-(sec-butyl)quinazoline-2, 4-diamine (“CRL13”)
Figure imgf000108_0003
2-(benzo[b]thiophen-3-yl)-4-((4-hydroxyphenethyl)amino)-7-isopropyl-7H-pyrrolo[2,3- 6/|pyrimidine-5-carbonitrile (“CRL14”)
Figure imgf000108_0004
N-(2-(1H-indol-3-yl)ethyl)-6-(benzo[b]thiophen-3-yl)-3-isopropybmidazo[1,5-α]pyrazin-8- amine (“CRL15”)
Figure imgf000109_0001
4-(2-((6-(benzo[b]thiophen-3-yl)-3-isopropylimidazo[1,5-α]pyrazin-8-yl)amino)ethyl)phenol
(“CRL16”)
Figure imgf000109_0002
5-(4-((2-(1H-indol-3-yl)ethyl)amino)-7-isopropylthieno[2,3-d] pyrimidin-2-yl)nicotinonitrile (“CRL17”)
Figure imgf000109_0003
N-(2-(1H-indol-3-yl)ethyl)-2-(5-fluoropyridin-3-yl)-7-isopropylthieno[2,3-d]pyrimidin-4- amine (“CRL18”)
Figure imgf000109_0004
N-(2-(1H-indol-3-yl)ethyl)-2-(5-fluoropyridin-3-yl)furo[2,3-d]pyrimidin-4-amine (“CRL19”)
Figure imgf000110_0001
N-(2-(1H-indol-3-yl)ethyl)-2-(5-methylpyridin-3-yl)furo[2,3-d]pyrimidin-4-amine
Figure imgf000110_0002
N-(2-(1H-indol-3-yl)ethyl)-7-isopropyl-2-(5-methylpyridin-3-yl)thieno[2,3-d]pyrimidin-4- amine (“CRL21”)
Figure imgf000110_0003
5-(4-((2-(1H-indol-3-yl)ethyl)amino)furo[2,3-d]pyrimi din-2 -yl)nicotinonitrile (“CRL22”)
Figure imgf000110_0004
7.3. Example 3: Characterization of three-stage NK cells METHODS
[00379] UCB CD34+ cells were cultivated in presence of cytokines including thrombopoietin, SCF, Flt3 ligand, IL-7, IL-15 and IL-2 for 35 days to produce three-stage NK cells, as described in Example 1. Multi-color flow cytometry was used to determine the phenotypic characteristics of three-stage NK cells.
[00380] For biological testing, the compounds were provided to culture to evaluate their effects on NK cell expansion and differentiation. Specifically, donors of CD34+ cells (StemCell Technology) were thawed and expanded in vitro following NK culture protocol. During the first 14 days of the culture, each CRL compounds was dissolved in DMSO and added to the culture at 10 mM concentration. SRI (at 10 pM) served as a positive control compound, while DMSO alone without any compound served as a negative control. At the end of the culture on Day 35, cell expansion, natural killer (NK) cell differentiation and cytotoxicity of the cells against K562 tumor cell line were characterized. Due to the large number of the compounds, the testing was performed in two experiments, CRLl-11 and CRL 12-22. The same donors were used for each experiment. Positive and negative controls were also included in both experiments.
Results
[00381] Cell expansion data showed that 20 out of the 22 compounds supported NK expansion at 10 pM concentration. Except for CRL7 and CRL 13, the rest of the compounds all resulted in a NK expansion of 2,000 ~ 15,000 fold over 35 days (FIG. 1 and FIG. 2). Among all the compounds, CRL 19, 20 and 22 supported cell expansion the best, and they demonstrated a similar level of expansion compared to SRI at Day 35 (FIG. 3). CD34 cell expansion at Day 14 of the culture showed a similar trend that most of the compounds supported CD34 cells expansion, and CRL19, 20 and 22 achieved the highest CD34 cell expansion at Day 14 (FIG. 4).
[00382] Cytotoxicity assay was run using compound cultured cells against K562 tumor cells at 10: 1 effector to target ratio (FIGS. 5 A - 5B) to evaluate cell functions. The results showed that the cells cultured with compounds killed 30-60% of K562 cells at 10:1 E:T ratio, indicating that the cells present NK functions. For both donors, cells cultured with CRL 17, 18, 19 and 21 demonstrated similar or greater killing activities compared to those cultured with SRI .
Conclusions: [00383] In summary, we found that all the compounds except CRL7 and CRL13 supported PNK-007 expansion and differentiation. Expansion with the compounds ranged from 2,000 ~ 15, 000 fold over 35 days, and the culture achieved more than 70% of NK cells. Among these compounds, CRL 19, 20 and 22 demonstrated very similar expansion, differentiation and cytotoxicity profiles as SRI for PNK-007 culture. CRL 17, 18, and 21 resulted in slightly less expansion compared to SRI but increased CD56+/CDlla+ subpopulation, and also increased killing activities of the cells.
7.4 Example 4: Further characterization of three-stage NK cells METHODS
[00384] Cells: Frozen PBMC were acquired from Stem Cell Technologies. Peripheral blood derived NKs (PB-NK) cells were isolated from fresh blood of healthy donors using the Human NK Cell Enrichment Kit (Stem Cell Technologies) according to manufacturer’s instructions. CYNK cells were generated from umbilical cord blood-derived CD34+ stem cells (Ref: Zhang et al. J Immunother Cancer. 2015). Briefly, the CD34+ cells were cultivated in the presence of cytokines including thromobopoietin, SCF, Flt3 ligand, IL-7, IL-15 and IL- 2 for 35 days. PBNK and CYNK cells were cryopreserved until analysis.
[00385] Magnetic-activated cell sorting: PNK cells were stained with PE Mouse Anti- Human CD11a (BD) and CD11a+ PNK cells concentrated using anti-PE MicroBeads according to manufacturer’s instructions (Miltenyi Biotec).
[00386] Single cell RNA sequencing: CYNK cells were combined with PB-NK at 1 : 1 ratio and gene expression analyzed on single cell level using 10X Genomics Chromium platform and Illumina sequencing. Bioinformatics analysis utilized 10X Genomics Cell Ranger analysis pipeline.
[00387] Flow Cytometry: Cryopreserved cells were rapidly thawed in a 37°C water bath and washed once in RPMI1640 + 10% hiFBS (heat inactivated Fetal Bovine Serum, Gibco), followed by LIVE/DEAD™ Fixable Aqua Stain in PBS. Cells were washed with FACS buffer (PBS + 2% FBS) followed by incubation in blocking solution (Brilliant Stain buffer, Mouse IgG2a isotype k control and Human BD Fc Block (all from BD)). Cells were washed with FACS buffer and incubated with fluorophore-coupled antibodies in FACS buffer for 25 min on ice. Cells were washed with FACS buffer before analysis on Fortessa X20 flow cytometer (BD).
[00388] qRT-PCR: RNA was isolated from cells using Quick-RNA Miniprep kit
(Qiagen) according to the manufacturer’s instructions. cDNA was synthesized using
Superscript IV Reverse Transcriptase (Thermo Fisher Scientific) in a standard reaction. RT- PCR was performed using Taqman Gene expression assays (Applied Biosystems). Expression levels were calculated relative to GAPDH (Hs02758991) using the AACt method.
RESULTS
[00389] CYNK cells efficiently kill various tumor cell lines in vitro, however, the mechanisms CYNK cells use to induce cell death remains poorly understood (rel). To elucidate on the activating NK cell receptors, the intracellular signaling pathways and molecular mechanisms CYNK cells employ to carry out their functional roles, we used single-cell RNA sequencing (scRNAseq) as an unbiased approach to compare CYNK cells to peripheral blood NK cells (PB-NK) (FIG. 6A). Unbiased transcriptional clustering revealed two distinct signatures differentiating between CYNK and PB-NK cells (FIG. 6B). Tables 1 and 2 list top 50 upregulated genes per cluster in PB-NK and CYNK cells, respectively. The gene set expressed higher in PB-NK cells included genes associated with NK cell functional roles, including FGFBP2, granzymes (GZMH, GZMM), CXCR4, KLRFl, KLF2, IFNG (Table 1).
□ FGFBP2, encoding fibroblast growth factor-binding protein, is known to be secreted by cytotoxic lymphocytes.
□ Granzymes are a group of serine proteases which are stored in the cytotoxic granules of NK cells and cytotoxic T lymphocytes (ref). While GzmA and GzmB induce target cell death upon release to their cytoplasm and have been extensively studied, less is known about the functional role of GzmH, GzmK and GzmM.
□ CXCR4 regulates NK cell homing to bone marrow.
□ KLRFl encodes NKp80, an activating C-type lectin-like immunoreceptor that is activated upon binding to activation-induced C-type lectin (AICL), inducing NK cell cytotoxicity and cytokine secretion.
□ Transcription factor KLF2 that regulates both NK cell proliferation and survival.
□ NK cell-derived IFN-g (IFNG gene) is a key immunoregulatory factor secreted from activated NK cells that promotes adaptive immune response by modulating dendritic cell and T cell responses.
Table 1. Top 50 upregulated genes per PB-NK cluster.
Feature CYNK PB-NK PB-NK Log2 PB-NK P-
Feature ID
Name Average Average Fold Change Value
Figure imgf000113_0001
Figure imgf000115_0002
[00390] Top differentially expressed genes in CYNK cluster that are encode factors associated with NK cell functional role include surface receptors and co-receptors (CD96, NCR3, CD59, KLRC1), TNFSF10, immune checkpoint genes (TNFRSF18, TNFRSF4, HAVCR2), NK cell receptor adaptor molecule genes (FCER1G and LAT2) (Table 2).
Table 2. Top 50 upregulated genes per CYNK cluster.
CYNK Log2
Feature PBNK CYNK CYNK P-
Feature ID Fold
Name Average Average Value
Change
Figure imgf000115_0001
Figure imgf000116_0001
[00391] To better understand how the cytotoxic response is initiated in CYNK cells, we specifically analyzed the expression of manually chosen genes encoding well characterized proteins leading from target detection to a cytolytic response, with main focus on NK cell receptors and adaptor molecule (Table 3). Differential gene expression analysis showed high expression of the two key cytotoxic molecules perforin (PRF1) and granzyme B (GZMB) in CYNK cells. Similarly, most receptors that were differentially expressed between CYNK and PB-NK cells, with the exception of KLRFl (encoding NKp80), were higher expressed on CYNK cells. Expression of selected NK cell effector and receptor genes is visualized on tSNE plots in FIG. 6C. Elevated expression of genes encoding components of the NK cell cytotoxic machinery correlate well with the high cytotoxic activity of CYNK cells against a broad range of target cells.
Table 3. Top differentially expressed genes encoding factors regulating NK cell cytolytic function. Genes that had <1 count per cell across the entire cluster were excluded.
CYNK
Feature CYNK PBNK Log2 CYNK P-
Feature ID Alias
Name Average Average Fold Value
Change
Figure imgf000116_0002
Figure imgf000117_0001
