US20170360907A1 - Compositions and methods for preventing and treating graft versus host disease - Google Patents

Compositions and methods for preventing and treating graft versus host disease Download PDF

Info

Publication number
US20170360907A1
US20170360907A1 US15/532,740 US201515532740A US2017360907A1 US 20170360907 A1 US20170360907 A1 US 20170360907A1 US 201515532740 A US201515532740 A US 201515532740A US 2017360907 A1 US2017360907 A1 US 2017360907A1
Authority
US
United States
Prior art keywords
cells
ilc2
expression
allo
subject
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/532,740
Inventor
Jonathan Serody
James Coghill
Danny Bruce
Bruce Blazar
Heather Stefanski
Benjamin Vincent
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of North Carolina at Chapel Hill
University of Minnesota
Original Assignee
University of North Carolina at Chapel Hill
University of Minnesota
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of North Carolina at Chapel Hill, University of Minnesota filed Critical University of North Carolina at Chapel Hill
Priority to US15/532,740 priority Critical patent/US20170360907A1/en
Assigned to THE UNIVERSITY OF NORTH CAROLINA AT CHAPEL HILL reassignment THE UNIVERSITY OF NORTH CAROLINA AT CHAPEL HILL ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SERODY, JONATHAN, VINCENT, BENJAMIN, BRUCE, DANNY, COGHILL, JAMES
Assigned to REGENTS OF THE UNIVERSITY OF MINNESOTA reassignment REGENTS OF THE UNIVERSITY OF MINNESOTA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BLAZAR, BRUCE, STEFANSKI, Heather
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIV OF NORTH CAROLINA CHAPEL HILL
Publication of US20170360907A1 publication Critical patent/US20170360907A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/001Preparations to induce tolerance to non-self, e.g. prior to transplantation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • 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
    • C12N5/0651Lymph nodes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/122Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells for inducing tolerance or supression of immune responses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/124Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5154Antigen presenting cells [APCs], e.g. dendritic cells or macrophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/577Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 tolerising response
    • 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
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2307Interleukin-7 (IL-7)
    • 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
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2333Interleukin-33 (IL-33)