[00392] We next analyzed the transcriptional profile of CYNK and PB-NK cells by quantitative real-time PCR (qRT-PCR) focusing on selected NK cell-associated genes that were highly and/or differentially expressed in the scRNAseq dataset (FIG. 7). RNA was extracted from freshly thawed naive cells post isolation or culture. qRT-PCR demonstrated high expression of CD69, KLRK1 and KLRB1 relative to the housekeeping gene GAPDH in both CYNK and PB-NK cells, whereas, KLRKl and KLRBl, encoding for NKG2D and CD161/KLRB1, respectively, were significantly higher expressed in PB-NK cells. Significant differential expression of NKp80, encoded by KLRFl gene, earlier seen by scRNAseq (Table 3), was confirmed by qRT-PCR. Similarly, KLRDl was higher expressed on PB-NK compared to CYNK cells. Together, the data show higher expression of the inhibitory killer cell lectin-like receptor (KLRBl, KLRDl, KLRFl) expression on PB-NK cells when compared to CYNK cells. The two C-type lectin receptor genes KLRCl and KLRC2, encoding the inhibitory NKG2A and the activating NKG2C, were higher expressed in CYNK cells. Of the natural cytotoxicity receptors (NCRs), only NCR2 (encoding NKp44) was differentially expressed with high expression in CYNK cells and almost no expression in PB- NK cells. Two co-activating NK cell receptor genes CD244 (2B4) and CD226 (DNAM-1) were slightly higher expressed in PB-NK compared to CYNK cells. Alongside the typical ligand-activated NK cell receptor genes, we also analyzed the expression of FCGR3A encoding an Fc receptor CD16 that is required for antibody-dependent cell-mediated cytotoxicity. Whereas scRNAseq data demonstrated no significant differential expression of FCGR3A, by qRT-PCR it was highly expressed in the PB-NK cells and at a very low level in CYNK cells. The expression of two genes TNFRSF18 and TNFSF10 that were highly differentially expressed by scRNAseq and elevated in the CYNK cluster, were also analyzed by qRT-PCR. The PCR data confirms high expression of these genes encoding for GITR and TRAIL, respectively, on CYNK cells relative to low level expression in PB-NK cells.
[00393] Lastly, we characterized CYNK cells relative to PB-NK by surface protein expression using flow cytometry. Antibodies targeting various NK cell receptors were chosen based on the transcriptional characterization by scRNAseq and qRT-PCR (Tables 1-3, GIG. 6 and FIG. 7). NK cells express high level of the NK cell marker CD56 and lack the expression of T cell, B cell and myeloid cell markers CD3, CD19 and CD14, respectively (FIG. 8). Whereas a majority of PB-NK cells express CD56 at a low level, a small subset of PB-NK cells express CD56 at a level seen in CYNK cells (FIG. 9). NCR analysis demonstrated a high expression of NKp44 in CYNK cells, whereas, NKp44 was expressed at a low level in PB-NK, corresponding well to our transcriptional analysis (FIG. 7). NKp80, on the other hand, was expressed on PB-NK cell and little on CYNK, also confirming the transcriptional data of KLRFl expression (Table 1 and FIG. 7). CD 16 was virtually not expressed on CYNK cells, whereas the majority of PB-NK cells expressed CD16 at a high level. CD16 protein expression, therefore, also corresponds well to transcriptional analysis (Table 1 and FIG. 7). The expression of killer cell lectin-like receptors was comparable between CYNK and PB- NK cells, with CYNK cells demonstrating higher mean fluorescence intensity compared to PB-NK cells for NKG2D, NKG2C, CD94 (NKG2C) and NKG2A. GITR, a checkpoint inhibitor molecule, encoded by TNFRSF18, was not expressed on PB-NK cells but highly on all CYNK cells, correlating well to qRT-PCR data.
[00394] We used the flow cytometry dataset (FIG. 8 and FIG. 9) to perform an unbiased analysis of the surface marker expression on CYNK and PB-NK cell populations (FIG. 10). Antibody-stained CYNK and PBMC cells were mixed for acquisition and analyzed by flow cytometry. It is evident from the tSNE plots that CYNK and PB-NK cells cluster separately from each other and other peripheral blood cells when looking at the localization of CD56- and CD3/CD14/CD 19-positive cells on the plot. High expression of NKp44 (CD336) and GITR (CD357) enable the identification of CYNK cells as GITR is virtually not expressed in any cell type in the PBMC subsets. PB-NK cells on the other hand, highly express CD 16 and NKp80 that are not expressed on CYNK cells. Altogether, we have identified cell surface markers that allow to distinguish CYNK cells from PB-NK with high confidence.
7.5 Example 5: Cleavage Resistant CD16 Expressing NK Cells [00395] Celularity, Inc. is developing human placental hematopoietic stem cells- derived, cryopreserved, off-the shelf, ex-vivo expanded and allogenic Natural killer (PNK) cells for various hematological malignancies and solid tumors. NK cells play a central role in antibody dependent cell mediated cytotoxicity (ADCC) through Fc receptor CD16 in monoclonal antibody mediated anti -tumor therapies. Two allelic forms of CD 16 have been identified with the 158Val/Val form has shown to have higher IgG binding affinity comparing with the 158Phe/Phe form. The high IgG binding allele are found in about 10-20% of the normal population. In addition, activation of NK cells induces CD16 shedding by matrix metalloprotease ADAM17 at 197Ser, thus limiting ADCC responses. A single mutation (Serl97Pro) prevents CD 16 shedding and increases ADCC activity in NK cells. Since the antibody binding affinity and CD 16 expression of PNK could vary with different donors, we hypothesize that expressing a high affinity (158Val) and proteinase cleavage resistant (197Pro) CD16 variant (CD16VP) augments anti-tumor ADCC activity.
Methods:
[00396] Lentivirus expressing CD16VP was used to transduce human placental CD34+ cells. After transduction, the cells were cultured in the presence of cytokines including thrombopoietin, SCF, Flt3 ligand, IL-7, IL-15 and IL-2, for 35 days to generate PNK- CD16VP cells. Non-transduced PNK cells (NT) served as a control. Expression of CD16VP was evaluated by activating cells with PMA/ionomycin to induce CD16 cleavage (CD16 shedding assay) followed by immunostaining with CD 16 antibody and analyzed using flowcytometry. ADCC of PNK-CD16VP cells was assessed against Daratumumab (anti- CD38) or Rituximab (anti-CD20) opsonized lymphoma cell lines at various effector to target (E:T) ratios. IgG was used as ADCC control. In vivo anti-tumor activity was assessed in a Daudi disseminated Xenograft model in NSG mice. Luciferase-expressing Daudi cells (3x106) were intravenously (IV) administered at day 0, followed by PNK-CD16VP cells (10x106) IV at day 1 and day 3, and Daratumumab at day 3. Tumor burden in mice was monitored by Bioluminescence Imaging (BLI). Statistical differences between the groups were calculated using paired t-test using Prism.
[00397] Cell culture: Human placental CD34+ cells were isolated and cultured in the presence of cytokines including thrombopoietin, SCF, Flt3 ligand, IL-7, IL-15 and IL-2, for 35 days to generate NK cells.
[00398] Cell Expansion and Characterization: Cell expansion was recorded during the culture process. On day 35, CD16VP cells were evaluated for NK surface markers
CD56+/CD3-, and CD16, using flow cytometry. [00399] CD16VP Shedding Assay: Expression of CD16VP was evaluated by activating cells with PMA/ionomycin to induce CD 16 cleavage followed by immunostaining with CD 16 antibody and analyzed using flow cytometry.
[00400] In vitro ADCC Assay: ADCC activity of CD16VP cells was assessed against
Daratumumab (anti-CD38) or Rituximab (anti-CD20) opsonized lymphoma cell lines at various effector to target (E/T) ratios. IgG was used as ADCC control. In sustained ADCC assay, CD16VP cells were treated with PMA/ionomycin and then evaluated for ADCC activity as described above.
[00401] Animal Study: In vivo anti-tumor activity was assessed in a Daudi disseminated Xenograft model in NSG mice. Luciferase-expressing Daudi cells (3x106) were intravenously (IV) administered at day 0, followed by CD16VP cells (10x106) IV at day 1 and day 3, and Daratumumab at day 3. Tumor burden in mice was monitored by Bioluminescence Imaging (BLI).
[00402] Statistical Analysis: Statistical analysis was performed using Prism/Excel program. Data are presented as mean ± standard deviation. Paired or unpaired two-tailed Student’s test were used for comparing two groups.
Results:
[00403] Lentiviral transduction of CD16VP achieved high expression efficiency in multiple placental CD34+ donors. These cells expanded [7095 ± 2998 folds (n=8)] and differentiated into PNK cells (>90% CD56+CD3-) at day 35. PNK-CD16VP expressed 64.6 ± 10.3% (n=8) of CD16, while the NT expressed 12.1± 3.3% (n=8) CD16. PMA/ionomycin induced >89% shedding of CD16 in NT cells, while significantly less (<11%) CD16 shedding was observed in PNK-CD16VP cells. These results indicated that CD16VP was expressed and maintained throughout the culture process. In vitro ADCC assay demonstrated improved anti -tumor activity of PNK-CD16VP cells over NT cells against Daratumumab or Rituximab opsonized lymphoma cell lines. At 10:1 E:T ratio PNK-CD16VP cells elicited higher cytotoxicity compared to NT: 47±13% against Daratumumab opsonized Daudi cells versus 25±5% (n=5; p<0.05); 30±13% against Daratumumab opsonized HS-Sultan cells versus 21±14% (n=3; p<0.05); 30±7% against Daratumumab opsonized Sudhl6 cells versus 16±10% (n=3; p<0.05). Improved ADCC activities in PNK-CD16VP were also observed in other cell lines including Raji, and Sudhl4 with Daratumumab and Rituximab antibodies. [00404] In this study, we genetically modified placental CD34+ cells to over-express CD16VP and evaluated the phenotype and functions of CD16VP cells regarding enhancement of ADCC and cleavage resistance. [00405] -7000-fold expansion and >90% NK purity were achieved by the process.
[00406] -65% of CD16VP expression was maintained during the culture.
[00407] CD16VP expression was shown to be resistant to shedding after activation.
[00408] CD16VP cells demonstrated enhanced ADCC in vitro against lymphoma cell lines in combination with Daratumumab or Rituximab.
[00409] CD16VP resistance to activation induced shedding supported sustained killing in vitro.
[00410] CD16VP cells showed in vivo anti-tumor activities at early time points in an
ADCC lymphoma model.
[00411] CD16VP provides a promising approach to augment the anti-tumor activities in combination with monoclonal antibodies. Further investigation is perused to support [00412] development of CD16VP in combination with therapeutic antibodies for various hematological malignancies and solid tumors.
[00413] PNK-CD16VP were used to test anti-tumor ADCC in vivo using a disseminated Daudi Xenograft model. The preliminary data demonstrated that PNK-CD16VP combined with Daratumumab reduced BLI signal (>50%) compared to vehicle or Daratumumab alone at day 10 after treatment. This observation suggested that PNK-CD16VP demonstrated in vivo ADCC anti-tumor activity.
Conclusions:
[00414] In this study, we genetically modified PNK to express high affinity and cleavage resistant CD16 variant using lentivirus. The PNK-CD16VP cells demonstrated enhanced ADCC function against lymphoma cell lines in vitro and in vivo. Further development of PNK-CD16VP for immune-oncology therapeutics is warranted.
[00415]
7.6 Example 6: Cleavage Resistant CD16 Expressing CYNK Cells [00416] CYNK cells were transduced with a CD16VP lentivirus and expanded as set forth above followed by analysis of cell surface marker expression, CD 16 evpression and CD16 shedding. See, FIG. 15A, FIG. 15B, FIG. 16, and FIG. 17.
Table 4: NK Surface Marker Expression
Figure imgf000122_0001
[00417] The results of this analysis showed the following: Average of 94.7% ± 2.1% of CD56+/CD3-. Less than 1% CD3+ and CD19+. Average of 74.1% ± 5.6% of CD 16 [00418] Post thaw viability: 89.1% ± 3.6%. Expressing activating receptors such as CD226, NKG2D, CD11a, NKp30, NKp44 and NKp46, and maturation marker CD94.
[00419] Shedding assays demonstrated shedding resistance of CYNK-101 (n=7 donors) induced by PMAi at both post thaw and after 2 days recovery.
[00420] CYNK-101 showed greater than 90% CD56+CD3-, less than 1% CD3 or CD 19, greater than 65% CD 16, and expression of NK surface markers such as CD226, NKG2D, CDlla, NKp30, NKp44, NKp46, and CD94.
[00421] CYNK-101 was resistant to CD 16 shedding following PMAi stimulation. [00422] CYNK-101 displayed cytotoxicity against K562 cells with a dose dependent manner. In the mixed targets culture system of K562 plus normal PBMCs, CYNK-101 can specifically kill K562 while sparing normal PBMCs even at the E:T ratio up to 100:1.
[00423] Increased GM-CSF, IFN-g, and TNF-a secretion was observed from CYNK- 101 in the presence of K562 compared to that of CYNK-101 alone.
[00424] Increased intracellular cytokine production such as GM-CSF, TNF-a, IFN-g was shown from CYNK-101 in the presence of K562, or stimulated with PMAi, or IL-12+IL- 18 compared to that of CYNK-101 alone.
7.7 Example 7: Cleavage Resistant CD 16 Expressing CYNK Cells Are Effective Against HER2+ Cancers
[00425] CYNK-101 is a human placental hematopoietic stem cell derived natural killer (NK) cell product, that is genetically modified to express a variant of CD 16, Fc gamma receptor III (FcgRIII), via lentiviral vector transduction. CD16 plays a central role in antibody dependent cell mediated cytotoxicity (ADCC) in NK cells through binding to the Fc portion of IgG antibodies. FIG. 18 shows the rationale of ADCC by engineering CD 16 on NK cells. CYNK-101 was designed to express a high-affinity proteolytic cleavage-resistant CD16 variant with a Valine at amino acid position 158 and Proline at position 197 as demonstrated in the Construct Design in FIG. 19 [Wu, 1997; Sugita, 1999; Koene, 1997;
Jing, 2015],
[00426] To assess the anti-tumor ADCC activity of CYNK-101, extensive in vitro, ex vivo and in vivo studies have been conducted using CYNK-101 in combination with trastuzumab (Herceptin) against HER2+ Gastric/Gastroesophageal Junction (G/GEJ) cancer cell lines, and HER2+ breast cancer cell lines. The in vitro study examined CYNK-101 multiparameter phenotypic characterization, CD16 shedding resistance evaluation of CYNK-101 followed by PMAi stimulation, cytotoxicity evaluation of CYNK-101 in combination with Herceptin against G/GEJ cancer cell lines as NCI-N87 and OE19; and breast cancer cell lines as AU565, BT-474, HCC-1954, SKBR-3, ZR-75-30; and assessment of cytokine secretion such as IFN-g, TNF-a, and GM-CSF from CYNK-101 in combination with Herceptin in the presence of the above tumor cell lines. An ex vivo study was conducted to further evaluate ADCC activity of CYNK-101 administered to NSG mice. CYNK-101 cells were isolated from mouse liver thirteen days post injection and evaluated for phenotypic characterization, CD 16 shedding resistance and anti -tumor ADCC activity. An in vivo study was further performed to evaluate the anti -tumor activity of CYNK-101 in combination with Trastuzumab in a subcutaneous gastric tumor mouse model. In addition, the safety of CYNK- 101 in combination with Trastuzumab was also evaluated in this study. The results summarized here provide a rationale to support clinical development of CYNK-101 in combination with trastuzumab against HER2 overexpressing G/GEJ cancer.
Methods and materials xCELLigence real time cell analysis-based antibody dependent cellular cytotoxicity assay against tumor cell lines
[00427] CYNK-101 cells (donor IDs: 2000112643, 2000113629, 2000113366, 2000113511, 2000113472, 2000113880, 2000114102) with purity of >85% CD56+CD3' were served as effector cells in this assay. The following HER2 overexpressing tumor cell lines were used as target cells: NCI-N87 (ATCC, Cat# CRL-5822), OE19 (Sigma, Cat#
96071721), HCC-1954 (ATCC, Cat# CRL-2338), SKBR-3 (ATCC, Cat# HTB-30), BT-474 (ATCC, Cat# CRL-3247), AU565 (ATCC, Cat# CRL-2351), and ZR-75-30 (ATCC, Cat# CRL-1504). The anti-HER2 antibody Herceptin (Blue Door Pharma, Rockville, MD, Cat# 50242-132-01) or the Ultra-LEAF purified human IgGi isotype control recombinant antibody (Biolegend, Cat# 403501) was used at lpg/mL. The xCELLigence real time cell analysis (RTCA) system (ACEA Biosciences, San Diego, CA) was used for the determination of
ADCC activities mediated by the combination of effector cells and antibody. 8 x 104 /well NCI-N87, 6 x 104 /well OE19, 1 x 104 /well HCC-1954, 2 x 104 /well SKBR-3, 2 x 104 /well BT-474, 2 x 104 /well AU565, or 2 x 104 /well ZR-75-30 target cells were seeded in the E- plates in 100 pL of assay buffer (RPMI 1640 media supplemented with 10% fetal bovine serum (FBS), 1% penicillin and 1% streptomycin) for 24 hours at 37°C in 5% CCh. IgGi or Herceptin was then added 30 minutes prior to the addition of different amounts of CYNK- 101 cells to achieve various E:T ratios (10:1, 5:1, 2.5:1, 1:1 and 0.6:1) for each donor in duplicate. Target cells were incubated with CYNK-101 cells in the presence of the antibodies in 200 pL final volume. The xCELLigence software was then used to determine the percent cytotoxicity at 4h and 24h after effector cells added. The results from different experiments were reported as mean of donors ± standard deviation of the mean (SD).
Flow cvtometrv-based cytotoxicity assay with tumor cell lines [00428] CYNK-101 from seven different donors (donor IDs: 2000112643, 2000113629, 2000113366, 2000113511, 2000113472, 2000113880, 2000114102) used in all the assays were > 85% CD56+CD3" cells, > 70% viability by 7-AAD". Cytotoxicity assays were performed by using CYNK-101 cells as effector cells and the tumor cell lines, such as K562 (ATCC, Cat# CCL-243) as the target cells. Target cell number was fixed at 1 x 104 while CYNK-101 cells were used in different amounts to achieve various E:T ratios (10:1, 5:1, 2.5:1, and 1.25:1). To distinguish target cells from CYNK-101 cells on the flow cytometer, target cells were labeled with 7.5 pM PKH26 fluorescent dye (Sigma- Aldrich, St Louis, MO, Cat# PKH26-GL). Target cells were incubated with NK cells in 96-well U- bottom tissue culture plates in 200 pL of assay buffer for 4 hours at 37°C in 5% CO2. After incubation, cells were harvested and TO-PRO-3 (Invitrogen, Carlsbad, CA Cat# T3605), a membrane-impermeable DNA stain, was added to the cultures at 1 pM final concentration in order to identify dead cells (TO-PRO-34). To determine spontaneous target cell death, PKH26-labeled target cells were cultured alone for the duration of the assay. As a positive control for dead cells, 1 x 105 labeled target cells were permeabilized with 350 pL of Cytofix/Cytoperm buffer (BD Biosciences, Cat# 554722) for 20 minutes at 4°C. The data were acquired on a FACSCanto X (BD Biosciences, San Jose, CA) and analyzed by FlowJo (Tree Star, Ashland, OR).
[00429] The percentage of dead target cells in each sample was calculated as follows: %TO-PRO-3+PKH26+ cells / (%TO-PRO-3+PKH26+ + %TO-PRO-3 PKH26+) * 100%. Percent cytotoxicity reported was calculated by subtracting the percent of dead target cells in cultures of target cells alone from the percent of dead target cells in co-cultures of effector and target cells. The results are reported as the mean of the donors ± standard deviation of the mean (SD).
CD 16 shedding resistance assay
[00430] To evaluate the CD 16 shedding resistance of CYNK-101 followed by cell stimulation, 5 x 105 CYNK-101 cells (donor IDs: 2000112643, 2000113629, 2000113366, 2000113511, 2000113472, 2000113880, 2000114102) were incubated for 2h with either Phorbol 12-myristate 13-acetate plus ionomycin (PMAi) (eBioscience, Cat# 00-4970-93) or ethanol vehicle control in RPMI-1640 media with 10% FBS. NK cells without CD 16 transduction were served as assay control. Cells from each condition were then incubated at room temperature for 10 minutes in FACS buffer (PBS with 0.5 mM EDTA and 2% FBS) containing human Fc block (BD Biosciences, Cat# 564220). The cells were stained with anti- CD16 PE (BD Biosciences, Cat# 556619), anti-CD56 APC (BD Biosciences, Cat# 555518), and anti-CDl la FITC (BD Biosciences, Cat# 555383) for 30 minutes at 4 °C. After washing, 7-AAD (BD Biosciences, Cat# 559925) was added to distinguish live and dead cells, the data were acquired on FACSCanto X (BD Biosciences, San Jose, CA) and analyzed by FlowJo (Tree Star, Ashland, OR). The data were reported as % CD56+CD16+ cells gated under 7- AAD" cells. Setting of the % positive gate was done using isotype stained samples as controls. Flow cytometric detection of NK surface receptors
[00431] 1 x 106 CYNK-101 cells from each donor (donor IDs: 2000112643,
2000113629, 2000113366, 2000113511, 2000113472, 2000113880, 2000114102) were first labeled with a viability stain (LIVE/DEAD Aqua, Invitrogen, Cat# L34957) according to the manufacturer’s protocol (Invitrogen). Cells were then stained with anti-CD56 Alexa Fluor 700 (BD Biosciences, Cat# 557919), anti-CD3 APC-H7 (BD Biosciences, Cat# 560176), and anti-CD16-PE (BD Biosciences, Cat# 556619) antibodies, as well as with an additional seven antibodies against NK receptors: NKG2D (BD Biosciences, Cat# 562364), NKp46 (BD Biosciences, Cat# 563230), NKp44 (BD Biosciences, Cat# 744305), NKp30 (BD Biosciences, Cat# 563385), CD94 (R & D Systems, Cat# FAB1058A), CDlla (BD Biosciences, Cat# 561387), and DNAM-1 (BD Biosciences, Cat# 559788). Samples were washed and immediately analyzed on BDLSR Fortessa X-20. Data were analyzed using the FlowJo software. The data were expressed as % positive cells gated under CD56+CD3"Aqua" single cells. Setting of the % positive gate was done using unstained and isotype stained samples as controls.
Intracellular cytokine staining (ICCS) assay [00432] CYNK-101 cells (donor IDs: 2000112643, 2000113629, 2000113366, 2000113511, 2000113472, 2000113880, 2000114102) were plated in 96-well plates at 2 x 105 cells/well in 200 μL of RPMI 1640 medium with 10% FBS and antibiotics. Stimulating agents were added to the corresponding wells as followings: IX Cell Stimulation Cocktail containing PMAi (eBiosciences, Cat# 00-4970-03), or IL-12 (20 ng/mL) (R&D Systems, Minneapolis, MN, Cat# 219-IL-025) plus IL-18 (100 ng/mL) (R&D Systems, Cat# B003-2). Additionally, 1 x 105 K562 tumor cells were added with the CYNK-101 cells at E:T ratio of 2: 1. Control wells were left unstimulated for the duration of stimulation. After 1 hour, the protein transport inhibitor Brefeldin A (BD Fastlmmune, San Diego, CA, Cat# 347688) was added to all the wells at 10 μg/mL final concentration. Stimulation was performed for an additional 4 hours, after which cells were labeled with a viability stain, LIVE/DEAD Aqua, according to the manufacturer’s protocol (Invitrogen, Cat# L34957). Cells were then stained with anti-CD56 PE-Cy7 (Biolegend, Cat# 362510), anti-CD3 APC-H7 (BD Biosciences,