Definitions

  • the presently disclosed subject matter relates to compositions and methods for preventing and treating graft versus host disease (GvHD) and/or idiopathic pneumonia syndrome (IPS).
  • the presently disclosed subject matter further relates to methods of making compositions for preventing and treating GvHD and/or IPS.
  • corticosteroid therapy is associated with a substantial number of significant side-effects including infectious complications, bone disturbances and profound metabolic changes.
  • the outcome for patients with steroid-refractory aGvHD is dismal with approximately 10% of these patients surviving one year after the diagnosis and little progress in improving this outcome over the past 20 years.
  • idiopathic pneumonia syndrome is a common pulmonary complication and remains a severe threat to survival after allo-SCT and allo-BMT. IPS often coincides with the onset of GvHD.
  • the compositions comprise a therapeutically effective amount of a cell preparation of IL-33 activated type 2 innate immune cells (ILC2), and a pharmaceutically acceptable carrier.
  • ILC2 cells have the following properties: test positive for expression of lymphoid marker CD90, test positive for expression of lymphoid marker ICOS, and test negative for expression of lympohoid marker lin.
  • the ILC2 cells test positive for expression of CD127, ST2, Sca-1, CD25, CD90, and ICOS, and test negative for expression of lin.
  • the ILC2 cells are activated by cytokines IL-33, IL-25, IL-7 and/or combinations thereof.
  • the ILC2 cells are derived from human cord blood cells.
  • the pharmaceutical composition is substantially free of MPP2 cells.
  • ILC2 cells in vitro methods for generating activated ILC2 cells, comprising in some embodiments collecting human cord blood from a subject, isolating c-Kit positive cells from the cord blood, and culturing the c-Kit positive cells in the presence of an IL-33 cytokine, whereby IL-33 activated ILC2 cells are generated.
  • the ILC2 cells are activated by exposing the c-Kit positive cells in culture to cytokines IL-33, IL-25, IL-7 and/or combinations thereof.
  • the in vitro methods further comprise analyzing the generated ILC2 cells by measuring expression of ST2 and CD127 in the ILC2 cells, whereby expression of ST2 and CD127 is indicative of the phenotype of ILC2 cells.
  • the ILC2 cells are substantially free of MPP2 cells.
  • the subject is a human patient or donor.
  • graft versus host disease GvHD
  • IPS idiopathic pneumonia syndrome
  • the ILC2 cells have the following properties: test positive for expression of lymphoid marker CD90, test positive for expression of lymphoid marker ICOS, and test negative for expression of lympohoid marker lin.
  • the ILC2 cells test positive for expression of CD127, ST2, Sca-1, CD25, CD90, and ICOS, and test negative for expression of lin.
  • the subject is a human subject receiving an allogeneic stem cell transplant (allo-SCT) or allogeneic bone marrow transplant (allo-BMT).
  • allo-SCT allogeneic stem cell transplant
  • allo-BMT allogeneic bone marrow transplant
  • the cell preparation of IL-33 activated ILC2 cells is administerd to the subject within 1 to 30 days after receiving the allo-SCT or allo-BMT.
  • the risk and/or severity of acute GVHD associated with allo-SCT or allo-BMT is reduced.
  • the cell preparation of IL-33 activated ILC2 cells is administered as a co-infusion with the allo-SCT or allo-BMT.
  • production of Th2 cytokines in the subject is increased, and/or production of Th1 and/or Th17 cytokines in the subject is decreased.
  • kits for preventing GvHD or IPS in a subject comprising in some embodiments providing a subject receiving allo-SCT or allo-BMT, and administering to the subject a therapeutically effective amount of a cell preparation of IL-33 activated ILC2 cells.
  • the cell preparation of IL-33 activated ILC2 cells is administered as a co-infusion with the allo-SCT or allo-BMT.
  • the co-infusion of ILC2 cells with the allo-SCT or allo-BMT comprises infusion of the ILC2 cells at a time sufficiently proximate to the allo-SCT or allo-BMT to prevent or substantially prevent onset of GvHD or IPS.
  • the ILC2 cells have the following properties: test positive for expression of lymphoid marker CD90; test positive for expression of lymphoid marker ICOS; and test negative for expression of lympohoid marker lin.
  • the ILC2 cells test positive for expression of CD127, ST2, Sca-1, CD25, CD90, and ICOS, and test negative for expression of lin.
  • production of Th2 cytokines in the subject is increased, and/or production of Th1 and/or Th17 cytokines in the subject is decreased.
  • the subject comprises a human subject receiving allo-SCT or allo-BMT.
  • production of Th1 and/or Th17 cytokines is decreased in the subject.
  • the ILC2 cells have the following properties: test positive for expression of lymphoid marker CD90, test positive for expression of lymphoid marker ICOS, and test negative for expression of lympohoid marker lin.
  • the ILC2 cells test positive for expression of CD127, ST2, Sca-1, CD25, CD90, and ICOS, and test negative for expression of lin.
  • FIGS. 1A-1E are a series of bar graphs showing that ILC2 cells are radiation sensitive. Comparison of unconditioned cells (white bars) or 24 hours after receiving radiation (950cGy) (grey bars) as analyzed by flow cytometry.
  • FIG. 1A ILC3 from the LP are radiation resistant.
  • ILC3 from MLN FIG. 1B
  • LP CD4 T cells FIG. 1C
  • ILC2 cells from the LP FIG. 1D
  • MLN FIG. 1E
  • This represents two independent experiments, n 4 for each. T test using GraphPad TM software (GraphPad Software, Inc., La Jolla, Calif., United States of America), ** p ⁇ 0.05.
  • FIG. 2 is a line graph showing that IL-33 with IL-7 (open circles) treatment increases ILC2 proliferation in vitro compared to IL-25 with IL-7 (solid squares) (10 ng/ml ea.), adjusted average cell number ⁇ SEM.
  • FIGS. 3A-3D are a series of plots showing that co-transplant of ILC2 cells reduces aGvHD.
  • Lethally irradiated B6D2 mice (950cGy) received 3.0 ⁇ 106 TCD BM (BM only), BM plus 4.0 ⁇ 106 total splenic T cells (Ctrl) or BM plus T cells with 4.0 ⁇ 106 IL-33 activated ILC2 cells (Treatment) in an ILC2 (Top row) or recipients received 4.0 ⁇ 106 IL-33 activated ILC2 cells at the time of allo-BMT or 7 days later (Bottom row)
  • FIG. 3A Percent survival after allo-SCT.
  • FIG. 3B GvHD scores over time.
  • FIG. 3C Percent survival in timed injection study.
  • FIGS. 4A-4E are a series of bar graphs showing that co-transplant of ILC2 cells reduces donor T cells.
  • Lethally irradiated B6D2 mice (950cGy) received 3.0 ⁇ 106 TCD BM (BM only), BM plus 4.0 ⁇ 106 total splenic GFP+ T cells (Ctrl), BM plus GFP+ T cells with 4.0 ⁇ 106 ILC2 cells (Treat 1:1) or BM plus GFP+ T cells with 8.0 ⁇ 106 ILC2 cells (Treat 1:2).
  • Donor T cells were evaluated in the GI tract 12 days after BMT.
  • FIG. 4A ELISA quantification of GFP (Cell Biolabs: AKR-121; Cell Biolabs, Inc., San Diego, Calif., United States of America) in tissue homogenates on day 12 post BMT from Ctrl (open bars) and Treat 1:1 (grey bars) normalized to grams of tissue.
  • Donor T cells from BMT (as described for FIG. 3A ) were evaluated by flow cytometry 12 days after BMT, Ctrl (open bars) and Treatment (grey bars).
  • FIG. 4B Percentage of donor T cells in the LP lymphocyte compartment (LPL).
  • FIG. 4C The total number of CD4+ and CD8+ donor T cells in the LP.
  • FIG. 4D Total number of IFN- ⁇ producing donor Th1 and Tc1 and the number of donor IL-17A producing Th17 cells.
  • FIGS. 5A and 5B are a set of bar graphs showing that IL-33 activated ILC2 migrate to GvHD target organs and secrete Th2 cytokines.
  • ILC2 cells were tracked using B6-GFP ILC2 donor mice for injection of IL-25 and in vitro expansion with IL-33 prior to co-infusion at BMT.
  • FIG. 5A ELISA quantification of ILC2 derived GFP in GvHD target organs.
  • FIG. 5B Bar graph of average IL-13 and IL5 IC cytokine ⁇ SEM in donor ILC2 cells day 12 post BMT.
  • FIG. 6 is a bar graph showing enumeration of ILC2 cells after infusion from lung parenchyma.
  • 1 ⁇ 10 6 ILC2 cells from eGFP+ mice were infused i.v. and 48 hours later evaluated from the lung parenchyma in B6D2 mice that had been lethally irradiated and received 3 ⁇ 10 6 B6 TCD bone marrow cells and 3.5 ⁇ 106 B 6 T cells.
  • ILC2 cells were distinguished by the expression of GFP and in addition expression of ST2, CD127 and CD90 and absence of lineage markers. There was a strong statistically significant increase in ILC2 cells in the lung after i.v. infusion (hatched bar) that was not found in the sham treated animals (open bar) (p ⁇ 0.001).
  • FIG. 7A is a bar graph indicating that there is no difference in the generation of T cells making IL-4 (Th2) after the infusion of ILC2 cells when comparing mice that received ILC2 cells (grey bar) and those receiving sham treatment (open bar) 12 days after bone marrow transplantation.
  • FIG. 7B illustrates the outcome of recipient mice receiving lethal irradiation (BM only) and infused with haploidentical wild type T cells with ILC2 cells (2:1) ratio (Control) or haploidentical STAT6 knockout T cells with ILC2 cells (2:1) ratio (Treated).
  • FIG. 7A is a bar graph indicating that there is no difference in the generation of T cells making IL-4 (Th2) after the infusion of ILC2 cells when comparing mice that received ILC2 cells (grey bar) and those receiving sham treatment (open bar) 12 days after bone marrow transplantation.
  • FIG. 7B illustrates the outcome of recipient mice receiving lethal irradiation (BM only) and infused with
  • FIG. 7C is a plot showing the clinical scoring of recipient mice receiving lethal irradiation (triangle—BM only) and infused with either haploidentical WT T cells (closed circle—Control) or STAT6 knockout T cells plus ILC2 cells (open circle—Treated).
  • FIG. 7D is a bar graph showing an evaluation for FoxP3 expressing T cells in the GI tract 12 days after the administration of haploidentical WT T cells (open bar) or WT T cells supplemented 2:1 with ILC2 cells (closed bar).
  • FIGS. 8A to 8C are a series of graphs and plots showing the results of an evaluating for the presence of donor antigen presenting cells in the GI tract in recipient mice after bone marrow transplant receiving ILC2 cells plus haploidentical T cells versus those receiving only haploidentical T cells.
  • FIG. 8A is a plot showing the results of tolerogeneic dendritic cells from donor mice being enumerated using CD11c and CD103 as markers.
  • FIG. 8B is a bar graph demonstrating that there is no increase in FoxP3-expressing T cells in the GI tract after bone marrow transplantation comparing mice receiving ILC2 cells (grey bar) with control treatment (open bar).
  • FIG. 8C is a plot showing the results of haplioidentical mice receiving lethal irradiation (BM only) and transplantation using wild type bone marrow cells supplemented with T cells with (Treatment) and without (Control) ILC2 cells.
  • FIGS. 9A to 9E are a series of plots and graphs showing the results of the permeability of the GI tract from day 4 to 20 by administering FITC-dextran to mice after bone marrow transplantation which received either T cells with sham ILC2 infusion versus T cells with ILC2 infusion.
  • Treatment groups in FIGS. 9A to 9C are as follows: Normal (lined bar); BM only (checkered bar); Control (open bar), Treated (grey bar).
  • Treatment gropus in FIGS. 9D and 9E are as follows: BM only (triangle); Control (closed circle); Treatment (closed circle).
  • GvHD is mediated by donor T cells that recognize major and minor histocompatibility differences in the host. Evaluations using pre-clinical models and clinical studies have indicated that Th1 T cells and perhaps Th17 cells may be mediators of aGvHD. Moreover, in accordance with the presently disclosed subject matter, Th1 cells have been found to be involved in the pathogenesis of aGvHD involving the small bowel and colon.
  • Th1 bias for GvHD involving the GI tract is not clear, although previous investigations have implicated the release of LPS and cytokine activation by host innate immune cells as a result of bacterial translocation after conditioning therapy. However, targeting this axis has not been substantially beneficial in the treatment of patients with steroid-refractory aGvHD.
  • innate immune cells that express similar cytokines and transcription factors compared to polarized T cells but lack germ-line encoded recombined receptors have been identified.
  • An innate immune population of cells that generates Th2 cytokines is termed nuocytes or type 2 innate immune cells (ILC2). These cells generate substantial quantities of IL-5, and IL-13 with more modest expression of IL-4. These cells are involved in Th2 cell generation of IL-13 and the host response to specific parasitic infections.
  • Human ILC2 cells can be found in the GI tract and lung and can be defined by their expression of CD161 and the chemoattractant, CRTH2 (Mjosberg et al., Nat Immunol. 2011; 12(11):1055-1062). Like murine ILCs, human ILC2 cells can be expanded in vitro and produce significant quantities of IL-5 and IL-13 in response to IL-7 and IL-33 (Yu et al., Mucosal Immunol. 2013).
  • T lymphocytes are characterized by the expression of the T cell receptor (TCR), which requires recombination of variable, diversity, joining and constant gene segments to produce a TCR capable of binding to peptide/MHC proteins.
  • TCR T cell receptor
  • APCs APCs
  • T cell activation, proliferation and differentiation leading to the generation of the adaptive immune response As this process takes 7-10 days, mechanisms are needed to control pathogen growth during this time frame.
  • the immune system has innate immune cells that can recognize pathogen-associated molecular patterns and/or damage-associated molecular patterns and initiate an immune response.
  • This aspect of the immune response which includes neutrophils, macrophages, dendritic cells and monocytes, is a critical part of the initial immune response.
  • NK cells that generate IFN- ⁇ were discovered (Kiessling et al., 1976). Following this, other populations of innate cells were found. These cells were often located at mucosal sites such as the GI tract and lung, where they served a critical role as “first responders” to infectious pathogens.
  • ILC1 gene IFN- ⁇ similar to NK cells
  • ILC2 ILC3 Is
  • the first population of innate cells are lymphoid tissue inducer cells (LTi). These cells express the transcription factor RORyt and are involved in the generation of lymphoid tissue.
  • lymph nodes LN and Peyer's patches (PP).
  • LN lymph nodes
  • PP Peyer's patches
  • LTi cells generated IL-17A and could be identified in mice after birth.
  • a second group of cells that resemble LTi cells but are phenotypically distinct has been identified in mice after birth. These cells generate IL-22 but not IL-17A and are found at mucosal sites mostly in the small bowel and colon.
  • ILCs express NK cell-associated cytotoxicity receptors NKp44 in humans and NKp46 in mice. They are distinct from a population of ILCs that generates IL-17A and lacks the expression of NK receptors.
  • These IL-22 generating ILCs critically require the expression of the transcription factor Ahr56.
  • ILC3 cells Innate immune cells that generate IL-17 and/or IL-22 have been termed ILC3 cells. Following the description of these cells, a second entirely different population of innate lymphoid cells was characterized. Previous work had suggested that the cytokine, IL-25, affected a non-T, B or NK cell.
  • the instant disclosure pertains, at least in part, to ILC2 cells.
  • ILC2 cells Such cells have in some embodiments been found to express CD127, CD117, CD90 and CD25, respond to IL-25 and IL-33, and generate IL-5 and IL-13.
  • MPP2 multipotent progenitor 2
  • IL-25-responsive a second but distinct population of cells that is IL-25-responsive is termed multipotent progenitor 2 (MPP2) cells and can generate multiple different myeloid cells.
  • MPP2 multipotent progenitor 2
  • ILC2 cells in the presence of IL-33 significantly increases their expression of IL-4, IL-5 and IL-13 and enhanced cell proliferation.
  • ILC2 and MPP2 cells share common characteristics such as cytokine profile, lack of lineage markers and response to IL-25 and IL-33, their discordant expression of CD25, CD127 and CD90 and distinct multipotent potential suggest they are distinct populations.
  • Type 2 ILC share similarities to Th2 and Treg cells expressing high levels of Th2 cytokines as well as regulatory receptors that are constitutively expressed on Tregs.
  • MPP2 cells can give rise to monocytes, macrophages, eosinophils and mast cells. These progeny are competent antigen presenting cells and the adoptive transfer of MPP2 cells promoted Th2 cytokine responses. IL-25 and IL-33 stimulation of ILC2 cells promoted M2 macrophage recruitment to visceral associated adipose tissue.
  • ILC2 cells require the expression of the transcription factor Id268. Like Th2 cells, ILC2 cells express GATA-3, MAF and STATE. ILC2 cells appear to be important for the generation of T cells making IL-13. ILC2 cells are important to the immune response to helminths such as Nippostrongyloides brasiliensis, which require IL-25 for an adequate immune response. These data support a framework by which ILC2 cells are relevant in providing a suitable environment for the generation of Th2 T cells. These Th2 cells along with MPP2 cells can assist in the recruitment and activation of mast cells, basophils, eosinophils and B cells allowing for clearance of parasitic infections. However, overexuberant ILC2/Th2 responses may be detrimental as clinical syndromes such as asthma may be increased after ILC2/Th2 activation.
  • therapeutic treatments and composition based on the role ILC2 cells play in the pathogenesis of GvHD and IPS both of which are associated with allo-SCT and/or allo-BMT.
  • protocols and methods are provided for isolating, expanding, activating and identifying ILC2 cells, in some embodiments in vitro and/or ex vivo, to be used in therapeutic methods and compositions for GvHD and IPS.
  • ILC2 cells in some embodiments in vitro and/or ex vivo, to be used in therapeutic methods and compositions for GvHD and IPS.
  • IL-25 and/or IL-33 in some embodiments with IL-7) can activate ILC2 cells inducing production of IL-4, IL-5 and IL-13.
  • Activated ILC2 cells can in some embodiments be used in therapeutic methods and compositions to treat and/or prevent GvHD and IPS by, at least in some aspects, shifting the cytokine micro-environment away form Th1/Th17 to Th2.
  • ILC2 cells can in some embodiments be highly sensitive to radiation.
  • the absence of recipient ILC2 cells in the colon and MLN may lead to alterations in the microenvironment leading to Th1 polarization in the lower GI tract induced after allo-SCT or allo-BMT.
  • compositions for treating GvHD and/or IPS can comprise a therapeutically effective amount of a cell preparation of IL-33 activated ILC2 cells.
  • the ILC2 cells in such a composition can be further characterized as testing positive for expression of lymphoid marker CD90 and/or ICOS, and/or testing negative for expression of lineage-specific lymphoid markers.
  • the ILC2 cells can in some embodiments test positive for expression of CD127, ST2, Sca-1, CD25, CD90, and/or ICOS, and/or test negative for expression of lineage markers.
  • ILC2 cells in addition to being activated by IL-33, ILC2 cells can in some aspects be activated by cytokines IL-25 and/or IL-7, alone or in one or more combinations with IL-33.
  • the pharmaceutical compositions provided herein, including those comprising activated ILC2 cells can be screened for and confirmed to be completely or substantially free of MPP2 cells.
  • the ILC2 cells that are grown and/or activated in vitro can be derived from any suitable source of ILC2 cells, including for example mammalian blood and/or blood products.
  • the ILC2 cells can be derived from human cord blood cells.
  • human cord blood cells can be derived from a donor subject and/or from a patient's own cord blood.
  • compositions provided herein can in some embodiments comprise a pharmaceutically acceptable carrier.
  • in vitro, or otherwise ex vivo, methods are also provided herein for generating activated ILC2 cells, comprising expansion of ILC2 cells from a small number of cells, and activation of such cells to generate cytokines.
  • Such methods can provide the activated ILC2 cells for use in the disclosed pharmaceutical compositions and related therapeutic methods and approaches.
  • These in vitro methods can in some embodiments comprise collecting human cord blood (or other potential source of ILC2 cells from a subject such as peripheral blood stem cells and/or bone marrow), isolating c-Kit positive cells from the cord blood, and culturing the c-Kit positive cells in the presence of an IL-33 cytokine.
  • the ILC2 cells can be activated.
  • additional cytokines can exposed to the cultured ILC2 cells to further elicit an activated state, including for example IL-25 and IL-7.
  • the IL-33, IL-25 and IL-7 cytokines can be exposed to the ILC2 cells in culture any combination thereof.
  • a further step can comprise analyzing the generated ILC2 cells by measuring expression of ST2 and CD127 in the ILC2 cells. Expression of ST2 and CD127 is indicative of the phenotype of ILC2 cells. Such a confirmatory step can rule out the presence of MPP2 cells such that a composition can be characterized as being free or substantially free of MPP2 cells. Additionally, characterization of human ILC2 cells found that the cells expressed CRTH2 and CD161 characteristic surface markers for the expression of ILC2 cells.
  • treatment or therapeutic methods are provided herein, including treatments and therapeutic approaches for GvHD and/or IPS.
  • methods for treating GvHD or IPS in a subject can comprise providing a subject in need of treatment for GvHD and/or IPS, and administering to the subject a therapeutically effective amount of a cell preparation of IL-33 activated ILC2 cells.
  • the cell preparation administered to a subject can comprise any cell preparation or composition as disclosed herein.
  • ILC2 cells can have properties including, but not limited to expression of lymphoid markers CD90 and/or ICOS, and/or absence of lymphoid marker lin.
  • the ILCS cell composition administered to a subject can be characterized by testing positive for expression of CD127, ST2, Sca-1, CD25, CD90, and ICOS, and test negative for expression of lin.
  • the subjects to be treated with the disclosed methods of treatment can comprise any subject in need of therapy, including any subject suffering from or susceptible to GvHD, IPS or related condition.
  • the subject can in some embodiments be a human subject receiving an allo-SCT or allo-BMT
  • the cell preparation of IL-33 activated ILC2 cells can be administered to the subject within 1 to 30 days after receiving the allo-SCT or allo-BMT. In some embodiments, the cell preparation of IL-33 activated ILC2 cells can be administered to the subject within 1 to 365 days after the onset or diagnosis of lower GI tract inflammatory GvHD or IPS.
  • the methods of treatment disclosed herein can in some embodiments reduce the risk and/or severity of acute GvHD associated with allo-SCT or allo-BM by preventing the onset of these conditions or significantly improving the survivability and/or treatability of the conditions.
  • GvHD and/or IPS can be completely or substantially reversed or overcome.
  • the administration of activated ILC2 cells as provided herein can completely eliminate or substantially decrease the clinical score of lower GI tract GVHD commensurate with a marked decrease in diarrhea and/or abdominal pain or blood in the stool.
  • treatement methods and therapeutic compositions as disclsoed herein can also reverse the symptoms and signs of IPS, such as but not limited to shortness of breath, hypoxemia, or increased work of breathing.
  • Such preventative methods can comprise providing a subject receiving allo-SCT or allo-BMT, and administering to the subject a therapeutically effective amount of a cell preparation of IL-33 activated ILC2 cells as a co-infusion to the allo-SCT or allo-BMT.
  • a cell preparation of IL-33 activated ILC2 cells as a co-infusion with the allo-SCT or allo-BMT, i.e. before, during or shortly after, the onset of GvHD and/or IPS can be prevented or at least substantially minimized.
  • the disclosed treatments and related methods can comprise administering the cell preparations of IL-33 activated ILC2 cells as a co-infusion with the allo-SCT or allo-BMT.
  • a co-infusion of ILC2 cells with the allo-SCT or allo-BMT can in some aspects comprise infusion of the ILC2 cells at a time sufficiently proximate to the allo-SCT or allo-BMT to prevent or substantially prevent onset of GvHD or IPS.
  • a co-infusion of ILC2 cells with the allo-SCT or allo-BMT can comprise infusion of the ILC2 cells at a time sufficiently proximate to the allo-SCT or allo-BMT to treat and/or substantially reduce any GvHD or IPS symptoms that arise as a result of the allo-SCT or allo-BMT.