Cat# 560176), and anti-CDlla BV711 (BD Biosciences, Cat# 563935) antibodies for 15 min at 4° C in brilliant stain buffer (BD Biosciences, Cat# 563794), fixed and permeabilized using Cytofix/Cytoperm kit according to the manufacturer’s protocol (BD Biosciences, Cat# 554722), and then washed with perm wash buffer (BD Biosciences, Cat# 554723). Permeabilized cells were stained with anti-IFN-g APC (BD Biosciences, Cat# 554702), anti- TNF-a BV650 (BD Biosciences, Cat# 563418), anti-GM-CSF PE (BD Biosciences, Cat# 554507), anti-Perforin BV421 (Biolegend, Cat# 308122), and anti-Granzyme B AF700 (BD Biosciences, Cat# 560213) antibodies in brilliant stain buffer. Data were acquired on BD LSRFortessaX-20 and analyzed using the FlowJo software. Data were expressed as % positive cells gated under CD56+CD3-Aqua- single cells. The gate that delineated cells positive for a given cytokine was set by utilizing unstimulated samples, isotypes, and/or separation of stained populations. The data were analyzed and presented using GraphPad Prism (GraphPad Software, La Jolla, CA) and depicted as mean of the donors ± SD.
Cytokine secretion assay
[00433] CYNK-101 cells (donor IDs: 2000112643, 2000113629, 2000113366, 2000113511, 2000113472, 2000113880, 2000114102) were incubated with tumor cell lines, or tumor cell lines with either the IgGi or Herceptin antibodies at 1 pg/mL in 96-well U- bottom tissue culture plates at an E:T ratio of 1:1 (1 x 105 cells each) in 200 pL of RPMI 1640 supplemented with 10% FBS and antibiotics. After 24 hours incubation at 37°C and 5% CO2, the supernatant was collected and cytokine concentrations were determined by Luminex analysis using MILLIPLEX MAP magnetic bead kits (EMD Millipore, Burlington, MA, Cat# HCD8MAG-15K-07 for granulocyte-macrophage colony-stimulating factor (GM-CSF), perforin, TNF-a, IL-10, granzyme A, granzyme B and IFN-g) according to the protocol provided by the manufacturer. The data were analyzed using Milliplex Xponent and Analyst software (EMD Millipore). The results from different experiments were depicted as mean of donors ± SD.
Ex vivo characterization of CYNK-101
[00434] 2 x 107 CYNK-101 cells were intravenous (IV) injected to busulfan pretreated
NOD-scid IL2Rgammanu11 immunodeficient (NSG) mice at DayO. Recombinant human IL-15 was intraperitoneally injected at Days 0, 2, 4, 6, 8, 10 and 12 to support NK cell proliferation and survival. At thirteen days post IV injection, CYNK-101 cells were isolated from mouse liver and evaluated for phenotype characterization, CD 16 shedding resistance followed by PMAi stimulation and ADCC activity against NCI-N87. The phenotyping panel included the following: AQUA, mCD45, hCD45, CD3, CD56, CD16, CDlla, CD94, CD158a, CD158el/e2, CD158bl/b2, NKG2D, NKp46, andNKp44.
In vivo efficacy and safety study of CYNK-101
[00435] The anti-tumor activity and safety of CYNK-101 in combination with Trastuzumab were evaluated in a subcutaneous gastric tumor model using the NSG mice with transgenic expression of human IL-15 (NSG-Tg-hIL15).
[00436] A total of eighty female NSG-Tg-hIL15 mice were preconditioned with busulfan (30mg/kg) to deplete myeloid cells on Day -3. As shown in FIG. 20, NCI-N87 gastric cancer cell line was subcutaneously inoculated at the dose of 3 x 106 cells/mouse on Day 0. One week later (Day 7), seventy -three mice with palpable tumors were randomized to the following four groups: Vehicle control (n=19), Trastuzumab (n=17), CYNK-101 (n=19), CYNK-101 + Trastuzumab (n=18). Trastuzumab (lOmg/kg) was administered intraperitonially (IP) once on Day 7, and CYNK-101 at the dose of 10 x 106 cells/mouse was administered intravenously (IV) three times on Days 7, 14 and 21.
In vitro anti-tumor ADCC activity of CYNK-101 in combination with Herceptin against HER2+ solid tumor cancer cells
Enhanced in vitro ADCC activities of CYNK-101 against G/GEJ tumor cell lines [00437] To assess the in vitro ADCC activity of CYNK-101 in combination with the anti-HER2 antibody Herceptin against HER2 overexpressing G/GEJ cancer cell lines, NCI- N87 and OE-19 were used as targets in a xCELLigence real time cell analysis-based assay. To assess NK cell dose dependent effects, varying E:T ratios from 10:1 to 0.6:1 for 7 different CYNK-101 donors were used in the assay. The Herceptin alone or IgGl control alone against the targets were included as controls. In addition, low HER2 expression normal human dermal fibroblasts cells (NHDF) were used as target cells to determine whether cytotoxic effects of CYNK-101 plus Herceptin were specific for HER2+ tumor cells.
[00438] The results showed that CYNK-101, when combined with Herceptin, displayed enhanced cytotoxic activity against both NCI-N87 and OE19 G/GEJ tumor cell lines at an E:T ratio as low as 0.6: 1 compared to when the IgG antibody was added. This activity increased with increasing E:T ratios in a dose-dependent manner at the early timepoint of 4h for both tumor lines and at 24h for OE19 (FIGS. 21A - 21B). By 24h at an E:T ratio of 0.6:1, CYNK-101 cells (n=7 donors) lysed 91.3% ± 15.0% of NCI-N87 cells in the presence of Herceptin compared to 7.5% ± 9.3% of NCI-N87 cells in the presence of IgG control, and 44.6% ± 20.1% of OE19 cells in the presence of Herceptin compared to 14.7% ± 14.8% in the presence of IgG control (FIGS. 21A - 21B Error! Reference source not found.)
[00439] FIGS. 21 A - 21 B shows the average cytotoxic activity of CYNK-101 cells in combination with Herceptin or a control IgG against the indicated gastric tumor cell lines, NCI-N87 and OE19. The error bars represent the SD from the mean calculated from seven different CYNK-101 donors. Cytotoxicity from Herceptin or IgG alone is included for reference. * indicate significantly higher activity of CYNK-101 in combination with Herceptin compared to that of IgG (***p<0.001 and *p<0.05).
[00440] Furthermore, when using mixed target of NCI-N87 with Low HER2 expression NHDF (FIG. 22A), CYNK-101 cells in combination with Herceptin specifically kill NCI-N87, while spare NHDF cells at any of the E:T ratios tested up to 100: 1 (FIG. 22B), indicating that CYNK-101 cells in combination with Herceptin were capable not only of lysing HER2+ tumor cells but also of discriminating between HER2+ normal and tumor targets.
Secretion of IFN -gamma, TNF-alpha and GM-CSF cytokines in response to gastric cancer tumor cell lines
[00441] Because NK cell cytokine secretion is implicated in anti-tumor immune responses [Marcus, 20141. CYNK-101 cytokine secretion was tested following co-culture with gastric cancer tumor cell lines. After 24 hours of co-culture, supernatant was analyzed for the presence of IFN-g, TNF-a, and GM-CSF. The results are summarized in Table 5. [00442] IFN-γ was significantly induced from CYNK-101 in the presence of both NCI-N87 and OE19 when Herceptin was added (Table 5). Specifically, 1 x 106 CYNK-101 cells secreted 803 ± 559 pg IFN-g with NCI-N87 and 88 ± 72 pg with OE19 when Herceptin was present compared to 15 ± 20 pg and 0 ± 0 pg when IgGi was present, respectively; the secretion in the presence of IgG was not significantly different from that of the baseline for CYNK-101 cells of 13 ± 16 pg. In addition, TNF-a and GM-CSF were significantly induced in the presence of both gastric cancer tumor cell lines when Herceptin was added in combination (Table 5).
[00443] In summary, CYNK-101 cells secreted relevant immunomodulatory cytokines in the presence of Herceptin and gastric cancer tumor cell lines. Thus, CYNK-101 cells in combination with Herceptin were not only capable of directly lysing gastric cancer tumor cells but also capable of indirectly eliciting anti-tumor responses through their ability to secrete IFN-g, TNF-a, and GM-CSF.
Table 5: Summary of cytokine secretion from CYNK-101 in response to gastric cancer tumor cells
Figure imgf000129_0001
a) Number of CYNK-101 donors b) Average
* P<0.05 (Student’s T-test, in the presence of tumor cells vs. none) CYNK-101 is resistant to CD16 shedding post PMAi stimulation
[00444] High expression of CD16 was detected from CYNK-101 as seen in FIG. 23, the average of CD16 expression of CYNK-101 (n=7 donors) is 74.1% ± 5.6%. To assess if CYNK-101 is resistant to CD 16 shedding, CYNK-101 was stimulated with PMAi for 2h, followed by surface staining of CD16, non-transduced NK cells were served as control. As shown in FIGS. 24 A - 24B, followed by PMAi stimulation, the percentage of CD 16 expression on CYNK-101 maintained compared to that of control, while around 70% CD 16 expression decreased from non-transduced NK cells. In summary, CYNK-101 demonstrates CD 16 shedding resistant post PMAi stimulation.
Expression of cytotoxic mediators, perforin and granzyme B, and NK activating receptors in CYNK-101
[00445] To confirm that CYNK-101 cells express molecules required for cytotoxic activity, we analyzed intracellular expression of perforin, granzyme B, and cell surface expression of activating receptors, such as CD 16, DNAM-1 (CD226), NKG2D, CD94, NKp30, NKp44, and NKp46, which are known to be important for cytotoxic effects of NK cells [Marcus, 20141 by flow cytometry. Data for CYNK-101 from seven donors were summarized in Table 6.
[00446] Results indicated that CYNK-101 cells expressed all of the factors required for cytotoxic activity tested: perforin (25.8% ± 23.0% cells), granzyme B (30.4% ± 30.4% cells), CD16 (74.1% ± 5.6% cells), CD226 (49.6% ± 9.7 % cells), NKG2D (40.6% ± 14.0% cells), CD94 (48.9% ± 10.7% cells), , NKp30 (37.4% ± 16.8% cells), NKp44 (93.8% ± 3.1% cells), andNKp46 (15.7% ± 5.0% cells). Results suggested that CYNK-101 cells exerted cytotoxic effects onto their targets by utilizing pathways that have been well described for NK cells involving recognition of tumor targets via activating receptor engagement and secretion of perforin and granzyme B-containing granules.
Table 6: Assessment of expression of perforin, Granzyme B, and activation receptors of
CYNK-101 by Flow Cytometry
Figure imgf000130_0001
Figure imgf000131_0001
Enhanced in vitro ADCC activities of CYNK-101 against HER2+ breast cancer cell lines [00447] To assess the in vitro ADCC activity of CYNK-101 in combination with Herceptin against HER2+ breast cancer cell lines, AU565, BT-474, HCC-1954, SKBR-3, ZR- 75-30 were used as targets in axCELLigence real time cell analysis-based assay. To assess NK cell dose dependent effects, varying E:T ratios from 10:1 to 0.6:1 for 7 different CYNK- 101 donors (unless specified) were used in the assay.
[00448] As shown in FIGS. 25 A - 25B, enhanced anti -tumor ADCC activity of CYNK-101 in combination with Herceptin against the HER2+ breast cancer cell lines was demonstrated at both 4h and 24h with the indicated E:T ratios compared to that of CYNK- 101 plus IgG control.
Secretion of IFN -gamma, TNF-alpha and GM-CSF cytokines in response to breast cancer cell lines