  • activated ILC2 cells can be infused at the time of transplantation (allo-SCT or allo-BMT) or after transplantation at the onset of symptoms suggestive of GVHD or IPS.
  • the methods comprising the administration of activated ILC2 cells can impact and cause a shift in the cytokine micro-environment in a subject away from Th1/Th17 to Th2.
  • subjects suffering from GvHD and/or IPS can have an overabundance of Th1/Th17 and reduced Th2.
  • shifting or rebalancing these cytokine levels by increasing the production of Th2 cytokines in the subject and/or decreasing production of Th1 and/or Th17 cytokines in the subject can ameliorate and/or treat one or more symptoms of GvHD and/or IPS.
  • methods of increasing production of Th2 cytokines in a subject comprising providing a subject in need of increased Th2 cytokine production, and administering to the subject a therapeutically effective amount of a cell preparation of IL-33 activated ILC2 cells.
  • a subject receiving such a treatment method can comprise a human subject receiving allo-SCT or allo-BMT.
  • the term “about,” when referring to a value or to an amount of a composition, dose, sequence identity (e.g., when comparing two or more nucleotide or amino acid sequences), mass, weight, temperature, time, volume, concentration, percentage, etc., is meant to encompass variations of in some embodiments ⁇ 20%, in some embodiments ⁇ 10%, in some embodiments ⁇ 5%, in some embodiments ⁇ 1%, in some embodiments ⁇ 0.5%, and in some embodiments ⁇ 0.1% from the specified amount, as such variations are appropriate to perform the disclosed methods or employ the disclosed compositions.
  • the phrase “consisting of” excludes any element, step, or ingredient not specified in the claim.
  • the phrase “consists of” appears in a clause of the body of a claim, rather than immediately following the preamble, it limits only the element set forth in that clause; other elements are not excluded from the claim as a whole.
  • the phrase “A, B, C, and/or D” includes A, B, C, and D individually, but also includes any and all combinations and subcombinations of A, B, C, and D.
  • the subject treated, screened, tested, or from which a sample is taken is desirably a human subject, although it is to be understood that the principles of the disclosed subject matter indicate that the compositions and methods are effective with respect to invertebrate and to all vertebrate species, including mammals, which are intended to be included in the term “subject”.
  • a mammal is understood to include any mammalian species in which screening is desirable, particularly agricultural and domestic mammalian species.
  • the disclosed methods and treatments are particularly useful in the testing, screening and/or treatment of warm-blooded vertebrates.
  • the presently disclosed subject matter concerns mammals and birds.
  • mammals such as humans, as well as those mammals of importance due to being endangered (such as Siberian tigers), of economical importance (animals raised on farms for consumption by humans) and/or social importance (animals kept as pets or in zoos) to humans, for instance, carnivores other than humans (such as cats and dogs), swine (pigs, hogs, and wild boars), ruminants (such as cattle, oxen, sheep, giraffes, deer, goats, bison, and camels), and horses.
  • carnivores other than humans such as cats and dogs
  • swine pigs, hogs, and wild boars
  • ruminants such as cattle, oxen, sheep, giraffes, deer, goats, bison, and camels
  • domesticated fowl i.e., poultry, such as turkeys, chickens, ducks, geese, guinea fowl, and the like, as they are also of economical importance to humans.
  • livestock including, but not limited to, domesticated swine (pigs and hogs), ruminants, horses, poultry, and the like.
  • the subject to be used in accordance with the presently disclosed subject matter is a subject in need of treatment and/or diagnosis.
  • a subject can have or be believed to GvHD, IPS or related condition or phenotype, particularly those associated with allo-SCT and/or allo-BMT.
  • compositions of the presently disclosed subject matter comprise in some embodiments a composition that includes a pharmaceutically acceptable carrier. Any suitable pharmaceutical formulation can be used to prepare the adenovirus vectors for administration to a subject.
  • suitable formulations can include aqueous and non-aqueous sterile injection solutions which can contain anti-oxidants, buffers, bacteriostats, bactericidal antibiotics and solutes which render the formulation isotonic with the bodily fluids of the intended recipient; and aqueous and non-aqueous sterile suspensions which can include suspending agents and thickening agents.
  • the formulations can be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and can be stored in a frozen or freeze-dried (lyophilized) condition requiring only the addition of sterile liquid carrier, for example water for injections, immediately prior to use.
  • Some exemplary ingredients are SDS, mannitol or another sugar, and phosphate-buffered saline (PBS).
  • formulations of this presently disclosed subject matter can include other agents conventional in the art having regard to the type of formulation in question.
  • sterile pyrogen-free aqueous and non-aqueous solutions can be used.
  • compositions of the presently disclosed subject matter can be used with additional adjuvants or biological response modifiers including, but not limited to, the cytokines.
  • compositions of the presently disclosed subject matter can be by any method known to one of ordinary skill in the art, including, but not limited to intravenous administration, intrasynovial administration, transdermal administration, intramuscular administration, subcutaneous administration, topical administration, rectal administration, intravaginal administration, intratumoral administration, oral administration, buccal administration, nasal administration, parenteral administration, inhalation, and insufflation.
  • suitable methods for administration of a composition of the presently disclosed subject matter include, but are not limited to intravenous.
  • a composition can be deposited at a site in need of treatment in any other manner. The particular mode of administering a composition of the presently disclosed subject matter depends on various factors, including the distribution and abundance of cells to be treated, additional tissue- or cell-targeting features of the composition, and mechanisms for metabolism or removal of the composition from its site of administration.
  • a “therapeutically effective amount” is an amount of the composition sufficient to produce a measurable response (e.g., a reduction in GvHD symptoms).
  • a therapeutically effective amount is an amount sufficient to reduction and/or ameliorate symptoms in a subject suffering from GvHD and/or IPS.
  • a therapeutically effective amount is an amount sufficient to improve the health, well-being, prognosis and/or survivability of a subject suffering from GvHD and/or IPS.
  • a therapeutically effective amount is an amount sufficient to reverse GvHD and/or IPS in a subject suffering from GvHD and/or IPS.
  • a therapeutically effective amount is an amount sufficient to prevent GvHD and/or IPS in a subject susceptible to GvHD and/or IPS, such as a subject receiving allo-BMT or allo-SCT. In some embodiments, a therapeutically effective amount is an amount sufficient to increase Th2 cytokine production in a subject suffering from GvHD and/or IPS, and/or decrease Th1 and/or Th17 cytokine production in a subject suffering from GvHD and/or IPS. In some embodiments a therapeutically effective amount can comprise an amount that allows for the tapering and discontinuation of other immunosuppressive drugs used to treat/prevent GVHD.
  • compositions of the presently disclosed subject matter can be varied so as to administer an amount of the active compound(s) that is effective to achieve the desired therapeutic response for a particular subject.
  • the selected dosage level can depend upon the activity of the therapeutic composition, the route of administration, combination with other drugs or treatments, the severity of the condition being treated, and the condition and prior medical history of the subject being treated. However, it is within the skill of the art to start doses of the compositions at levels lower than required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved.
  • one of ordinary skill in the art can tailor the dosages to an individual patient, taking into account the particular formulation, method for administration to be used with the composition, and severity of the condition, e.g. GvHD and/or IPS. Further calculations of dose can consider patient height and weight, severity and stage of symptoms, and the presence of additional deleterious physical conditions. Such adjustments or variations, as well as evaluation of when and how to make such adjustments or variations, are well known to those of ordinary skill in the art of medicine.
  • C57BL/6J (H2b) (termed WT), BALB/c, and C57BL/6J ⁇ DBA/2 F1 (termed B6D2) were purchased from The Jackson Laboratory, Bar Harbor, Me., United States of America.
  • the generation of enhanced GFP-expressing C57BL/6 mice has been described previously (Panoskaltsis-Mortari et al., Blood 2004; 103(9):3590-8).
  • Donor and recipient mice were age-matched males between 8 and 16 weeks. All experiments were performed in accordance with protocols approved by the University of North Carolina Institutional Animal and Care Use Committee, Chapel Hill, N.C., United States of America.
  • mice 10 to 12 week old C57BL/6 mice are given 0.4 ⁇ g of recombinant mouse IL-17E/IL-25 (R&D systems) for 4 days. On day 5, the cells are isolated from the mesenteric lymph node (MLN) and by peritoneal lavage using 10% serum complete RPMI.
  • MNN mesenteric lymph node
  • ILC2 cells are isolated by negative selection with MAC column using biotinylated antibodies [(Anti-CD8 ⁇ (53-6.7), anti-CD4 (RM 4.4), anti-CD3 ⁇ (145-2C11), anti- ⁇ TCR (UC7-13DS), anti-TER119 (TER-119), anti-B220 (RA3-6B2), anti-CD11c (N418), anti-CD11b (M1/70), anti-NK1.1 (PK136), anti-CD19 (MB19-1), anti-Ly6G (1A8) and anti-CD49b (DX5)] and Streptavidin Microbeads (Miltenyi 130-048-101, Miltenyi Biotec, Cambridge, Mass., United States of America).
  • Cells are cultured for six days in 10% RPMI-c with rIL-7 and rIL-33 (10 ng/ml) changing media every 2 days. On the sixth day the cell purity is determined by flow cytometry and purified if needed as described above. ILC2 activation was evaluated on day 6 by IC staining of IL-5 and IL-13 by flow cytometry.
  • T cells were isolated using Cedarlane T cell recovery column kit (Cedarlane Laboratories, Burlington, N.C., United States of America), followed by Ab depletion using PE-conjugated anti-mouse B220 and anti-mouse CD25 Abs (eBioscience, San Diego, Calif., United States of America) and magnetic bead selection using anti-PE beads (Miltenyi Biotec, Cambridge, Mass., United States of America).
  • T cell depleted bone marrow was prepared as described by Coghill et al., Blood. 2010; 115(23):4914-22. The day prior to transplantation, recipient mice received either 950 cGy (B6D2) or 800cGy (BALB/c) of total body irradiation.
  • mice were intravenously injected with either 4 ⁇ 106 T cells and 3 ⁇ 106 TCD BM cells, or 5 ⁇ 105 total T cells and 5 ⁇ 106 TCD BM cells, respectively.
  • B6 or BALB/c mice also received 4 ⁇ 106 or 1 ⁇ 106 CD90+ ILC2 respectively. Mice were observed twice weekly for clinical GVHD signs and symptoms based on a previously established clinical scoring system (van Den Brink et al., Journal of Immunology. 2000; 164(1):469-80).
  • ILC2 cells were given either at the time of bone marrow transplantation (supplemented with T cells) or 7 days after bone marrow transplantation.
  • mice were given P815 or mBC-CML tumor cells at the time of bone marrow transplantation.
  • B6D2 mice received 950 cGy of total body irradiation. 24 hours later cells were isolated and evaluated by flow cytometry. Total cell numbers were established from the absolute event count of live cells gated as Lineage negative, CD90+, ICOS+ or ROR ⁇ t+ for ILC2 or ILC3 respectively or as CD4+ for T cells.
  • Lamina propria lymphocytes were isolated using the Miltenyi Lamina basement dissociation kit (130-097-410) as per the manufacturer's instructions (Miltenyi Biotec, Cambridge, Mass., United States of America). Livers and lungs were digested in a solution of 1 mg/ml collagenase A (Roche) and 75 U of DNase I (Sigma-Aldrich, St. Louis, Mo., United States of America) in RPMI 1640/5% newborn calf serum. Digested tissues were treated with ACK lysis buffer to remove RBCs and were passed through 100- ⁇ m pore size cell strainers.
  • Leukocytes were collected at the interface of a 40%/80% Percoll (Sigma-Aldrich) in RPMI 1640/5% newborn calf serum. The pelleted cells were washed in PBS/2% FBS. Spleens were teased apart, treated with ACK lysis buffer, and washed in PBS/2% FBS.
  • Type 2 Innate Lymphoid Cells are Radiation Sensitive.
  • ILC2 cells in the MLN were extremely sensitive to the effects of irradiation. In fact, these cells were as radiation sensitive as conventional CD4+ T cells ( FIGS. 1C and 1D ).
  • the absence of recipient ILC2 cells in the colon and MLN may critically lead to alterations in the microenvironment leading to Th1 polarization in the lower GI tract induced after allo-SCT.
  • Th2 cytokines and the data indicating that shifting the cytokine micro-environment away form Th1/Th17 to Th2 reduces the severity of aGVHD, it was hypothesized that co-transplantation of ILC2 cells would reduce aGvHD and increase survival after allo-SCT.
  • ILC2 cells in normal lymphoid tissue.
  • a protocol was established for the isolation, expansion, and identification of ILC2 cells isolated from the mouse MLN and pertoneal cavity.
  • ILC2 cells can be identified as CD90+, lin ⁇ , ICOS+ with additional expression of CD127, ST2, Sca-1, and CD25.
  • Injection of IL-25 will elicit an increase in ILC2 cells in the MLN and peritoneal cavity and this method was adapted to expand ILC2 in vivo. After 4 days of IL-25 treatment intraperitoneal injection (i.p.) a significant increase in the number of ILC2 cells compared to control mice was found.
  • ILC2 cells were expanded in vitro with IL-33 and IL-7 treatment.
  • IL-33 and IL-7 greater than 10 fold expansion of ILC2 cells in vitro after 6 days compared to IL-25 treated cultures ( FIG. 2 ) was observed.
  • ILC2 specific surface markers including CD127, ST2 and CD44, it was determined that the cells were bona fide ILC2. It was also determined that ILC2 cells lose expression of CD90 in culture.
  • IL-33 and IL-7 treatment also activates the ILC2 cells increasing the percentage of cultured cells that produced IL-4, IL-5 and IL-13 with no evidence of expansion of other ILC groups; IFN- ⁇ in ILC1 cells; or IL-22 in ILC3 cells.
  • IL-25 and IL-33 activate ILC2 inducing production of IL-4, IL-5 and IL-13.
  • IL-25 (0.4 ng/injection for 4 days) elicits increased ILC2 in the MLN of B6 mice.
  • IL-33 with IL-7 treatment increases ILC2 proliferation in vitro compared to IL-25 with IL-7 (10 ng/ml each), adjusted average cell number ⁇ SEM. See FIG. 2 .
  • Lineage negative, IL-33 activated cells express ILC2 specific surface markers CD127, ST2 and CD44.
  • IL-33 treatment activates ILC2 cells increasing cytokine production. Cells were isolated from MLN and by peritoneal lavage on day 5 of IL-25 injection and lineage depleted by bead separation (Miltenyi Biotec, Cambridge, Mass., United States of America).
  • the cells were analyzed for the expression of ST2 and CD127, which are not expressed by MPP2 cells. IT was shown that over 92% of the cells that were generated expressed ST2 and CD127, consistent with the phenotype of ILC2 cells.
  • ILC2 cells Considering activated ILC2 cells expressed high levels of Th2 inducing cytokines, survival studies after allo-SCT co-infusing ILC2 cells were performed. For these experiments, splenic T cells were transplanted with (treatment) or without (ctrl) ILC2 cells. Interestingly, co-transplantation of one dose of ILC2 cells significantly improved survival in the B6 to B6D2 model of aGvHD with 70% of recipients surviving over 60 days ( FIG. 3A ). Recipients were monitored and scored for GVHD symptoms as previously described (van Den Brink et al., Journal of Immunology. 2000; 164(1):469-80, Fulton et al., Journal of immunology. 2012; 189(4):1765-72).
  • mice receiving ILC2 cells there was a reduction in the clinical GvHD score in mice receiving ILC2 cells, beginning at day 10.
  • a significant prolongation of survival was found in B6 into BALB/c transplants, a much more stringent MHC completely mis-matched model.
  • ILC2 Cells Reduce GI Tract T Cell Burden and Limit Th1 and Th17 Induction
  • Donor T cell migration were evaluated using B6-GFP donor T cells and fluorescence stereomicroscopy and GFP ELISA. It was determined that co-infusion of ILC2 cells reduced donor T cells in Peyer's Patches (PP) in the colon and small bowel in a dose dependent manor as determined by GFP intensity. Using Zeiss SteREO microscopy with eGFP bandpass filter, GFP images were taken for each organ and GFP intensities were determined by software analysis. In addition, there is significantly less GFP in tissue homogenates. T cell infiltration into GvHD target organs was analyzed by flow cytometry and it was determined that those recipients of ILC2 cells had reduced lymphocytes in the LP of the colon ( FIG. 4A ).
  • LC2 migration to GvHD target organs was evaluated by infusing B6-GFP ILC2 cells.
  • ILC2 cells were tracked using B6-GFP ILC2 donor mice for injection of IL-25 and in vitro expansion with IL-33 prior to co-infusion at BMT.
  • IVIS imaging of GFP expressing ILC2 cells in Peyer's patches 12 days after BMT in B6D2 mice was employed. While only limited GFP signal can be can be found in the lamina intestinal, large clusters of GFP-ILC2 cells were observed in the Peyer's patches on the intestine. Additionally, it was found that ILC2 cells migrate not only to the colon and small bowel but other GvHD target organs by measuring GFP in tissue by ELISA ( FIG.
  • IL-33 activated cells that have been co-infused are innate lymphoid cells as determined by their expression of the lymphoid marker CD90 in the absence of other lineage defining markers including CD3, CD19, CD11 b, CD11 c, B220, Gr-1, NK1.1 or CD49b. It was also confirmed that these innate lymphoid cells are ILC2 by evaluating cytokine expression by intracellular staining IL-5 and IL-13 in ILC2 12 days after transplant. Greater than 90% of donor GFP positive IL-33 activated cells continue to express IL-13 in all target organs ( FIG. 5B ).
  • IL-5 expression varies among GvHD target organs with more than 50% of ILC2 cells in the GI tract maintaining IL-5 expression.
  • Th2 inducing cytokines we would expect an increase in Th2 expansion.
  • IL-4 producing T cells there is not an increase in IL-4 producing T cells in mice that receive ILC2 cells.
  • ILC2 cells were expanded from cord blood cells using the AhR antagonist SP1. Frozen umbilical cord blood cells were taken and stained with antibodies specific for CD34, c-Kit, CD90 and IL-7R ⁇ . The presence of endogenous ILC2 cells in cord blood was analyzed and very few cells were found present. Thus, c-Kit+ cells were isolated from umbilical cord blood and put in the presence of the cytokines IL-33, IL-25 and IL-2 for cell expansion. There was robust expansion of ILC2 cells in the presence of the cytokines IL-2 and IL-33 and the presence of all three cytokines. This was a 10-fold increase over the 20 days in culture.
  • ILC2 cells can migrate into the pulmonary parenchyma ( FIG. 6 ).
  • 1 ⁇ 10 6 ILC2 cells from eGFP+ mice were infused i.v. and 48 hours later evaluated from the lung parenchyma in B6D2 mice that had been lethally irradiated and received 3 ⁇ 10 6 B6 TCD bone marrow cells and 3.5 ⁇ 10 6 B6 T cells.
  • ILC2 cells were distinguished by the expression of GFP and in addition expression of ST2, CD127 and CD90 and absence of lineage markers. There was a strong statistically significant increase in ILC2 cells in the lung after i.v. infusion that was not found in the sham treated animals (p ⁇ 0.001).
  • Pulmonary GvHD and idiopathic pneumonia syndrome are mediated predominantly by Th17 donor T cells. Yi et al., Blood. 2009; 114(14):3101-3112; Carlson et al., Blood. 2009; 113(6):1365-1374.
  • ILC2 cells can be used to treat ongoing IPS by blocking the generation of Th17 cells.
  • B6D2 Recipient mice were lethally irradiated and administered donor T cells from B6 wild type or mice lacking the STAT6 gene which is critical for the generation of Th2 T cells.
  • one group of mice receiving STAT6 null T cells received ILC2 cells at a 1:1 ratio with T cells (T cell dose was 4 ⁇ 10 6 cells).
  • the other group received PBS as a control.
  • Overall survival and clinical score of GVHD were evaluated.
  • N 8 mice/group. ILC2 cells are equally effective in preventing GVHD even when using T cells that cannot become Th2 cells.
  • FIG. 7A is a bar graph indicating that there is no difference in the generation of T cells making IL-4 (Th2) after the infusion of ILC2 cells when comparing mice that received ILC2 cells (grey bar) and those receiving sham treatment (open bar) 12 days after bone marrow transplantation.
  • FIG. 7B illustrates the outcome of recipient mice receiving lethal irradiation (BM only) and infused with haploidentical wild type T cells with ILC2 cells (2:1) ratio (Control) or haploidentical STAT6 knockout T cells with ILC2 cells (2:1) ratio (Treated). STAT6 null T cells cannot generate Th2 cells. This figure demonstrates that the function of ILC2 cells to block GVHD is independent of the generation of Th2 cells.
  • FIG. 7A is a bar graph indicating that there is no difference in the generation of T cells making IL-4 (Th2) after the infusion of ILC2 cells when comparing mice that received ILC2 cells (grey bar) and those receiving sham treatment (open bar
  • FIG. 7C is a plot showing the clinical scoring of recipient mice receiving lethal irradiation (triangle—BM only) and infused with either haploidentical WT T cells (closed circle—Control) or STAT6 knockout T cells plus ILC2 cells (open circle—Treated). Clinical scores are significantly improved in recipient mice given ILC2 cells with STAT6 knockout T cells compared to those given only WT T cells.
  • FIG. 7D is a bar graph showing an evaluation for FoxP3 expressing T cells in the GI tract 12 days after the administration of haploidentical WT T cells (open bar) or WT T cells supplemented 2:1 with ILC2 cells (closed bar). This demonstrates that there is no quantitative increase in FoxP3-expressing regulatory T cells in the GI tract after ILC2 administration.
  • ILC2 Cells Function by Enhancing GI Tract Myeloid Derived Suppressor Cells
  • B6D2 recipient mice were lethally irradiated and administered 4 ⁇ 10 6 B6 T cells with/without a 1:2 ratio (ILC2 cells:T cells) of donor ILC2 cells. Following transplantation, mice were administered anti-GR-1 or anti-Ly6C mAb 2 ⁇ per week for three weeks. As shown in FIGS. 8A to 8C , the activity of ILC2 cells to block GVHD was completely lost by depleting myeloid cells with these antibodies. In FIG. 8A tolerogeneic dendritic cells from the donor have been enumerated using CD11c and CD103 as markers. Mice receiving ILC2 cells have an increase in the number of tolerogenic DCs in the GI tract after transplantation. FIG.
  • FIG. 8B again demonstrates that there is no increase in FoxP3-expressing T cells in the GI tract after bone marrow transplantation comparing mice receiving ILC2 cells (grey bar) with control treatment (open bar).
  • FIG. 8C haplioidentical mice received lethal irradiation (BM only) and transplantation using wild type bone marrow cells supplemented with T cells with (Treatment) and without (Control) ILC2 cells. 2-3 ⁇ per week after transplantation receipient mice received anti-Gr-1 monoclonal antibody by intraperitoneal injection (i.p.). Depletion of Gr-1-expressing cells completely eliminated the benefit of infusion of ILC2 cells to prevent GVHD. This indicates that ILC2 cell infusion is dependent on the presence of donor myeloid cells for its function.
  • ILC2 cell infusion diminishes GI tract permeability and infectious complications that are associated with lower GI tract GVHD.
  • FIG. 9A to 9E the permeability of the GI tract we from day 4 to 20 was evaluated by administering FITC-dextran to mice after bone marrow transplantation which received either T cells with sham ILC2 infusion versus T cells with ILC2 infusion.
  • FIGS. 9A to 9C are as follows: Normal (lined bar); BM only (checkered bar); Control (open bar), Treated (grey bar).
  • Treatment gropus in FIGS. 9D and 9E are as follows: BM only (triangle); Control (closed circle); Treatment (closed circle).

Abstract

ILC2 cells play a role in the pathogenesis of graft versus host disease (GvHD) and idiopathic pneumonia syndrome (IPS), both conditions associated with allogeneic stem cell transplantation. Infusion of IL-33 activated ILC2 cells into patients with ongoing GvHD or IPS, or prior to onset of GvHD or IPS in susceptible patients, substantially ameliorates the disease and improves survival.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims benefit of U.S. Provisional Patent Application Ser. No. 62/087,340, filed Dec. 4, 2014, herein incorporated by reference in its entirety.
  • GRANT STATEMENT
  • This invention was made with government support under Grant Nos. CA166794 and HL115761 awarded by the National Institutes of Health. The government has certain rights in the invention.
  • TECHNICAL FIELD
  • The presently disclosed subject matter relates to compositions and methods for preventing and treating graft versus host disease (GvHD) and/or idiopathic pneumonia syndrome (IPS). The presently disclosed subject matter further relates to methods of making compositions for preventing and treating GvHD and/or IPS.
  • BACKGROUND
  • Graft versus host disease remains a major obstacle to the widespread utilization of allogeneic stem cell transplant (allo-SCT) and allogeneic bone marrow transplant (allo-BMT) for the treatment of high-risk or relapsed leukemia, lymphoid malignancies, myelodysplastic syndromes, myeloproliferative neoplasms, and bone marrow failure syndromes. While there has been progress in approaches to prevent acute GvHD (aGvHD), there has been little advancement in clinical approaches for the treatment of patients with aGvHD, which rely predominantly on systemic corticosteroids.
  • Unfortunately, corticosteroid therapy is associated with a substantial number of significant side-effects including infectious complications, bone disturbances and profound metabolic changes. The outcome for patients with steroid-refractory aGvHD is dismal with approximately 10% of these patients surviving one year after the diagnosis and little progress in improving this outcome over the past 20 years.
  • Similarly, idiopathic pneumonia syndrome (IPS) is a common pulmonary complication and remains a severe threat to survival after allo-SCT and allo-BMT. IPS often coincides with the onset of GvHD.
  • What are needed are therapies, methods of treatment and compositions for use in treating and/or preventing GvHD and IPS. Methods of making such therapeutics and effectively administering the same are also needed. Such advantages, and others disclosed herein, are provided by the instant disclosure.
  • SUMMARY
  • This summary lists several embodiments of the presently disclosed subject matter, and in many cases lists variations and permutations of these embodiments. This summary is merely exemplary of the numerous and varied embodiments. Mention of one or more representative features of a given embodiment is likewise exemplary. Such an embodiment can typically exist with or without the feature(s) mentioned; likewise, those features can be applied to other embodiments of the presently disclosed subject matter, whether listed in this summary or not. To avoid excessive repetition, this Summary does not list or suggest all possible combinations of such features.
  • Provided herein are pharmaceutical compositions for treating graft versus host disease (GvHD) or idiopathic pneumonia syndrome (IPS). In some embodiments the compositions comprise a therapeutically effective amount of a cell preparation of IL-33 activated type 2 innate immune cells (ILC2), and a pharmaceutically acceptable carrier. In some embodiments the ILC2 cells have the following properties: test positive for expression of lymphoid marker CD90, test positive for expression of lymphoid marker ICOS, and test negative for expression of lympohoid marker lin. In some embodiments the ILC2 cells test positive for expression of CD127, ST2, Sca-1, CD25, CD90, and ICOS, and test negative for expression of lin. In some embodiments the ILC2 cells are activated by cytokines IL-33, IL-25, IL-7 and/or combinations thereof. In some embodiments the ILC2 cells are derived from human cord blood cells. In some embodiments the pharmaceutical composition is substantially free of MPP2 cells.
  • Provided herein are in vitro methods for generating activated ILC2 cells, comprising in some embodiments collecting human cord blood from a subject, isolating c-Kit positive cells from the cord blood, and culturing the c-Kit positive cells in the presence of an IL-33 cytokine, whereby IL-33 activated ILC2 cells are generated. In some embodiments the ILC2 cells are activated by exposing the c-Kit positive cells in culture to cytokines IL-33, IL-25, IL-7 and/or combinations thereof. In some embodiments the in vitro methods further comprise analyzing the generated ILC2 cells by measuring expression of ST2 and CD127 in the ILC2 cells, whereby expression of ST2 and CD127 is indicative of the phenotype of ILC2 cells. In some embodiments the ILC2 cells are substantially free of MPP2 cells. In some embodiments the subject is a human patient or donor.
  • Provided herein are methods for treating graft versus host disease (GvHD) or idiopathic pneumonia syndrome (IPS) in a subject, comprising in some embodiments providing a subject in need of treatment for GvHD and/or IPS, and administering to the subject a therapeutically effective amount of a cell preparation of IL-33 activated ILC2 cells. In some embodiments the ILC2 cells have the following properties: test positive for expression of lymphoid marker CD90, test positive for expression of lymphoid marker ICOS, and test negative for expression of lympohoid marker lin. In some embodiments the ILC2 cells test positive for expression of CD127, ST2, Sca-1, CD25, CD90, and ICOS, and test negative for expression of lin. In some embodiments the subject is a human subject receiving an allogeneic stem cell transplant (allo-SCT) or allogeneic bone marrow transplant (allo-BMT). In some embodiments the cell preparation of IL-33 activated ILC2 cells is administerd to the subject within 1 to 30 days after receiving the allo-SCT or allo-BMT. In some embodiments the risk and/or severity of acute GVHD associated with allo-SCT or allo-BMT is reduced. In some embodiments the cell preparation of IL-33 activated ILC2 cells is administered as a co-infusion with the allo-SCT or allo-BMT. In some embodiments production of Th2 cytokines in the subject is increased, and/or production of Th1 and/or Th17 cytokines in the subject is decreased.
  • Provided herein are methods for preventing GvHD or IPS in a subject, comprising in some embodiments providing a subject receiving allo-SCT or allo-BMT, and administering to the subject a therapeutically effective amount of a cell preparation of IL-33 activated ILC2 cells. In some embodiments the cell preparation of IL-33 activated ILC2 cells is administered as a co-infusion with the allo-SCT or allo-BMT. In some embodiments the co-infusion of ILC2 cells with the allo-SCT or allo-BMT comprises infusion of the ILC2 cells at a time sufficiently proximate to the allo-SCT or allo-BMT to prevent or substantially prevent onset of GvHD or IPS. In some embodiments the ILC2 cells have the following properties: test positive for expression of lymphoid marker CD90; test positive for expression of lymphoid marker ICOS; and test negative for expression of lympohoid marker lin. In some embodiments the ILC2 cells test positive for expression of CD127, ST2, Sca-1, CD25, CD90, and ICOS, and test negative for expression of lin. In some embodiments production of Th2 cytokines in the subject is increased, and/or production of Th1 and/or Th17 cytokines in the subject is decreased.
  • Provided herein are methods of increasing production of Th2 cytokines in a subject, comprising: providing a subject in need of increased Th2 cytokine production, and administering to the subject a therapeutically effective amount of a cell preparation of IL-33 activated ILC2 cells. In some embodiments the subject comprises a human subject receiving allo-SCT or allo-BMT. In some embodiments production of Th1 and/or Th17 cytokines is decreased in the subject. In some embodiments the ILC2 cells have the following properties: test positive for expression of lymphoid marker CD90, test positive for expression of lymphoid marker ICOS, and test negative for expression of lympohoid marker lin. In some embodiments the ILC2 cells test positive for expression of CD127, ST2, Sca-1, CD25, CD90, and ICOS, and test negative for expression of lin.
  • Accordingly, it is an object of the presently disclosed subject matter to provide compositions and methods for preventing and treating GvHD and/or IPS. This and other objects are achieved in whole or in part by the presently disclosed subject matter. Further, an object of the presently disclosed subject matter having been stated above, other objects and advantages of the presently disclosed subject matter will become apparent to those skilled in the art after a study of the following description, Drawings and Examples.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The presently disclosed subject matter can be better understood by referring to the following figures. The components in the figures are not necessarily to scale, emphasis instead being placed upon illustrating the principles of the presently disclosed subject matter (often schematically). In the figures, like reference numerals designate corresponding parts throughout the different views. A further understanding of the presently disclosed subject matter can be obtained by reference to an embodiment set forth in the illustrations of the accompanying drawings. Although the illustrated embodiment is merely exemplary of systems for carrying out the presently disclosed subject matter, both the organization and method of operation of the presently disclosed subject matter, in general, together with further objectives and advantages thereof, may be more easily understood by reference to the drawings and the following description. The drawings are not intended to limit the scope of this presently disclosed subject matter, which is set forth with particularity in the claims as appended or as subsequently amended, but merely to clarify and exemplify the presently disclosed subject matter.
  • For a more complete understanding of the presently disclosed subject matter, reference is now made to the following drawings in which:
  • FIGS. 1A-1E are a series of bar graphs showing that ILC2 cells are radiation sensitive. Comparison of unconditioned cells (white bars) or 24 hours after receiving radiation (950cGy) (grey bars) as analyzed by flow cytometry. (FIG. 1A) ILC3 from the LP are radiation resistant. ILC3 from MLN (FIG. 1B) and LP CD4 T cells (FIG. 1C) are sensitive to radiation. ILC2 cells from the LP (FIG. 1D) and MLN (FIG. 1E) are sensitive to radiation. This represents two independent experiments, n=4 for each. T test using GraphPadTM software (GraphPad Software, Inc., La Jolla, Calif., United States of America), ** p<0.05.
  • FIG. 2 is a line graph showing that IL-33 with IL-7 (open circles) treatment increases ILC2 proliferation in vitro compared to IL-25 with IL-7 (solid squares) (10 ng/ml ea.), adjusted average cell number±SEM.
  • FIGS. 3A-3D are a series of plots showing that co-transplant of ILC2 cells reduces aGvHD. Lethally irradiated B6D2 mice (950cGy) received 3.0×106 TCD BM (BM only), BM plus 4.0×106 total splenic T cells (Ctrl) or BM plus T cells with 4.0×106 IL-33 activated ILC2 cells (Treatment) in an ILC2 (Top row) or recipients received 4.0×106 IL-33 activated ILC2 cells at the time of allo-BMT or 7 days later (Bottom row) (FIG. 3A) Percent survival after allo-SCT. (FIG. 3B) GvHD scores over time. (FIG. 3C) Percent survival in timed injection study. (FIG. 3D) GvHD scores of timed injection study. These represents 4 independent experiments, n=42. ** p=0.01, by Log-rank (Mantel-cox) test using GraphPad Prism TM 5 software (GraphPad Software, Inc., La Jolla, Calif., United States of America).
  • FIGS. 4A-4E are a series of bar graphs showing that co-transplant of ILC2 cells reduces donor T cells. Lethally irradiated B6D2 mice (950cGy) received 3.0×106 TCD BM (BM only), BM plus 4.0×106 total splenic GFP+ T cells (Ctrl), BM plus GFP+ T cells with 4.0×106 ILC2 cells (Treat 1:1) or BM plus GFP+ T cells with 8.0×106 ILC2 cells (Treat 1:2). Donor T cells were evaluated in the GI tract 12 days after BMT. (FIG. 4A) ELISA quantification of GFP (Cell Biolabs: AKR-121; Cell Biolabs, Inc., San Diego, Calif., United States of America) in tissue homogenates on day 12 post BMT from Ctrl (open bars) and Treat 1:1 (grey bars) normalized to grams of tissue. Donor T cells from BMT (as described for FIG. 3A) were evaluated by flow cytometry 12 days after BMT, Ctrl (open bars) and Treatment (grey bars). (FIG. 4B) Percentage of donor T cells in the LP lymphocyte compartment (LPL). (FIG. 4C) The total number of CD4+ and CD8+ donor T cells in the LP. (FIG. 4D) Total number of IFN-γ producing donor Th1 and Tc1 and the number of donor IL-17A producing Th17 cells. (FIG. 4E) The percentage of donor Treg as determined by intranuclear staining of FoxP3. These represents 2 independent experiments, n=4 each. Student T test using GraphPad PrismTM software (GraphPad Software, Inc., La Jolla, Calif., United States of America), * p<0.05.
  • FIGS. 5A and 5B are a set of bar graphs showing that IL-33 activated ILC2 migrate to GvHD target organs and secrete Th2 cytokines. ILC2 cells were tracked using B6-GFP ILC2 donor mice for injection of IL-25 and in vitro expansion with IL-33 prior to co-infusion at BMT. (FIG. 5A) ELISA quantification of ILC2 derived GFP in GvHD target organs. (FIG. 5B) Bar graph of average IL-13 and IL5 IC cytokine±SEM in donor ILC2 cells day 12 post BMT.
  • FIG. 6 is a bar graph showing enumeration of ILC2 cells after infusion from lung parenchyma. 1×106 ILC2 cells from eGFP+ mice were infused i.v. and 48 hours later evaluated from the lung parenchyma in B6D2 mice that had been lethally irradiated and received 3×106 B6 TCD bone marrow cells and 3.5×106 B6 T cells. ILC2 cells were distinguished by the expression of GFP and in addition expression of ST2, CD127 and CD90 and absence of lineage markers. There was a strong statistically significant increase in ILC2 cells in the lung after i.v. infusion (hatched bar) that was not found in the sham treated animals (open bar) (p <0.001).
  • FIG. 7A is a bar graph indicating that there is no difference in the generation of T cells making IL-4 (Th2) after the infusion of ILC2 cells when comparing mice that received ILC2 cells (grey bar) and those receiving sham treatment (open bar) 12 days after bone marrow transplantation. FIG. 7B illustrates the outcome of recipient mice receiving lethal irradiation (BM only) and infused with haploidentical wild type T cells with ILC2 cells (2:1) ratio (Control) or haploidentical STAT6 knockout T cells with ILC2 cells (2:1) ratio (Treated). FIG. 7C is a plot showing the clinical scoring of recipient mice receiving lethal irradiation (triangle—BM only) and infused with either haploidentical WT T cells (closed circle—Control) or STAT6 knockout T cells plus ILC2 cells (open circle—Treated). FIG. 7D is a bar graph showing an evaluation for FoxP3 expressing T cells in the GI tract 12 days after the administration of haploidentical WT T cells (open bar) or WT T cells supplemented 2:1 with ILC2 cells (closed bar).
  • FIGS. 8A to 8C are a series of graphs and plots showing the results of an evaluating for the presence of donor antigen presenting cells in the GI tract in recipient mice after bone marrow transplant receiving ILC2 cells plus haploidentical T cells versus those receiving only haploidentical T cells. FIG. 8A is a plot showing the results of tolerogeneic dendritic cells from donor mice being enumerated using CD11c and CD103 as markers. FIG. 8B is a bar graph demonstrating that there is no increase in FoxP3-expressing T cells in the GI tract after bone marrow transplantation comparing mice receiving ILC2 cells (grey bar) with control treatment (open bar). FIG. 8C is a plot showing the results of haplioidentical mice receiving lethal irradiation (BM only) and transplantation using wild type bone marrow cells supplemented with T cells with (Treatment) and without (Control) ILC2 cells.
  • FIGS. 9A to 9E are a series of plots and graphs showing the results of the permeability of the GI tract from day 4 to 20 by administering FITC-dextran to mice after bone marrow transplantation which received either T cells with sham ILC2 infusion versus T cells with ILC2 infusion. Treatment groups in FIGS. 9A to 9C are as follows: Normal (lined bar); BM only (checkered bar); Control (open bar), Treated (grey bar). Treatment gropus in FIGS. 9D and 9E are as follows: BM only (triangle); Control (closed circle); Treatment (closed circle).
  • DETAILED DESCRIPTION
  • The presently disclosed subject matter now will be described more fully hereinafter, in which some, but not all embodiments of the presently disclosed subject matter are described. Indeed, the disclosed subject matter can be embodied in many different forms and should not be construed as limited to the embodiments set forth herein; rather, these embodiments are provided so that this disclosure will satisfy applicable legal requirements.
  • GvHD is mediated by donor T cells that recognize major and minor histocompatibility differences in the host. Evaluations using pre-clinical models and clinical studies have indicated that Th1 T cells and perhaps Th17 cells may be mediators of aGvHD. Moreover, in accordance with the presently disclosed subject matter, Th1 cells have been found to be involved in the pathogenesis of aGvHD involving the small bowel and colon.