[00449] CYNK-101 cytokine secretion was tested following co-culture with breast cancer cell lines in the presence of Herceptin or IgG control. After 24 hours of co-culture, supernatant was analyzed for the production of IFN-g, TNF-a, and GM-CSF.
[00450] As seen inFIG. 26, IFN-g was significantly induced from CYNK-101 in the presence of the breast cancer cell lines as indicated compared to that of IgG control. In addition, TNF-a and GM-CSF were significantly induced in the presence of the breast cancer cell lines when Herceptin was added in combination.
[00451] In summary, CYNK-101 cells secreted relevant immunomodulatory cytokines in the presence of Herceptin and breast cancer cell lines. Thus, CYNK-101 cells in combination with Herceptin were not only capable of directly lysing breast cancer cells but also capable of indirectly eliciting anti-tumor responses through their ability to secrete IFN-g, TNF-a, and GM-CSF.
Ex vivo anti-tumor ADCC activity of CYNK-101 in combination with Herceptin against HER2+ gastric cancer cells [00452] An ex vivo model was developed to further evaluate ADCC activity of CYNK- 101 administered to NSG mice. In brief, CYNK-101 was IV-injected at the dose of 2 x 107 cells/animal into busulfan-pretreated NSG mice at Day 0. Recombinant human IL-15 was intraperitoneally injected at Days 0, 2, 4, 6, 8, 10 and 12 to support CYNK-101 in vivo expansion and persistence. At Day 13, CYNK-101 cells were isolated from mouse livers (ex v/voCYNK-101) and assessed for phenotype and ADCC activity. FIG. 27 shows the percentage of CYNK-101 cells from the livers pre and post mouse cell depletion in the ex vivo study. In this study, among the cells from post mouse cell depletion, 69.3% of which were live hCD45+, and of those 97.6% were CD56+CD3" cells. Surface marker expression profiling revealed that the ex vivo isolated cells have increased CD94 and CD158 expression, retained their CD 16 expression, and decreased NKp44 expression compared to the corresponding prior infusion CYNK-101 donor (FIG. 28). Thus, the increased expression of CD94 and CD158 indicated the maturation of CYNK-101 cells in NSG mice after 13 days IV administration.
[00453] After stimulated by PMAi for 2h, both ex vivo and in vitro CYNK-101 demonstrated CD 16 shedding resistance, while 60% CD 16 shedding observed from control sample (FIG. 29). FIG. 30 demonstrated that enhanced ADCC activity of ex v/voCYNK-101 in combination with Herceptin against NCI-N87 at E:T ratio of 0.5:1 compared to that of IgG, or in vitro CYNK-101 plus Herceptin, in vitro CYNK-101 plus IgG. Increased cytokine secretion such as IFN-g, TNF-a and GM-CSF was observed from ex vivo CYNK-101 in combination with Herceptin against NCI-N87 at E:T ratio of 0.5:1 (FIG. 31).
[00454] In summary, the study demonstrated that ex v/vo-CYNK-101 can be enriched from mouse liver followed by 13 days post injection. Ex v/vo-CYNK-101 was CD 16 shedding resistance post PMAi stimulation. Ex vivo-CYNK-101 in combination with Herceptin showed enhanced ADCC activity and enhanced cytokine secretion against NCI- N87 compared to that of ex v/vo-CYNK-101+IgG, in vitro CYNK-101+Herceptin, and in vitro CYNK-lOl+IgG.
[00455] Taken together, the in vitro and ex vivo results demonstrated ADCC activity of CYNK-101 in combination with Herceptin against HER2 overexpressing G/GEJ tumor cell lines. The data outlined here support the clinical development of CYNK-101 in combination with trastuzumab for patients with HER2 overexpressing G/GEJ adenocarcinoma.
In vivo efficacy of CYNK-101 in combination with Trastuzumab in a subcutaneous gastric tumor mouse model [00456] In this study, the anti -tumor activity and safety of CYNK-101 in combination with Trastuzumab were evaluated in a subcutaneous gastric tumor model using NSG-Tg- hIL15. As shown in FIG. 32, CYNK-101 alone had no impact on tumor growth, whereas Trastuzumab alone significantly inhibited tumor growth from Day 14 to Day 28 compared with vehicle control. In consistent with our in vitro and ex vivo findings, CYNK-101 in combination with Trastuzumab demonstrated a synergistic anti-tumor activity and significantly suppressed tumor growth compared to Trastuzumab or CYNK-101 alone. More significantly, CYNK-101 plus Trastuzumab treatment continuously reduced tumor volume from Day 7 to Day 28, and approximately 30% tumor volume reduction was achieved on Day 28 compared to Day 7 baseline.
[00457] Three repeated weekly IV administration of CYNK-101 at the dose of 10 x 106 cells/mouse was safe and well tolerated with or without Trastuzumab combination therapy. No abnormal clinical symptoms, morbidity and mortality occurred throughout the study. No CYNK-101 or CYNK-101 + Trastuzumab related abnormal effects on body weight and gross pathology at necropsy were observed.
[00458] Taken together, this study demonstrated that CYNK-101 in combination with Trastuzumab had a synergistic in vivo anti-tumor activity against gastric cancer, and the combination therapy was safe and well tolerated.
[00459] In summary, our results demonstrated synergistic anti-tumor ADCC activities of CYNK-101 plus Trastuzumab against HER2+ gastric cancer cells in vitro, ex vivo and in vivo. Further development of CYNK-101 in combination with Trastuzumab for HER2+ gastric cancer immunotherapy is underway.
References
Jing Y, Ni Z, Wu J, Higgins L, Markowski TW, Kaufman DS, Walcheck B. Identification of an ADAM17 cleavage region in human CD16 (FcyRIII) and the engineering of anon- cleavable version of the receptor in NK cells. PLoS One. 2015 Mar 27;10(3):e0121788.
Koene HR, Kleijer M, Algra J, Roos D, von dem Borne AE, de Haas M. Fc gammaRIIIa- 158V/F polymorphism influences the binding of IgG by natural killer cell Fc gammaRIIIa, independently of the Fc gammaRIIIa-48L/R/H phenotype. Blood. 1997 Aug 1;90(3): 1109-14.
Marcus A, Gowen BG, Thompson TW, Iannello A, Ardolino M, Deng W, et al. Recognition of tumors by the innate immune system and natural killer cells. Adv Immunol. 2014;122:91- 128. Sugita N, Yamamoto K, Kobayashi T, Van Der Pol W, Horigome T, Yoshie H, Van De Winkel JG, Hara K. Relevance of Fc gamma RIIIa-158V-F polymorphism to recurrence of adult periodontitis in Japanese patients. Clin Exp Immunol.1999 Aug;l 17(2):350-4.
Wu J, Edberg JC, Redecha PB, Bansal V, Guyre PM, Coleman K, Salmon JE, Kimberly RP. A novel polymorphism of FcgammaRIIIa (CD 16) alters receptor function and predisposes to autoimmune disease. J Clin Invest. 1997 Sep l;100(5):1059-70.
7.8 Example 8: Monitoring T cell reactivity to allogeneic placental cell products using multiple lymphocyte reaction and TCR sequencing approaches [00460] Allogeneic stem cell-derived therapies offer patients a readily accessible source of off-the-shelf product. However, allogeneic cells may be recognized by the recipient’s immune system thus negatively affecting persistence and efficacy. Among people, allelic differences in human leukocyte antigen (HLA) complex genes are a major source of alloantigen which can elicit immunity. While there are established assays for measuring alloantibodies, monitoring T cell alloreactivity is more challenging. Unique clonal populations vary between individuals and functional assays are difficult to scale and standardize if alloreactive populations are rare/infrequent. Here, we evaluate how alloreactive T cells from patients may be identified using multiple lymphocyte reaction (MLR) utilizing dendritic cells (DC) derived from the placental blood of donors used for drug product. In transplantation, next generation sequencing of the TCR locus permits the subsequent identification and monitoring of allogeneic T cell clones in a patient over time1. We evaluate if similar approaches can be applied to monitoring alloreactivity to allogeneic cell therapies using cryopreserved drug donor and recipient patient leukocytes.
[00461] Placental leukocytes depleted of CD34 cells or peripheral blood mononuclear cells (PBMC) were enriched for monocytes using magnetic-bead isolation followed by dendritic cell (DC) maturation with or without cytokine restimulation. DC were activated with lipopoly saccharide (LPS) for 16 hours then cultured in the presence of 3rd party CD3+ T cells for 5 days in the MLR. Proliferating alloantigen-specific T cells were observed by bright field microscopy and by flow cytometry. T cell cytokine production was observed by multiplex analysis of supernatants from the MLR. TCR locus sequencing was performed on cDNA libraries generated from unstimulated and post-MLR cultures using the Illumina platform.
[00462] Well-established MLR protocols using GM-CSF+IL4 monocyte derived DCs as professional antigen-presenting cells are insufficient for placental blood monocytes indicated by low levels of HLA-ABC and HLA-DR. We demonstrate placental monocytes can be fully matured to DC using additional cytokine restimulation resulting in clonal cell expansion visible by brightfield microscopy and IFNg, IL-2, and TNFa cytokine release. [00463] TCR sequencing analysis from one responding T cell donor following stimulation with 3 different donor DCs demonstrated robust expansion of one dominant clone followed by several conserved T cell clones. These outgrowing T cells were the same across different DC donors suggesting that expanding T cell clones from this individual likely recognize conserved epitopes shared across these non-self HLA alleles. Structural studies suggest that conserved contact points between TCR and HLA alleles are dominant predictors of TCR binding, rather than the specificity of non-self peptides2.
[00464] Immune rejection is a potential limitation of allogeneic cell therapy. We demonstrate that monocytes can be cryopreserved from placental and peripheral blood in anticipation of peforming MLR functional assays. This permits the downstream monitoring of T cell alloreactivity to cell therapy products using a treated patient’s blood sample as a source of responding T cells. Our data highlight the opportunity to identify alloreactive T cell clones from patients, followed by clone tracking over time following exposure to allogeneic cell therapy.
[00465] 1. DeWolf S, Sykes M. Alloimmune T cells in transplantation. J Clin Invest.
2017;127(7):2473-2481.
[00466] 2. Coif LA, Bankovich AJ, Hanick NA, et al. How a single T cell receptor recognizes both self and foreign MHC. Cell. 2007;129(1): 135-146.
[00467] The present invention is not to be limited in scope by the specific embodiments described herein. Indeed, various modifications of the invention in addition to those described will become apparent to those skilled in the art from the foregoing description and accompanying figures. Such modifications are intended to fall within the scope of the appended claims.
[00468] All references cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes. The citation of any publication is for its disclosure prior to the filing date and should not be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention.