  • The reason for the Th1 bias for GvHD involving the GI tract is not clear, although previous investigations have implicated the release of LPS and cytokine activation by host innate immune cells as a result of bacterial translocation after conditioning therapy. However, targeting this axis has not been substantially beneficial in the treatment of patients with steroid-refractory aGvHD.
  • New populations of innate cells that express similar cytokines and transcription factors compared to polarized T cells but lack germ-line encoded recombined receptors have been identified. An innate immune population of cells that generates Th2 cytokines is termed nuocytes or type 2 innate immune cells (ILC2). These cells generate substantial quantities of IL-5, and IL-13 with more modest expression of IL-4. These cells are involved in Th2 cell generation of IL-13 and the host response to specific parasitic infections. Human ILC2 cells can be found in the GI tract and lung and can be defined by their expression of CD161 and the chemoattractant, CRTH2 (Mjosberg et al., Nat Immunol. 2011; 12(11):1055-1062). Like murine ILCs, human ILC2 cells can be expanded in vitro and produce significant quantities of IL-5 and IL-13 in response to IL-7 and IL-33 (Yu et al., Mucosal Immunol. 2013).
  • T lymphocytes are characterized by the expression of the T cell receptor (TCR), which requires recombination of variable, diversity, joining and constant gene segments to produce a TCR capable of binding to peptide/MHC proteins. The generation of a T cell repertoire, with limited activity against self, occurs in the thymus. These naïve T cells recognize antigens presented by APCs, which leads to T cell activation, proliferation and differentiation leading to the generation of the adaptive immune response. As this process takes 7-10 days, mechanisms are needed to control pathogen growth during this time frame. Thus, the immune system has innate immune cells that can recognize pathogen-associated molecular patterns and/or damage-associated molecular patterns and initiate an immune response. This aspect of the immune response, which includes neutrophils, macrophages, dendritic cells and monocytes, is a critical part of the initial immune response.
  • New populations of innate immune cells have been discovered. Previously, NK cells that generate IFN-γ were discovered (Kiessling et al., 1976). Following this, other populations of innate cells were found. These cells were often located at mucosal sites such as the GI tract and lung, where they served a critical role as “first responders” to infectious pathogens. Three new groups of innate immune cells, ILC1 (generate IFN-γ similar to NK cells), ILC2 and ILC3 Is have been described. The first population of innate cells are lymphoid tissue inducer cells (LTi). These cells express the transcription factor RORyt and are involved in the generation of lymphoid tissue. Previous work has shown that fetal CD4+/CD3−/CD45+/IL-7Rα+ cells were important for the development of lymph nodes (LN) and Peyer's patches (PP). These cells, which are generated around day 12.5 during development, require the transcription factors Id2 and RORγt for their development. They express surface lymphotoxin (LT) α1β2, and were termed LTi cells. The absence of these cells or of the transcription factors necessary for their generation (Id2, RORγt), or proteins that mediate the interaction of LTα1β2 with LTβR on stromal cells leads to absence of LNs and PP.
  • It has also been determined that these LTi cells generated IL-17A and could be identified in mice after birth. A second group of cells that resemble LTi cells but are phenotypically distinct has been identified in mice after birth. These cells generate IL-22 but not IL-17A and are found at mucosal sites mostly in the small bowel and colon. These ILCs express NK cell-associated cytotoxicity receptors NKp44 in humans and NKp46 in mice. They are distinct from a population of ILCs that generates IL-17A and lacks the expression of NK receptors. These IL-22 generating ILCs critically require the expression of the transcription factor Ahr56.
  • Innate immune cells that generate IL-17 and/or IL-22 have been termed ILC3 cells. Following the description of these cells, a second entirely different population of innate lymphoid cells was characterized. Previous work had suggested that the cytokine, IL-25, affected a non-T, B or NK cell.
  • The instant disclosure pertains, at least in part, to ILC2 cells. Such cells have in some embodiments been found to express CD127, CD117, CD90 and CD25, respond to IL-25 and IL-33, and generate IL-5 and IL-13. Notably, a second but distinct population of cells that is IL-25-responsive is termed multipotent progenitor 2 (MPP2) cells and can generate multiple different myeloid cells. However, they appear to be different from true ILC2 cells as ILC2 cells do not give rise to myeloid progenitors.
  • As disclosed herein, cultures of ILC2 cells in the presence of IL-33 significantly increases their expression of IL-4, IL-5 and IL-13 and enhanced cell proliferation. Although ILC2 and MPP2 cells share common characteristics such as cytokine profile, lack of lineage markers and response to IL-25 and IL-33, their discordant expression of CD25, CD127 and CD90 and distinct multipotent potential suggest they are distinct populations. Type 2 ILC share similarities to Th2 and Treg cells expressing high levels of Th2 cytokines as well as regulatory receptors that are constitutively expressed on Tregs.
  • MPP2 cells can give rise to monocytes, macrophages, eosinophils and mast cells. These progeny are competent antigen presenting cells and the adoptive transfer of MPP2 cells promoted Th2 cytokine responses. IL-25 and IL-33 stimulation of ILC2 cells promoted M2 macrophage recruitment to visceral associated adipose tissue.
  • The generation of ILC2 cells requires the expression of the transcription factor Id268. Like Th2 cells, ILC2 cells express GATA-3, MAF and STATE. ILC2 cells appear to be important for the generation of T cells making IL-13. ILC2 cells are important to the immune response to helminths such as Nippostrongyloides brasiliensis, which require IL-25 for an adequate immune response. These data support a framework by which ILC2 cells are relevant in providing a suitable environment for the generation of Th2 T cells. These Th2 cells along with MPP2 cells can assist in the recruitment and activation of mast cells, basophils, eosinophils and B cells allowing for clearance of parasitic infections. However, overexuberant ILC2/Th2 responses may be detrimental as clinical syndromes such as asthma may be increased after ILC2/Th2 activation.
  • Thus, disclosed herein are therapeutic treatments and composition based on the role ILC2 cells play in the pathogenesis of GvHD and IPS, both of which are associated with allo-SCT and/or allo-BMT. Particularly, disclosed herein for the first time are therapeutic approaches and compositions based on the discovery that infusion of IL-33 activated ILC2 cells into patients with ongoing GvHD or IPS, or prior to onset of GvHD or IPS in a susceptible patient, substantially ameliorates the diseases and improves survival.
  • In some embodiments protocols and methods are provided for isolating, expanding, activating and identifying ILC2 cells, in some embodiments in vitro and/or ex vivo, to be used in therapeutic methods and compositions for GvHD and IPS. For example, and as detailed in the Examples below, in some embodiments IL-25 and/or IL-33 (in some embodiments with IL-7) can activate ILC2 cells inducing production of IL-4, IL-5 and IL-13. Activated ILC2 cells can in some embodiments be used in therapeutic methods and compositions to treat and/or prevent GvHD and IPS by, at least in some aspects, shifting the cytokine micro-environment away form Th1/Th17 to Th2.
  • Indeed, as described in detail in the Examples, it is demonstrated herein that ILC2 cells can in some embodiments be highly sensitive to radiation. Thus, without being bound by any particular theory or mechanism of action, it was surmised that the absence of recipient ILC2 cells in the colon and MLN, may lead to alterations in the microenvironment leading to Th1 polarization in the lower GI tract induced after allo-SCT or allo-BMT. Given ILC2 production of Th2 cytokines and the data indicating that shifting the cytokine micro-environment away form Th1/Th17 to Th2 reduces the severity of GVHD, it was hypothesized that co-transplantation of ILC2 cells could reduce aGvHD and IPS and increase survival after allo-SCT and allo-BMT.
  • As such, provided herein are pharmaceutical composition for treating GvHD and/or IPS. Such pharmaceutical compositions can comprise a therapeutically effective amount of a cell preparation of IL-33 activated ILC2 cells. The ILC2 cells in such a composition can be further characterized as testing positive for expression of lymphoid marker CD90 and/or ICOS, and/or testing negative for expression of lineage-specific lymphoid markers. Moreover, the ILC2 cells can in some embodiments test positive for expression of CD127, ST2, Sca-1, CD25, CD90, and/or ICOS, and/or test negative for expression of lineage markers. As described further in the Examples herein, in addition to being activated by IL-33, ILC2 cells can in some aspects be activated by cytokines IL-25 and/or IL-7, alone or in one or more combinations with IL-33. Moreover, the pharmaceutical compositions provided herein, including those comprising activated ILC2 cells, can be screened for and confirmed to be completely or substantially free of MPP2 cells.
  • The ILC2 cells that are grown and/or activated in vitro can be derived from any suitable source of ILC2 cells, including for example mammalian blood and/or blood products. In some embodiments, the ILC2 cells can be derived from human cord blood cells. Such human cord blood cells can be derived from a donor subject and/or from a patient's own cord blood.
  • The compositions provided herein, particularly those to be administered to a subject or patient, can in some embodiments comprise a pharmaceutically acceptable carrier.
  • In vitro, or otherwise ex vivo, methods are also provided herein for generating activated ILC2 cells, comprising expansion of ILC2 cells from a small number of cells, and activation of such cells to generate cytokines. Such methods can provide the activated ILC2 cells for use in the disclosed pharmaceutical compositions and related therapeutic methods and approaches. These in vitro methods can in some embodiments comprise collecting human cord blood (or other potential source of ILC2 cells from a subject such as peripheral blood stem cells and/or bone marrow), isolating c-Kit positive cells from the cord blood, and culturing the c-Kit positive cells in the presence of an IL-33 cytokine. By culturing the ILC2 cells in the presence of at least IL-33 cytokines the ILC2 cells can be activated. In some embodiments additional cytokines can exposed to the cultured ILC2 cells to further elicit an activated state, including for example IL-25 and IL-7. The IL-33, IL-25 and IL-7 cytokines can be exposed to the ILC2 cells in culture any combination thereof.
  • To confirm the presence of activated ILC2 cells in some embodiments a further step can comprise analyzing the generated ILC2 cells by measuring expression of ST2 and CD127 in the ILC2 cells. Expression of ST2 and CD127 is indicative of the phenotype of ILC2 cells. Such a confirmatory step can rule out the presence of MPP2 cells such that a composition can be characterized as being free or substantially free of MPP2 cells. Additionally, characterization of human ILC2 cells found that the cells expressed CRTH2 and CD161 characteristic surface markers for the expression of ILC2 cells.
  • In some embodiments treatment or therapeutic methods are provided herein, including treatments and therapeutic approaches for GvHD and/or IPS. For example, in some embodiments methods for treating GvHD or IPS in a subject are provided and can comprise providing a subject in need of treatment for GvHD and/or IPS, and administering to the subject a therapeutically effective amount of a cell preparation of IL-33 activated ILC2 cells. The cell preparation administered to a subject can comprise any cell preparation or composition as disclosed herein. For example, such ILC2 cells can have properties including, but not limited to expression of lymphoid markers CD90 and/or ICOS, and/or absence of lymphoid marker lin. In some embodiments, the ILCS cell composition administered to a subject can be characterized by testing positive for expression of CD127, ST2, Sca-1, CD25, CD90, and ICOS, and test negative for expression of lin.
  • The subjects to be treated with the disclosed methods of treatment can comprise any subject in need of therapy, including any subject suffering from or susceptible to GvHD, IPS or related condition. In some embodiments the subject can in some embodiments be a human subject receiving an allo-SCT or allo-BMT
  • In some embodiments the cell preparation of IL-33 activated ILC2 cells can be administered to the subject within 1 to 30 days after receiving the allo-SCT or allo-BMT. In some embodiments, the cell preparation of IL-33 activated ILC2 cells can be administered to the subject within 1 to 365 days after the onset or diagnosis of lower GI tract inflammatory GvHD or IPS.
  • The methods of treatment disclosed herein can in some embodiments reduce the risk and/or severity of acute GvHD associated with allo-SCT or allo-BM by preventing the onset of these conditions or significantly improving the survivability and/or treatability of the conditions. In some embodiments, GvHD and/or IPS can be completely or substantially reversed or overcome. For example, in some embodiments the administration of activated ILC2 cells as provided herein can completely eliminate or substantially decrease the clinical score of lower GI tract GVHD commensurate with a marked decrease in diarrhea and/or abdominal pain or blood in the stool. In some embodiments treatement methods and therapeutic compositions as disclsoed herein can also reverse the symptoms and signs of IPS, such as but not limited to shortness of breath, hypoxemia, or increased work of breathing.
  • In addition to treating GvHD and/or IPS, in some embodiments methods are provided for preventing GvHD or IPS in a subject. Such preventative methods can comprise providing a subject receiving allo-SCT or allo-BMT, and administering to the subject a therapeutically effective amount of a cell preparation of IL-33 activated ILC2 cells as a co-infusion to the allo-SCT or allo-BMT. By administering a cell preparation of IL-33 activated ILC2 cells as a co-infusion with the allo-SCT or allo-BMT, i.e. before, during or shortly after, the onset of GvHD and/or IPS can be prevented or at least substantially minimized. Thus, in some embodiments the disclosed treatments and related methods can comprise administering the cell preparations of IL-33 activated ILC2 cells as a co-infusion with the allo-SCT or allo-BMT. A co-infusion of ILC2 cells with the allo-SCT or allo-BMT can in some aspects comprise infusion of the ILC2 cells at a time sufficiently proximate to the allo-SCT or allo-BMT to prevent or substantially prevent onset of GvHD or IPS. In some embodiments a co-infusion of ILC2 cells with the allo-SCT or allo-BMT can comprise infusion of the ILC2 cells at a time sufficiently proximate to the allo-SCT or allo-BMT to treat and/or substantially reduce any GvHD or IPS symptoms that arise as a result of the allo-SCT or allo-BMT. For example, activated ILC2 cells can be infused at the time of transplantation (allo-SCT or allo-BMT) or after transplantation at the onset of symptoms suggestive of GVHD or IPS.
  • In some embodiments the methods comprising the administration of activated ILC2 cells can impact and cause a shift in the cytokine micro-environment in a subject away from Th1/Th17 to Th2. As discussed herein, subjects suffering from GvHD and/or IPS can have an overabundance of Th1/Th17 and reduced Th2. As such, shifting or rebalancing these cytokine levels by increasing the production of Th2 cytokines in the subject and/or decreasing production of Th1 and/or Th17 cytokines in the subject can ameliorate and/or treat one or more symptoms of GvHD and/or IPS. Accordingly, in some embodiments methods of increasing production of Th2 cytokines in a subject are provided, comprising providing a subject in need of increased Th2 cytokine production, and administering to the subject a therapeutically effective amount of a cell preparation of IL-33 activated ILC2 cells. In some embodiments a subject receiving such a treatment method can comprise a human subject receiving allo-SCT or allo-BMT.
  • Definitions
  • The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the presently disclosed subject matter.
  • While the following terms are believed to be well understood by one of ordinary skill in the art, the following definitions are set forth to facilitate explanation of the presently disclosed subject matter.
  • All technical and scientific terms used herein, unless otherwise defined below, are intended to have the same meaning as commonly understood by one of ordinary skill in the art. References to techniques employed herein are intended to refer to the techniques as commonly understood in the art, including variations on those techniques or substitutions of equivalent techniques that would be apparent to one of skill in the art. While the following terms are believed to be well understood by one of ordinary skill in the art, the following definitions are set forth to facilitate explanation of the presently disclosed subject matter.
  • In describing the presently disclosed subject matter, it will be understood that a number of techniques and steps are disclosed. Each of these has individual benefit and each can also be used in conjunction with one or more, or in some cases all, of the other disclosed techniques.
  • Accordingly, for the sake of clarity, this description will refrain from repeating every possible combination of the individual steps in an unnecessary fashion. Nevertheless, the specification and claims should be read with the understanding that such combinations are entirely within the scope of the invention and the claims.
  • Following long-standing patent law convention, the terms “a”, “an”, and “the” refer to “one or more” when used in this application, including the claims. Thus, for example, reference to “a cell” includes a plurality of such cells, and so forth.
  • Unless otherwise indicated, all numbers expressing quantities of ingredients, reaction conditions, and so forth used in the specification and claims are to be understood as being modified in all instances by the term “about”. Accordingly, unless indicated to the contrary, the numerical parameters set forth in this specification and attached claims are approximations that can vary depending upon the desired properties sought to be obtained by the presently disclosed subject matter.
  • As used herein, the term “about,” when referring to a value or to an amount of a composition, dose, sequence identity (e.g., when comparing two or more nucleotide or amino acid sequences), mass, weight, temperature, time, volume, concentration, percentage, etc., is meant to encompass variations of in some embodiments ±20%, in some embodiments ±10%, in some embodiments ±5%, in some embodiments ±1%, in some embodiments ±0.5%, and in some embodiments ±0.1% from the specified amount, as such variations are appropriate to perform the disclosed methods or employ the disclosed compositions.
  • The term “comprising”, which is synonymous with “including” “containing” or “characterized by” is inclusive or open-ended and does not exclude additional, unrecited elements or method steps. “Comprising” is a term of art used in claim language which means that the named elements are essential, but other elements can be added and still form a construct within the scope of the claim.
  • As used herein, the phrase “consisting of” excludes any element, step, or ingredient not specified in the claim. When the phrase “consists of” appears in a clause of the body of a claim, rather than immediately following the preamble, it limits only the element set forth in that clause; other elements are not excluded from the claim as a whole.
  • As used herein, the phrase “consisting essentially of” limits the scope of a claim to the specified materials or steps, plus those that do not materially affect the basic and novel characteristic(s) of the claimed subject matter.
  • With respect to the terms “comprising”, “consisting of”, and “consisting essentially of”, where one of these three terms is used herein, the presently disclosed and claimed subject matter can include the use of either of the other two terms.
  • As used herein, the term “and/or” when used in the context of a listing of entities, refers to the entities being present singly or in combination. Thus, for example, the phrase “A, B, C, and/or D” includes A, B, C, and D individually, but also includes any and all combinations and subcombinations of A, B, C, and D.
  • Subjects
  • The subject treated, screened, tested, or from which a sample is taken, is desirably a human subject, although it is to be understood that the principles of the disclosed subject matter indicate that the compositions and methods are effective with respect to invertebrate and to all vertebrate species, including mammals, which are intended to be included in the term “subject”. Moreover, a mammal is understood to include any mammalian species in which screening is desirable, particularly agricultural and domestic mammalian species.
  • The disclosed methods and treatments are particularly useful in the testing, screening and/or treatment of warm-blooded vertebrates. Thus, the presently disclosed subject matter concerns mammals and birds.
  • More particularly, provided herein is the testing, screening and/or treatment of mammals such as humans, as well as those mammals of importance due to being endangered (such as Siberian tigers), of economical importance (animals raised on farms for consumption by humans) and/or social importance (animals kept as pets or in zoos) to humans, for instance, carnivores other than humans (such as cats and dogs), swine (pigs, hogs, and wild boars), ruminants (such as cattle, oxen, sheep, giraffes, deer, goats, bison, and camels), and horses. Also provided is the treatment of birds, including the treatment of those kinds of birds that are endangered, kept in zoos, as well as fowl, and more particularly domesticated fowl, i.e., poultry, such as turkeys, chickens, ducks, geese, guinea fowl, and the like, as they are also of economical importance to humans. Thus, provided herein is the treatment of livestock, including, but not limited to, domesticated swine (pigs and hogs), ruminants, horses, poultry, and the like.
  • In some embodiments, the subject to be used in accordance with the presently disclosed subject matter is a subject in need of treatment and/or diagnosis. In some embodiments, a subject can have or be believed to GvHD, IPS or related condition or phenotype, particularly those associated with allo-SCT and/or allo-BMT.
  • Formulations
  • The compositions of the presently disclosed subject matter comprise in some embodiments a composition that includes a pharmaceutically acceptable carrier. Any suitable pharmaceutical formulation can be used to prepare the adenovirus vectors for administration to a subject.
  • For example, suitable formulations can include aqueous and non-aqueous sterile injection solutions which can contain anti-oxidants, buffers, bacteriostats, bactericidal antibiotics and solutes which render the formulation isotonic with the bodily fluids of the intended recipient; and aqueous and non-aqueous sterile suspensions which can include suspending agents and thickening agents. The formulations can be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and can be stored in a frozen or freeze-dried (lyophilized) condition requiring only the addition of sterile liquid carrier, for example water for injections, immediately prior to use. Some exemplary ingredients are SDS, mannitol or another sugar, and phosphate-buffered saline (PBS).
  • It should be understood that in addition to the ingredients particularly mentioned above the formulations of this presently disclosed subject matter can include other agents conventional in the art having regard to the type of formulation in question. For example, sterile pyrogen-free aqueous and non-aqueous solutions can be used.
  • The therapeutic regimens and compositions of the presently disclosed subject matter can be used with additional adjuvants or biological response modifiers including, but not limited to, the cytokines.
  • Administration
  • Administration of the compositions of the presently disclosed subject matter can be by any method known to one of ordinary skill in the art, including, but not limited to intravenous administration, intrasynovial administration, transdermal administration, intramuscular administration, subcutaneous administration, topical administration, rectal administration, intravaginal administration, intratumoral administration, oral administration, buccal administration, nasal administration, parenteral administration, inhalation, and insufflation. In some embodiments, suitable methods for administration of a composition of the presently disclosed subject matter include, but are not limited to intravenous. Alternatively, a composition can be deposited at a site in need of treatment in any other manner. The particular mode of administering a composition of the presently disclosed subject matter depends on various factors, including the distribution and abundance of cells to be treated, additional tissue- or cell-targeting features of the composition, and mechanisms for metabolism or removal of the composition from its site of administration.
  • Dosage
  • An effective dose of a composition of the presently disclosed subject matter is administered to a subject in need thereof. A “therapeutically effective amount” is an amount of the composition sufficient to produce a measurable response (e.g., a reduction in GvHD symptoms). In some embodiments, a therapeutically effective amount is an amount sufficient to reduction and/or ameliorate symptoms in a subject suffering from GvHD and/or IPS. In some embodiments, a therapeutically effective amount is an amount sufficient to improve the health, well-being, prognosis and/or survivability of a subject suffering from GvHD and/or IPS. In some embodiments, a therapeutically effective amount is an amount sufficient to reverse GvHD and/or IPS in a subject suffering from GvHD and/or IPS. In some embodiments, a therapeutically effective amount is an amount sufficient to prevent GvHD and/or IPS in a subject susceptible to GvHD and/or IPS, such as a subject receiving allo-BMT or allo-SCT. In some embodiments, a therapeutically effective amount is an amount sufficient to increase Th2 cytokine production in a subject suffering from GvHD and/or IPS, and/or decrease Th1 and/or Th17 cytokine production in a subject suffering from GvHD and/or IPS. In some embodiments a therapeutically effective amount can comprise an amount that allows for the tapering and discontinuation of other immunosuppressive drugs used to treat/prevent GVHD.
  • Actual dosage levels of active ingredients in the compositions of the presently disclosed subject matter can be varied so as to administer an amount of the active compound(s) that is effective to achieve the desired therapeutic response for a particular subject. The selected dosage level can depend upon the activity of the therapeutic composition, the route of administration, combination with other drugs or treatments, the severity of the condition being treated, and the condition and prior medical history of the subject being treated. However, it is within the skill of the art to start doses of the compositions at levels lower than required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved.
  • After review of the disclosure of the presently disclosed subject matter presented herein, one of ordinary skill in the art can tailor the dosages to an individual patient, taking into account the particular formulation, method for administration to be used with the composition, and severity of the condition, e.g. GvHD and/or IPS. Further calculations of dose can consider patient height and weight, severity and stage of symptoms, and the presence of additional deleterious physical conditions. Such adjustments or variations, as well as evaluation of when and how to make such adjustments or variations, are well known to those of ordinary skill in the art of medicine.
  • EXAMPLES
  • The following Examples are included to further illustrate various embodiments of the presently disclosed subject matter. However, those of ordinary skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the presently disclosed subject matter.
  • Materials and Methods for Examples 1-6 Mice
  • C57BL/6J (H2b) (termed WT), BALB/c, and C57BL/6J×DBA/2 F1 (termed B6D2) were purchased from The Jackson Laboratory, Bar Harbor, Me., United States of America. The generation of enhanced GFP-expressing C57BL/6 mice has been described previously (Panoskaltsis-Mortari et al., Blood 2004; 103(9):3590-8). Donor and recipient mice were age-matched males between 8 and 16 weeks. All experiments were performed in accordance with protocols approved by the University of North Carolina Institutional Animal and Care Use Committee, Chapel Hill, N.C., United States of America.
  • Isolation and Expansion of ILC2 Cells
  • 10 to 12 week old C57BL/6 mice are given 0.4 μg of recombinant mouse IL-17E/IL-25 (R&D systems) for 4 days. On day 5, the cells are isolated from the mesenteric lymph node (MLN) and by peritoneal lavage using 10% serum complete RPMI. ILC2 cells are isolated by negative selection with MAC column using biotinylated antibodies [(Anti-CD8α (53-6.7), anti-CD4 (RM 4.4), anti-CD3ε (145-2C11), anti-γδTCR (UC7-13DS), anti-TER119 (TER-119), anti-B220 (RA3-6B2), anti-CD11c (N418), anti-CD11b (M1/70), anti-NK1.1 (PK136), anti-CD19 (MB19-1), anti-Ly6G (1A8) and anti-CD49b (DX5)] and Streptavidin Microbeads (Miltenyi 130-048-101, Miltenyi Biotec, Cambridge, Mass., United States of America). Cells are cultured for six days in 10% RPMI-c with rIL-7 and rIL-33 (10 ng/ml) changing media every 2 days. On the sixth day the cell purity is determined by flow cytometry and purified if needed as described above. ILC2 activation was evaluated on day 6 by IC staining of IL-5 and IL-13 by flow cytometry.
  • Transplantation Models
  • Total T cells were isolated using Cedarlane T cell recovery column kit (Cedarlane Laboratories, Burlington, N.C., United States of America), followed by Ab depletion using PE-conjugated anti-mouse B220 and anti-mouse CD25 Abs (eBioscience, San Diego, Calif., United States of America) and magnetic bead selection using anti-PE beads (Miltenyi Biotec, Cambridge, Mass., United States of America). T cell depleted bone marrow was prepared as described by Coghill et al., Blood. 2010; 115(23):4914-22. The day prior to transplantation, recipient mice received either 950 cGy (B6D2) or 800cGy (BALB/c) of total body irradiation. For B6 to B6D2 or B6 to BALB/c transplants, recipients were intravenously injected with either 4×106 T cells and 3×106 TCD BM cells, or 5×105 total T cells and 5×106 TCD BM cells, respectively. For ILC2 treatment groups, B6 or BALB/c mice also received 4×106 or 1×106 CD90+ ILC2 respectively. Mice were observed twice weekly for clinical GVHD signs and symptoms based on a previously established clinical scoring system (van Den Brink et al., Journal of Immunology. 2000; 164(1):469-80). ILC2 cells were given either at the time of bone marrow transplantation (supplemented with T cells) or 7 days after bone marrow transplantation. For tumor studies mice were given P815 or mBC-CML tumor cells at the time of bone marrow transplantation.
  • Evaluation of Radiation Sensitivity
  • B6D2 mice received 950 cGy of total body irradiation. 24 hours later cells were isolated and evaluated by flow cytometry. Total cell numbers were established from the absolute event count of live cells gated as Lineage negative, CD90+, ICOS+ or RORγt+ for ILC2 or ILC3 respectively or as CD4+ for T cells.
  • Stereomicroscopy
  • Organs from anesthetized animals were imaged with a Zeiss SteReo Lumar V12 microscope with GFP bandpass filter (Carl Zeiss Microlmaging,
  • Inc., Thornwood, N.Y., United States of America) at room temperature. AxioVision (Carl Zeiss) software was used to determine GFP intensities. Control and treatment recipient organs were imaged using the identical magnification (mag) and exposure (exp) times on day +12: PP-exp 200 ms, mag 40×.
  • Organ GFP Quantification
  • Organs from recipient animals were homogenized and absolute GFP levels determine by ELISA (Cell Biolabs, Inc., San Diego, Calif., United States of America). Detailed experimental procedures were conducted as described by Coghill et al., Blood. 2010; 115(23):4914-22.
  • Cell Isolation from GvHD Target Organs
  • Spleen, liver, lung, and colon were excised and weighed. Lamina propria lymphocytes were isolated using the Miltenyi Lamina propria dissociation kit (130-097-410) as per the manufacturer's instructions (Miltenyi Biotec, Cambridge, Mass., United States of America). Livers and lungs were digested in a solution of 1 mg/ml collagenase A (Roche) and 75 U of DNase I (Sigma-Aldrich, St. Louis, Mo., United States of America) in RPMI 1640/5% newborn calf serum. Digested tissues were treated with ACK lysis buffer to remove RBCs and were passed through 100-μm pore size cell strainers. Leukocytes were collected at the interface of a 40%/80% Percoll (Sigma-Aldrich) in RPMI 1640/5% newborn calf serum. The pelleted cells were washed in PBS/2% FBS. Spleens were teased apart, treated with ACK lysis buffer, and washed in PBS/2% FBS.
  • Example 1
  • Type 2 Innate Lymphoid Cells are Radiation Sensitive.
  • Initially, the effect of radiation therapy on the survival of ILCs was investigated. For this, B6D2 mice were given a lethal dose of radiation and cell survival was determined by flow cytometry. As shown by Hanash et al., ILC3 cells in the LP of the colon but not MLN are resistant to radiation (FIGS. 1A and 1B) (Hanash et al., Immunity. 2012;37(2):339-50). In addition, CD4+ T cells in the LP, spleen and MLN are sensitive to radiation showing significant reduction of total cells within 24 hrs (FIG. 1C). Interestingly, unlike ILC3 cells, ILC2 cells from the LP of the colon were highly sensitive to radiation. Additionally, similar to ILC3 cells, ILC2 cells in the MLN were exquisitely sensitive to the effects of irradiation. In fact, these cells were as radiation sensitive as conventional CD4+ T cells (FIGS. 1C and 1D). Thus, the absence of recipient ILC2 cells in the colon and MLN, may critically lead to alterations in the microenvironment leading to Th1 polarization in the lower GI tract induced after allo-SCT. Given ILC2 production of Th2 cytokines and the data indicating that shifting the cytokine micro-environment away form Th1/Th17 to Th2 reduces the severity of aGVHD, it was hypothesized that co-transplantation of ILC2 cells would reduce aGvHD and increase survival after allo-SCT.