Claims

WHAT IS CLAIMED IS:
1. A method of suppressing the proliferation of tumor cells comprising contacting the tumor cells with a population of placental-derived natural killer cells comprising a cleavage resistant CD 16 and an antibody, wherein the tumor cells are HER2+, and wherein the antibody is an anti-HER2 antibody.
2. The method of suppressing the proliferation of tumor cells of claim 1, wherein the placental-derived natural killer (NK) cells are CYNK cells.
3. The method of suppressing the proliferation of tumor cells of claim 2, wherein the CYNK cells are placental CD34+ cell-derived natural killer (NK) cells.
4. The method of suppressing the proliferation of tumor cells of any one of claims 2-3, wherein the CYNK cells are characterized by expression of one or more markers selected from the group consisting of FGFBP2, GZMH, CCL3L3, GZMM, CXCR4, ZEB2, KLF2, LITAF, RORA, LYAR, CNOT1, IFNG, DUSP2, ATG2A, CD7, PMAIP1, PPP2R5C, NR4A2, ZFP36L2, PIK3R1, KLRFl, SNHG9, MT2A, RGS2, CHD1, DUSP1, EML4, ZFP36, ZC3H12A, DNAJB6, SBDS, IRF1, TSC22D3, TSPYL2, PNRC1, ISCA1, JUNB, WHAMM, RICTOR, TNFAIP3, EPC1, MVD, CLK1, ARL4C, REL, KMT2E, YPEL5, AMD1, BTG2, and IDS which is lower than expression of said markers in peripheral blood natural killer cells and / or expression of one or more markers selected from the group consisting of NDFIP2, LINC00996, MAL, CCL1, MB, SPINK2, C15orf48, CAMK1, KLRCl, TNFSF10, TNFRSF18, IL32, CAPG, AC092580.4, S100A11, TNFRSF4, ENOl, FCER1G, CCND2, KRT81, MRPS6, ANXA2, PTGER2, GLOl, HAVCR2, PYCARD, LAT2, SLC16A3, COTL1, PKM, TALDOl, CD96, NCR3, KRT86, STMN1, LTB, ARPC1B, ARPC5, FKBPIA, TIMP1, GZMK, CD59, PGK1, RGS10, EVL, RAC2,
LGALS1, ITGB7, TUBB, PGAM1, PRF1, GZMB, IL2RB, KLRC2, and KLRBl which is higher than expression of said markers in peripheral blood natural killer cells.
5. The method of suppressing the proliferation of tumor cells of any one of claims 2-4, wherein the CYNK cells are characterized by expression of one or more markers selected from the group consisting of FGFBP2, GZMH, CCL3L3, GZMM, CXCR4, ZEB2, KLF2, LITAF, RORA, LYAR, CNOT1, IFNG, DUSP2, ATG2A, CD7, PMAIP1, PPP2R5C, NR4A2, ZFP36L2, PIK3R1, KLRFl, SNHG9, MT2A, RGS2, CHD1, DUSP1, EML4, ZFP36, ZC3H12A, DNAJB6, SBDS, IRF1, TSC22D3, TSPYL2, PNRC1, ISCA1, JUNB, WHAMM, RICTOR, TNFAIP3, EPC1, MVD, CLK1, ARL4C, REL, KMT2E, YPEL5, AMD1, BTG2, and IDS which is lower than expression of said markers in peripheral blood natural killer cells.
6. The method of suppressing the proliferation of tumor cells of claim 4 or claim 5, wherein expression of 2, 3, 4, 5, 6, 7, 8, 9, 10, or more markers selected from the group consisting of FGFBP2, GZMH, CCL3L3, GZMM, CXCR4, ZEB2, KLF2, LITAF, RORA, LYAR, CNOT1, IFNG, DUSP2, ATG2A, CD7, PMAIP1, PPP2R5C, NR4A2, ZFP36L2, PIK3R1, KLRFl, SNHG9, MT2A, RGS2, CHD1, DUSP1, EML4, ZFP36, ZC3H12A, DNAJB6, SBDS, IRF1, TSC22D3, TSPYL2, PNRC1, ISCA1, JUNB, WHAMM, RICTOR, TNFAIP3, EPC1, MVD, CLK1, ARL4C, REL, KMT2E, YPEL5, AMD1, BTG2, and IDS is lower than expression of said markers in peripheral blood natural killer cells.
7. The method of suppressing the proliferation of tumor cells of any one of claims 2-6, wherein the CYNK cells are characterized by expression of one or more markers selected from the group consisting of NDFIP2, LINC00996, MAL, CCL1, MB, SPINK2, C15orf48, CAMK1, KLRC1, TNFSF10, TNFRSF18, IL32, CAPG, AC092580.4, S100A11, TNFRSF4, ENOl, FCER1G, CCND2, KRT81, MRPS6, ANXA2, PTGER2, GLOl, HAVCR2, PYCARD, LAT2, SLC16A3, COTL1, PKM, TALDOl, CD96, NCR3, KRT86, STMN1, LTB, ARPCIB, ARPC5, FKBP1A, TIMP1, GZMK, CD59, PGK1, RGS10, EVL, RAC2, LGALS1, ITGB7, TUBB, PGAM1, PRF1, GZMB, IL2RB, KLRC2, and KLRBl which is higher than expression of said markers in peripheral blood natural killer cells.
8. The method of suppressing the proliferation of tumor cells of claims 2-7, wherein expression of 2, 3, 4, 5, 6, 7, 8, 9, 10, or more markers selected from the group consisting of NDFIP2, LINC00996, MAL, CCL1, MB, SPINK2, C15orf48, CAMK1, KLRC1, TNFSF10, TNFRSF18, IL32, CAPG, AC092580.4, S100A11, TNFRSF4, ENOl, FCER1G, CCND2, KRT81, MRPS6, ANXA2, PTGER2, GLOl, HAVCR2, PYCARD, LAT2, SLC16A3, COTL1, PKM, TALDOl, CD96, NCR3, KRT86, STMN1, LTB, ARPCIB, ARPC5, FKBP1A, TIMP1, GZMK, CD59, PGK1, RGS10, EVL, RAC2, LGALS1, ITGB7, TUBB, PGAM1, PRF1, GZMB, IL2RB, KLRC2, and KLRBl is higher than expression of said markers in peripheral blood natural killer cells.
9. The method of suppressing the proliferation of tumor cells of any one of claims 1-8, wherein a nucleic acid encoding the cleavage resistant CD 16 has been introduced into the NK cells by transfection.
10. The method of suppressing the proliferation of tumor cells of any one of claims 1-8, wherein a nucleic acid encoding the cleavage resistant CD 16 has been introduced into the NK cells by transduction.
11. The method of suppressing the proliferation of tumor cells of claim 10, wherein a nucleic acid encoding the cleavage resistant CD 16 has been introduced into the NK cells by retroviral transduction.
12. The method of suppressing the proliferation of tumor cells of claim 10, wherein a nucleic acid encoding the cleavage resistant CD 16 has been introduced into the NK cells by lentiviral transduction.
13. The method of suppressing the proliferation of tumor cells of any one of claims 1-12, wherein greater than 90% of the cells in the population are CD56+ and CD3-.
14. The method of suppressing the proliferation of tumor cells of any one of claims 1-13, wherein less than 1% of the cells in the population are CD3+.
15. The method of suppressing the proliferation of tumor cells of any one of claims 1-14, wherein less than 1% of the cells in the population are CD19+.
16. The method of suppressing the proliferation of tumor cells of any one of claims 1-15, wherein greater than 65% of the cells in the population are CD16+.
17. The method of suppressing the proliferation of tumor cells of any one of claims 1-16, wherein the population of cells comprises cells which express one or more surface markers selected from the group consisting of CD226, NKG2D, CD11a, NKp30, NKp44, NKp46, CD94, and combinations thereof.
18. The method of suppressing the proliferation of tumor cells of any one of claims 1-16, wherein the population of cells exhibit greater antibody-dependent cellular cytotoxicity than a population of placental-derived natural killer cells lacking expression of the cleavage resistant CD 16.
19. The method of suppressing the proliferation of tumor cells of claim 1, wherein said contacting is contacting in vitro.
20. The method of suppressing the proliferation of tumor cells of claim 1, wherein said contacting is contacting in vivo.
21. The method of suppressing the proliferation of tumor cells of claim 8, wherein said contacting is in a human.
22. The method of suppressing the proliferation of tumor cells of any one of claims 1-16, wherein the cleavage resistant CD 16 comprises a valine residue at position 158.
23. The method of suppressing the proliferation of tumor cells of any one of claims 1-16, wherein the cleavage resistant CD16 comprises a proline residue at position 197.
24. The method of suppressing the proliferation of tumor cells of any one of claims 1-16, wherein the cleavage resistant CD 16 is CD16VP.
25. The method of suppressing the proliferation of tumor cells of any one of claims 1-16, wherein the cleavage resistant CD 16 does not comprise a proline residue at position 197.
26. The method of suppressing the proliferation of tumor cells of any one of claims 1-16, wherein the tumor cells are tumor cells from a cancer selected from the group consisting of Bladder cancers, Breast cancers, Cervical cancers, Cholangiocarcinomas (extrahepatic), Cholangiocarcinomas (intrahepatic), Colorectal cancers, Esophageal or esophagogastric junction cancers, Gallbladder cancers, Gastric adenocarcinomas, Gastrointestinal stromal tumors, Glioblastoma multiforme, high grade gliomas, Gliomas (low grade), Head and neck carcinomas, Hepatocellular carcinomas, Intestinal (small) malignancies, Kidney cancers, Lung cancers (non small cells), Lung cancers (small cells), Melanomas, Melanomas (uveal), Neuroendocrine tumors, Oligodendrogliomas, Ovarian (epithelial) cancers, Ovarian (non-epithelial) cancers, Pancreatic adenocarcinomas, Penile cancers, Pituitary cancers, Prostate cancers, Sarcomas (peritoneal, retroperitoneal), Sarcomas (soft tissues), Solitary fibrous tumors, Testicular cancers, Thymic cancers, Thyroid cancers, Uterine cancers, and combinations thereof.
27. The method of suppressing the proliferation of tumor cells of any one of claims 1-16, wherein the tumor cells are gastric cancer cells.