  • Example 2
  • Isolation, Expansion and Activation of ILC2 Cells.
  • One obstacle to be addressed is the limited number of ILC2 cells in normal lymphoid tissue. To address this, a protocol was established for the isolation, expansion, and identification of ILC2 cells isolated from the mouse MLN and pertoneal cavity. ILC2 cells can be identified as CD90+, lin−, ICOS+ with additional expression of CD127, ST2, Sca-1, and CD25. Injection of IL-25 will elicit an increase in ILC2 cells in the MLN and peritoneal cavity and this method was adapted to expand ILC2 in vivo. After 4 days of IL-25 treatment intraperitoneal injection (i.p.) a significant increase in the number of ILC2 cells compared to control mice was found. However, this increase in ILC2 cells was insufficient to generate a significant population to use for in vivo transplantation. After isolation ILC2 cells were expanded in vitro with IL-33 and IL-7 treatment. With the use of IL-33 and IL-7 greater than 10 fold expansion of ILC2 cells in vitro after 6 days compared to IL-25 treated cultures (FIG. 2) was observed. There was a concern that an expansion of a second Lineage negative cell type in our culture had occurred. However, based their expression of additional ILC2 specific surface markers including CD127, ST2 and CD44, it was determined that the cells were bona fide ILC2. It was also determined that ILC2 cells lose expression of CD90 in culture. Further, IL-33 and IL-7 treatment also activates the ILC2 cells increasing the percentage of cultured cells that produced IL-4, IL-5 and IL-13 with no evidence of expansion of other ILC groups; IFN-γ in ILC1 cells; or IL-22 in ILC3 cells.
  • Thus, IL-25 and IL-33 activate ILC2 inducing production of IL-4, IL-5 and IL-13. IL-25 (0.4 ng/injection for 4 days) elicits increased ILC2 in the MLN of B6 mice. IL-33 with IL-7 treatment increases ILC2 proliferation in vitro compared to IL-25 with IL-7 (10 ng/ml each), adjusted average cell number ±SEM. See FIG. 2. Lineage negative, IL-33 activated cells express ILC2 specific surface markers CD127, ST2 and CD44. IL-33 treatment activates ILC2 cells increasing cytokine production. Cells were isolated from MLN and by peritoneal lavage on day 5 of IL-25 injection and lineage depleted by bead separation (Miltenyi Biotec, Cambridge, Mass., United States of America).
  • To determine if the ILC2 cells that were generated ex vivo were significantly contaminated with MPP2 cells, the cells were analyzed for the expression of ST2 and CD127, which are not expressed by MPP2 cells. IT was shown that over 92% of the cells that were generated expressed ST2 and CD127, consistent with the phenotype of ILC2 cells.
  • Example 3
  • Co-Infusion of ILC2 Cells Reduces aGvHD and Increases Recipient Survival
  • Considering activated ILC2 cells expressed high levels of Th2 inducing cytokines, survival studies after allo-SCT co-infusing ILC2 cells were performed. For these experiments, splenic T cells were transplanted with (treatment) or without (ctrl) ILC2 cells. Interestingly, co-transplantation of one dose of ILC2 cells significantly improved survival in the B6 to B6D2 model of aGvHD with 70% of recipients surviving over 60 days (FIG. 3A). Recipients were monitored and scored for GVHD symptoms as previously described (van Den Brink et al., Journal of Immunology. 2000; 164(1):469-80, Fulton et al., Journal of immunology. 2012; 189(4):1765-72). As shown in FIG. 3B, there was a reduction in the clinical GvHD score in mice receiving ILC2 cells, beginning at day 10. In addition, a significant prolongation of survival was found in B6 into BALB/c transplants, a much more stringent MHC completely mis-matched model.
  • Survival studies were performed to determine if ILC2 cells can suppress established aGvHD by infusing ILC2 cells 7 days after transplant. Interestingly, with a single dose of ILC2 cells, survival was improved over untreated recipients (FIG. 3C). In addition, we observed a significant reduction in GvHD scores in those recipients of day 7 ILC2 injection beginning at day 20 (FIG. 3D). These findings have significant clinical implications as there is no cellular treatment for aGvHD after inflammation has begun.
  • Example 4
  • Infused ILC2 Cells Reduce GI Tract T Cell Burden and Limit Th1 and Th17 Induction
  • Donor T cell migration were evaluated using B6-GFP donor T cells and fluorescence stereomicroscopy and GFP ELISA. It was determined that co-infusion of ILC2 cells reduced donor T cells in Peyer's Patches (PP) in the colon and small bowel in a dose dependent manor as determined by GFP intensity. Using Zeiss SteREO microscopy with eGFP bandpass filter, GFP images were taken for each organ and GFP intensities were determined by software analysis. In addition, there is significantly less GFP in tissue homogenates. T cell infiltration into GvHD target organs was analyzed by flow cytometry and it was determined that those recipients of ILC2 cells had reduced lymphocytes in the LP of the colon (FIG. 4A). This finding correlated with reduced total CD4+ T cells and a significant reduction in CD8+ T cells (FIG. 4C). In addition, a significant reduction in the total number of CD4+ and CD8+ T cells that express IFN-γ was observed, as was reduced IL-17A producing CD4+ T cells (FIG. 4D). Interestingly, an increase in IL-4 producing donor T cells was not seen. The development of Treg cells was analyzed and only modest enhancement of FoxP3 expression was found (FIG. 4E). These data indicate that co-infusion of ILC2 reduces aGvHD by limiting the development of inflammatory Th1 and Th17 cells.
  • Example 5
  • ILC2 Cells Migrate to Peyer's Patches in the GI Tract and Produce Th2 Inducing Cytokines
  • LC2 migration to GvHD target organs was evaluated by infusing B6-GFP ILC2 cells. Particularly, ILC2 cells were tracked using B6-GFP ILC2 donor mice for injection of IL-25 and in vitro expansion with IL-33 prior to co-infusion at BMT. IVIS imaging of GFP expressing ILC2 cells in Peyer's patches 12 days after BMT in B6D2 mice was employed. While only limited GFP signal can be can be found in the lamina propria, large clusters of GFP-ILC2 cells were observed in the Peyer's patches on the intestine. Additionally, it was found that ILC2 cells migrate not only to the colon and small bowel but other GvHD target organs by measuring GFP in tissue by ELISA (FIG. 5A). Evaluating GFP positive ILC2 cells after transplant by flow cytometry confirms that the IL-33 activated cells that have been co-infused are innate lymphoid cells as determined by their expression of the lymphoid marker CD90 in the absence of other lineage defining markers including CD3, CD19, CD11 b, CD11 c, B220, Gr-1, NK1.1 or CD49b. It was also confirmed that these innate lymphoid cells are ILC2 by evaluating cytokine expression by intracellular staining IL-5 and IL-13 in ILC2 12 days after transplant. Greater than 90% of donor GFP positive IL-33 activated cells continue to express IL-13 in all target organs (FIG. 5B). Interestingly, IL-5 expression varies among GvHD target organs with more than 50% of ILC2 cells in the GI tract maintaining IL-5 expression. With the increased production of Th2 inducing cytokines in the GI tract we would expect an increase in Th2 expansion. As mentioned above there is not an increase in IL-4 producing T cells in mice that receive ILC2 cells.
  • Example 6
  • Human ILC2 Expansion
  • ILC2 cells were expanded from cord blood cells using the AhR antagonist SP1. Frozen umbilical cord blood cells were taken and stained with antibodies specific for CD34, c-Kit, CD90 and IL-7Rα. The presence of endogenous ILC2 cells in cord blood was analyzed and very few cells were found present. Thus, c-Kit+ cells were isolated from umbilical cord blood and put in the presence of the cytokines IL-33, IL-25 and IL-2 for cell expansion. There was robust expansion of ILC2 cells in the presence of the cytokines IL-2 and IL-33 and the presence of all three cytokines. This was a 10-fold increase over the 20 days in culture. When the cells were evaluated in culture at day 20, two different populations of cells were found—one that was c-Kithi and a second that was c-Kitlo. When the two different cell populations were analyzed, it was found that the c-Kithi population had decreased expression of IL-7Rα compared to the c-Kitlo population.
  • Example 7
  • Use of ILC2 Cells to Treat IPS
  • In addition to their ability to migrate into the LP of the GI tract, it was found that expanded ILC2 cells can migrate into the pulmonary parenchyma (FIG. 6). 1×106 ILC2 cells from eGFP+ mice were infused i.v. and 48 hours later evaluated from the lung parenchyma in B6D2 mice that had been lethally irradiated and received 3×106 B6 TCD bone marrow cells and 3.5×106 B6 T cells. ILC2 cells were distinguished by the expression of GFP and in addition expression of ST2, CD127 and CD90 and absence of lineage markers. There was a strong statistically significant increase in ILC2 cells in the lung after i.v. infusion that was not found in the sham treated animals (p<0.001).
  • Pulmonary GvHD and idiopathic pneumonia syndrome (IPS) are mediated predominantly by Th17 donor T cells. Yi et al., Blood. 2009; 114(14):3101-3112; Carlson et al., Blood. 2009; 113(6):1365-1374. Thus, ILC2 cells can be used to treat ongoing IPS by blocking the generation of Th17 cells.
  • Example 8
  • ILC2 Cells Function in the Absence of Th2 Polarization
  • B6D2 Recipient mice were lethally irradiated and administered donor T cells from B6 wild type or mice lacking the STAT6 gene which is critical for the generation of Th2 T cells. At the time of transplantation, one group of mice receiving STAT6 null T cells received ILC2 cells at a 1:1 ratio with T cells (T cell dose was 4×106 cells). The other group received PBS as a control. Overall survival and clinical score of GVHD were evaluated. N=8 mice/group. ILC2 cells are equally effective in preventing GVHD even when using T cells that cannot become Th2 cells.
  • FIG. 7A is a bar graph indicating that there is no difference in the generation of T cells making IL-4 (Th2) after the infusion of ILC2 cells when comparing mice that received ILC2 cells (grey bar) and those receiving sham treatment (open bar) 12 days after bone marrow transplantation. FIG. 7B illustrates the outcome of recipient mice receiving lethal irradiation (BM only) and infused with haploidentical wild type T cells with ILC2 cells (2:1) ratio (Control) or haploidentical STAT6 knockout T cells with ILC2 cells (2:1) ratio (Treated). STAT6 null T cells cannot generate Th2 cells. This figure demonstrates that the function of ILC2 cells to block GVHD is independent of the generation of Th2 cells. FIG. 7C is a plot showing the clinical scoring of recipient mice receiving lethal irradiation (triangle—BM only) and infused with either haploidentical WT T cells (closed circle—Control) or STAT6 knockout T cells plus ILC2 cells (open circle—Treated). Clinical scores are significantly improved in recipient mice given ILC2 cells with STAT6 knockout T cells compared to those given only WT T cells. FIG. 7D is a bar graph showing an evaluation for FoxP3 expressing T cells in the GI tract 12 days after the administration of haploidentical WT T cells (open bar) or WT T cells supplemented 2:1 with ILC2 cells (closed bar). This demonstrates that there is no quantitative increase in FoxP3-expressing regulatory T cells in the GI tract after ILC2 administration.
  • Example 9
  • ILC2 Cells Function by Enhancing GI Tract Myeloid Derived Suppressor Cells
  • B6D2 recipient mice were lethally irradiated and administered 4×106 B6 T cells with/without a 1:2 ratio (ILC2 cells:T cells) of donor ILC2 cells. Following transplantation, mice were administered anti-GR-1 or anti-Ly6C mAb 2× per week for three weeks. As shown in FIGS. 8A to 8C, the activity of ILC2 cells to block GVHD was completely lost by depleting myeloid cells with these antibodies. In FIG. 8A tolerogeneic dendritic cells from the donor have been enumerated using CD11c and CD103 as markers. Mice receiving ILC2 cells have an increase in the number of tolerogenic DCs in the GI tract after transplantation. FIG. 8B again demonstrates that there is no increase in FoxP3-expressing T cells in the GI tract after bone marrow transplantation comparing mice receiving ILC2 cells (grey bar) with control treatment (open bar). In FIG. 8C haplioidentical mice received lethal irradiation (BM only) and transplantation using wild type bone marrow cells supplemented with T cells with (Treatment) and without (Control) ILC2 cells. 2-3× per week after transplantation receipient mice received anti-Gr-1 monoclonal antibody by intraperitoneal injection (i.p.). Depletion of Gr-1-expressing cells completely eliminated the benefit of infusion of ILC2 cells to prevent GVHD. This indicates that ILC2 cell infusion is dependent on the presence of donor myeloid cells for its function.
  • Example 10
  • ILC2 Cell Infusion Diminishes GI Tract Permeability
  • ILC2 cell infusion diminishes GI tract permeability and infectious complications that are associated with lower GI tract GVHD. As shown in FIG. 9A to 9E, the permeability of the GI tract we from day 4 to 20 was evaluated by administering FITC-dextran to mice after bone marrow transplantation which received either T cells with sham ILC2 infusion versus T cells with ILC2 infusion. There was a marked decrease in the leakage of FITC dextran from the GI tract in mice receiving T cells with ILC2 cells compared to T cells alone. This indicates that ILC2 cells decrease intestinal permeability and the translocation of GI tract flora after transplantation. Treatment groups in FIGS. 9A to 9C are as follows: Normal (lined bar); BM only (checkered bar); Control (open bar), Treated (grey bar). Treatment gropus in FIGS. 9D and 9E are as follows: BM only (triangle); Control (closed circle); Treatment (closed circle).
  • REFERENCES
  • 1. Panoskaltsis-Mortari A, Price A, Hermanson J R, Taras E, Lees C, Serody J S, Blazar B R. In vivo imaging of graft-versus-host-disease in mice. Blood. 2004;103(9):3590-8. doi: 10.1182/blood-2003-08-2827. PubMed PM ID: 14715632.
  • 2. Coghill J M, Carlson M J, Panoskaltsis-Mortari A, West M L, Burgents J E, Blazar B R, Serody J S. Separation of graft-versus-host disease from graft-versus-leukemia responses by targeting CC-chemokine receptor 7 on donor T cells. Blood. 2010;115(23):4914-22. doi: 10.1182/blood-2009-08-239848. PubMed PMID: 20185583; PubMed Central PMCID: PMC2890182.
  • 3. van Den Brink M R, Moore E, Horndasch K J, Crawford J M, Hoffman J, Murphy G F, Burakoff S J. Fas-deficient Ipr mice are more susceptible to graft-versus-host disease. Journal of immunology. 2000;164(1):469-80. PubMed PMID: 10605044.
  • 4. Hanash A M, Dudakov J A, Hua G, O'Connor M H, Young L F, Singer N V, West M L, Jenq R R, Holland A M, Kappel L W, Ghosh A, Tsai J J, Rao U K, Yim N L, Smith O M, Velardi E, Hawryluk E B, Murphy G F, Liu C, Fouser L A, Kolesnick R, Blazar B R, van den Brink M R. Interleukin-22 protects intestinal stem cells from immune-mediated tissue damage and regulates sensitivity to graft versus host disease. Immunity. 2012;37(2):339-50. doi: 10.1016/j.immuni.2012.05.028. PubMed PMID: 22921121; PubMed Central PMCID: PMC3477611.
  • 5. Neill D R, Wong S H, Bellosi A, Flynn R J, Daly M, Langford T K, Bucks C, Kane C M, Fallon P G, Pannell R, Jolin H E, McKenzie A N. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature. 2010;464(7293):1367-70. doi: 10.1038/nature08900. PubMed PMID: 20200518; PubMed Central PMCID: PMC2862165.
  • 6. Fulton L M, Carlson M J, Coghill J M, Ott L E, West M L, Panoskaltsis-Mortari A, Littman D R, Blazar B R, Serody J S. Attenuation of acute graft-versus-host disease in the absence of the transcription factor RORgammat. Journal of immunology. 2012;189(4):1765-72. doi: 10.4049/jimmuno1.1200858. PubMed PMID: 22778391; PubMed Central PMCID: PMC3411855.
  • 7. Yi T, Chen Y, Wang L, et al. Reciprocal differentiation and tissue-specific pathogenesis of Th1, Th2, and Th17 cells in graft-versus-host disease. Blood. 2009;114(14):3101-3112.
  • 8. Carlson M J, West M L, Coghill J M, Panoskaltsis-Mortari A, Blazar B R, Serody J S. In vitro-differentiated TH17 cells mediate lethal acute graft-versus-host disease with severe cutaneous and pulmonary pathologic manifestations. Blood. 2009;113(6):1365-1374.
  • 9. Mjosberg et al., Nat Immunol. 2011; 12(11):1055-1062.
  • 10. Yu et al., Mucosal Immunol. 2013.
  • 11. Kiessling et al., J Exp Med. 1976;143(4):772-780.
  • All references listed herein including but not limited to all patents, patent applications and publications thereof, scientific journal articles, and database entries (e.g., GENBANK® database entries and all annotations available therein) are incorporated herein by reference in their entireties to the extent that they supplement, explain, provide a background for, or teach methodology, techniques, and/or compositions employed herein.
  • It will be understood that various details of the presently disclosed subject matter may be changed without departing from the scope of the presently disclosed subject matter. Furthermore, the foregoing description is for the purpose of illustration only, and not for the purpose of limitation.

Claims (31)

1. A pharmaceutical composition for treating graft versus host disease (GvHD) or idiopathic pneumonia syndrome (IPS), the composition comprising:
a therapeutically effective amount of a cell preparation of IL-33 activated type 2 innate immune cells (ILC2); and
a pharmaceutically acceptable carrier.
2. The pharmaceutical composition of claim 1, wherein the ILC2 cells have the following properties:
test positive for expression of lymphoid marker CD90;
test positive for expression of lymphoid marker ICOS; and
test negative for expression of lympohoid marker lin.
3. The pharmaceutical composition of claim 2, wherein the ILC2 cells test positive for expression of CD127, ST2, Sca-1, CD25, CD90, and ICOS, and test negative for expression of lin.
4. The pharmaceutical composition of claim 3, wherein the ILC2 cells are activated by cytokines IL-33, IL-25, IL-7 and/or combinations thereof.
5. The pharmaceutical composition of claim 1, wherein the ILC2 cells are derived from human cord blood cells.
6. The pharmaceutical composition of claim 1, wherein the pharmaceutical composition is substantially free of MPP2 cells.
7. An in vitro method for generating activated ILC2 cells, comprising:
collecting human cord blood from a subject;
isolating c-Kit positive cells from the cord blood; and
culturing the c-Kit positive cells in the presence of an IL-33 cytokine, whereby IL-33 activated ILC2 cells are generated.
8. The method of claim 7, wherein the ILC2 cells are activated by exposing the c-Kit positive cells in culture to cytokines IL-33, IL-25, IL-7 and/or combinations thereof.
9. The method of claim 7, further comprising analyzing the generated ILC2 cells by measuring expression of ST2 and CD127 in the ILC2 cells, whereby expression of ST2 and CD127 is indicative of the phenotype of ILC2 cells.
10. The method of claim 7, wherein the ILC2 cells are substantially free of MPP2 cells.
11. The method of claim 7, wherein the subject is a human patient or donor.
12. A composition comprising activated ILC2 cells produced by the method of claim 7.
13. A method for treating graft versus host disease (GvHD) or idiopathic pneumonia syndrome (IPS) in a subject, comprising:
providing a subject in need of treatment for GvHD and/or IPS; and
administering to the subject a therapeutically effective amount of a cell preparation of IL-33 activated ILC2 cells.
14. The method of claim 13, wherein the ILC2 cells have the following properties:
test positive for expression of lymphoid marker CD90;
test positive for expression of lymphoid marker ICOS; and
test negative for expression of lympohoid marker lin.
15. The method of claim 14, wherein the ILC2 cells test positive for expression of CD127, ST2, Sca-1, CD25, CD90, and ICOS, and test negative for expression of lin.
16. The method of claim 13, wherein the subject is a human subject receiving an allogeneic stem cell transplant (allo-SCT) or allogeneic bone marrow transplant (allo-BMT).
17. The method of claim 13, wherein the cell preparation of IL-33 activated ILC2 cells is administerd to the subject within 1 to 30 days after receiving the allo-SCT or allo-BMT.
18. The method of claim 13, wherein the risk and/or severity of acute GVHD associated with allo-SCT or allo-BMT is reduced.
19. The method of claim 13, wherein the cell preparation of IL-33 activated ILC2 cells is administered as a co-infusion with the allo-SCT or allo-BMT.
20. The method of claim 13, wherein production of Th2 cytokines in the subject is increased, and/or production of Th1 and/or Th17 cytokines in the subject is decreased.
21. A method for preventing GvHD or IPS in a subject, comprising:
providing a subject receiving allo-SCT or allo-BMT; and
administering to the subject a therapeutically effective amount of a cell preparation of IL-33 activated ILC2 cells.
22. The method of claim 21, wherein the cell preparation of IL-33 activated ILC2 cells is administered as a co-infusion with the allo-SCT or allo-BMT.
23. The method of claim 22, wherein the co-infusion of ILC2 cells with the allo-SCT or allo-BMT comprises infusion of the ILC2 cells at a time sufficiently proximate to the allo-SCT or allo-BMT to prevent or substantially prevent onset of GvHD or IPS.
24. The method of claim 21, wherein the ILC2 cells have the following properties:
test positive for expression of lymphoid marker CD90;
test positive for expression of lymphoid marker ICOS; and
test negative for expression of lympohoid marker lin.
25. The method of claim 24, wherein the ILC2 cells test positive for expression of CD127, ST2, Sca-1, CD25, CD90, and ICOS, and test negative for expression of lin.
26. The method of claim 21, wherein production of Th2 cytokines in the subject is increased, and/or production of Th1 and/or Th17 cytokines in the subject is decreased.
27. A method of increasing production of Th2 cytokines in a subject, comprising:
providing a subject in need of increased Th2 cytokine production; and
administering to the subject a therapeutically effective amount of a cell preparation of IL-33 activated ILC2 cells.
28. The method of claim 27, wherein the subject comprises a human subject receiving allo-SCT or allo-BMT.
29. The method of claim 27, wherein production of Th1 and/or Th17 cytokines is decreased in the subject.
30. The method of claim 27, wherein the ILC2 cells have the following properties:
test positive for expression of lymphoid marker CD90;
test positive for expression of lymphoid marker ICOS; and
test negative for expression of lympohoid marker lin.
31. The method of claim 30, wherein the ILC2 cells test positive for expression of CD127, ST2, Sca-1, CD25, CD90, and ICOS, and test negative for expression of lin.
US15/532,740 2014-12-04 2015-12-04 Compositions and methods for preventing and treating graft versus host disease Abandoned US20170360907A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/532,740 US20170360907A1 (en) 2014-12-04 2015-12-04 Compositions and methods for preventing and treating graft versus host disease

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201462087340P 2014-12-04 2014-12-04
US15/532,740 US20170360907A1 (en) 2014-12-04 2015-12-04 Compositions and methods for preventing and treating graft versus host disease
PCT/US2015/064001 WO2016090250A1 (en) 2014-12-04 2015-12-04 Compositions and methods for preventing and treating graft versus host disease

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/064001 A-371-Of-International WO2016090250A1 (en) 2014-12-04 2015-12-04 Compositions and methods for preventing and treating graft versus host disease

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/598,914 Division US11471517B2 (en) 2014-12-04 2019-10-10 Compositions and methods for preventing and treating graft versus host disease

Publications (1)

Publication Number Publication Date
US20170360907A1 true US20170360907A1 (en) 2017-12-21

Family

ID=56092529

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/532,740 Abandoned US20170360907A1 (en) 2014-12-04 2015-12-04 Compositions and methods for preventing and treating graft versus host disease
US16/598,914 Active 2036-06-02 US11471517B2 (en) 2014-12-04 2019-10-10 Compositions and methods for preventing and treating graft versus host disease

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/598,914 Active 2036-06-02 US11471517B2 (en) 2014-12-04 2019-10-10 Compositions and methods for preventing and treating graft versus host disease

Country Status (2)

Country Link
US (2) US20170360907A1 (en)
WO (1) WO2016090250A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019213660A3 (en) * 2018-05-04 2019-11-28 The Broad Institute, Inc. Compositions and methods for modulating cgrp signaling to regulate innate lymphoid cell inflammatory responses
WO2020197984A1 (en) * 2019-03-22 2020-10-01 Albany Medical College Methods for improving cognitive function
CN112805017A (en) * 2018-07-16 2021-05-14 詹森药业有限公司 Use of innate lymphoid cells to inhibit microglial activation
US11471517B2 (en) 2014-12-04 2022-10-18 The University Of North Carolina At Chapel Hill Compositions and methods for preventing and treating graft versus host disease

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP7231326B2 (en) 2014-11-10 2023-03-01 ジェネンテック, インコーポレイテッド Therapeutic and diagnostic methods for IL-33-mediated disorders
US10093730B2 (en) 2014-11-10 2018-10-09 Genentech, Inc. Anti-interleukin-33 antibodies and uses thereof
MX2019006886A (en) * 2016-12-13 2019-10-30 Limm Therapeutics Sa Methods of treating diseases associated with ilc2 cells.
MX2022011156A (en) 2020-03-13 2022-10-13 Genentech Inc Anti-interleukin-33 antibodies and uses thereof.
WO2022251952A1 (en) * 2021-06-01 2022-12-08 Jefferies Wilfred A Serum free culture for the expansion of type 2 innate lymphoid cells

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7691821B2 (en) * 2001-09-19 2010-04-06 University Of South Florida Inhibition of SHIP to enhance stem cell harvest and transplantation
US7651855B2 (en) * 2003-04-17 2010-01-26 The Trustees Of The University Of Pennsylvania Regulatory T cells and their use in immunotherapy and suppression of autoimmune responses
US20090208517A1 (en) * 2004-08-19 2009-08-20 Bernhard Moser Preparation Of Antigen-Presenting Human Gamma-Delta T Cells And Use In Immunotherapy
EP1795588A1 (en) * 2005-12-07 2007-06-13 Cellerix, S.L. Use of adipose tissue derived mesenchymal stem cells for the treatment of graft versus host disease
WO2014072446A1 (en) * 2012-11-08 2014-05-15 INSERM (Institut National de la Santé et de la Recherche Médicale) Method for inducing il-2-free proliferation of gamma delta t cells
WO2015042521A1 (en) * 2013-09-20 2015-03-26 University Of Virginia Patent Foundation Compositions and methods for treatment of autoimmune and inflammatory diseases and disorders
WO2016090250A1 (en) 2014-12-04 2016-06-09 The University Of North Carolina At Chapel Hill Compositions and methods for preventing and treating graft versus host disease
US9864138B2 (en) 2015-01-05 2018-01-09 The Research Foundation For The State University Of New York Integrated photonics including germanium

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11471517B2 (en) 2014-12-04 2022-10-18 The University Of North Carolina At Chapel Hill Compositions and methods for preventing and treating graft versus host disease
WO2019213660A3 (en) * 2018-05-04 2019-11-28 The Broad Institute, Inc. Compositions and methods for modulating cgrp signaling to regulate innate lymphoid cell inflammatory responses
CN112805017A (en) * 2018-07-16 2021-05-14 詹森药业有限公司 Use of innate lymphoid cells to inhibit microglial activation
WO2020197984A1 (en) * 2019-03-22 2020-10-01 Albany Medical College Methods for improving cognitive function

Also Published As

Publication number Publication date
US20200155656A1 (en) 2020-05-21
US11471517B2 (en) 2022-10-18
WO2016090250A1 (en) 2016-06-09

Similar Documents

Publication Publication Date Title
US11471517B2 (en) Compositions and methods for preventing and treating graft versus host disease
Hao et al. Central nervous system (CNS)–resident natural killer cells suppress Th17 responses and CNS autoimmune pathology
JP2023159331A (en) Therapeutic pooled blood apoptotic cell preparations and uses thereof
ES2765884T3 (en) Expansion methods and evaluation of B lymphocytes and use of expanded B lymphocytes for the treatment of diseases
Lee et al. Phenotype and function of nasal dendritic cells
Kurihara et al. Crosstalk between nonclassical monocytes and alveolar macrophages mediates transplant ischemia-reperfusion injury through classical monocyte recruitment
JP6096677B2 (en) Enhanced allogeneic hematopoietic stem cell transplantation
Hülsdünker et al. Insights into the pathogenesis of GvHD: what mice can teach us about man
WO2014074852A1 (en) Compositions and methods for modulating an immune response
JP2022550132A (en) COMPOSITIONS COMPRISING REGULATORY T CELLS AND METHODS OF MAKING AND USING SAME
Mathä et al. Migration of lung resident group 2 innate lymphoid cells link allergic lung inflammation and liver immunity
JPWO2009145238A1 (en) Immunomodulators and uses thereof
Wirsdörfer et al. Dendritic cell-like cells accumulate in regenerating murine skeletal muscle after injury and boost adaptive immune responses only upon a microbial challenge
JP2024511052A (en) Enriched regulatory T cell population and method for producing the same
CN112805017A (en) Use of innate lymphoid cells to inhibit microglial activation
JPWO2010050167A1 (en) Th2 cell inducing composition, Th2 type therapeutic composition, and use thereof
López-Rodríguez et al. Type I interferons drive MAIT cell functions against bacterial pneumonia
Patel et al. Mouse models of graft-versus-host disease
Lineburg The contribution of cytokine signals, autophagy and antigen presentation to complications associated with allogeneic stem cell transplantation
Gallizioli Identity and functions of dendritic cell subsets in ischaemia-induced neuroinflammation
Hsu The Role of Meningeal Lymphangiogenic Vessels as an Immune-Regulatory Niche during Neuroinflammation
US20200108091A1 (en) Gut-protective effect of rig-1/mavs and sting activation
Lok Imaging innate immune cell behaviour in lymphoid organs
KR20230160305A (en) New treatment for sepsis
JP2024510649A (en) Compositions containing regulatory T cells and methods of using the same

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE UNIVERSITY OF NORTH CAROLINA AT CHAPEL HILL, N

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SERODY, JONATHAN;COGHILL, JAMES;BRUCE, DANNY;AND OTHERS;SIGNING DATES FROM 20160223 TO 20160224;REEL/FRAME:043101/0417

Owner name: REGENTS OF THE UNIVERSITY OF MINNESOTA, MINNESOTA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BLAZAR, BRUCE;STEFANSKI, HEATHER;SIGNING DATES FROM 20160223 TO 20160224;REEL/FRAME:043101/0584

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIV OF NORTH CAROLINA CHAPEL HILL;REEL/FRAME:044002/0362

Effective date: 20170922

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

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

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

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

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