28. The method of suppressing the proliferation of tumor cells of any one of claims 1-16, wherein the anti-HER2 antibody is Trastuzumab.
29. A method of treating a HER2+ cancer in a subject, comprising administering to the subject a population of placental-derived natural killer cells comprising a cleavage resistant CD 16 and an antibody, wherein the antibody is an anti-HER2 antibody.
30. The method of suppressing the proliferation of tumor cells of claim 1, wherein the placental-derived natural killer (NK) cells are CYNK cells.
31. The method of claim 2, wherein the CYNK cells are placental CD34+ cell- derived natural killer (NK) cells.
32. The method of any one of claims 2-3, wherein the CYNK cells are characterized by expression of one or more markers selected from the group consisting of FGFBP2, GZMH, CCL3L3, GZMM, CXCR4, ZEB2, KLF2, LITAF, RORA, LYAR, CNOT1, IFNG, DUSP2, ATG2A, CD7, PMAIP1, PPP2R5C, NR4A2, ZFP36L2, PIK3R1, KLRFl, SNHG9, MT2A, RGS2, CHD1, DUSP1, EML4, ZFP36, ZC3H12A, DNAJB6, SBDS, IRFl, TSC22D3, TSPYL2, PNRC1, ISCA1, JUNB, WHAMM, RICTOR, TNFAIP3,
EPC1, MVD, CLK1, ARL4C, REL, KMT2E, YPEL5, AMD1, BTG2, and IDS which is lower than expression of said markers in peripheral blood natural killer cells and / or expression of one or more markers selected from the group consisting of NDFIP2, LINC00996, MAL, CCL1, MB, SPINK2, C15orf48, CAMK1, KLRC1, TNFSF10, TNFRSF18, IL32, CAPG, AC092580.4, S100A11, TNFRSF4, ENOl, FCER1G, CCND2, KRT81, MRPS6, ANXA2, PTGER2, GLOl, HAVCR2, PYCARD, LAT2, SLC16A3, COTL1, PKM, TALDOl, CD96, NCR3, KRT86, STMN1, LTB, ARPC1B, ARPC5, FKBP1A, TIMP1, GZMK, CD59, PGK1, RGS10, EVL, RAC2, LGALS1, ITGB7, TUBB, PGAM1, PRF1, GZMB, IL2RB, KLRC2, and KLRBl which is higher than expression of said markers in peripheral blood natural killer cells.
33. The method of any one of claims 2-4, wherein the CYNK cells are characterized by expression of one or more markers selected from the group consisting of FGFBP2, GZMH, CCL3L3, GZMM, CXCR4, ZEB2, KLF2, LITAF, RORA, LYAR, CNOT1, IFNG, DUSP2, ATG2A, CD7, PMAIP1, PPP2R5C, NR4A2, ZFP36L2, PIK3R1, KLRFl, SNHG9, MT2A, RGS2, CHD1, DUSP1, EML4, ZFP36, ZC3H12A, DNAJB6, SBDS, IRF1, TSC22D3, TSPYL2, PNRC1, ISCA1, JUNB, WHAMM, RICTOR, TNFAIP3, EPC1, MVD, CLK1, ARL4C, REL, KMT2E, YPEL5, AMD1, BTG2, and IDS which is lower than expression of said markers in peripheral blood natural killer cells.
34. The method of claim 4 or claim 5, wherein expression of 2, 3, 4, 5, 6, 7, 8, 9, 10, or more markers selected from the group consisting of FGFBP2, GZMH, CCL3L3, GZMM, CXCR4, ZEB2, KLF2, LITAF, RORA, LYAR, CNOT1, IFNG, DUSP2, ATG2A, CD7, PMAIP1, PPP2R5C, NR4A2, ZFP36L2, PIK3R1, KLRFl, SNHG9, MT2A, RGS2, CHD1, DUSP1, EML4, ZFP36, ZC3H12A, DNAJB6, SBDS, IRF1, TSC22D3, TSPYL2, PNRC1, ISCA1, JUNB, WHAMM, RICTOR, TNFAIP3, EPC1, MVD, CLK1, ARL4C, REL, KMT2E, YPEL5, AMD1, BTG2, and IDS is lower than expression of said markers in peripheral blood natural killer cells.
35. The method of any one of claims 2-6, wherein the CYNK cells are characterized by expression of one or more markers selected from the group consisting of NDFIP2, LINC00996, MAL, CCL1, MB, SPINK2, C15orf48, CAMK1, KLRCl, TNFSF10, TNFRSF18, IL32, CAPG, AC092580.4, S100A11, TNFRSF4, ENOl, FCER1G, CCND2, KRT81, MRPS6, ANXA2, PTGER2, GLOl, HAVCR2, PYCARD, LAT2, SLC16A3, COTL1, PKM, TALDOl, CD96, NCR3, KRT86, STMNl, LTB, ARPC1B, ARPC5, FKBPIA, TIMP1, GZMK, CD59, PGK1, RGS10, EVL, RAC2, LGALS1, ITGB7, TUBB, PGAM1, PRF1, GZMB, IL2RB, KLRC2, and KLRBl which is higher than expression of said markers in peripheral blood natural killer cells.
36. The method of claims 2-7, wherein expression of 2, 3, 4, 5, 6, 7, 8, 9, 10, or more markers selected from the group consisting ofNDFIP2, LINC00996, MAL, CCL1, MB, SPINK2, C15orf48, CAMK1, KLRC1, TNFSF10, TNFRSF18, IL32, CAPG, AC092580.4, S100A11, TNFRSF4, ENOl, FCER1G, CCND2, KRT81, MRPS6, ANXA2, PTGER2, GLOl, HAVCR2, PYCARD, LAT2, SLC16A3, COTL1, PKM, TALDOl, CD96, NCR3, KRT86, STMN1, LTB, ARPC1B, ARPC5, FKBP1A, TIMP1, GZMK, CD59, PGK1,
RGS10, EVL, RAC2, LGALS1, ITGB7, TUBB, PGAM1, PRF1, GZMB, IL2RB, KLRC2, and KLRBl is higher than expression of said markers in peripheral blood natural killer cells.
37. The method of any one of claims 1-8, wherein a nucleic acid encoding the cleavage resistant CD 16 has been introduced into the NK cells by transfection.
38. The method of any one of claims 1-8, wherein a nucleic acid encoding the cleavage resistant CD 16 has been introduced into the NK cells by transduction.
39. The method of claim 10, wherein a nucleic acid encoding the cleavage resistant CD 16 has been introduced into the NK cells by retroviral transduction.
40. The method of claim 10, wherein a nucleic acid encoding the cleavage resistant CD 16 has been introduced into the NK cells by lentiviral transduction.
41. The method of any one of claims 1-12, wherein greater than 90% of the cells in the population are CD56+ and CD3-.
42. The method of any one of claims 1-13, wherein less than 1% of the cells in the population are CD3+.
43. The method of any one of claims 1-14, wherein less than 1% of the cells in the population are CD19+.
44. The method of any one of claims 1-15, wherein greater than 65% of the cells in the population are CD16+.
45. The method of any one of claims 1-16, wherein the population of cells comprises cells which express one or more surface markers selected from the group consisting of CD226, NKG2D, CDlla, NKp30, NKp44, NKp46, CD94, and combinations thereof.
46. The method of any one of claims 1-16, wherein the population of cells exhibit greater antibody-dependent cellular cytotoxicity than a population of placental-derived natural killer cells lacking expression of the cleavage resistant CD 16.
47. The method of claim 1, wherein said contacting is contacting in vitro.
48. The method of claim 1, wherein said contacting is contacting in vivo.
49. The method of claim 8, wherein said contacting is in a human.
50. The method of any one of claims 1-16, wherein the cleavage resistant CD 16 comprises a valine residue at position 158.
51. The method of any one of claims 1-16, wherein the cleavage resistant CD 16 comprises a proline residue at position 197.
52. The method of any one of claims 1-16, wherein the cleavage resistant CD 16 is CD16VP.
53. The method of any one of claims 1-16, wherein the cleavage resistant CD 16 does not comprise a proline residue at position 197.
54. The method of any one of claims 1-16, wherein the tumor cells are tumor cells from a cancer selected from the group consisting of Bladder cancers, Breast cancers, Cervical cancers, Cholangiocarcinomas (extrahepatic), Cholangiocarcinomas (intrahepatic), Colorectal cancers, Esophageal or esophagogastric junction cancers, Gallbladder cancers, Gastric adenocarcinomas, Gastrointestinal stromal tumors, Glioblastoma multiforme, high grade gliomas, Gliomas (low grade), Head and neck carcinomas, Hepatocellular carcinomas, Intestinal (small) malignancies, Kidney cancers, Lung cancers (non small cells), Lung cancers (small cells), Melanomas, Melanomas (uveal), Neuroendocrine tumors, Oligodendrogliomas, Ovarian (epithelial) cancers, Ovarian (non-epithelial) cancers, Pancreatic adenocarcinomas, Penile cancers, Pituitary cancers, Prostate cancers, Sarcomas (peritoneal, retroperitoneal), Sarcomas (soft tissues), Solitary fibrous tumors, Testicular cancers, Thymic cancers, Thyroid cancers, Uterine cancers, and combinations thereof.
55. The method of any one of claims 1-16, wherein the tumor cells are gastric cancer cells.
56. The method of any one of claims 1-16, wherein the anti-HER2 antibody is Trastuzumab.
57. The method of claim 27, wherein said population of placental-derived natural killer cells comprising a cleavage resistant CD 16 and/or said anti-HER2 antibody are administered intravenously.
58. The method of claim 27 or 36, wherein the population of placental-derived natural killer cells comprising a cleavage resistant CD16 and the anti-HER2 antibody are administered sequentially.
59. The method of claim 43, wherein the population of placental-derived natural killer cells comprising a cleavage resistant CD 16 and the anti-HER2 antibody are administered concurrently.
60. A population of human placental-derived natural killer cells comprising a cleavage resistant CD 16 for use in the manufacture of a medicament for treatment of a HER2+ cancer in a subject.
61. Use of a composition comprising a population of human placental-derived natural killer cells comprising a cleavage resistant CD 16 for treatment of a HER2+ cancer in a subject.
PCT/US2020/063670 2019-12-05 2020-12-07 Her2+ cancer treatment with populations of natural killer cells comprising a cleavage resistant cd16 WO2021113849A1 (en)

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US201962944334P 2019-12-05 2019-12-05
US201962944341P 2019-12-05 2019-12-05
US62/944,334 2019-12-05
US62/944,341 2019-12-05
US202063035496P 2020-06-05 2020-06-05
US202063035377P 2020-06-05 2020-06-05
US63/035,496 2020-06-05
US63/035,377 2020-06-05
US202063115953P 2020-11-19 2020-11-19
US63/115,953 2020-11-19

Publications (1)

Publication Number Publication Date
WO2021113849A1 true WO2021113849A1 (en) 2021-06-10

Family

ID=74125670

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/063670 WO2021113849A1 (en) 2019-12-05 2020-12-07 Her2+ cancer treatment with populations of natural killer cells comprising a cleavage resistant cd16

Country Status (1)

Country Link
WO (1) WO2021113849A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115521913A (en) * 2022-02-22 2022-12-27 北京景达生物科技有限公司 Combined application of NK cells and CD20, CD38 and Her2 antibodies
WO2023198162A1 (en) * 2022-04-13 2023-10-19 星奕昂(上海)生物科技有限公司 Anti-splicing mutant of cd16 for enhancing cell function

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5372581A (en) 1993-07-21 1994-12-13 Minneapolis Children's Services Corporation Method and apparatus for placental blood collection
US5415665A (en) 1991-03-19 1995-05-16 Utah Medical Products, Inc. Umbilical cord clamping, cutting, and blood collecting device and method
US20040048796A1 (en) 2002-03-26 2004-03-11 Hariri Robert J. Collagen biofabric and methods of preparation and use therefor
US7045148B2 (en) 2000-12-06 2006-05-16 Anthrogenesis Corporation Method of collecting placental stem cells
US7147626B2 (en) 2004-09-23 2006-12-12 Celgene Corporation Cord blood and placenta collection kit
US7255879B2 (en) 2000-12-06 2007-08-14 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
US20070190042A1 (en) 2005-12-29 2007-08-16 Edinger James W Composition for collecting and preserving placental stem cells and methods of using the composition
US20070275362A1 (en) 2000-12-06 2007-11-29 James Edinger Placental stem cell populations
US7498171B2 (en) 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US20090104164A1 (en) 2007-09-26 2009-04-23 Celgene Cellular Therapeutics Angiogenic cells from human placental perfusate
US20170174743A1 (en) * 2014-03-28 2017-06-22 Regents Of The University Of Minnesota Polypeptides, cells, and methods involving engineered cd16
WO2018126074A1 (en) * 2016-12-30 2018-07-05 Celularity, Inc. Genetically modified natural killer cells

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5415665A (en) 1991-03-19 1995-05-16 Utah Medical Products, Inc. Umbilical cord clamping, cutting, and blood collecting device and method
US5372581A (en) 1993-07-21 1994-12-13 Minneapolis Children's Services Corporation Method and apparatus for placental blood collection
US7468276B2 (en) 2000-12-06 2008-12-23 Anthrogenesis Corporation Placental stem cells
US7045148B2 (en) 2000-12-06 2006-05-16 Anthrogenesis Corporation Method of collecting placental stem cells
US8057788B2 (en) 2000-12-06 2011-11-15 Anthrogenesis Corporation Placental stem cell populations
US7255879B2 (en) 2000-12-06 2007-08-14 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
US20070275362A1 (en) 2000-12-06 2007-11-29 James Edinger Placental stem cell populations
US20040048796A1 (en) 2002-03-26 2004-03-11 Hariri Robert J. Collagen biofabric and methods of preparation and use therefor
US7498171B2 (en) 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
US7147626B2 (en) 2004-09-23 2006-12-12 Celgene Corporation Cord blood and placenta collection kit
US20070190042A1 (en) 2005-12-29 2007-08-16 Edinger James W Composition for collecting and preserving placental stem cells and methods of using the composition
US20090104164A1 (en) 2007-09-26 2009-04-23 Celgene Cellular Therapeutics Angiogenic cells from human placental perfusate
US20170174743A1 (en) * 2014-03-28 2017-06-22 Regents Of The University Of Minnesota Polypeptides, cells, and methods involving engineered cd16
WO2018126074A1 (en) * 2016-12-30 2018-07-05 Celularity, Inc. Genetically modified natural killer cells

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
COIF LABANKOVICH AJHANICK NA ET AL.: "How a single T cell receptor recognizes both self and foreign MHC", CELL, vol. 129, no. 1, 2007, pages 135 - 146, XP055103907, DOI: 10.1016/j.cell.2007.01.048
DEWOLF SSYKES M: "Alloimmune T cells in transplantation", J CLIN INVEST, vol. 127, no. 7, 2017, pages 2473 - 2481
IRENE RAITMAN ET AL: "Abstract 2218: High affinity and cleavage resistant CD16 enhances Trastuzumab mediated killing of gastric cancer targets by human placental CD34+-derived natural killer cells", IMMUNOLOGY, 15 August 2020 (2020-08-15), US, pages 2218 - 2218, XP055740096, ISSN: 0953-4954, DOI: 10.1158/1538-7445.AM2020-2218 *
IUPAC-IUB COMMISSION ON BIOCHEMICAL NOMENCLATURE, BIOCHEM, vol. 11, 1972, pages 942 - 944
JING YNI ZWU JHIGGINS LMARKOWSKI TWKAUFMAN DSWALCHECK B: "Identification of an ADAM17 cleavage region in human CD16 (FcyRIII) and the engineering of a non-cleavable version of the receptor in NK cells", PLOS ONE, vol. 10, no. 3, 27 March 2015 (2015-03-27), pages e0121788, XP055218471, DOI: 10.1371/journal.pone.0121788
KOENE HRKLEIJER MALGRA JROOS DVON DEM BORNE AEDE HAAS M: "Fc gammaRIIIa-158V/F polymorphism influences the binding of IgG by natural killer cell Fc gammaRIIIa, independently of the Fc gammaRIIIa-48L/R/H phenotype", BLOOD, vol. 90, no. 3, 1 August 1997 (1997-08-01), pages 1109 - 14
MARCUS AGOWEN BGTHOMPSON TWIANNELLO AARDOLINO MDENG W ET AL.: "Recognition of tumors by the innate immune system and natural killer cells", ADV IMMUNOL, vol. 122, 2014, pages 91 - 128, XP055536910, DOI: 10.1016/B978-0-12-800267-4.00003-1
SHUYANG HE ET AL: "EXTH-16. HUMAN PLACENTAL HEMATOPOIETIC STEM CELL DERIVED NATURAL KILLER CELLS FOR GLIOBLASTOMA IMMUNOTHERAPY", NEURO-ONCOLOGY, vol. 21, no. Supplement_6, 11 November 2019 (2019-11-11) - 24 November 2009 (2009-11-24), US, pages vi85 - vi85, XP055723628, ISSN: 1522-8517, DOI: 10.1093/neuonc/noz175.350 *
SUGITA NYAMAMOTO KKOBAYASHI TVAN DER POL WHORIGOME TYOSHIE HVAN DE WINKEL JGHARA K: "Relevance of Fc gamma RIIIa-158V-F polymorphism to recurrence of adult periodontitis in Japanese patients", CLIN EXP IMMUNOL, vol. 117, no. 2, August 1999 (1999-08-01), pages 350 - 4
WU JEDBERG JCREDECHA PBBANSAL VGUYRE PMCOLEMAN KSALMON JEKIMBERLY RP: "A novel polymorphism of FcgammaRIIIa (CD 16) alters receptor function and predisposes to autoimmune disease", J CLIN INVEST, vol. 100, no. 5, 1 September 1997 (1997-09-01), pages 1059 - 70, XP002921994, DOI: 10.1172/JCI119616
XUAN GUO ET AL: "Celularity Engineering High Affinity and Cleavage Resistant CD16 to Augment ADCC of Placental Hematopoietic Stem Cells-Derived Natural Killer Cells", 13 November 2019 (2019-11-13), XP055740076, Retrieved from the Internet <URL:https://www.celularityinc.com/wp-content/uploads/2020/05/Engineering-High-Affinity-Cleavage-Resistant-CD16-to-Augment-ADCC-of-Placental-Derived-NK-Cells.pdf> [retrieved on 20201014] *
ZHANG ET AL., J IMMUNOTHER CANCER, 2015

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115521913A (en) * 2022-02-22 2022-12-27 北京景达生物科技有限公司 Combined application of NK cells and CD20, CD38 and Her2 antibodies
WO2023198162A1 (en) * 2022-04-13 2023-10-19 星奕昂(上海)生物科技有限公司 Anti-splicing mutant of cd16 for enhancing cell function

Similar Documents

Publication Publication Date Title
JP2022062020A (en) Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
JP6797803B2 (en) How to use natural killer cells to treat blood disorders, solid tumors, or infectious diseases
JP2022023148A (en) Natural killer cells and ilc3 cells as well as uses thereof
JP2022078099A (en) Method for generating natural killer cells
Hofmeister et al. Ex vivo expansion of umbilical cord blood stem cells for transplantation: growing knowledge from the hematopoietic niche
JP2020188782A (en) Natural killer cell and use of the same
AU2018253115B2 (en) Aryl hydrocarbon receptor antagonists and uses thereof
JP2022511786A (en) Proliferation of natural killer cells and ILC3 cells with novel aromatic compounds
CA3152281A1 (en) Cbl inhibitors and compositions for expansion of immune cells
WO2021113849A1 (en) Her2+ cancer treatment with populations of natural killer cells comprising a cleavage resistant cd16
AU2022287584A1 (en) Methods of treating acute myeloid leukemia and multiple myeloma using natural killer cells
US20230355759A1 (en) Populations of natural killer cells comprising a cleavage resistant cd16
US20220265712A1 (en) Populations of natural killer cells for treating cancers
CN113853204A (en) Expansion of expanded NK cell fractions suitable for transplantation combination therapy and uses thereof
US20190231823A1 (en) Rejuvenated aged hematopoietic stem cells and methods of use
US20220273716A1 (en) Populations of natural killer cells comprising a cd38 chimeric antigen receptor
KR20190039988A (en) Substituted azole derivatives for the production, proliferation and differentiation of hematopoietic stem and progenitor cells
Liu et al. Cotransplantation of human umbilical cord blood-derived stromal cells enhances hematopoietic reconstitution and engraftment in irradiated BABL/c mice
WO2023137344A1 (en) Cleavage resistant cd16 constructs and uses thereof
CA3063674A1 (en) Selection and use of umbilical cord cell fractions suitable for transplantation
WO2023010123A1 (en) Placenta-dervied nk cells as a senolytic for therapeutic and other uses
AU2021201896A1 (en) Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
WO2023278628A1 (en) Human placental hematopoietic stem cell derived natural killer cells in acute myeloid leukemia (aml) remission with minimal residual disease (mrd) or relapsed/refractory aml
US20220000919A1 (en) Placental derived natural killer cells for treatment of coronavirus infections
Tocci et al. CD34+ cells from first-trimester fetal blood are enriched in primitive hemopoietic progenitors

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: 20835929

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: 20835929

Country of ref document: EP

Kind code of ref document: A1